1
|
Zhang Y, Zhou C, Xie Q, Xia L, Liu L, Bao W, Lin H, Xiong X, Zhang H, Zheng Z, Zhao J, Liang W. Dual release scaffolds as a promising strategy for enhancing bone regeneration: an updated review. Nanomedicine (Lond) 2025; 20:371-388. [PMID: 39891431 PMCID: PMC11812394 DOI: 10.1080/17435889.2025.2457317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025] Open
Abstract
Advancements in tissue regeneration, particularly bone regeneration is key area of research due to potential of novel therapeutic approaches. Efforts to reduce reliance on autologous and allogeneic bone grafts have led to the development of biomaterials that promote synchronized and controlled bone healing. However, the use of growth factors is limited by their short half-life, slow tissue penetration, large molecular size and potential toxicity. These factors suggest that traditional delivery methods may be inadequate hence, to address these challenges, new strategies are being explored. These novel approaches include the use of bioactive substances within advanced delivery systems that enable precise spatiotemporal control. Dual-release composite scaffolds offer a promising solution by reducing the need for multiple surgical interventions and simplifying the treatment process. These scaffolds allow for sustained and controlled drug release, enhancing bone repair while minimizing the drawbacks of conventional methods. This review explores various dual-drug release systems, discussing their modes of action, types of drugs used and release mechanisms to improve bone regeneration.
Collapse
Affiliation(s)
- Yongtao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, Zhejiang, China
| | - Qiong Xie
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hongming Lin
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Xiaochun Xiong
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Zeping Zheng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| |
Collapse
|
2
|
Shaikh S, Mehrotra S, van Bochove B, Teotia AK, Singh P, Laurén I, Lindfors NC, Seppälä J, Kumar A. Strontium-Substituted Nanohydroxyapatite Containing Biodegradable 3D Printed Composite Scaffolds for Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:65378-65393. [PMID: 39556416 DOI: 10.1021/acsami.4c16195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Treatment of large-size bone defects is difficult, and acquiring autografts may be challenging due to limited availability. A synthetic patient-specific bone substitute can be developed by using 3D printing technologies in such cases. In the present study, we have developed photocurable composite resins with poly(trimethylene carbonate) (PTMC) containing a high percentage of biodegradable bioactive strontium-substituted nanohydroxyapatite (SrHA, size 30-70 nm). These photocurable resins have then been employed to develop high-surface-area 3D-printed bone substitutes using the digital light processing (DLP) technique. To enhance the surface area of the 3D-printed substitute, cryogels alone and functionalized with bioactive components of bone morphogenetic protein (BMP) and zoledronic acid (ZA) were filled within the 3D-printed scaffold/substitute. The scaffolds were tested in vitro for biocompatibility and functionality in vivo in two therapeutically relevant rat models with large bone defects (4 mm). The porosities of 3D printed scaffolds were found to be 60.1 ± 0.9%, 72.9 ± 0.5%, and 74.3 ± 1.6% for PTMC, PTMC-HA, and PTMC-SrHA, respectively, which is in the range of cancellous bone (50-95%). The thermogravimetric analysis demonstrated the fabrication of 3D printed composites with HA and SrHA concentrations of 51.5 and 57.4 wt %, respectively, in the PTMC matrix. The tensile Young's modulus (E), compressive moduli, and wettability increased post incorporation of SrHA and HA in the PTMC matrix. In vitro and in vivo results revealed that SrHA integrated into the PTMC matrix exhibited good physicochemical and biological properties. Furthermore, the osteoactive molecule-functionalized 3D printed composite scaffolds were found to have an adequate osteoconductive and osteoinductive surface that has shown increased bone regeneration and defect repair in both tibial and cranial bone defects. Our findings thus support the use of PTMC-SrHA composites as next-generation patient-specific synthetic bioactive biodegradable bone substitutes.
Collapse
Affiliation(s)
- Shazia Shaikh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Shreya Mehrotra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Bas van Bochove
- Advanced Organ bioengineering and Therapeutics, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, Enschede 7522 NB, The Netherlands
- Polymer Technology, School of Chemical Engineering, Aalto University, Espoo, FI-00076 Finland
| | - Arun Kumar Teotia
- Polymer Technology, School of Chemical Engineering, Aalto University, Espoo, FI-00076 Finland
| | - Prerna Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Isabella Laurén
- Polymer Technology, School of Chemical Engineering, Aalto University, Espoo, FI-00076 Finland
| | - Nina C Lindfors
- Department of Musculoskeletal and Plastic Surgery, University of Helsinki, Helsinki 00014, Finland
- Helsinki University Central Hospital, Helsinki 00280, Finland
| | - Jukka Seppälä
- Polymer Technology, School of Chemical Engineering, Aalto University, Espoo, FI-00076 Finland
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- Center for Nanosciences, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- The Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- Center of Excellence for Materials in Medicine, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| |
Collapse
|
3
|
Jian X, Han J, Chen J, Xiao S, Deng C. Therapeutic potential of microRNA-engineered exosomes in diabetic wound healing: a meta-analysis. Arch Dermatol Res 2024; 316:493. [PMID: 39066806 DOI: 10.1007/s00403-024-03234-3] [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: 05/12/2024] [Revised: 05/12/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
Diabetic wounds, a prevalent diabetes complication, pose significant challenges in treatment. MicroRNA-engineered exosomes (miR-exo) are a promising new treatment for diabetic wounds; however, their mechanism remains to be completely understood. Therefore, we aimed to conduct a meta-analysis to evaluate the efficacy of miR-exo treatment in the management of diabetic wounds. To achieve this aim, academic databases, including PubMed, Embase, Web of Science, and the Cochrane Library, were searched for papers published before July 4, 2023. Outcome indicators (e.g., rate of wound healing, neovascular count, rate of re-epithelialization, deposition of collagen, breadth of scar, and inflammatory factors) were assessed. Six studies (total of 72 animals) met inclusion criteria and were analyzed. The amalgamated data revealed that miR-exo treatment exhibited superior results compared to those of control therapy. miR-exo treatment significantly enhanced the rate of wound healing, increased the number of neovascular formations, accelerated the rate of re-epithelialization, increased collagen deposition, reduced scar width, while significantly downregulating the expression of inflammatory factors. Our findings indicate that miR-exo treatment augments overall diabetic wound healing, especially when administered in conjunction with innovative dressings. To ascertain the optimal parameters for miR-exo treatment in managing diabetic wounds, future studies must encompass rigorous, large-scale, double-blinded clinical trials while incorporating long-term follow-up assessments for enhanced reliability and accuracy.
Collapse
Affiliation(s)
- Xichao Jian
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, P. R. China
| | - Jiansu Han
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, P. R. China
| | - Junzhe Chen
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, P. R. China
| | - Shune Xiao
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, P. R. China
- Collaborative Innovation Center of Tissue Repair and Regenerative Medicine, Zunyi, 563003, Guizhou, P. R. China
| | - Chengliang Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, P. R. China.
- Collaborative Innovation Center of Tissue Repair and Regenerative Medicine, Zunyi, 563003, Guizhou, P. R. China.
| |
Collapse
|
4
|
Tenkumo T, Koide R, Ogawa T, Yamaguchi H, Suzuki S, Miyashita M, Nakamura K, Wang H, Yoda N, Sasaki K. A triple growth factor strategy for optimizing bone augmentation in mice. J Biomed Mater Res B Appl Biomater 2024; 112:e35447. [PMID: 38997799 DOI: 10.1002/jbm.b.35447] [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: 12/05/2023] [Revised: 04/07/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024]
Abstract
With dental implant treatment becoming the gold standard, the need for effective bone augmentation prior to implantation has grown. This study aims to evaluate a bone augmentation strategy integrating three key growth factors: bone morphogenetic protein-2 (BMP-2), insulin-like growth factor 1 (IGF-1), and vascular endothelial growth factor (VEGF). Collagen scaffolds incorporating BMP-2, IGF-1, or VEGF were fabricated and categorized into five groups based on their content: scaffold alone; BMP-2 alone (BMP-2); BMP-2 and IGF-1 (BI); BMP-2, IGF-1, and VEGF (BIV); and BMP-2 and IGF-1 with an earlier release of VEGF (BI + V). The prepared scaffolds were surgically implanted into the calvarias of C57BL/6JJcl mice, and hard tissue formation was assessed after 10 and 28 days through histological, tomographic, and biochemical analyses. The combination of BMP-2 and IGF-1 induced a greater volume of hard tissue augmentation compared with that of BMP-2 alone, regardless of VEGF supplementation, and these groups had increased levels of cartilage compared with others. The volume of hard tissue formation was greatest in the BIV group. In contrast, the BI + V group exhibited a hard tissue volume similar to that of the BI group. While VEGF and CD31 levels were highest in the BIV group at 10 days, there was no correlation at the same time point between hard tissue formation and the quantity of M2 macrophages. In conclusion, the simultaneous release of BMP-2, IGF-1, and VEGF proved to be effective in promoting bone augmentation.
Collapse
Affiliation(s)
- Taichi Tenkumo
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Rie Koide
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Toru Ogawa
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Hirofumi Yamaguchi
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Shigeki Suzuki
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Makiko Miyashita
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Keisuke Nakamura
- Department of Advanced Free Radical Science, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Han Wang
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Nobuhiro Yoda
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Keiichi Sasaki
- Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
5
|
Remy M, Upara C, Ding QJ, Miszuk JM, Sun H, Hong L. MicroRNA-200c Release from Gelatin-Coated 3D-Printed PCL Scaffolds Enhances Bone Regeneration. ACS Biomater Sci Eng 2024; 10:2337-2350. [PMID: 38531043 PMCID: PMC11005014 DOI: 10.1021/acsbiomaterials.3c01105] [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: 08/08/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
The fabrication of clinically relevant synthetic bone grafts relies on combining multiple biodegradable biomaterials to create a structure that supports the regeneration of defects while delivering osteogenic biomolecules that enhance regeneration. MicroRNA-200c (miR-200c) functions as a potent osteoinductive biomolecule to enhance osteogenic differentiation and bone formation; however, synthetic tissue-engineered bone grafts that sustain the delivery of miR-200c for bone regeneration have not yet been evaluated. In this study, we created novel, multimaterial, synthetic bone grafts from gelatin-coated 3D-printed polycaprolactone (PCL) scaffolds. We attempted to optimize the release of pDNA encoding miR-200c by varying gelatin types, concentrations, and polymer crosslinking materials to improve its functions for bone regeneration. We revealed that by modulating gelatin type, coating material concentration, and polymer crosslinking, we effectively altered the release rates of pDNA encoding miR-200c, which promoted osteogenic differentiation in vitro and bone regeneration in a critical-sized calvarial bone defect animal model. We also demonstrated that crosslinking the gelatin coatings on the PCL scaffolds with low-concentration glutaraldehyde was biocompatible and increased cell attachment. These results strongly indicate the potential use of gelatin-based systems for pDNA encoding microRNA delivery in gene therapy and further demonstrate the effectiveness of miR-200c for enhancing bone regeneration from synthetic bone grafts.
Collapse
Affiliation(s)
- Matthew
T. Remy
- Iowa
Institute for Oral Health Research, College
of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
- Roy
J. Carver Department of Biomedical Engineering, College of Engineering, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Chawin Upara
- Iowa
Institute for Oral Health Research, College
of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Qiong J. Ding
- Iowa
Institute for Oral Health Research, College
of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Jacob M. Miszuk
- Iowa
Institute for Oral Health Research, College
of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Hongli Sun
- Iowa
Institute for Oral Health Research, College
of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Liu Hong
- Iowa
Institute for Oral Health Research, College
of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| |
Collapse
|
6
|
Kolliopoulos V, Harley BA. Mineralized collagen scaffolds for regenerative engineering applications. Curr Opin Biotechnol 2024; 86:103080. [PMID: 38402689 PMCID: PMC10947798 DOI: 10.1016/j.copbio.2024.103080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 02/27/2024]
Abstract
Collagen is a primary constituent of the tissue extracellular matrix. As a result, collagen has been a common component of tissue engineering biomaterials, including those to promote bone regeneration or to investigate cell-material interactions in the context of bone homeostasis or disease. This review summarizes key considerations regarding current state-of-the-art design and use of collagen biomaterials for these applications. We also describe strategic opportunities for collagen biomaterials to address a new era of challenges, including immunomodulation and appropriate consideration of sex and other patient characteristics in biomaterial design.
Collapse
Affiliation(s)
- Vasiliki Kolliopoulos
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Brendan Ac Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
7
|
Poerio A, Mano JF, Cleymand F. Advanced 3D Printing Strategies for the Controlled Delivery of Growth Factors. ACS Biomater Sci Eng 2023; 9:6531-6547. [PMID: 37968925 DOI: 10.1021/acsbiomaterials.3c00873] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
The controlled delivery of growth factors (GFs) from tissue engineered constructs represents a promising strategy to improve tissue repair and regeneration. However, despite their established key role in tissue regeneration, the use of GFs is limited by their short half-life in the in vivo environment, their dose-dependent effectiveness, and their space- and time-dependent activity. Promising results have been obtained both in vitro and in vivo in animal models. Nevertheless, the clinical application of tissue engineered constructs releasing GFs is still challenging due to the several limitations and risks associated with their use. 3D printing and bioprinting, by allowing the microprecise spatial deposition of multiple materials and the fabrication of complex geometries with high resolution, offer advanced strategies for an optimal release of GFs from tissue engineered constructs. This review summarizes the strategies that have been employed to include GFs and their delivery system into biomaterials used for 3D printing applications to optimize their controlled release and to improve both the in vitro and in vivo regeneration processes. The approaches adopted to overcome the above-mentioned limitations are presented, showing the potential of the technology of 3D printing to get one step closer to clinical applications.
Collapse
Affiliation(s)
- Aurelia Poerio
- Institut Jean Lamour, University of Lorraine, Nancy 54011, France
| | - João F Mano
- Department of Chemistry, CICECO─Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - Franck Cleymand
- Institut Jean Lamour, University of Lorraine, Nancy 54011, France
| |
Collapse
|
8
|
Hong YR, Kim TH, Park KH, Kang J, Lee K, Park EK, Kwon TG, Lim JO, Oh CW. rhBMP-2-Conjugated Three-Dimensional-Printed Poly(L-lactide) Scaffold is an Effective Bone Substitute. Tissue Eng Regen Med 2022; 20:69-81. [PMID: 36512177 PMCID: PMC9852414 DOI: 10.1007/s13770-022-00506-9] [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: 09/27/2022] [Revised: 10/27/2022] [Accepted: 10/30/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Bone growth factors, particularly bone morphogenic protein-2 (BMP-2), are required for effective treatment of significant bone loss. Despite the extensive development of bone substitutes, much remains to be desired for wider application in clinical settings. The currently available bone substitutes cannot sustain prolonged BMP-2 release and are inconvenient to use. In this study, we developed a ready-to-use bone substitute by sequential conjugation of BMP to a three-dimensional (3D) poly(L-lactide) (PLLA) scaffold using novel molecular adhesive materials that reduced the operation time and sustained prolonged BMP release. METHODS A 3D PLLA scaffold was printed and BMP-2 was conjugated with alginate-catechol and collagen. PLLA scaffolds were conjugated with different concentrations of BMP-2 and evaluated for bone regeneration in vitro and in vivo using a mouse calvarial model. The BMP-2 release kinetics were analyzed using ELISA. Histological analysis and micro-CT image analysis were performed to evaluate new bone formation. RESULTS The 3D structure of the PLLA scaffold had a pore size of 400 µm and grid thickness of 187-230 µm. BMP-2 was released in an initial burst, followed by a sustained release for 14 days. Released BMP-2 maintained osteoinductivity in vitro and in vivo. Micro-computed tomography and histological findings demonstrate that the PLLA scaffold conjugated with 2 µg/ml of BMP-2 induced optimal bone regeneration. CONCLUSION The 3D-printed PLLA scaffold conjugated with BMP-2 enhanced bone regeneration, demonstrating its potential as a novel bone substitute.
Collapse
Affiliation(s)
- Yu Ri Hong
- Joint Institute for Regenerative Medicine, Kyungpook National University, Bio-Medical Research Institute, Kyungpook National University Hospital, Daegu, 41940, Republic of Korea
| | - Tae-Ho Kim
- Joint Institute for Regenerative Medicine, Kyungpook National University, Bio-Medical Research Institute, Kyungpook National University Hospital, Daegu, 41940, Republic of Korea
| | - Kyeong-Hyeon Park
- School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
- Department of Orthopedic Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Jumi Kang
- Department of Chemistry and Green-Nano Materials Research Center, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Kyueui Lee
- Department of Chemistry and Green-Nano Materials Research Center, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu, 41940, Republic of Korea
| | - Tae-Geon Kwon
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University, Daegu, 41940, Republic of Korea
| | - Jeong Ok Lim
- Joint Institute for Regenerative Medicine, Kyungpook National University, Bio-Medical Research Institute, Kyungpook National University Hospital, Daegu, 41940, Republic of Korea.
- School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
| | - Chang-Wug Oh
- Department of Orthopedic Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea.
| |
Collapse
|
9
|
Susin C, Lee J, Fiorini T, Koo KT, Schüpbach P, Finger Stadler A, Wikesjö UME. Screening of Hydroxyapatite Biomaterials for Alveolar Augmentation Using a Rat Calvaria Critical-Size Defect Model: Bone Formation/Maturation and Biomaterials Resolution. Biomolecules 2022; 12:1677. [PMID: 36421691 PMCID: PMC9687935 DOI: 10.3390/biom12111677] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Natural (bovine-/equine-/porcine-derived) or synthetic hydroxyapatite (HA) biomaterials appear to be the preferred technologies among clinicians for bone augmentation procedures in preparation for implant dentistry. The aim of this study was to screen candidate HA biomaterials intended for alveolar ridge augmentation relative to their potential to support local bone formation/maturation and to assess biomaterial resorption using a routine critical-size rat calvaria defect model. METHODS Eighty adult male Sprague Dawley outbred rats obtained from a approved-breeder, randomized into groups of ten, were used. The calvaria defects (ø8 mm) either received sham surgery (empty control), Bio-Oss (bovine HA/reference control), or candidate biomaterials including bovine HA (Cerabone, DirectOss, 403Z013), and bovine (403Z014) or synthetic HA/ß-TCP (Reprobone, Ceraball) constructs. An 8 wk healing interval was used to capture the biomaterials' resolution. RESULTS All biomaterials displayed biocompatibility. Strict HA biomaterials showed limited, if any, signs of biodegradation/resorption, with the biomaterial area fraction ranging from 22% to 42%. Synthetic HA/ß-TCP constructs showed limited evidence of biodegradation/erosion (biomaterial area fraction ≈30%). Mean linear defect closure in the sham-surgery control approximated 40%. Mean linear defect closure for the Bio-Oss reference control approximated 18% compared with 15-35% for the candidate biomaterials without significant differences between the controls and candidate biomaterials. CONCLUSIONS None of the candidate HA biomaterials supported local bone formation/maturation beyond the native regenerative potential of this rodent model, pointing to their limitations for regenerative procedures. Biocompatibility and biomaterial dimensional stability could suggest their potential utility as long-term defect fillers.
Collapse
Affiliation(s)
- Cristiano Susin
- Laboratory for Applied Periodontal & Craniofacial Research (LAPCR), Division of Comprehensive Oral Health, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jaebum Lee
- Laboratory for Applied Periodontal & Craniofacial Research (LAPCR), Division of Comprehensive Oral Health, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tiago Fiorini
- Laboratory for Applied Periodontal & Craniofacial Research (LAPCR), Division of Comprehensive Oral Health, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Section of Periodontology, School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre 90000-000, Brazil
| | - Ki-Tae Koo
- Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 110-460, Korea
| | | | - Amanda Finger Stadler
- Laboratory for Applied Periodontal & Craniofacial Research (LAPCR), Division of Comprehensive Oral Health, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ulf ME Wikesjö
- Laboratory for Applied Periodontal & Craniofacial Research (LAPCR), Division of Comprehensive Oral Health, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
10
|
Guo A, Zheng Y, Zhong Y, Mo S, Fang S. Effect of chitosan/inorganic nanomaterial scaffolds on bone regeneration and related influencing factors in animal models: A systematic review. Front Bioeng Biotechnol 2022; 10:986212. [PMID: 36394038 PMCID: PMC9643585 DOI: 10.3389/fbioe.2022.986212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/11/2022] [Indexed: 09/19/2023] Open
Abstract
Bone tissue engineering (BTE) provides a promising alternative for transplanting. Due to biocompatibility and biodegradability, chitosan-based scaffolds have been extensively studied. In recent years, many inorganic nanomaterials have been utilized to modify the performance of chitosan-based materials. In order to ascertain the impact of chitosan/inorganic nanomaterial scaffolds on bone regeneration and related key factors, this study presents a systematic comparison of various scaffolds in the calvarial critical-sized defect (CSD) model. A total of four electronic databases were searched without publication date or language restrictions up to April 2022. The Animal Research Reporting of In Vivo Experiments 2.0 guidelines (ARRIVE 2.0) were used to assess the quality of the included studies. Moreover, the risk of bias (RoB) was evaluated via the Systematic Review Center for Laboratory Animal Experimentation (SYRCLE) tool. After the screening, 22 studies were selected. None of these studies achieved high quality or had a low RoB. In the available studies, scaffolds reconstructed bone defects in radically different extensions. Several significant factors were identified, including baseline characteristics, physicochemical properties of scaffolds, surgery details, and scanning or reconstruction parameters of micro-computed tomography (micro-CT). Further studies should focus on not only improving the osteogenic performance of the scaffolds but also increasing the credibility of studies through rigorous experimental design and normative reports.
Collapse
Affiliation(s)
| | | | | | - Shuixue Mo
- College of Stomatology, Guangxi Medical University, Nanning, China
| | - Shanbao Fang
- College of Stomatology, Guangxi Medical University, Nanning, China
| |
Collapse
|
11
|
V. K. AD, Ray S, Arora U, Mitra S, Sionkowska A, Jaiswal AK. Dual drug delivery platforms for bone tissue engineering. Front Bioeng Biotechnol 2022; 10:969843. [PMID: 36172012 PMCID: PMC9511792 DOI: 10.3389/fbioe.2022.969843] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/23/2022] [Indexed: 11/22/2022] Open
Abstract
The dual delivery platforms used in bone tissue engineering provide supplementary bioactive compounds that include distinct medicines and growth factors thereby aiding enhanced bone regeneration. The delivery of these compounds can be adjusted for a short or prolonged time based on the requirement by altering various parameters of the carrier platform. The platforms thus used are fabricated to mimic the niche of the bone microenvironment, either in the form of porous 3D structures, microspheres, or films. Thus, this review article focuses on the concept of dual drug delivery platform and its importance, classification of various platforms for dual drug delivery specific to bone tissue engineering, and finally highlights the foresight into the future direction of these techniques for better clinical applications.
Collapse
Affiliation(s)
- Anupama Devi V. K.
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, India
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | - Sarbajit Ray
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | - Udita Arora
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | - Sunrito Mitra
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | | | - Amit Kumar Jaiswal
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, India
- *Correspondence: Amit Kumar Jaiswal,
| |
Collapse
|
12
|
Howard MT, Wang S, Berger AG, Martin JR, Jalili-Firoozinezhad S, Padera RF, Hammond PT. Sustained release of BMP-2 using self-assembled layer-by-layer film-coated implants enhances bone regeneration over burst release. Biomaterials 2022; 288:121721. [PMID: 35981926 PMCID: PMC10396073 DOI: 10.1016/j.biomaterials.2022.121721] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/08/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022]
Abstract
Current clinical products delivering the osteogenic growth factor bone morphogenetic protein 2 (BMP-2) for bone regeneration have been plagued by safety concerns due to a high incidence of off-target effects resulting from bolus release and supraphysiological doses. Layer-by-layer (LbL) film deposition offers the opportunity to coat bone defect-relevant substrates with thin films containing proteins and other therapeutics; however, control of release kinetics is often hampered by interlayer diffusion of drugs throughout the film during assembly, which causes burst drug release. In this work, we present the design of different laponite clay diffusional barrier layer architectures in self-assembled LbL films to modulate the release kinetics of BMP-2 from the surface of a biodegradable implant. Release kinetics were tuned by incorporating laponite in different film arrangements and with varying deposition techniques to achieve release of BMP-2 over 2 days, 4 days, 14 days, and 30 days. Delivery of a low dose (0.5 μg) of BMP-2 over 2 days and 30 days using these LbL film architectures was then compared in an in vivo rat critical size calvarial defect model to determine the effect of BMP-2 release kinetics on bone regeneration. After 6 weeks, sustained release of BMP-2 over 30 days induced 3.7 times higher bone volume and 7.4 times higher bone mineral density as compared with 2-day release of BMP-2, which did not induce more bone growth than the uncoated scaffold control. These findings represent a crucial step in the understanding of how BMP-2 release kinetics influence treatment efficacy and underscore the necessity to optimize protein delivery methods in clinical formulations for bone regeneration. This work could be applied to the delivery of other therapeutic proteins for which careful tuning of the release rate is a key optimization parameter.
Collapse
Affiliation(s)
- MayLin T Howard
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Sheryl Wang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Adam G Berger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - John R Martin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Sasan Jalili-Firoozinezhad
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| | - Robert F Padera
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02215, United States.
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States.
| |
Collapse
|
13
|
Yuan L, Xu X, Song X, Hong L, Zhang Z, Ma J, Wang X. Effect of bone-shaped nanotube-hydrogel drug delivery system for enhanced osseointegration. BIOMATERIALS ADVANCES 2022; 137:212853. [PMID: 35929281 DOI: 10.1016/j.bioadv.2022.212853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 04/10/2022] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
Anodic titanium dioxide nanotubes (TNT) have a range of beneficial theranostic properties. However, a lack of effective osseointegration is a problem frequently associated with the titanium dental implant surface. Here, we investigated whether bone-shaped nanotube titanium implants could enhance osseointegration via promoting initial release of vascular endothelial growth factor 165 (VEGF165) and dual release of recombinant human bone morphogenetic protein-2 (rhBMP-2). Thus, we generated cylindrical-shaped nanotubes (TNT1) and bone-shaped nanotubes (TNT2) through voltage-varying and time-varying electrochemical anodization methods, respectively. Additionally, we prepared rhBMP-2-loaded cylindrical-shaped nanotubes/VEGF165-loaded hydrogel (TNT-F1) and rhBMP-2-loaded bone-shaped nanotubes/VEGF165-loaded hydrogel (TNT-F2) drug delivery systems. We evaluated the characteristics and release kinetics of the drug delivery systems, and then analyzed the cytocompatibility and osteogenic differentiation of these specimens with mesenchymal stem cells (MSCs) in vitro. Finally, we utilized a rat femur defect model to test the bone formation capacity of nanotube-hydrogel drug delivery system in vivo. Among these different nanotubes structures, the bone-shaped one was the optimum structure for growth factor release.
Collapse
Affiliation(s)
- Lichan Yuan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, China
| | - Xiaoxu Xu
- Nanjing Children's Hospital, Nanjing Medical University, Nanjing 210093, China
| | - Xiaotong Song
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, China
| | - Leilei Hong
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, China
| | - Zhongyin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, China
| | - Junqing Ma
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, China.
| | - Xiaoliang Wang
- Key Laboratory of High Performance Polymer Materials and Technology of Ministry of Education, Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, State Key Laboratory of coordination Chemistry, Nanjing National Laboratory of Nanostructures, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
14
|
Li Y, Fraser D, Mereness J, Van Hove A, Basu S, Newman M, Benoit DSW. Tissue Engineered Neurovascularization Strategies for Craniofacial Tissue Regeneration. ACS APPLIED BIO MATERIALS 2022; 5:20-39. [PMID: 35014834 PMCID: PMC9016342 DOI: 10.1021/acsabm.1c00979] [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] [Indexed: 02/08/2023]
Abstract
Craniofacial tissue injuries, diseases, and defects, including those within bone, dental, and periodontal tissues and salivary glands, impact an estimated 1 billion patients globally. Craniofacial tissue dysfunction significantly reduces quality of life, and successful repair of damaged tissues remains a significant challenge. Blood vessels and nerves are colocalized within craniofacial tissues and act synergistically during tissue regeneration. Therefore, the success of craniofacial regenerative approaches is predicated on successful recruitment, regeneration, or integration of both vascularization and innervation. Tissue engineering strategies have been widely used to encourage vascularization and, more recently, to improve innervation through host tissue recruitment or prevascularization/innervation of engineered tissues. However, current scaffold designs and cell or growth factor delivery approaches often fail to synergistically coordinate both vascularization and innervation to orchestrate successful tissue regeneration. Additionally, tissue engineering approaches are typically investigated separately for vascularization and innervation. Since both tissues act in concert to improve craniofacial tissue regeneration outcomes, a revised approach for development of engineered materials is required. This review aims to provide an overview of neurovascularization in craniofacial tissues and strategies to target either process thus far. Finally, key design principles are described for engineering approaches that will support both vascularization and innervation for successful craniofacial tissue regeneration.
Collapse
Affiliation(s)
- Yiming Li
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - David Fraser
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jared Mereness
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Amy Van Hove
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Sayantani Basu
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Maureen Newman
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States.,Materials Science Program, University of Rochester, Rochester, New York 14627, United States.,Department of Chemical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Biomedical Genetics and Center for Oral Biology, University of Rochester Medical Center, Rochester, New York 14642, United States
| |
Collapse
|
15
|
Dalisson B, Charbonnier B, Aoude A, Gilardino M, Harvey E, Makhoul N, Barralet J. Skeletal regeneration for segmental bone loss: Vascularised grafts, analogues and surrogates. Acta Biomater 2021; 136:37-55. [PMID: 34626818 DOI: 10.1016/j.actbio.2021.09.053] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 09/25/2021] [Accepted: 09/28/2021] [Indexed: 02/08/2023]
Abstract
Massive segmental bone defects (SBD) are mostly treated by removing the fibula and transplanting it complete with blood supply. While revolutionary 50 years ago, this remains the standard treatment. This review considers different strategies to repair SBD and emerging potential replacements for this highly invasive procedure. Prior to the technical breakthrough of microsurgery, researchers in the 1960s and 1970s had begun to make considerable progress in developing non autologous routes to repairing SBD. While the breaktthrough of vascularised bone transplantation solved the immediate problem of a lack of reliable repair strategies, much of their prior work is still relevant today. We challenge the assumption that mimicry is necessary or likely to be successful and instead point to the utility of quite crude (from a materials technology perspective), approaches. Together there are quite compelling indications that the body can regenerate entire bone segments with few or no exogenous factors. This is important, as there is a limit to how expensive a bone repair can be and still be widely available to all patients since cost restraints within healthcare systems are not likely to diminish in the near future. STATEMENT OF SIGNIFICANCE: This review is significant because it is a multidisciplinary view of several surgeons and scientists as to what is driving improvement in segmental bone defect repair, why many approaches to date have not succeeded and why some quite basic approaches can be as effective as they are. While there are many reviews of the literature of grafting and bone repair the relative lack of substantial improvement and slow rate of progress in clinical translation is often overlooked and we seek to challenge the reader to consider the issue more broadly.
Collapse
|
16
|
Cheng WX, Liu YZ, Meng XB, Zheng ZT, Li LL, Ke LQ, Li L, Huang CS, Zhu GY, Pan HD, Qin L, Wang XL, Zhang P. PLGA/β-TCP composite scaffold incorporating cucurbitacin B promotes bone regeneration by inducing angiogenesis. J Orthop Translat 2021; 31:41-51. [PMID: 34804799 PMCID: PMC8571783 DOI: 10.1016/j.jot.2021.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Vascularization is an essential step in successful bone tissue engineering. The induction of angiogenesis in bone tissue engineering can be enhanced through the delivery of therapeutic agents that stimulate vessel and bone formation. In this study, we show that cucurbitacin B (CuB), a tetracyclic terpene derived from Cucurbitaceae family plants, facilitates the induction of angiogenesis in vitro. METHODS We incorporated CuB into a biodegradable poly (lactide-co-glycolide) (PLGA) and β-tricalcium phosphate (β-TCP) biomaterial scaffold (PT/CuB) Using 3D low-temperature rapid prototyping (LT-RP) technology. A rat skull defect model was used to verify whether the drug-incorporated scaffold has the effects of angiogenesis and osteogenesis in vivo for the regeneration of bone defect. Cytotoxicity assay was performed to determine the safe dose range of the CuB. Tube formation assay and western blot assay were used to analyze the angiogenesis effect of CuB. RESULTS PT/CuB scaffold possessed well-designed bio-mimic structure and improved mechanical properties. CuB was linear release from the composite scaffold without affecting pH value. The results demonstrated that the PT/CuB scaffold significantly enhanced neovascularization and bone regeneration in a rat critical size calvarial defect model compared to the scaffold implants without CuB. Furthermore, CuB stimulated angiogenic signaling via up-regulating VEGFR2 and VEGFR-related signaling pathways. CONCLUSION CuB can serve as promising candidate compound for promoting neovascularization and osteogenesis, especially in tissue engineering for repair of bone defects. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE This study highlights the potential use of CuB as a therapeutic agent and strongly support its adoption as a component of composite scaffolds for tissue-engineering of bone repair.
Collapse
Affiliation(s)
- Wen-Xiang Cheng
- Centre for Translational Medicine Research & Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
- Shenzhen Engineering Research Center for Medical Bioactive Materials, Shenzhen, PR China
| | - Yan-Zhi Liu
- Centre for Translational Medicine Research & Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Marine Medical Research Institute, Guangdong Medical University, Zhanjiang, PR China
| | - Xiang-Bo Meng
- Centre for Translational Medicine Research & Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Zheng-Tan Zheng
- Centre for Translational Medicine Research & Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Ling-Li Li
- Centre for Translational Medicine Research & Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Li-Qing Ke
- Centre for Translational Medicine Research & Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Ling Li
- Centre for Translational Medicine Research & Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Cui-Shan Huang
- Centre for Translational Medicine Research & Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Guo-Yuan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, PR China
| | - Hu-Dan Pan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, PR China
| | - Ling Qin
- Centre for Translational Medicine Research & Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Xin-Luan Wang
- Centre for Translational Medicine Research & Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Peng Zhang
- Centre for Translational Medicine Research & Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
- Shenzhen Engineering Research Center for Medical Bioactive Materials, Shenzhen, PR China
| |
Collapse
|
17
|
Personalized Baghdadite scaffolds: stereolithography, mechanics and in vivo testing. Acta Biomater 2021; 132:217-226. [PMID: 33711527 DOI: 10.1016/j.actbio.2021.03.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/10/2021] [Accepted: 03/04/2021] [Indexed: 01/04/2023]
Abstract
An ongoing challenge in the field of orthopedics is to produce a clinically relevant synthetic ceramic scaffold for the treatment of 'critical-sized' bone defects, which cannot heal without intervention. We had developed a bioactive ceramic (baghdadite, Ca₃ZrSi₂O₉) and demonstrated its outstanding bioactivity using traditional manufacturing techniques. Here, we report on the development of a versatile stereolithography printing technology that enabled fabrication of anatomically-shaped and -sized Baghdadite scaffolds. We assessed the in vivo bioactivity of these scaffolds in co-delivering of bone morphogenetic protein-2 (BMP2) and zoledronic acid (ZA) through bioresorbable coatings to induce bone formation and increase retention in a rat model of heterotopic ossification. Micro-computed tomography, histology, mechanical tests pre- and post-implantation, and mechanical modelling were used to assess bone ingrowth and its effects on the mechanics of the scaffolds. Bone ingrowth and the consequent mechanical properties of the scaffolds improved with increasing BMP2 dose. Co-delivery of ZA with BMP2 further improved this outcome. The significant bone formation within the scaffolds functionalized with 10 µg BMP2 and 2 µg ZA made them 2.3 × stiffer and 2.7 × stronger post-implantation and turned these inherently brittle scaffolds into a tough and deformable material. The effects of bone ingrowth on the mechanical properties of scaffolds were captured in a mechanical model that can be used in future clinical studies for non-destructive evaluation of scaffold's stiffness and strength as new bone forms. These results support the practical utilization of our versatile stereolithographic printing methods and BMP2/ZA functionalization to create fit-for-purpose personalized implants for clinical trials. STATEMENT OF SIGNIFICANCE: In this study, we addressed a long-standing challenge of developing a ceramic printing technology that enables fabrication of customizable anatomically-shaped and -sized bioceramic scaffolds with precise internal architectures using an inexpensive desktop printer. We also addressed another challenge related to delivery of pharmaceuticals. BMP2, currently available as a bone-inducing bioactive protein, is clinically administered in a collagen scaffold that has limited moldability and poor mechanical properties. The comparably stiffer and stronger 3D printed personalized Baghdadite scaffolds developed here can be readily functionalized with bioresorbable coatings containing BMP2 ± ZA. These innovations considerably improve on the prior art and are scalable for use in human surgery.
Collapse
|
18
|
Kauffmann P, Raschke D, Tröltzsch M, Santander P, Brockmeyer P, Schliephake H. The use of rhBMP2 for augmentation of established horizontal/vertical defects may require additional use of rhVEGF to achieve significant bone regeneration: An in vivo experimental study. Clin Oral Implants Res 2021; 32:1228-1240. [PMID: 34352150 DOI: 10.1111/clr.13820] [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: 09/01/2020] [Revised: 02/08/2021] [Accepted: 04/07/2021] [Indexed: 11/28/2022]
Abstract
AIM To test the hypothesis that the use of rhBMP2 in established defects requires additional growth factors such as rhVEGF to accomplish effective bone repair. MATERIALS AND METHODS Horizontal/vertical defects of 2 cm length and 1 cm height were created bilaterally in the alveolar crest of the maxillae of 18 minipigs together with the extraction of all premolar teeth and one molar tooth on both sides. After 3 months of healing, defects were augmented with 0.5 g particulate PDLLA/CaCO3 composite loaded with 400 µg rhBMP2/50 µg rhVEGF165 on one side and 800 µg rhBMP2 on the other in 12 test animals, whereas defects in six control animals were sham operated and left unfilled on one side and augmented with blank carriers on the other. After 4 and 13 weeks, the animals were evaluated each for area of new bone formation (mm²) and bone density (area %). RESULTS Augmentations with carriers loaded with 800 g µrhBMP2 failed to induce significantly more bone than in the augmentations with unloaded carrier after 4 and 13 weeks (p = .1000, p = .381). Augmentations with carriers loaded with 400 µg rhBMP2 and 50 µg erhVEGF165 resulted in significantly increased bone formation after 13 weeks (p = .024) compared to blank carriers. Soft tissue in augmentations with combined rhBMP2/rhVEGF165 loading exhibited numerous microvessels compared to soft tissue in augmentations with rhBMP2. CONCLUSIONS It is concluded that effective bone regeneration in augmentations of established alveolar ridge defects may require the application of rhVEGF additionally to rhBMP2.
Collapse
Affiliation(s)
- Philipp Kauffmann
- Department for Oral & Maxillofacial Surgery, Universitätsmedizin Goettingen, Goettingen, Germany
| | - David Raschke
- Department for Oral & Maxillofacial Surgery, Universitätsmedizin Goettingen, Goettingen, Germany
| | - Markus Tröltzsch
- Private Office Ansbach, Germany & Department for Oral & Maxillofacial Surgery, Universitätsmedizin Goettingen, Goettingen, Germany
| | - Petra Santander
- Department of Orthodontics, Universitätsmedizin Göttingen, Goettingen, Germany
| | - Phillip Brockmeyer
- Department for Oral & Maxillofacial Surgery, Universitätsmedizin Goettingen, Goettingen, Germany
| | - Henning Schliephake
- Department for Oral & Maxillofacial Surgery, Universitätsmedizin Goettingen, Goettingen, Germany
| |
Collapse
|
19
|
Subbiah R, Ruehle MA, Klosterhoff BS, Lin AS, Hettiaratchi MH, Willett NJ, Bertassoni LE, García AJ, Guldberg RE. Triple growth factor delivery promotes functional bone regeneration following composite musculoskeletal trauma. Acta Biomater 2021; 127:180-192. [PMID: 33823326 DOI: 10.1016/j.actbio.2021.03.066] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/28/2021] [Accepted: 03/31/2021] [Indexed: 12/20/2022]
Abstract
Successful bone healing in severe trauma depends on early revascularization to restore oxygen, nutrient, growth factor, and progenitor cell supply to the injury. Therapeutic angiogenesis strategies have therefore been investigated to promote revascularization following severe bone injuries; however, results have been inconsistent. This is the first study investigating the effects of dual angiogenic growth factors (VEGF and PDGF) with low-dose bone morphogenetic protein-2 (BMP-2; 2.5 µg) on bone healing in a clinically challenging composite bone-muscle injury model. Our hydrogel-based delivery systems demonstrated a more than 90% protein entrapment efficiency and a controlled simultaneous release of three growth factors over 28 days. Co-stimulation of microvascular fragment constructs with VEGF and PDGF promoted vascular network formation in vitro compared to VEGF or PDGF alone. In an in vivo model of segmental bone and volumetric muscle loss injury, combined VEGF (5 µg) and PDGF (7.5 µg or 15 µg) delivery with a low dose of BMP-2 significantly enhanced regeneration of vascularized bone compared to BMP-2 treatment alone. Notably, the regenerated bone mechanics reached ~60% of intact bone, a value that was previously only achieved by delivery of high-dose BMP-2 (10 µg) in this injury model. Overall, sustained delivery of VEGF, PDFG, and BMP-2 is a promising strategy to promote functional vascularized bone tissue regeneration following severe composite musculoskeletal injury. Although this study is conducted in a clinically relevant composite injury model in rats using a simultaneous release strategy, future studies are necessary to test the regenerative potential of spatiotemporally controlled delivery of triple growth factors on bone healing using large animal models. STATEMENT OF SIGNIFICANCE: Volumetric muscle loss combined with delayed union or non-union bone defect causes deleterious effects on bone regeneration even with the supplementation of bone morphogenetic protein-2 (BMP-2). In this study, the controlled delivery of dual angiogenic growth factors (vascular endothelial growth factor [VEGF] + Platelet-derived growth factor [PDGF]) increases vascular growth in vitro. Co-delivering VEGF+PDGF significantly increase the bone formation efficacy of low-dose BMP-2 and improves the mechanics of regenerated bone in a challenging composite bone-muscle injury model.
Collapse
|
20
|
Ni M, Sun W, Li Y, Ding L, Lin W, Peng H, Zheng Q, Sun J, Li J, Liu H, Yang Y, Xu L, Zhang G. Sox11 Modified Tendon-Derived Stem Cells Promote the Repair of Osteonecrosis of Femoral Head. Cell Transplant 2021; 30:9636897211053870. [PMID: 34699265 PMCID: PMC8552377 DOI: 10.1177/09636897211053870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/26/2021] [Accepted: 09/29/2021] [Indexed: 11/23/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a leading cause of mobility impairment which may lead to a total hip replacement. Recent studies have found tendon derived stem cells (TDSCs) might be an ideal cell source for musculoskeletal tissue regeneration. And our previous study has shown Sox11 could promote osteogenesis of bone marrow-derived MSCs. However, the effect of TDSCs or Sox11 over-expressing TDSCs (TDSCs-Sox11) on bone regeneration in ONFH has not been investigated. In the present study, TDSCs were infected with AAV carrying Sox11 or empty vector. We showed that Sox11 could promote the proliferation and osteogenic differentiation of TDSCs, as well as angiogenesis in vitro. The western blot analysis showed that Sox11 could activate the PI3K/Akt signaling pathway to promote osteogenesis of TDSCs. Finally, using a rabbit model of hormone-induced ONFH, our result demonstrated that local administration of TDSCs or TDSCs overexpressing Sox11 could accelerate bone regeneration in necrotic femoral heads, and TDSCs overexpressing Sox11 showed better effects. TDSCs over-expressing Sox11 might be a promising cell source for stem cell therapy to promote bone regeneration, such as ONFH, fracture, bone defect, and so on.
Collapse
Affiliation(s)
- Ming Ni
- Department of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing, P.R. China
- Contributed equally as first authors
| | - Weiwei Sun
- Department of Anesthesiology, the First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, P.R. China
- Contributed equally as first authors
| | - Yucong Li
- Department of Orthopedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Lingli Ding
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiping Lin
- Department of Orthopedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Haiwen Peng
- Department of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing, P.R. China
| | - Qingyuan Zheng
- Department of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing, P.R. China
| | - Jingyang Sun
- Department of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing, P.R. China
| | - Juncheng Li
- Department of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing, P.R. China
| | - Hao Liu
- Department of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing, P.R. China
| | - Yi Yang
- Central Laboratory, Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Liangliang Xu
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guoqiang Zhang
- Department of Orthopedics, the First Medical Center, Chinese PLA General Hospital, Beijing, P.R. China
| |
Collapse
|
21
|
Subbiah R, Cheng A, Ruehle MA, Hettiaratchi MH, Bertassoni LE, Guldberg RE. Effects of controlled dual growth factor delivery on bone regeneration following composite bone-muscle injury. Acta Biomater 2020; 114:63-75. [PMID: 32688092 DOI: 10.1016/j.actbio.2020.07.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 10/23/2022]
Abstract
The objective of this study was to investigate the controlled release of two growth factors (BMP-2 and VEGF) as a treatment strategy for bone healing in clinically challenging composite injuries, consisting of a femoral segmental bone defect and volumetric muscle loss. This is the first investigation of dual growth factor delivery in a composite injury model using an injectable delivery system consisting of heparin microparticles and alginate gel. The loading efficiency of growth factors into these biomaterials was found to be >90%, revealing a strong affinity of VEGF and BMP-2 to heparin and alginate. The system could achieve simultaneous or tunable release of VEGF and BMP-2 by varying the loading strategy. Single growth factor delivery (VEGF or BMP-2 alone) significantly enhanced vascular growth in vitro. However, no synergistic effect was observed for dual growth factor (BMP-2 + VEGF) delivery in vitro. Effective bone healing was achieved in all treatment groups (BMP-2, simultaneous or tunable delivery of BMP-2 and VEGF) in the composite injury model. The mechanics of the regenerated bone reached a maximum strength of ~52% of intact bone with tunable delivery of VEGF and BMP-2. Overall, simultaneous or tunable co-delivery of low-dose BMP-2 and VEGF failed to fully restore the mechanics of bone in this injury model. Given the severity of the composite injury, VEGF alone may not be sufficient to establish mature and stable blood vessels when compared with previous studies co-delivering BMP-2+VEGF enhanced bone tissue regeneration. Hence, future studies are warranted to develop an alternative treatment strategy focusing on better control over growth factor dose, spatiotemporal delivery, and additional growth factors to regenerate fully functional bone tissue. STATEMENT OF SIGNIFICANCE: We have developed an injectable delivery system consisting of heparin microparticles and an alginate hydrogel that is capable of delivering multiple growth factors in a tunable manner. We used this delivery system to deliver BMP-2 and VEGF in a rodent model of composite bone-muscle injury that mimics clinical type III open fractures. An advanced treatment strategy is necessary for these injuries in order to avoid the negative side effects of high doses of growth factors and because it has been shown that the addition of a muscle injury in this model attenuates the bone regenerative effect of BMP-2. This is the first study to test the effects of dual growth factor delivery (BMP-2/VEGF) on bone healing in a composite bone-muscle injury model and is expected to open up new directions in protein delivery for regenerative medicine.
Collapse
|
22
|
Umbilical Cord Mesenchymal Stem Cell-Derived Nanovesicles Potentiate the Bone-Formation Efficacy of Bone Morphogenetic Protein 2. Int J Mol Sci 2020; 21:ijms21176425. [PMID: 32899307 PMCID: PMC7504262 DOI: 10.3390/ijms21176425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/22/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
Recombinant human bone morphogenetic protein 2 (rhBMP-2) is one of the most potent osteogenic factors used to treat bone loss. However, at higher doses, rhBMP-2 does not necessarily increase bone formation but rather increases the incidence of adverse side effects. Here, we investigated whether umbilical cord mesenchymal stem cell (UCMSC)-derived nanovesicles (NVs) further increase the in vivo bone formation at high doses of rhBMP-2. In the presence of UCMSC-derived NVs, proliferation, migration, and tube formation of human umbilical vein endothelial cells were stimulated in vitro. Furthermore, migration and osteogenesis of human bone marrow-derived mesenchymal stem cells were stimulated. To examine the efficacy of UCMSC-derived NVs on in vivo bone formation, collagen sponges soaked with rhBMP-2 and UCMSC-derived NVs were used in athymic nude mice with calvarial defects. At a high rhBMP-2 dosage (500 ng/mL), UCMSC-derived NVs significantly promoted bone formation in calvarial defects; however, the UCMSC-derived NVs alone did not induce in vivo bone formation. Our results indicate that UCMSC-derived NVs can potentiate the bone formation efficacy of rhBMP-2 at a high dosage.
Collapse
|
23
|
Aghali A, Arman HE. Photoencapsulated-mesenchymal stromal cells in biodegradable thiol-acrylate hydrogels enhance regeneration of craniofacial bone tissue defects. Regen Med 2020; 15:2115-2127. [PMID: 33211632 DOI: 10.2217/rme-2020-0061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 10/20/2020] [Indexed: 01/09/2023] Open
Abstract
Aim: This study investigated biodegradable thiol-acrylate hydrogels as stem cell carriers to facilitate cranial bone regeneration. Materials & methods: Two formulations of thiol-acrylate hydrogels (5 and 15 wt% Poly[ethylene glycol]-diacrylate [PEGDA] hydrogels) were used as stem cell carriers. Bone marrow mesenchymal stromal cells and dental pulp mesenchymal stromal cells were photoencapsulated and cultured in basal or osteogenic medium 3 days before the surgery. Using New Zealand White Rabbits, four defects (5 mm diameter and 2 mm thickness) were created and hydrogel scaffolds were implanted in each rabbit cranium for 6 weeks. Results & Conclusion: AlamarBlue assay showed increasing metabolic activity levels in 5 wt% PEGDA hydrogels than 15 wt% PEGDA hydrogels. Photoencapsulated-mesenchymal stromal cells in 15 wt% PEGDA hydrogels demonstrated significantly increasing alkaline phosphatase activity levels on day 7 compared with days 1 and 3. Histological diagnosis showed 5 wt% PEGDA hydrogels resulted in lower averaged residual gel areas than 15 wt% PEGDA hydrogel specimens and control groups 6 weeks postimplantation.
Collapse
Affiliation(s)
- Arbi Aghali
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47908, USA
| | - Huseyin E Arman
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
24
|
Ding MH, Lozoya EG, Rico RN, Chew SA. The Role of Angiogenesis-Inducing microRNAs in Vascular Tissue Engineering. Tissue Eng Part A 2020; 26:1283-1302. [PMID: 32762306 DOI: 10.1089/ten.tea.2020.0170] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is an important process in tissue repair and regeneration as blood vessels are integral to supply nutrients to a functioning tissue. In this review, the application of microRNAs (miRNAs) or anti-miRNAs that can induce angiogenesis to aid in blood vessel formation for vascular tissue engineering in ischemic diseases such as peripheral arterial disease and stroke, cardiac diseases, and skin and bone tissue engineering is discussed. Endothelial cells (ECs) form the endothelium of the blood vessel and are recognized as the primary cell type that drives angiogenesis and studied in the applications that were reviewed. Besides ECs, mesenchymal stem cells can also play a pivotal role in these applications, specifically, by secreting growth factors or cytokines for paracrine signaling and/or as constituent cells in the new blood vessel formed. In addition to delivering miRNAs or cells transfected/transduced with miRNAs for angiogenesis and vascular tissue engineering, the utilization of extracellular vesicles (EVs), such as exosomes, microvesicles, and EVs collectively, has been more recently explored. Proangiogenic miRNAs and anti-miRNAs contribute to angiogenesis by targeting the 3'-untranslated region of targets to upregulate proangiogenic factors such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor, and hypoxia-inducible factor-1 and increase the transduction of VEGF signaling through the PI3K/AKT and Ras/Raf/MEK/ERK signaling pathways such as phosphatase and tensin homolog or regulating the signaling of other pathways important for angiogenesis such as the Notch signaling pathway and the pathway to produce nitric oxide. In conclusion, angiogenesis-inducing miRNAs and anti-miRNAs are promising tools for vascular tissue engineering for several applications; however, future work should emphasize optimizing the delivery and usage of these therapies as miRNAs can also be associated with the negative implications of cancer.
Collapse
Affiliation(s)
- May-Hui Ding
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Eloy G Lozoya
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Rene N Rico
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Sue Anne Chew
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| |
Collapse
|
25
|
Dashtimoghadam E, Fahimipour F, Tongas N, Tayebi L. Microfluidic fabrication of microcarriers with sequential delivery of VEGF and BMP-2 for bone regeneration. Sci Rep 2020; 10:11764. [PMID: 32678204 PMCID: PMC7366644 DOI: 10.1038/s41598-020-68221-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 05/26/2020] [Indexed: 12/21/2022] Open
Abstract
Wound instability and poor functional vascularization in bone tissue engineering lead to lack of tissue integration and ultimate failure of engineered grafts. In order to harness the regenerative potential of growth factors and stimulate bone healing, present study aims to design multifunctional cell therapy microcarriers with the capability of sequential delivery of essential growth factors, bone morphogenetic protein 2 (BMP-2) and vascular endothelial growth factor (VEGF). An on-chip double emulsion method was implemented to generate monodisperse VEGF encapsulated microcarriers. Bio-inspired poly(3,4-dihydroxyphenethylamine) (PDA) was then functionalized to the microcarriers surface for BMP-2 conjugation. The microcarriers were seeded with mesenchymal stem cells (MSCs) using a dynamic culture technique for cells expansion. Finally, the microcarriers were incorporated into an injectable alginate-RGD hydrogel laden with endothelial cells (ECs) for further analysis. The DNA and calcium content, as well as ALP activity of the construct were analyzed. The confocal fluorescent microscopy was employed to monitor the MSCs and tunneling structure of ECs. Eventually, the capability of developed microcarriers for bone tissue formation was examined in vivo. Microfluidic platform generated monodisperse VEGF-loaded PLGA microcarriers with size-dependent release patterns. Microcarriers generated with the on-chip technique showed more sustained VEGF release profiles compared to the conventional bulk mixing method. The PDA functionalization of microcarriers surface not only provided immobilization of BMP-2 with prolonged bioavailability, but also enhanced the attachment and proliferation of MSCs. Dynamic culturing of microcarriers showcased their great potential to boost MSCs population required for stem cell therapy of bone defects. ALP activity and calcium content analysis of MSCs-laden microcarriers loaded into injectable hydrogels revealed their capability of tunneling formation, vascular cell growth and osteogenic differentiation. The in vivo histology and real-time polymerase chain reaction analysis revealed that transplantation of MSC-laden microcarriers supports ectopic bone formation in the rat model. The presented approach to design bioactive microcarriers offer sustained sequential delivery of bone ECM chemical cues and offer an ideal stabilized 3D microenvironment for patient-specific cell therapy applications. The proposed methodology is readily expandable to integrate other cells and cytokines in a tuned spatiotemporal manner for personalized regenerative medicine.
Collapse
Affiliation(s)
| | - Farahnaz Fahimipour
- Marquette University School of Dentistry, Milwaukee, WI, USA
- Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nikita Tongas
- Marquette University School of Dentistry, Milwaukee, WI, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, USA.
| |
Collapse
|
26
|
Zhang W, Shi W, Wu S, Kuss M, Jiang X, Untrauer JB, Reid SP, Duan B. 3D printed composite scaffolds with dual small molecule delivery for mandibular bone regeneration. Biofabrication 2020; 12:035020. [PMID: 32369796 PMCID: PMC8059098 DOI: 10.1088/1758-5090/ab906e] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Functional reconstruction of craniomaxillofacial defects is challenging, especially for the patients who suffer from traumatic injury, cranioplasty, and oncologic surgery. Three-dimensional (3D) printing/bioprinting technologies provide a promising tool to fabricate bone tissue engineering constructs with complex architectures and bioactive components. In this study, we implemented multi-material 3D printing to fabricate 3D printed PCL/hydrogel composite scaffolds loaded with dual bioactive small molecules (i.e. resveratrol and strontium ranelate). The incorporated small molecules are expected to target several types of bone cells. We systematically studied the scaffold morphologies and small molecule release profiles. We then investigated the effects of the released small molecules from the drug loaded scaffolds on the behavior and differentiation of mesenchymal stem cells (MSCs), monocyte-derived osteoclasts, and endothelial cells. The 3D printed scaffolds, with and without small molecules, were further implanted into a rat model with a critical-sized mandibular bone defect. We found that the bone scaffolds containing the dual small molecules had combinational advantages in enhancing angiogenesis and inhibiting osteoclast activities, and they synergistically promoted MSC osteogenic differentiation. The dual drug loaded scaffolds also significantly promoted in vivo mandibular bone formation after 8 week implantation. This work presents a 3D printing strategy to fabricate engineered bone constructs, which can likely be used as off-the-shelf products to promote craniomaxillofacial regeneration.
Collapse
Affiliation(s)
- Wenhai Zhang
- First Hip Department of Orthopedics, Tianjin Hospital, Tianjin, 300211, China
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shaohua Wu
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- College of Textiles & Clothing; Collaborative Innovation Center of Marine Biomass Fibers, Qingdao University, Qingdao, China
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xiping Jiang
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- College of Medicine, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jason B Untrauer
- Division of Oral & Maxillofacial Surgery, Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - St Patrick Reid
- College of Medicine, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Mechanical and Materials Engineering, University of Nebraska- Lincoln, Lincoln, NE, USA
| |
Collapse
|
27
|
Evaluation of BMP-2 and VEGF loaded 3D printed hydroxyapatite composite scaffolds with enhanced osteogenic capacity in vitro and in vivo. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 112:110893. [PMID: 32409051 DOI: 10.1016/j.msec.2020.110893] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/02/2020] [Accepted: 03/20/2020] [Indexed: 11/22/2022]
Abstract
Large-sized bone defect repair is a challenging task in orthopedic surgery. Porous scaffolds with controlled release of growth factors have been investigated for many years. In this study, a hydroxyapatite composite scaffold was prepared by 3D printing at low temperature and coating with layer-by-layer (LBL) assembly. Bone morphogenic protein-2 (BMP-2) and vascular endothelial growth factors (VEGF) were loaded into the composite scaffolds. The release of dual growth factors was analyzed in vitro. The cell growth and osteogenic differentiation were assessed by culturing MC3T3-E1 cells onto the scaffolds. In an established rabbit model of critical-sized calvarial defect (15 mm in diameter), the osteogenic and angiogenic properties after implantation of scaffolds were evaluated by micro-computed tomography (micro-CT) and stained sections. Our results showed that the scaffolds possessed well-designed porous structure and could release two growth factors in a sustained way. The micro-CT analysis showed that the scaffolds with BMP-2/VEGF could accelerate new bone formation. Findings of immunochemical staining of collagen type I and lectin indicated that better osteogenic and angiogenic properties induced by BMP-2 and VEGF. These results suggested that the novel composite scaffolds combined with BMP-2/VEGF had both osteogenic and angiogenic abilities which could enhance new bone formation with good quality. Thus, the combination of 3D printed scaffolds loaded with BMP-2/VEGF might provide a potential solution for bone repair and regeneration in clinical applications.
Collapse
|
28
|
Piotrowski SL, Wilson L, Dharmaraj N, Hamze A, Clark A, Tailor R, Hill LR, Lai S, Kasper FK, Young S. Development and Characterization of a Rabbit Model of Compromised Maxillofacial Wound Healing. Tissue Eng Part C Methods 2020; 25:160-167. [PMID: 30747042 PMCID: PMC6457326 DOI: 10.1089/ten.tec.2018.0361] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
IMPACT STATEMENT Maxillofacial defects often present the clinical challenge of a compromised wound bed. Preclinical evaluation of tissue engineering techniques developed to facilitate healing and reconstruction typically involves animal models with ideal wound beds. The healthy wound bed scenario does not fully mimic the complex clinical environment in patients, which can lead to technology failure when translating from preclinical in vivo research to clinical use. The reported preclinical animal model of compromised wound healing enables investigation of tissue engineering technologies in a more clinically relevant scenario, potentially fostering translation of promising results in preclinical research to patients.
Collapse
Affiliation(s)
- Stacey L Piotrowski
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, The University of Texas Health Science Center, Houston, Texas.,2 Center for Laboratory Animal Medicine and Care, The University of Texas Health Science Center, Houston, Texas.,3 Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lindsay Wilson
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, The University of Texas Health Science Center, Houston, Texas
| | - Neeraja Dharmaraj
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, The University of Texas Health Science Center, Houston, Texas
| | - Amani Hamze
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, The University of Texas Health Science Center, Houston, Texas
| | - Ashley Clark
- 4 Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center, Houston, Texas
| | - Ramesh Tailor
- 5 Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lori R Hill
- 3 Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephen Lai
- 6 Division of Surgery, Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - F Kurtis Kasper
- 7 Department of Orthodontics, School of Dentistry, Graduate School of Biomedical Sciences, The University of Texas Health Science Center, Houston, Texas
| | - Simon Young
- 1 Department of Oral and Maxillofacial Surgery, School of Dentistry, The University of Texas Health Science Center, Houston, Texas
| |
Collapse
|
29
|
Rothe R, Schulze S, Neuber C, Hauser S, Rammelt S, Pietzsch J. Adjuvant drug-assisted bone healing: Part I – Modulation of inflammation. Clin Hemorheol Microcirc 2020; 73:381-408. [PMID: 31177205 DOI: 10.3233/ch-199102] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Rebecca Rothe
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany
| | - Sabine Schulze
- University Center of Orthopaedics & Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany
- Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany
| | - Sandra Hauser
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics & Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany
- Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Dresden, Germany
- Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
30
|
Juhl O, Zhao N, Merife AB, Cohen D, Friedman M, Zhang Y, Schwartz Z, Wang Y, Donahue H. Aptamer-Functionalized Fibrin Hydrogel Improves Vascular Endothelial Growth Factor Release Kinetics and Enhances Angiogenesis and Osteogenesis in Critically Sized Cranial Defects. ACS Biomater Sci Eng 2019; 5:6152-6160. [PMID: 32190730 PMCID: PMC7079287 DOI: 10.1021/acsbiomaterials.9b01175] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An aging population, decreased activity levels and increased combat injuries, have led to an increase in critical sized bone defects. As more defects are treated using allografts, which is the current standard of care, the deficiencies of allografts are becoming more evident. Allografts lack the angiogenic potential to induce sufficient vasculogenesis to counteract the hypoxic environment associated with critical sized bone defects. In this study, aptamer-functionalized fibrin hydrogels (AFH), engineered to release vascular endothelial growth factor (VEGF), were evaluated for their material properties, growth factor release kinetics, and angiogenic and osteogenic potential in vivo. Aptamer functionalization to native fibrin did not result in significant changes in biocompatibility or hydrogel gelation. However, aptamer functionalization of fibrin did improve the release kinetics of VEGF from AFH and, when compared to FH, reduced the diffusivity and extended the release of VEGF several days longer. VEGF released from AFH, in vivo, increased vascularization to a greater degree, relative to VEGF released from FH, in a murine critical-sized cranial defect. Defects treated with AFH loaded with VEGF, relative to nonhydrogel loaded controls, showed a nominal increase in osteogenesis. Together, these data suggest that AFH more efficiently incorporates and retains VEGF in vitro and in vivo, which then enhances angiogenesis and osteogenesis to a greater extent in vivo than FH.
Collapse
Affiliation(s)
- Otto Juhl
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Nan Zhao
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Anna-Blessing Merife
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - David Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Michael Friedman
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Yue Zhang
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| | - Yong Wang
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Henry Donahue
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia 23220, United States
| |
Collapse
|
31
|
Son J, Kim J, Lee K, Hwang J, Choi Y, Seo Y, Jeon H, Kang HC, Woo HM, Kang BJ, Choi J. DNA aptamer immobilized hydroxyapatite for enhancing angiogenesis and bone regeneration. Acta Biomater 2019; 99:469-478. [PMID: 31494292 DOI: 10.1016/j.actbio.2019.08.047] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/15/2022]
Abstract
In this study, we developed aptamer-conjugated hydroxyapatite (Apt-HA) that promotes bone regeneration and angiogenesis. The 3R02 bivalent aptamer specific to vascular endothelial growth factor (VEGF) was grafted to the hydroxyapatite (HA) surface. Apt-HA was tested for its VEGF protein capture ability to determine the optimal aptamer concentration immobilized on the HA. Apt-HA showed higher VEGF protein capture ability, and faster growth of human umbilical vein endothelial cell (HUVEC) compared to a neat HA with no cytotoxic effects on human osteoblasts. To examine in vivo angiogenesis and bone regeneration, Apt-HA and HA were bilaterally implanted into rabbit tibial metaphyseal defects and analyzed after eight weeks using micro-CT, histology, and histomorphometry. Apt-HA showed significantly increased the volume of new bones, the percentage of bone, and the density of bone mineral in cortical bone. Apt-HA also exhibited the enhanced bone formation at the cortical region in a histomorphometric analysis. Finally, Apt-HA showed significantly increased blood vessel number compared to a neat HA. In summary, the engineered Apt-HA has potential as a bone graft material that may simultaneously promote bone regeneration and angiogenesis. STATEMENT OF SIGNIFICANCE: This work presents a functional hydroxyapatite bone graft using a DNA-based aptamer which overcomes the limitations of existing bone graft materials, which use bound signaling peptides. DNA aptamer immobilized hydroxyapatite enhances the in vitro proliferation of human umbilical vascular endothelial cells as well as in vivo angiogenesis and bone regeneration. DNA aptamer immobilized hydroxyapatite shows no cytotoxic effect on human osteoblasts.
Collapse
|
32
|
Stuckensen K, Lamo-Espinosa JM, Muiños-López E, Ripalda-Cemboráin P, López-Martínez T, Iglesias E, Abizanda G, Andreu I, Flandes-Iparraguirre M, Pons-Villanueva J, Elizalde R, Nickel J, Ewald A, Gbureck U, Prósper F, Groll J, Granero-Moltó F. Anisotropic Cryostructured Collagen Scaffolds for Efficient Delivery of RhBMP-2 and Enhanced Bone Regeneration. MATERIALS 2019; 12:ma12193105. [PMID: 31554158 PMCID: PMC6804013 DOI: 10.3390/ma12193105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 01/22/2023]
Abstract
In the treatment of bone non-unions, an alternative to bone autografts is the use of bone morphogenetic proteins (BMPs), e.g., BMP–2, BMP–7, with powerful osteoinductive and osteogenic properties. In clinical settings, these osteogenic factors are applied using absorbable collagen sponges for local controlled delivery. Major side effects of this strategy are derived from the supraphysiological doses of BMPs needed, which may induce ectopic bone formation, chronic inflammation, and excessive bone resorption. In order to increase the efficiency of the delivered BMPs, we designed cryostructured collagen scaffolds functionalized with hydroxyapatite, mimicking the structure of cortical bone (aligned porosity, anisotropic) or trabecular bone (random distributed porosity, isotropic). We hypothesize that an anisotropic structure would enhance the osteoconductive properties of the scaffolds by increasing the regenerative performance of the provided rhBMP–2. In vitro, both scaffolds presented similar mechanical properties, rhBMP–2 retention and delivery capacity, as well as scaffold degradation time. In vivo, anisotropic scaffolds demonstrated better bone regeneration capabilities in a rat femoral critical-size defect model by increasing the defect bridging. In conclusion, anisotropic cryostructured collagen scaffolds improve bone regeneration by increasing the efficiency of rhBMP–2 mediated bone healing.
Collapse
Affiliation(s)
- Kai Stuckensen
- Department for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, D-97070 Würzburg, Germany
| | - José M Lamo-Espinosa
- Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Emma Muiños-López
- Cell Therapy Area. Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Purificación Ripalda-Cemboráin
- Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Cell Therapy Area. Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | | | - Elena Iglesias
- Cell Therapy Area. Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Gloria Abizanda
- Cell Therapy Area. Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Ion Andreu
- Department of Materials CEIT-TECNUN, Universidad de Navarra, 20018 San Sebastian, Spain
| | | | - Juan Pons-Villanueva
- Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Reyes Elizalde
- Department of Materials CEIT-TECNUN, Universidad de Navarra, 20018 San Sebastian, Spain
| | - Joachim Nickel
- Department Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, D-97070 Würzburg, Germany
| | - Andrea Ewald
- Department for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, D-97070 Würzburg, Germany
| | - Uwe Gbureck
- Department for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, D-97070 Würzburg, Germany
| | - Felipe Prósper
- Cell Therapy Area. Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Department of Haematology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, D-97070 Würzburg, Germany.
| | - Froilán Granero-Moltó
- Department of Orthopaedic Surgery and Traumatology, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- Cell Therapy Area. Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| |
Collapse
|
33
|
Donos N, Dereka X, Calciolari E. The use of bioactive factors to enhance bone regeneration: A narrative review. J Clin Periodontol 2019; 46 Suppl 21:124-161. [DOI: 10.1111/jcpe.13048] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/08/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Nikos Donos
- Centre for Oral Immunobiology & Regenerative Medicine & Centre for Oral Clinical Research (COCR); Institute of Dentistry, Barts & The London School of Medicine & Dentistry; Queen Mary University of London (QMUL); London UK
| | - Xanthippi Dereka
- Centre for Oral Immunobiology & Regenerative Medicine & Centre for Oral Clinical Research (COCR); Institute of Dentistry, Barts & The London School of Medicine & Dentistry; Queen Mary University of London (QMUL); London UK
- Department of Periodontology; School of Dentistry; National and Kapodistrian University of Athens; Athens Greece
| | - Elena Calciolari
- Centre for Oral Immunobiology & Regenerative Medicine & Centre for Oral Clinical Research (COCR); Institute of Dentistry, Barts & The London School of Medicine & Dentistry; Queen Mary University of London (QMUL); London UK
| |
Collapse
|
34
|
He R, Chen J, Jiang J, Liu B, Liang D, Zhou W, Chen W, Wang Y. Synergies of accelerating differentiation of bone marrow mesenchymal stem cells induced by low intensity pulsed ultrasound, osteogenic and endothelial inductive agent. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:674-684. [PMID: 30835554 DOI: 10.1080/21691401.2019.1576704] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In terms to investigate the effect of low-intensity pulsed ultrasound (LIPUS) for differentiation of bone marrow mesenchymal stem cells (BMSCs) and the feasibility of simultaneously inducing into osteoblasts and vascular endothelial cells within the cell culture medium in which two inductive agents are added at the same time with or without LIPUS. Cells were divided into a non-induced group, an osteoblast-induced group, a vascular endothelial-induced group, and a bidirectional differentiation-induced group. Each group was further subdivided into LIPUS and non-LIPUS groups. The cell proliferation in each group was measured by MTT assay. Cell morphological and ultrastructural changes were observed by inverted phase contrast microscopy and transmission electron microscopy. The differentiation of BMSCs was detected by confocal microscopy, flow cytometry and quantitative RT-PCR. Results demonstrated that both osteoblast and vascular endothelial cell differentiation markers were expressed in the bidirectional differentiation induction group and early osteogenesis and angiogenesis appeared. The cell proliferation, differentiation rate and expression of osteocalcin and vWF in the LIPUS groups were all significantly higher than those in the corresponding non-LIPUS group (p < .05), suggesting LIPUS treatment can promote the differentiation efficiency and rate of BMSCs, especially in the bidirectional differentiation induction group. This study suggests the combination of LIPUS and dual-inducing agents could induce and accelerate simultaneous differentiation of BMSCs to osteoblasts and vascular endothelial cells. These findings indicate the method could be applied to research on generating vascularized bone tissue with a shape and function that mimics natural bone to accelerate early osteogenesis and angiogenesis.
Collapse
Affiliation(s)
- Ruixin He
- a State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine , Chongqing Medical University , Chongqing , P.R.China
| | - Junlin Chen
- a State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine , Chongqing Medical University , Chongqing , P.R.China
| | - Jingwei Jiang
- a State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine , Chongqing Medical University , Chongqing , P.R.China
| | - Baoru Liu
- a State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine , Chongqing Medical University , Chongqing , P.R.China
| | - Dandan Liang
- a State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine , Chongqing Medical University , Chongqing , P.R.China
| | - Weichen Zhou
- a State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine , Chongqing Medical University , Chongqing , P.R.China
| | - Wenzhi Chen
- a State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine , Chongqing Medical University , Chongqing , P.R.China.,b The Second Affiliated Hospital of Chongqing Medical University , Chongqing , P.R.China
| | - Yan Wang
- a State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine , Chongqing Medical University , Chongqing , P.R.China
| |
Collapse
|
35
|
Goonoo N, Bhaw-Luximon A. Mimicking growth factors: role of small molecule scaffold additives in promoting tissue regeneration and repair. RSC Adv 2019; 9:18124-18146. [PMID: 35702423 PMCID: PMC9115879 DOI: 10.1039/c9ra02765c] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/02/2019] [Indexed: 12/31/2022] Open
Abstract
The primary aim of tissue engineering scaffolds is to mimic the in vivo environment and promote tissue growth. In this quest, a number of strategies have been developed such as enhancing cell-material interactions through modulation of scaffold physico-chemical parameters. However, more is required for scaffolds to relate to the cell natural environment. Growth factors (GFs) secreted by cells and extracellular matrix (ECM) are involved in both normal repair and abnormal remodeling. The direct use of GFs on their own or when incorporated within scaffolds represent a number of challenges such as release rate, stability and shelf-life. Small molecules have been proposed as promising alternatives to GFs as they are able to minimize or overcome many shortcomings of GFs, in particular immune response and instability. Despite the promise of small molecules in various TE applications, their direct use is limited by nonspecific adverse effects on non-target tissues and organs. Hence, they have been incorporated within scaffolds to localize their actions and control their release to target sites. However, scanty rationale is available which links the chemical structure of these molecules with their mode of action. We herewith review various small molecules either when used on their own or when incorporated within polymeric carriers/scaffolds for bone, cartilage, neural, adipose and skin tissue regeneration.
Collapse
Affiliation(s)
- Nowsheen Goonoo
- Biomaterials, Drug Delivery and Nanotechnology (BDDN) Unit, Centre for Biomedical and Biomaterials Research, University of Mauritius Réduit Mauritius
| | - Archana Bhaw-Luximon
- Biomaterials, Drug Delivery and Nanotechnology (BDDN) Unit, Centre for Biomedical and Biomaterials Research, University of Mauritius Réduit Mauritius
| |
Collapse
|
36
|
Casap N, Rushinek H, Jensen OT. Vertical Alveolar Augmentation Using BMP-2/ACS/Allograft with Printed Titanium Shells to Establish an Early Vascular Scaffold. Oral Maxillofac Surg Clin North Am 2019; 31:473-487. [PMID: 31133506 DOI: 10.1016/j.coms.2019.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Traditional reconstruction of major alveolar ridge deficiency has required autogenous cortical cancellous particulate bone grafts, often augmented with particulate allogeneic components. Now there is a new concept to consider, that of orthoalveolar form. This paradigm shift involves components of the tissue engineering triad of inductive growth factors combined with a matrix and stem cells, together with osteotomies or devices designed for space maintenance. Reported here is early experience with computer technology used to redesign deficient alveolar ridges deriving ideal alveolar-shaped bone-forms made from powdered titanium, sintered by laser at high temperature using rapid prototype technology.
Collapse
Affiliation(s)
- Nardy Casap
- Department of Oral and Maxillofacial Surgery, Hebrew University-Hadassah School of Dental Medicine, PO Box 12272, Jerusalem 91120, Israel.
| | - Heli Rushinek
- Department of Oral and Maxillofacial Surgery, Hebrew University-Hadassah School of Dental Medicine, PO Box 12272, Jerusalem 91120, Israel
| | - Ole T Jensen
- Department of Oral Maxillofacial Surgery, University of Utah, School of Dentistry, 530 Wakara Way, Salt Lake City, Utah 84108, USA
| |
Collapse
|
37
|
Wu Y, Zhang C, Wu J, Han Y, Wu C. Angiogenesis and bone regeneration by mesenchymal stem cell transplantation with danshen in a rabbit model of avascular necrotic femoral head. Exp Ther Med 2019; 18:163-171. [PMID: 31258650 PMCID: PMC6566092 DOI: 10.3892/etm.2019.7556] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/03/2017] [Indexed: 12/27/2022] Open
Abstract
The present study aimed to explore the potential of combined treatment with mesenchymal stem cells (MSCs) and danshen for angiogenesis and bone regeneration in a rabbit model of avascular necrosis of femoral head (ANFH). A rabbit model of ANFH was established using the Shwartzman reaction with methylprednisolone and Escherichia coli endotoxin injection. Magnetic resonance imaging (MRI) and histopathological examination were used to evaluate the rabbit model of ANFH. The rabbits were randomly divided into the danshen group, the MSCs group, the danshen combined with MSCs group and the model group (treated with physiological saline). The expression level of monocyte chemoattractant protein-1 (MCP-1) and stromal cell-derived factor-1 (SDF-1) were determined by reverse transcription polymerase chain reaction (RT-PCR). The expression level of bone morphogenetic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF) were detected by immunofluorescence and the mRNA expression of BMP-2 and VEGF were detected by RT-PCR. Typical osteonecrosis occurred in the rabbit model of ANFH, which indicated that the model was successfully established. MCP-1 and SDF-1 were significantly increased in the model group compared with the normal group (P<0.05). Following the administration of MSCs and Salvia miltiorrhiza (danshen), MSCs labeled with 5-bromo-2-deoxyuridine were observed to be gathered in the necrotic area. The increased migration of MSCs to the necrotic area may be due to the upregulated expression of the chemokines MCP-1 and SDF-1. ANFH treated with danshen combined with MSCs may promote revascularization by increasing the expression of VEGF and BMP-2 in the femoral head, promoting re-ossification and revascularization. Danshen combined with the transplantation of MSCs may be regarded as a novel therapy for the treatment of ANFH in a clinical setting.
Collapse
Affiliation(s)
- Yungang Wu
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Chunwu Zhang
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jianjing Wu
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yan Han
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Chunlei Wu
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
38
|
Sharma S, Xue Y, Xing Z, Yassin MA, Sun Y, Lorens JB, Finne-Wistrand A, Sapkota D, Mustafa K. Adenoviral mediated mono delivery of BMP2 is superior to the combined delivery of BMP2 and VEGFA in bone regeneration in a critical-sized rat calvarial bone defect. Bone Rep 2019; 10:100205. [PMID: 31193299 PMCID: PMC6525280 DOI: 10.1016/j.bonr.2019.100205] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/11/2019] [Accepted: 04/10/2019] [Indexed: 01/30/2023] Open
Abstract
Apart from osteogenesis, neovascularization of the defect area is an important determinant for successful bone healing. Accordingly, several studies have employed the combined delivery of VEGFA and BMP2 for bone regeneration. Nevertheless, the outcomes of these studies are highly variable. The aim of our study was to compare the effectiveness of adenoviral mediated delivery of BMP2 alone and in combination with VEGFA in rat bone marrow stromal cells (rBMSC) seeded on a poly(LLA-co-CL) scaffold in angiogenesis and osteogenesis using a critical-sized rat calvarial defect model. Both mono delivery of BMP2 and the combined delivery of a lower ratio of VEGFA and BMP2 (1:4) led to up-regulation of osteogenic genes (Alpl and Runx2) and increased calcium deposition in vitro, compared with the GFP control. Micro computed tomography (microCT) analysis of the rat calvarial defect at 8 weeks showed that the mono delivery of BMP2 (43.37 ± 3.55% defect closure) was the most effective in healing the bone defect, followed by the combined delivery of BMP2 and VEGFA (27.86 ± 2.89%) and other controls. Histological and molecular analyses supported the microCT findings. Analysis of the angiogenesis, however, showed that both mono delivery of BMP2 and combined delivery of BMP2 and VEGFA had similar angiogenic effect in the calvarial defects. Examination of the key genes related to host response against the adenoviral vectors showed that the current model system was not associated with adverse immune response. Overall, the results show that the mono delivery of BMP2 was superior to the combined delivery of BMP2 and VEGFA in healing the critical-sized rat calvarial bone defect. These findings underscore the importance of appropriate growth factor combination for the successful outcome in bone regeneration.
Collapse
Affiliation(s)
- Sunita Sharma
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Ying Xue
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Zhe Xing
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Mohammed A Yassin
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Yang Sun
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - James B Lorens
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Dipak Sapkota
- Department of Oral Biology, Faculty of Dentistry, 0316 Oslo, Norway
| | - Kamal Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
39
|
Subbiah R, Guldberg RE. Materials Science and Design Principles of Growth Factor Delivery Systems in Tissue Engineering and Regenerative Medicine. Adv Healthc Mater 2019; 8:e1801000. [PMID: 30398700 DOI: 10.1002/adhm.201801000] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/13/2018] [Indexed: 01/22/2023]
Abstract
Growth factors (GFs) are signaling molecules that direct cell development by providing biochemical cues for stem cell proliferation, migration, and differentiation. GFs play a key role in tissue regeneration, but one major limitation of GF-based therapies is dosage-related adverse effects. Additionally, the clinical applications and efficacy of GFs are significantly affected by the efficiency of delivery systems and other pharmacokinetic factors. Hence, it is crucial to design delivery systems that provide optimal activity, stability, and tunable delivery for GFs. Understanding the physicochemical properties of the GFs and the biomaterials utilized for the development of biomimetic GF delivery systems is critical for GF-based regeneration. Many different delivery systems have been developed to achieve tunable delivery kinetics for single or multiple GFs. The identification of ideal biomaterials with tunable properties for spatiotemporal delivery of GFs is still challenging. This review characterizes the types, properties, and functions of GFs, the materials science of widely used biomaterials, and various GF loading strategies to comprehensively summarize the current delivery systems for tunable spatiotemporal delivery of GFs aimed for tissue regeneration applications. This review concludes by discussing fundamental design principles for GF delivery vehicles based on the interactive physicochemical properties of the proteins and biomaterials.
Collapse
Affiliation(s)
- Ramesh Subbiah
- Parker H. Petit Institute for Bioengineering and Bioscience; George W. Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
| | - Robert E. Guldberg
- Parker H. Petit Institute for Bioengineering and Bioscience; George W. Woodruff School of Mechanical Engineering; Georgia Institute of Technology; Atlanta GA 30332 USA
- Phil and Penny Knight Campus for Accelerating Scientific Impact; 6231 University of Oregon; Eugene OR 97403 USA
| |
Collapse
|
40
|
White KA, Olabisi RM. Spatiotemporal Control Strategies for Bone Formation through Tissue Engineering and Regenerative Medicine Approaches. Adv Healthc Mater 2019; 8:e1801044. [PMID: 30556328 DOI: 10.1002/adhm.201801044] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/06/2018] [Indexed: 02/06/2023]
Abstract
Global increases in life expectancy drive increasing demands for bone regeneration. The gold standard for surgical bone repair is autografting, which enjoys excellent clinical outcomes; however, it possesses significant drawbacks including donor site morbidity and limited availability. Although collagen sponges delivered with bone morphogenetic protein, type 2 (BMP2) are a common alternative or supplement, they do not efficiently retain BMP2, necessitating extremely high doses to elicit bone formation. Hence, reports of BMP2 complications are rising, including cancer promotion and ectopic bone formation, the latter inducing complications such as breathing difficulties and neurologic impairments. Thus, efforts to exert spatial control over bone formation are increasing. Several tissue engineering approaches have demonstrated the potential for targeted and controlled bone formation. These approaches include biomaterial scaffolds derived from synthetic sources, e.g., calcium phosphates or polymers; natural sources, e.g., bone or seashell; and immobilized biofactors, e.g., BMP2. Although BMP2 is the only protein clinically approved for use in a surgical device, there are several proteins, small molecules, and growth factors that show promise in tissue engineering applications. This review profiles the tissue engineering advances in achieving control over the location and onset of bone formation (spatiotemporal control) toward avoiding the complications associated with BMP2.
Collapse
Affiliation(s)
- Kristopher A. White
- Department of Chemical and Biochemical Engineering; Rutgers University; 98 Brett Road Piscataway NJ 08854 USA
| | - Ronke M. Olabisi
- Department of Biomedical Engineering; Rutgers University; 599 Taylor Road Piscataway NJ 08854 USA
| |
Collapse
|
41
|
Zhang Q, Qin M, Zhou X, Nie W, Wang W, Li L, He C. Porous nanofibrous scaffold incorporated with S1P loaded mesoporous silica nanoparticles and BMP-2 encapsulated PLGA microspheres for enhancing angiogenesis and osteogenesis. J Mater Chem B 2018; 6:6731-6743. [PMID: 32254690 DOI: 10.1039/c8tb02138d] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Repair of bone defects remains a major clinical challenge due to inadequate or abnormal vascularization in bone substitutes, which commonly leads to inferior bone formation or bone nonunion. Therefore, healing of bone defects requires the coordinated processes of angiogenesis and osteogenesis. In this study, sphingosine-1-phosphate (S1P) was initially loaded into mesoporous silica nanoparticles (MSNs) to form angiogenic microcarriers, which were subsequently embedded into porous nanofibrous poly-l-lactide (PLLA) scaffolds during a thermally induced phase separation (TIPS) process, while bone morphogenetic protein-2 (BMP-2) was encapsulated into poly(lactic-co-glycolic acid) (PLGA) microspheres to obtain osteogenic microcarriers, which were then integrated onto a MSNs/PLLA nanofibrous scaffold by a post seeding method. The osteogenic and angiogenic activities of the resulting dual-bioactive factor containing scaffolds were evaluated both in vitro and in vivo. The simulated drug release studies indicated that both bioactive factors will be released simultaneously and continuously from the fabricated composite scaffold. Moreover, the ectopic bone formation results showed that the sustained release of S1P and BMP-2 from the composite scaffold resulted in a synergistic effect on blood vessel formation and bone regeneration. Taken together, the results showed the promising application of the dual-bioactive factor loaded nanofibrous scaffold for enhanced bone regeneration.
Collapse
Affiliation(s)
- Qianqian Zhang
- Key Laboratory of Science and Technology of Eco-Textiles, Ministry of Education, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| | | | | | | | | | | | | |
Collapse
|
42
|
Chen R, Yu Y, Zhang W, Pan Y, Wang J, Xiao Y, Liu C. Tuning the bioactivity of bone morphogenetic protein-2 with surface immobilization strategies. Acta Biomater 2018; 80:108-120. [PMID: 30218780 DOI: 10.1016/j.actbio.2018.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/31/2018] [Accepted: 09/11/2018] [Indexed: 01/07/2023]
Abstract
Bone morphogenetic protein-2 (BMP-2) involved therapy is of great potential for bone regeneration. However, its clinical application is restricted due to the undesirable bioactivity and relevant complications in vivo. Immobilization of recombinant BMP-2 (rhBMP-2) is an efficient strategy to mimic natural microenvironment and retain its bioactivity. Herein, we present evidences indicating that osteoinductive capacity of rhBMP-2 can be regulated via variant immobilizing approaches. Three representative superficial immobilizing models were employed to fabricate rhBMP-2-immobilized surfaces including physical adsorption (Au/rhBMP-2), covalent grafting (rhBMP-2-SAM-Au) and heparin binding (Hep-SAM-Au/rhBMP-2) (SAM: self-assembled monolayer). Loading capacity, releasing behavior, osteogenic differentiation and signaling pathways involved, as well as the cellular recognition of rhBMP-2 under various immobilization modes were systematically investigated. As a result, disparate immobilizing approaches not only have effects on loading capacity, but also lead to disparity of osteoinduction at the same dosage. Notably, heparin could reinforce the recognition between rhBMP-2 and its receptors (BMPRs) whereas weaken its binding to its antagonist Noggin. Owing to this "selective" binding feature, the favorable osteoinduction and maximum ectopic bone formation can be achieved with the heparin-binding approach. In particular, manipulation of orientation-mediated BMP-2-cell recognition efficiency may be a potential target to design more therapeutic efficient rhBMP-2 delivery system. STATEMENT OF SIGNIFICANCE: Bone morphogenetic protein-2 (BMP-2) is crucial in bone regeneration. However, its clinical application is challenged due to its shorten half-life and supra-physiological dose associated complications. In this study, three representative superficial immobilizing patterns were fabricated through physical adsorption, covalent grafting and electrostatic interaction with heparin respectively. We provided evidences indicating an dose-dependent osteoinductive capacity of immobilized BMP-2. Further, a possible mechanism of rhBMP-2-cell recognition at the interface was presented, highlighting the superior effect of heparin on rhBMP-2 bioactivity. Finally, We proposed a dual mechanism of tuning the bioactivity of immobilized rhBMP-2 through surface immobilization approaches: regulation of the saturated loading capacity and orientation-mediated rhBMP-2-cell recognition. These results provide novel insights into designing criterion of efficient delivery vehicle for rhBMP-2.
Collapse
|
43
|
Sharmin F, O'Sullivan M, Malinowski S, Lieberman JR, Khan Y. Large scale segmental bone defect healing through the combined delivery of VEGF and BMP‐2 from biofunctionalized cortical allografts. J Biomed Mater Res B Appl Biomater 2018; 107:1002-1010. [DOI: 10.1002/jbm.b.34193] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/18/2017] [Accepted: 08/22/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Farzana Sharmin
- Department of Materials Science and EngineeringUniversity of Connecticut Storrs Connecticut
- Institute for Regenerative EngineeringUConn Health Farmington Connecticut
| | | | - Seth Malinowski
- Department of Biomedical EngineeringUniversity of Connecticut Storrs Connecticut
| | - Jay R. Lieberman
- Department of Orthopedic SurgeryKeck School of Medicine of the University of Southern California California Los Angeles
| | - Yusuf Khan
- Department of Materials Science and EngineeringUniversity of Connecticut Storrs Connecticut
- Institute for Regenerative EngineeringUConn Health Farmington Connecticut
- Department of Orthopaedic SurgeryUConn Health Farmington Connecticut
- Department of Biomedical EngineeringUniversity of Connecticut Storrs Connecticut
- UConn Musculoskeletal Institute Farmington Connecticut
| |
Collapse
|
44
|
Calabrese V, Santoro A, Trovato Salinaro A, Modafferi S, Scuto M, Albouchi F, Monti D, Giordano J, Zappia M, Franceschi C, Calabrese EJ. Hormetic approaches to the treatment of Parkinson's disease: Perspectives and possibilities. J Neurosci Res 2018; 96:1641-1662. [PMID: 30098077 DOI: 10.1002/jnr.24244] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/21/2018] [Accepted: 03/21/2018] [Indexed: 01/17/2023]
Abstract
Age-related changes in the brain reflect a dynamic interaction of genetic, epigenetic, phenotypic, and environmental factors that can be temporally restricted or more longitudinally present throughout the lifespan. Fundamental to these mechanisms is the capacity for physiological adaptation through modulation of diverse molecular and biochemical signaling occurring from the intracellular to the network-systemic level throughout the brain. A number of agents that affect the onset and progression of Parkinson's disease (PD)-like effects in experimental models exhibit temporal features, and mechanisms of hormetic dose responses. These findings have particular significance since the hormetic dose response describes the amplitude and range of potential therapeutic effects, thereby affecting the design and conduct of studies of interventions against PD (and other neurodegenerative diseases), and may also be important to a broader consideration of hormetic processes in resilient adaptive responses that might afford protection against the onset and/or progression of PD and related disorders.
Collapse
Affiliation(s)
- Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania.,IBREGENS, Nutraceuticals and Functional Food Biotechnologies Research Associated, University of Catania, Italy
| | - Aurelia Santoro
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania
| | - Sergio Modafferi
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania
| | - Maria Scuto
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania
| | - Ferdaous Albouchi
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania
| | - Daniela Monti
- Department of Experimental, Clinical and Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - James Giordano
- Departments of Neurology and Biochemistry, and Neuroethics Studies Program, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Mario Zappia
- Department of Medical Sciences, Surgical and Advanced Technologies G.F. Ingrassia, Section of Neurosciences, University of Catania, Italy
| | | | - Edward J Calabrese
- Environmental Health Sciences Division, School of Public Health, University of Massachusetts, Amherst, Massachusetts, USA
| |
Collapse
|
45
|
De Witte TM, Fratila-Apachitei LE, Zadpoor AA, Peppas NA. Bone tissue engineering via growth factor delivery: from scaffolds to complex matrices. Regen Biomater 2018; 5:197-211. [PMID: 30094059 PMCID: PMC6077800 DOI: 10.1093/rb/rby013] [Citation(s) in RCA: 305] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023] Open
Abstract
In recent years, bone tissue engineering has emerged as a promising solution to the limitations of current gold standard treatment options for bone related-disorders such as bone grafts. Bone tissue engineering provides a scaffold design that mimics the extracellular matrix, providing an architecture that guides the natural bone regeneration process. During this period, a new generation of bone tissue engineering scaffolds has been designed and characterized that explores the incorporation of signaling molecules in order to enhance cell recruitment and ingress into the scaffold, as well as osteogenic differentiation and angiogenesis, each of which is crucial to successful bone regeneration. Here, we outline and critically analyze key characteristics of successful bone tissue engineering scaffolds. We also explore candidate materials used to fabricate these scaffolds. Different growth factors involved in the highly coordinated process of bone repair are discussed, and the key requirements of a growth factor delivery system are described. Finally, we concentrate on an analysis of scaffold-based growth factor delivery strategies found in the recent literature. In particular, the incorporation of two-phase systems consisting of growth factor-loaded nanoparticles embedded into scaffolds shows great promise, both by providing sustained release over a therapeutically relevant timeframe and the potential to sequentially deliver multiple growth factors.
Collapse
Affiliation(s)
- Tinke-Marie De Witte
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Additive Manufacturing Laboratory, Department of Biomechanical Engineering, Delft University of Technology (TU Delft), CD Delft, The Netherlands
| | - Lidy E Fratila-Apachitei
- Additive Manufacturing Laboratory, Department of Biomechanical Engineering, Delft University of Technology (TU Delft), CD Delft, The Netherlands
| | - Amir A Zadpoor
- Additive Manufacturing Laboratory, Department of Biomechanical Engineering, Delft University of Technology (TU Delft), CD Delft, The Netherlands
| | - Nicholas A Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, and Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
46
|
Kowalczewski CJ, Saul JM. Biomaterials for the Delivery of Growth Factors and Other Therapeutic Agents in Tissue Engineering Approaches to Bone Regeneration. Front Pharmacol 2018; 9:513. [PMID: 29896102 PMCID: PMC5986909 DOI: 10.3389/fphar.2018.00513] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/27/2018] [Indexed: 12/14/2022] Open
Abstract
Bone fracture followed by delayed or non-union typically requires bone graft intervention. Autologous bone grafts remain the clinical "gold standard". Recently, synthetic bone grafts such as Medtronic's Infuse Bone Graft have opened the possibility to pharmacological and tissue engineering strategies to bone repair following fracture. This clinically-available strategy uses an absorbable collagen sponge as a carrier material for recombinant human bone morphogenetic protein 2 (rhBMP-2) and a similar strategy has been employed by Stryker with BMP-7, also known as osteogenic protein-1 (OP-1). A key advantage to this approach is its "off-the-shelf" nature, but there are clear drawbacks to these products such as edema, inflammation, and ectopic bone growth. While there are clinical challenges associated with a lack of controlled release of rhBMP-2 and OP-1, these are among the first clinical examples to wed understanding of biological principles with biochemical production of proteins and pharmacological principles to promote tissue regeneration (known as regenerative pharmacology). After considering the clinical challenges with such synthetic bone grafts, this review considers the various biomaterial carriers under investigation to promote bone regeneration. This is followed by a survey of the literature where various pharmacological approaches and molecular targets are considered as future strategies to promote more rapid and mature bone regeneration. From the review, it should be clear that pharmacological understanding is a key aspect to developing these strategies.
Collapse
Affiliation(s)
| | - Justin M Saul
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, United States
| |
Collapse
|
47
|
Yao Q, Liu Y, Selvaratnam B, Koodali RT, Sun H. Mesoporous silicate nanoparticles/3D nanofibrous scaffold-mediated dual-drug delivery for bone tissue engineering. J Control Release 2018; 279:69-78. [PMID: 29649529 DOI: 10.1016/j.jconrel.2018.04.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/01/2018] [Accepted: 04/06/2018] [Indexed: 12/20/2022]
Abstract
Controlled delivery systems play a critical role in the success of bone morphogenetic proteins (i.e., BMP2 and BMP7) for challenged bone repair. Instead of single-drug release that is currently and commonly prevalent, dual-drug delivery strategies are highly desired to achieve effective bone regeneration because natural bone repair process is driven by multiple factors. Particularly, angiogenesis is essential for osteogenesis and requires more than just one factor (e.g., Vascular Endothelial Growth Factor, VEGF). Therefore, we developed a novel mesoporous silicate nanoparticles (MSNs) incorporated-3D nanofibrous gelatin (GF) scaffold for dual-delivery of BMP2 and deferoxamine (DFO). DFO is a hypoxia-mimetic drug that can activate hypoxia-inducible factor-1 alpha (HIF-1α), and trigger subsequent angiogenesis. Sustained BMP2 release system was achieved through encapsulation into large-pored MSNs, while the relative short-term release of DFO was engineered through covalent conjugation with chitosan to reduce its cytotoxicity and elongate its half-life. Both MSNs and DFO were incorporated onto a porous 3D GF scaffold to serve as a biomimetic osteogenic microenvironment. Our data indicated that DFO and BMP2 were released from a scaffold at different release rates (10 vs 28 days) yet maintained their angiogenic and osteogenic ability, respectively. Importantly, our data indicated that the released DFO significantly improved BMP2-induced osteogenic differentiation where the dose/duration was important for its effects in both mouse and human stem cell models. Thus, we developed a novel and tunable MSNs/GF 3D scaffold-mediated dual-drug delivery system and studied the potential application of the both FDA-approved DFO and BMP2 for bone tissue engineering.
Collapse
Affiliation(s)
- Qingqing Yao
- Department of Biomedical Engineering, University of South Dakota, BioSNTR, Sioux Falls, SD 57107, USA; School of Ophthalmology and Optometry, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou, Zhejiang 325027, China; Institute of Advanced Materials for Nano-Bio Applications, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yangxi Liu
- Department of Biomedical Engineering, University of South Dakota, BioSNTR, Sioux Falls, SD 57107, USA
| | - Balaranjan Selvaratnam
- Department of Chemistry, University of South Dakota, 414 E. Clark Street, Vermillion 57069, SD, USA
| | - Ranjit T Koodali
- Department of Chemistry, University of South Dakota, 414 E. Clark Street, Vermillion 57069, SD, USA
| | - Hongli Sun
- Department of Biomedical Engineering, University of South Dakota, BioSNTR, Sioux Falls, SD 57107, USA.
| |
Collapse
|
48
|
Lam J, Lee EJ, Clark EC, Mikos AG. Honing Cell and Tissue Culture Conditions for Bone and Cartilage Tissue Engineering. Cold Spring Harb Perspect Med 2017; 7:a025734. [PMID: 28348176 PMCID: PMC5710100 DOI: 10.1101/cshperspect.a025734] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
An avenue of tremendous interest and need in health care encompasses the regeneration of bone and cartilage. Over the years, numerous tissue engineering strategies have contributed substantial progress toward the realization of clinically relevant therapies. Cell and tissue culture protocols, however, show many variations that make experimental results among different publications challenging to compare. This collection surveys prevalent cell sources, soluble factors, culture medium formulations, environmental factors, and genetic modification approaches in the literature. The intent of consolidating this information is to provide a starting resource for scientists considering how to optimize the parameters for cell differentiation and tissue culture procedures within the context of bone and cartilage tissue engineering.
Collapse
Affiliation(s)
- Johnny Lam
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Esther J Lee
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Elisa C Clark
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Antonios G Mikos
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas 77251
| |
Collapse
|
49
|
Segredo-Morales E, García-García P, Évora C, Delgado A. BMP delivery systems for bone regeneration: Healthy vs osteoporotic population. Review. J Drug Deliv Sci Technol 2017. [DOI: 10.1016/j.jddst.2017.05.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
50
|
Kim HD, Amirthalingam S, Kim SL, Lee SS, Rangasamy J, Hwang NS. Biomimetic Materials and Fabrication Approaches for Bone Tissue Engineering. Adv Healthc Mater 2017; 6. [PMID: 29171714 DOI: 10.1002/adhm.201700612] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 10/09/2017] [Indexed: 01/14/2023]
Abstract
Various strategies have been explored to overcome critically sized bone defects via bone tissue engineering approaches that incorporate biomimetic scaffolds. Biomimetic scaffolds may provide a novel platform for phenotypically stable tissue formation and stem cell differentiation. In recent years, osteoinductive and inorganic biomimetic scaffold materials have been optimized to offer an osteo-friendly microenvironment for the osteogenic commitment of stem cells. Furthermore, scaffold structures with a microarchitecture design similar to native bone tissue are necessary for successful bone tissue regeneration. For this reason, various methods for fabricating 3D porous structures have been developed. Innovative techniques, such as 3D printing methods, are currently being utilized for optimal host stem cell infiltration, vascularization, nutrient transfer, and stem cell differentiation. In this progress report, biomimetic materials and fabrication approaches that are currently being utilized for biomimetic scaffold design are reviewed.
Collapse
Affiliation(s)
- Hwan D. Kim
- School of Chemical and Biological Engineering; The Institute of Chemical Processes; Seoul National University; Seoul 151-742 Republic of Korea
| | | | - Seunghyun L. Kim
- Interdisciplinary Program in Bioengineering; Seoul National University; Seoul 151-742 Republic of Korea
| | - Seunghun S. Lee
- Interdisciplinary Program in Bioengineering; Seoul National University; Seoul 151-742 Republic of Korea
| | - Jayakumar Rangasamy
- Centre for Nanosciences and Molecular Medicine; Amrita University; Kochi 682041 India
| | - Nathaniel S. Hwang
- School of Chemical and Biological Engineering; The Institute of Chemical Processes; Seoul National University; Seoul 151-742 Republic of Korea
- Interdisciplinary Program in Bioengineering; Seoul National University; Seoul 151-742 Republic of Korea
- The BioMax Institute of Seoul National University; Seoul 151-742 Republic of Korea
| |
Collapse
|