1
|
Lo KWH, Murray M, Laurencin CT. A Scientific Program in Regenerative Engineering (ASPIRE): A Prospective Program Aimed at Tackling Health Disparities in the USA. J Racial Ethn Health Disparities 2024:10.1007/s40615-024-02140-8. [PMID: 39392567 DOI: 10.1007/s40615-024-02140-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/11/2024] [Accepted: 08/15/2024] [Indexed: 10/12/2024]
Abstract
The continued low numbers of Blacks in STEMM (Science, Technology, Engineering, Mathematics, and Medicine) represent an American crisis that threatens growing awareness and efforts to effectively address health disparities that affect the Black population. Regenerative engineering is an emerging STEMM field that seeks to combine principles from engineering, life sciences, physics, and medicine to develop new technologies for repairing and regenerating damaged tissues and organs. We believe that regenerative engineering has the potential to address some of the root causes of health disparities by developing new approaches that are more accessible and affordable, particularly for low-income communities and people living in rural areas. We have developed a new education program targeting to K-12 groups "A Scientific Program in Regenerative Engineering (ASPIRE)" that supports the mentoring and education of Black K-12 students to enter successfully and thrive as professionals in STEMM particularly in the area of regenerative engineering. We have been collaborating with several public-school systems in Connecticut, especially among the regions with health disparities to implement the program. We believe our new educational K-12 program would serve as a vehicle to reduce health disparities in the region.
Collapse
Affiliation(s)
- Kevin W-H Lo
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT, 06268, USA
| | - Marsha Murray
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT, 06268, USA
| | - Cato T Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT, 06268, USA.
| |
Collapse
|
2
|
杨 开, 宋 成, 马 智, 汪 杰. [The effect of surface modification strategies on biological activity of titanium implant]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2024; 41:604-611. [PMID: 38932548 PMCID: PMC11208658 DOI: 10.7507/1001-5515.202308049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/24/2024] [Indexed: 06/28/2024]
Abstract
The surface morphology of titanium metal is an important factor affecting its hydrophilicity and biocompatibility, and exploring the surface treatment strategy of titanium metal is an important way to improve its biocompatibility . In this study , titanium (TA4) was firstly treated by large particle sand blasting and acid etching (SLA) technology, and then the obtained SLA-TA4 was treated by single surface treatments such as alkali-heat, ultraviolet light and plasma bombardment. According to the experimental results, alkali-heat treatment is the best treatment method to improve and maintain surface hydrophilicity of titanium. Then, the nanowire network morphology of titanium surface and its biological property, formed by further surface treatments on the basis of alkali-heat treatment, were investigated. Through the cell adhesion experiment of mouse embryonic osteoblast cells (MC3T3-E1), the ability of titanium material to support cell adhesion and cell spreading was investigated after different surface treatments. The mechanism of biological activity difference of titanium surface formed by different surface treatments was investigated according to the contact angle, pit depth and roughness of the titanium sheet surface. The results showed that the SLA-TA4 titanium sheet after a treatment of alkali heat for 10 h and ultraviolet irradiation for 1 h has the best biological activity and stability. From the perspective of improving surface bioactivity of medical devices, this study has important reference value for relevant researches on surface treatment of titanium implantable medical devices.
Collapse
Affiliation(s)
- 开通 杨
- 江苏大学 材料科学与工程学院(江苏镇江 212013)School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, P. R. China
| | - 成龙 宋
- 江苏大学 材料科学与工程学院(江苏镇江 212013)School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, P. R. China
| | - 智豪 马
- 江苏大学 材料科学与工程学院(江苏镇江 212013)School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, P. R. China
| | - 杰 汪
- 江苏大学 材料科学与工程学院(江苏镇江 212013)School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, P. R. China
| |
Collapse
|
3
|
Mitchell J, Lo KWH. The Use of Small-Molecule Compounds for Cell Adhesion and Migration in Regenerative Medicine. Biomedicines 2023; 11:2507. [PMID: 37760948 PMCID: PMC10525671 DOI: 10.3390/biomedicines11092507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/22/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Cell adhesion is essential for cell survival, communication, and regulation, and it is of fundamental importance in the development and maintenance of tissues. Cell adhesion has been widely explored due to its many important roles in the fields of tissue regenerative engineering and cell biology. This is because the mechanical interactions between a cell and its extracellular matrix (ECM) can influence and control cell behavior and function. Currently, biomaterials for regenerative medicine have been heavily investigated as substrates for promoting a cells' adhesive properties and subsequent proliferation, tissue differentiation, and maturation. Specifically, the manipulation of biomaterial surfaces using ECM coatings such as fibronectin extracted from animal-derived ECM have contributed significantly to tissue regenerative engineering as well as basic cell biology research. Additionally, synthetic and natural bioadhesive agents with pronounced abilities to enhance adhesion in numerous biological components and molecules have also been assessed in the field of tissue regeneration. Research into the use of facilitative bioadhesives has aimed to further optimize the biocompatibility, biodegradability, toxicity levels, and crosslinking duration of bioadhesive materials for improved targeted delivery and tissue repair. However, the restrictive drawbacks of some of these bioadhesive and animal-derived materials include the potential risk of disease transmission, immunogenicity, poor reproducibility, impurities, and instability. Therefore, it is necessary for alternative strategies to be sought out to improve the quality of cell adhesion to biomaterials. One promising strategy involves the use of cell-adhesive small molecules. Small molecules are relatively inexpensive, stable, and low-molecular-weight (<1000 Da) compounds with great potential to serve as efficient alternatives to conventional bioadhesives, ECM proteins, and other derived peptides. Over the past few years, a number of cell adhesive small molecules with the potential for tissue regeneration have been reported. In this review, we discuss the current progress using cell adhesive small molecules to regulate tissue regeneration.
Collapse
Affiliation(s)
- Juan Mitchell
- School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA;
| | - Kevin W.-H. Lo
- Connecticut Convergence Institute for Translation in Regenerative Engineering, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Medicine, Division of Endocrinology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Biomedical Engineering, School of Engineering, University of Connecticut, Storrs, CT 06268, USA
- Institute of Materials Science (IMS), School of Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
4
|
Awale GM, Barajaa MA, Kan HM, Seyedsalehi A, Nam GH, Hosseini FS, Ude CC, Schmidt TA, Lo KWH, Laurencin CT. Regenerative engineering of long bones using the small molecule forskolin. Proc Natl Acad Sci U S A 2023; 120:e2219756120. [PMID: 37216527 PMCID: PMC10235978 DOI: 10.1073/pnas.2219756120] [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: 11/18/2022] [Accepted: 04/10/2023] [Indexed: 05/24/2023] Open
Abstract
Bone grafting procedures have become increasingly common in the United States, with approximately 500,000 cases occurring each year at a societal cost exceeding $2.4 billion. Recombinant human bone morphogenetic proteins (rhBMPs) are therapeutic agents that have been widely used by orthopedic surgeons to stimulate bone tissue formation alone and when paired with biomaterials. However, significant limitations such as immunogenicity, high production cost, and ectopic bone growth from these therapies remain. Therefore, efforts have been made to discover and repurpose osteoinductive small-molecule therapeutics to promote bone regeneration. Previously, we have demonstrated that a single-dose treatment with the small-molecule forskolin for just 24 h induces osteogenic differentiation of rabbit bone marrow-derived stem cells in vitro, while mitigating adverse side effects attributed with prolonged small-molecule treatment schemes. In this study, we engineered a composite fibrin-PLGA [poly(lactide-co-glycolide)]-sintered microsphere scaffold for the localized, short-term delivery of the osteoinductive small molecule, forskolin. In vitro characterization studies showed that forskolin released out of the fibrin gel within the first 24 h and retained its bioactivity toward osteogenic differentiation of bone marrow-derived stem cells. The forskolin-loaded fibrin-PLGA scaffold was also able to guide bone formation in a 3-mo rabbit radial critical-sized defect model comparable to recombinant human bone morphogenetic protein-2 (rhBMP-2) treatment, as demonstrated through histological and mechanical evaluation, with minimal systemic off-target side effects. Together, these results demonstrate the successful application of an innovative small-molecule treatment approach within long bone critical-sized defects.
Collapse
Affiliation(s)
- Guleid M. Awale
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Department of Chemical Engineering, University of Connecticut, Storrs, CT06269
| | - Mohammed A. Barajaa
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT06030
- Department of Biomedical Engineering, Imam Abdulrahman Bin Faisal University,31451Dammam, Saudi Arabia
| | - Ho-Man Kan
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
| | - Amir Seyedsalehi
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT06030
| | - Ga Hie Nam
- Department of Pathology and Laboratory Medicine, UConn Health, Farmington, CT06030
| | - Fatemeh S. Hosseini
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Department of Skeletal Biology and Regeneration, UConn Health, Farmington, CT06030
| | - Chinedu C. Ude
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
| | - Tannin A. Schmidt
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT06030
| | - Kevin W.-H. Lo
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Division of Endocrinology, Department of Medicine, UConn Health, Farmington, CT06030
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT06030
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Department of Chemical Engineering, University of Connecticut, Storrs, CT06269
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT06030
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT06030
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT06269
| |
Collapse
|
5
|
Bakhshandeh B, Ranjbar N, Abbasi A, Amiri E, Abedi A, Mehrabi M, Dehghani Z, Pennisi CP. Recent progress in the manipulation of biochemical and biophysical cues for engineering functional tissues. Bioeng Transl Med 2023; 8:e10383. [PMID: 36925674 PMCID: PMC10013802 DOI: 10.1002/btm2.10383] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/28/2022] [Accepted: 07/16/2022] [Indexed: 11/11/2022] Open
Abstract
Tissue engineering (TE) is currently considered a cutting-edge discipline that offers the potential for developing treatments for health conditions that negatively affect the quality of life. This interdisciplinary field typically involves the combination of cells, scaffolds, and appropriate induction factors for the regeneration and repair of damaged tissue. Cell fate decisions, such as survival, proliferation, or differentiation, critically depend on various biochemical and biophysical factors provided by the extracellular environment during developmental, physiological, and pathological processes. Therefore, understanding the mechanisms of action of these factors is critical to accurately mimic the complex architecture of the extracellular environment of living tissues and improve the efficiency of TE approaches. In this review, we recapitulate the effects that biochemical and biophysical induction factors have on various aspects of cell fate. While the role of biochemical factors, such as growth factors, small molecules, extracellular matrix (ECM) components, and cytokines, has been extensively studied in the context of TE applications, it is only recently that we have begun to understand the effects of biophysical signals such as surface topography, mechanical, and electrical signals. These biophysical cues could provide a more robust set of stimuli to manipulate cell signaling pathways during the formation of the engineered tissue. Furthermore, the simultaneous application of different types of signals appears to elicit synergistic responses that are likely to improve functional outcomes, which could help translate results into successful clinical therapies in the future.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Nika Ranjbar
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Ardeshir Abbasi
- Department of Immunology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Elahe Amiri
- Department of Life Science Engineering, Faculty of New Sciences and TechnologyUniversity of TehranTehranIran
| | - Ali Abedi
- Department of Life Science Engineering, Faculty of New Sciences and TechnologyUniversity of TehranTehranIran
| | - Mohammad‐Reza Mehrabi
- Department of Microbial Biotechnology, School of Biology, College of ScienceUniversity of TehranTehranIran
| | - Zahra Dehghani
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and TechnologyAalborg UniversityAalborgDenmark
| |
Collapse
|
6
|
Li G, Li Y, Zhang X, Gao P, Xia X, Xiao S, Wen J, Guo T, Yang W, Li J. Strontium and simvastatin dual loaded hydroxyapatite microsphere reinforced poly(ε-caprolactone) scaffolds promote vascularized bone regeneration. J Mater Chem B 2023; 11:1115-1130. [PMID: 36636931 DOI: 10.1039/d2tb02309a] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The promotion of vascular network formation in the early stages of implantation is considered a prerequisite for successful functional bone regeneration. In this study, we successfully constructed 3D printed scaffolds with strong mechanical strength and a controllable pore structure that can sustainably release strontium (Sr) ions and simvastatin (SIM) for up to 28 days by incorporation of Sr2+ and SIM-loaded hydroxyapatite microspheres (MHA) into a poly(ε-caprolactone) (PCL) matrix. In vitro cell experiments showed that Sr-doped scaffolds were beneficial to the proliferation and osteogenic differentiation of bone mesenchymal stem cells (BMSCs), an appropriate dose of SIM was beneficial to cell proliferation and angiogenesis, and a high dose of SIM was cytotoxic. The Sr- and SIM-dual-loaded scaffolds with an appropriate dose significantly induced osteogenic differentiation of BMSCs and tube formation of human umbilical vein endothelial cells (HUVECs) in vitro and promoted vascular network and functional bone formation in vivo. Ribose nucleic acid (RNA) sequencing analysis suggested that the mechanism of promotion of vascularized bone regeneration by fabricated scaffolds is that dual-loaded Sr2+ and SIM can upregulate osteogenic and vasculogenic-related genes and downregulate osteoclast-related genes, which is beneficial for vascular and new bone regeneration. The 3D printed composite scaffolds loaded with high-stability and low-cost inorganic Sr2+ ions and SIM small-molecule drugs hold great promise in the field of promoting vascularized bone regeneration.
Collapse
Affiliation(s)
- Gen Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China.
| | - Yubao Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China.
| | - Xianhui Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Pengfei Gao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Xue Xia
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China.
| | - Shiqi Xiao
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China.
| | - Jing Wen
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China.
| | - Tao Guo
- Department of Orthopaedics, Guizhou Provincial People's hospital, Guiyang 550002, China
| | - Weihu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Jidong Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
7
|
Nun N, Joy A. Fabrication and Bioactivity of Peptide-Conjugated Biomaterial Tissue Engineering Constructs. Macromol Rapid Commun 2023; 44:e2200342. [PMID: 35822458 DOI: 10.1002/marc.202200342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/22/2022] [Indexed: 01/11/2023]
Abstract
Tissue engineering combines materials engineering, cells and biochemical factors to improve, restore or replace various types of biological tissues. A nearly limitless combination of these strategies can be combined, providing a means to augment the function of a number of biological tissues such as skin tissue, neural tissue, bones, and cartilage. Compounds such as small molecule therapeutics, proteins, and even living cells have been incorporated into tissue engineering constructs to influence biological processes at the site of implantation. Peptides have been conjugated to tissue engineering constructs to circumvent limitations associated with conjugation of proteins or incorporation of cells. This review highlights various contemporary examples in which peptide conjugation is used to overcome the disadvantages associated with the inclusion of other bioactive compounds. This review covers several peptides that are commonly used in the literature as well as those that do not appear as frequently to provide a broad scope of the utility of the peptide conjugation technique for designing constructs capable of influencing the repair and regeneration of various bodily tissues. Additionally, a brief description of the construct fabrication techniques encountered in the covered examples and their advantages in various tissue engineering applications is provided.
Collapse
Affiliation(s)
- Nicholas Nun
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, OH, 44321, USA
| | - Abraham Joy
- School of Polymer Science and Polymer Engineering, The University of Akron, Akron, OH, 44321, USA
| |
Collapse
|
8
|
Awale G, Kan HM, Laurencin CT, Lo KWH. Molecular Mechanisms Underlying the Short-Term Intervention of Forskolin-Mediated Bone Regeneration. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00285-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
9
|
Mitchell J, Lo KWH. Small molecule-mediated regenerative engineering for craniofacial and dentoalveolar bone. Front Bioeng Biotechnol 2022; 10:1003936. [PMID: 36406208 PMCID: PMC9667056 DOI: 10.3389/fbioe.2022.1003936] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/18/2022] [Indexed: 09/29/2023] Open
Abstract
The comprehensive reconstruction of extensive craniofacial and dentoalveolar defects remains a major clinical challenge to this day, especially in complex medical cases involving cancer, cranioplasty, and traumatic injury. Currently, osteogenic small molecule-based compounds have been explored extensively to repair and regenerate bone tissue because of their unique advantages. Over the past few years, a number of small molecules with the potential of craniofacial and periodontal bone tissue regeneration have been reported in literature. In this review, we discuss current progress using small molecules to regulate cranial and periodontal bone regeneration. Future directions of craniofacial bone regenerative engineering using the small molecule-based compounds will be discussed as well.
Collapse
Affiliation(s)
- Juan Mitchell
- School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Kevin W. H. Lo
- School of Medicine, Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT, United States
- Department of Medicine, Division of Endocrinology, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
- Department of Biomedical Engineering, School of Engineering, University of Connecticut, Storrs, CT, United States
- School of Engineering, Institute of Materials Science (IMS), University of Connecticut, Storrs, CT, United States
| |
Collapse
|
10
|
Noohi P, Abdekhodaie MJ, Nekoofar MH, Galler KM, Dummer PMH. Advances in Scaffolds Used for Pulp-Dentine Complex Tissue Engineering - A Narrative Review. Int Endod J 2022; 55:1277-1316. [PMID: 36039729 DOI: 10.1111/iej.13826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 07/28/2022] [Accepted: 08/10/2022] [Indexed: 11/27/2022]
Abstract
Pulp necrosis in immature teeth disrupts root development and predisposes roots to fracture as a consequence of their thin walls and open apices. Regenerative endodontics is a developing treatment modality whereby necrotic pulps are replaced with newly formed healthy tissue inside the root canal. Many clinical studies have demonstrated the potential of this strategy to stimulate root maturation and apical root-end closure. However, clinical outcomes are patient-dependent and unpredictable. The development of predictable clinical protocols is achieved through the interplay of the three classical elements of tissue engineering, namely, stem cells, signaling molecules, and scaffolds. Scaffolds provide structural support for cells to adhere and proliferate and also regulate cell differentiation and metabolism. Hence, designing and fabricating an appropriate scaffold is a crucial step in tissue engineering. In this review, four main classes of scaffolds used to engineer pulp-dentine complexes, including bioceramic-based scaffolds, synthetic polymer-based scaffolds, natural polymer-based scaffolds, and composite scaffolds, are covered. Additionally, recent advances in the design, fabrication, and application of such scaffolds are analysed along with their advantages and limitations. Finally, the importance of vascular network establishment in the success of pulp-dentine complex regeneration and strategies used to create scaffolds to address this challenge are discussed.
Collapse
Affiliation(s)
- Parisa Noohi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Mohammad J Abdekhodaie
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Mohammad H Nekoofar
- Department of Endodontics, School of Dentistry, Tehran University of Medical Sciences Tehran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Endodontic, Bahçeşehir University School of Dentistry, Istanbul, Turkey
| | - Kerstin M Galler
- Department of Conservative Dentistry and Periodontology, University Hospital Erlangen-Nürnberg, Erlangen, Germany
| | - Paul M H Dummer
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
11
|
Yang Z, Feng L, Wang M, Li Y, Bai S, Lu X, Wang H, Zhang X, Wang Y, Lin S, Tortorella MD, Li G. Sesamin Promotes Osteoporotic Fracture Healing by Activating Chondrogenesis and Angiogenesis Pathways. Nutrients 2022; 14:nu14102106. [PMID: 35631249 PMCID: PMC9147588 DOI: 10.3390/nu14102106] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/31/2022] Open
Abstract
Osteoporotic fracture has been regarded as one of the most common bone disorders in the aging society. The natural herb-derived small molecules were revealed as potential treatment approaches for osteoporotic fracture healing. Sesamin is a member of lignan family, which possesses estrogenic activity and plays a significant role in modulating bone homeostasis. Our previous study reported the promoting effect of sesamin on postmenopausal osteoporosis treatment. However, the role of sesamin in osteoporotic fracture healing has not been well studied yet. In this study, we further investigated the putative treatment effect of sesamin on osteoporotic fracture healing. Our study indicated that sesamin could activate bone morphogenetic protein 2 (BMP2) signaling pathway and further promotes in vitro chondrogenesis and angiogenesis activities. This promoting effect was abolished by the treatment of ERα inhibitor. In the osteoporotic bone fracture model, we demonstrated that sesamin markedly improves the callus formation and increases the cartilaginous area at the early-stage, as well as narrowing the fracture gap, and expands callus volume at the late-stage fracture healing site of the OVX mice femur. Furthermore, the angiogenesis at the osteoporotic fracture site was also significantly improved by sesamin treatment. In conclusion, our research illustrated the therapeutic potential and underlying regulation mechanisms of sesamin on osteoporotic fracture healing. Our studies shed light on developing herb-derived bioactive compounds as novel drugs for the treatment of osteoporotic fracture healing, especially for postmenopausal women with low estrogen level.
Collapse
Affiliation(s)
- Zhengmeng Yang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Lu Feng
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China; (L.F.); (Y.W.)
| | - Ming Wang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Yucong Li
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Shanshan Bai
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Xuan Lu
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Haixing Wang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Xiaoting Zhang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Yaofeng Wang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China; (L.F.); (Y.W.)
| | - Sien Lin
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Micky D. Tortorella
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China; (L.F.); (Y.W.)
- Correspondence: (M.D.T.); (G.L.)
| | - Gang Li
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
- Correspondence: (M.D.T.); (G.L.)
| |
Collapse
|
12
|
Rabiei M, Kalhor N, Farhadi A, Ramezanpour S, Tahamtani Y, Azarnia M. Synthetic Small Molecules to Induce Insulin Secretion and Pancreatic Beta Cell Specific Gene Expression. Cells Tissues Organs 2022:000522154. [DOI: 10.1159/000522154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/17/2022] [Indexed: 11/19/2022] Open
Abstract
Despite various efficient pharmaceuticals which are already used to manage diabetes, new drugs are needed to preserve and restore the function of pancreatic β-cells (pβCs) including cell specific gene expression and insulin production and secretion. Newly developed small molecules (SMs) with potential anti-diabetic activity need to be preliminary tested. Mice insulinoma MIN6 cells (MIN6) can be utilized as an in vitro screening model. These cells have pancreatic β-cells characteristics and can secrete insulin in response to glucose level changes. As well, β-cell-specific gene expression pattern of these cells is similar to that of mouse pancreatic islet cells. It is possible to use this cell line as a research tool to study the function of the pancreatic β-cells. To date, approximately 60 genes have been identified which, are effective in the pβCs embryonic development and insulin production and secretion during puberty, including pancreas/duodenum homeobox protein 1 (Pdx1), neuronal differentiation 1 (Neurod1), neurogenin3 (Ngn3), and insulin-1 precursor (Ins1). In this study, a family of new SMs that are structurally similar to glinides was synthesized through three different synthetic methods and categorized into three categories (C1-C3). Then, these novel SMs were characterized by testing their effects on cell viability, pβCs-specific gene expression, and insulin secretion of MIN6 in four different concentrations and three time points. According to our results, SMs of C1 (1j, 1k, and 1l) and two SMs of C3 (1f, 1I), at 200 μM concentration, were able to increase the expression levels of Pdx1, Neurod1, Ngn3, and Ins1 as well as the insulin secretion after 24 hours. However, C2 (1a, 1b, 1c and 1d) did not show significant bio-activity of MIN6 cells. These investigated molecules can provide a tool for exploring pseudo-islet functionality in MIN6 cells or provide a possible basis for future therapeutic interventions for diabetes.
Collapse
|
13
|
Sun Z, Xu X, Lv Z, Li J, Shi T, Sun H, Sun K, Tan G, Yan W, Yang YX, Wu R, Xu J, Guo H, Jiang Q, Shi D. Intraarticular injection of SHP2 inhibitor SHP099 promotes the repair of rabbit full-thickness cartilage defect. J Orthop Translat 2022; 32:112-120. [PMID: 35228993 PMCID: PMC8857578 DOI: 10.1016/j.jot.2022.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 01/06/2022] [Accepted: 01/13/2022] [Indexed: 11/19/2022] Open
Abstract
Background Cartilage repair has been a challenge in the field of orthopaedics for decades, highlighting the significance of investigating potential therapeutic drugs. In this study, we explored the effect of the SHP2 inhibitor SHP099, a small-molecule drug, on cartilage repair. Methods Human synovial mesenchymal stem cells (SMSCs) were isolated, and their three-way differentiation potential was examined. After treatment with chondrogenic medium, the chondrogenic effect of SHP099 on SMSCs was examined by western blot, qPCR, and immunofluorescence (IF). Micro-mass culture was also used to detect the effect of SHP099. To explore the chondrogenic effects of SHP099 in vivo, full-thickness cartilage defects with microfractures were constructed in the right femoral trochlea of New Zealand White rabbits. Intraarticular injection of SHP099 or normal saline was performed twice a week for 6 weeks. Cartilage repair was evaluated by haematoxylin and eosin (HE) staining and safranin O/fast green staining. Immunohistochemistry (IHC) for collagen II (COL2) was also conducted to verify the abundance of cartilage extracellular matrix after SHP099 treatment. The mechanism involving yes-associated protein (YAP) and WNT signalling was investigated in vitro. Results SMSCs isolated from human synovium have optimal multi-differentiation potential. SHP099 increased chondrogenic marker (SOX9, COL2) expression and decreased hypertrophic marker (COL10, RUNX2) expression in SMSCs. In micro-mass culture, the SHP099-induced cartilage tissues had a better result of Safranin O and Toluidine blue staining and are enriched in cartilage-specific collagen II. Inhibition of YAP and WNT signalling was also observed. Moreover, compared to the normal saline group at 6 weeks, intraarticular injection of SHP099 resulted in better defect filling, forming increased hyaline cartilage-like tissue with higher levels of glycosaminoglycan (GAG) and COL2. Conclusion SHP099 promotes the repair of rabbit full-thickness cartilage defects, representing a potential therapeutic drug for cartilage repair. The Translational potential of this article This study provides evidence that SHP2 inhibition promotes chondrogenesis and the repair of cartilage in defect area, which could be a novel therapeutic approach for cartilage repair.
Collapse
Affiliation(s)
- Ziying Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Xingquan Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Zhongyang Lv
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Jiawei Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Tianshu Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Heng Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Kuoyang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Guihua Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Wenqiang Yan
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Yannick Xiaofan Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Drum Tower of Clinical Medicine, Nanjing Medical University, Nanjing, 210008, Jiangsu, PR China
| | - Rui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Jia Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Drum Tower of Clinical Medicine, Nanjing Medical University, Nanjing, 210008, Jiangsu, PR China
| | - Hu Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
| | - Dongquan Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, Jiangsu, PR China
- Corresponding author. Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| |
Collapse
|
14
|
Hayaei Tehrani RS, Sayahpour FA, Esfandiari F. A comparison between BMP4 and SB4 in inducing germ line gene expression pattern during embryonic stem cells differentiation. Differentiation 2021; 123:9-17. [PMID: 34864442 DOI: 10.1016/j.diff.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 11/15/2021] [Accepted: 11/21/2021] [Indexed: 11/03/2022]
Abstract
Germ cell production from stem cells allows for studying the mechanisms involved in gamete development with the aim of helping infertile couples with the generation of healthy gametes. In this context, improving the protocols for in-vitro germ cell induction from stem cells is very important. Recently, SB4 small molecule has been introduced as a potent agonist for bone morphogenic protein 4 (BMP4). Herein, we investigated whether BMP4, is replaceable by SB4 for having affordable protocol for in vitro germ cell differentiation. We demonstrated that SB4 can induce Blimp1 (as the first gene induced germ line differentiation) expression significantly but at a lower level compared to BMP4. However, Tfap2c (a putative downstream target of Blimp1 during germ cell differentiation) expression level in SB4-induced aggregates was significantly higher than in BMP4-induced aggregates. Moreover, co-presence of both BMP4 and SB4 could increase the expression level of Prdm14, Nnose3 and Stella (Dppa3), and thereby improve establishment of the germ cell fate during in-vitro differentiation of embryonic stem cells. In summary, our data suggest that SB4 could improve germ line gene expression pattern induced by BMP4 during embryonic stem cells in-vitro differentiation.
Collapse
Affiliation(s)
- Reyhaneh Sadat Hayaei Tehrani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fereshteh Esfandiari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
15
|
Dong H, Zhu T, Zhang M, Wang D, Wang X, Huang G, Wang S, Zhang M. Polymer Scaffolds-Enhanced Bone Regeneration in Osteonecrosis Therapy. Front Bioeng Biotechnol 2021; 9:761302. [PMID: 34631688 PMCID: PMC8498195 DOI: 10.3389/fbioe.2021.761302] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022] Open
Abstract
Osteonecrosis without effective early treatment eventually leads to the collapse of the articular surface and causes arthritis. For the early stages of osteonecrosis, core decompression combined with bone grafting, is a procedure worthy of attention and clinical trial. And the study of bone graft substitutes has become a hot topic in the area of osteonecrosis research. In recent years, polymers have received more attention than other materials due to their excellent performance. However, because of the harsh microenvironment in osteonecrosis, pure polymers may not meet the stringent requirements of osteonecrosis research. The combined application of polymers and various other substances makes up for the shortcomings of polymers, and to meet a broad range of requirements for application in osteonecrosis therapy. This review focuses on various applying polymers in osteonecrosis therapy, then discusses the development of biofunctionalized composite polymers based on the polymers combined with different bioactive substances. At the end, we discuss their prospects for translation to clinical practice.
Collapse
Affiliation(s)
- Hengliang Dong
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Tongtong Zhu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Mingran Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Dapeng Wang
- Department of Orthopedics, Siping Central Hospital, Siping, China
| | - Xukai Wang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Guanning Huang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shuaishuai Wang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Minglei Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Zhang N, Wang W, Zhang X, Nune KC, Zhao Y, Liu N, Misra R, Yang K, Tan L, Yan J. The effect of different coatings on bone response and degradation behavior of porous magnesium-strontium devices in segmental defect regeneration. Bioact Mater 2021; 6:1765-1776. [PMID: 33313453 PMCID: PMC7718143 DOI: 10.1016/j.bioactmat.2020.11.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
Regeneration of long-bone segmental defects remains a challenge for orthopedic surgery. Current treatment options often require several revision procedures to maintain acceptable alignment and achieve osseous healing. A novel hollow tubular system utilizing magnesium-strontium (Mg-Sr) alloy with autogenous morselized bone filled inside to repair segmental defects was developed. To improve the corrosion and biocompatible properties, two coatings, Ca-P and Sr-P coatings, were prepared on surface of the implants. Feasibility of applying these coated implants was systematically evaluated in vitro and in vivo, and simultaneously to have a better understanding on the relationship of degradation and bone regeneration on the healing process. According to the in vitro corrosion study by electrochemical measurements, greater corrosion resistance was obtained for Ca-P coated sample, and attributed to the double-layer protective structure. The cytotoxicity and alkaline phosphatase (ALP) assays demonstrated enhanced bioactivity for Sr-P coated group because of the long-lasting release of beneficial Sr2+. At 12 weeks post-implantation with Mg-Sr alloy porous device, the segmental defects were effectively repaired with respect to both integrity and continuity. In addition, compared with the Ca-P coated implant, the Sr-P coated implant was more proficient at promoting bone formation and mineralization. In summary, the Sr-P coated implants have bioactive properties and exceptional durability, and promote bone healing that is close to the natural rate, implying their potential application for the regeneration of segmental defects.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Orthopedic Surgery, The 2nd Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, China
- Department of Orthopedic Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Weidan Wang
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Xiuzhi Zhang
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, China
| | - Krishna. C. Nune
- Department of Metallurgical, Material and Biomedical Engineering, The University of Texas at EI Paso, TX, 79968, USA
| | - Ying Zhao
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Na Liu
- Department of Orthopedic Surgery, The 2nd Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, China
| | - R.D.K. Misra
- Department of Metallurgical, Material and Biomedical Engineering, The University of Texas at EI Paso, TX, 79968, USA
| | - Ke Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Lili Tan
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Jinglong Yan
- Department of Orthopedic Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
17
|
Gurel Pekozer G, Abay Akar N, Cumbul A, Beyzadeoglu T, Torun Kose G. Investigation of Vasculogenesis Inducing Biphasic Scaffolds for Bone Tissue Engineering. ACS Biomater Sci Eng 2021; 7:1526-1538. [PMID: 33740374 DOI: 10.1021/acsbiomaterials.0c01071] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vascularization is the main obstacle for the bone tissue engineering strategies since the defect size is generally large. Incorporation of angiogenic factors is one of the strategies employed in order to accelerate vascularization and improve bone healing. In this study, a biphasic scaffold consisting of fibrous poly(lactide-co-glycolide) (PLGA) and poly(lactide-co-glycolide)-block-poly(ethylene glycol)-block-poly(lactide-co-glycolide) (PLGA-PEG-PLGA) hydrogel loaded with vascular endothelial growth factor-A (VEGF) inducer, GS4012, was constructed. Mesenchymal stem cells isolated from rat bone marrow (rBMSCs) were used for differentiation into osteogenic cells, and endothelial cells isolated from rat peripheral blood (rPBECs) were used to test the in vitro endothelial cell recruitment. The biphasic scaffold was tested for cell proliferation, ALP expression, VEGF induction, expression of osteogenic genes by rBMSCs, and recruitment of rPBECs in vitro and for improved bone healing and vascularization in vivo on critical size rat cranial defects. Endothelial migration through porous insert and VEGF induction were obtained in vitro in response to GS4012 as well as the upregulation of ALP, Runx2, Col I, and OC gene expressions. The biphasic scaffold was also shown to be effective in improving endothelial cell recruitment, vascularization, and bone healing in vivo. Thus, the proposed design has a great potential for the healing of critical size bone defect in tissue engineering studies according to both in vitro and in vivo investigations.
Collapse
Affiliation(s)
- Gorke Gurel Pekozer
- Biomedical Engineering Department, Faculty of Electrical and Electronics Engineering, Yildiz Technical University, Istanbul 34220, Turkey
| | - Nergis Abay Akar
- Genetics and Bioengineering Department, Faculty of Engineering, Yeditepe University, Istanbul 34755, Turkey
| | - Alev Cumbul
- Histology and Embryology Department, Faculty of Medicine, Yeditepe University, Istanbul 34755, Turkey
| | - Tahsin Beyzadeoglu
- Orthopaedics and Traumatology, Facuty of Health Sciences, Halic University Beyzadeoglu Clinic, Istanbul 34738, Turkey
| | - Gamze Torun Kose
- Genetics and Bioengineering Department, Faculty of Engineering, Yeditepe University, Istanbul 34755, Turkey
| |
Collapse
|
18
|
Moreno-Manzano V, Zaytseva-Zotova D, López-Mocholí E, Briz-Redón Á, Løkensgard Strand B, Serrano-Aroca Á. Injectable Gel Form of a Decellularized Bladder Induces Adipose-Derived Stem Cell Differentiation into Smooth Muscle Cells In Vitro. Int J Mol Sci 2020; 21:E8608. [PMID: 33203120 PMCID: PMC7696281 DOI: 10.3390/ijms21228608] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 01/03/2023] Open
Abstract
Biologic scaffolds composed of extracellular matrix components have been proposed to repair and reconstruct a variety of tissues in clinical and pre-clinical studies. Injectable gels can fill and conform any three-dimensional shape and can be delivered to sites of interest by minimally invasive techniques. In this study, a biological gel was produced from a decellularized porcine urinary bladder by enzymatic digestion with pepsin. The enzymatic digestion was confirmed by visual inspection after dissolution in phosphate-buffered saline solution and Fourier-transform infrared spectroscopy. The rheological and biological properties of the gel were characterized and compared to those of the MatrigelTM chosen as a reference material. The storage modulus G' reached 19.4 ± 3.7 Pa for the 30 mg/mL digested decellularized bladder gels after ca. 3 h at 37 °C. The results show that the gel formed of the porcine urinary bladder favored the spontaneous differentiation of human and rabbit adipose-derived stem cells in vitro into smooth muscle cells to the detriment of cell proliferation. The results support the potential of the developed injectable gel for tissue engineering applications to reconstruct for instance the detrusor muscle part of the human urinary bladder.
Collapse
Affiliation(s)
- Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Lab, Centro de Investigación Príncipe Felipe, c/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain;
| | - Daria Zaytseva-Zotova
- NOBIPOL, Department of Biotechnology and Food Science, NTNU Norwegian University of Science and Technology, Sem Sælands vei 6-8, N-7491 Trondheim, Norway; (D.Z.-Z.); (B.L.S.)
| | - Eric López-Mocholí
- Neuronal and Tissue Regeneration Lab, Centro de Investigación Príncipe Felipe, c/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain;
| | - Álvaro Briz-Redón
- Statistics Office, City Council of Valencia, Plaza Ayuntamiento 1, 46002 Valencia, Spain;
| | - Berit Løkensgard Strand
- NOBIPOL, Department of Biotechnology and Food Science, NTNU Norwegian University of Science and Technology, Sem Sælands vei 6-8, N-7491 Trondheim, Norway; (D.Z.-Z.); (B.L.S.)
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain
| |
Collapse
|
19
|
Frassica MT, Jones SK, Suriboot J, Arabiyat AS, Ramirez EM, Culibrk RA, Hahn MS, Grunlan MA. Enhanced Osteogenic Potential of Phosphonated-Siloxane Hydrogel Scaffolds. Biomacromolecules 2020; 21:5189-5199. [DOI: 10.1021/acs.biomac.0c01293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Michael T. Frassica
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-3120, United States
| | - Sarah K. Jones
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-3120, United States
| | - Jakkrit Suriboot
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-3120, United States
| | - Ahmad S. Arabiyat
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180-31590, United States
| | - Esteban M. Ramirez
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-3120, United States
| | - Robert A. Culibrk
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180-31590, United States
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180-31590, United States
| | - Melissa A. Grunlan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-3120, United States
- Department of Materials Science & Engineering, Texas A&M University, College Station, Texas 77843-3003, United States
- Department of Chemistry, Texas A&M University, College Station, Texas 77843-3255, United States
| |
Collapse
|
20
|
Reid G, Magarotto F, Marsano A, Pozzobon M. Next Stage Approach to Tissue Engineering Skeletal Muscle. Bioengineering (Basel) 2020; 7:E118. [PMID: 33007935 PMCID: PMC7711907 DOI: 10.3390/bioengineering7040118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/18/2020] [Accepted: 09/26/2020] [Indexed: 02/08/2023] Open
Abstract
Large-scale muscle injury in humans initiates a complex regeneration process, as not only the muscular, but also the vascular and neuro-muscular compartments have to be repaired. Conventional therapeutic strategies often fall short of reaching the desired functional outcome, due to the inherent complexity of natural skeletal muscle. Tissue engineering offers a promising alternative treatment strategy, aiming to achieve an engineered tissue close to natural tissue composition and function, able to induce long-term, functional regeneration after in vivo implantation. This review aims to summarize the latest approaches of tissue engineering skeletal muscle, with specific attention toward fabrication, neuro-angiogenesis, multicellularity and the biochemical cues that adjuvate the regeneration process.
Collapse
Affiliation(s)
- Gregory Reid
- Department of Cardiac Surgery, University Hospital Basel, 4031 Basel, Switzerland; (G.R.); (A.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Fabio Magarotto
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
- Institute of Pediatric Research, Città della Speranza, 35127 Padova, Italy
| | - Anna Marsano
- Department of Cardiac Surgery, University Hospital Basel, 4031 Basel, Switzerland; (G.R.); (A.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Michela Pozzobon
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
- Institute of Pediatric Research, Città della Speranza, 35127 Padova, Italy
| |
Collapse
|
21
|
Rahmati M, Frank MJ, Walter SM, Monjo MC, Satué M, Reseland JE, Lyngstadaas SP, Haugen HJ. Osteoimmunomodulatory Effects of Enamel Matrix Derivate and Strontium Coating Layers: A Short- and Long-Term In Vivo Study. ACS APPLIED BIO MATERIALS 2020; 3:5169-5181. [PMID: 32954227 PMCID: PMC7493216 DOI: 10.1021/acsabm.0c00608] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/23/2020] [Indexed: 12/22/2022]
Abstract
Over the past few years, surface modification of implant surfaces has gained substantial attention as a promising solution to avoid the failure of biomaterials after implantation. Although researchers suggest several strategies for surface functionalization of titanium-based implants, only a few studies have compared the osteoimmunomodulatory effects of ionic nanostructures and biofunctionalization in the same biological model. Enamel matrix derivate (EMD) and strontium are both known for their positive influences on bone cell responses. In this study, we functionalized the titanium-zirconium implant surface with EMD and strontium using an electrochemical cathodic polarization method. Afterward, we evaluated the osteoimmunomodulatory effects of EMD or strontium coated titanium-zirconium implants in the tibia of eight Gray Bastard Chinchilla rabbits. We performed 2 and 3D micro-CT, wound fluid, histologic, and histomorphometric analyses on bone tissues after 4- and 8-weeks of implantation. Although the results could indicate some differences between groups regarding the bone quality, there was no difference in bone amount or volume. EMD stimulated higher ALP activity and lower cytotoxicity in wound fluid, as well as a lower expression of inflammatory markers after 8 weeks indicating its osteoimmunomodulatory effects after implantation. Overall, the results suggested that ionic nanostructure modification and biofunctionalization might be useful in regulating the immune responses to implants.
Collapse
Affiliation(s)
- Maryam Rahmati
- Department
of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109
Blindern, NO-0317 Oslo, Norway
| | - Matthias Johannes Frank
- Department
of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109
Blindern, NO-0317 Oslo, Norway
- Institute
of Medical and Polymer Engineering, Chair of Medical Engineering, Technische Universität München, Boltzmannstrasse 15, 85748 Garching, Germany
| | - Sebastian Martin Walter
- Department
of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109
Blindern, NO-0317 Oslo, Norway
- Institute
of Medical and Polymer Engineering, Chair of Medical Engineering, Technische Universität München, Boltzmannstrasse 15, 85748 Garching, Germany
| | - Marta Cabrer Monjo
- Department
of Fundamental Biology and Health Sciences, Research Institute on Health Sciences (IUNICS), University of Balearic
Islands, ES-07122 Palma, Spain
- Balearic
Islands Health Institute (IdISBa), ES-07010 Palma, Spain
| | - Maria Satué
- Department
of Fundamental Biology and Health Sciences, Research Institute on Health Sciences (IUNICS), University of Balearic
Islands, ES-07122 Palma, Spain
- Balearic
Islands Health Institute (IdISBa), ES-07010 Palma, Spain
- Department
of Biomedical Sciences, University of Veterinary
Medicine, 1210 Vienna, Austria
| | - Janne Elin Reseland
- Department
of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109
Blindern, NO-0317 Oslo, Norway
| | - Ståle Petter Lyngstadaas
- Department
of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109
Blindern, NO-0317 Oslo, Norway
| | - Håvard Jostein Haugen
- Department
of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109
Blindern, NO-0317 Oslo, Norway
| |
Collapse
|
22
|
Cottrill E, Lazzari J, Pennington Z, Ehresman J, Schilling A, Dirckx N, Theodore N, Sciubba D, Witham T. Oxysterols as promising small molecules for bone tissue engineering: Systematic review. World J Orthop 2020; 11:328-344. [PMID: 32908817 PMCID: PMC7453739 DOI: 10.5312/wjo.v11.i7.328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/08/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bone tissue engineering is an area of continued interest within orthopaedic surgery, as it promises to create implantable bone substitute materials that obviate the need for autologous bone graft. Recently, oxysterols – oxygenated derivatives of cholesterol – have been proposed as a novel class of osteoinductive small molecules for bone tissue engineering. Here, we present the first systematic review of the in vivo evidence describing the potential therapeutic utility of oxysterols for bone tissue engineering.
AIM To systematically review the available literature examining the effect of oxysterols on in vivo bone formation.
METHODS We conducted a systematic review of the literature following PRISMA guidelines. Using the PubMed/MEDLINE, Embase, and Web of Science databases, we queried all publications in the English-language literature investigating the effect of oxysterols on in vivo bone formation. Articles were screened for eligibility using PICOS criteria and assessed for potential bias using an expanded version of the SYRCLE Risk of Bias assessment tool. All full-text articles examining the effect of oxysterols on in vivo bone formation were included. Extracted data included: Animal species, surgical/defect model, description of therapeutic and control treatments, and method for assessing bone growth. Primary outcome was fusion rate for spinal fusion models and percent bone regeneration for critical-sized defect models. Data were tabulated and described by both surgical/defect model and oxysterol employed. Additionally, data from all included studies were aggregated to posit the mechanism by which oxysterols may mediate in vivo bone formation.
RESULTS Our search identified 267 unique articles, of which 27 underwent full-text review. Thirteen studies (all preclinical) met our inclusion/exclusion criteria. Of the 13 included studies, 5 employed spinal fusion models, 2 employed critical-sized alveolar defect models, and 6 employed critical-sized calvarial defect models. Based upon SYRCLE criteria, the included studies were found to possess an overall “unclear risk of bias”; 54% of studies reported treatment randomization and 38% reported blinding at any level. Overall, seven unique oxysterols were evaluated: 20(S)-hydroxycholesterol, 22(R)-hydroxycholesterol, 22(S)-hydroxycholesterol, Oxy4/Oxy34, Oxy18, Oxy21/Oxy133, and Oxy49. All had statistically significant in vivo osteoinductive properties, with Oxy4/Oxy34, Oxy21/Oxy133, and Oxy49 showing a dose-dependent effect in some cases. In the eight studies that directly compared oxysterols to rhBMP-2-treated animals, similar rates of bone growth occurred in the two groups. Biochemical investigation of these effects suggests that they may be primarily mediated by direct activation of Smoothened in the Hedgehog signaling pathway.
CONCLUSION Present preclinical evidence suggests oxysterols significantly augment in vivo bone formation. However, clinical trials are necessary to determine which have the greatest therapeutic potential for orthopaedic surgery patients.
Collapse
Affiliation(s)
- Ethan Cottrill
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Julianna Lazzari
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Zach Pennington
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Jeff Ehresman
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Andrew Schilling
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Naomi Dirckx
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Nicholas Theodore
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Daniel Sciubba
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Timothy Witham
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| |
Collapse
|
23
|
Chen J, Ashames A, Buabeid MA, Fahelelbom KM, Ijaz M, Murtaza G. Nanocomposites drug delivery systems for the healing of bone fractures. Int J Pharm 2020; 585:119477. [PMID: 32473968 DOI: 10.1016/j.ijpharm.2020.119477] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/20/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022]
Abstract
The skeletal system is fundamental for the structure and support of the body consisting of bones, cartilage, and connective tissues. Poor fracture healing is a chief clinical problem leading to disability, extended hospital stays and huge financial liability. Even though most fractures are cured using standard clinical methods, about 10% of fractures are delayed or non-union. Despite decades of progress, the bone-targeted delivery system is still restricted due to the distinctive anatomical bone features. Recently, various novel nanocomposite systems have been designed for the cell-specific targeting of bone, enhancing drug solubility, improving drug stability and inhibiting drug degradation so that it can reach its target site without being removed in the systemic circulation. Such targeting systems could consist of biological compounds i.e. bone marrow stem cells (BMSc), growth factors, RNAi, parathyroid hormone or synthetic compounds, i.e. bisphosphonates (BPs) and calcium phosphate cement. Hydrogels and nanoparticles are also being employed for fracture healing. In this review, we discussed the normal mechanism of bone healing and all the possible drug delivery systems being employed for the healing of the bone fracture.
Collapse
Affiliation(s)
- Jianxian Chen
- School of Economics, Capital University of Economics and Business, Beijing, China
| | - Akram Ashames
- College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates.
| | - Manal Ali Buabeid
- College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Khairi Mustafa Fahelelbom
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain, United Arab Emirates
| | - Muhammad Ijaz
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, 54000, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, 54000, Pakistan.
| |
Collapse
|
24
|
Kuttappan S, Jo JI, Sabu CK, Menon D, Tabata Y, Nair MB. Bioinspired nanocomposite fibrous scaffold mediated delivery of ONO-1301 and BMP2 enhance bone regeneration in critical sized defect. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110591. [DOI: 10.1016/j.msec.2019.110591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 01/08/2023]
|
25
|
Zhu T, Cui Y, Zhang M, Zhao D, Liu G, Ding J. Engineered three-dimensional scaffolds for enhanced bone regeneration in osteonecrosis. Bioact Mater 2020; 5:584-601. [PMID: 32405574 PMCID: PMC7210379 DOI: 10.1016/j.bioactmat.2020.04.008] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/11/2020] [Accepted: 04/11/2020] [Indexed: 12/15/2022] Open
Abstract
Osteonecrosis, which is typically induced by trauma, glucocorticoid abuse, or alcoholism, is one of the most severe diseases in clinical orthopedics. Osteonecrosis often leads to joint destruction, and arthroplasty is eventually required. Enhancement of bone regeneration is a critical management strategy employed in osteonecrosis therapy. Bone tissue engineering based on engineered three-dimensional (3D) scaffolds with appropriate architecture and osteoconductive activity, alone or functionalized with bioactive factors, have been developed to enhance bone regeneration in osteonecrosis. In this review, we elaborate on the ideal properties of 3D scaffolds for enhanced bone regeneration in osteonecrosis, including biocompatibility, degradability, porosity, and mechanical performance. In addition, we summarize the development of 3D scaffolds alone or functionalized with bioactive factors for accelerating bone regeneration in osteonecrosis and discuss their prospects for translation to clinical practice. Engineered three-dimensional scaffolds boost bone regeneration in osteonecrosis. The ideal properties of three-dimensional scaffolds for osteonecrosis treatment are discussed. Bioactive factors-functionalized three-dimensional scaffolds are promising bone regeneration devices for osteonecrosis management. The challenges and opportunities of engineered three-dimensional scaffolds for osteonecrosis therapy are predicted.
Collapse
Affiliation(s)
- Tongtong Zhu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, PR China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Yutao Cui
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, 130041, PR China
| | - Mingran Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, PR China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Duoyi Zhao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Guangyao Liu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, PR China
- Corresponding author.
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| |
Collapse
|
26
|
Basu S, Pacelli S, Paul A. Self-healing DNA-based injectable hydrogels with reversible covalent linkages for controlled drug delivery. Acta Biomater 2020; 105:159-169. [PMID: 31972367 DOI: 10.1016/j.actbio.2020.01.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/11/2020] [Accepted: 01/14/2020] [Indexed: 01/08/2023]
Abstract
Injectable hydrogels represent a valuable tool for the delivery of therapeutic molecules aimed to restore the functionality of damaged tissues. In this study, we report the design of a nanocomposite DNA-based hydrogel crosslinked with oxidized alginate (OA) via the formation of reversible imine linkages. The formulated hydrogel functioned as an injectable carrier for the sustained delivery of a small molecule drug, simvastatin. The degree of oxidation of alginate and the concentration of silicate-based nanoparticles (nSi) were varied to modulate the rheological properties of the hydrogels. Specifically, the formulations consisting of OA with higher degree of oxidation displayed the highest value of storage moduli, yield stress, yield strain, and rapid recovery after removal of cyclic stress. The hydrogel formulations exhibited self-healing and shear-thinning properties due to the reversible nature of the covalent imine bonds formed between the aldehyde groups of OA and the amine groups present in the DNA nucleotides. Moreover, the incorporation of charged nSi further enhanced the shear strength of the formulated hydrogels by establishing electrostatic interactions with the phosphate groups of the DNA network. The optimized hydrogel was able to promote the sustained release of simvastatin for more than a week. The bioactivity of the released drug was confirmed by testing its ability to induce osteogenic differentiation and migration of human adipose-derived stem cells in vitro. Overall, the results obtained from this study demonstrate that DNA could be used as a natural biopolymer to fabricate self-healing injectable hydrogels with sustained release properties for minimally invasive therapeutic approaches. STATEMENT OF SIGNIFICANCE: Dynamic covalent chemistry, especially Schiff base reactions have emerged as a promising route for the formation of injectable hydrogels. Our study demonstrated the development of a DNA-based self-healing hydrogel formed via Schiff base reaction occurring at physiological conditions. The hydrogels functioned as sustained delivery vehicles for the hydrophobic drug simvastatin, which requires a polymeric carrier for controlled delivery of therapeutic concentrations of the drug without exhibiting cytotoxic effects. Presently available hydrogel-based drug delivery systems encounter major challenges for the delivery of hydrophobic drugs due to the hydrophilic nature of the base matrix. Our strategy presents a platform technology for the design of minimally invasive approaches for the sustained delivery of hydrophobic drugs similar to simvastatin.
Collapse
Affiliation(s)
- Sayantani Basu
- Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, KS, USA
| | - Settimio Pacelli
- Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Arghya Paul
- Department of Chemical and Biochemical Engineering, Department of Chemistry, The University of Western Ontario, London, ON N6A 5B9, Canada.
| |
Collapse
|
27
|
Liu Y, Hu J, Sun H. Mineralized nanofibrous scaffold promotes phenamil-induced osteoblastic differentiation while mitigating adipogenic differentiation. J Tissue Eng Regen Med 2019; 14:464-474. [PMID: 31840422 DOI: 10.1002/term.3007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 10/23/2019] [Accepted: 10/27/2019] [Indexed: 12/20/2022]
Abstract
Large bone defects represent a significant unmet medical challenge. Cost effectiveness and better stability make small molecule organic compounds a more promising alternative compared with biomacromolecules, for example, growth factors/hormones, in regenerative medicine. However, one common challenge for the application of these small compounds is their side-effect issue. Phenamil is emerging as an intriguing small molecule to promote bone repair by strongly activating bone morphogenetic protein signaling pathway. In addition to osteogenesis, phenamil also induces significant adipogenesis based on some in vitro studies, which is a concern that impedes it from potential clinical applications. Besides the soluble chemical signals, cellular differentiation is heavily dependent on the microenvironments provided by the 3D scaffolds. Therefore, we developed a 3D nanofibrous biomimetic scaffold-based strategy to harness the phenamil-induced stem cell lineage differentiation. Based on the gene expression, alkaline phosphatase activity, and mineralization data, we indicated that bone-matrix mimicking mineralized-gelatin nanofibrous scaffold effectively improved phenamil-induced osteoblastic differentiation, while mitigating the adipogenic differentiation in vitro. In addition to normal culture conditions, we also indicated that mineralized matrix can significantly improve phenamil-induced osteoblastic differentiation in simulated inflammatory condition. In viewing of the crucial role of mineralized matrix, we developed an innovative and facile mineral deposition-based strategy to sustain release of phenamil from 3D scaffolds for efficient local bone regeneration. Overall, our study demonstrated that biomaterials played a crucial role in modulating small molecule drug phenamil-induced osteoblastic differentiation by providing a bone-matrix mimicking mineralized gelatin nanofibrous scaffolds.
Collapse
Affiliation(s)
- Yangxi Liu
- Department of Biomedical Engineering, University of South Dakota, BioSNTR, Sioux Falls, South Dakota
| | - Jue Hu
- Department of Oral and Maxillofacial Surgery, College of Dentistry, University of Iowa, Iowa City, Iowa
| | - Hongli Sun
- Department of Oral and Maxillofacial Surgery, College of Dentistry, University of Iowa, Iowa City, Iowa
| |
Collapse
|
28
|
Kuttappan S, Jo JI, Menon D, Ishimoto T, Nakano T, Nair SV, Tabata Y, Nair MB. ONO-1301 loaded nanocomposite scaffolds modulate cAMP mediated signaling and induce new bone formation in critical sized bone defect. Biomater Sci 2019; 8:884-896. [PMID: 31822874 DOI: 10.1039/c9bm01352k] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent studies on bone regeneration demonstrate the use of low cost and stable small molecules, which avoid the adverse effect and high cost of growth factors. Herein, we investigate the chemotactic, angiogenic and osteoinductive potential of a prostacyclin analogue, ONO-1301, when delivered through a biomimetic nanocomposite scaffold (nanohydroxyapatite-gelatin matrix reinforced with fibers) for bone tissue regeneration. The small molecule was loaded onto the scaffold in three different concentrations. There was burst release from all the groups of scaffolds within 24 h followed by a sustained release up to 14 days, but the concentration was dependent on loading percentage. ONO-1301 loaded scaffolds augmented the migration, proliferation and osteogenic differentiation of mesenchymal stem cells (MSCs), but increasing the concentration beyond a certain dose did not show any effect. The osteoinduction was mediated through the prostaglandin I2 receptor and cyclic AMP (cAMP) signaling pathway. They also promoted new bone formation in large sized calvarial defects in rats compared to the scaffold alone, but did not show any impact on angiogenesis. Hence, this study suggests the chemotactic and osteoinductive capability of ONO-1301 for the repair and regeneration of critical sized bone defects.
Collapse
Affiliation(s)
- Shruthy Kuttappan
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, India.
| | - Jun-Ichiro Jo
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Japan.
| | - Deepthy Menon
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, India.
| | - Takuya Ishimoto
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Takayoshi Nakano
- Division of Materials and Manufacturing Science, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Shantikumar V Nair
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, India.
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Japan.
| | - Manitha B Nair
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, India.
| |
Collapse
|
29
|
Amano H, Iwaki F, Oki M, Aoki K, Ohba S. An osteogenic helioxanthin derivative suppresses the formation of bone-resorbing osteoclasts. Regen Ther 2019; 11:290-296. [PMID: 31667208 PMCID: PMC6813560 DOI: 10.1016/j.reth.2019.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/08/2019] [Accepted: 08/30/2019] [Indexed: 02/08/2023] Open
Abstract
Objective The helioxanthin derivative 4-(4-methoxyphenyl)thieno[2,3-b:5,4-c′]dipyridine-2-carboxamide (TH) is a low-molecular-weight compound that was identified through screening for osteogenic compounds that enhance the activity of mouse preosteoblastic MC3T3-E1 cells. In the present study, we found that TH suppressed osteoclast differentiation. Methods Using the hematopoietic stem cells of ddY mice, TH was added to the culture in the experimental group, and the number of osteoclasts was measured with rhodamine phalloidin staining and TRAP staining. In osteo assay, bone resorption area was compared by the von Kossa staining. Results Specifically, TH inhibited the cyclic guanosine monophosphate (cGMP)-degrading activity of phosphodiesterase (PDE), promoted nitric oxide (NO) production, and dose-dependently suppressed osteoclast differentiation in an osteoclast formation culture of mouse bone marrow cells. The NO-competitive guanylyl cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) attenuated the suppressive activity of TH on osteoclast differentiation. Conclusion: Given the previously reported suppressive action of cGMP on osteoclastogenesis, our data suggest that TH negatively impacts osteoclast differentiation at least to some extent by stimulating NO production and inhibiting PDE activity, both of which lead to the upregulation of intracellular cGMP. This study supports the potential use of TH as a novel antiosteoporotic reagent that not only stimulates bone formation but also inhibits bone resorption.
Collapse
Affiliation(s)
- Hitoshi Amano
- Department of Basic Oral Health Engineering, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549 Japan
| | - Futoshi Iwaki
- Department of Oral and Maxillofacial Surgery, Kobe City Nishi-Kobe Medical Center, 5-7-1, Koujidai Nishi-ku, Kobe, 651-2273, Japan
| | - Meiko Oki
- Department of Basic Oral Health Engineering, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549 Japan
| | - Kazuhiro Aoki
- Department of Basic Oral Health Engineering, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549 Japan
| | - Shinsuke Ohba
- Department of Cell Biology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
30
|
Zhang S, Xing M, Li B. Recent advances in musculoskeletal local drug delivery. Acta Biomater 2019; 93:135-151. [PMID: 30685475 PMCID: PMC6615977 DOI: 10.1016/j.actbio.2019.01.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/18/2018] [Accepted: 01/22/2019] [Indexed: 12/17/2022]
Abstract
Musculoskeletal disorders are a significant burden on the global economy and public health. Advanced drug delivery plays a key role in the musculoskeletal field and holds the promise of enhancing the repair of degenerated and injured musculoskeletal tissues. Ideally, drug delivery should have the ability to directly deliver therapeutic agents to the diseased/injured sites with a desirable drug level over a period of time. Here, we present a mini-review of the current state-of-the-art research associated with local drug delivery and its use for the treatment of musculoskeletal disorders. First, an overview of drug delivery strategies, with a focus on issues related to musculoskeletal pathology, potential therapeutic strategies, conventional and non-conventional drugs, and various delivery systems, is introduced. Then, we highlight recent advances in the emerging fields of musculoskeletal local drug delivery, involving therapeutic drugs (e.g., genes, small molecule therapeutics, and stem cells), novel delivery vehicles (e.g., 3D printing and tissue engineering techniques), and innovative delivery approaches (e.g., multi-drug delivery and smart stimuli-responsive delivery). The review concludes with future perspectives and associated challenges for developing local drug delivery for musculoskeletal applications. STATEMENT OF SIGNIFICANCE: Three important aspects are highlighted in this manuscript: 1) The advanced musculoskeletal drug delivery is introduced from the aspects ranging from musculoskeletal disorders, potential therapeutic solutions, and various drug delivery systems. 2) The recent advances in the emerging fields of musculoskeletal local drug delivery, involving therapeutic drugs (e.g., genes, small molecule therapeutics, and stem cells), novel delivery vehicles (e.g., 3D printing and tissue engineering technique), and innovative delivery approaches (e.g., multi-drug delivery and smart stimuli-responsive delivery), are highlighted. 3) The challenges and perspectives of future research directions in the development of musculoskeletal local drug delivery are presented.
Collapse
Affiliation(s)
- Shichao Zhang
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV 26506-9196, United States
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Bingyun Li
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV 26506-9196, United States.
| |
Collapse
|
31
|
Haugen HJ, Lyngstadaas SP, Rossi F, Perale G. Bone grafts: which is the ideal biomaterial? J Clin Periodontol 2019; 46 Suppl 21:92-102. [DOI: 10.1111/jcpe.13058] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Håvard Jostein Haugen
- Department of Biomaterials; Institute of Clinical Dentistry; Faculty of Dentistry; University of Oslo; Oslo Norway
| | - Ståle Petter Lyngstadaas
- Department of Biomaterials; Institute of Clinical Dentistry; Faculty of Dentistry; University of Oslo; Oslo Norway
- Corticalis AS; Oslo Science Park Oslo Norway
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”; Politecnico di Milano; Milano Italy
| | - Giuseppe Perale
- Industrie Biomediche Insubri SA; Mezzovico-Vira Switzerland
- Biomaterials Laboratory; Institute for Mechanical Engineering and Materials Technology; University of Applied Sciences and Arts of Southern Switzerland; Manno Switzerland
- Department of Surgical Sciences; Faculty of Medical Sciences; Orthopaedic Clinic-IRCCS A.O.U. San Martino; Genova Italy
| |
Collapse
|
32
|
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: 5.0] [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
|
33
|
Tang X, Daneshmandi L, Awale G, Nair LS, Laurencin CT. Skeletal Muscle Regenerative Engineering. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019; 5:233-251. [PMID: 33778155 DOI: 10.1007/s40883-019-00102-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Skeletal muscles have the intrinsic ability to regenerate after minor injury, but under certain circumstances such as severe trauma from accidents, chronic diseases or battlefield injuries the regeneration process is limited. Skeletal muscle regenerative engineering has emerged as a promising approach to address this clinical issue. The regenerative engineering approach involves the convergence of advanced materials science, stem cell science, physical forces, insights from developmental biology, and clinical translation. This article reviews recent studies showing the potential of the convergences of technologies involving biomaterials, stem cells and bioactive factors in concert with clinical translation, in promoting skeletal muscle regeneration. Several types of biomaterials such as electrospun nanofibers, hydrogels, patterned scaffolds, decellularized tissues, and conductive matrices are being investigated. Detailed discussions are given on how these biomaterials can interact with cells and modulate their behavior through physical, chemical and mechanical cues. In addition, the application of physical forces such as mechanical and electrical stimulation are reviewed as strategies that can further enhance muscle contractility and functionality. The review also discusses established animal models to evaluate regeneration in two clinically relevant muscle injuries; volumetric muscle loss (VML) and muscle atrophy upon rotator cuff injury. Regenerative engineering approaches using advanced biomaterials, cells, and physical forces, developmental cues along with insights from immunology, genetics and other aspects of clinical translation hold significant potential to develop promising strategies to support skeletal muscle regeneration.
Collapse
Affiliation(s)
- Xiaoyan Tang
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Leila Daneshmandi
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Guleid Awale
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Lakshmi S Nair
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA.,Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA.,Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA.,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
34
|
Chen X, Song Z, Chen R, Tan S, Huang C, Liu Y, Cheng B, Fu Q. Lysophosphatidic acid enhanced the osteogenic and angiogenic capability of osteoblasts via LPA1/3 receptor. Connect Tissue Res 2019; 60:85-94. [PMID: 29447019 DOI: 10.1080/03008207.2018.1439485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Lysophosphatidic acid is a serum-derived growth factor that is involved in wound healing. Although in its infancy, a growing body of evidence has demonstrated that lysophosphatidic acid exerts a potentially significant role in regulating bone cell biology. However, previous studies mainly focused on the osteoinductive potential of lysophosphatidic acid, its effects on bone tissue vascularization, another essential element during bone regeneration, remains ill-defined so far. Here in this study, we examined the effects of lysophosphatidic acid on osteogenic differentiation as well as the angiogenesis-inducing capacity of pre-osteoblasts, a cell population that coordinates osteogenic and angiogenic processes in bone regenerating niche. Our results showed that treatment of MC3T3-E1 pre-osteoblastic cells with lysophosphatidic acid enhanced alkaline phosphatase activity and matrix mineralization, demonstrating in vitro osteoblastic differentiation. Of particular importance was the finding that vascular endothelial growth factor secretion also increased after lysophosphatidic acid treatment. Lysophosphatidic acid conditioned media of MC3T3-E1 cells was capable of promoting angiogenic behavior of endothelial cells, as evidenced by stimulating proliferation, migration, and tube formation. Besides, inhibition of LPA1/3 receptor abolished lysophosphatidic acid-induced elevation of the osteogenic and angiogenic capability of pre-osteoblasts. Our research demonstrated the important role of lysophosphatidic acid in coupling osteogenesis and angiogenesis during bone remodeling through orchestrating pre-osteoblast behavior, and implications therein for novel and effective treatment strategies for bone regeneration success.
Collapse
Affiliation(s)
- Xiaodan Chen
- a Guanghua School of Stomatology, Hospital of Stomatology , Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology , Guangzhou , Guangdong , P.R. China
| | - Zijun Song
- a Guanghua School of Stomatology, Hospital of Stomatology , Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology , Guangzhou , Guangdong , P.R. China
| | - Rui Chen
- b Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute , Guangdong Second Provincial General Hospital , Guangzhou , Guangdong , P.R. China
| | - Shuyi Tan
- a Guanghua School of Stomatology, Hospital of Stomatology , Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology , Guangzhou , Guangdong , P.R. China.,c The Affiliated Stomatological Hospital of Southern Medical University & Guangdong Provincial Stomatological Hospital , Guangzhou , Guangdong , P.R. China
| | - Chunhuang Huang
- a Guanghua School of Stomatology, Hospital of Stomatology , Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology , Guangzhou , Guangdong , P.R. China
| | - Yanhui Liu
- d The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine , Guangzhou , Guangdong , P.R. China
| | - Bin Cheng
- a Guanghua School of Stomatology, Hospital of Stomatology , Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology , Guangzhou , Guangdong , P.R. China
| | - Qiang Fu
- a Guanghua School of Stomatology, Hospital of Stomatology , Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology , Guangzhou , Guangdong , P.R. China
| |
Collapse
|
35
|
Nakayama KH, Shayan M, Huang NF. Engineering Biomimetic Materials for Skeletal Muscle Repair and Regeneration. Adv Healthc Mater 2019; 8:e1801168. [PMID: 30725530 PMCID: PMC6589032 DOI: 10.1002/adhm.201801168] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/21/2018] [Indexed: 11/12/2022]
Abstract
Although skeletal muscle is highly regenerative following injury or disease, endogenous self-regeneration is severely impaired in conditions of volume traumatic muscle loss. Consequently, tissue engineering approaches are a promising means to regenerate skeletal muscle. Biological scaffolds serve as not only structural support for the promotion of cellular ingrowth but also impart potent modulatory signaling cues that may be beneficial for tissue regeneration. In this work, the progress of tissue engineering approaches for skeletal muscle engineering and regeneration is overviewed, with a focus on the techniques to create biomimetic engineered tissue using extracellular cues. These factors include mechanical and electrical stimulation, geometric patterning, and delivery of growth factors or other bioactive molecules. The progress of evaluating the therapeutic efficacy of these approaches in preclinical models of muscle injury is further discussed.
Collapse
Affiliation(s)
- Karina H Nakayama
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Mahdis Shayan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
36
|
O'Neill E, Rajpura K, Carbone EJ, Awale G, Kan HM, Lo KWH. Repositioning Tacrolimus: Evaluation of the Effect of Short-Term Tacrolimus Treatment on Osteoprogenitor Cells and Primary Cells for Bone Regenerative Engineering. Assay Drug Dev Technol 2019; 17:77-88. [DOI: 10.1089/adt.2018.876] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Edward O'Neill
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
| | - Komal Rajpura
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Connecticut Institute for Clinical and Translational Science, University of Connecticut Health Center, Farmington, Connecticut
| | - Erica J. Carbone
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- UConn Stem Cell Institute, University of Connecticut Health Center, Farmington, Connecticut
| | - Guleid Awale
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
| | - Ho-Man Kan
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Department of Orthopaedic Surgery, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
| | - Kevin W.-H. Lo
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, School of Medicine, Farmington, Connecticut
- Connecticut Institute for Clinical and Translational Science, University of Connecticut Health Center, Farmington, Connecticut
- UConn Stem Cell Institute, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
37
|
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: 2.8] [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
|
38
|
Bone Regenerative Engineering Using a Protein Kinase A-Specific Cyclic AMP Analogue Administered for Short Term. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018. [DOI: 10.1007/s40883-018-0063-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Ferracini R, Martínez Herreros I, Russo A, Casalini T, Rossi F, Perale G. Scaffolds as Structural Tools for Bone-Targeted Drug Delivery. Pharmaceutics 2018; 10:pharmaceutics10030122. [PMID: 30096765 PMCID: PMC6161191 DOI: 10.3390/pharmaceutics10030122] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/19/2022] Open
Abstract
Although bone has a high potential to regenerate itself after damage and injury, the efficacious repair of large bone defects resulting from resection, trauma or non-union fractures still requires the implantation of bone grafts. Materials science, in conjunction with biotechnology, can satisfy these needs by developing artificial bones, synthetic substitutes and organ implants. In particular, recent advances in materials science have provided several innovations, underlying the increasing importance of biomaterials in this field. To address the increasing need for improved bone substitutes, tissue engineering seeks to create synthetic, three-dimensional scaffolds made from organic or inorganic materials, incorporating drugs and growth factors, to induce new bone tissue formation. This review emphasizes recent progress in materials science that allows reliable scaffolds to be synthesized for targeted drug delivery in bone regeneration, also with respect to past directions no longer considered promising. A general overview concerning modeling approaches suitable for the discussed systems is also provided.
Collapse
Affiliation(s)
- Riccardo Ferracini
- Department of Surgical Sciences, Orthopaedic Clinic-IRCCS A.O.U. San Martino, 16132 Genova, Italy.
| | - Isabel Martínez Herreros
- Department of Surgical Sciences, Orthopaedic Clinic-IRCCS A.O.U. San Martino, 16132 Genova, Italy.
| | - Antonio Russo
- Department of Surgical Sciences, Orthopaedic Clinic-IRCCS A.O.U. San Martino, 16132 Genova, Italy.
| | - Tommaso Casalini
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Vladimir-Prelog-Weg 1, 8093 Zurich, Switzerland.
- Biomaterials Laboratory, Institute for Mechanical Engineering and Materials Technology, University of Applied Sciences and Arts of Southern Switzerland, Via Cantonale 2C, Galleria, 26928 Manno, Switzerland.
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, 20131 Milano, Italy.
| | - Giuseppe Perale
- Department of Surgical Sciences, Orthopaedic Clinic-IRCCS A.O.U. San Martino, 16132 Genova, Italy.
- Biomaterials Laboratory, Institute for Mechanical Engineering and Materials Technology, University of Applied Sciences and Arts of Southern Switzerland, Via Cantonale 2C, Galleria, 26928 Manno, Switzerland.
- Industrie Biomediche Insubri SA, Via Cantonale 67, 6805 Mezzovico-Vira, Switzerland.
| |
Collapse
|
40
|
Shi R, Huang Y, Ma C, Wu C, Tian W. Current advances for bone regeneration based on tissue engineering strategies. Front Med 2018; 13:160-188. [PMID: 30047029 DOI: 10.1007/s11684-018-0629-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/14/2017] [Indexed: 01/07/2023]
Abstract
Bone tissue engineering (BTE) is a rapidly developing strategy for repairing critical-sized bone defects to address the unmet need for bone augmentation and skeletal repair. Effective therapies for bone regeneration primarily require the coordinated combination of innovative scaffolds, seed cells, and biological factors. However, current techniques in bone tissue engineering have not yet reached valid translation into clinical applications because of several limitations, such as weaker osteogenic differentiation, inadequate vascularization of scaffolds, and inefficient growth factor delivery. Therefore, further standardized protocols and innovative measures are required to overcome these shortcomings and facilitate the clinical application of these techniques to enhance bone regeneration. Given the deficiency of comprehensive studies in the development in BTE, our review systematically introduces the new types of biomimetic and bifunctional scaffolds. We describe the cell sources, biology of seed cells, growth factors, vascular development, and the interactions of relevant molecules. Furthermore, we discuss the challenges and perspectives that may propel the direction of future clinical delivery in bone regeneration.
Collapse
Affiliation(s)
- Rui Shi
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Yuelong Huang
- Department of Spine Surgery of Beijing Jishuitan Hospital, The Fourth Clinical Medical College of Peking University, Beijing, 100035, China
| | - Chi Ma
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Chengai Wu
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Wei Tian
- Institute of Traumatology and Orthopaedics, Beijing Laboratory of Biomedical Materials, Beijing Jishuitan Hospital, Beijing, 100035, China. .,Department of Spine Surgery of Beijing Jishuitan Hospital, The Fourth Clinical Medical College of Peking University, Beijing, 100035, China.
| |
Collapse
|
41
|
O’Neill E, Awale G, Daneshmandi L, Umerah O, Lo KWH. The roles of ions on bone regeneration. Drug Discov Today 2018; 23:879-890. [DOI: 10.1016/j.drudis.2018.01.049] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/04/2018] [Accepted: 01/29/2018] [Indexed: 12/16/2022]
|
42
|
Wang Y, Newman MR, Benoit DSW. Development of controlled drug delivery systems for bone fracture-targeted therapeutic delivery: A review. Eur J Pharm Biopharm 2018; 127:223-236. [PMID: 29471078 DOI: 10.1016/j.ejpb.2018.02.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/17/2018] [Accepted: 02/17/2018] [Indexed: 01/09/2023]
Abstract
Impaired fracture healing is a major clinical problem that can lead to patient disability, prolonged hospitalization, and significant financial burden. Although the majority of fractures heal using standard clinical practices, approximately 10% suffer from delayed unions or non-unions. A wide range of factors contribute to the risk for nonunions including internal factors, such as patient age, gender, and comorbidities, and external factors, such as the location and extent of injury. Current clinical approaches to treat nonunions include bone grafts and low-intensity pulsed ultrasound (LIPUS), which realizes clinical success only to select patients due to limitations including donor morbidities (grafts) and necessity of fracture reduction (LIPUS), respectively. To date, therapeutic approaches for bone regeneration rely heavily on protein-based growth factors such as INFUSE, an FDA-approved scaffold for delivery of bone morphogenetic protein 2 (BMP-2). Small molecule modulators and RNAi therapeutics are under development to circumvent challenges associated with traditional growth factors. While preclinical studies has shown promise, drug delivery has become a major hurdle stalling clinical translation. Therefore, this review overviews current therapies employed to stimulate fracture healing pre-clinically and clinically, including a focus on drug delivery systems for growth factors, parathyroid hormone (PTH), small molecules, and RNAi therapeutics, as well as recent advances and future promise of fracture-targeted drug delivery.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Maureen R Newman
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Danielle S W Benoit
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Chemical Engineering, 4517 Wegmans Hall, University of Rochester, Rochester, NY 14627, USA; Department of Orthopaedics, 601 Elmwood Ave, University of Rochester, Rochester, NY 14642, USA; Department of Biomedical Genetics, 601 Elmwood Ave, University of Rochester, Rochester, NY 14642, USA; Center for Oral Biology, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
43
|
Narayanan G, Nair LS, Laurencin CT. Regenerative Engineering of the Rotator Cuff of the Shoulder. ACS Biomater Sci Eng 2018; 4:751-786. [PMID: 33418763 DOI: 10.1021/acsbiomaterials.7b00631] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Rotator cuff tears often heal poorly, leading to re-tears after repair. This is in part attributed to the low proliferative ability of the resident cells (tendon fibroblasts and tendon-stem cells) upon injury to the rotator cuff tissue and the low vascularity of the tendon insertion. In addition, surgical outcomes of current techniques used in clinical settings are often suboptimal, leading to the formation of neo-tissue with poor biomechanics and structural characteristics, which results in re-tears. This has prompted interest in a new approach, which we term as "Regenerative Engineering", for regenerating rotator cuff tendons. In the Regenerative Engineering paradigm, roles played by stem cells, scaffolds, growth factors/small molecules, the use of local physical forces, and morphogenesis interplayed with clinical surgery techniques may synchronously act, leading to synergistic effects and resulting in successful tissue regeneration. In this regard, various cell sources such as tendon fibroblasts and adult tissue-derived stem cells have been isolated, characterized, and investigated for regenerating rotator cuff tendons. Likewise, numerous scaffolds with varying architecture, geometry, and mechanical characteristics of biologic and synthetic origin have been developed. Furthermore, these scaffolds have been also fabricated with biochemical cues (growth factors and small molecules), facilitating tissue regeneration. In this Review, various strategies to regenerate rotator cuff tendons using stem cells, advanced materials, and factors in the setting of physical forces under the Regenerative Engineering paradigm are described.
Collapse
Affiliation(s)
- Ganesh Narayanan
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut 06030, United States.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, Connecticut 06030, United States.,Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
| | - Lakshmi S Nair
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut 06030, United States.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, Connecticut 06030, United States.,Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, Connecticut 06030, United States.,Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States.,Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Cato T Laurencin
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut 06030, United States.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, Connecticut 06030, United States.,Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, Connecticut 06030, United States.,Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut 06030, United States.,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, Connecticut 06269, United States.,Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269, United States.,Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut 06269, United States.,Connecticut Institute for Clinical and Translational Science, University of Connecticut Health Center, Farmington, Connecticut 06030, United States
| |
Collapse
|
44
|
Wang Y, Newman MR, Ackun-Farmmer M, Baranello MP, Sheu TJ, Puzas JE, Benoit DSW. Fracture-Targeted Delivery of β-Catenin Agonists via Peptide-Functionalized Nanoparticles Augments Fracture Healing. ACS NANO 2017; 11:9445-9458. [PMID: 28881139 PMCID: PMC5736386 DOI: 10.1021/acsnano.7b05103] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Despite several decades of progress, bone-specific drug delivery is still a major challenge. Current bone-acting drugs require high-dose systemic administration which decreases therapeutic efficacy and increases off-target tissue effects. Here, a bone-targeted nanoparticle (NP) delivery system for a β-catenin agonist, 3-amino-6-(4-((4-methylpiperazin-1-yl)sulfonyl)phenyl)-N-(pyridin-3-yl)pyrazine-2-carboxamide, a glycogen synthase kinase 3 beta (GSK-3β) inhibitor, was developed to enhance fracture healing. The GSK-3β inhibitor loading capacity was found to be 15 wt % within highly stable poly(styrene-alt-maleic anhydride)-b-poly(styrene) NPs, resulting in ∼50 nm particles with ∼ -30 mV surface charge. A peptide with high affinity for tartrate-resistant acid phosphatase (TRAP), a protein deposited by osteoclasts on bone resorptive surfaces, was introduced to the NP corona to achieve preferential delivery to fractured bone. Targeted NPs showed improved pharmacokinetic profiles with greater accumulation at fractured bone, accompanied by significant uptake in regenerative cell types (mesenchymal stem cells (MSCs) and osteoblasts). MSCs treated with drug-loaded NPs in vitro exhibited 2-fold greater β-catenin signaling than free drug that was sustained for 5 days. To verify similar activity in vivo, TOPGAL reporter mice bearing fractures were treated with targeted GSK-3β inhibitor-loaded NPs. Robust β-galactosidase activity was observed in fracture callus and periosteum treated with targeted carriers versus controls, indicating potent β-catenin activation during the healing process. Enhanced bone formation and microarchitecture were observed in mice treated with GSK-3β inhibitor delivered via TRAP-binding peptide-targeted NPs. Specifically, increased bone bridging, ∼4-fold greater torsional rigidity, and greater volumes of newly deposited bone were observed 28 days after treatment, indicating expedited fracture healing.
Collapse
Affiliation(s)
- Yuchen Wang
- 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 R. 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
| | - Marian Ackun-Farmmer
- 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
| | - Michael P. Baranello
- Department of Chemical Engineering, University of Rochester, Rochester, New York 14627, United States
| | - Tzong-Jen Sheu
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - J. Edward Puzas
- 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 Chemical 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
| |
Collapse
|
45
|
Abstract
BACKGROUND Nonhealing bone defects represent an immense biomedical burden. Despite recent advances in protein-based bone regeneration, safety concerns over bone morphogenetic protein-2 have prompted the search for alternative factors. Previously, the authors examined the additive/synergistic effects of hedgehog and Nel-like protein-1 (NELL-1) on the osteogenic differentiation of mesenchymal stem cells in vitro. In this study, the authors sought to leverage their previous findings by applying the combination of Smoothened agonist (SAG), hedgehog signal activator, and NELL-1 to an in vivo critical-size bone defect model. METHODS A 4-mm parietal bone defect was created in mixed-gender CD-1 mice. Treatment groups included control (n = 6), SAG (n = 7), NELL-1 (n = 7), and SAG plus NELL-1 (n = 7). A custom fabricated poly(lactic-co-glycolic acid) disk with hydroxyapatite coating was used as an osteoinductive scaffold. RESULTS Results at 4 and 8 weeks showed increased bone formation by micro-computed tomographic analyses with either stimulus alone (SAG or NELL-1), but significantly greater bone formation with both components combined (SAG plus NELL-1). This included greater bone healing scores and increased bone volume and bone thickness. Histologic analyses confirmed a significant increase in new bone formation with the combination therapy SAG plus NELL-1, accompanied by increased defect vascularization. CONCLUSIONS In summary, the authors' results suggest that combining the hedgehog signaling agonist SAG and NELL-1 has potential as a novel therapeutic strategy for the healing of critical-size bone defects. Future directions will include optimization of dosage and delivery strategy for an SAG and NELL-1 combination product.
Collapse
|
46
|
Narayanan G, Bhattacharjee M, Nair LS, Laurencin CT. Musculoskeletal Tissue Regeneration: the Role of the Stem Cells. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2017. [DOI: 10.1007/s40883-017-0036-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
47
|
Ifegwu OC, Awale G, Rajpura K, Lo KWH, Laurencin CT. Harnessing cAMP signaling in musculoskeletal regenerative engineering. Drug Discov Today 2017; 22:1027-1044. [PMID: 28359841 PMCID: PMC7440772 DOI: 10.1016/j.drudis.2017.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/08/2017] [Accepted: 03/20/2017] [Indexed: 01/28/2023]
Abstract
This paper reviews the most recent findings in the search for small molecule cyclic AMP analogues regarding their potential use in musculoskeletal regenerative engineering.
Collapse
Affiliation(s)
- Okechukwu Clinton Ifegwu
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Guleid Awale
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Chemical and Biomolecular Engineering, University of Connecticut, School of Engineering, Storrs, CT 06030, USA
| | - Komal Rajpura
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Connecticut Institute for Clinical and Translational Science, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Kevin W-H Lo
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA; Connecticut Institute for Clinical and Translational Science, University of Connecticut Health Center, Farmington, CT 06030, USA; UConn Stem Cell Institute, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, School of Engineering, Storrs, CT 06268, USA
| | - Cato T Laurencin
- Institute for Regenerative Engineering, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; The Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA; Connecticut Institute for Clinical and Translational Science, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Medicine, Division of Endocrinology, University of Connecticut Health Center, School of Medicine, Farmington, CT 06030, USA; UConn Stem Cell Institute, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Biomedical Engineering, University of Connecticut, School of Engineering, Storrs, CT 06268, USA.
| |
Collapse
|
48
|
Patel R, Patel M, Kwak J, Iyer AK, Karpoormath R, Desai S, Rarh V. Polymeric microspheres: a delivery system for osteogenic differentiation. POLYM ADVAN TECHNOL 2017. [DOI: 10.1002/pat.4084] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Rajkumar Patel
- School of Electrical and Computer Engineering; The University of Seoul; Seoul 02504 Korea
| | - Madhumita Patel
- Department of Chemistry and Nano Science; Ewha Womans University; Seodaemun-gu Seoul 120-750 South Korea
| | - Jeonghun Kwak
- School of Electrical and Computer Engineering; The University of Seoul; Seoul 02504 Korea
| | - Arun K. Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-Bind) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health, Sciences; Wayne State University; 259 Mack Ave Detroit MI 48201 USA
| | - Rajshekhar Karpoormath
- Department of Pharmaceutical Chemistry, College of Health Sciences; University of Kwa Zulu Natal; Durban 4000 Africa
| | - Shrojal Desai
- Global Infusion Systems R&D at Hospira; Chicago, IL USA
| | - Vimal Rarh
- Department of Chemistry, S.G.T.B. Khalsa College; University of Delhi; Delhi 110007 India
| |
Collapse
|
49
|
Zhang M, Zhang P, Liu Y, Zhou Y. GSK3 inhibitor AR-A014418 promotes osteogenic differentiation of human adipose-derived stem cells via ERK and mTORC2/Akt signaling pathway. Biochem Biophys Res Commun 2017; 490:182-188. [PMID: 28602697 DOI: 10.1016/j.bbrc.2017.06.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 06/07/2017] [Indexed: 01/01/2023]
Abstract
Small molecule-based bone tissue engineering is emerging as a promising strategy for bone defects restoration. In this study, we intended to identify the roles and mechanisms of AR-A014418, a highly selective inhibitor of GSK3, on the osteogenic differentiation. We found that AR-A014418 exhibited a dose-dependent effect on osteogenic differentiation of human adipose-derived stem cells (hASCs). hASCs treated with AR-A014418 showed higher activity of ERK and mTORC2/Akt signaling. Administration of ERK inhibitor U0126 or knockdown of RICTOR by siRNA attenuated AR-A014418 induced osteogenic differentiation of hASCs. Our results suggested that AR-A014418 significantly promoted osteogenic potential of hASCs partially by the activation of ERK and mTORC2/Akt signaling pathway, and might be used for bone tissue engineering as an osteo-inductive factor.
Collapse
Affiliation(s)
- Min Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; National Engineering Lab for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| |
Collapse
|
50
|
Balmayor ER, Geiger JP, Koch C, Aneja MK, van Griensven M, Rudolph C, Plank C. Modified mRNA for BMP-2 in Combination with Biomaterials Serves as a Transcript-Activated Matrix for Effectively Inducing Osteogenic Pathways in Stem Cells. Stem Cells Dev 2017; 26:25-34. [DOI: 10.1089/scd.2016.0171] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Elizabeth R. Balmayor
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Ethris GmbH, Planegg, Germany
| | | | | | | | - Martijn van Griensven
- Experimental Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | - Christian Plank
- Ethris GmbH, Planegg, Germany
- Institute of Molecular Immunology and Experimental Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|