1
|
Bell JA, Mayfield CK, Collon K, Chang S, Gallo MC, Lechtholz-Zey E, Ayad M, Sugiyam O, Tang AH, Park SH, Lieberman JR. In vivo effects of cell seeding technique in an ex vivo regional gene therapy model for bone regeneration. J Biomed Mater Res A 2024; 112:1688-1698. [PMID: 38602243 DOI: 10.1002/jbm.a.37718] [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: 10/04/2023] [Revised: 03/14/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024]
Abstract
When delivering cells on a scaffold to treat a bone defect, the cell seeding technique determines the number and distribution of cells within a scaffold, however the optimal technique has not been established. This study investigated if human adipose-derived stem cells (ASCs) transduced with a lentiviral vector to overexpress bone morphogenetic protein 2 (BMP-2) and loaded on a scaffold using dynamic orbital shaker could reduce the total cell dose required to heal a critical sized bone defect when compared with static seeding. Human ASCs were loaded onto a collagen/biphasic ceramic scaffold using static loading and dynamic orbital shaker techniques, compared with our labs standard loading technique, and implanted into femoral defects of nude rats. Both a low dose and standard dose of transduced cells were evaluated. Outcomes investigated included BMP-2 production, radiographic healing, micro-computerized tomography, histologic assessment, and biomechanical torsional testing. BMP-2 production was higher in the orbital shaker cohort compared with the static seeding cohort. No statistically significant differences were noted in radiographic, histomorphometric, and biomechanical outcomes between the low-dose static and dynamic seeding groups, however the standard-dose static seeding cohort had superior biomechanical properties. The standard-dose 5 million cell dose standard loading cohort had superior maximum torque and torsional stiffness on biomechanical testing. The use of orbital shaker technique was labor intensive and did not provide equivalent biomechanical results with the use of fewer cells.
Collapse
Affiliation(s)
- Jennifer A Bell
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Cory K Mayfield
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Kevin Collon
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Stephanie Chang
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Matthew C Gallo
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Elizabeth Lechtholz-Zey
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Mina Ayad
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Osamu Sugiyam
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Amy H Tang
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Sang-Hyun Park
- J. Vernon Luck Orthopaedic Research Center, Orthopaedic Institute for Children, Los Angeles, California, USA
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| |
Collapse
|
2
|
Mirzavandi Z, Poursamar SA, Amiri F, Bigham A, Rafienia M. 3D printed polycaprolactone/gelatin/ordered mesoporous calcium magnesium silicate nanocomposite scaffold for bone tissue regeneration. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:58. [PMID: 39348082 PMCID: PMC11442632 DOI: 10.1007/s10856-024-06828-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024]
Abstract
Tissue engineering scaffolds are three-dimensional structures that provide an appropriate environment for cellular attachment, proliferation, and differentiation. Depending on their specific purpose, these scaffolds must possess distinct features, including appropriate mechanical properties, porosity, desired degradation rate, and cell compatibility. This investigation aimed to fabricate a new nanocomposite scaffold using a 3D printing technique composed of poly(ε-caprolactone) (PCL)/Gelatin (GEL)/ordered mesoporous calcium-magnesium silicate (om-CMS) particles. Different weight ratios of om-CMS were added and optimized, and a series of scaffolds were constructed for comparison purposes, including PCL 50%/Gel 50%, PCL 50%/Gel 45%/om-CMS%5, and PCL 50%/Gel 40%/om-CMS%10. The optimized weight ratio of om-CMS was 10% without leaving behind negative effects on the filaments' structure. The scaffolds' physical and chemical properties were assessed using various techniques, and their degradation rate, bioactivity potential, cell viability, attachment, and ALP activity were evaluated in vitro. The results demonstrated that the PCL 50%/Gel 40%/om-CMS10% scaffold had promising potential for further studies in bone tissue regeneration.
Collapse
Affiliation(s)
- Zahra Mirzavandi
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Ali Poursamar
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farshad Amiri
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ashkan Bigham
- Institute of Polymers, Composites, and Biomaterials, National Research Council, Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
| | - Mohammad Rafienia
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
3
|
Nelson AL, Mancino C, Gao X, Choe JA, Chubb L, Williams K, Czachor M, Marcucio R, Taraballi F, Cooke JP, Huard J, Bahney C, Ehrhart N. β-catenin mRNA encapsulated in SM-102 lipid nanoparticles enhances bone formation in a murine tibia fracture repair model. Bioact Mater 2024; 39:273-286. [PMID: 38832305 PMCID: PMC11145078 DOI: 10.1016/j.bioactmat.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 06/05/2024] Open
Abstract
Fractures continue to be a global economic burden as there are currently no osteoanabolic drugs approved to accelerate fracture healing. In this study, we aimed to develop an osteoanabolic therapy which activates the Wnt/β-catenin pathway, a molecular driver of endochondral ossification. We hypothesize that using an mRNA-based therapeutic encoding β-catenin could promote cartilage to bone transformation formation by activating the canonical Wnt signaling pathway in chondrocytes. To optimize a delivery platform built on recent advancements in liposomal technologies, two FDA-approved ionizable phospholipids, DLin-MC3-DMA (MC3) and SM-102, were used to fabricate unique ionizable lipid nanoparticle (LNP) formulations and then tested for transfection efficacy both in vitro and in a murine tibia fracture model. Using firefly luciferase mRNA as a reporter gene to track and quantify transfection, SM-102 LNPs showed enhanced transfection efficacy in vitro and prolonged transfection, minimal fracture interference and no localized inflammatory response in vivo over MC3 LNPs. The generated β-cateninGOF mRNA encapsulated in SM-102 LNPs (SM-102-β-cateninGOF mRNA) showed bioactivity in vitro through upregulation of downstream canonical Wnt genes, axin2 and runx2. When testing SM-102-β-cateninGOF mRNA therapeutic in a murine tibia fracture model, histomorphometric analysis showed increased bone and decreased cartilage composition with the 45 μg concentration at 2 weeks post-fracture. μCT testing confirmed that SM-102-β-cateninGOF mRNA promoted bone formation in vivo, revealing significantly more bone volume over total volume in the 45 μg group. Thus, we generated a novel mRNA-based therapeutic encoding a β-catenin mRNA and optimized an SM-102-based LNP to maximize transfection efficacy with a localized delivery.
Collapse
Affiliation(s)
- Anna Laura Nelson
- Steadman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine, Vail, CO, USA
- Colorado State University, School of Biomedical Engineering, Fort Collins CO, USA
| | - Chiara Mancino
- Houston Methodist Research Institute, Center for Musculoskeletal Regeneration, Houston TX, USA
| | - Xueqin Gao
- Steadman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine, Vail, CO, USA
| | - Joshua A. Choe
- University of Wisconsin-Madison, Department of Orthopedics and Rehabilitation, Department of Biomedical Engineering, Medical Scientist Training Program, Madison, WI, USA
| | - Laura Chubb
- Colorado State University, Department of Clinical Sciences, Fort Collins CO, USA
| | - Katherine Williams
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| | - Molly Czachor
- Steadman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine, Vail, CO, USA
| | - Ralph Marcucio
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
| | - Francesca Taraballi
- Houston Methodist Research Institute, Center for Musculoskeletal Regeneration, Houston TX, USA
| | - John P. Cooke
- Houston Methodist Research Institute, Center for RNA Therapeutics, Department of Cardiovascular Sciences, Houston, TX, USA
| | - Johnny Huard
- Steadman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine, Vail, CO, USA
- Colorado State University, Department of Clinical Sciences, Fort Collins CO, USA
| | - Chelsea Bahney
- Steadman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine, Vail, CO, USA
- Colorado State University, Department of Clinical Sciences, Fort Collins CO, USA
- University of California, San Francisco (UCSF), Orthopaedic Trauma Institute, San Francisco, CA, USA
| | - Nicole Ehrhart
- Colorado State University, School of Biomedical Engineering, Fort Collins CO, USA
- Colorado State University, Department of Clinical Sciences, Fort Collins CO, USA
- Colorado State University, Department of Microbiology, Immunology, and Pathology, Fort Collins, CO, USA
| |
Collapse
|
4
|
Wang J, Liu M, Yang C, Pan Y, Ji S, Han N, Sun G. Biomaterials for bone defect repair: Types, mechanisms and effects. Int J Artif Organs 2024; 47:75-84. [PMID: 38166512 DOI: 10.1177/03913988231218884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Bone defects or bone discontinuities caused by trauma, infection, tumours and other diseases have led to an increasing demand for bone grafts and biomaterials. Autologous bone grafts, bone grafts with vascular tips, anastomosed vascular bone grafts and autologous bone marrow components are all commonly used in clinical practice, while oversized bone defects require the use of bone tissue engineering-related biomaterials to repair bone defects and promote bone regeneration. Currently, inorganic components such as polysaccharides and bioceramics, as well as a variety of bioactive proteins, metal ions and stem cells can be loaded into hydrogels or 3D printed scaffold materials to achieve better therapeutic results. In this review, we provide an overview of the types of materials, applications, potential mechanisms and current developments in the repair of bone defects.
Collapse
Affiliation(s)
- Jiaming Wang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Mingchong Liu
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chensong Yang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yutao Pan
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shengchao Ji
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ning Han
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guixin Sun
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
5
|
Nelson AL, Fontana G, Chubb L, Choe J, Williams K, Regan D, Huard J, Murphy W, Ehrhart N, Bahney C. Mineral coated microparticles doped with fluoride and complexed with mRNA prolong transfection in fracture healing. Front Bioeng Biotechnol 2024; 11:1295313. [PMID: 38264578 PMCID: PMC10803474 DOI: 10.3389/fbioe.2023.1295313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024] Open
Abstract
Introduction: Impaired fracture healing, specifically non-union, has been found to occur up to 14% in tibial shaft fractures. The current standard of care to treat non-union often requires additional surgeries which can result in long recovery times. Injectable-based therapies to accelerate fracture healing have the potential to mitigate the need for additional surgeries. Gene therapies have recently undergone significant advancements due to developments in nanotechnology, which improve mRNA stability while reducing immunogenicity. Methods: In this study, we tested the efficacy of mineral coated microparticles (MCM) and fluoride-doped MCM (FMCM) to effectively deliver firefly luciferase (FLuc) mRNA lipoplexes (LPX) to the fracture site. Here, adult mice underwent a tibia fracture and stabilization method and all treatments were locally injected into the fracture. Level of osteogenesis and amount of bone formation were assessed using gene expression and histomorphometry respectively. Localized and systemic inflammation were measured through gene expression, histopathology scoring and measuring C-reactive protein (CRP) in the serum. Lastly, daily IVIS images were taken to track and measure transfection over time. Results: MCM-LPX-FLuc and FMCM-LPX-FLuc were not found to cause any cytotoxic effects when tested in vitro. When measuring the osteogenic potential of each mineral composition, FMCM-LPX-FLuc trended higher in osteogenic markers through qRT-PCR than the other groups tested in a murine fracture and stabilization model. Despite FMCM-LPX-FLuc showing slightly elevated il-1β and il-4 levels in the fracture callus, inflammation scoring of the fracture callus did not result in any differences. Additionally, an acute systemic inflammatory response was not observed in any of the samples tested. The concentration of MCM-LPX-FLuc and FMCM-LPX-FLuc that was used in the murine fracture model did not stimulate bone when analyzed through stereological principles. Transfection efficacy and kinetics of delivery platforms revealed that FMCM-LPX-FLuc prolongs the luciferase signal both in vitro and in vivo. Discussion: These data together reveal that FMCM-LPX-FLuc could serve as a promising mRNA delivery platform for fracture healing applications.
Collapse
Affiliation(s)
- Anna Laura Nelson
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, CO, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Gianluca Fontana
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, United States
| | - Laura Chubb
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Josh Choe
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, United States
| | - Katherine Williams
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Colorado State University, Fort Collins, CO, United States
| | - Dan Regan
- Department of Microbiology, Colorado State University, Fort Collins, CO, United States
| | - Johnny Huard
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, CO, United States
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - William Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Nicole Ehrhart
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Chelsea Bahney
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute (SPRI), Vail, CO, United States
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
- Orthopaedic Trauma Institute, University of California, San Francisco, CA, United States
| |
Collapse
|
6
|
Bell JA, Collon K, Mayfield C, Gallo MC, Chang SW, Sugiyama O, Tang AH, Hollis R, Chopra S, Kohn DB, Lieberman JR. Biodistribution of lentiviral transduced adipose-derived stem cells for "ex-vivo" regional gene therapy for bone repair. Gene Ther 2023; 30:826-834. [PMID: 37568039 DOI: 10.1038/s41434-023-00415-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/31/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023]
Abstract
Ex-vivo gene therapy has been shown to be an effective method for treating bone defects in pre-clinical models. As gene therapy is explored as a potential treatment option in humans, an assessment of the safety profile becomes an important next step. The purpose of this study was to evaluate the biodistribution of viral particles at the defect site and various internal organs in a rat femoral defect model after implantation of human ASCs transduced with lentivirus (LV) with two-step transcriptional activation (TSTA) of bone morphogenetic protein-2 (LV-TSTA-BMP-2). Animals were sacrificed at 4-, 14-, 56-, and 84-days post implantation. The defects were treated with either a standard dose (SD) of 5 million cells or a high dose (HD) of 15 million cells to simulate a supratherapeutic dose. Treatment groups included (1) SD LV-TSTA-BMP-2 (2) HD LV-TSTA-BMP-2, (3) SD LV-TSTA-GFP (4) HD LV-TSTA-GFP and (5) SD nontransduced cells. The viral load at the defect site and ten organs was assessed at each timepoint. Histology of all organs, ipsilateral tibia, and femur were evaluated at each timepoint. There were nearly undetectable levels of LV-TSTA-BMP-2 transduced cells at the defect site at 84-days and no pathologic changes in any organ at all timepoints. In conclusion, human ASCs transduced with a lentiviral vector were both safe and effective in treating critical size bone defects in a pre-clinical model. These results suggest that regional gene therapy using lentiviral vector to treat bone defects has the potential to be a safe and effective treatment in humans.
Collapse
Affiliation(s)
- Jennifer A Bell
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, CA, 90089, USA.
| | - Kevin Collon
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, CA, 90089, USA
| | - Cory Mayfield
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, CA, 90089, USA
| | - Matthew C Gallo
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, CA, 90089, USA
| | - Stephanie W Chang
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, CA, 90089, USA
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, CA, 90089, USA
| | - Amy H Tang
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, CA, 90089, USA
| | - Roger Hollis
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Shefali Chopra
- Department of Pathology, Keck School of Medicine of University of Southern California, 1975 Zonal Ave, Los Angeles, CA, 90089, USA
| | - Donald B Kohn
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, CA, 90089, USA
| |
Collapse
|
7
|
Ball JR, Shelby T, Hernandez F, Mayfield CK, Lieberman JR. Delivery of Growth Factors to Enhance Bone Repair. Bioengineering (Basel) 2023; 10:1252. [PMID: 38002376 PMCID: PMC10669014 DOI: 10.3390/bioengineering10111252] [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: 09/15/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
The management of critical-sized bone defects caused by nonunion, trauma, infection, malignancy, pseudoarthrosis, and osteolysis poses complex reconstruction challenges for orthopedic surgeons. Current treatment modalities, including autograft, allograft, and distraction osteogenesis, are insufficient for the diverse range of pathology encountered in clinical practice, with significant complications associated with each. Therefore, there is significant interest in the development of delivery vehicles for growth factors to aid in bone repair in these settings. This article reviews innovative strategies for the management of critical-sized bone loss, including novel scaffolds designed for controlled release of rhBMP, bioengineered extracellular vesicles for delivery of intracellular signaling molecules, and advances in regional gene therapy for sustained signaling strategies. Improvement in the delivery of growth factors to areas of significant bone loss has the potential to revolutionize current treatment for this complex clinical challenge.
Collapse
Affiliation(s)
- Jacob R. Ball
- Department of Orthopaedic Surgery, University of Southern California Keck School of Medicine, 1500 San Pablo St., Los Angeles, CA 90033, USA
| | | | | | | | | |
Collapse
|
8
|
Zhou J, See CW, Sreenivasamurthy S, Zhu D. Customized Additive Manufacturing in Bone Scaffolds-The Gateway to Precise Bone Defect Treatment. RESEARCH (WASHINGTON, D.C.) 2023; 6:0239. [PMID: 37818034 PMCID: PMC10561823 DOI: 10.34133/research.0239] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023]
Abstract
In the advancing landscape of technology and novel material development, additive manufacturing (AM) is steadily making strides within the biomedical sector. Moving away from traditional, one-size-fits-all implant solutions, the advent of AM technology allows for patient-specific scaffolds that could improve integration and enhance wound healing. These scaffolds, meticulously designed with a myriad of geometries, mechanical properties, and biological responses, are made possible through the vast selection of materials and fabrication methods at our disposal. Recognizing the importance of precision in the treatment of bone defects, which display variability from macroscopic to microscopic scales in each case, a tailored treatment strategy is required. A patient-specific AM bone scaffold perfectly addresses this necessity. This review elucidates the pivotal role that customized AM bone scaffolds play in bone defect treatment, while offering comprehensive guidelines for their customization. This includes aspects such as bone defect imaging, material selection, topography design, and fabrication methodology. Additionally, we propose a cooperative model involving the patient, clinician, and engineer, thereby underscoring the interdisciplinary approach necessary for the effective design and clinical application of these customized AM bone scaffolds. This collaboration promises to usher in a new era of bioactive medical materials, responsive to individualized needs and capable of pushing boundaries in personalized medicine beyond those set by traditional medical materials.
Collapse
Affiliation(s)
- Juncen Zhou
- Department of Biomedical Engineering,
Stony Brook University, Stony Brook, NY, USA
| | - Carmine Wang See
- Department of Biomedical Engineering,
Stony Brook University, Stony Brook, NY, USA
| | - Sai Sreenivasamurthy
- Department of Biomedical Engineering,
Stony Brook University, Stony Brook, NY, USA
| | - Donghui Zhu
- Department of Biomedical Engineering,
Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
9
|
Su D, Swearson S, Krongbaramee T, Sun H, Hong L, Amendt BA. Exploring microRNAs in craniofacial regenerative medicine. Biochem Soc Trans 2023; 51:841-854. [PMID: 37073783 PMCID: PMC11244734 DOI: 10.1042/bst20221448] [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: 11/14/2022] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/20/2023]
Abstract
microRNAs (miRs) have been reported over the decades as important regulators in bone development and bone regeneration. They play important roles in maintaining the stem cell signature as well as regulating stem cell fate decisions. Thus, delivering miRs and miR inhibitors to the defect site is a potential treatment towards craniofacial bone defects. However, there are challenges in translation of basic research to clinics, including the efficiency, specificity, and efficacy of miR manipulation methods and the safety of miR delivery systems. In this review, we will compare miR oligonucleotides, mimics and antagomirs as therapeutic reagents to treat disease and regenerate tissues. Newer technology will be discussed as well as the efficiency and efficacy of using these technologies to express or inhibit miRs in treating and repairing oral tissues. Delivery of these molecules using extracellular vesicles and nanoparticles can achieve different results and depending on their composition will elicit specific effects. We will highlight the specificity, toxicity, stability, and effectiveness of several miR systems in regenerative medicine.
Collapse
Affiliation(s)
- Dan Su
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
| | - Samuel Swearson
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
| | - Tadkamol Krongbaramee
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
- Division of Endodontics, Department of Restorative Dentistry & Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Hongli Sun
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| | - Liu Hong
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| | - Brad A Amendt
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| |
Collapse
|
10
|
Jahangiri S, Rahimnejad M, Nasrollahi Boroujeni N, Ahmadi Z, Motamed Fath P, Ahmadi S, Safarkhani M, Rabiee N. Viral and non-viral gene therapy using 3D (bio)printing. J Gene Med 2022; 24:e3458. [PMID: 36279107 DOI: 10.1002/jgm.3458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/05/2022] [Accepted: 10/15/2022] [Indexed: 12/30/2022] Open
Abstract
The overall success in launching discovered drugs is tightly restricted to the high rate of late-stage failures, which ultimately inhibits the distribution of medicines in markets. As a result, it is imperative that methods reliably predict the effectiveness and, more critically, the toxicity of medicine early in the drug development process before clinical trials be continuously innovated. We must stay up to date with the fast appearance of new infections and diseases by rapidly developing the requisite vaccinations and medicines. Modern in vitro models of disease may be used as an alternative to traditional disease models, and advanced technology can be used for the creation of pharmaceuticals as well as cells, drugs, and gene delivery systems to expedite the drug discovery procedure. Furthermore, in vitro models that mimic the spatial and chemical characteristics of native tissues, such as a 3D bioprinting system or other technologies, have proven to be more effective for drug screening than traditional 2D models. Viral and non-viral gene delivery vectors are a hopeful tool for combinatorial gene therapy, suggesting a quick way of simultaneously deliver multiple genes. A 3D bioprinting system embraces an excellent potential for gene delivery into the different cells or tissues for different diseases, in tissue engineering and regeneration medicine, in which the precise nucleic acid is located in the 3D printed tissues and scaffolds. Non-viral nanocarriers, in combination with 3D printed scaffolds, are applied to their delivery of genes and controlled release properties. There remains, however, a big obstacle in reaching the full potential of 3D models because of a lack of in vitro manufacturing of live tissues. Bioprinting advancements have made it possible to create biomimetic constructions that may be used in various drug discovery research applications. 3D bioprinting also benefits vaccinations, medicines, and relevant delivery methods because of its flexibility and adaptability. This review discusses the potential of 3D bioprinting technologies for pharmaceutical studies.
Collapse
Affiliation(s)
- Sepideh Jahangiri
- Department of Biomedical Sciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Maedeh Rahimnejad
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Biomedical Engineering Institute, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Narges Nasrollahi Boroujeni
- Bioprocess Engineering Research Group, Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Zarrin Ahmadi
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, VIC, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - Puria Motamed Fath
- Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Sepideh Ahmadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moein Safarkhani
- Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Navid Rabiee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, South Korea.,School of Engineering, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
11
|
Collon K, Bell JA, Chang SW, Gallo MC, Sugiyama O, Marks C, Lieberman JR. Effects of cell seeding technique and cell density on BMP-2 production in transduced human mesenchymal stem cells. J Biomed Mater Res A 2022; 110:1944-1952. [PMID: 35950648 PMCID: PMC9804863 DOI: 10.1002/jbm.a.37430] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 01/09/2023]
Abstract
Small animal models have demonstrated the efficacy of ex vivo regional gene therapy using scaffolds loaded with BMP-2-expressing mesenchymal stem cells (MSCs). Prior to clinical translation, optimization of seeding techniques of the transduced cells will be important to minimize time and resource expenditure, while maximizing cell delivery and BMP-2 production. No prior studies have investigated cell-seeding techniques in the setting of transduced cells for gene therapy applications. Using BMP-2-expressing transduced adipose-derived MSCs and a porous ceramic scaffold, this study compared previously described static and dynamic seeding techniques with respect to cell seeding efficiency, uniformity of cell distribution, and in vitro BMP-2 production. Static and negative pressure seeding techniques demonstrated the highest seeding efficiency, while orbital shaking was associated with the greatest increases in BMP-2 production per cell. Low density cell suspensions were associated with the highest seeding efficiency and uniformity of cell distribution, and the greatest increases in BMP-2 production from 2 to 7 days after seeding. Our results highlight the potential for development of an optimized cell density and seeding technique that could greatly reduce the number of MSCs needed to produce therapeutic BMP-2 levels in clinical situations. Further studies are needed to investigate in vivo effects of cell seeding techniques on bone healing.
Collapse
Affiliation(s)
- Kevin Collon
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Jennifer A. Bell
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Stephanie W. Chang
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Matthew C. Gallo
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Osamu Sugiyama
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| | - Carolyn Marks
- Core Center of Excellence in Nano ImagingUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Jay R. Lieberman
- Department of Orthopaedic SurgeryKeck School of Medicine of USCLos AngelesCaliforniaUSA
| |
Collapse
|
12
|
Mayfield CK, Ayad M, Lechtholz-Zey E, Chen Y, Lieberman JR. 3D-Printing for Critical Sized Bone Defects: Current Concepts and Future Directions. Bioengineering (Basel) 2022; 9:680. [PMID: 36421080 PMCID: PMC9687148 DOI: 10.3390/bioengineering9110680] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/15/2023] Open
Abstract
The management and definitive treatment of segmental bone defects in the setting of acute trauma, fracture non-union, revision joint arthroplasty, and tumor surgery are challenging clinical problems with no consistently satisfactory solution. Orthopaedic surgeons are developing novel strategies to treat these problems, including three-dimensional (3D) printing combined with growth factors and/or cells. This article reviews the current strategies for management of segmental bone loss in orthopaedic surgery, including graft selection, bone graft substitutes, and operative techniques. Furthermore, we highlight 3D printing as a technology that may serve a major role in the management of segmental defects. The optimization of a 3D-printed scaffold design through printing technique, material selection, and scaffold geometry, as well as biologic additives to enhance bone regeneration and incorporation could change the treatment paradigm for these difficult bone repair problems.
Collapse
Affiliation(s)
- Cory K. Mayfield
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Mina Ayad
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Elizabeth Lechtholz-Zey
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Yong Chen
- Department of Aerospace and Mechanical Engineering, Viterbi School of Engineering, University of Southern California, Los Angleles, CA 90089, USA
| | - Jay R. Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| |
Collapse
|
13
|
Novel structural designs of 3D-printed osteogenic graft for rapid angiogenesis. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00212-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Watson-Levings RS, Palmer GD, Levings PP, Dacanay EA, Evans CH, Ghivizzani SC. Gene Therapy in Orthopaedics: Progress and Challenges in Pre-Clinical Development and Translation. Front Bioeng Biotechnol 2022; 10:901317. [PMID: 35837555 PMCID: PMC9274665 DOI: 10.3389/fbioe.2022.901317] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
In orthopaedics, gene-based treatment approaches are being investigated for an array of common -yet medically challenging- pathologic conditions of the skeletal connective tissues and structures (bone, cartilage, ligament, tendon, joints, intervertebral discs etc.). As the skeletal system protects the vital organs and provides weight-bearing structural support, the various tissues are principally composed of dense extracellular matrix (ECM), often with minimal cellularity and vasculature. Due to their functional roles, composition, and distribution throughout the body the skeletal tissues are prone to traumatic injury, and/or structural failure from chronic inflammation and matrix degradation. Due to a mixture of environment and endogenous factors repair processes are often slow and fail to restore the native quality of the ECM and its function. In other cases, large-scale lesions from severe trauma or tumor surgery, exceed the body’s healing and regenerative capacity. Although a wide range of exogenous gene products (proteins and RNAs) have the potential to enhance tissue repair/regeneration and inhibit degenerative disease their clinical use is hindered by the absence of practical methods for safe, effective delivery. Cumulatively, a large body of evidence demonstrates the capacity to transfer coding sequences for biologic agents to cells in the skeletal tissues to achieve prolonged delivery at functional levels to augment local repair or inhibit pathologic processes. With an eye toward clinical translation, we discuss the research progress in the primary injury and disease targets in orthopaedic gene therapy. Technical considerations important to the exploration and pre-clinical development are presented, with an emphasis on vector technologies and delivery strategies whose capacity to generate and sustain functional transgene expression in vivo is well-established.
Collapse
Affiliation(s)
- Rachael S. Watson-Levings
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Glyn D. Palmer
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Padraic P. Levings
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - E. Anthony Dacanay
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Christopher H. Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MI, United States
| | - Steven C. Ghivizzani
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
- *Correspondence: Steven C. Ghivizzani,
| |
Collapse
|
15
|
Shibahara K, Hayashi K, Nakashima Y, Ishikawa K. Effects of Channels and Micropores in Honeycomb Scaffolds on the Reconstruction of Segmental Bone Defects. Front Bioeng Biotechnol 2022; 10:825831. [PMID: 35372306 PMCID: PMC8971796 DOI: 10.3389/fbioe.2022.825831] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/01/2022] [Indexed: 01/17/2023] Open
Abstract
The reconstruction of critical-sized segmental bone defects is a key challenge in orthopedics because of its intractability despite technological advancements. To overcome this challenge, scaffolds that promote rapid bone ingrowth and subsequent bone replacement are necessary. In this study, we fabricated three types of carbonate apatite honeycomb (HC) scaffolds with uniaxial channels bridging the stumps of a host bone. These HC scaffolds possessed different channel and micropore volumes. The HC scaffolds were implanted into the defects of rabbit ulnar shafts to evaluate the effects of channels and micropores on bone reconstruction. Four weeks postoperatively, the HC scaffolds with a larger channel volume promoted bone ingrowth compared to that with a larger micropore volume. In contrast, 12 weeks postoperatively, the HC scaffolds with a larger volume of the micropores rather than the channels promoted the scaffold resorption by osteoclasts and bone formation. Thus, the channels affected bone ingrowth in the early stage, and micropores affected scaffold resorption and bone formation in the middle stage. Furthermore, 12 weeks postoperatively, the HC scaffolds with large volumes of both channels and micropores formed a significantly larger amount of new bone than that attained using HC scaffolds with either large volume of channels or micropores, thereby bridging the host bone stumps. The findings of this study provide guidance for designing the pore structure of scaffolds.
Collapse
Affiliation(s)
- Keigo Shibahara
- Department of Biomaterials Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichiro Hayashi
- Department of Biomaterials Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kunio Ishikawa
- Department of Biomaterials Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
16
|
Remy MT, Akkouch A, He L, Eliason S, Sweat ME, Krongbaramee T, Fei F, Qian F, Amendt BA, Song X, Hong L. Rat Calvarial Bone Regeneration by 3D-Printed β-Tricalcium Phosphate Incorporating MicroRNA-200c. ACS Biomater Sci Eng 2021; 7:4521-4534. [PMID: 34437807 PMCID: PMC8441974 DOI: 10.1021/acsbiomaterials.0c01756] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
![]()
Advanced fabrication
methods for bone grafts designed to match
defect sites that combine biodegradable, osteoconductive materials
with potent, osteoinductive biologics would significantly impact the
clinical treatment of large bone defects. In this study, we engineered
synthetic bone grafts using a hybrid approach that combined three-dimensional
(3D-)printed biodegradable, osteoconductive β-tricalcium phosphate
(β-TCP) with osteoinductive microRNA(miR)-200c. 3D-printed β-TCP
scaffolds were fabricated utilizing a suspension-enclosing projection-stereolithography
(SEPS) process to produce constructs with reproducible microarchitectures
that enhanced the osteoconductive properties of β-TCP. Collagen
coating on 3D-printed β-TCP scaffolds slowed the release of
plasmid DNA encoding miR-200c compared to noncoated
constructs. 3D-printed β-TCP scaffolds coated with miR-200c-incorporated collagen increased the transfection efficiency of miR-200c of both rat and human BMSCs and additionally increased
osteogenic differentiation of hBMSCs in vitro. Furthermore, miR-200c-incorporated scaffolds significantly enhanced bone
regeneration in critical-sized rat calvarial defects. These results
strongly indicate that bone grafts combining SEPS 3D-printed osteoconductive
biomaterial-based scaffolds with osteoinductive miR-200c can be used
as superior bone substitutes for the clinical treatment of large bone
defects.
Collapse
Affiliation(s)
- Matthew T Remy
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Adil Akkouch
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Li He
- Department of Industrial and Systems Engineering, College of Engineering, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Steven Eliason
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Mason E Sweat
- Department of Industrial and Systems Engineering, College of Engineering, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Tadkamol Krongbaramee
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Fan Fei
- Department of Industrial and Systems Engineering, College of Engineering, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Fang Qian
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Brad A Amendt
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States.,Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, United States.,Center for Craniofacial Anomalies Research, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Xuan Song
- Department of Industrial and Systems Engineering, College of Engineering, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Liu Hong
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States.,Center for Craniofacial Anomalies Research, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, United States
| |
Collapse
|
17
|
Sakemi Y, Hayashi K, Tsuchiya A, Nakashima Y, Ishikawa K. Reconstruction of critical-size segmental defects in rat femurs using carbonate apatite honeycomb scaffolds. J Biomed Mater Res A 2021; 109:1613-1622. [PMID: 33644971 DOI: 10.1002/jbm.a.37157] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/01/2020] [Accepted: 02/10/2021] [Indexed: 12/19/2022]
Abstract
Critical-size segmental defects are formidable challenges in orthopedic surgery. Various scaffolds have been developed to facilitate bone reconstruction within such defects. Many previously studied scaffolds achieved effective outcomes with a combination of high cost, high-risk growth factors or stem cells. Herein, we developed honeycomb scaffolds (HCSs) comprising carbonate apatite (CO3 Ap) containing 8% carbonate, identical to human bone composition. The CO3 Ap HCSs were white-columned blocks harboring regularly arranged macropore channels of a size and wall thickness of 156 ± 5 μm and 102 ± 10 μm, respectively. The compressive strengths of the HCSs parallel and perpendicular to the macropore channel direction were 51.0 ± 11.8 and 15.6 ± 2.2 MPa, respectively. The HCSs were grafted into critical-sized segmental defects in rat femurs. The HCSs bore high-load stresses without any observed breakage. Two-weeks post-implantation, calluses formed around the HCSs and immature bone formed in the HCS interior. The calluses and immature bone matured until 8 weeks via endochondral ossification. At 12 weeks post-implantation, large parts of the HCSs were gradually replaced by newly formed bone. The bone reconstruction efficacy of the CO3 Ap HCSs alone was comparable to that of protein and cell scaffolds, while achieving a lower cost and increased safety.
Collapse
Affiliation(s)
- Yuta Sakemi
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichiro Hayashi
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Akira Tsuchiya
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kunio Ishikawa
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
18
|
Ihn H, Kang H, Iglesias B, Sugiyama O, Tang A, Hollis R, Bougioukli S, Skorka T, Park S, Longjohn D, Oakes DA, Kohn DB, Lieberman JR. Regional Gene Therapy with Transduced Human Cells: The Influence of "Cell Dose" on Bone Repair. Tissue Eng Part A 2021; 27:1422-1433. [PMID: 33882718 DOI: 10.1089/ten.tea.2020.0382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Regional gene therapy using a lentiviral vector containing the BMP-2 complementary DNA (cDNA) has been shown to heal critical-sized bone defects in rodent models. An appropriate "cellular dose" needs to be defined for eventual translation into human trials. The purpose of this study was to evaluate bone defect healing potential and quality using three different doses of transduced human bone marrow cells (HBMCs). HBMCs were transduced with a lentiviral vector containing either BMP-2 or green fluorescent protein (GFP). All cells were loaded onto compression-resistant matrices and implanted in the bone defect of athymic rats. Treatment groups included femoral defects that were treated with a low-dose (1 × 106 cells), standard-dose (5 × 106 cells), and high-dose (1.5 × 107 cells) HBMCs transduced with lentiviral vector containing BMP-2 cDNA. The three control groups were bone defects treated with HBMCs that were either nontransduced or transduced with vector containing GFP. All animals were sacrificed at 12 weeks. The bone formed in each defect was evaluated with plain radiographs, microcomputed tomography (microCT), histomorphometric analysis, and biomechanical testing. Bone defects treated with higher doses of BMP-2-producing cells were more likely to have healed (6/14 of the low-dose group; 12/14 of the standard-dose group; 14/14 of the high-dose group; χ2(2) = 15.501, p < 0.001). None of the bone defects in the control groups had healed. Bone defects treated with high dose and standard dose of BMP-2-producing cells consistently outperformed those treated with a low dose in terms of bone formation, as assessed by microCT and histomorphometry, and biomechanical parameters. However, statistical significance was only seen between defects treated with high dose and low dose. Larger doses of BMP-2-producing cells were associated with a higher likelihood of forming heterotopic ossification. Femurs treated with a standard- and high-dose BMP-2-producing cells demonstrated similar healing and biomechanical properties. Increased doses of BMP-2 delivered through higher cell doses have the potential to heal large bone defects. Adapting regional gene therapy for use in humans will require a balance between promoting bone repair and limiting heterotopic ossification. Impact statement Critical bone loss may result from complex traumatic bone injury (i.e., open fracture or blast injury), revision total joint arthroplasty, and spine pseudoarthrosis. This is a challenging clinical problem to treat and regional gene therapy is an innovative means of addressing it. This study provides information regarding the quantity of cells or "cell dose" of transduced cells needed to treat a critical-sized bone defect in a rat model. This information may be extrapolated for use in humans in future trials.
Collapse
Affiliation(s)
- Hansel Ihn
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| | - Hyunwoo Kang
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| | - Brenda Iglesias
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| | - Osamu Sugiyama
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| | - Amy Tang
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| | - Roger Hollis
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, California, USA
| | - Sofia Bougioukli
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| | - Tautis Skorka
- USC Molecular Imaging Center, Los Angeles, California, USA
| | - Sanghyun Park
- Orthopaedic Institute for Children, J. Vernon Luck. Sr., Orthopedic Research Center, Los Angeles, California, USA
| | - Donald Longjohn
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| | - Daniel A Oakes
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| | - Donald B Kohn
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, California, USA.,Department of Molecular & Medical Pharmacology, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California, USA.,Eli & Edythe Broad Center for Regenerative Medicine & Stem Cell Research, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Jay R Lieberman
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
19
|
Xu H, Wang C, Liu C, Li J, Peng Z, Guo J, Zhu L. Stem cell-seeded 3D-printed scaffolds combined with self-assembling peptides for bone defect repair. Tissue Eng Part A 2021; 28:111-124. [PMID: 34157886 DOI: 10.1089/ten.tea.2021.0055] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Bone defects caused by infection, tumor, trauma and so on remain difficult to treat clinically. Bone tissue engineering (BTE) has great application prospect in promoting bone defect repair. Polycaprolactone (PCL) is a commonly used material for creating BTE scaffolds. In addition, self-assembling peptides (SAPs) can function as the extracellular matrix and promote osteogenesis and angiogenesis. In the work, a PCL scaffold was constructed by 3D printing, then integrated with bone marrow mesenchymal stem cells (BMSCs) and SAPs. The research aimed to assess the bone repair ability of PCL/BMSC/SAP implants. BMSC proliferation in PCL/SAP scaffolds was assessed via Cell Counting Kit-8. In vitro osteogenesis of BMSCs cultured in PCL/SAP scaffolds was assessed by alkaline phosphatase staining and activity assays. Enzyme linked immunosorbent assays were also performed to detect the levels of osteogenic factors. The effects of BMSC-conditioned medium from 3D culture systems on the migration and angiogenesis of human umbilical vein endothelial cells (HUVECs) were assessed by scratch, transwell, and tube formation assays. After 8 weeks of in vivo transplantation, radiography and histology were used to evaluate bone regeneration, and immunohistochemistry staining was utilized to detect neovascularization. In vitro results demonstrated that PCL/SAP scaffolds promoted BMSC proliferation and osteogenesis compared to PCL scaffolds, and the PCL/BMSC/SAP conditional medium (CM) enhanced HUVEC migration and angiogenesis compared to the PCL/BMSC CM. In vivo results showed that, compared to the blank control, PCL, and PCL/BMSC groups, the PCL/BMSC/SAP group had significantly increased bone and blood vessel formation. Thus, the combination of BMSC-seeded 3D-printed PCL and SAPs can be an effective approach for treating bone defects.
Collapse
Affiliation(s)
- Haixia Xu
- Department of Spine Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China;
| | - Chengqiang Wang
- Department of Spine Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China;
| | - Chun Liu
- Department of Spine Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China;
| | - Jianjun Li
- Department of Spine Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China;
| | - Ziyue Peng
- Department of Spine Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China;
| | - Jiasong Guo
- Department of Spine Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Histology and Embryology, Southern Medical University, Guangzhou, China.,Key Laboratory of Tissue Construction and Detection of Guangdong Province, Guangzhou, China.,Institute of Bone Biology, Academy of Orthopaedics, Guangdong Province, Guangzhou, China;
| | - Lixin Zhu
- Department of Spine Surgery, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China;
| |
Collapse
|
20
|
Huang CC. New Designed Decellularized Scaffolds for Scaffold-based Gene Therapy from Elastic Cartilages via Supercritical Carbon Dioxide Fluid and Alkaline/Protease Treatments. Curr Gene Ther 2021; 22:162-167. [PMID: 34148537 DOI: 10.2174/1566523219666210618151843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/26/2021] [Accepted: 04/02/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Scaffold-based gene therapy provides a promising approach for tissue engineering, which is important and popular, that combines medical applications with engineering materials knowledge. OBJECTIVE The decellularization techniques were employed to remove the cellular components from porcine elastic cartilages, leaving a native decellularized extracellular matrix(dECM) composition and architecture integrity of largely insoluble collagen, elastin, and tightly bound glycosaminoglycans. For newly designed collagen scaffold samples, elastic cartilages was hydrolyzed by protease with different concentrations. In this way, it could gain state completely and clearly. METHODS An extraction process of supercritical carbon dioxide(ScCO2) was used to remove cellular components from porcine elastic cartilage. The dECM scaffolds with collagen must be characterized by Fourier transform infrared spectroscopy(FTIR), thermo-gravimetric analysis (TGA), and scanning electron microscope(SEM). RESULTS The study provided a new treatment combined with supercritical carbon dioxide and alkaline/protease to prepare dECM scaffolds with hole-scaffold microstructures and introduce into a potential application on osteochondral tissue engineering using scaffold-based gene therapy. The new process is simple and efficient. The pore-scaffold microstructures were observed in dECM scaffolds derived from porcine elastic cartilages. The Tdmax values of the resulting dECM scaffolds were observed over 330oC. CONCLUSION A series of new scaffolds were successfully obtained from porcine tissue by using ScCO2 and alkaline/enzyme treatments such as an aqueous mixing solution of NH4OH and papain. The dECM scaffolds with high thermal stability were obtained. The resulting scaffold with clean pore-scaffold microstructure could be a potential application for scaffold-based gene therapy.
Collapse
|
21
|
Kang HP, Ihn H, Robertson DM, Chen X, Sugiyama O, Tang A, Hollis R, Skorka T, Longjohn D, Oakes D, Shah R, Kohn D, Jakus AE, Lieberman JR. Regional gene therapy for bone healing using a 3D printed scaffold in a rat femoral defect model. J Biomed Mater Res A 2021; 109:2346-2356. [PMID: 34018305 DOI: 10.1002/jbm.a.37217] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/27/2021] [Accepted: 05/02/2021] [Indexed: 11/07/2022]
Abstract
At the present time there are no consistently satisfactory treatment options for some challenging bone loss scenarios. We have previously reported on the properties of a novel 3D-printed hydroxyapatite-composite material in a pilot study, which demonstrated osteoconductive properties but was not tested in a rigorous, clinically relevant model. We therefore utilized a rat critical-sized femoral defect model with a scaffold designed to match the dimensions of the bone defect. The scaffolds were implanted in the bone defect after being loaded with cultured rat bone marrow cells (rBMC) transduced with a lentiviral vector carrying the cDNA for BMP-2. This experimental group was compared against 3 negative and positive control groups. The experimental group and positive control group loaded with rhBMP-2 demonstrated statistically equivalent radiographic and histologic healing of the defect site (p > 0.9), and significantly superior to all three negative control groups (p < 0.01). However, the healed defects remained biomechanically inferior to the unoperated, contralateral femurs (p < 0.01). When combined with osteoinductive signals, the scaffolds facilitate new bone formation in the defect. However, the scaffold alone was not sufficient to promote adequate healing, suggesting that it is not substantially osteoinductive as currently structured. The combination of gene therapy with 3D-printed scaffolds is quite promising, but additional work is required to optimize scaffold geometry, cell dosage and delivery.
Collapse
Affiliation(s)
- H Paco Kang
- Department of Orthopaedic Surgery, University of Southern California; Los Angeles, California, USA
| | - Hansel Ihn
- Department of Orthopaedic Surgery, University of Southern California; Los Angeles, California, USA
| | - Djani M Robertson
- Department of Orthopaedic Surgery, University of Southern California; Los Angeles, California, USA
| | - Xiao Chen
- Department of Orthopaedic Surgery, University of Southern California; Los Angeles, California, USA
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, University of Southern California; Los Angeles, California, USA
| | - Amy Tang
- Department of Orthopaedic Surgery, University of Southern California; Los Angeles, California, USA
| | | | - Tautis Skorka
- Department of Orthopaedic Surgery, University of Southern California; Los Angeles, California, USA
| | - Donald Longjohn
- Department of Orthopaedic Surgery, University of Southern California; Los Angeles, California, USA
| | - Daniel Oakes
- Department of Orthopaedic Surgery, University of Southern California; Los Angeles, California, USA
| | | | - Donald Kohn
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
| | | | | |
Collapse
|
22
|
Musculoskeletal tissue engineering: Regional gene therapy for bone repair. Biomaterials 2021; 275:120901. [PMID: 34091300 DOI: 10.1016/j.biomaterials.2021.120901] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/24/2021] [Accepted: 05/14/2021] [Indexed: 02/07/2023]
Abstract
Bone loss associated with fracture nonunion, revision total joint arthroplasty (TJA), and pseudoarthrosis of the spine presents a challenging clinical scenario for the orthopaedic surgeon. Current treatment options including autograft, allograft, bone graft substitutes, and bone transport techniques are associated with significant morbidity, high costs, and prolonged treatment regimens. Unfortunately, these treatment strategies have proven insufficient to safely and consistently heal bone defects in the stringent biological environments often encountered in clinical cases of bone loss. The application of tissue engineering (TE) to musculoskeletal pathology has uncovered exciting potential treatment strategies for challenging bone loss scenarios in orthopaedic surgery. Regional gene therapy involves the local implantation of nucleic acids or genetically modified cells to direct specific protein expression, and has shown promise as a potential TE technique for the regeneration of bone. Preclinical studies in animal models have demonstrated the ability of regional gene therapy to safely and effectively heal critical sized bone defects which otherwise do not heal. The purpose of the present review is to provide a comprehensive overview of the current status of gene therapy applications for TE in challenging bone loss scenarios, with an emphasis on gene delivery methods and models, scaffold biomaterials, preclinical results, and future directions.
Collapse
|
23
|
Yao C, Lai Y, Chen Y, Cheng C. Bone Morphogenetic Protein‐2‐Activated 3D‐Printed Polylactic Acid Scaffolds to Promote Bone Regrowth and Repair. Macromol Biosci 2020; 20:e2000161. [DOI: 10.1002/mabi.202000161] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/28/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Chun‐Hsu Yao
- Department of Biomedical Imaging and Radiological Science China Medical University Taichung Taiwan 404 Republic of China
- School of Chinese Medicine China Medical University Taichung Taiwan 404 Republic of China
- Biomaterials Translational Research Center China Medical University Hospital Taichung Taiwan 404 Republic of China
- Department of Biomedical Informatics Asia University Taichung Taiwan 413 Republic of China
| | - Yi‐Hui Lai
- Department of Biomedical Imaging and Radiological Science China Medical University Taichung Taiwan 404 Republic of China
| | - Yi‐Wen Chen
- Graduate Institute of Clinical Medical Science China Medical University Taichung Taiwan 404 Republic of China
- Graduate Institute of Biomedical Sciences China Medical University Taichung Taiwan 404 Republic of China
- 3D Printing Medical Research Institute Asia University Taichung Taiwan 413 Republic of China
| | - Cheng‐Hsin Cheng
- Department of Neurosurgery An Nan Hospital China Medical University Tainan Taiwan 709 Republic of China
- Graduate Institute of Medical Science Chang Jung Christian University Tainan Taiwan 711 Republic of China
| |
Collapse
|
24
|
BMP-2 Gene Delivery-Based Bone Regeneration in Dentistry. Pharmaceutics 2019; 11:pharmaceutics11080393. [PMID: 31387267 PMCID: PMC6723260 DOI: 10.3390/pharmaceutics11080393] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/22/2019] [Accepted: 08/02/2019] [Indexed: 02/06/2023] Open
Abstract
Bone morphogenetic protein-2 (BMP-2) is a potent growth factor affecting bone formation. While recombinant human BMP-2 (rhBMP-2) has been commercially available in cases of non-union fracture and spinal fusion in orthopaedics, it has also been applied to improve bone regeneration in challenging cases requiring dental implant treatment. However, complications related to an initially high dosage for maintaining an effective physiological concentration at the defect site have been reported, although an effective and safe rhBMP-2 dosage for bone regeneration has not yet been determined. In contrast to protein delivery, BMP-2 gene transfer into the defect site induces BMP-2 synthesis in vivo and leads to secretion for weeks to months, depending on the vector, at a concentration of nanograms per milliliter. BMP-2 gene delivery is advantageous for bone wound healing process in terms of dosage and duration. However, safety concerns related to viral vectors are one of the hurdles that need to be overcome for gene delivery to be used in clinical practice. Recently, commercially available gene therapy has been introduced in orthopedics, and clinical trials in dentistry have been ongoing. This review examines the application of BMP-2 gene therapy for bone regeneration in the oral and maxillofacial regions and discusses future perspectives of BMP-2 gene therapy in dentistry.
Collapse
|