1
|
Gallo MC, Elias A, Reynolds J, Ball JR, Lieberman JR. Regional Gene Therapy for Bone Tissue Engineering: A Current Concepts Review. Bioengineering (Basel) 2025; 12:120. [PMID: 40001640 PMCID: PMC11852166 DOI: 10.3390/bioengineering12020120] [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: 12/24/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
The management of segmental bone defects presents a complex reconstruction challenge for orthopedic surgeons. Current treatment options are limited by efficacy across the spectrum of injury, morbidity, and cost. Regional gene therapy is a promising tissue engineering strategy for bone repair, as it allows for local implantation of nucleic acids or genetically modified cells to direct specific protein expression. In cell-based gene therapy approaches, a variety of different cell types have been described including mesenchymal stem cells (MSCs) derived from multiple sources-bone marrow, adipose, skeletal muscle, and umbilical cord tissue, among others. MSCs, in particular, have been well studied, as they serve as a source of osteoprogenitor cells in addition to providing a vehicle for transgene delivery. Furthermore, MSCs possess immunomodulatory properties, which may support the development of an allogeneic "off-the-shelf" gene therapy product. Identifying an optimal cell type is paramount to the successful clinical translation of cell-based gene therapy approaches. Here, we review current strategies for the management of segmental bone loss in orthopedic surgery, including bone grafting, bone graft substitutes, and operative techniques. We also highlight regional gene therapy as a tissue engineering strategy for bone repair, with a focus on cell types and cell sources suitable for this application.
Collapse
Affiliation(s)
- Matthew C. Gallo
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Aura Elias
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Julius Reynolds
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Jacob R. Ball
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
| | - Jay R. Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (A.E.); (J.R.); (J.R.B.)
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
2
|
Sarkar A, Gallo MC, Bell JA, Mayfield CK, Ball JR, Ayad M, Lechtholz-Zey E, Chang SW, Sugiyama O, Evseenko D, Lieberman JR. Ex Vivo Regional Gene Therapy Compared to Recombinant BMP-2 for the Treatment of Critical-Size Bone Defects: An In Vivo Single-Cell RNA-Sequencing Study. Bioengineering (Basel) 2025; 12:29. [PMID: 39851303 PMCID: PMC11762083 DOI: 10.3390/bioengineering12010029] [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: 11/10/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 01/26/2025] Open
Abstract
Ex vivo regional gene therapy is a promising tissue-engineering strategy for bone regeneration: osteogenic mesenchymal stem cells (MSCs) can be genetically modified to express an osteoinductive stimulus (e.g., bone morphogenetic protein-2), seeded onto an osteoconductive scaffold, and then implanted into a bone defect to exert a therapeutic effect. Compared to recombinant human BMP-2 (rhBMP-2), which is approved for clinical use, regional gene therapy may have unique benefits related to the addition of MSCs and the sustained release of BMP-2. However, the cellular and transcriptional mechanisms regulating the response to these two strategies for BMP-2 mediated bone regeneration are largely unknown. Here, for the first time, we performed single-cell RNA sequencing (10x Genomics) of hematoma tissue in six rats with critical-sized femoral defects that were treated with either regional gene therapy or rhBMP-2. Our unbiased bioinformatic analysis of 2393 filtered cells in each group revealed treatment-specific differences in their cellular composition, transcriptional profiles, and cellular communication patterns. Gene therapy treatment induced a more robust chondrogenic response, as well as a decrease in the proportion of fibroblasts and the expression of profibrotic pathways. Additionally, gene therapy was associated with an anti-inflammatory microenvironment; macrophages expressing canonical anti-inflammatory markers were more common in the gene therapy group. In contrast, pro-inflammatory markers were more highly expressed in the rhBMP-2 group. Collectively, the results of our study may offer insights into the unique pathways through which ex vivo regional gene therapy can augment bone regeneration compared to rhBMP-2. Furthermore, an improved understanding of the cellular pathways involved in segmental bone defect healing may allow for the further optimization of regional gene therapy or other bone repair strategies.
Collapse
Affiliation(s)
- Arijita Sarkar
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Matthew C. Gallo
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Jennifer A. Bell
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Cory K. Mayfield
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Jacob R. Ball
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Mina Ayad
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Elizabeth Lechtholz-Zey
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Stephanie W. Chang
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
| | - Jay R. Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA; (M.C.G.); (J.A.B.); (C.K.M.); (J.R.B.); (M.A.); (E.L.-Z.); (S.W.C.); (O.S.); (D.E.); (J.R.L.)
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
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
|
6
|
Wang W, Chen B, Yang J, Li Y, Ding H, Liu H, Yuan C. Sema3A Modified PDLSCs Exhibited Enhanced Osteogenic Capabilities and Stimulated Differentiation of Pre-Osteoblasts. Cell Biochem Biophys 2023; 81:543-552. [PMID: 37421591 DOI: 10.1007/s12013-023-01148-x] [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] [Accepted: 06/23/2023] [Indexed: 07/10/2023]
Abstract
Genetically engineered stem cells, not only acting as vector delivering growth factors or cytokines but also exhibiting improved cell properties, are promising cells for periodontal tissue regeneration. Sema3A is a power secretory osteoprotective factor. In this study, we aimed to construct Sema3A modified periodontal ligament stem cells (PDLSCs) and evaluated their osteogenic capability and crosstalk with pre-osteoblasts MC3T3-E1. First, Sema3A modified PDLSCs was constructed using lentivirus infection system carrying Sema3A gene and the transduction efficiency was analyzed. The osteogenic differentiation and proliferation of Sema3A-PDLSCs was evaluated. Then, MC3T3-E1 was directly co-cultured with Sema3A-PDLSCs or cultured in condition medium of Sema3A-PDLSCs and the osteogenic ability of MC3T3-E1 was assessed. The results showed that Sema3A-PDLSCs expressed and secreted upregulated Sema3A protein, which confirmed successful construction of Sema3A modified PDLSCs. After osteogenic induction, Sema3A-PDLSCs expressed upregulated ALP, OCN, RUNX2, and SP7 mRNA, expressed higher ALP activity, and produced more mineralization nodes, compared with Vector-PDLSCs. Whereas, there was no obvious differences in proliferation between Sema3A-PDLSCs and Vector-PDLSCs. MC3T3-E1 expressed upregulated mRNA of ALP, OCN, RUNX2, and SP7 when directly co-cultured with Sema3A-PDLSCs than Vector-PDLSCs. MC3T3-E1 also expressed upregulated osteogenic markers, showed higher ALP activity, and produced more mineralization nodes when cultured using condition medium of Sema3A-PDLSCs instead of Vector-PDLSCs. In conclusion, our results indicated that Sema3A modified PDLSCs showed enhanced osteogenic capability, and also facilitated differentiation of pre-osteoblasts.
Collapse
Affiliation(s)
- Wen Wang
- School of Stomatology, Xuzhou Medical University, Xuzhou, China
- Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Banghui Chen
- School of Stomatology, Xuzhou Medical University, Xuzhou, China
- Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jintao Yang
- School of Stomatology, Xuzhou Medical University, Xuzhou, China
| | - Yizhou Li
- School of Stomatology, Xuzhou Medical University, Xuzhou, China
| | - Haonan Ding
- School of Stomatology, Nanjing Medical University, Nanjing, China
| | - Hao Liu
- School of Stomatology, Xuzhou Medical University, Xuzhou, China.
- Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Changyong Yuan
- School of Stomatology, Xuzhou Medical University, Xuzhou, China.
- Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
7
|
Liu H, Müller PE, Aszódi A, Klar RM. Osteochondrogenesis by TGF-β3, BMP-2 and noggin growth factor combinations in an ex vivo muscle tissue model: Temporal function changes affecting tissue morphogenesis. Front Bioeng Biotechnol 2023; 11:1140118. [PMID: 37008034 PMCID: PMC10060664 DOI: 10.3389/fbioe.2023.1140118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
In the absence of clear molecular insight, the biological mechanism behind the use of growth factors applied in osteochondral regeneration is still unresolved. The present study aimed to resolve whether multiple growth factors applied to muscle tissue in vitro, such as TGF-β3, BMP-2 and Noggin, can lead to appropriate tissue morphogenesis with a specific osteochondrogenic nature, thereby revealing the underlying molecular interaction mechanisms during the differentiation process. Interestingly, although the results showed the typical modulatory effect of BMP-2 and TGF-β3 on the osteochondral process, and Noggin seemingly downregulated specific signals such as BMP-2 activity, we also discovered a synergistic effect between TGF-β3 and Noggin that positively influenced tissue morphogenesis. Noggin was observed to upregulate BMP-2 and OCN at specific time windows of culture in the presence of TGF-β3, suggesting a temporal time switch causing functional changes in the signaling protein. This implies that signals change their functions throughout the process of new tissue formation, which may depend on the presence or absence of specific singular or multiple signaling cues. If this is the case, the signaling cascade is far more intricate and complex than originally believed, warranting intensive future investigations so that regenerative therapies of a critical clinical nature can function properly.
Collapse
Affiliation(s)
- Heng Liu
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, Munich, Germany
- Department of Orthopaedics and Traumatology, Beijing Jishuitan Hospital, The Fourth Medical College of Peking University, Beijing, China
- *Correspondence: Heng Liu, ; Roland M. Klar,
| | - Peter E. Müller
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, Munich, Germany
| | - Attila Aszódi
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, Munich, Germany
| | - Roland M. Klar
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, Munich, Germany
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, MO, United States
- *Correspondence: Heng Liu, ; Roland M. Klar,
| |
Collapse
|
8
|
Hosseinkhani H, Domb AJ, Sharifzadeh G, Nahum V. Gene Therapy for Regenerative Medicine. Pharmaceutics 2023; 15:856. [PMID: 36986717 PMCID: PMC10057434 DOI: 10.3390/pharmaceutics15030856] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
The development of biological methods over the past decade has stimulated great interest in the possibility to regenerate human tissues. Advances in stem cell research, gene therapy, and tissue engineering have accelerated the technology in tissue and organ regeneration. However, despite significant progress in this area, there are still several technical issues that must be addressed, especially in the clinical use of gene therapy. The aims of gene therapy include utilising cells to produce a suitable protein, silencing over-producing proteins, and genetically modifying and repairing cell functions that may affect disease conditions. While most current gene therapy clinical trials are based on cell- and viral-mediated approaches, non-viral gene transfection agents are emerging as potentially safe and effective in the treatment of a wide variety of genetic and acquired diseases. Gene therapy based on viral vectors may induce pathogenicity and immunogenicity. Therefore, significant efforts are being invested in non-viral vectors to enhance their efficiency to a level comparable to the viral vector. Non-viral technologies consist of plasmid-based expression systems containing a gene encoding, a therapeutic protein, and synthetic gene delivery systems. One possible approach to enhance non-viral vector ability or to be an alternative to viral vectors would be to use tissue engineering technology for regenerative medicine therapy. This review provides a critical view of gene therapy with a major focus on the development of regenerative medicine technologies to control the in vivo location and function of administered genes.
Collapse
Affiliation(s)
- Hossein Hosseinkhani
- Innovation Center for Advanced Technology, Matrix, Inc., New York, NY 10019, USA
| | - Abraham J. Domb
- The Center for Nanoscience and Nanotechnology, Alex Grass Center for Drug Design and Synthesis and Cannabinoids Research, School of Pharmacy, Faculty of Medicine, Institute of Drug Research, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Ghorbanali Sharifzadeh
- Department of Polymer Engineering, School of Chemical Engineering, Universiti Teknologi Malaysia, Skudai 81310, Johor, Malaysia
| | - Victoria Nahum
- The Center for Nanoscience and Nanotechnology, Alex Grass Center for Drug Design and Synthesis and Cannabinoids Research, School of Pharmacy, Faculty of Medicine, Institute of Drug Research, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| |
Collapse
|
9
|
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: 3] [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
|
10
|
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: 24] [Impact Index Per Article: 8.0] [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
|
11
|
Collon K, Bell JA, Gallo MC, Chang SW, Bougioukli S, Sugiyama O, Tassey J, Hollis R, Heckmann N, Oakes DA, Longjohn DB, Evseenko D, Kohn DB, Lieberman JR. Influence of donor age and comorbidities on transduced human adipose-derived stem cell in vitro osteogenic potential. Gene Ther 2022; 30:369-376. [PMID: 36216880 PMCID: PMC10086075 DOI: 10.1038/s41434-022-00367-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 09/24/2022] [Accepted: 09/27/2022] [Indexed: 01/17/2023]
Abstract
Human adipose-derived mesenchymal stem cells (ASCs) transduced with a lentiviral vector system to express bone morphogenetic protein 2 (LV-BMP-2) have been shown to reliably heal bone defects in animal models. However, the influence of donor characteristics such as age, sex, race, and medical co-morbidities on ASC yield, growth and bone regenerative capacity, while critical to the successful clinical translation of stem cell-based therapies, are not well understood. Human ASCs isolated from the infrapatellar fat pads in 122 ASC donors were evaluated for cell growth characteristics; 44 underwent additional analyses to evaluate in vitro osteogenic potential, with and without LV-BMP-2 transduction. We found that while female donors demonstrated significantly higher cell yield and ASC growth rates, age, race, and the presence of co-morbid conditions were not associated with differences in proliferation. Donor demographics or the presence of comorbidities were not associated with differences in in vitro osteogenic potential or stem cell differentiation, except that transduced ASCs from healthy donors produced more BMP-2 at day 2. Overall, donor age, sex, race, and the presence of co-morbid conditions had a limited influence on cell yield, proliferation, self-renewal capacity, and osteogenic potential for non-transduced and transduced (LV-BMP-2) ASCs. These results suggest that ASCs are a promising resource for both autologous and allogeneic cell-based gene therapy applications.
Collapse
Affiliation(s)
- 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.
| | - 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
| | - 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
| | - Sofia Bougioukli
- 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
| | - Jade Tassey
- 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
| | - Nathanael Heckmann
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave,HMR 702, Los Angeles, CA, 90089, USA
| | - Daniel A Oakes
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave,HMR 702, Los Angeles, CA, 90089, USA
| | - Donald B Longjohn
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave,HMR 702, Los Angeles, CA, 90089, USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave,HMR 702, 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
|
12
|
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: 16] [Impact Index Per Article: 5.3] [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
|
13
|
Bougioukli S, Chateau M, Morales H, Vakhshori V, Sugiyama O, Oakes D, Longjohn D, Cannon P, Lieberman JR. Limited potential of AAV-mediated gene therapy in transducing human mesenchymal stem cells for bone repair applications. Gene Ther 2021; 28:729-739. [PMID: 32807899 DOI: 10.1038/s41434-020-0182-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 07/01/2020] [Accepted: 07/22/2020] [Indexed: 12/26/2022]
Abstract
Adeno-associated viral vectors (AAV) are unique in their ability to transduce a variety of both dividing and nondividing cells, with significantly lower risk of random genomic integration and with no known pathogenicity in humans, but their role in ex vivo regional gene therapy for bone repair has not been definitively established. The goal of this study was to test the ability of AAV vectors carrying the cDNA for BMP-2 to transduce human mesenchymal stem cells (MSCs), produce BMP-2, and induce osteogenesis in vitro as compared with lentiviral gene therapy with a two-step transcriptional amplification system lentiviral vector (LV-TSTA). To this end, we created two AAV vectors (serotypes 2 and 6) expressing the target transgene; eGFP or BMP-2. Transduction of human MSCs isolated from bone marrow (BMSCs) or adipose tissue (ASCs) with AAV2-eGFP and AAV6-eGFP led to low transduction efficiency (BMSCs: 3.57% and 8.82%, respectively, ASCs: 6.17 and 20.2%, respectively) and mean fluorescence intensity as seen with FACS analysis 7 days following transduction, even at MOIs as high as 106. In contrast, strong eGFP expression was detectable in all of the cell types post transduction with LV-TSTA-eGFP. Transduction with BMP-2 producing vectors led to minimal BMP-2 production in AAV-transduced cells 2 and 7 days following transduction. In addition, transduction of ASCs and BMSCs with AAV2-BMP-2 and AAV6-BMP-2 did not enhance their osteogenic potential as seen with an alizarin red assay. In contrast, the LV-TSTA-BMP-2-transduced cells were characterized by an abundant BMP-2 production and induction of the osteogenic phenotype in vitro (p < 0.001 vs. AAV2 and 6). Our results demonstrate that the AAV2 and AAV6 vectors cannot induce a significant transgene expression in human BMSCs and ASCs, even at MOIs as high as 106. The LV-TSTA vector is significantly superior in transducing human MSCs; thus this vector would be preferable when developing an ex vivo regional gene therapy strategy for clinical use in orthopedic surgery applications.
Collapse
Affiliation(s)
- Sofia Bougioukli
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Morgan Chateau
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Heidy Morales
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Venus Vakhshori
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daniel Oakes
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Donald Longjohn
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paula Cannon
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
14
|
De la Vega RE, Atasoy-Zeybek A, Panos JA, VAN Griensven M, Evans CH, Balmayor ER. Gene therapy for bone healing: lessons learned and new approaches. Transl Res 2021; 236:1-16. [PMID: 33964474 PMCID: PMC8976879 DOI: 10.1016/j.trsl.2021.04.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022]
Abstract
Although gene therapy has its conceptual origins in the treatment of Mendelian disorders, it has potential applications in regenerative medicine, including bone healing. Research into the use of gene therapy for bone healing began in the 1990s. Prior to this period, the highly osteogenic proteins bone morphogenetic protein (BMP)-2 and -7 were cloned, produced in their recombinant forms and approved for clinical use. Despite their promising osteogenic properties, the clinical usefulness of recombinant BMPs is hindered by delivery problems that necessitate their application in vastly supraphysiological amounts. This generates adverse side effects, some of them severe, and raises costs; moreover, the clinical efficacy of the recombinant proteins is modest. Gene delivery offers a potential strategy for overcoming these limitations. Our research has focused on delivering a cDNA encoding human BMP-2, because the recombinant protein is Food and Drug Administration approved and there is a large body of data on its effects in people with broken bones. However, there is also a sizeable literature describing experimental results obtained with other transgenes that may directly or indirectly promote bone formation. Data from experiments in small animal models confirm that intralesional delivery of BMP-2 cDNA is able to heal defects efficiently and safely while generating transient, local BMP-2 concentrations 2-3 log orders less than those needed by recombinant BMP-2. The next challenge is to translate this information into a clinically expedient technology for bone healing. Our present research focuses on the use of genetically modified, allografted cells and chemically modified messenger RNA.
Collapse
Affiliation(s)
- Rodolfo E De la Vega
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota; Musculoskeletal Gene Therapy Research Laboratory, Mayo Clinic, Rochester, Minnesota; cBITE, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Aysegul Atasoy-Zeybek
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota; Musculoskeletal Gene Therapy Research Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Joseph A Panos
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota; Musculoskeletal Gene Therapy Research Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Martijn VAN Griensven
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota; Musculoskeletal Gene Therapy Research Laboratory, Mayo Clinic, Rochester, Minnesota; cBITE, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Christopher H Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota; Musculoskeletal Gene Therapy Research Laboratory, Mayo Clinic, Rochester, Minnesota.
| | - Elizabeth R Balmayor
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, Minnesota; Musculoskeletal Gene Therapy Research Laboratory, Mayo Clinic, Rochester, Minnesota; IBE, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
15
|
Abstract
» Orthopaedics pioneered the expansion of gene therapy beyond its traditional scope of diseases that are caused by rare single-gene defects. Orthopaedic applications of gene therapy are most developed in the areas of arthritis and regenerative medicine, but several additional possibilities exist. » Invossa, an ex vivo gene therapeutic for osteoarthritis, was approved in South Korea in 2017, but its approval was retracted in 2019 and remains under appeal; a Phase-III clinical trial of Invossa has restarted in the U.S. » There are several additional clinical trials for osteoarthritis and rheumatoid arthritis that could lead to approved gene therapeutics for arthritis. » Bone-healing and cartilage repair are additional areas that are attracting considerable research; intervertebral disc degeneration and the healing of ligaments, tendons, and menisci are other applications of interest. Orthopaedic tumors, genetic diseases, and aseptic loosening are additional potential targets. » If successful, these endeavors will expand the scope of gene therapy from providing expensive medicines for a few patients to providing affordable medicines for many.
Collapse
|
16
|
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: 11] [Impact Index Per Article: 2.8] [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
|
17
|
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: 1.5] [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
|
18
|
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: 34] [Impact Index Per Article: 8.5] [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
|
19
|
Betz VM, Ren B, Betz OB, Jansson V, Müller PE. Osteoinduction within adipose tissue fragments by heterodimeric bone morphogenetic Proteins-2/6 and -2/7 versus homodimeric bone morphogenetic protein-2: Therapeutic implications for bone regeneration. J Gene Med 2021; 23:e3311. [PMID: 33527563 DOI: 10.1002/jgm.3311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Fragments of subcutaneous adipose tissue that have been genetically modified to express bone morphogenetic protein-2 (BMP-2) regenerate large segmental osseous lesions in rodents. Gene-activated adipose tissue can be implanted into osseous defects without prior cell extraction and cell culture. The present study aimed to explore whether the heterodimers BMP-2/6 or BMP-2/7 exceed the osteoinductive effect of BMP-2 on adipose tissue. METHODS In an in vitro tissue culture system, freshly harvested rat subcutaneous adipose tissue was cultivated in the presence of either BMP-2 or BMP-2/6 or BMP-2/7 at a high (200 ng/ml) and low (50 ng/ml) concentration. Gene expression analysis as well as histological and immunohistochemical methods were applied to test for osteoinduction. RESULTS A concentration of 200 ng/ml of homodimeric BMP-2 induced osteogenic differentiation most potently, showing more calcification and a higher expression level of bone markers than both concentrations of BMP-2/6 or -2/7. A concentration of 50 ng/ml of BMP-2 was a significantly stronger osteogenic inducer than both concentrations of BMP-2/6 and the low concentration of BMP-2/7. The most potent heterodimeric driver of osteoinduction was BMP-2/7 at a high concentration, demonstrating effects similar to those of BMP-2 at a low concentration. CONCLUSIONS Homodimeric BMP-2 evoked osteoinduction within adipose tissue more potently and at a lower concentration than heterodimeric BMP-2/6 or BMP-2/7. This result agrees well with the fact that it might be easier to translate adipose grafts activated by homodimeric BMP-2 clinically. Preclinical in vivo gene transfer studies are necessary to confirm the results of the present study.
Collapse
Affiliation(s)
- Volker M Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Munich, Campus Grosshadern, LMU, Munich, Bavaria, Germany
| | - Bin Ren
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Munich, Campus Grosshadern, LMU, Munich, Bavaria, Germany
| | - Oliver B Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Munich, Campus Grosshadern, LMU, Munich, Bavaria, Germany.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Volkmar Jansson
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Munich, Campus Grosshadern, LMU, Munich, Bavaria, Germany
| | - Peter E Müller
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Munich, Campus Grosshadern, LMU, Munich, Bavaria, Germany
| |
Collapse
|
20
|
Vakhshori V, Bougioukli S, Sugiyama O, Kang HP, Tang AH, Park SH, Lieberman JR. Ex vivo regional gene therapy with human adipose-derived stem cells for bone repair. Bone 2020; 138:115524. [PMID: 32622870 PMCID: PMC7423694 DOI: 10.1016/j.bone.2020.115524] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 06/20/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The treatment of complex bone loss scenarios remains challenging. This study evaluates the efficacy of ex vivo regional gene therapy using transduced human adipose-derived stem cells (ASCs) overexpressing bone morphogenetic protein-2 (BMP-2) to treat critical-sized bone defects. METHODS Critical-sized femoral defects created surgically in immunocompromised rats were treated with ASCs transduced with a lentivirus encoding BMP-2 (Group 1, n = 14), or green fluorescent protein (Group 2, n = 5), nontransduced ASCs (Group 3, n = 5), or rhBMP-2 (Group 4, n = 14). At 12 weeks, femurs were evaluated for quantity and quality of bone formation with plain radiographs, micro-computed tomography, histology/histomorphometry, and biomechanical strength testing. RESULTS Thirteen of 14 samples in Group 1 and all 14 samples in Group 4 showed radiographic healing, while no samples in either Groups 2 or 3 healed. Groups 1 and 4 had significantly higher radiographic scores (p < 0.001), bone volume fraction (BV/TV) (p < 0.001), and bone area fraction (BA/TA) than Groups 2 and 3 (p < 0.001). Radiographic scores, BV/TV, and BA/TA were not significantly different between Groups 1 and 4. No difference with regards to mean torque, rotation at failure, torsional stiffness, and energy to failure was seen between Groups 1 and 4. CONCLUSIONS Human ASCs modified to overexpress BMP-2 resulted in abundant bone formation, with the quality of bone comparable to that of rhBMP-2. This strategy represents a promising approach in the treatment of large bone defects in the clinical setting. CLINICAL RELEVANCE Large bone defects may require sustained protein production to induce an appropriate osteoinductive response. Ex vivo regional gene therapy using a lentiviral vector has the potential to be part of a comprehensive tissue engineering strategy for treating osseous defects.
Collapse
Affiliation(s)
- Venus Vakhshori
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, 1520 San Pablo Street, Suite 2000, Los Angeles, CA 90033, United States of America.
| | - Sofia Bougioukli
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, 1520 San Pablo Street, Suite 2000, Los Angeles, CA 90033, United States of America
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, 1520 San Pablo Street, Suite 2000, Los Angeles, CA 90033, United States of America
| | - Hyunwoo P Kang
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, 1520 San Pablo Street, Suite 2000, Los Angeles, CA 90033, United States of America
| | - Amy H Tang
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, 1520 San Pablo Street, Suite 2000, Los Angeles, CA 90033, United States of America
| | - Sang-Hyun Park
- Orthopaedic Institute for Children, J. Vernon Luck Sr. Orthopaedic Research Center, University of California, Los Angeles, 403 West Adams Boulevard, Los Angeles, CA 90007, United States of America
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, 1520 San Pablo Street, Suite 2000, Los Angeles, CA 90033, United States of America.
| |
Collapse
|
21
|
Virk MS, Luo W, Sikes KJ, Li J, Plaas A, Cole BJ. Gene expression profiling of progenitor cells isolated from rat rotator cuff musculotendinous junction. BMC Musculoskelet Disord 2020; 21:194. [PMID: 32222148 PMCID: PMC7102440 DOI: 10.1186/s12891-020-03190-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 03/05/2020] [Indexed: 01/08/2023] Open
Abstract
Background Rotator cuff tendon tears are typically degenerative and usually affect the region of tendon insertion on bone. The remnant torn tendon is degenerative and may not be an ideal source for progenitor cells for cell-based therapies. Therefore, the aim of this study was to determine if musculotendinous junction (MTJ), which is adjacent to tendon would be a viable alternate source of progenitor stem cells. We also sought to study the gene expression profile MTJ progenitors and compare it with progenitors isolated from RC tendon, RC muscle and other existing tissue sources (bone marrow, adipose tissue, and Achilles tendon). Methods Rotator cuff tendon (RCT), muscle (RCM), and RCMTJ as well as Achilles tendon (AT) tissues were harvested from healthy male Lewis rats and progenitor cultures were established from these tissues and also from bone marrow and adipose tissue. Quantitative RT-PCR was performed on RNA extracts from intact tissues and progenitor cells using a custom array for the mesenchymal stem cell (MSC) differentiation marker genes. The gene expression profile of MSC differentiation markers within four tissues types, six progenitor cells, and between tissue and their corresponding progenitors were compared. Results Progenitors cells can be isolated from rat rotator cuff musculotendinous tissue and their pattern of MSC gene expression was similar to the rotator cuff tendon progenitors for majority of the genes tested. However, there were significant differences between the MSC gene expression patterns of RCMTJ and RCM progenitors. Furthermore, there were differences in gene expression between the RCMTJ tissue and its progenitor cells with respect to MSC differentiation markers. The gene expression pattern of RCMTJ tissue was similar to RCM tissue with respect to markers of chondrogenesis, myogenesis, tenogenesis, and MSC specific markers. Conclusion We demonstrate that the musculotendinous junction contains distinct set of progenitor cells and their MSC gene expression pattern is similar to rotator cuff tendon progenitors. RCMTJ progenitors will be an attractive option for cell-based regenerative treatment of chronic rotator cuff tears.
Collapse
Affiliation(s)
- Mandeep S Virk
- Department of Orthopaedic Surgery, Division of Shoulder & Elbow, New York University Langone Orthopedic Hospital, 301 East 17th street, New York, 10003, NY, USA.
| | - Wei Luo
- Dept of Internal Medicine, Rush University Medical Center, 1735 W Harrison St Jelke 1302, Chicago, 60612, IL, USA
| | - Katie J Sikes
- Department of Clinical Sciences, Colorado State University, Translational Medicine Institute, Fort Collins, 80523, CO, USA
| | - Jun Li
- Dept of Internal Medicine, Rush University Medical Center, 1735 W Harrison St Jelke 1302, Chicago, 60612, IL, USA
| | - Anna Plaas
- Dept of Internal Medicine, Rush University Medical Center, 1735 W Harrison St Jelke 1302, Chicago, 60612, IL, USA
| | - Brian J Cole
- Department of Orthopaedic Surgery, Sports and Shoulder and Elbow Division, Midwest Orthopaedics at Rush University, 1611 West Harrison Suite 300, Chicago, IL, USA
| |
Collapse
|
22
|
Park SY, Kim KH, Kim S, Rhee SH, Yeo IS, Heo SJ, Lee YM, Seol YJ. Comparison of experimental peri-implantitis models after application of ex vivo BMP2 gene therapy using periodontal ligament stem cells. Sci Rep 2020; 10:3590. [PMID: 32108172 PMCID: PMC7046768 DOI: 10.1038/s41598-020-60341-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/11/2020] [Indexed: 11/21/2022] Open
Abstract
Peri-implantitis is an inflammatory disease that results in bone destruction around dental implants. A preclinical study using beagle models is frequently performed prior to clinical application in dentistry. Previously, we proposed an immediate peri-implantitis experimental model with a shorter experimental duration and less expense than the conventional experimental model. However, the differences in the regenerative outcomes between the immediate and conventional models were not fully revealed. In this study, we aimed to compare the regenerative outcomes between both models when ex vivo BMP2 gene therapy using autologous periodontal ligament stem cells (B2/PDLSCs) was applied to peri-implantitis defects. The results showed that the defect depths were significantly different between both models. New bone formation occurred in both models, but there were significant differences between the models. More than 70% of the defects were filled with newly formed bone in the conventional model, whereas 30-40% of the defects were filled in the immediate model. However, after adjustment for the differences in the defect depths between the models, the statistically significant differences in the regenerative outcomes between the models were lost. In conclusion, the inferior regenerative outcome of an immediate peri-implantitis model at B2/PDLSCs transplantation resulted from the defect depths, not the model itself.
Collapse
Affiliation(s)
- Shin-Young Park
- Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Korea
- Department of Dentistry and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Korea
| | - Kyoung-Hwa Kim
- Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Korea
| | - Sungtae Kim
- Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Korea
| | - Sang-Hoon Rhee
- Department of Dental Biomaterials Science, School of Dentistry, Seoul National University, Seoul, 03080, Korea
| | - In-Sung Yeo
- Department of Prosthodontics and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Korea
| | - Seong-Joo Heo
- Department of Prosthodontics and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Korea
| | - Yong-Moo Lee
- Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Korea
| | - Yang-Jo Seol
- Department of Periodontology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Korea.
| |
Collapse
|
23
|
Kong L, Zuo R, Wang M, Wang W, Xu J, Chai Y, Guan J, Kang Q. Silencing MicroRNA-137-3p, which Targets RUNX2 and CXCL12 Prevents Steroid-induced Osteonecrosis of the Femoral Head by Facilitating Osteogenesis and Angiogenesis. Int J Biol Sci 2020; 16:655-670. [PMID: 32025213 PMCID: PMC6990928 DOI: 10.7150/ijbs.38713] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/14/2019] [Indexed: 12/13/2022] Open
Abstract
The main pathogenesis of steroid-induced osteonecrosis of the femoral head (SONFH) includes decreased osteogenic capacity of bone marrow-derived mesenchymal stem cells (BMSCs) and damaged blood supply to the femoral head. MicroRNAs (miRNAs) have been shown to play prominent roles in SONFH development. However, there is no report that a specific miRNA targeting two genes in two different pathogenic pathways has been applied to this disease. The present study investigated the effects of transplantation of miR-137-3p-silenced BMSCs on the prevention and early treatment of SONFH. First, western blotting and dual luciferase assays were employed to verify that miR-137-3p directly targets Runx2 and CXCL12. Then, silencing of miR-137-3p was found to facilitate osteogenic differentiation of BMSCs, which was confirmed by alkaline phosphatase (ALP) staining, alizarin red staining and qRT-PCR. Silencing of miR-137-3p also promoted angiogenesis by human umbilical vein endothelial cells (HUVECs) in the presence or absence of glucocorticoids. Thereafter, overexpression of Runx2 and CXCL12 without the 3′ untranslated region (3′UTR) partially rescued the effects of miR-137-3p on osteogenesis and angiogenesis, respectively. This finding further supported the hypothesis that miR-137-3p exerts its functions partly by regulating the genes, Runx2 and CXCL12. We also demonstrated that SONFH was partially prevented by transplantation of miR-137-3p-silenced BMSCs into a rat model. Micro-CT and histology showed that the transplantation of miR-137-3p-silenced BMSCs significantly improved bone regeneration. Additionally, the results of enzyme-linked immunosorbent assays (ELISA) and flow cytometry suggested that stromal cell-derived factor-1α (SDF-1α) and endothelial progenitor cells (EPCs) participated in the process of vascular repair. Taken together, these findings show that silencing of miR-137-3p directly targets the genes, Runx2 and CXCL12, which can play critical roles in SONFH repair by facilitating osteogenic differentiation and mobilizing EPCs.
Collapse
Affiliation(s)
- Lingchi Kong
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Rongtai Zuo
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Mengwei Wang
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Wenbo Wang
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Jia Xu
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yimin Chai
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Junjie Guan
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Qinglin Kang
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
24
|
Bougioukli S, Alluri R, Pannell W, Sugiyama O, Vega A, Tang A, Skorka T, Park SH, Oakes D, Lieberman JR. Ex vivo gene therapy using human bone marrow cells overexpressing BMP-2: "Next-day" gene therapy versus standard "two-step" approach. Bone 2019; 128:115032. [PMID: 31398502 PMCID: PMC6813891 DOI: 10.1016/j.bone.2019.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 01/13/2023]
Abstract
Traditionally, ex vivo gene therapy involves a two-step approach, with culture expansion of cells prior to transduction and implantation. We have tried to simplify this strategy and eliminate the time and cost associated with culture expansion, by introducing "next-day" regional gene therapy using human bone marrow cells. The purpose of this study was to determine whether a lentiviral vector (LV) carrying the cDNA for BMP-2 can transduce freshly isolated human BM cells, leading to abundant BMP production and bone formation in vivo, and evaluate the in vivo osteoinductive potential of "next-day" gene therapy and the standard "two-step" tissue culture expansion approach. To this end, human bone marrow cells (HBMC) from patients undergoing total hip arthroplasty were harvested, transduced with a BMP-2-expressing LV either overnight ("next day" gene therapy; ND) or after culture expansion (cultured "two-step" approach; C) and then implanted into a rat critical-sized femoral defect. The animals were randomly assigned to one of the following groups: I; ND-HBMC transduced with LV-TSTA BMP-2, II; ND-HBMC transduced with LV-TSTA GFP, III; non-transduced ND-HBMC; IV; C-HBMC transduced with LV-TSTA BMP-2, V; C-HBMC transduced with LV-TSTA-GFP, VI; non-transduced C-HBMC. Treatment with either "next-day" or cultured HBMC demonstrated a significant increase in new bone formation compared with all negative control groups as seen in plain radiographs, microCT and histologic/histomorphometric analysis. At 12 weeks post-op, complete defect union on plain X-rays occurred in 7/14 animals in the ND-HBMC/BMP-2 group and 12/14 in the C-HBMC/BMP-2 treated rats. The two-step approach was associated with more consistent results, a higher union rate, and superiority with regards to all of the studied bone healing parameters. In this study we demonstrate proof of concept that BMP-2-transduced human bone marrow cells can be used to enhance bone healing in segmental bone defects, and that regional gene therapy using lentiviral transduction has the osteoinductive potential to heal large bone defects in clinical settings.
Collapse
Affiliation(s)
- Sofia Bougioukli
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ram Alluri
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - William Pannell
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andrew Vega
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Amy Tang
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Sang Hyun Park
- Orthopaedic Institute for Children, J. Vernon Luck. Sr., Orthopaedic Research Center, Los Angeles, CA, USA
| | - Daniel Oakes
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
25
|
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: 61] [Impact Index Per Article: 10.2] [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
|
26
|
Kelly DC, Raftery RM, Curtin CM, O'Driscoll CM, O'Brien FJ. Scaffold-Based Delivery of Nucleic Acid Therapeutics for Enhanced Bone and Cartilage Repair. J Orthop Res 2019; 37:1671-1680. [PMID: 31042304 DOI: 10.1002/jor.24321] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/09/2019] [Indexed: 02/04/2023]
Abstract
Recent advances in tissue engineering have made progress toward the development of biomaterials capable of the delivery of growth factors, such as bone morphogenetic proteins, in order to promote enhanced tissue repair. However, controlling the release of these growth factors on demand and within the desired localized area is a significant challenge and the associated high costs and side effects of uncontrolled delivery have proven increasingly problematic in clinical orthopedics. Gene therapy may be a valuable tool to avoid the limitations of local delivery of growth factors. Following a series of setbacks in the 1990s, the field of gene therapy is now seeing improvements in safety and efficacy resulting in substantial clinical progress and a resurgence in confidence. Biomaterial scaffold-mediated gene therapy provides a template for cell infiltration and tissue formation while promoting transfection of cells to engineer therapeutic proteins in a sustained but ultimately transient fashion. Additionally, scaffold-mediated delivery of RNA-based therapeutics can silence specific genes associated with orthopedic pathological states. This review will provide an overview of the current state-of-the-art in the field of gene-activated scaffolds and their use within orthopedic tissue engineering applications. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1671-1680, 2019.
Collapse
Affiliation(s)
- Domhnall C Kelly
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Rosanne M Raftery
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Caroline M Curtin
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Caitriona M O'Driscoll
- Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland.,Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.,Trinity Centre of Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.,Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| |
Collapse
|
27
|
Loozen LD, Kruyt MC, Kragten AHM, Schoenfeldt T, Croes M, Oner CF, Dhert WJA, Alblas J. BMP-2 gene delivery in cell-loaded and cell-free constructs for bone regeneration. PLoS One 2019; 14:e0220028. [PMID: 31365542 PMCID: PMC6668905 DOI: 10.1371/journal.pone.0220028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 07/08/2019] [Indexed: 11/18/2022] Open
Abstract
To induce osteogenicity in bone graft substitutes, plasmid-based expression of BMP-2 (pBMP-2) has been successfully applied in gene activated matrices based on alginate polymer constructs. Here, we investigated whether cell seeding is necessary for non-viral BMP-2 gene expression in vivo. Furthermore, to gain insight in the role of BMP-producing cells, we compared inclusion of bone progenitor cells with non-osteogenic target cells in gene delivery constructs. Plasmid DNA encoding GFP (pGFP) was used to trace transfection of host tissue cells and seeded cells in a rat model. Transgene expression was followed in both cell-free alginate-ceramic constructs as well as constructs seeded with syngeneic fibroblasts or multipotent mesenchymal stromal cells (MSCs). Titration of pGFP revealed that the highest pGFP dose resulted in frequent presence of positive host cells in the constructs. Both cell-loaded groups were associated with transgene expression, most effectively in the MSC-loaded constructs. Subsequently, we investigated effectiveness of cell-free and cell-loaded alginate-ceramic constructs with pBMP-2 to induce bone formation. Local BMP-2 production was found in all groups containing BMP-2 plasmid DNA, and was most pronounced in the groups with MSCs transfected with high concentration pBMP-2. Bone formation was only apparent in the recombinant protein BMP-2 group. In conclusion, we show that non-viral gene delivery of BMP-2 is a potentially effective way to induce transgene expression in vivo, both in cell-seeded as well as cell-free conditions. However, alginate-based gene delivery of BMP-2 to host cells or seeded cells did not result in protein levels adequate for bone formation in this setting, calling for more reliable scaffold compatible transfection methods.
Collapse
Affiliation(s)
- Loek D. Loozen
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Moyo C. Kruyt
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Ted Schoenfeldt
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michiel Croes
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cumhur F. Oner
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Wouter J. A. Dhert
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jacqueline Alblas
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
28
|
Alluri R, Song X, Bougioukli S, Pannell W, Vakhshori V, Sugiyama O, Tang A, Park SH, Chen Y, Lieberman JR. Regional gene therapy with 3D printed scaffolds to heal critical sized bone defects in a rat model. J Biomed Mater Res A 2019; 107:2174-2182. [PMID: 31112357 DOI: 10.1002/jbm.a.36727] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/29/2019] [Accepted: 05/20/2019] [Indexed: 01/23/2023]
Abstract
The objective of the present study was to assess the ability of transduced rat bone marrow cells (RBMCs) that overexpress BMP-2 loaded on a three-dimensionally (3D) printed scaffold to heal a critical sized rat femoral defect. Tricalcium phosphate (TCP) scaffolds were 3D printed to fit a critical sized rat femoral defect. The RBMCs were transduced with a lentiviral (LV) vector expressing BMP-2 or GFP. The rats were randomized into the following treatment groups: (1) RBMC/LV-BMP-2 + TCP, (2) RBMC/LV-GFP + TCP, (3) nontransduced RBMCs + TCP, (4) TCP scaffold alone. The animals were euthanized at 12 weeks and evaluated with plain radiographs, microcomputed tomography (micro-CT), histology, histomorphometry, and biomechanically. Each LV-BMP-2 + TCP treated specimen demonstrated complete healing of the femoral defect on plain radiographs and micro-CT. No femurs healed in the control groups. Micro-CT demonstrated that LV-BMP-2 + TCP treated femoral defects formed 197% more bone volume compared to control groups (p < 0.05). Histologic analysis demonstrated bone formation across the TCP scaffold, uniting the femoral defect on both ends in the LV-BMP-2 + TCP treated specimens. Biomechanical assessment demonstrated similar stiffness (p = 0.863), but lower total energy to failure, peak torque, and peak displacement (p < 0.001) of the femurs treated with LV-BMP-2 + TCP when compared to the contralateral control femur. Regional gene therapy induced overexpression of BMP-2 via transduced RBMCs combined with an osteoconductive 3D printed TCP scaffold can heal a critically sized femoral defect in an animal model. The combination of regional gene therapy and 3D printed osteoconductive scaffolds has significant clinical potential to enhance bone regeneration.
Collapse
Affiliation(s)
- Ram Alluri
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Xuan Song
- Department of Industrial and Systems Engineering, The University of Iowa, Iowa City, Iowa
| | - Sofia Bougioukli
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - William Pannell
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Venus Vakhshori
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Amy Tang
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Sang-Hyun Park
- Orthopaedic Institute for Children, J. Vernon Luck. Sr. Orthopaedic Research Center, Los Angeles, California
| | - Yong Chen
- Viterbi School of Engineering, University of Southern California, Los Angeles, California
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|
29
|
Ball AN, Phillips JN, McIlwraith CW, Kawcak CE, Samulski RJ, Goodrich LR. Genetic modification of scAAV-equine-BMP-2 transduced bone-marrow-derived mesenchymal stem cells before and after cryopreservation: An "off-the-shelf" option for fracture repair. J Orthop Res 2019; 37:1310-1317. [PMID: 30578639 PMCID: PMC8366205 DOI: 10.1002/jor.24209] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/12/2018] [Indexed: 02/04/2023]
Abstract
Optimizing the environment of complex bone healing and improving treatment of catastrophic bone fractures and segmental bone defects remains an unmet clinical need both human and equine veterinary medical orthopaedics. The objective of this study was to determine whether scAAV-equine-BMP-2 transduced cells would induce osteogenesis in equine bone marrow derived mesenchymal stem cells (BMDMSCs) in vitro, and if these cells could be cryopreserved in an effort to osteogenically prime them as an "off-the-shelf" gene therapeutic approach for fracture repair. Our study found that transgene expression is altered by cell expansion, as would be expected by a transduction resulting in episomal transgene expression, and that osteoinductive levels could still be achieved 5 days after recovery, and protein expression would continue up to 14 days after transduction. This is the first evidence that cryopreservation of genetically modified BMDMSCs would not alter the osteoinductive potential or clinical use of allogeneic donor cells in cases of equine fracture repair. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1310-1317, 2019.
Collapse
Affiliation(s)
- Alyssa N. Ball
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA,Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Jennifer N. Phillips
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA,Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - C. Wayne McIlwraith
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA,Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Christopher E. Kawcak
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA,Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Richard J. Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Laurie R. Goodrich
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA,Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
30
|
Bougioukli S, Saitta B, Sugiyama O, Tang AH, Elphingstone J, Evseenko D, Lieberman JR. Lentiviral Gene Therapy for Bone Repair Using Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells. Hum Gene Ther 2019; 30:906-917. [PMID: 30773946 DOI: 10.1089/hum.2018.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Umbilical cord blood (UCB) has been increasingly explored as an alternative source of stem cells for use in regenerative medicine due to several advantages over other stem-cell sources, including the need for less stringent human leukocyte antigen matching. Combined with an osteoinductive signal, UCB-derived mesenchymal stem cells (MSCs) could revolutionize the treatment of challenging bone defects. This study aimed to develop an ex vivo regional gene-therapy strategy using BMP-2-transduced allogeneic UCB-MSCs to promote bone repair. To this end, human UCB-MSCs were transduced with a lentiviral vector carrying the cDNA for BMP-2 (LV-BMP-2). In vitro assays to determine the UCB-MSC osteogenic potential and BMP-2 production were followed by in vivo implantation of LV-BMP-2-transduced UCB-MSCs in a mouse hind-limb muscle pouch. Non-transduced and LV-GFP-transduced UCB-MSCs were used as controls. Transduction with LV-BMP-2 was associated with abundant BMP-2 production and induction of osteogenic differentiation in vitro. Implantation of BMP-2-transduced UCB-MSCs led to robust heterotopic bone formation 4 weeks postoperatively, as seen on radiographs and histology. These results, along with the fact that UCB-MSCs can be easily collected with no donor-site morbidity and low immunogenicity, suggest that UCB might be a preferable allogeneic source of MSCs to develop an ex vivo gene-therapy approach to treat difficult bone-repair scenarios.
Collapse
Affiliation(s)
- Sofia Bougioukli
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Biagio Saitta
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Amy H Tang
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Joseph Elphingstone
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
31
|
Bougioukli S, Vakhshori V, Ortega B, Sugiyama O, Lieberman J. Regulated ex vivo regional gene therapy for bone repair using an inducible caspase-9 suicide gene system. Gene Ther 2019; 26:230-239. [PMID: 30962534 DOI: 10.1038/s41434-019-0069-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/04/2019] [Accepted: 03/01/2019] [Indexed: 12/25/2022]
Abstract
In order to adapt ex vivo regional gene therapy for clinical applications in orthopaedic surgery, safety issues must be considered. In this study we developed a suicide approach using a dual gene expression two step transcriptional amplification lentiviral vector (LV-TSTA) encoding BMP-2 and an inducible caspase 9 (iC9) system that selectively induces apoptosis upon activation with a chemical inducer of dimerization (CID). Transduction of rat bone marrow stromal cells (RBMSCs) with LV-TSTA-iC9/BMP-2 led to abundant BMP-2 production (90.3 ± 7.9 ng/24 h/106 cells) in vitro and stimulated bone formation in a mouse muscle pouch in the absence of CID. Moreover it was shown that CID could be used to selectively induce apoptosis in iC9-transduced cells both in vitro and in vivo. Double exposure to serial dilutions of CID decreased in vitro production of BMP-2 by 85-87% and Luc activity by 97-99% in iC9/BMP-2 or iC9/Luc-transduced cells respectively. Early administration of CID (Days 0-1 post-op) in mice implanted with iC9/BMP-2-transduced RBMSCs was effective in blocking bone formation, indicating that CID was toxic to the transduced cells. In iC9/Luc-implanted mice, late administration of two doses of CID (Days 27-28 post-op) significantly reduced the luciferase signal. The current study provides proof of concept for the potential clinical application of regulated gene therapy to promote bone repair.
Collapse
Affiliation(s)
- Sofia Bougioukli
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Venus Vakhshori
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brandon Ortega
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jay Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
32
|
Koons GL, Mikos AG. Progress in three-dimensional printing with growth factors. J Control Release 2019; 295:50-59. [PMID: 30579982 PMCID: PMC6358495 DOI: 10.1016/j.jconrel.2018.12.035] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 11/06/2018] [Accepted: 12/19/2018] [Indexed: 12/19/2022]
Abstract
Incorporation of growth factors in biomedical constructs can encourage cellular activities necessary for tissue regeneration within an implant system. Three-dimensional printing offers a capacity for spatial dictation and dosage control of incorporated growth factors which promises to minimize complications from the supraphysiologic doses and burst release involved in current growth factor delivery systems. Successful implementation of three-dimensional printing with growth factors requires preservation of the bioactivity of printed growth factors, spatial localization of growth factors within the construct architecture during printing, and controlled release of growth factors after printing. This review describes demonstrated approaches for addressing each of these goals, including direct inclusion of growth factors with the biomaterial during printing, or intermediary encapsulation of growth factors in delivery vehicles such as microparticles or nanoparticles.
Collapse
Affiliation(s)
- Gerry L Koons
- Department of Bioengineering, Rice University, Houston, TX, USA; Center for Engineering Complex Tissues, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA.
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX, USA; Center for Engineering Complex Tissues, USA.
| |
Collapse
|
33
|
Ren B, Betz VM, Thirion C, Salomon M, Klar RM, Jansson V, Müller PE, Betz OB. Gene activated adipose tissue fragments as advanced autologous biomaterials for bone regeneration: osteogenic differentiation within the tissue and implications for clinical translation. Sci Rep 2019; 9:224. [PMID: 30659209 PMCID: PMC6338750 DOI: 10.1038/s41598-018-36283-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/26/2018] [Indexed: 02/06/2023] Open
Abstract
Cost-effective, expedited approaches for bone regeneration are urgently needed in an ageing population. Bone Morphogenetic Proteins (BMPs) stimulate osteogenesis but their efficacy is impeded by their short half-life. Delivery by genetically modified cells can overcome this problem. However, cell isolation and propagation represent significant obstacles for the translation into the clinic. Instead, complete gene activated fragments of adipose tissue hold great potential for bone repair. Here, using an in-vitro culture system, we investigated whether adenoviral transduction with human BMP-2 can promote osteogenic differentiation within adipose tissue fragments. Osteoinduction in adipose tissue fragments was evaluated by quantitative reverse transcriptase polymerase chain reaction, immunohistology and histomorphometry. BMP-2 transduced adipose tissue synthesized BMP-2 protein over 30 days peaking by day six, which significantly promoted osteogenic differentiation as indicated by increased calcium depositions, up-regulation of bone marker genes, and bone-related protein expression. Our results demonstrate that cells within adipose tissue fragments can differentiate osteogenically after BMP-2 transduction of cells on the surface of the adipose tissue. BMP-2 gene activated adipose tissue represents an advanced osteo-regenerative biomaterial that can actively contribute to osteogenesis and potentially enable the development of a novel, cost-effective, one-step surgical approach to bone repair without the need for cell isolation.
Collapse
Affiliation(s)
- Bin Ren
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Marchioninistr 15, 81377, Munich, Germany.
| | - Volker M Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Marchioninistr 15, 81377, Munich, Germany
| | - Christian Thirion
- Sirion Biotech GmbH, Am Klopferspitz 19, 82152, Martinsried, Germany
| | - Michael Salomon
- Sirion Biotech GmbH, Am Klopferspitz 19, 82152, Martinsried, Germany
| | - Roland M Klar
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Marchioninistr 15, 81377, Munich, Germany
| | - Volkmar Jansson
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Marchioninistr 15, 81377, Munich, Germany
| | - Peter E Müller
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Marchioninistr 15, 81377, Munich, Germany
| | - Oliver B Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Marchioninistr 15, 81377, Munich, Germany
| |
Collapse
|
34
|
Osteoinduction within BMP-2 transduced muscle tissue fragments with and without a fascia layer: implications for bone tissue engineering. Gene Ther 2018; 26:16-28. [PMID: 30368527 DOI: 10.1038/s41434-018-0047-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 08/27/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022]
Abstract
Bone can be engineered in vivo by implantation of gene-activated muscle tissue fragments. This expedited approach may be further improved by use of muscle tissue with attached fascia. The aim of this in vitro study was to provide an in depth comparison of the osteogenic differentiation capacity of muscle alone and muscle with fascia after BMP-2 transduction. Skeletal muscle tissue from rats was cut into pieces with and without a fascia layer on the surface. Adenoviral BMP-2 or GFP vectors were used for transduction. Osteogenic differentiation within the tissue fragments was evaluated and compared by qRT-PCR, alizarin red S staining, histomorphometry and immunohistology. Transduction efficiency and level of transgene expression were higher for muscle with fascia than muscle alone. Transduction with BMP-2 led to a significant upregulation of bone marker genes, proteins, and calcium deposition in both groups. Interestingly, histological evaluation revealed that osteoinduction did not occur within the fascia layer itself. The upregulation of bone marker genes in muscle with fascia was significantly lower after 2 weeks but similar after 4 weeks of in vitro culture in comparison to muscle alone. The fascia layer led to higher transduction efficiency and enhanced BMP-2 expression. Despite fascia's lower capacity for osteogenic differentiation, muscle implants may benefit from the fascia layer by the improved ability to deliver BMP-2. The presented data may contribute to the development of a novel, cost-effective, single-surgery bone engineering technology and encourage the evaluation of the osteoregenerative potential of muscle with fascia in an animal model.
Collapse
|
35
|
Sharmin F, O'Sullivan M, Malinowski S, Lieberman JR, Khan Y. Large scale segmental bone defect healing through the combined delivery of VEGF and BMP‐2 from biofunctionalized cortical allografts. J Biomed Mater Res B Appl Biomater 2018; 107:1002-1010. [DOI: 10.1002/jbm.b.34193] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/18/2017] [Accepted: 08/22/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Farzana Sharmin
- Department of Materials Science and EngineeringUniversity of Connecticut Storrs Connecticut
- Institute for Regenerative EngineeringUConn Health Farmington Connecticut
| | | | - Seth Malinowski
- Department of Biomedical EngineeringUniversity of Connecticut Storrs Connecticut
| | - Jay R. Lieberman
- Department of Orthopedic SurgeryKeck School of Medicine of the University of Southern California California Los Angeles
| | - Yusuf Khan
- Department of Materials Science and EngineeringUniversity of Connecticut Storrs Connecticut
- Institute for Regenerative EngineeringUConn Health Farmington Connecticut
- Department of Orthopaedic SurgeryUConn Health Farmington Connecticut
- Department of Biomedical EngineeringUniversity of Connecticut Storrs Connecticut
- UConn Musculoskeletal Institute Farmington Connecticut
| |
Collapse
|
36
|
Betz VM, Ren B, Messmer C, Jansson V, Betz OB, Müller PE. Bone morphogenetic protein-2 is a stronger inducer of osteogenesis within muscle tissue than heterodimeric bone morphogenetic protein-2/6 and -2/7: Implications for expedited gene-enhanced bone repair. J Gene Med 2018; 20:e3042. [DOI: 10.1002/jgm.3042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 11/09/2022] Open
Affiliation(s)
- Volker M. Betz
- Department of Gene Therapy; University of Ulm; Ulm Germany
- Center for Rehabilitation; RKU - University and Rehabilitation Hospitals Ulm; Ulm Germany
| | - Bin Ren
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation; University Hospital Grosshadern, Ludwig-Maximilians-University Munich; Munich Germany
| | - Carolin Messmer
- Center for Rehabilitation; RKU - University and Rehabilitation Hospitals Ulm; Ulm Germany
| | - Volkmar Jansson
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation; University Hospital Grosshadern, Ludwig-Maximilians-University Munich; Munich Germany
| | - Oliver B. Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation; University Hospital Grosshadern, Ludwig-Maximilians-University Munich; Munich Germany
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA USA
| | - Peter E. Müller
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation; University Hospital Grosshadern, Ludwig-Maximilians-University Munich; Munich Germany
| |
Collapse
|
37
|
Loozen LD, Kruyt MC, Vandersteen A, Kragten AHM, Croes M, Öner FC, Alblas J. Osteoinduction by Ex Vivo Nonviral Bone Morphogenetic Protein Gene Delivery Is Independent of Cell Type. Tissue Eng Part A 2018; 24:1423-1431. [PMID: 29766760 DOI: 10.1089/ten.tea.2018.0032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Ex vivo nonviral gene delivery of bone inductive factors has the potential to heal bone defects. Due to their inherent role in new bone formation, multipotent stromal cells (MSCs) have been studied as the primary target cell for gene delivery in a preclinical setting. The relative contribution of autocrine and paracrine mechanisms, and the need of osteogenic cells, remains unclear. This study investigates the contribution of MSCs as producer of transgenic bone morphogenetic proteins (BMPs) and to what extent the seeded MSCs participate in actual osteogenesis. Rat-derived MSCs or fibroblasts (FBs) were cotransfected with pBMP-2 and pBMP-6 or pBMP-7 via nucleofection. The bioactivity of BMP products was shown through in vitro osteogenic differentiation assays. To investigate their role in new bone formation, transfected cells were seeded on ceramic scaffolds and implanted subcutaneously in rats. Bone formation was assessed by histomorphometry after 8 weeks. As a proof of principle, we also investigated the suitability of bone marrow-derived mononuclear cells and the stromal vascular fraction isolated from adipose tissue for a one-stage gene delivery strategy. Bone formation was induced in all conditions containing cells overexpressing BMP heterodimers. Constructs seeded with FBs transfected with BMP-2/6 and MSCs transfected with BMP-2/6 showed comparable bone volumes, both significantly higher than controls. Single-stage gene delivery proved possible and resulted in some bone formation. We conclude that bone formation as a result of ex vivo BMP gene delivery can be achieved even without direct osteogenic potential of the transfected cell type, suggesting that transfected cells mainly function as a production facility for osteoinductive proteins. In addition, single-stage transfection and reimplantation of cells appeared feasible, thus facilitating future clinical translation of the method.
Collapse
Affiliation(s)
- Loek D Loozen
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Moyo C Kruyt
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Angela Vandersteen
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Angela H M Kragten
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Michiel Croes
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - F Cumhur Öner
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Jacqueline Alblas
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| |
Collapse
|
38
|
In vitro evaluation of a lentiviral two-step transcriptional amplification system using GAL4FF transactivator for gene therapy applications in bone repair. Gene Ther 2018; 25:260-268. [PMID: 29907876 DOI: 10.1038/s41434-018-0024-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/02/2018] [Accepted: 05/23/2018] [Indexed: 01/20/2023]
Abstract
In this study, we developed a lentiviral two-step transcriptional amplification (TSTA) system expressing bone morphogenetic protein-2 (BMP-2) under the control of a GAL4FF transactivator to enhance gene expression and limit toxicity for bone repair applications. To this end human MSCs, isolated from bone marrow or adipose tissue, were transduced overnight with a LV-TSTA system (GAL4FF or GAL4vp16) expressing BMP-2 or GFP and evaluated in vitro for transduction efficiency, mean fluorescence intensity, cell viability, and BMP-2 production. FACS analysis of GFP-transduced MSCs confirmed successful transduction with the GAL4FF+GFP vector. Moreover, ELISA demonstrated abundant BMP-2 production by GAL4FF+BMP2-transduced human MSCs over a period of 8 weeks, with minimal cytotoxicity at all time points. Compared to GAL4vp16, GAL4FF was superior with respect to BMP production at 1, 2, 4, 6, and 8 weeks in BMSCs. In ASCs, GAL4FF was still associated with higher BMP-2 production at weeks 2-8, but this difference was not as prominent as in BMSCs. To our knowledge, this is the first report of GAL4FF-mediated BMP-2 production by human BMSCs and ASCs. Compared to the standard GAL4vp16TSTA vector, GAL4FF was associated with lower cytotoxicity and higher in vitro gene expression in both BMSCs and ASCs.
Collapse
|
39
|
Ball AN, Donahue SW, Wojda SJ, McIlwraith CW, Kawcak CE, Ehrhart N, Goodrich LR. The challenges of promoting osteogenesis in segmental bone defects and osteoporosis. J Orthop Res 2018; 36:1559-1572. [PMID: 29280510 PMCID: PMC8354209 DOI: 10.1002/jor.23845] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 12/04/2017] [Indexed: 02/04/2023]
Abstract
Conventional clinical management of complex bone healing scenarios continues to result in 5-10% of fractures forming non-unions. Additionally, the aging population and prevalence of osteoporosis-related fractures necessitate the further exploration of novel ways to augment osteogenesis in this special population. This review focuses on the current clinical modalities available, and the ongoing clinical and pre-clinical research to promote osteogenesis in segmental bone defects, delayed unions, and osteoporosis. In summary, animal models of fracture repair are often small animals as historically significant large animal models, like the dog, continue to gain favor as companion animals. Small rodents have well-documented limitations in comparing to fracture repair in humans, and few similarities exist. Study design, number of studies, and availability of funding continue to limit large animal studies. Osteoinduction with rhBMP-2 results in robust bone formation, although long-term quality is scrutinized due to poor bone mineral quality. PTH 1-34 is the only FDA approved osteo-anabolic treatment to prevent osteoporotic fractures. Limited to 2 years of clinical use, PTH 1-34 has further been plagued by dose-related ambiguities and inconsistent results when applied to pathologic fractures in systematic human clinical studies. There is limited animal data of PTH 1-34 applied locally to bone defects. Gene therapy continues to gain popularity among researchers to augment bone healing. Non-integrating viral vectors and targeted apoptosis of genetically modified therapeutic cells is an ongoing area of research. Finally, progenitor cell therapies and the content variation of patient-side treatments (e.g., PRP and BMAC) are being studied. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1559-1572, 2018.
Collapse
Affiliation(s)
- Alyssa N. Ball
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, 1678 Campus Delivery, Fort Collins, Colorado 80523-1678
| | - Seth W. Donahue
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, 1678 Campus Delivery, Fort Collins, Colorado 80523-1678,,Department of Mechanical Engineering, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| | - Samantha J. Wojda
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, 1678 Campus Delivery, Fort Collins, Colorado 80523-1678,,Department of Mechanical Engineering, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| | - C. Wayne McIlwraith
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, 1678 Campus Delivery, Fort Collins, Colorado 80523-1678
| | - Christopher E. Kawcak
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, 1678 Campus Delivery, Fort Collins, Colorado 80523-1678
| | - Nicole Ehrhart
- Department of Clinical Sciences, Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado
| | - Laurie R. Goodrich
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, 1678 Campus Delivery, Fort Collins, Colorado 80523-1678
| |
Collapse
|
40
|
Betz VM, Kochanek S, Rammelt S, Müller PE, Betz OB, Messmer C. Recent advances in gene-enhanced bone tissue engineering. J Gene Med 2018; 20:e3018. [PMID: 29601661 DOI: 10.1002/jgm.3018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/18/2018] [Accepted: 03/18/2018] [Indexed: 12/13/2022] Open
Abstract
The loss of bone tissue represents a critical clinical condition that is frequently faced by surgeons. Substantial progress has been made in the area of bone research, providing insight into the biology of bone under physiological and pathological conditions, as well as tools for the stimulation of bone regeneration. The present review discusses recent advances in the field of gene-enhanced bone tissue engineering. Gene transfer strategies have emerged as highly effective tissue engineering approaches for supporting the repair of the musculoskeletal system. By contrast to treatment with recombinant proteins, genetically engineered cells can release growth factors at the site of injury over extended periods of time. Of particular interest are the expedited technologies that can be applied during a single surgical procedure in a cost-effective manner, allowing translation from bench to bedside. Several promising methods based on the intra-operative genetic manipulation of autologous cells or tissue fragments have been developed in preclinical studies. Moreover, gene therapy for bone regeneration has entered the clinical stage with clinical trials for the repair of alveolar bone. Current trends in gene-enhanced bone engineering are also discussed with respect to the movement of the field towards expedited, translational approaches. It is possible that gene-enhanced bone tissue engineering will become a clinical reality within the next few years.
Collapse
Affiliation(s)
- Volker M Betz
- Department of Gene Therapy, University of Ulm, Ulm, Germany.,Center for Rehabilitation, RKU - University and Rehabilitation Hospitals Ulm, Ulm, Germany
| | | | - Stefan Rammelt
- University Center of Orthopedics and Traumatology and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, Dresden, Germany
| | - Peter E Müller
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Oliver B Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Carolin Messmer
- Center for Rehabilitation, RKU - University and Rehabilitation Hospitals Ulm, Ulm, Germany
| |
Collapse
|
41
|
Bougioukli S, Sugiyama O, Pannell W, Ortega B, Tan MH, Tang AH, Yoho R, Oakes DA, Lieberman JR. Gene Therapy for Bone Repair Using Human Cells: Superior Osteogenic Potential of Bone Morphogenetic Protein 2-Transduced Mesenchymal Stem Cells Derived from Adipose Tissue Compared to Bone Marrow. Hum Gene Ther 2018; 29:507-519. [PMID: 29212377 DOI: 10.1089/hum.2017.097] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Ex vivo regional gene therapy strategies using animal mesenchymal stem cells genetically modified to overexpress osteoinductive growth factors have been successfully used in a variety of animal models to induce both heterotopic and orthotopic bone formation. However, in order to adapt regional gene therapy for clinical applications, it is essential to assess the osteogenic capacity of transduced human cells and choose the cell type that demonstrates the best clinical potential. Bone-marrow stem cells (BMSC) and adipose-derived stem cells (ASC) were selected in this study for in vitro evaluation, before and after transduction with a lentiviral two-step transcriptional amplification system (TSTA) overexpressing bone morphogenetic protein 2 (BMP-2; LV-TSTA-BMP-2) or green fluorescent protein (GFP; LV-TSTA-GFP). Cell growth, transduction efficiency, BMP-2 production, and osteogenic capacity were assessed. The study demonstrated that BMSC were characterized by a slower cell growth compared to ASC. Fluorescence-activated cell sorting analysis of GFP-transduced cells confirmed successful transduction with the vector and revealed an overall higher but not statistically significant transduction efficiency in ASC versus BMSC (90.2 ± 4.06% vs. 80.4 ± 8.51%, respectively; p = 0.146). Enzyme-linked immunosorbent assay confirmed abundant BMP-2 production by both cell types transduced with LV-TSTA-BMP-2, with BMP-2 production being significantly higher in ASC versus BMSC (239.5 ± 116.55 ng vs. 70.86 ± 24.7 ng; p = 0.001). Quantitative analysis of extracellular deposition of calcium (Alizarin red) and alkaline phosphatase activity showed that BMP-2-transduced cells had a higher osteogenic differentiation capacity compared to non-transduced cells. When comparing the two cell types, ASC/LV-TSTA-BMP-2 demonstrated a significantly higher mineralization potential compared to BMSC/LV-TSTA-BMP-2 7 days post transduction (p = 0.014). In conclusion, this study demonstrates that transduction with LV-TSTA-BMP-2 can significantly enhance the osteogenic potential of both human BMSC and ASC. BMP-2-treated ASC exhibited higher BMP-2 production and greater osteogenic differentiation capacity compared to BMP-2-treated BMSC. These results, along with the fact that liposuction is an easy procedure with lower donor-site morbidity compared to BM aspiration, indicate that adipose tissue might be a preferable source of MSCs to develop a regional gene therapy approach to treat difficult bone-repair scenarios.
Collapse
Affiliation(s)
- Sofia Bougioukli
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Osamu Sugiyama
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - William Pannell
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Brandon Ortega
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Matthew H Tan
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Amy H Tang
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Robert Yoho
- 2 Cosmetic Surgery Practice , Pasadena, California
| | - Daniel A Oakes
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Jay R Lieberman
- 1 Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California , Los Angeles, California
| |
Collapse
|
42
|
Alluri R, Jakus A, Bougioukli S, Pannell W, Sugiyama O, Tang A, Shah R, Lieberman JR. 3D printed hyperelastic "bone" scaffolds and regional gene therapy: A novel approach to bone healing. J Biomed Mater Res A 2018; 106:1104-1110. [PMID: 29266747 DOI: 10.1002/jbm.a.36310] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/09/2017] [Accepted: 12/15/2017] [Indexed: 01/11/2023]
Abstract
The purpose of this study was to evaluate the viability of human adipose-derived stem cells (ADSCs) transduced with a lentiviral (LV) vector to overexpress bone morphogenetic protein-2 (BMP-2) loaded onto a novel 3D printed scaffold. Human ADSCs were transduced with a LV vector carrying the cDNA for BMP-2. The transduced cells were loaded onto a 3D printed Hyperelastic "Bone" (HB) scaffold. In vitro BMP-2 production was assessed using enzyme-linked immunosorbent assay analysis. The ability of ADSCs loaded on the HB scaffold to induce in vivo bone formation in a hind limb muscle pouch model was assessed in the following groups: ADSCs transduced with LV-BMP-2, LV-green fluorescent protein, ADSCs alone, and empty HB scaffolds. Bone formation was assessed using radiographs, histology and histomorphometry. Transduced ADSCs BMP-2 production on the HB scaffold at 24 hours was similar on 3D printed HB scaffolds versus control wells with transduced cells alone, and continued to increase after 1 and 2 weeks of culture. Bone formation was noted in LV-BMP-2 animals on plain radiographs at 2 and 4 weeks after implantation; no bone formation was noted in the other groups. Histology demonstrated that the LV-BMP-2 group was the only group that formed woven bone and the mean bone area/tissue area was significantly greater when compared with the other groups. 3D printed HB scaffolds are effective carriers for transduced ADSCs to promote bone repair. The combination of gene therapy and tissue engineered scaffolds is a promising multidisciplinary approach to bone repair with significant clinical potential. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 1104-1110, 2018.
Collapse
Affiliation(s)
- Ram Alluri
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, California, 90089
| | - Adam Jakus
- Department of Materials Science and Engineering, Northwestern University, 303 E. Superior St., 11th Floor, Chicago, Illinois, 60611.,Department of Materials Science and Engineering, Northwestern University, 2220 Campus Dr., Evanston, IL, 60208.,Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E Superior St., Chicago, IL, 60611
| | - Sofia Bougioukli
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, California, 90089
| | - William Pannell
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, California, 90089
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, California, 90089
| | - Amy Tang
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, California, 90089
| | - Ramille Shah
- Department of Materials Science and Engineering, Northwestern University, 2220 Campus Dr., Evanston, IL, 60208.,Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E Superior St., Chicago, IL, 60611.,Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Rd., Evanston, IL, 60208.,Department of Surgery, Division of Organ Transplantation, Northwestern University, 251 E Huron St., Chicago, IL, 60611
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, 2011 Zonal Ave, HMR 702, Los Angeles, California, 90089
| |
Collapse
|
43
|
Ren B, Betz VM, Thirion C, Salomon M, Jansson V, Müller PE, Betz OB. Gene‐activated tissue grafts for sustained bone morphogenetic protein‐2 delivery and bone engineering: Is muscle with fascia superior to muscle and fat? J Tissue Eng Regen Med 2017; 12:1002-1011. [DOI: 10.1002/term.2575] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/06/2017] [Accepted: 09/12/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Bin Ren
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital GrosshadernLudwig‐Maximilians‐University Munich Munich Germany
| | - Volker M. Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital GrosshadernLudwig‐Maximilians‐University Munich Munich Germany
| | | | | | - Volkmar Jansson
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital GrosshadernLudwig‐Maximilians‐University Munich Munich Germany
| | - Peter E. Müller
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital GrosshadernLudwig‐Maximilians‐University Munich Munich Germany
| | - Oliver B. Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital GrosshadernLudwig‐Maximilians‐University Munich Munich Germany
| |
Collapse
|
44
|
Adkar SS, Brunger JM, Willard VP, Wu CL, Gersbach CA, Guilak F. Genome Engineering for Personalized Arthritis Therapeutics. Trends Mol Med 2017; 23:917-931. [PMID: 28887050 PMCID: PMC5657581 DOI: 10.1016/j.molmed.2017.08.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/06/2017] [Accepted: 08/08/2017] [Indexed: 02/06/2023]
Abstract
Arthritis represents a family of complex joint pathologies responsible for the majority of musculoskeletal conditions. Nearly all diseases within this family, including osteoarthritis, rheumatoid arthritis, and juvenile idiopathic arthritis, are chronic conditions with few or no disease-modifying therapeutics available. Advances in genome engineering technology, most recently with CRISPR-Cas9, have revolutionized our ability to interrogate and validate genetic and epigenetic elements associated with chronic diseases such as arthritis. These technologies, together with cell reprogramming methods, including the use of induced pluripotent stem cells, provide a platform for human disease modeling. We summarize new evidence from genome-wide association studies and genomics that substantiates a genetic basis for arthritis pathogenesis. We also review the potential contributions of genome engineering in the development of new arthritis therapeutics.
Collapse
Affiliation(s)
- Shaunak S Adkar
- Department of Orthopedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | | - Chia-Lung Wu
- Department of Orthopedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Cytex Therapeutics, Inc., Durham, NC 27705, USA.
| |
Collapse
|
45
|
Betz VM, Keller A, Foehr P, Thirion C, Salomon M, Rammelt S, Zwipp H, Burgkart R, Jansson V, Müller PE, Betz OB. BMP-2 gene activated muscle tissue fragments for osteochondral defect regeneration in the rabbit knee. J Gene Med 2017; 19. [PMID: 28744947 DOI: 10.1002/jgm.2972] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Previously published data indicate that BMP-2 gene activated muscle tissue grafts can repair large bone defects in rats. This innovative abbreviated ex vivo gene therapy is appealing because it does not require elaborative and time-consuming extraction and expansion of cells. Hence, in the present study, we evaluated the potential of this expedited tissue engineering approach for regenerating osteochondral defects in rabbits. METHODS Autologous muscle tissue grafts from female White New Zealand rabbits were directly transduced with an adenoviral BMP-2 vector or remained unmodified. Osteochondral defects in the medial condyle of rabbit knees were treated with either BMP-2 activated muscle tissue implants or unmodified muscle tissue or remained empty. After 13 weeks, repair of osteochondral defects was examined by biomechanical indentation testing and by histology/imunohistochemistry applying an extended O'Driscoll scoring system and histomorphometry. RESULTS Biomechanical investigations revealed a trend towards slightly improved mechanical properties of the group receiving BMP-2 activated muscle tissue compared to unmodified muscle treatment and empty defect controls. However, a statistically significant difference was noted only between BMP-2 muscle and unmodified muscle treatment. Also, histological evaluation resulted in slightly higher histological scores and improved collagen I/II ratio without statistical significance in the BMP-2 treatment group. Histomorphometry indicated enhanced repair of subchondral bone after treatment with BMP-2 muscle, with a significantly larger bone area compared to untreated defects. CONCLUSIONS Gene activated muscle tissue grafts showed potential for osteochondral defect repair. There is room for improvement via the use of appropriate growth factor combinations.
Collapse
Affiliation(s)
- Volker M Betz
- University Center of Orthopaedics and Traumatology and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, Dresden, Germany
| | - Alexander Keller
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Peter Foehr
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | | | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, Dresden, Germany.,DFG-Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Hans Zwipp
- University Center of Orthopaedics and Traumatology and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, Dresden, Germany.,DFG-Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Rainer Burgkart
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Volkmar Jansson
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Peter E Müller
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Oliver B Betz
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
46
|
Bondarava M, Cattaneo C, Ren B, Thasler WE, Jansson V, Müller PE, Betz OB. Osseous differentiation of human fat tissue grafts: From tissue engineering to tissue differentiation. Sci Rep 2017; 7:39712. [PMID: 28054585 PMCID: PMC5213995 DOI: 10.1038/srep39712] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/25/2016] [Indexed: 12/27/2022] Open
Abstract
Conventional bone tissue engineering approaches require isolation and in vitro propagation of autologous cells, followed by seeding on a variety of scaffolds. Those protracted procedures impede the clinical applications. Here we report the transdifferentiation of human fat tissue fragments retrieved from subcutaneous fat into tissue with bone characteristics in vitro without prior cell isolation and propagation. 3D collagen-I cultures of human fat tissue were cultivated either in growth medium or in osteogenic medium (OM) with or without addition of Bone Morphogenetic Proteins (BMPs) BMP-2, BMP-7 or BMP-9. Ca2+ depositions were observed after two weeks of osteogenic induction which visibly increased when either type of BMP was added. mRNA levels of alkaline phosphatase (ALP) and osteocalcin (OCN) increased when cultured in OM alone but addition of BMP-2, BMP-7 or BMP-9 caused significantly higher expression levels of ALP and OCN. Immunofluorescent staining for OCN, osteopontin and sclerostin supported the observed real-time-PCR data. BMP-9 was the most effective osteogenic inducer in this system. Our findings reveal that tissue regeneration can be remarkably simplified by omitting prior cell isolation and propagation, therefore removing significant obstacles on the way to clinical applications of much needed regeneration treatments.
Collapse
Affiliation(s)
- Maryna Bondarava
- University Hospital of Munich (LMU), Campus Grosshadern, Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, Munich, DE, Germany
| | - Chiara Cattaneo
- University Hospital of Munich (LMU), Campus Grosshadern, Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, Munich, DE, Germany
| | - Bin Ren
- University Hospital of Munich (LMU), Campus Grosshadern, Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, Munich, DE, Germany
| | - Wolfgang E Thasler
- University Hospital of Munich (LMU), Biobank under the administration of the Human Tissue and Cell Research (HTCR) Foundation, Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Munich, DE, Germany
| | - Volkmar Jansson
- University Hospital of Munich (LMU), Campus Grosshadern, Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, Munich, DE, Germany
| | - Peter E Müller
- University Hospital of Munich (LMU), Campus Grosshadern, Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, Munich, DE, Germany
| | - Oliver B Betz
- University Hospital of Munich (LMU), Campus Grosshadern, Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, Munich, DE, Germany
| |
Collapse
|
47
|
Liao JC. Bone Marrow Mesenchymal Stem Cells Expressing Baculovirus-Engineered Bone Morphogenetic Protein-7 Enhance Rabbit Posterolateral Fusion. Int J Mol Sci 2016; 17:ijms17071073. [PMID: 27399674 PMCID: PMC4964449 DOI: 10.3390/ijms17071073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/20/2016] [Accepted: 06/27/2016] [Indexed: 02/06/2023] Open
Abstract
Previous studies have suggested that bone marrow-derived mesenchymal stem cells (BMDMSCs) genetically modified with baculoviral bone morphogenetic protein-2 (Bac-BMP-2) vectors could achieve successful fusion in a femur defect model or in a spinal fusion model. In this study, BMDMSCs expressing BMP-7 (Bac-BMP-7-BMDMSCs) were generated. We hypothesized that Bac-BMP-7-BMDMSCs could secrete more BMP-7 than untransduced BMDMSCs in vitro and achieve spinal posterolateral fusion in a rabbit model. Eighteen rabbits underwent posterolateral fusion at L4-5. Group I (n = 6) was implanted with collagen-β-tricalcium phosphate (TCP)-hydroxyapatite (HA), Group II (n = 6) was implanted with collagen-β-TCP-HA plus BMDMSCs, and Group III (n = 6) was implanted with collagen-β-TCP-HA plus Bac-BMP-7-BMDMSCs. In vitro production of BMP-7 was quantified with an enzyme-linked immunosorbent assay (ELISA). Spinal fusion was examined using computed tomography (CT), manual palpation, and histological analysis. ELISA demonstrated that Bac-BMP-7-BMDMSCs produced four-fold to five-fold more BMP-7 than did BMDMSCs. In the CT results, 6 fused segments were observed in Group I (50%, 6/12), 8 in Group II (67%, 8/12), and 12 in Group III (100%, 12/12). The fusion rate, determined by manual palpation, was 0% (0/6) in Group I, 0% (0/6) in Group II, and 83% (5/6) in Group III. Histology showed that Group III had more new bone and matured marrow formation. In conclusion, BMDMSCs genetically transduced with the Bac-BMP-7 vector could express more BMP-7 than untransduced BMDMSCs. These Bac-BMP-7-BMDMSCs on collagen-β-TCP-HA scaffolds were able to induce successful spinal fusion in rabbits.
Collapse
Affiliation(s)
- Jen-Chung Liao
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan.
| |
Collapse
|
48
|
Murray IR, LaPrade RF, Musahl V, Geeslin AG, Zlotnicki JP, Mann BJ, Petrigliano FA. Biologic Treatments for Sports Injuries II Think Tank-Current Concepts, Future Research, and Barriers to Advancement, Part 2: Rotator Cuff. Orthop J Sports Med 2016; 4:2325967116636586. [PMID: 27099865 PMCID: PMC4820026 DOI: 10.1177/2325967116636586] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Rotator cuff tears are common and result in considerable morbidity. Tears within the tendon substance or at its insertion into the humeral head represent a considerable clinical challenge because of the hostile local environment that precludes healing. Tears often progress without intervention, and current surgical treatments are inadequate. Although surgical implants, instrumentation, and techniques have improved, healing rates have not improved, and a high failure rate remains for large and massive rotator cuff tears. The use of biologic adjuvants that contribute to a regenerative microenvironment have great potential for improving healing rates and function after surgery. This article presents a review of current and emerging biologic approaches to augment rotator cuff tendon and muscle regeneration focusing on the scientific rationale, preclinical, and clinical evidence for efficacy, areas for future research, and current barriers to advancement and implementation.
Collapse
Affiliation(s)
| | | | - Volker Musahl
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Andrew G Geeslin
- Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, Michigan, USA
| | - Jason P Zlotnicki
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Barton J Mann
- Author deceased.; American Orthopaedic Society for Sports Medicine, Rosemont, Illinois, USA
| | | |
Collapse
|
49
|
Bougioukli S, Jain A, Sugiyama O, Tinsley BA, Tang AH, Tan MH, Adams DJ, Kostenuik PJ, Lieberman JR. Combination therapy with BMP-2 and a systemic RANKL inhibitor enhances bone healing in a mouse critical-sized femoral defect. Bone 2016; 84:93-103. [PMID: 26723577 PMCID: PMC4903101 DOI: 10.1016/j.bone.2015.12.052] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/08/2015] [Accepted: 12/22/2015] [Indexed: 11/26/2022]
Abstract
Recombinant human BMP-2 (rhBMP-2) is a potent osteoinductive agent, but has been associated not only with bone formation, but also osteoclastogenesis and bone resorption. Osteoprotegerin (OPG) is a RANKL inhibitor that blocks differentiation and function of osteoclasts. We hypothesized that the combination of local BMP-2 (recombinant protein or a product of gene therapy) plus systemic OPG-Fc is more effective than BMP-2 alone in promoting bone repair. To test this hypothesis we used a mouse critical-sized femoral defect model. Col2.3eGFP (osteoblastic marker) male mice were treated with rhBMP-2 (group I), rhBMP-2 and systemic OPG (group II), rhBMP-2 and delayed administration of OPG (group III), mouse BM cells transduced with a lentiviral vector containing the BMP-2 gene (LV-BMP-2; group IV), LV-BMP-2 and systemic OPG (group V), a carrier alone (group VI) and administration of OPG alone (group VII). All bone defects treated with BMP-2 (alone or combined with OPG) healed, whereas minimal bone formation was noted in animals treated with the carrier alone or OPG alone. MicroCT analysis showed that bone volume (BV) in rhBMP-2+OPG and LV-BMP-2+OPG groups was significantly higher compared to rhBMP-2 alone (p<0.01) and LV-BMP-2 alone (p<0.001). Similar results were observed in histomorphometry, with rhBMP-2 alone defects exhibiting significantly lower bone area (B.Ar) compared to rhBMP-2+OPG defects (p<0.005) and LV-BMP-2 defects having a significantly lower B.Ar compared to all BMP-2+OPG treated groups (p≤0.01). TRAP staining demonstrated a major osteoclast response in the groups that did not receive OPG (rhBMP-2, LV-BMP-2 and sponge alone) beginning as early as 7days post-operatively. In conclusion, we demonstrated that locally delivered BMP-2 (recombinant protein or gene therapy) in combination with systemically administered OPG improved bone healing compared to BMP-2 alone in a mouse critical-sized bone defect. These data indicate that osteoclasts can diminish healing responses to BMP-2 and that RANKL inhibition may thus accentuate BMP-2 efficacy.
Collapse
Affiliation(s)
- Sofia Bougioukli
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ashish Jain
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, University of Connecticut Health, Farmington, CT, USA
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brian A Tinsley
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, University of Connecticut Health, Farmington, CT, USA
| | - Amy H Tang
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Matthew H Tan
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Douglas J Adams
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, University of Connecticut Health, Farmington, CT, USA
| | - Paul J Kostenuik
- Phylon Pharma Services, Newbury Park, CA, USA; Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
50
|
Yin PT, Han E, Lee KB. Engineering Stem Cells for Biomedical Applications. Adv Healthc Mater 2016; 5:10-55. [PMID: 25772134 PMCID: PMC5810416 DOI: 10.1002/adhm.201400842] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/14/2015] [Indexed: 12/19/2022]
Abstract
Stem cells are characterized by a number of useful properties, including their ability to migrate, differentiate, and secrete a variety of therapeutic molecules such as immunomodulatory factors. As such, numerous pre-clinical and clinical studies have utilized stem cell-based therapies and demonstrated their tremendous potential for the treatment of various human diseases and disorders. Recently, efforts have focused on engineering stem cells in order to further enhance their innate abilities as well as to confer them with new functionalities, which can then be used in various biomedical applications. These engineered stem cells can take on a number of forms. For instance, engineered stem cells encompass the genetic modification of stem cells as well as the use of stem cells for gene delivery, nanoparticle loading and delivery, and even small molecule drug delivery. The present Review gives an in-depth account of the current status of engineered stem cells, including potential cell sources, the most common methods used to engineer stem cells, and the utilization of engineered stem cells in various biomedical applications, with a particular focus on tissue regeneration, the treatment of immunodeficiency diseases, and cancer.
Collapse
Affiliation(s)
- Perry T Yin
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08854, USA
| | - Edward Han
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, M5S 3G9, Canada
| | - Ki-Bum Lee
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08854, USA
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 610 Taylor Road, Piscataway, NJ, 08854, USA
| |
Collapse
|