1
|
Amini M, Venkatesan JK, Liu W, Leroux A, Nguyen TN, Madry H, Migonney V, Cucchiarini M. Advanced Gene Therapy Strategies for the Repair of ACL Injuries. Int J Mol Sci 2022; 23:ijms232214467. [PMID: 36430947 PMCID: PMC9695211 DOI: 10.3390/ijms232214467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/07/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022] Open
Abstract
The anterior cruciate ligament (ACL), the principal ligament for stabilization of the knee, is highly predisposed to injury in the human population. As a result of its poor intrinsic healing capacities, surgical intervention is generally necessary to repair ACL lesions, yet the outcomes are never fully satisfactory in terms of long-lasting, complete, and safe repair. Gene therapy, based on the transfer of therapeutic genetic sequences via a gene vector, is a potent tool to durably and adeptly enhance the processes of ACL repair and has been reported for its workability in various experimental models relevant to ACL injuries in vitro, in situ, and in vivo. As critical hurdles to the effective and safe translation of gene therapy for clinical applications still remain, including physiological barriers and host immune responses, biomaterial-guided gene therapy inspired by drug delivery systems has been further developed to protect and improve the classical procedures of gene transfer in the future treatment of ACL injuries in patients, as critically presented here.
Collapse
Affiliation(s)
- Mahnaz Amini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Jagadeesh K. Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Amélie Leroux
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Tuan Ngoc Nguyen
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
| | - Véronique Migonney
- Laboratoire CSPBAT UMR CNRS 7244, Université Sorbonne Paris Nord, Avenue JB Clément, 93430 Villetaneuse, France
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany
- Correspondence: or
| |
Collapse
|
2
|
Aimaletdinov A, Mindubaeva G, Khalikova S, Kabwe E, Salmakova A, Alexandrova N, Rutland C, Rizvanov A, Zakirova E. Application of gene therapy in the treatment of superficial digital flexor tendon injury in horses. Open Vet J 2020; 10:261-266. [PMID: 33282696 PMCID: PMC7703612 DOI: 10.4314/ovj.v10i3.3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/02/2020] [Indexed: 11/29/2022] Open
Abstract
Background: Tendon injuries are one of the most common causes of orthopedic disorders in horses. Such injuries involve a long course of treatment and recovery. The most promising method of treating these injuries is the use of recombinant proteins and gene therapy. Aim: In this work, we evaluated the therapeutic efficacy of plasmid DNA (pDNA) containing two species-specific coding sequences, i.e. vascular endothelial growth factor 164 (VEGF164) and fibroblast growth factor 2 (FGF2), in the treatment of severe damage to the tendon of the superficial digital flexor. Methods: A pDNA construct was used to restore the damaged superficial digital flexor tendon in the horse. Results: This study showed that the administration of pDNA encoding VEGF164 and FGF2 genes at the injury area increased the regenerative activities of the damaged tendon. Conclusion: This study shows the therapeutic properties of genetic constructs (pDNA) and contributes to the advancements in the use of these therapies.
Collapse
Affiliation(s)
- Alexandr Aimaletdinov
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | | | | | - Emmanuel Kabwe
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation.,Kazan Research Institute of Epidemiology and Microbiology, Kazan, the Republic of Tatarstan, Russian Federation
| | - Alexandra Salmakova
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Natalia Alexandrova
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Catrin Rutland
- Faculty of Medicine, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Albert Rizvanov
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Elena Zakirova
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| |
Collapse
|
3
|
Ahmed MF, El-Sayed AK, Chen H, Zhao R, Jin K, Zuo Q, Zhang Y, Li B. Direct conversion of mouse embryonic fibroblast to osteoblast cells using hLMP-3 with Yamanaka factors. Int J Biochem Cell Biol 2018; 106:84-95. [PMID: 30453092 DOI: 10.1016/j.biocel.2018.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/05/2018] [Accepted: 11/16/2018] [Indexed: 01/14/2023]
Abstract
Large bone defects and bone loss after fractures remain significant challenges for orthopedic surgeons. Our study aims to find an available, applicable and biological treatment for bone regeneration overcoming the limitations in ESC/iPSC technology. We directly reprogrammed the mouse embryonic fibroblast (MEF) into osteoblast cells using different combinations of Yamanaka factors with human lim mineralization protein-3 (hLMP-3). LMP is an intracellular LIM-domain protein acting as an effective positive regulator of the osteoblast differentiation. After transduction, cells were cultured in osteogenic medium, and then examined for osteoblast formation. The expression of osteogenic markers (BMP2, Runx2 and Osterix) during reprogramming and in vitro mineralization assay revealed that the best reprogramming cocktail was (c-Myc - Oct4) with hLMP-3. In addition, both immunofluorescent staining and western blot analysis confirmed that osteocalcin (OCN) expression increased in the cells treated with the c-Myc/Oct4/hLMP3 cocktail than using hLMP-3 alone. Furthermore, this reprogramming cocktail showed efficient healing in an induced femoral bone defect in rat animal model one month after transplantation. In the present study, we reported for the first time the effect of combining Yamanaka factors with hLMP-3 to induce osteoblast cells from MEF both in vitro and in vivo.
Collapse
Affiliation(s)
- Mahmoud F Ahmed
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China; College of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | | | - Hao Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, 215006, China
| | - Ruifeng Zhao
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Kai Jin
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Qisheng Zuo
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yani Zhang
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Bichun Li
- Key Laboratory of Animal Breeding, Reproduction and Molecular Design for Jiangsu Provience, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
4
|
Kovac M, Litvin YA, Aliev RO, Zakirova EY, Rutland CS, Kiyasov AP, Rizvanov AA. Gene Therapy Using Plasmid DNA Encoding VEGF164 and FGF2 Genes: A Novel Treatment of Naturally Occurring Tendinitis and Desmitis in Horses. Front Pharmacol 2018; 9:978. [PMID: 30233367 PMCID: PMC6127648 DOI: 10.3389/fphar.2018.00978] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 08/08/2018] [Indexed: 11/30/2022] Open
Abstract
This clinical study describes the intralesional application of the plasmid DNA encoding two therapeutic species-specific growth factors: vascular endothelial growth factor (VEGF164) and fibroblast growth factor 2 (FGF2) in seven horses to restore naturally occurring injuries of the superficial digital flexor tendon (SDFT) (tendinitis) and in three horses with suspensory ligament branch desmitis. Following application all horses were able to commence a more rapid exercise program in comparison to standardized exercise programs. Clinical observation and ultrasonic imaging was used to evaluate the regeneration rate of the tendon and ligament injury recovery and to confirm the safety of this gene therapy in horses, throughout a 12 month period. Follow-up data of the horses revealed a positive outcome including significant ultrasonographic and clinical improvements in 8 out of 10 horses with SDFT and suspensory ligament branch lesions, with return to their pre-injury level of performance by 2–6 months after the completion of treatment. The ninth horse initially presenting with severe suspensory ligament branch desmopathy, showed no significant ultrasonographic improvements in the first 2 months after treatment, however, it improved clinically and became less lame. The final horse, presenting with severe tendinitis of the SDFT returned to their pre-injury level of performance, but experienced re-injury 6 months after treatment. This data is highly promising, however, further research in experimental models, with the histopathological, immunohistochemical and gene expression evaluation of the equine tendon/ligament after gene therapy application is required in order to fully understand the mechanisms of action. This treatment and the significant clinical impacts observed represents an important advancement in the field of medicine.
Collapse
Affiliation(s)
- Milomir Kovac
- Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | - Yaroslav A Litvin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Ruslan O Aliev
- Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | - Elena Y Zakirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Catrin S Rutland
- School of Veterinary Medicine and Science, Faculty of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Andrey P Kiyasov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
5
|
Gene Therapy Strategies in Bone Tissue Engineering and Current Clinical Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1119:85-101. [DOI: 10.1007/5584_2018_253] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
6
|
Ma J, Guo W, Gao M, Huang B, Qi Q, Ling Z, Chen Y, Hu H, Zhou H, Yu F, Chen K, Richards G, Lin J, Zhou Z, Xiao D, Zou X. Biomimetic matrix fabricated by LMP-1 gene-transduced MC3T3-E1 cells for bone regeneration. Biofabrication 2017; 9:045010. [PMID: 28930090 DOI: 10.1088/1758-5090/aa8dd1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bone healing is regulated by multiple microenvironmental signals provided by the extracellular matrix (ECM). This study aimed to mimic the native osteoinductive microenvironment by developing an ECM using gene-transduced cells. The LIM mineralization protein-1 (LMP-1) gene was transferred to murine pre-osteoblast cells (MC3T3-E1) using lentiviral vectors. Western blotting assay indicated that the MC3T3-E1 cells expressed an increased level of bone morphologic protein-2, -4 and -7 (BMP-2, -4 and -7) after LMP-1 gene transduction. The transduced cells were then seeded into calcined bovine bone scaffolds and cultured for 7, 14, and 21 days to construct ECMs on the scaffolds. The ECM-scaffold composites were then decellularized using the freeze-drying method. Scaffolds without ECM deposition were used as controls. The composites and controls were implanted into critical-sized bone defects created in the distal femurs of New Zealand rabbits. Twelve weeks after the surgery, both microcomputed tomography and histologic results indicated that the 7-day-cell-modified ECM-scaffold composites induced bone regeneration with significantly larger volume, trabecular thickness and connectivity than the controls. However, the 14- and 21-day-cell-modified ECM-scaffold composites triggered sustained inflammation response even at 12 weeks after the surgery and showed less bone ingrowth and integration than their 7-day-cell-modified counterparts. In conclusion, these results highlight the viable gene transfer techniques for manipulating cells in a constructed microenvironment of ECM for bone regeneration. However, the unresolved inflammation relating to the duration of ECM modification needs to be considered.
Collapse
Affiliation(s)
- Junxuan Ma
- Department of Orthopedic, Peking University Shenzhen Hospital, Shenzhen, People's Republic of China. Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliate Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Kovac M, Litvin YA, Aliev RO, Zakirova EY, Rutland CS, Kiyasov AP, Rizvanov AA. Gene Therapy Using Plasmid DNA Encoding Vascular Endothelial Growth Factor 164 and Fibroblast Growth Factor 2 Genes for the Treatment of Horse Tendinitis and Desmitis: Case Reports. Front Vet Sci 2017; 4:168. [PMID: 29067288 PMCID: PMC5641304 DOI: 10.3389/fvets.2017.00168] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/25/2017] [Indexed: 11/16/2022] Open
Abstract
In this clinical study, for the first time we used the direct gene therapy to restore severe injuries of the suspensory ligament branch and superficial digital flexor tendon in horses (Equus caballus). We injected the plasmid DNA encoding two therapeutic species-specific growth factors: vascular endothelial growth factor 164 and fibroblast growth factor 2 at the site of injury in the suspensory ligament branch and tendon. Treatment effects were evaluated with the use of clinical observation and ultrasound imaging during a period of a few months. We showed that gene therapy used within a period of 2–3 months after the injury resulted in the complete recovery of functions and full restoration of the severely damaged suspensory ligament and superficial digital flexor tendon.
Collapse
Affiliation(s)
- Milomir Kovac
- Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | | | - Ruslan O Aliev
- Moscow State Academy of Veterinary Medicine and Biotechnology, Moscow, Russia
| | | | - Catrin S Rutland
- Faculty of Medicine, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | | | | |
Collapse
|
8
|
Basic FGF or VEGF gene therapy corrects insufficiency in the intrinsic healing capacity of tendons. Sci Rep 2016; 6:20643. [PMID: 26865366 PMCID: PMC4749961 DOI: 10.1038/srep20643] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 01/07/2016] [Indexed: 12/11/2022] Open
Abstract
Tendon injury during limb motion is common. Damaged tendons heal poorly and frequently undergo unpredictable ruptures or impaired motion due to insufficient innate healing capacity. By basic fibroblast growth factor (bFGF) or vascular endothelial growth factor (VEGF) gene therapy via adeno-associated viral type-2 (AAV2) vector to produce supernormal amount of bFGF or VEGF intrinsically in the tendon, we effectively corrected the insufficiency of the tendon healing capacity. This therapeutic approach (1) resulted in substantial amelioration of the low growth factor activity with significant increases in bFGF or VEGF from weeks 4 to 6 in the treated tendons (p < 0.05 or p < 0.01), (2) significantly promoted production of type I collagen and other extracellular molecules (p < 0.01) and accelerated cellular proliferation, and (3) significantly increased tendon strength by 68–91% from week 2 after AAV2-bFGF treatment and by 82–210% from week 3 after AAV2-VEGF compared with that of the controls (p < 0.05 or p < 0.01). Moreover, the transgene expression dissipated after healing was complete. These findings show that the gene transfers provide an optimistic solution to the insufficiencies of the intrinsic healing capacity of the tendon and offers an effective therapeutic possibility for patients with tendon disunion.
Collapse
|
9
|
Fu TS, Chang YH, Wong CB, Wang IC, Tsai TT, Lai PL, Chen LH, Chen WJ. Mesenchymal stem cells expressing baculovirus-engineered BMP-2 and VEGF enhance posterolateral spine fusion in a rabbit model. Spine J 2015; 15:2036-44. [PMID: 25463976 DOI: 10.1016/j.spinee.2014.11.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/11/2014] [Accepted: 11/05/2014] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Mesenchymal stem cell (MSC)-based cell therapy and gene transfer have converged and show great potential for accelerating bone healing. Gene therapy can provide more sustained expression of osteogenic factors such as bone morphogenetic protein-2 (BMP-2). We previously demonstrated that low-dose BMP-2 enhanced spinal posterolateral fusion by MSCs in a rabbit model. Herein, we genetically modified rabbit MSCs with a recombinant baculovirus encoding BMP-2 (Bac-CB) and vascular endothelial growth factor (Bac-VEGF) seeded into porous scaffolds to enhance spinal fusion. PURPOSE This study evaluates the success rate of the MSC-based cell therapy and gene transfer approach for single-level posterolateral spine fusion. We hypothesize that combining three-dimensional tricalcium phosphate (TCP) scaffolds and genetically modified allogeneic MSCs with baculovirus-mediated growth factor expression would increase the success rate of spinal fusion. STUDY DESIGN The study design was based on an animal model (approved by the Institutional Animal Care and Use Committee) using 18 adult male New Zealand rabbits. METHODS This study included 18 male New Zealand rabbits, weighing 3.5 to 4 kg. Allogeneic bone marrow-derived MSCs were isolated and genetically modified with Bac-CB and Bac-CV seeded onto TCP scaffolds (MSC/Bac/TCP). The animals were divided into three groups according to the material implanted into the bilateral L4-L5 intertransverse space: TCP scaffold (n=6), MSC/TCP (n=6), and MSC/Bac/TCP (n=6). After 12 weeks, the rabbits were euthanized for radiographic examination, manual palpation, and histologic study. RESULTS Bilateral fusion areas in each animal were evaluated independently. The radiographic fusion rates at 12 sites were 0 of 12 in the TCP scaffold group, 4 of 12 in the MSC/TCP group, and 10 of 12 in the MSC/Bac/TCP group. By manual palpation, there were zero solid fusions in the TCP scaffold group, two solid fusions in the MSC/TCP group, and five solid fusions in the MSC/Bac/TCP group. Fusion rates were significantly greater in the MSC/Bac/TCP group. CONCLUSIONS The results indicate the potential of using baculovirus as a vector for gene/cell therapy approaches to improve bone healing and support the feasibility of using allogeneic MSCs for inducing bone formation and intertransverse process fusion.
Collapse
Affiliation(s)
- Tsai-Sheng Fu
- Department of Orthopaedic Surgery, Keelung Chang Gung Memorial Hospital, Chang Gung University, 7F, No. 222, Maijin Rd, Keelung 20401, Taiwan.
| | - Yu-Han Chang
- Department of Orthopaedic Surgery, Linkou Chang Gung Memorial Hospital, Chang Gung University, No. 5, Fusing St., Gueishan, Taoyuan 333, Taiwan
| | - Chak-Bor Wong
- Department of Orthopaedic Surgery, Keelung Chang Gung Memorial Hospital, Chang Gung University, 7F, No. 222, Maijin Rd, Keelung 20401, Taiwan
| | - I-Chun Wang
- Department of Orthopaedic Surgery, Keelung Chang Gung Memorial Hospital, Chang Gung University, 7F, No. 222, Maijin Rd, Keelung 20401, Taiwan
| | - Tsung-Ting Tsai
- Department of Orthopaedic Surgery, Linkou Chang Gung Memorial Hospital, Chang Gung University, No. 5, Fusing St., Gueishan, Taoyuan 333, Taiwan
| | - Po-Liang Lai
- Department of Orthopaedic Surgery, Linkou Chang Gung Memorial Hospital, Chang Gung University, No. 5, Fusing St., Gueishan, Taoyuan 333, Taiwan
| | - Lih-Huei Chen
- Department of Orthopaedic Surgery, Linkou Chang Gung Memorial Hospital, Chang Gung University, No. 5, Fusing St., Gueishan, Taoyuan 333, Taiwan
| | - Wen-Jer Chen
- Department of Orthopaedic Surgery, Linkou Chang Gung Memorial Hospital, Chang Gung University, No. 5, Fusing St., Gueishan, Taoyuan 333, Taiwan
| |
Collapse
|
10
|
Li KC, Hu YC. Cartilage tissue engineering: recent advances and perspectives from gene regulation/therapy. Adv Healthc Mater 2015; 4:948-68. [PMID: 25656682 DOI: 10.1002/adhm.201400773] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/10/2015] [Indexed: 12/16/2022]
Abstract
Diseases in articular cartilages affect millions of people. Despite the relatively simple biochemical and cellular composition of articular cartilages, the self-repair ability of cartilage is limited. Successful cartilage tissue engineering requires intricately coordinated interactions between matrerials, cells, biological factors, and phycial/mechanical factors, and still faces a multitude of challenges. This article presents an overview of the cartilage biology, current treatments, recent advances in the materials, biological factors, and cells used in cartilage tissue engineering/regeneration, with strong emphasis on the perspectives of gene regulation (e.g., microRNA) and gene therapy.
Collapse
Affiliation(s)
- Kuei-Chang Li
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| | - Yu-Chen Hu
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| |
Collapse
|
11
|
Lin CY, Chang YH, Sung LY, Chen CL, Lin SY, Li KC, Yen TC, Lin KJ, Hu YC. Long-term tracking of segmental bone healing mediated by genetically engineered adipose-derived stem cells: focuses on bone remodeling and potential side effects. Tissue Eng Part A 2014; 20:1392-402. [PMID: 24367947 DOI: 10.1089/ten.tea.2013.0314] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
We previously showed that transplantation of adipose-derived stem cells (ASCs) engineered with hybrid baculovirus (BV) persistently expressing bone morphogenetic protein 2 (BMP2)/vascular endothelial growth factor (VEGF) into segmental defects in New Zealand White (NZW) rabbits led to successful defect reunion. By using microcomputed tomography and histology, here we further demonstrated that transplanting the hybrid BV-engineered ASCs into the massive defects (10 mm in length) at the femoral diaphysis of NZW rabbits resulted in trabecular bone formation in the interior via endochondral ossification and bone remodeling at 3 months post-transplantation. The progression of bone remodeling gave rise to the resorption of trabecular bone and conspicuous reconstruction of medullary cavity and cortical bone with lamellar structure at 8 months post-transplantation, hence conferring mechanical properties that were comparable to those of nonoperated femora. Importantly, X-ray, positron emission tomography/computed tomography scans, and histopathology revealed no signs of heterotopic bone formation and tumor formation. These data altogether attested that the genetically engineered ASCs and prolonged BMP2/VEGF expression not only healed and remodeled the stringent segmental defects, but also revitalized the defects into living bone tissues that structurally and biomechanically resembled intact bones without appreciable side effects, making it one step closer to translate this technology to the clinical setting.
Collapse
Affiliation(s)
- Chin-Yu Lin
- 1 Department of Chemical Engineering, National Tsing Hua University , Hsinchu, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Therapeutic potential of a low-cost device for wound healing: a study of three cases of healing after lower-extremity amputation in patients with diabetes. Am J Ther 2014; 20:394-8. [PMID: 23344093 DOI: 10.1097/mjt.0b013e318235f309] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Diabetic foot ulcers constitute a tremendous challenge for patients, caregivers, and health care systems. The high incidence and high financial costs associated with their treatment have transformed them in a health and economic worldwide problem. The increase in population life expectancy and lifestyle changes have facilitated the spreading of diabetes, rising diabetic foot ulcer incidence. Only 60%-80% of the patients achieve healing of ulcers, and the incidence of a second ulcer, in the same or different site of the foot that has had a previous ulcer, is approximately 50% in 2-5 years. In addition, ulcers with duration longer than 4 weeks are commonly associated with bad results in healing and an increased risk of amputation. Three patients with type 2 diabetes mellitus have been subjected to treatment with NL.1.2, a low-cost, biocompatible solid device that presented pro-angiogenic properties. The selected patients had undergone amputation, and their wounds, classified as Wagner II, did not show a significant progress in healing after a period of 2-5 months before treatment with NL.1.2. Complete closure of their wounds was achieved in 42-60 days.
Collapse
|
13
|
Recent progresses in gene delivery-based bone tissue engineering. Biotechnol Adv 2013; 31:1695-706. [DOI: 10.1016/j.biotechadv.2013.08.015] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 07/24/2013] [Accepted: 08/19/2013] [Indexed: 12/18/2022]
|
14
|
Gong L, Zhou X, Wu Y, Zhang Y, Wang C, Zhou H, Guo F, Cui L. Proteomic analysis profile of engineered articular cartilage with chondrogenic differentiated adipose tissue-derived stem cells loaded polyglycolic acid mesh for weight-bearing area defect repair. Tissue Eng Part A 2013; 20:575-87. [PMID: 24044689 DOI: 10.1089/ten.tea.2013.0205] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The present study was designed to investigate the possibility of full-thickness defects repair in porcine articular cartilage (AC) weight-bearing area using chondrogenic differentiated autologous adipose-derived stem cells (ASCs) with a follow-up of 3 and 6 months, which is successive to our previous study on nonweight-bearing area. The isolated ASCs were seeded onto the phosphoglycerate/polylactic acid (PGA/PLA) with chondrogenic induction in vitro for 2 weeks as the experimental group prior to implantation in porcine AC defects (8 mm in diameter, deep to subchondral bone), with PGA/PLA only as control. With follow-up time being 3 and 6 months, both neo-cartilages of postimplantation integrated well with the neighboring normal cartilage and subchondral bone histologically in experimental group, whereas only fibrous tissue in control group. Immunohistochemical and toluidine blue staining confirmed similar distribution of COL II and glycosaminoglycan in the regenerated cartilage to the native one. A vivid remolding process with repair time was also witnessed in the neo-cartilage as the compressive modulus significantly increased from 70% of the normal cartilage at 3 months to nearly 90% at 6 months, which is similar to our former research. Nevertheless, differences of the regenerated cartilages still could be detected from the native one. Meanwhile, the exact mechanism involved in chondrogenic differentiation from ASCs seeded on PGA/PLA is still unknown. Therefore, proteome is resorted leading to 43 proteins differentially identified from 20 chosen two-dimensional spots, which do help us further our research on some committed factors. In conclusion, the comparison via proteome provided a thorough understanding of mechanisms implicating ASC differentiation toward chondrocytes, which is further substantiated by the present study as a perfect supplement to the former one in nonweight-bearing area.
Collapse
Affiliation(s)
- Lunli Gong
- 1 Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Niemansburg SL, van Delden JJ, Dhert WJ, Bredenoord AL. Regenerative medicine interventions for orthopedic disorders: ethical issues in the translation into patients. Regen Med 2013; 8:65-73. [PMID: 23259806 DOI: 10.2217/rme.12.112] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Regenerative medicine (RM) technologies, such as cell therapy, gene transfer and tissue engineering, are expected to move the field of orthopedics into a new era. Now that more and more attempts are underway to translate preclinical research into clinical studies, it is time to proactively discuss the ethical issues associated with first-in-human applications of RM interventions for musculoskeletal disorders. The design and launch of early clinical trials will be ethically challenging due to the specific features of RM in general, and the application for musculoskeletal disorders specifically. In this paper, we identify three sets of ethical issues that need to be addressed when considering initiating early clinical trials: assessment of risks and benefits; designing a study in terms of outcome measures and comparators; and participant selection. These issues are particularly emphasized in RM research that aims to apply these approaches in an early stage of degenerative musculoskeletal disorders.
Collapse
Affiliation(s)
- Sophie L Niemansburg
- Department of Medical Humanities, Julius Center, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
16
|
Yuan QY, Huang J, Li XJ, Peng JB, Li XS, Chen ZP, Si LY. A transendocardial delivery and intracardiac ultrasound irradiation treatment catheter. Drug Deliv 2013; 20:252-7. [DOI: 10.3109/10717544.2013.801048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
17
|
Guo H, Zhang J, Hao S, Jin Q. Adenovirus-mediated small interfering RNA targeting tumor necrosis factor-α inhibits titanium particle-induced osteoclastogenesis and bone resorption. Int J Mol Med 2013; 32:296-306. [PMID: 23760678 DOI: 10.3892/ijmm.2013.1416] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 03/04/2013] [Indexed: 11/05/2022] Open
Abstract
Wear particles are phagocytosed by macrophages, resulting in cellular activation and the release of pro-inflammatory factors, which cause periprosthetic osteolysis and subsequent aseptic loosening, the most common causes of total joint arthroplasty (TJA) failure. During this pathological process, tumor necrosis factor (TNF)-α plays an important role in wear particle-induced osteolysis. Therefore, in this study, we used adenovirus-mediated small interfering RNA (siRNA) targeting TNF-α to suppress the TNF-α release from activated macrophages in response to titanium particles. Our results showed that recombinant adenovirus (Ad-TNF-α-siRNA) suppressed the TNF-α release from activated macrophages in response to titanium particles, and reduced titanium particle-induced osteoclastogenesis and bone resorption in the presence of receptor activator of nuclear factor-κB ligand (RANKL). In addition, the conditioned medium of macrophages challenged with titanium particles (Ti CM) stimulated osteoprogenitor RANKL expression. The conditioned medium of macrophages challenged with titanium particles and Ad-TNF-α-siRNA (Ti-Ad CM) reduced the mRNA expression in MC3T3-E1 cells compared to Ti CM. Based on these data, TNF-α strongly synergizes with RANKL to promote osteoclast differentiation. Furthermore, TNF-α promoted osteoclast differentiation by stimulating osteoprogenitor RANKL expression. Ad-TNF-α-siRNA effectively suppressed osteoclast differentiation and bone resorption following exposure to titanium particles in the presence of RANKL. In addition, recombinant adenovirus (Ad-TNF-α-siRNA) does not have a toxic effect on the murine macrophage cell line, RAW264.7. Consequently, it can be concluded that recombinant adenovirus-mediated siRNA targeting TNF-α (Ad-TNF-α-siRNA) may provide a novel therapeutic approach for the treatment of periprosthetic osteolysis.
Collapse
Affiliation(s)
- Haohui Guo
- Department of Orthopedic Surgery, Ningxia Medical University Affiliated Hospital, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | | | | | | |
Collapse
|
18
|
Zhao X, Li Z, Pan H, Liu W, Lv M, Leung F, Lu WW. Enhanced gene delivery by chitosan-disulfide-conjugated LMW-PEI for facilitating osteogenic differentiation. Acta Biomater 2013; 9:6694-703. [PMID: 23395816 DOI: 10.1016/j.actbio.2013.01.039] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 01/25/2013] [Accepted: 01/30/2013] [Indexed: 10/27/2022]
Abstract
Chitosan-disulfide-conjugated LMW-PEI (CS-ss-PEI) was designed to combine the biocompatibility of chitosan and the gene delivery ability of polyethylenimine (PEI) using bio-reducible disulfide for bone morphogenetic protein (BMP2) gene delivery in mediating osteogenic differentiation. It was prepared by conjugating low molecular weight PEI (LMW-PEI) to chitosan through oxidization of thiols introduced for the formation of disulfide linkage. The structure, molecular weight and buffer capacity were characterized by Fourier transform infrared (FTIR), light scattering and acid-base titration, respectively. The reduction in molecular weight of CS-ss-PEI by the reducing agent indicated its bio-reducible property. With the increment in the LMW-PEI component, the copolymer showed increased DNA binding ability and formed denser nanocomplexes. CS-ss-PEI exhibited low cytotoxicity in COS-1, HepG2 and 293T cells over the different weight ratios. The transfection efficiency of CS-ss-PEI4 was significantly higher than that of PEI 25k and comparable with Lipofectamine in mediating luciferase expression. Its application for BMP2 gene delivery was confirmed in C2C12 cells by BMP2 expression. For inducing in vitro osteogenic differentiation, CS-ss-PEI4 mediated BMP2 gene delivery showed a stronger effect in MG-63 osteoblast cells and stem cells in terms of alkaline phosphatase activity and mineralization compared with PEI25k and Lipofectamine. This study provides a potential gene delivery system for orthopedic-related disease.
Collapse
|
19
|
Mashayekhi K, O’Brien M, Zugun-Eloae F, Labusca L. Novel approaches for treating musculoskeletal diseases: molecular orthopedics and systems medicine. Open Orthop J 2013; 7:144-51. [PMID: 23798982 PMCID: PMC3664448 DOI: 10.2174/1874325001307010144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 01/25/2013] [Accepted: 02/02/2012] [Indexed: 02/07/2023] Open
Abstract
Molecular medicine uses knowledge about cell structure and function for disease, diagnostics, stage characterisation and treatment. The advent of genomic technologies is considerably leading to developments in the field of molecular medicine. The accumulation of detailed information about gene expression, epigenetic variability, protein transcription and functional modulation is contributing to a new era in medicine. Rapid and early diagnostic procedures, molecular characterisation of degenerative and proliferative diseases and personalized therapies are predicted to lead to advancements in health prevention and treatment of disease. Diagnostic tools and therapies based on local and /or general modulation of cellular processes for traumatic or degenerative musculoskeletal conditions are becoming available. A logical consequence of the information derived from extensive data gathering, systems biology and systemic medicine has lead to significant improvements in understanding biological structure and function in a simultaneous bottom top and integrative, holistic manner. The description of disease mechanism at an intimate, subcellular level has a dual benefit. A thorough understanding of the crosstalk involved in molecular pathways both in the normal and the diseased state are expanding scientific knowledge and simultaneously are enabling design cell-targeted and individualized therapies. This paper presents a brief overview of current molecular based treatments available to the orthopedic surgeon and introduces the concept of systemic medicine from the perspective of musculoskeletal pathology.
Collapse
Affiliation(s)
- Kaveh Mashayekhi
- Systems Bioinformatics and Modelling GMBH,39 Basaltstrase D-62540, Tel+4915154924033 Frankfurt, Germany
- BioTalentum Ltd, Aulich Lajos str. 26.2100 Gödöllő, Hungary
- REMEDI National Centre for Biomedical Engineering Science (NCBES), Orbsen Building, NUI Galway, Ireland
| | - Margaret O’Brien
- National Centre for Biomedical Engineering Science (NCBES), Orbsen Building, NUI Galway, Ireland
| | - Florin Zugun-Eloae
- Molecular Genetics and Immunology Department, ”Gr. T. Popa” University of Medicine and Pharmacy, Iasi, Romania, Str. Universitatii nr.16 700115 Iasi, Romania
| | - Luminita Labusca
- Systems Bioinformatics and Modelling GMBH,39 Basaltstrase D-62540, Tel+4915154924033 Frankfurt, Germany
- University Hospital Saint Spiridon Iasi Romania 1st Independentei Boulevard Iasi, Romania
- Address correspondence to this author at the University Hospital Saint Spiridon Iasi Romania 1st Independentei Boulevard Iasi, Romania; Tel: +40749162219; E-mails: ,
| |
Collapse
|
20
|
Watson RS, Broome TA, Levings PP, Rice BL, Kay JD, Smith AD, Gouze E, Gouze JN, Dacanay EA, Hauswirth WW, Nickerson DM, Dark MJ, Colahan PT, Ghivizzani SC. scAAV-mediated gene transfer of interleukin-1-receptor antagonist to synovium and articular cartilage in large mammalian joints. Gene Ther 2012; 20:670-7. [PMID: 23151520 PMCID: PMC3577988 DOI: 10.1038/gt.2012.81] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the long-term goal of developing a gene-based treatment for osteoarthritis (OA), we performed studies to evaluate the equine joint as a model for adeno-associated virus (AAV)-mediated gene transfer to large, weight-bearing human joints. A self-complementary AAV2 vector containing the coding regions for human interleukin-1-receptor antagonist (hIL-1Ra) or green fluorescent protein was packaged in AAV capsid serotypes 1, 2, 5, 8 and 9. Following infection of human and equine synovial fibroblasts in culture, we found that both were only receptive to transduction with AAV1, 2 and 5. For these serotypes, however, transgene expression from the equine cells was consistently at least 10-fold higher. Analyses of AAV surface receptor molecules and intracellular trafficking of vector genomes implicate enhanced viral uptake by the equine cells. Following delivery of 1 × 10(11) vector genomes of serotypes 2, 5 and 8 into the forelimb joints of the horse, all three enabled hIL-1Ra expression at biologically relevant levels and effectively transduced the same cell types, primarily synovial fibroblasts and, to a lesser degree, chondrocytes in articular cartilage. These results provide optimism that AAV vectors can be effectively adapted for gene delivery to large human joints affected by OA.
Collapse
Affiliation(s)
- R S Watson
- Department of Orthopaedics and Rehabilitation, University of Florida, Gainesville, FL 32608-0137, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Zhang Y, Wu C, Luo T, Li S, Cheng X, Miron RJ. Synthesis and inflammatory response of a novel silk fibroin scaffold containing BMP7 adenovirus for bone regeneration. Bone 2012; 51:704-13. [PMID: 22796416 DOI: 10.1016/j.bone.2012.06.029] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 06/15/2012] [Accepted: 06/18/2012] [Indexed: 11/25/2022]
Abstract
Gene therapy has garnished tremendous awareness for the repair of osseous defects. It exhibits high efficiency gene transfer and osteogenic differentiation potential making it well suitable for the sustained delivery of growth factors to local tissues. In the present study a simplified solution-based in situ biomimetic synthesis method is demonstrated for bone morphogenetic protein 7 (BMP7) adenovirus combined with silk fibroin scaffolds. This scaffold not only provides the three dimensional space for bone ingrowth, but also releases the BMP7 adenovirus which targets its secretion by host cells in vivo. Scaffolds were tested both in vitro for their osteogenic potential as well as in vivo in a critical-size calvarial defect in mice. Scaffolds loaded with bone morphogenetic protein 7 adenovirus (adBMP7) were able to sustain release of adBMP7 for up to 21 days and support cell proliferation and differentiation to bone forming osteoblasts. Calvarial defects treated with scaffolds containing adBMP7 significantly induced new bone formation in vivo. To demonstrate immuno-compatibility with host tissues, IL-2, IL-6 and TNF-α were measured up to 4 weeks post-implantation. Although these scaffolds demonstrated an initial pro-inflammatory response, levels of IL-2, IL-6 and TNF-α returned to baseline control values at either 2 or 4 weeks post-implantation demonstrating long term compatibility for growth factor delivery via gene therapy. The results from the present study indicate the promise of gene delivery scaffold systems for robust, low cost, and high quality bone tissue engineering applications.
Collapse
Affiliation(s)
- Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Wuhan University, 237 Luoyu Road, Wuhan 430079, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
22
|
Perera JR, Gikas PD, Bentley G. The present state of treatments for articular cartilage defects in the knee. Ann R Coll Surg Engl 2012; 94:381-7. [PMID: 22943326 PMCID: PMC3954317 DOI: 10.1308/003588412x13171221592573] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2011] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Chondral and osteochondral lesions of the knee are notoriously difficult to treat due to the poor healing capacity of articular cartilage and the hostile environment of moving joints, ultimately causing disabling pain and early osteoarthritis. There are many different reconstructive techniques used currently but few are proven to be of value. However, some have been shown to produce a better repair with hyaline-like cartilage rather than fibrocartilage. METHODS A systematic search of all available online databases including PubMed, MEDLINE(®) and Embase™ was undertaken using several keywords. All the multiple treatment options and methods available were considered. These were summarised, and the evidence for and against them was scrutinised. RESULTS A total of 460 articles were identified after cross-referencing the database searches using the keywords. These revealed that autologous and matrix assisted chondrocyte implantation demonstrated both 'good to excellent' histological results and significant improvement in clinical outcomes. CONCLUSIONS Autologous and matrix assisted chondrocyte implantation have been shown to treat symptomatic lesions successfully with significant histological and clinical improvement. There is, however, still a need for further randomised clinical trials, perfecting the type of scaffold and the use of adjuncts such as growth factors. A list of recommendations for treatment and the potential future trends of managing these lesions are given.
Collapse
Affiliation(s)
- J R Perera
- Royal National Orthopaedic Hospital NHS Trust, Stanmore, Middlesex HA7 4LP, UK.
| | | | | |
Collapse
|
23
|
rAAV Vectors as Safe and Efficient Tools for the Stable Delivery of Genes to Primary Human Chondrosarcoma Cells In Vitro and In Situ. Sarcoma 2012; 2012:347417. [PMID: 22645415 PMCID: PMC3356986 DOI: 10.1155/2012/347417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 02/17/2012] [Accepted: 02/17/2012] [Indexed: 12/11/2022] Open
Abstract
Treatment of chondrosarcoma remains a major challenge in orthopaedic oncology. Gene transfer strategies based on recombinant adenoassociated viral (rAAV) vectors may provide powerful tools to develop new, efficient therapeutic options against these tumors. In the present study, we tested the hypothesis that rAAV is adapted for a stable and safe delivery of foreign sequences in human chondrosarcoma tissue by transducing primary human chondrosarcoma cells in vitro and in situ with different reporter genes (E. coli lacZ, firefly luc, Discosoma sp. RFP). The effects of rAAV administration upon cell survival and metabolic activities were also evaluated to monitor possibly detrimental effects of the gene transfer method. Remarkably, we provide evidence that efficient and prolonged expression of transgene sequences via rAAV can be safely achieved in all the systems investigated, demonstrating the potential of the approach of direct application of therapeutic gene vectors as a means to treat chondrosarcoma.
Collapse
|
24
|
Moran CJ, Barry FP, Maher SA, Shannon FJ, Rodeo SA. Advancing regenerative surgery in orthopaedic sports medicine: the critical role of the surgeon. Am J Sports Med 2012; 40:934-44. [PMID: 22085730 DOI: 10.1177/0363546511426677] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The constant desire to improve outcomes in orthopaedic sports medicine requires us to continuously consider the challenges faced in the surgical repair or reconstruction of soft tissue and cartilaginous injury. In many cases, surgical efforts targeted at restoring normal anatomy and functional status are ultimately impaired by the biological aspect of the natural history of these injuries, which acts as an obstacle to a satisfactory repair process after surgery. The clinical management of sports injuries and the delivery of appropriate surgical intervention are continuously evolving, and it is likely that the principles of regenerative medicine will have an increasing effect in this specialized field of orthopaedic practice going forward. Ongoing advances in arthroscopy and related surgical techniques should facilitate this process. In contrast to the concept of engineered replacement of entire tissues, it is probable that the earliest effect of regenerative strategies seen in clinical practice will involve biological augmentation of current operative techniques via a synergistic process that might be best considered "regenerative surgery." This article provides an overview of the principles of regenerative surgery in cartilage repair and related areas of orthopaedic surgery sports medicine. The possibilities and challenges of a gradual yet potential paradigm shift in treatment through the increased use of biological augmentation are considered. The translational process and critical role to be played by the specialist surgeon are also addressed. We conclude that increased understanding of the potential and challenges of regenerative surgery should allow those specializing in orthopaedic surgery sports medicine to lead the way in advancing the frontiers of biological strategies to enhance modern clinical care in an evidence-based manner.
Collapse
Affiliation(s)
- Cathal J Moran
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland.
| | | | | | | | | |
Collapse
|
25
|
Augmented healing of critical-size calvarial defects by baculovirus-engineered MSCs that persistently express growth factors. Biomaterials 2012; 33:3682-92. [PMID: 22361095 DOI: 10.1016/j.biomaterials.2012.02.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 02/02/2012] [Indexed: 02/08/2023]
Abstract
Repair of large calvarial bony defects remains clinically challenging because successful spontaneous calvarial re-ossification rarely occurs. Although bone marrow-derived mesenchymal stem cells (BMSCs) genetically engineered with baculovirus (BV) for transient expression of osteogenic/angiogenic factors hold promise for bone engineering, we hypothesized that calvarial bone healing necessitates prolonged growth factor expression. Therefore, we employed a hybrid BV vector system whereby one BV expressed FLP while the other harbored the BMP2 (or VEGF) cassette flanked by Frt sequences. Transduction of rabbit BMSCs with the FLP/Frt-based BV vector led to FLP-mediated episome formation, which not only extended the BMP2/VEGF expression beyond 28 days but augmented the BMSCs osteogenesis. After allotransplantation into rabbits, X-ray, PET/CT, μCT and histological analyses demonstrated that the sustained BMP2/VEGF expression remarkably ameliorated the angiogenesis and regeneration of critical-size (8 mm) calvarial defects, when compared with the group implanted with BMSCs transiently expressing BMP2/VEGF. The prolonged expression by BMSCs accelerated the bone remodeling and regenerated the bone through the natural intramembranous pathway, filling ≈83% of the area and ≈63% of the volume in 12 weeks. These data implicated the potential of the hybrid BV vector to engineer BMSCs for sustained BMP2/VEGF expression and the repair of critical-size calvarial defects.
Collapse
|
26
|
Evans CH, Ghivizzani SC, Robbins PD. Orthopedic gene therapy--lost in translation? J Cell Physiol 2012; 227:416-20. [PMID: 21948071 DOI: 10.1002/jcp.23031] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Orthopedic gene therapy has been the topic of considerable research for two decades. The preclinical data are impressive and many orthopedic conditions are well suited to genetic therapies. But there have been few clinical trials and no FDA-approved product exists. This paper examines why this is so. The reasons are multifactorial. Clinical translation is expensive and difficult to fund by traditional academic routes. Because gene therapy is viewed as unsafe and risky, it does not attract major funding from the pharmaceutical industry. Start-up companies are burdened by the complex intellectual property environment and difficulties in dealing with the technology transfer offices of major universities. Successful translation requires close interactions between scientists, clinicians and experts in regulatory and compliance issues. It is difficult to create such a favorable translational environment. Other promising fields of biological therapy have contemplated similar frustrations approximately 20 years after their founding, so there seem to be more general constraints on translation that are difficult to define. Gene therapy has noted some major clinical successes in recent years, and a sense of optimism is returning to the field. We hope that orthopedic applications will benefit collaterally from this upswing and move expeditiously into advanced clinical trials.
Collapse
Affiliation(s)
- C H Evans
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | | | | |
Collapse
|
27
|
Inflammation and immune response of intra-articular serotype 2 adeno-associated virus or adenovirus vectors in a large animal model. ARTHRITIS 2012; 2012:735472. [PMID: 22288012 PMCID: PMC3263587 DOI: 10.1155/2012/735472] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 10/05/2011] [Indexed: 12/02/2022]
Abstract
Intra-articular gene therapy has potential for the treatment of osteoarthritis and rheumatoid arthritis. To quantify in vitro relative gene transduction, equine chondrocytes and synovial cells were treated with adenovirus vectors (Ad), serotype 2 adeno-associated virus vectors (rAAV2), or self-complementary (sc) AAV2 vectors carrying green fluorescent protein (GFP). Using 6 horses, bilateral metacarpophalangeal joints were injected with Ad, rAAV2, or scAAV2 vectors carrying GFP genes to assess the in vivo joint inflammation and neutralizing antibody (NAb) titer in serum and joint fluid. In vitro, the greater transduction efficiency and sustained gene expression were achieved by scAAV2 compared to rAAV2 in equine chondrocytes and synovial cells. In vivo, AAV2 demonstrated less joint inflammation than Ad, but similar NAb titer. The scAAV2 vectors can induce superior gene transduction than rAAV2 in articular cells, and both rAAV2 and scAAV2 vectors were showed to be safer for intra-articular use than Ad vectors.
Collapse
|
28
|
Darabos N, Haspl M, Moser C, Darabos A, Bartolek D, Groenemeyer D. Intraarticular application of autologous conditioned serum (ACS) reduces bone tunnel widening after ACL reconstructive surgery in a randomized controlled trial. Knee Surg Sports Traumatol Arthrosc 2011; 19 Suppl 1:S36-46. [PMID: 21360125 DOI: 10.1007/s00167-011-1458-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2010] [Accepted: 02/14/2011] [Indexed: 01/11/2023]
Abstract
PURPOSE Pro-inflammatory cytokines play a pivotal role in osteoarthritis, as well as in bone tunnel widening after ACL reconstructive surgery. A new treatment option is to administer autologous conditioned serum (ACS) containing endogenous anti-inflammatory cytokines including IL-1Ra and growth factors (IGF-1, PDGF, and TGF-β1, among others) in the liquid blood phase. The purpose of this trial was to establish whether the postoperative outcome could be affected by intraarticular application of ACS. METHODS In a prospective, randomized, double-blinded, placebo-controlled trial with two parallel groups, 62 patients were treated. Bone tunnel width was measured by CT scans, while clinical efficacy was assessed by patient-administered outcome instruments (WOMAC, IKDC 2000) up to 1 year following the ACL reconstruction in patients receiving either ACS (Group A) or placebo (Group B). We compared the levels and dynamics of IL-1β concentrations in the synovial liquid and examined the correlation between the levels of IL-1β at three different postoperative points. RESULTS Bone tunnel enlargement was significantly less (6 months: 8%, 12 months: 13%) in Group A than in Group B (6 months: 31%, 12 months: 38%). Clinical outcomes (WOMAC, IKDC 2000) were consistently better in patients treated with ACS at all data points and for all outcome parameters, and there were statistically significant differences in the WOMAC stiffness subscale after 1 year. The decrease in IL-1β synovial fluid concentration was more pronounced in the ACS group, and values were lower, to a statistically significant degree, in the ACS group at day 10. CONCLUSION The intraarticular administration/injection of ACS results in decreased bone tunnel widening after ACL reconstructive surgery.
Collapse
Affiliation(s)
- Nikica Darabos
- University Clinic for Traumatology, Medical School, University of Zagreb, Draskoviceva 19, 10000, Zagreb, Croatia.
| | | | | | | | | | | |
Collapse
|
29
|
Qiu L, Zhang L, Wang L, Jiang Y, Luo Y, Peng Y, Lin L. Ultrasound-targeted microbubble destruction enhances naked plasmid DNA transfection in rabbit Achilles tendons in vivo. Gene Ther 2011; 19:703-10. [DOI: 10.1038/gt.2011.165] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
30
|
Wagner DE, Bhaduri SB. Progress and outlook of inorganic nanoparticles for delivery of nucleic acid sequences related to orthopedic pathologies: a review. TISSUE ENGINEERING PART B-REVIEWS 2011; 18:1-14. [PMID: 21707439 DOI: 10.1089/ten.teb.2011.0081] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The anticipated growth in the aging population will drastically increase medical needs of society; of which, one of the largest components will undoubtedly be from orthopedic-related pathologies. There are several proposed solutions being investigated to cost-effectively prepare for the future--pharmaceuticals, implant devices, cell and gene therapies, or some combination thereof. Gene therapy is one of the more promising possibilities because it seeks to correct the root of the problem, thereby minimizing treatment duration and cost. Currently, viral vectors have shown the highest efficacies, but immunological concerns remain. Nonviral methods show reduced immune responses but are regarded as less efficient. The nonviral paradigms consist of mechanical and chemical approaches. While organic-based materials have been used more frequently in particle-based methods, inorganic materials capable of delivery have distinct advantages, especially advantageous in orthopedic applications. The inorganic gene therapy field is highly interdisciplinary in nature, and requires assimilation of knowledge across the broad fields of cell biology, biochemistry, molecular genetics, materials science, and clinical medicine. This review provides an overview of the role each area plays in orthopedic gene therapy as well as possible future directions for the field.
Collapse
Affiliation(s)
- Darcy E Wagner
- Department of Biomedical Engineering, Colleges of Medicine and Engineering, University of Toledo, Toledo, Ohio 43606, USA.
| | | |
Collapse
|
31
|
The role of adipose-derived stem cells engineered with the persistently expressing hybrid baculovirus in the healing of massive bone defects. Biomaterials 2011; 32:6505-14. [DOI: 10.1016/j.biomaterials.2011.05.059] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 05/19/2011] [Indexed: 01/01/2023]
|
32
|
Abstract
Light-activated gene transduction (LAGT) is an approach to localize gene therapy via preactivation of cells with UV light, which facilitates transduction by recombinant adeno-associated virus vectors. Prior studies demonstrated that UVC induces LAGT secondary to pyrimidine dimer formation, while UVA induces LAGT secondary to reactive oxygen species (ROS) generation. However, the empirical UVB boundary of these UV effects is unknown. Thus, we aimed to define the action spectra for UV-induced LAGT independent of DNA damage, and determine an optimal wavelength to maximize safety and efficacy. Results: UV at 288, 311 and 320nm produced significant dose-dependent LAGT effects, of which the maximum (800-fold) was observed with 4kJ/m2 at 311nm. Consistent with its robust cytotoxicity, 288nm produced significantly high levels of DNA damage at all doses tested, while 311, 320 and 330nm did not generate pyrimidine dimers and produced low levels of DNA damage detected by comet assay. While 288nm failed to induce ROS, the other wavelengths were effective, with the maximum (10-fold) effect observed with 30 kJ/m2 at 311nm. An in vivo pilot study assessing 311nm-induced LAGT of rabbit articular chondrocytes demonstrated a significant 6.6-fold (p<0.05) increase in transduction with insignificant cytotoxicity. Conclusion: 311nm was found to be the optimal wavelength for LAGT based on its superior efficacy at the peak dose, and its broad safety range that is remarkably wider than the other UV wavelengths tested.
Collapse
|
33
|
Shi Q, Zhang XL, Dai KR, Benderdour M, Fernandes JC. siRNA therapy for cancer and non-lethal diseases such as arthritis and osteoporosis. Expert Opin Biol Ther 2010; 11:5-16. [DOI: 10.1517/14712598.2010.532483] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
34
|
Okabayashi T, Nakanishi K, Tsuchihara T, Arino H, Yoshihara Y, Tominaga S, Uenoyama M, Suzuki S, Asagiri M, Nemoto K. Axonal-transport-mediated gene transduction in the interior of rat bone. PLoS One 2010; 5. [PMID: 20927397 PMCID: PMC2946924 DOI: 10.1371/journal.pone.0013034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 09/06/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Gene transduction has been considered advantageous for the sustained delivery of proteins to specific target tissues. However, in the case of hard tissues, such as bone, local gene delivery remains problematic owing to anatomical accessibility limitations of the target sites. METHODOLOGY/PRINCIPAL FINDINGS Here, we evaluated the feasibility of exogenous gene transduction in the interior of bone via axonal transport following intramuscular administration of a nonviral vector. A high expression level of the transduced gene was achieved in the tibia ipsilateral to the injected tibialis anterior muscle, as well as in the ipsilateral sciatic nerve and dorsal root ganglia. In sciatic transection rats, the gene expression level was significantly lowered in bone. CONCLUSIONS/SIGNIFICANCE These results suggest that axonal transport is critical for gene transduction. Our study may provide a basis for developing therapeutic methods for efficient gene delivery into hard tissues.
Collapse
Affiliation(s)
- Toshitaka Okabayashi
- Department of Orthopaedic Surgery, National Defense Medical College, Saitama, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Moran CJ, Shannon FJ, Barry FP, O’Byrne JM, O’Brien T, Curtin W. Translation of science to surgery. ACTA ACUST UNITED AC 2010; 92:1195-202. [DOI: 10.1302/0301-620x.92b9.23651] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Orthopaedic surgery is in an exciting transitional period as modern surgical interventions, implants and scientific developments are providing new therapeutic options. As advances in basic science and technology improve our understanding of the pathology and repair of musculoskeletal tissue, traditional operations may be replaced by newer, less invasive procedures which are more appropriately targeted at the underlying pathophysiology. However, evidence-based practice will remain a basic requirement of care. Orthopaedic surgeons can and should remain at the forefront of the development of novel therapeutic interventions and their application. Progression of the potential of bench research into an improved array of orthopaedic treatments in an effective yet safe manner will require the development of a subgroup of specialists with extended training in research to play an important role in bridging the gap between laboratory science and clinical practice. International regulations regarding the introduction of new biological treatments will place an additional burden on the mechanisms of this translational process, and orthopaedic surgeons who are trained in science, surgery and the regulatory environment will be essential. Training and supporting individuals with these skills requires special consideration and discussion by the orthopaedic community. In this paper we review some traditional approaches to the integration of orthopaedic science and surgery, the therapeutic potential of current regenerative biomedical science for cartilage repair and ways in which we may develop surgeons with the skills required to translate scientific discovery into effective and properly assessed orthopaedic treatments.
Collapse
Affiliation(s)
- C. J. Moran
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| | - F. J. Shannon
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| | - F. P. Barry
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| | - J. M. O’Byrne
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| | - T. O’Brien
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| | - W. Curtin
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Sciences, National University of Ireland, University Road, Galway, Ireland
| |
Collapse
|
36
|
Abstract
"Gene doping" is the term used to describe the potential abuse of gene therapy as a performance-enhancing agent. Gene doping would apply the techniques used in gene therapy to provide altered expression of genes that would promote physical superiority. For example, insulin-like growth factor 1 (IGF-1) is a primary target for growth hormone; overexpression of IGF-1 can lead to increased muscle mass and power. Although gene doping is still largely theoretical, its implications for sports, health, ethics, and medical genetics are significant.
Collapse
|
37
|
Comparative efficacy of dermal fibroblast-mediated and direct adenoviral bone morphogenetic protein-2 gene therapy for bone regeneration in an equine rib model. Gene Ther 2010; 17:733-44. [DOI: 10.1038/gt.2010.13] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
38
|
Abstract
Traumatic bone defects and nonunion represent a significant source of morbidity and socioeconomic burden in trauma patients. The treatment of these conditions is currently hampered by inadequate therapies. This has prompted a new era of investigation into biologic therapies for augmenting fracture healing. Within this body of research, gene therapy has arisen as a novel and effective method of delivering therapeutic proteins at a site of desired bone regeneration. Gene therapy has shown tremendous potential in preclinical studies of fracture healing, but to date, no clinical trials have occurred. This article reviews the scientific basis for gene therapy in fracture healing, provides an overview of important preclinical studies that have been performed to date, and discusses the current barriers and future directions of gene therapy as it applies to fracture healing.
Collapse
|
39
|
Ishihara A, Zekas LJ, Litsky AS, Weisbrode SE, Bertone AL. Dermal fibroblast-mediated BMP2 therapy to accelerate bone healing in an equine osteotomy model. J Orthop Res 2010; 28:403-11. [PMID: 19777486 DOI: 10.1002/jor.20978] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This study evaluated healing of equine metacarpal/metatarsal osteotomies in response to percutaneous injection of autologous dermal fibroblasts (DFbs) genetically engineered to secrete bone morphogenetic protein-2 (BMP2) or demonstrate green fluorescent protein (GFP) gene expression administered 14 days after surgery. Radiographic assessment of bone formation indicated greater and earlier healing of bone defects treated with DFb with BMP2 gene augmentation. Quantitative computed tomography and biomechanical testing revealed greater mineralized callus and torsional strength of DFb-BMP2-treated bone defects. On the histologic evaluation, the bone defects with DFb-BMP2 implantation had greater formation of mature cartilage and bone nodules within the osteotomy gap and greater mineralization activity on osteotomy edges. Autologous DFbs were successfully isolated in high numbers by a skin biopsy, rapidly expanded without fastidious culture techniques, permissive to adenoviral vectors, and efficient at in vitro BMP2 protein production and BMP2-induced osteogenic differentiation. This study demonstrated an efficacy and feasibility of DFb-mediated BMP2 therapy to accelerate the healing of osteotomies. Skin cell-mediated BMP2 therapy may be considered as a potential treatment for various types of fractures and bone defects.
Collapse
Affiliation(s)
- Akikazu Ishihara
- Comparative Orthopedic Research Laboratories, Department of Veterinary Clinical Sciences, The Ohio State University, 601 Tharp Street, Columbus, Ohio 43210, USA
| | | | | | | | | |
Collapse
|
40
|
Lin CY, Chang YH, Lin KJ, Yen TC, Tai CL, Chen CY, Lo WH, Hsiao IT, Hu YC. The healing of critical-sized femoral segmental bone defects in rabbits using baculovirus-engineered mesenchymal stem cells. Biomaterials 2010; 31:3222-30. [PMID: 20144476 DOI: 10.1016/j.biomaterials.2010.01.030] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2009] [Accepted: 01/09/2010] [Indexed: 12/22/2022]
Abstract
Management of massive segmental bone defects remains a challenging clinical problem and bone marrow-derived mesenchymal stem cells (BMSCs) hold promise for bone regeneration. To explore whether BMSCs engineered by baculovirus (an emerging gene delivery vector) can heal large bone defects, New Zealand White (NZW) rabbit BMSCs were transduced with the BMP2-expressing baculovirus or VEGF-expressing baculovirus, and co-implanted into critical-sized (10mm) femoral segmental defects in NZW rabbits. X-ray analysis revealed that the baculovirus-engineered BMSCs not only bridged the defects at as early as week 2, but also healed the defects in 100% of rabbits (13/13) at week 4. The osteogenic metabolism, as monitored by positron emission tomography (PET) also suggested the completion of bone healing at week 8. When compared with other control groups, the BMP2/VEGF-expressing BMSCs remarkably enhanced the segmental bone repair and mechanical properties, as evidenced by micro-computed tomography (microCT), histochemical staining and biomechanical testing. The ameliorated bone healing concurred with the augmented angiogenesis. These data demonstrated, that BMSCs engineered to express BMP2 and VEGF accelerate the repair of large femoral bone defects and improve the quality of the regenerated bone, which paves an avenue to utilizing baculovirus as a vector for BMSCs modification and regenerative medicine.
Collapse
Affiliation(s)
- Chin-Yu Lin
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chuang CK, Lin KJ, Lin CY, Chang YH, Yen TC, Hwang SM, Sung LY, Chen HC, Hu YC. Xenotransplantation of Human Mesenchymal Stem Cells into Immunocompetent Rats for Calvarial Bone Repair. Tissue Eng Part A 2010; 16:479-88. [DOI: 10.1089/ten.tea.2009.0401] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ching-Kuang Chuang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medical Imaging, Chang Gung University, Taoyuan, Taiwan
| | - Chin-Yu Lin
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Han Chang
- Department of Orthopaedic, Chang Gung University, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tzu-Chen Yen
- Department of Nuclear Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medical Imaging, Chang Gung University, Taoyuan, Taiwan
| | - Shiaw-Min Hwang
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan
| | - Li-Yu Sung
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Huang-Chi Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
42
|
Abstract
The clinical augmentation of bone currently involves the use of autogenous or allogeneic bone grafts and synthetic materials, all of which are associated with limitations. Research on the safe enhancement of bone formation concerns the potential value of scaffolds, stem cells, gene therapy, and chemical and mechanical signals. Optimal scaffolds are engineered to provide mechanical stability while supporting osteogenesis, osteoconduction and/or osteoinduction. Scaffold materials include natural or synthetic polymers, ceramics, and composites. The resorption, mechanical strength and efficacy of these materials can be manipulated through structural and chemical design parameters. Cell-seeded scaffolds contain stem cells or progenitor cells, such as culture-expanded marrow stromal cells and multipotent skeletal progenitor cells sourced from other tissues. Despite extensive evidence from proof-of-principle studies, bone tissue engineering has not translated to clinical practice. Much of the research involves in vitro and animal models that do not replicate potential clinical applications. Problem areas include cell sources and numbers, over-reliance on existing scaffold materials, optimum delivery of factors, control of transgene expression, vascularization, integration with host bone, and the capacity to form bone and marrow structures in vivo. Current thinking re-emphasizes the potential of biomimetic materials to stimulate, enhance, or control bone's innate regenerative capacity at the implantation site.
Collapse
Affiliation(s)
- Ericka M Bueno
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
43
|
Evans CH, Ghivizzani SC, Robbins PD. Progress and Prospects: genetic treatments for disorders of bones and joints. Gene Ther 2009; 16:944-52. [DOI: 10.1038/gt.2009.73] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
44
|
Baculovirus transduction of mesenchymal stem cells triggers the toll-like receptor 3 pathway. J Virol 2009; 83:10548-56. [PMID: 19656899 DOI: 10.1128/jvi.01250-09] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) can be genetically modified with viral vectors and hold promise as a cell source for regenerative medicine, yet how hMSCs respond to viral vector transduction remains poorly understood, leaving the safety concerns unaddressed. Here, we explored the responses of hMSCs against an emerging DNA viral vector, baculovirus (BV), and discovered that BV transduction perturbed the transcription of 816 genes associated with five signaling pathways. Surprisingly, Toll-like receptor-3 (TLR3), a receptor that generally recognizes double-stranded RNA, was apparently upregulated by BV transduction, as confirmed by microarray, PCR array, flow cytometry, and confocal microscopy. Cytokine array data showed that BV transduction triggered robust secretion of interleukin-6 (IL-6) and IL-8 but not of other inflammatory cytokines and beta interferon (IFN-beta). BV transduction activated the signaling molecules (e.g., Toll/interleukin-1 receptor domain-containing adaptor-inducing IFN-beta, NF-kappaB, and IFN regulatory factor 3) downstream of TLR3, while silencing the TLR3 gene with small interfering RNA considerably abolished cytokine expression and promoted cell migration. These data demonstrate, for the first time, that a DNA viral vector can activate the TLR3 pathway in hMSCs and lead to a cytokine expression profile distinct from that in immune cells. These findings underscore the importance of evaluating whether the TLR3 signaling cascade plays roles in the immune response provoked by other DNA vectors (e.g., adenovirus). Nonetheless, BV transduction barely disturbed surface marker expression and induced only transient and mild cytokine responses, thereby easing the safety concerns of using BV for hMSCs engineering.
Collapse
|
45
|
|