1
|
Abstract
Mesenchymal stromal cell (MSC) therapy has produced very promising results for multiple diseases in animal models, with over 780 clinical trials on going or completed. However, most of the human clinical trials have not been as successful as trials using preclinical models. To improve the therapeutic potential of MSCs, different research groups have used gene transfer vectors to express factors involved in migration, survival, differentiation, and immunomodulation. The ideal gene transfer vector for most applications should achieve long-term, stable (constitutive or inducible) transgene expression in MSCs and their progeny. Given their efficiency and low impact on transduced cells, lentiviral vectors (LVs) are the vectors of choice. In this chapter we will describe a detailed protocol for the generation of genetically modified MSCs using lentiviral vectors (LVs). Although this protocol has been optimized for MSC lentiviral transduction, it can be easily adapted to other stem cells by changing culture conditions while maintaining volumes and incubation times.
Collapse
|
2
|
Eltorai AEM, Susai CJ, Daniels AH. Mesenchymal stromal cells in spinal fusion: Current and future applications. J Orthop 2016; 14:1-3. [PMID: 27821993 DOI: 10.1016/j.jor.2016.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/13/2016] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been a promising area of study for regenerative medicine. These cells can be harvested from bone marrow, adipose tissue, and other areas allowing for autologous transplantation of these cells into the area of degeneration or injury. With the proper signals, these cells may be able to regenerate healthy tissue. Recent studies have yielded promising evidence supporting translational mesenchymal stromal cell applications particularly in spinal fusion surgery.
Collapse
Affiliation(s)
- Adam E M Eltorai
- Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Cynthia J Susai
- Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Alan H Daniels
- Division of Spine Surgery, Department of Orthopaedic Surgery, Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
3
|
Ding Y, Ruan D, Luk KDK, He Q, Wang C. The effect of gamma irradiation on the biological properties of intervertebral disc allografts: in vitro and in vivo studies in a beagle model. PLoS One 2014; 9:e100304. [PMID: 24959720 PMCID: PMC4069010 DOI: 10.1371/journal.pone.0100304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 04/22/2014] [Indexed: 12/26/2022] Open
Abstract
Study Design An animal experiment about intervertebral disc allograft. Objective To explore the feasibility to decellularize disc allografts treated by 6°Co Gamma Irradiation, and simultaneously, to assess the possibility to make use of the decellularized natural disc scaffold for disc degeneration biotherapy. Summary of Background Data Studies of both animal and human disc allograft transplantation indicated that the disc allograft may serve as a scaffold to undertake the physiological responsibility of the segment. Methods Experiment in vitro: 48 discs of beagles were harvested and divided randomly into four groups including a control group and three irradiated groups. Immediate cell viability and biomechanical properties of the discs were checked and comparisons were made among these groups. Experiment in vivo: 24 beagles accepted single-level allografted disc treated with different doses of gamma irradiation. Plain X-rays and MRIs were taken before and after surgery. Then, the spinal columns were harvested en bloc from the sacrificed beagles and were examined morphologically. Results There were significant differences of both the annulus fibrosus and nucleus pulposus immediate cell viabilities among the various groups. There were no obvious differences of the biomechanical properties among the four groups. The disc height and range of motion decreased significantly in all groups as time went on. The observed indexes in irradiated groups were much smaller than those in the control group, but the indexes in 18-kGy group were larger than those in 25-kGy and 50-kGy groups. Both MRI and macroscopic findings showed that the segmental degeneration in the control and 18-kGy group was less severe than that in 25-kGy and 50-kGy groups. Conclusion Gamma Irradiation can decellularize disc allograft successfully to provide natural scaffold for the study of degenerative disc disease therapy, and also can be used as an effective method to produce adjustable animal models.
Collapse
Affiliation(s)
- Yu Ding
- Department of Rehabilitation Medicine and Pain Management Center, Navy General Hospital, Beijing, China
| | - Dike Ruan
- Department of Orthopaedics, Navy General Hospital, Beijing, China
- * E-mail:
| | - Keith D. K. Luk
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Qing He
- Department of Orthopaedics, Navy General Hospital, Beijing, China
| | - Chaofeng Wang
- Department of Orthopaedics, Navy General Hospital, Beijing, China
| |
Collapse
|
4
|
Wegman F, van der Helm Y, Öner FC, Dhert WJ, Alblas J. Bone Morphogenetic Protein-2 Plasmid DNA as a Substitute for Bone Morphogenetic Protein-2 Protein in Bone Tissue Engineering. Tissue Eng Part A 2013; 19:2686-92. [DOI: 10.1089/ten.tea.2012.0569] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Fiona Wegman
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Yvonne van der Helm
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F. Cumhur Öner
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wouter J.A. Dhert
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jacqueline Alblas
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
5
|
Lathuilière A, Cosson S, Lutolf MP, Schneider BL, Aebischer P. A high-capacity cell macroencapsulation system supporting the long-term survival of genetically engineered allogeneic cells. Biomaterials 2013; 35:779-91. [PMID: 24103654 DOI: 10.1016/j.biomaterials.2013.09.071] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 09/20/2013] [Indexed: 11/27/2022]
Abstract
The rapid increase in the number of approved therapeutic proteins, including recombinant antibodies, for diseases necessitating chronic treatments raises the question of the overall costs imposed on healthcare systems. It is therefore important to investigate alternative methods for recombinant protein administration. The implantation of genetically engineered cells is an attractive strategy for the chronic long-term delivery of recombinant proteins. Here, we have developed a high-capacity cell encapsulation system for the implantation of allogeneic myoblasts, which survive at high density for at least one year. This flat sheet device is based on permeable polypropylene membranes sealed to a mechanically resistant frame which confine cells seeded in a tailored biomimetic poly(ethylene glycol) (PEG)-based hydrogel matrix. In order to quantitate the number of cells surviving in the device and optimize initial conditions leading to high-density survival, we implant devices containing C2C12 mouse myoblasts expressing a luciferase reporter in the mouse subcutaneous tissue. We show that initial cell load, hydrogel stiffness and permeable membrane porosity are critical parameters to achieve long-term implant survival and efficacy. Optimization of these parameters leads to the survival of encapsulated myogenic cells at high density for several months, with minimal inflammatory response and dense neovascularization in the adjacent host tissue. Therefore, this encapsulation system is an effective platform for the implantation of genetically engineered cells in allogeneic conditions, which could be adapted to the chronic administration of recombinant proteins.
Collapse
Affiliation(s)
- Aurélien Lathuilière
- Neurodegenerative Studies Laboratory, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
6
|
Evans NR, Davies EM, Dare CJ, Oreffo RO. Tissue engineering strategies in spinal arthrodesis: the clinical imperative and challenges to clinical translation. Regen Med 2013; 8:49-64. [PMID: 23259805 DOI: 10.2217/rme.12.106] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Skeletal disorders requiring the regeneration or de novo production of bone present considerable reconstructive challenges and are one of the main driving forces for the development of skeletal tissue engineering strategies. The skeletal or mesenchymal stem cell is a fundamental requirement for osteogenesis and plays a pivotal role in the design and application of these strategies. Research activity has focused on incorporating the biological role of the mesenchymal stem cell with the developing fields of material science and gene therapy in order to create a construct that is not only capable of inducing host osteoblasts to produce bone, but is also osteogenic in its own right. This review explores the clinical need for reparative approaches in spinal arthrodesis, identifying recent tissue engineering strategies employed to promote spinal fusion, and considers the ongoing challenges to successful clinical translation.
Collapse
Affiliation(s)
- Nick R Evans
- Bone & Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Human Development & Health, Institute of Developmental Sciences, Southampton General Hospital, Southampton, UK.
| | | | | | | |
Collapse
|
7
|
Salehinejad P, Alitheen NB, Nematollahi-Mahani SN, Ali AM, Omar AR, Janzamin E, Hajghani M. Effect of culture media on expansion properties of human umbilical cord matrix-derived mesenchymal cells. Cytotherapy 2012; 14:948-53. [PMID: 22587592 DOI: 10.3109/14653249.2012.684377] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSC) have been isolated from a number of different tissues, including umbilical cord. Because of the lack of a uniform approach to human umbilical cord matrix-derived mesenchymal (hUCM) cell expansion, we attempted to identify the optimum conditions for the production of a high quantity of hUCM cells by comparing two media. METHODS We compared the ability of Dulbecco's Modified Eagle's Medium/F12 (DMEM/F12) and Alpha Minimum Essential Medium (α-MEM) with Glutamax (GL) (α-MEM/GL) to expand hUCM cells. For this purpose, hUCM cells were cultured in plates containing different culture media supplemented with 10% fetal bovine serum (FBS). Culture dishes were left undisturbed for 10-14 days to allow propagation of the newly formed hUCM cells. The expansion properties, CD marker expression, differentiation potential, population doubling time (PDT) and cell activity were compared between the two groups. RESULTS The hUCM cells harvested from each group were positive for MSC markers, including CD44, CD90 and CD105, while they were negative for the hematopoietic cell surface marker CD34. Differentiation into adipogenic and osteogenic lineages was confirmed for both treatments. Cell activity was higher in the α-MEM/GL group than the DMEM/F12 group. PDT was calculated to be 60 h for the DMEM/F12 group, while for the α-MEM/GL group it was 47 h. CONCLUSIONS Our data reveal that α-MEM/GL with 10% FBS supports hUCM cell growth more strongly than DMEM/F12 with 10% FBS.
Collapse
Affiliation(s)
- Parvin Salehinejad
- Institute of Bioscience, University Putra Malaysia, Kuala Lumpur, Malaysia
| | | | | | | | | | | | | |
Collapse
|
8
|
Sheyn D, Kallai I, Tawackoli W, Cohn Yakubovich D, Oh A, Su S, Da X, Lavi A, Kimelman-Bleich N, Zilberman Y, Li N, Bae H, Gazit Z, Pelled G, Gazit D. Gene-modified adult stem cells regenerate vertebral bone defect in a rat model. Mol Pharm 2011; 8:1592-601. [PMID: 21834548 DOI: 10.1021/mp200226c] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vertebral compression fractures (VCFs), the most common fragility fractures, account for approximately 700,000 injuries per year. Since open surgery involves morbidity and implant failure in the osteoporotic patient population, a new minimally invasive biological solution to vertebral bone repair is needed. Previously, we showed that adipose-derived stem cells (ASCs) overexpressing a BMP gene are capable of inducing spinal fusion in vivo. We hypothesized that a direct injection of ASCs, designed to transiently overexpress rhBMP6, into a vertebral bone void defect would accelerate bone regeneration. Porcine ASCs were isolated and labeled with lentiviral vectors that encode for the reporter gene luciferase (Luc) under constitutive (ubiquitin) or inductive (osteocalcin) promoters. The ASCs were first labeled with reporter genes and then nucleofected with an rhBMP6-encoding plasmid. Twenty-four hours later, bone void defects were created in the coccygeal vertebrae of nude rats. The ASC-BMP6 cells were suspended in fibrin gel (FG) and injected into the bone void. A control group was injected with FG alone. The regenerative process was monitored in vivo using microCT, and cell survival and differentiation were monitored using tissue specific reporter genes and bioluminescence imaging (BLI). The surgically treated vertebrae were harvested after 12 weeks and subjected to histological and immunohistochemical (against porcine vimentin) analyses. In vivo BLI detected Luc-expressing cells at the implantation site over a 12-week period. Beginning 2 weeks postoperatively, considerable defect repair was observed in the group treated with ASC-BMP6 cells. The rate of bone formation in the stem cell-treated group was two times faster than that in the FG-treated group, and bone volume at the end point was 2-fold compared to the control group. Twelve weeks after cell injection the bone volume within the void reached the volume measured in native vertebrae. Immunostaining against porcine vimentin indicated that the ASC-BMP6 cells contributed to new bone formation. Here we show the potential of injections of BMP-modified ASCs to repair vertebral bone defects in a rat model. Our results could pave the way to a novel approach for the biological treatment of traumatic and osteoporosis-related vertebral bone injuries.
Collapse
Affiliation(s)
- Dmitriy Sheyn
- Department of Surgery and Cedars-Sinai Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Tang ZB, Cao JK, Wen N, Wang HB, Zhang ZW, Liu ZQ, Zhou J, Duan CM, Cui FZ, Wang CY. Posterolateral spinal fusion with nano-hydroxyapatite-collagen/PLA composite and autologous adipose-derived mesenchymal stem cells in a rabbit model. J Tissue Eng Regen Med 2011; 6:325-36. [PMID: 21751422 DOI: 10.1002/term.445] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2010] [Accepted: 05/12/2011] [Indexed: 11/10/2022]
Abstract
Spinal fusion is routinely performed to treat low back pain caused by degeneration of intervertebral discs. An autologous bone graft derived from the iliac crest is the standard procedure used for spinal fusion. However, several shortcomings, including pseudarthrosis, pain and the need for blood transfusion are known to be associated with the procedure. Our study analysed the effectiveness of a new mineralized collagen matrix, nano-hydroxyapatite-collagen-polylactic acid (nHAC-PLA), combined with autologous adipose-derived mesenchymal stem cells (ADMSCs) as a graft material for posterolateral spinal fusion in a rabbit model. Forty rabbits were randomly divided into four groups: autologous iliac crest bone group (ACB), nHAC-PLA composite group (nHAC-PLA), autologous iliac crest bone mixed with nHAC-PLA composite group (ACB + nHAC-PLA), and nHAC-PLA composite combined with ADMSCs (ADMSCs + nHAC-PLA). The viability and the proliferation of the ADMSCs seeded on the scaffolds were evaluated by live/dead kit and MTT assay in vitro, respectively. Lumbar posterolateral fusions were assessed by manual palpation, radiographical and histological procedures, mechanical strength and scanning electronic microscopy (SEM) in 10 weeks of observation. The results showed that the rate of fusion was significantly higher in the ACB and ADMSCs + nHAC-PLA groups than that in the nHAC-PLA and ACB + nHAC-PLA groups. It was not significantly higher in the ACB group than in the ADMSCs + nHAC-PLA group. From microstructural analysis of the samples using histological staining methods, there was more new bone-like tissue formation in the ACB and ADMSCs + nHAC-PLA groups than that in the other two groups at the 10th postoperative week. Our study demonstrated the effective impact of nHAC-PLA combined with ADMSCs in rabbit posterolateral spinal fusion.
Collapse
Affiliation(s)
- Zi-Bin Tang
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Centre, Academy of Military Medical Sciences, 27 Taiping Road, Beijing 100850, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Olabisi RM, Lazard Z, Heggeness MH, Moran KM, Hipp JA, Dewan AK, Davis AR, West JL, Olmsted-Davis EA. An injectable method for noninvasive spine fusion. Spine J 2011; 11:545-56. [PMID: 21292563 PMCID: PMC3327508 DOI: 10.1016/j.spinee.2010.12.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Revised: 12/01/2010] [Accepted: 12/17/2010] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Bone morphogenetic proteins (BMPs) induce bone formation but are difficult to localize, and subsequent diffusion from the site of interest and short half-life reduce the efficacy of the protein. Currently, spine fusion requires stripping, decortications of the transverse processes, and an autograft harvest procedure. Even in combination with BMPs, clinical spinal fusion has a high failure rate, presumably because of difficulties in localizing sufficient levels of BMP. PURPOSE The goal was to achieve reliable spine fusion through a single injection of a cell-based gene therapy system without the need for any surgical intervention. STUDY DESIGN Eighty-seven immunodeficient (n=44) and immune-competent (n=43) mice were injected along the paraspinous musculature to achieve rapid induction of heterotopic ossification (HO) and ultimately spinal arthrodesis. METHODS Immunodeficient and immune-competent mice were injected with fibroblasts, transduced with an adenoviral vector to express BMP2, along the paraspinous musculature. Bone formation was evaluated via radiographs, microcomputed tomography, and biomechanical analysis. RESULTS ew bridging bone between the vertebrae and the fusion to adjacent skeletal bone was obtained as early as 2 weeks. Reduction in spine flexion-extension also occurred as early as 2 weeks after injection of the gene therapy system, with greater than 90% fusion by 4 weeks in all animals regardless of their genetic background. CONCLUSIONS Injection of our cell-based system into the paraspinous musculature induces spinal fusion that is dependent neither on the cell type nor on the immune status. These studies are the first to harness HO in an immune-competent model as a noninvasive injectable system for clinically relevant spinal fusion and may one day impact human spinal arthrodesis.
Collapse
Affiliation(s)
- Ronke M. Olabisi
- Department of Bioengineering, Rice University, MS 142, 6100 Main St, Houston, TX 77005, USA
| | - ZaWaunyka Lazard
- Center for Cell and Gene Therapy, Baylor College of Medicine, Alkek Graduate School BCMN-N1010, One Baylor Plaza, Houston, TX 77030, USA
| | - Michael H. Heggeness
- Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA
| | - Kevin M. Moran
- Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA
| | - John A. Hipp
- Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA
| | - Ashvin K. Dewan
- Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA
| | - Alan R. Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Alkek Graduate School BCMN-N1010, One Baylor Plaza, Houston, TX 77030, USA,Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jennifer L. West
- Department of Bioengineering, Rice University, MS 142, 6100 Main St, Houston, TX 77005, USA
| | - Elizabeth A. Olmsted-Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Alkek Graduate School BCMN-N1010, One Baylor Plaza, Houston, TX 77030, USA,Department of Orthopaedic Surgery, Baylor College of Medicine, Medical Towers BCM615, One Baylor Plaza, Houston, TX 77030, USA,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA,Corresponding author. Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, MS BCM505, Houston, TX 77030, USA. Tel.: (713) 798-1253; fax: (713) 798-1230. (E.A. Olmsted-Davis)
| |
Collapse
|
11
|
Meyerrose T, Olson S, Pontow S, Kalomoiris S, Jung Y, Annett G, Bauer G, Nolta JA. Mesenchymal stem cells for the sustained in vivo delivery of bioactive factors. Adv Drug Deliv Rev 2010; 62:1167-74. [PMID: 20920540 DOI: 10.1016/j.addr.2010.09.013] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 09/24/2010] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSC) are a promising tool for cell therapy, either through direct contribution to the repair of bone, tendon and cartilage or as an adjunct therapy through protein production and immune mediation. They are an attractive vehicle for cellular therapies due to a variety of cell intrinsic and environmentally responsive properties. Following transplantation, MSC are capable of systemic migration, are not prone to tumor formation, and appear to tolerize the immune response across donor mismatch. These attributes combine to allow MSC to reside in many different tissue types without disrupting the local microenvironment and, in some cases, responding to the local environment with appropriate protein secretion. We describe work done by our group and others in using human MSC for the sustained in vivo production of supraphysiological levels of cytokines for the support of cotransplanted hematopoietic stem cells and enzymes that are deficient in animal models of lysosomal storage disorders such as MPSVII. In addition, the use of MSC engineered to secrete protein products has been reviewed in several fields of tissue injury repair, including but not limited to revascularization after myocardial infarction, regeneration of intervertebral disc defects and spine therapy, repair of stroke, therapy for epilepsy, skeletal tissue repair, chondrogenesis/knee and joint repair, and neurodegenerative diseases. Genetically engineered MSC have thus proven safe and efficacious in numerous animal models of disease modification and tissue repair and are poised to be tested in human clinical trials. The potential for these interesting cells to secrete endogenous or transgene products in a sustained and long-term manner is highly promising and is discussed in the current review.
Collapse
|
12
|
Seo HS, Jung JK, Lim MH, Hyun DK, Oh NS, Yoon SH. Evaluation of Spinal Fusion Using Bone Marrow Derived Mesenchymal Stem Cells with or without Fibroblast Growth Factor-4. J Korean Neurosurg Soc 2009; 46:397-402. [PMID: 19893733 DOI: 10.3340/jkns.2009.46.4.397] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2009] [Revised: 06/24/2009] [Accepted: 10/15/2009] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVE In this study, the authors assessed the ability of rat bone marrow derived mesenchymal stem cells (BMDMSCs), in the presence of a growth factor, fibroblast growth factor-4 (FGF-4) and hydroxyapatite, to act as a scaffold for posterolateral spinal fusion in a rat model. METHODS Using a rat posterolateral spine fusion model, the experimental study comprised 3 groups. Group 1 was composed of 6 animals that were implanted with 0.08 gram hydroxyapatite only. Group 2 was composed of 6 animals that were implanted with 0.08 gram hydroxyapatite containing 1 x 10(6)/ 60 microL rat of BMDMSCs. Group 3 was composed of 6 animals that were implanted with 0.08 gram hydroxyapatite containing 1 x 10(6)/ 60 microL of rat BMDMSCs and FGF-4 1 microG to induce the bony differentiation of the BMDMSCs. Rats were assessed using radiographs obtained at 4, 6, and 8 weeks postoperatively. After sacrifice, spines were explanted and assessed by manual palpation, high-resolution microcomputerized tomography, and histological analysis. RESULTS Radiographic, high-resolution microcomputerized tomographic, and manual palpation revealed spinal fusion in five rats (83%) in Group 2 at 8 weeks. However, in Group 1, three (60%) rats developed fusion at L4-L5 by radiography and two (40%) by manual palpation in radiographic examination. In addition, in Group 3, bone fusion was observed in only 50% of rats by manual palpation and radiographic examination at this time. CONCLUSION The present study demonstrates that 0.08 gram of hydroxyapatite with 1 x 10(6)/ 60 microL rat of BMDMSCs induced bone fusion. FGF-4, added to differentiate primitive 1 x 10(6)/ 60 microL rat of BMDMSCs did not induce fusion. Based on histologic data, FGF-4 appears to induce fibrotic change rather than differentiation to bone by 1 x 10(6)/ 60 microL rat of BMDMSCs.
Collapse
Affiliation(s)
- Hyun Sung Seo
- Department of Neurosurgery, School of Medicine, Inha University, Incheon, Korea
| | | | | | | | | | | |
Collapse
|