1
|
Li H, Li Y, Xiang L, Luo S, Zhang Y, Li S. Therapeutic potential of GDF-5 for enhancing tendon regenerative healing. Regen Ther 2024; 26:290-298. [PMID: 39022600 PMCID: PMC11252783 DOI: 10.1016/j.reth.2024.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 07/20/2024] Open
Abstract
Tendon injury is a common disorder of the musculoskeletal system, with a higher possibility of occurrence in elderly individuals and athletes. After a tendon injury, the tendon suffers from inadequate and slow healing, resulting in the formation of fibrotic scar tissue, ending up with inferior functional properties. Therapeutic strategies involving the application of growth factors have been advocated to promote tendon healing. Growth and differentiation-5 (GDF-5) represents one such factor that has shown promising effect on tendon healing in animal models and in vitro cultures. Although promising, these studies are limited as the molecular mechanisms by which GDF-5 exerts its effect remain incompletely understood. Starting from broadly introducing essential elements of current understanding about GDF-5, the present review aims to define the effect of GDF-5 and its possible mechanisms of action in tendon healing. Nevertheless, we still need more in vivo studies to explore dosage, application time and delivery strategy of GDF-5, so as to pave the way for future clinical translation.
Collapse
Affiliation(s)
- Hanyue Li
- School of Physical Education, Southwest Medical University, PR China
| | - Yini Li
- Department of Ultrasound, The Affiliated Hospital of Southwest Medical University, Sichuan, PR China
| | - Linmei Xiang
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Shengyu Luo
- School of Physical Education, Southwest Medical University, PR China
| | - Yan Zhang
- Luzhou Vocational and Technical College, PR China
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, PR China
| |
Collapse
|
2
|
Wang D, Pun CCM, Huang S, Tang TCM, Ho KKW, Rothrauff BB, Yung PSH, Blocki AM, Ker EDF, Tuan RS. Tendon-derived extracellular matrix induces mesenchymal stem cell tenogenesis via an integrin/transforming growth factor-β crosstalk-mediated mechanism. FASEB J 2020; 34:8172-8186. [PMID: 32301551 DOI: 10.1096/fj.201902377rr] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/26/2022]
Abstract
Treatment of tendon injuries is challenging. To develop means to augment tendon regeneration, we have previously prepared a soluble, low immunogenic (DNA-free), tendon extracellular matrix fraction (tECM) by urea extraction of juvenile bovine tendons, which is capable of enhancing transforming growth factor-β (TGF-β) mediated tenogenesis in human adipose-derived stem cells (hASCs). Here, we aimed to elucidate the mechanism of tECM-driven hASC tenogenic differentiation in vitro, focusing on the integrin and TGF-β/SMAD pathways. Our results showed that tECM promoted hASC proliferation and tenogenic differentiation in vitro based on tenogenesis-associated markers. tECM also induced higher expression of several integrin subunits and TGF-β receptors, and nuclear translocation of p-SMAD2 in hASCs. Pharmacological inhibition of integrin-ECM binding, focal adhesion kinase (FAK) signaling, or TGF-β signaling independently led to compromised pro-tenogenic effects of tECM and actin fiber polymerization. Additionally, integrin blockade inhibited tECM-driven TGFBR2 expression, while inhibiting TGF-β signaling decreased tECM-mediated expression of integrin α1, α2, and β1 in hASCs. Together, these findings suggest that the strong pro-tenogenic bioactivity of tECM is regulated via integrin/TGF-β signaling crosstalk. Understanding how integrins interact with signaling by TGF-β and/or other growth factors (GFs) within the tendon ECM microenvironment will provide a rational basis for an ECM-based approach for tendon repair.
Collapse
Affiliation(s)
- Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Charmaine C M Pun
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shuting Huang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Thomas C M Tang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kevin K W Ho
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Benjamin B Rothrauff
- Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick S H Yung
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Anna M Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Elmer D F Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Osteoblastic Differentiation of Functionalized Biphasic Hydroxyapatite and b-Tricalcium Phosphate with Recombinant Human Growth and Differentiation (rhGDF-5). Macromol Res 2018. [DOI: 10.1007/s13233-018-6142-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
4
|
Abstract
Adipose-derived stem/stromal cells (ASCs), together with adipocytes, vascular endothelial cells, and vascular smooth muscle cells, are contained in fat tissue. ASCs, like the human bone marrow stromal/stem cells (BMSCs), can differentiate into several lineages (adipose cells, fibroblast, chondrocytes, osteoblasts, neuronal cells, endothelial cells, myocytes, and cardiomyocytes). They have also been shown to be immunoprivileged, and genetically stable in long-term cultures. Nevertheless, unlike the BMSCs, ASCs can be easily harvested in large amounts with minimal invasive procedures. The combination of these properties suggests that these cells may be a useful tool in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Simone Ciuffi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Roberto Zonefrati
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Maria Luisa Brandi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| |
Collapse
|
5
|
Yang G, Rothrauff BB, Lin H, Yu S, Tuan RS. Tendon-Derived Extracellular Matrix Enhances Transforming Growth Factor-β3-Induced Tenogenic Differentiation of Human Adipose-Derived Stem Cells. Tissue Eng Part A 2017; 23:166-176. [PMID: 27809678 DOI: 10.1089/ten.tea.2015.0498] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Because of the limited and unsatisfactory outcomes of clinical tendon repair, tissue engineering approaches using adult mesenchymal stem cells are being considered a promising alternative strategy to heal tendon injuries. Successful and functional tendon tissue engineering depends on harnessing the biochemical cues presented by the native tendon extracellular matrix (ECM) and the embedded tissue-specific biofactors. In this study, we have prepared and characterized the biological activities of a soluble extract of decellularized tendon ECM (tECM) on adult adipose-derived stem cells (ASCs), on the basis of histological, biochemical, and gene expression analyses. The results showed that tECM enhances the proliferation and transforming growth factor (TGF)-β3-induced tenogenesis of ASCs in both plate and scaffold cultures in vitro, and modulates matrix deposition of ASCs seeded in scaffolds. These findings suggest that combining tendon ECM extract with TGF-β3 treatment is a possible alternative approach to induce tenogenesis for ASCs.
Collapse
Affiliation(s)
- Guang Yang
- 1 Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,3 Department of Bioengineering, University of Pittsburgh Swanson School of Engineering , Pittsburgh, Pennsylvania
| | - Benjamin B Rothrauff
- 1 Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,4 Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Hang Lin
- 1 Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,4 Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Shuting Yu
- 1 Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,5 School of Medicine, Tsinghua University , Beijing, China
| | - Rocky S Tuan
- 1 Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,3 Department of Bioengineering, University of Pittsburgh Swanson School of Engineering , Pittsburgh, Pennsylvania.,4 Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
6
|
Narayanan G, Vernekar VN, Kuyinu EL, Laurencin CT. Poly (lactic acid)-based biomaterials for orthopaedic regenerative engineering. Adv Drug Deliv Rev 2016; 107:247-276. [PMID: 27125191 PMCID: PMC5482531 DOI: 10.1016/j.addr.2016.04.015] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/09/2016] [Accepted: 04/17/2016] [Indexed: 02/07/2023]
Abstract
Regenerative engineering converges tissue engineering, advanced materials science, stem cell science, and developmental biology to regenerate complex tissues such as whole limbs. Regenerative engineering scaffolds provide mechanical support and nanoscale control over architecture, topography, and biochemical cues to influence cellular outcome. In this regard, poly (lactic acid) (PLA)-based biomaterials may be considered as a gold standard for many orthopaedic regenerative engineering applications because of their versatility in fabrication, biodegradability, and compatibility with biomolecules and cells. Here we discuss recent developments in PLA-based biomaterials with respect to processability and current applications in the clinical and research settings for bone, ligament, meniscus, and cartilage regeneration.
Collapse
Affiliation(s)
- Ganesh Narayanan
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Varadraj N Vernekar
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Emmanuel L Kuyinu
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Cato T Laurencin
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA; School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
7
|
Buser Z, Acosta FL. Stem cells and spinal fusion--are we there yet? Spine J 2016; 16:400-1. [PMID: 27063500 DOI: 10.1016/j.spinee.2015.11.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 10/23/2015] [Accepted: 11/10/2015] [Indexed: 02/03/2023]
Abstract
Wheeler DL, Fredericks DC, Dryer RF, Bae HW. Allogeneic mesenchymal precursor cells (MPCs) combined with an osteoconductive scaffold to promote lumbar interbody spine fusion in an ovine model. Spine J 2016:16:389-99 (in this issue).
Collapse
Affiliation(s)
- Zorica Buser
- Department of Orthopaedic Surgery, University of Southern California, 1450 San Pablo St, Suite 5400, Los Angeles, CA, 90033, USA
| | - Frank L Acosta
- Department of Orthopaedic Surgery, University of Southern California, 1450 San Pablo St, Suite 5400, Los Angeles, CA, 90033, USA; Department of Neurological Surgery, University of Southern California, 1450 San Pablo St, Suite 5400, Los Angeles, CA, 90033, USA.
| |
Collapse
|
8
|
The effective mode of growth and differentiation factor-5 in promoting the chondrogenic differentiation of adipose-derived stromal cells. Cell Tissue Bank 2015; 17:105-15. [PMID: 26084505 DOI: 10.1007/s10561-015-9517-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 06/08/2015] [Indexed: 01/06/2023]
Abstract
Our study aimed to find out the most effective mode for chondrogenic differentiation based on time, dose and culture method. ADSCs were cultured and identified by CD44, CD49d, and CD106 immumohistochemical staining method, and their differentiation potential to chondrocyte were detected by Alizarin red staining. ADSCs induced by different concentrations of GDF-5 for chondrogenic differentiation were detected by blue and toluidine blue staining and collagen type II and X immumohistochemical staining. The expression of collagen I, II, X and aggrecan gene in GDF-induced ADSCs cultured in 2- and 3-dimension was identified by real-time PCR. Cell microstructure and proliferation in three-dimensional scaffolds at day 7, 14, 21 and 28 were analyzed by scanning electron microscopy and MTS assay. The ADSCs were successfully identified by CD44 and CD49d, and their differentiation potential was detected by Alizarin red staining. Real-time PCR showed that collagen and aggrecan were expressed at high levels in 100 or 200 ng/mL GDF-5 treated cells. The collagen types (I, II) and aggrecan genes were higher expressed in GDF-5 induced scaffold group than that in monolayer group. MTS showed that the cell counts were not significantly different among different treated time. Both collagen type II and aggrecan gene were highly expressed at day 14, while collagen types I and X gene expressions peaked at day 21 and 28. The 100 ng/mL GDF-5 is effective and cost-effective for chondrogenic differentiation when cultured at day 14 in vitro under three-dimensional culture conditions.
Collapse
|
9
|
Skovrlj B, Guzman JZ, Al Maaieh M, Cho SK, Iatridis JC, Qureshi SA. Cellular bone matrices: viable stem cell-containing bone graft substitutes. Spine J 2014; 14:2763-72. [PMID: 24929059 PMCID: PMC4402977 DOI: 10.1016/j.spinee.2014.05.024] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 04/03/2014] [Accepted: 05/20/2014] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Advances in the field of stem cell technology have stimulated the development and increased use of allogenic bone grafts containing live mesenchymal stem cells (MSCs), also known as cellular bone matrices (CBMs). It is estimated that CBMs comprise greater than 17% of all bone grafts and bone graft substitutes used. PURPOSE To critically evaluate CBMs, specifically their technical specifications, existing published data supporting their use, US Food and Drug Administration (FDA) regulation, cost, potential pitfalls, and other aspects pertaining to their use. STUDY DESIGN Areview of literature. METHODS A series of Ovid, Medline, and Pubmed-National Library of Medicine/National Institutes of Health (www.ncbi.nlm.nih.gov) searches were performed. Only articles in English journals or published with English language translations were included. Level of evidence of the selected articles was assessed. Specific technical information on each CBM was obtained by direct communication from the companies marketing the individual products. RESULTS Five different CBMs are currently available for use in spinal fusion surgery. There is a wide variation between the products with regard to the average donor age at harvest, total cellular concentration, percentage of MSCs, shelf life, and cell viability after defrosting. Three retrospective studies evaluating CBMs and fusion have shown fusion rates ranging from 90.2% to 92.3%, and multiple industry-sponsored trials are underway. No independent studies evaluating spinal fusion rates with the use of CBMs exist. All the commercially available CBMs claim to meet the FDA criteria under Section 361, 21 CFR Part 1271, and are not undergoing FDA premarket review. The CBMs claim to provide viable MSCs and are offered at a premium cost. Numerous challenges exist in regard to MSCs' survival, function, osteoblastic potential, and cytokine production once implanted into the intended host. CONCLUSIONS Cellular bone matrices may be a promising bone augmentation technology in spinal fusion surgery. Although CBMs appear to be safe for use as bone graft substitutes, their efficacy in spinal fusion surgery remains highly inconclusive. Large, nonindustry sponsored studies evaluating the efficacy of CBMs are required. Without results from such studies, surgeons must be made aware of the potential pitfalls of CBMs in spinal fusion surgery. With the currently available data, there is insufficient evidence to support the use of CBMs as bone graft substitutes in spinal fusion surgery.
Collapse
Affiliation(s)
- Branko Skovrlj
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, 1 Gustave L Levy Place, Box 1136, New York, NY 10029, USA
| | - Javier Z. Guzman
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, 5 East 98th St, 9th Floor, Box 1188, New York, NY 10029, USA
| | - Motasem Al Maaieh
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, 5 East 98th St, 9th Floor, Box 1188, New York, NY 10029, USA
| | - Samuel K. Cho
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, 5 East 98th St, 9th Floor, Box 1188, New York, NY 10029, USA
| | - James C. Iatridis
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, 5 East 98th St, 9th Floor, Box 1188, New York, NY 10029, USA
| | - Sheeraz A. Qureshi
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, 5 East 98th St, 9th Floor, Box 1188, New York, NY 10029, USA,Corresponding author. Department of Orthopaedic Surgery, Ichan School of Medicine at Mount Sinai, 5 E. 98th St, Box 1188, New York, NY 10029, USA. Tel.: (212) 241-3909; fax: (212) 534-6202.
| |
Collapse
|
10
|
Isolation, characterization, differentiation, and application of adipose-derived stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2014; 123:55-105. [PMID: 20091288 DOI: 10.1007/10_2009_24] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
While bone marrow-derived mesenchymal stem cells are known and have been investigated for a long time, mesenchymal stem cells derived from the adipose tissue were identified as such by Zuk et al. in 2001. However, as subcutaneous fat tissue is a rich source which is much more easily accessible than bone marrow and thus can be reached by less invasive procedures, adipose-derived stem cells have moved into the research spotlight over the last 8 years.Isolation of stromal cell fractions involves centrifugation, digestion, and filtration, resulting in an adherent cell population containing mesenchymal stem cells; these can be subdivided by cell sorting and cultured under common conditions.They seem to have comparable properties to bone marrow-derived mesenchymal stem cells in their differentiation abilities as well as a favorable angiogenic and anti-inflammatory cytokine secretion profile and therefore have become widely used in tissue engineering and clinical regenerative medicine.
Collapse
|
11
|
Key role for the transcriptional factor, osterix, in spine development. Spine J 2014; 14:683-94. [PMID: 24268393 DOI: 10.1016/j.spinee.2013.08.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 08/06/2013] [Accepted: 08/23/2013] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Osterix (Osx) is an important transcriptional factor for bone formation; however, its role in spine development has not been determined. PURPOSE The goal of the present study is to observe the role of Osx in spine development. STUDY DESIGN/SETTING Deletion and overexpression of Osx were achieved in Osx knockout and transgenic mice, respectively, to determine the effect of Osx on spine development. METHODS With 2.3-kb type I collagen as a promoter, Osx were deleted in mice, and overexpression of Osx was obtained in Osx transgenic mice. Video, X-ray radiology, histology, tartrate-resistant acid phosphatase staining, Safranin O staining, and immunohistochemistry were used to assess the effect of Osx deletion and overexpression on spine development. This study was supported by National Science Foundation of China (81271935) and Foundation of State Key Laboratory of Trauma, Burns and combined injury (SKLZZ SKLZZ201124). No potential conflict of interest to disclose. RESULTS Overexpression of Osx did not have an obvious effect on spine development, whereas deletion of Osx led to severe spine deformities that included wedged vertebrae, spinal stenosis, and congenital scoliosis. Also, Osx deactivation resulted in shortened vertebrae and excessive bone volume in the vertebrae. TRAP staining showed that activity of osteoclasts decreased in Osx-null mice, and examination with TdT-mediated dUTP nick end labeling revealed that the apoptosis rate at the growth plate decreased significantly in Osx-null mice. Excessive formation of bone was positive for Safranin O staining. CONCLUSIONS Osx plays an important role in spine development, and its deactivation leads to severe spine deformities.
Collapse
|
12
|
Werner BC, Li X, Shen FH. Stem cells in preclinical spine studies. Spine J 2014; 14:542-51. [PMID: 24246748 DOI: 10.1016/j.spinee.2013.08.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 07/05/2013] [Accepted: 08/23/2013] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT The recent identification and characterization of mesenchymal stem cells have introduced a shift in the research focus for future technologies in spinal surgery to achieve spinal fusion and treat degenerative disc disease. Current and past techniques use allograft to replace diseased tissue or rely on host responses to recruit necessary cellular progenitors. Adult stem cells display long-term proliferation, efficient self-renewal, and multipotent differentiation. PURPOSE This review will focus on two important applications of stem cells in spinal surgery: spine fusion and the management of degenerative disc disease. STUDY DESIGN Review of the literature. METHODS Relevant preclinical literature regarding stem cell sources, growth factors, scaffolds, and animal models for both osteogenesis and chondrogenesis will be reviewed, with an emphasis on those studies that focus on spine applications of these technologies. RESULTS In both osteogenesis and chondrogenesis, adult stem cells derived from bone marrow or adipose show promise in preclinical studies. Various growth factors and scaffolds have also been shown to enhance the properties and eventual clinical potential of these cells. Although its utility in clinical applications has yet to be proven, gene therapy has also been shown to hold promise in preclinical studies. CONCLUSIONS The future of spine surgery is constantly evolving, and the recent advancements in stem cell-based technologies for both spine fusion and the treatment of degenerative disc disease is promising and indicative that stem cells will undoubtedly play a major role clinically. It is likely that these stem cells, growth factors, and scaffolds will play a critical role in the future for replacing diseased tissue in disease processes such as degenerative disc disease and in enhancing host tissue to achieve more reliable spine fusion.
Collapse
Affiliation(s)
- Brian C Werner
- Department of Orthopaedic Surgery, University of Virginia, PO Box 800159, Charlottesville, VA 22908-0159 USA
| | - Xudong Li
- Department of Orthopaedic Surgery, University of Virginia, PO Box 800159, Charlottesville, VA 22908-0159 USA
| | - Francis H Shen
- Department of Orthopaedic Surgery, University of Virginia, PO Box 800159, Charlottesville, VA 22908-0159 USA.
| |
Collapse
|
13
|
Liang H, Li X, Shimer AL, Balian G, Shen FH. A novel strategy of spine defect repair with a degradable bioactive scaffold preloaded with adipose-derived stromal cells. Spine J 2014; 14:445-54. [PMID: 24360747 DOI: 10.1016/j.spinee.2013.09.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/27/2013] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Although the use of mesenchymal stem cells (MSC) with scaffolds for bone repair has been considered an effective method, the interactions between implanted materials and bone tissues have not been fully elucidated. At some specific sites, such as the vertebral body (VB) of the spine, the process of bone repair with implanted biomaterials is rarely reported. Recently, adipose tissue was found to be an alternative source of MSC besides bone marrow. However, the strategy of using adipose-derived stromal (ADS) cells with bioactive scaffold for the repair of spinal bone defects has seldom been studied. PURPOSE To use a sintered poly(lactide-co-glycolide) acid (PLGA) microspheres scaffold seeded with induced rat ADS cells to repair a bone defect of the VB in a rat model. STUDY DESIGN Basic science and laboratory study. METHODS A sintered porous microspheres scaffold was manufactured by PLGA. ADS cells were isolated from Fischer 344 rats and then induced by osteogenic medium with growth and differentiation factor 5 (GDF5) in vitro. Before implantation, cells were cultured with inductive media for 2 weeks as a monolayer situation and 1 more week on a PLGA scaffold as a three-dimensional structure. These assembled bioactive scaffolds then were implanted in lumbar VB bone defects in Fischer 344 rats. The ex vivo differentiation of the cells was confirmed by von Kossa staining and real-time polymerase chain reaction. The performance of cells on the scaffold was detected by scanning electron microscopy and (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assay. In vivo bone formation was quantitatively measured by computed tomography study. And the effect of tissue repair was also evaluated by histological studies. RESULTS Proliferation and differentiation of cells were confirmed before in vivo implantation. Quantification of bone formation in vivo through serial three-dimensional computed tomography images revealed that the VB implanted with GDF5-induced cells demonstrated more bone formation than the control groups. Besides the bone formation period that occurred between 2 and 4 weeks in all groups, a second bone formation period was found to occur only in the groups that received cells with previous induction in vitro. This second period of significant bone formation happened simultaneously with collapsing of the scaffolds. It was then demonstrated histologically that vascularization early in the process and cooperation between host bone and implanted cells accompanied by collapse of the scaffold may be the factors that influence bone formation. This study not only provides a therapeutic strategy of using biomaterial for bone repair in the spine, but also may lead to a technological method for studying the relationship between implanted stem cells and host tissue. CONCLUSIONS Adipose-derived stromal cells maintained in culture on a scaffold and treated with osteogenic induction with growth factor ex vivo could be used to enhance bone repair in vivo.
Collapse
Affiliation(s)
- Haixiang Liang
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratories, University of Virginia School of Medicine, Cobb Hall, Room B057, Box 800159, Charlottesville, VA 22908, USA
| | - Xudong Li
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratories, University of Virginia School of Medicine, Cobb Hall, Room B057, Box 800159, Charlottesville, VA 22908, USA
| | - Adam L Shimer
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratories, University of Virginia School of Medicine, Cobb Hall, Room B057, Box 800159, Charlottesville, VA 22908, USA
| | - Gary Balian
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratories, University of Virginia School of Medicine, Cobb Hall, Room B057, Box 800159, Charlottesville, VA 22908, USA; Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Jordan Hall 6007, 1340 Jefferson Park Ave, Charlottesville, VA 22908, USA
| | - Francis H Shen
- Department of Orthopaedic Surgery, Orthopaedic Research Laboratories, University of Virginia School of Medicine, Cobb Hall, Room B057, Box 800159, Charlottesville, VA 22908, USA.
| |
Collapse
|
14
|
Abstract
In 2001, researchers at the University of California, Los Angeles, described the isolation of a new population of adult stem cells from liposuctioned adipose tissue. These stem cells, now known as adipose-derived stem cells or ADSCs, have gone on to become one of the most popular adult stem cells populations in the fields of stem cell research and regenerative medicine. As of today, thousands of research and clinical articles have been published using ASCs, describing their possible pluripotency in vitro, their uses in regenerative animal models, and their application to the clinic. This paper outlines the progress made in the ASC field since their initial description in 2001, describing their mesodermal, ectodermal, and endodermal potentials both in vitro and in vivo, their use in mediating inflammation and vascularization during tissue regeneration, and their potential for reprogramming into induced pluripotent cells.
Collapse
|
15
|
The challenge and advancement of annulus fibrosus tissue engineering. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2013; 22:1090-100. [PMID: 23361531 DOI: 10.1007/s00586-013-2663-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 12/26/2012] [Accepted: 01/07/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND Intervertebral disc degeneration, a main cause of back pain, is an endemic problem and a big economic burden for the health care system. Current treatments are symptom relieving but do not address underlying problems-biological and structural deterioration of the disc. Tissue engineering is an emerging approach for the treatment of intervertebral disc degeneration since it restores the functionality of native tissues. Although numerous studies have focused on the nucleus pulposus tissue engineering and achieved successes in laboratory settings, disc tissue engineering without annulus fibrosus for the end stage of disc degeneration is deemed to fail. The purpose of this article is to review the advancement of annulus fibrosus tissue engineering. MATERIAL AND METHODS Relevant articles regarding annulus fibrosus tissue engineering were identified in PubMed and Medline databases. RESULTS The ideal strategy for disc regeneration is to restore the function and integrity of the disc by using biomaterials, native matrices, growth factors, and cells that producing matrices. In the past decades there are tremendous advancement in annulus fibrosus tissue engineering including cell biology, biomaterials, and whole disc replacement. The recent promising results on whole disc tissue engineering-a composite of annulus fibrosus and nucleus pulposus-make the tissue engineering approach more appealing. CONCLUSION Despite the promising results in disc tissue engineering, there is still much work to be done regarding the clinical application.
Collapse
|
16
|
Shen FH, Werner BC, Liang H, Shang H, Yang N, Li X, Shimer AL, Balian G, Katz AJ. Implications of adipose-derived stromal cells in a 3D culture system for osteogenic differentiation: an in vitro and in vivo investigation. Spine J 2013; 13:32-43. [PMID: 23384881 DOI: 10.1016/j.spinee.2013.01.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 12/20/2012] [Accepted: 01/08/2013] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Healthy mammalian cells in normal tissues are organized in complex three-dimensional (3D) networks that display nutrient and signaling gradients. Conventional techniques that grow cells in a 2D monolayer fail to reproduce the environment that is observed in vivo. In recent years, 3D culture systems have been used to mimic tumor microenvironments in cancer research and to emulate embryogenesis in stem cell cultures. However, there have been no studies exploring the ability for adipose-derived stromal (ADS) cells in a 3D culture system to undergo osteogenic differentiation. PURPOSE To characterize and investigate the in vitro and in vivo potential for human ADS cells in a novel 3D culture system to undergo osteogenic differentiation. STUDY DESIGN Basic science and laboratory study. METHODS Human ADS cells were isolated and prepared as either a 2D monolayer or 3D multicellular aggregates (MAs). Multicellular aggregates were formed using the hanging droplet technique. Cells were treated in osteogenic medium in vitro, and cellular differentiation was investigated using gene expression, histology, and microCT at 1-, 2-, and 4-week time points. In vivo investigation involved creating a muscle pouch by developing the avascular muscular interval in the vastus lateralis of male athymic rats. Specimens were then pretreated with osteogenic medium and surgically implanted as (1) carrier (Matrigel) alone (control), (2) carrier with human ADS cells in monolayer, or (3) human ADS cells as MAs. In vivo evidence of osteogenic differentiation was evaluated with micro computed tomography and histologic sectioning at a 2-week time point. RESULTS Human ADS cells cultured by the hanging droplet technique successfully formed MAs at the air-fluid interface. Adipose-derived stromal cells cultured in monolayer or as 3D MAs retain their ability to self-replicate and undergo multilineage differentiation as confirmed by increased runx2/Cbfa2, ALP, and OCN and increased matrix mineralization on histologic sectioning. Multicellular aggregate cells expressed increased differentiation potential and extracellular matrix production over the same human ADS cells cultured in monolayer. Furthermore, MAs reseeded onto monolayer retained their stem cell capabilities. When implanted in vivo, significantly greater bone volume and extracellular matrix were present in the implanted specimens of MAs confirmed on both microCT and histological sectioning. CONCLUSIONS This is the first study to investigate the capability of human ADS cells in a 3D culture system to undergo osteogenic differentiation. The results confirm that MAs maintain their stem cell characteristics. Compared with analogous cells in monolayer culture, the human ADS cells as MAs exhibit elevated levels of osteogenic differentiation and increased matrix mineralization. Furthermore, the creation of uniform spheroids allows for improved handling and manipulation during transplantation. These findings strongly support the concept that 3D culture systems remain not only a viable option for stem cell culture but also possibly a more attractive alternative to traditional culture techniques to improve the osteogenic potential of human adipose stem cells.
Collapse
Affiliation(s)
- Francis H Shen
- Department of Orthopaedic Surgery, University of Virginia, Box 800159, Charlottesville, VA 22908-0159, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Chen J, Shi ZD, Ji X, Morales J, Zhang J, Kaur N, Wang S. Enhanced osteogenesis of human mesenchymal stem cells by periodic heat shock in self-assembling peptide hydrogel. Tissue Eng Part A 2012; 19:716-28. [PMID: 23072422 DOI: 10.1089/ten.tea.2012.0070] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The mechanisms for the heat-induced osteogenesis are not completely known and the thermal regulation of human mesenchymal stem cell (hMSC) differentiation is not well studied. In this study, the direct effects of mild heat shock (HS) on the differentiation of hMSCs into osteoblasts in self-assembling peptide hydrogel and on tissue culture plates were investigated. hMSCs isolated from human bone marrow were seeded in conventional culture plates (two-dimensional [2D] culture) and on the surface of three-dimensional (3D) PuraMatrix peptide hydrogel (3D culture), followed by 1 h HS at 41°C once a week during osteogenic differentiation. Alkaline phosphatase (ALP) activity was enhanced in both 2D and 3D cultures via periodic HS at early stage of differentiation; meanwhile, HS significantly increased the calcium deposition at day 19 and 27 of differentiation in both 2D and 3D cultures. The periodic HS also upregulated osteo-specific genes, osterix (OSX) on day 11, osteopontin (OP) on day 19, and bone morphogenetic protein 2 (BMP2) on day 25 in 2D culture. In 3D PuraMatrix culture, the runt-related transcription factor 2 (Runx2) was upregulated by HS on day 25 of differentiation. The heat shock protein 70 (HSP70) was significantly upregulated by HS in differentiated hMSCs analyzed at 24 h after HS. These results demonstrate that HS induced an earlier differentiation of hMSCs and enhanced the maturation of osteoblasts differentiated from hMSCs. Therefore, mild HS treatment may be potentially used to enhance the bone regeneration using hMSCs. Our data will guide the design of in vivo heating protocols and enable further investigations in thermal treatments of MSC osteogenesis for bone tissue engineering.
Collapse
Affiliation(s)
- Jing Chen
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Lubelski D, Abdullah KG, Benzel EC, Mroz TE. The Utility of Allograft Mesenchymal Stem Cells for Spine Fusion: A Literature Review. Global Spine J 2012; 2:109-14. [PMID: 27054055 PMCID: PMC4813091 DOI: 10.1055/s-0032-1307263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
More than 50% of patients complain of postoperative donor site morbidity following iliac crest bone graft harvest, and recent discoveries have identified adverse outcomes following bone morphogenetic protein use in spine fusion. This has led the spine community to turn toward alternative methods to promote fusion following spine surgery. The present article reviews numerous studies that have shown the osteogenic potential of mesenchymal stem cells (MSCs). MSCs have been used with both in vitro and in vivo models and have involved animal studies ranging from rats to macaque monkeys to successfully induce bone regeneration in lesions of the tibia and spine. There is no fear of graft rejection, as there may be with other allograft materials, because neither undifferentiated nor differentiated MSCs elicit lymphocyte response when transplanted; they tend to alter the cytokine profile to an anti-inflammatory state. Early clinical trials are underway with various commercially available MSC formulations. Although there is much enthusiasm, it is integral that the spine surgery community carefully evaluate the use of MSCs in spine fusion through well-designed and executed studies to determine the efficacy and safety profiles in spine surgery patients.
Collapse
Affiliation(s)
- Daniel Lubelski
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| | - Kalil G. Abdullah
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| | - Edward C. Benzel
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| | - Thomas E. Mroz
- Department of Neurological Surgery, Cleveland Clinic Center for Spine Health,
Cleveland, Ohio
| |
Collapse
|
19
|
Park JC, Wikesjö UME, Koo KT, Lee JS, Kim YT, Pippig SD, Bastone P, Kim CS, Kim CK. Maturation of alveolar bone following implantation of an rhGDF-5/PLGA composite into 1-wall intra-bony defects in dogs: 24-week histometric observations. J Clin Periodontol 2012; 39:565-73. [DOI: 10.1111/j.1600-051x.2012.01868.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2012] [Indexed: 11/29/2022]
Affiliation(s)
- Jung-Chul Park
- Department of Periodontology; Research Institute for Periodontal Regeneration; College of Dentistry; Yonsei University; Seoul; Korea
| | - Ulf M. E. Wikesjö
- Departments of Periodontics and Oral Biology; Laboratory for Applied Periodontal & Craniofacial Regeneration (LAPCR); Georgia Health Sciences University College of Dental Medicine; Augusta; GA; USA
| | - Ki-Tae Koo
- Department of Periodontology; College of Dentistry; Seoul National University; Seoul; Korea
| | - Jung-Seok Lee
- Department of Periodontology; Research Institute for Periodontal Regeneration; College of Dentistry; Yonsei University; Seoul; Korea
| | - Yong-Tae Kim
- Department of Periodontology; Research Institute for Periodontal Regeneration; College of Dentistry; Yonsei University; Seoul; Korea
| | | | | | - Chang-Sung Kim
- Department of Periodontology; Research Institute for Periodontal Regeneration; College of Dentistry; Yonsei University; Seoul; Korea
| | - Chong-Kwan Kim
- Department of Periodontology; Research Center for Orofacial Hard Tissue Regeneration (MRC); College of Dentistry; Yonsei University; Seoul; Korea
| |
Collapse
|
20
|
Cheng X, Yang T, Meng W, Liu H, Zhang T, Shi R. Overexpression of GDF5 through an adenovirus vector stimulates osteogenesis of human mesenchymal stem cells in vitro and in vivo. Cells Tissues Organs 2012; 196:56-67. [PMID: 22287558 DOI: 10.1159/000330791] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2011] [Indexed: 02/05/2023] Open
Abstract
The use of stem cells combined with gene therapy could be an important way to facilitate bone regeneration. In this study, the aim was to investigate the potential of growth and differentiation factor-5 (GDF5) to genetically manipulate human mesenchymal stem cells (hMSCs) for bone regeneration. Recombinant adenovirus Ad-GDF5 and Ad-GFP were constructed and identified, and the titer of both were determined. Third-passage hMSCs were infected with adenovirus, and the expression of GDF5 was confirmed by detection of GFP-positive cells, GDF5 mRNA levels, Western blotting, and enzyme-linked immunosorbent assay (ELISA). hMSCs at passage 3 were divided into four groups: (1) an experimental group infected with Ad-GDF5, (2) a positive control group cultured with osteogenic differentiation medium, (3) a control group infected with Ad-GFP cultured with standard medium, and (4) a blank control group cultured with standard medium. Evaluation of cell morphology and proliferation, analysis of the expression of genes related to osteogenic differentiation, von Kossa staining, and immunofluorescent staining of collagen I were used to investigate the osteogenesis of cells among the groups. After culturing the cells for 2 days under each corresponding condition, the cells were detached and subcutaneously injected into the backs of nude mice to evaluate bone formation. Samples were collected for histological staining, protein Western blotting, and micro-computer tomography. When infected with Ad-GDF5, hMSCs could overexpress GDF5 for a prolonged period in vitro and reach a concentration of 160 ng/ml. Cells infected with Ad-GDF5 or cultured in osteogenic medium displayed osteogenic differentiation based on their histological and cellular properties and on their gene and protein expression patterns. Furthermore, Ad-GDF5 showed a better ability to upregulate the expression of collagen I, alkaline phosphatase, and osteocalcin mRNA than the osteogenic medium. Furthermore, Ad-GDF5 expression was associated with enhanced bone formation in vivo. Our findings suggest that hMSCs infected with Ad-GDF5 can differentiate in an osteogenic direction and may be a promising cell source for bone regeneration.
Collapse
Affiliation(s)
- Xiangjun Cheng
- Department of Orthopaedic Surgery, West China Hospital, Sichuan University, Chengdu, PR China
| | | | | | | | | | | |
Collapse
|
21
|
Coleman CM, Scheremeta BH, Boyce AT, Mauck RL, Tuan RS. Delayed fracture healing in growth differentiation factor 5-deficient mice: a pilot study. Clin Orthop Relat Res 2011; 469:2915-24. [PMID: 21590487 PMCID: PMC3171561 DOI: 10.1007/s11999-011-1912-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 04/29/2011] [Indexed: 01/31/2023]
Abstract
BACKGROUND Growth differentiation factor-5 (GDF-5) is a key regulator of skeletogenesis and bone repair and induces bone formation in spinal fusions and nonunion applications by enhancing chondrocytic and osteocytic differentiation and stimulating angiogenesis. Elucidating the contribution of GDF-5 to fracture repair may support its clinical application in complex fractures. QUESTIONS/PURPOSE We therefore asked whether the absence of GDF-5 during fracture repair impaired bone healing as assessed radiographically, histologically, and mechanically. METHODS In this pilot study, we performed tibial osteotomies on 10-week-old male mice, stabilized by intramedullary and extramedullary nailing. Healing was assessed radiographically and histologically on Days 1 (n = 1 wild-type; n = 5 bp [brachopodism]), 5 (n = 3 wild-type; n = 3 bp), 10 (n = 6 wild-type; n = 3 bp), 14 (n = 6 wild-type; n = 6 bp), 21 (n = 6 wild-type; n = 6 bp), 28 (n = 7 wild-type; n = 6 bp), and 56 (n = 6 wild-type; n = 6 bp) after fracture. After 10 (n = 7 wild-type; n = 7 bp contralateral and n = 3 bp fractured tibiae), 14 (n = 6 wild-type; n = 6 bp), 21 (n = 6 wild-type; n = 6 bp), 28 (n = 6 wild-type; n = 3 bp), and 56 (n = 8 wild-type; n = 6 bp) days, the callus cross-sectional area was calculated. We characterized the mechanical integrity of the healing fracture by yield stress and Young's modulus at 28 (n = 6 wild-type; n = 3 bp) and 56 (n = 8 wild-type; n = 6 bp) days postfracture. RESULTS The absence of GDF-5 impaired cartilaginous matrix deposition in the callus and reduced callus cross-sectional area. After 56 days, the repaired bp fracture was mechanically comparable to that of controls. CONCLUSIONS Although GDF-5 deficiency did not compromise long-term fracture healing, a delay in cartilage formation and remodeling supports roles for GDF-5 in the early phase of bone repair. CLINICAL RELEVANCE Local delivery of GDF-5 to clinically difficult fractures may simulate cartilage formation in the callus and support subsequent remodeling.
Collapse
Affiliation(s)
- Cynthia M. Coleman
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Service, Bethesda, MD USA
- Regenerative Medicine Institute, National University of Ireland Galway, Galway City, County Galway Ireland
| | - Brooke H. Scheremeta
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Service, Bethesda, MD USA
- Schneider Children’s Hospital, New Hyde Park, NY USA
| | - Amanda T. Boyce
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Service, Bethesda, MD USA
| | - Robert L. Mauck
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Service, Bethesda, MD USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA USA
| | - Rocky S. Tuan
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Service, Bethesda, MD USA
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA 15219 USA
| |
Collapse
|
22
|
Scanning electron microscopy preparation protocol for differentiated stem cells. Anal Biochem 2011; 416:186-90. [DOI: 10.1016/j.ab.2011.05.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 05/19/2011] [Accepted: 05/20/2011] [Indexed: 01/29/2023]
|
23
|
Abstract
This is a review of the growing scientific interest in the developmental plasticity and therapeutic potential of stromal cells isolated from adipose tissue. Adipose-derived stem/stromal cells (ASCs) are multipotent somatic stem cells that are abundant in fat tissue. It has been shown that ASCs can differentiate into several lineages, including adipose cells, chondrocytes, osteoblasts, neuronal cells, endothelial cells, and cardiomyocytes. At the same time, adipose tissue can be harvested by a minimally invasive procedure, which makes it a promising source of adult stem cells. Therefore, it is believed that ASCs may become an alternative to the currently available adult stem cells (e.g. bone marrow stromal cells) for potential use in regenerative medicine. In this review, we present the basic information about the field of adipose-derived stem cells and their potential use in various applications.
Collapse
|
24
|
Min CK, Wikesjö UME, Park JC, Chae GJ, Pippig SD, Bastone P, Kim CS, Kim CK. Wound healing/regeneration using recombinant human growth/differentiation factor-5 in an injectable poly-lactide-co-glycolide-acid composite carrier and a one-wall intra-bony defect model in dogs. J Clin Periodontol 2011; 38:261-8. [DOI: 10.1111/j.1600-051x.2010.01691.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
25
|
Park A, Hogan MV, Kesturu GS, James R, Balian G, Chhabra AB. Adipose-derived mesenchymal stem cells treated with growth differentiation factor-5 express tendon-specific markers. Tissue Eng Part A 2010; 16:2941-51. [PMID: 20575691 DOI: 10.1089/ten.tea.2009.0710] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVES Adipose-derived mesenchymal stem cells (ADMSCs) are a unique population of stem cells with therapeutic potential in the treatment of connective tissue injuries. Growth differentiation factor-5 (GDF)-5 is known to play a role in tendon repair and maintenance. The aim of this study was to investigate the effects of GDF-5 on proliferation and tendonogenic gene expression of rat ADMSCs. METHODS ADMSCs were treated in culture with different concentrations of GDF-5 (0-1000 ng/mL) for 12 days. Biochemical, temporal, and concentration kinetic studies were done. Extracellular matrix (ECM) synthesis, tendonogenic differentiation, and matrix remodeling gene and protein expression were analyzed. RESULTS GDF-5 led to increased ADMSC proliferation in a dose- and time-dependent manner. ADMSCs demonstrated enhanced ECM (collagen type I, decorin, and aggrecan) and tendonogenic marker (scleraxis, tenomodulin, and tenascin-C) gene expression with 100 ng/mL of GDF-5 (p < 0.05). ECM and tendon-specific markers showed time-dependent increases at various time points (p < 0.05), although decorin decreased at day 9 (p < 0.05). GDF-5 did alter expression of matrix remodeling genes, with no specific trends observed. Western blot analysis confirmed dose- and time-dependent increases in protein expression of tenomodulin, tenascin-C, Smad-8, and matrix metalloproteinase-13. CONCLUSION In vitro GDF-5 treatment can induce cellular events leading to the tendonogenic differentiation of ADMSCs. The use of combined GDF-5 and ADMSCs tissue-engineered therapies may have a role in the future of tendon repair.
Collapse
Affiliation(s)
- Andrew Park
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Virginia Health System, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | |
Collapse
|
26
|
Seong JM, Kim BC, Park JH, Kwon IK, Mantalaris A, Hwang YS. Stem cells in bone tissue engineering. Biomed Mater 2010; 5:062001. [PMID: 20924139 DOI: 10.1088/1748-6041/5/6/062001] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bone tissue engineering has been one of the most promising areas of research, providing a potential clinical application to cure bone defects. Recently, various stem cells including embryonic stem cells (ESCs), bone marrow-derived mesenchymal stem cells (BM-MSCs), umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs), adipose tissue-derived stem cells (ADSCs), muscle-derived stem cells (MDSCs) and dental pulp stem cells (DPSCs) have received extensive attention in the field of bone tissue engineering due to their distinct biological capability to differentiate into osteogenic lineages. The application of these stem cells to bone tissue engineering requires inducing in vitro differentiation of these cells into bone forming cells, osteoblasts. For this purpose, efficient in vitro differentiation towards osteogenic lineage requires the development of well-defined and proficient protocols. This would reduce the likelihood of spontaneous differentiation into divergent lineages and increase the available cell source for application to bone tissue engineering therapies. This review provides a critical examination of the various experimental strategies that could be used to direct the differentiation of ESC, BM-MSC, UCB-MSC, ADSC, MDSC and DPSC towards osteogenic lineages and their potential applications in tissue engineering, particularly in the regeneration of bone.
Collapse
Affiliation(s)
- Jeong Min Seong
- Department of Preventive and Social Dentistry & Institute of Oral Biology, College of Dentistry, Kyung Hee University, Seoul 130-701, Korea
| | | | | | | | | | | |
Collapse
|
27
|
García Cruz DM, Gomes M, Reis RL, Moratal D, Salmerón-Sánchez M, Gómez Ribelles JL, Mano JF. Differentiation of mesenchymal stem cells in chitosan scaffolds with double micro and macroporosity. J Biomed Mater Res A 2010; 95:1182-93. [DOI: 10.1002/jbm.a.32906] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 04/22/2010] [Accepted: 05/25/2010] [Indexed: 11/12/2022]
|
28
|
Rada T, Reis RL, Gomes ME. Adipose tissue-derived stem cells and their application in bone and cartilage tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2010; 15:113-25. [PMID: 19196117 DOI: 10.1089/ten.teb.2008.0423] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The adipose tissue was considered a reserve of energy until the '80s, when it was found that this tissue was involved in the metabolism of sex steroids such as estrogens. From then on, the importance attributed to this tissue radically changed as it was then considered an active organ, involved in important functions of the human body. In 2001, for the first time, the existence of stem cells within this tissue was reported, and since then, this tissue has been gaining an increased importance as a stem cell source for a wide range of potential applications in cell therapies and/or tissue engineering and regenerative medicine strategies, mainly due to its wide availability and easy access. This manuscript provides an overview on adipose stem cells (i.e., adipose tissue-derived stem cells, ASCs) considering the tissue of origin, the niche of the ASCs, and their phenotype in all aspects. In this paper it is also discussed the markers that have been used for the characterization of these cells, their differentiation properties, and their immunological reactivity, reporting studies from 2001 until this date. The ASCs are also compared with bone marrow stem cells (BMSCs), until now considered as the gold standard source of stem cells, underlining the common characteristics and the differences between the stem cells obtained from these two sources, as well as the advantages and disadvantages of their potential use in different applications. Finally, this review will also focus on the potential application of ASCs in tissue engineering applications, particularly in the regeneration of bone and cartilage, commenting on the progress of this approach and future trends of the field.
Collapse
Affiliation(s)
- Tommaso Rada
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, University of Minho, Braga, Portugal
| | | | | |
Collapse
|
29
|
Lee JS, Wikesjö UME, Jung UW, Choi SH, Pippig S, Siedler M, Kim CK. Periodontal wound healing/regeneration following implantation of recombinant human growth/differentiation factor-5 in a beta-tricalcium phosphate carrier into one-wall intrabony defects in dogs. J Clin Periodontol 2010; 37:382-9. [PMID: 20447262 DOI: 10.1111/j.1600-051x.2010.01544.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Recombinant human growth/differentiation factor-5 (rhGDF-5) is being evaluated as a candidate therapy in support of periodontal regeneration. The objective of this study was to evaluate periodontal wound healing/regeneration following the application of rhGDF-5 on a particulate beta-tricalcium phosphate (beta-TCP) carrier using an established defect model. MATERIALS AND METHODS Bilateral 4 x 5 mm (width x depth), one-wall, critical-size, intrabony periodontal defects were surgically created at the mandibular second and fourth pre-molar teeth in 15 Beagle dogs. Unilateral defects in five animals received rhGDF-5/beta-TCP (Scil Technology GmbH); five animals received beta-TCP solo; and five animals served as sham-surgery controls. Contralateral sites received treatments reported elsewhere. The animals were sacrificed following an 8-week healing interval for histological examination. RESULTS Clinical healing was generally uneventful. Sites implanted with rhGDF-5/beta-TCP exhibited greater enhanced cementum and bone formation compared with beta-TCP and sham-surgery controls; cementum regeneration averaged (+/- SD) 3.83 +/- 0.73 versus 1.65 +/- 0.82 and 2.48 +/- 1.28 mm for the controls (p<0.05). Corresponding values for bone regeneration height averaged 3.26 +/- 0.30 versus 1.70 +/- 0.66 and 1.68 +/- 0.49 mm (p<0.05), and bone area 10.45 +/- 2.26 versus 6.31 +/- 2.41 and 3.00 +/- 1.97 mm(2) (p<0.05). Cementum regeneration included cellular/acellular cementum with or without a functionally oriented periodontal ligament. A non-specific connective tissue attachment was evident in the sham-surgery control. Controls exhibited mostly woven bone with primary osteons, whereas rhGDF-5/beta-TCP sites showed a noticeable extent of lamellar bone. Sites receiving rhGDF-5/beta-TCP or beta-TCP showed some residual beta-TCP granules apparently undergoing biodegradation without obvious differences between the sites. Sites receiving beta-TCP alone commonly showed residual beta-TCP granules sequestered in the connective tissue or fibrovascular marrow. CONCLUSION rhGDF-5/beta-TCP has a greater potential to support the regeneration of the periodontal attachment. Long-term studies are necessary to confirm the uneventful maturation of the regenerated tissues.
Collapse
Affiliation(s)
- Jung-Seok Lee
- Department of Periodontology, Research Institute for Periodontal Regeneration, College of Dentistry, Yonsei University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
30
|
Liang H, Wang K, Shimer AL, Li X, Balian G, Shen FH. Use of a bioactive scaffold for the repair of bone defects in a novel reproducible vertebral body defect model. Bone 2010; 47:197-204. [PMID: 20580872 DOI: 10.1016/j.bone.2010.05.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 05/16/2010] [Accepted: 05/17/2010] [Indexed: 11/16/2022]
Abstract
Bone defects in vertebral bodies (VB) usually occur after the reduction of fractures or are caused by bone disease. Besides the treatment of original disease, repair of the bone defect can restore the structure of VB and improve stabilization of the spine to protect the spinal cord nerves. To aid studies of the efficacy of bioengineering techniques for repair of VB, we developed a rat model with a critical size bone defect in VB. Air-motivated burrs were used to create two sizes of bone defect (2 x 3 x 1.5 mm; 2 x 3 x 3 mm) in the anterior part of VB in 6-month-old Fischer 344 rats. Quantitative CT analyses and histological assays demonstrated that neither defects self-repaired by 8 weeks post surgery. Moreover, the tendency of bone formation was monitored in the same animal by serial CT image evaluations, allowing us to demonstrate that there was significant bone growth during the 4- to 6-week period after the creation of the bone defect. We then implanted sintered poly(lactic-co-glycolic acid) (PLGA) microsphere scaffolds loaded with Matrigel with or without recombinant human bone morphogenetic protein 2 (rhBMP2; 2.0 microg rhBMP2/10 microL Matrigel/scaffold) into the bone defect (2 x 3 x 3 mm) in the VB. Bone formation was detected by quantitative analyses of serial CT images, which demonstrated bone growth in rats that received the rhBMP2 implant, in both surrounding areas and inside area of the scaffold. In addition to a rapid increase within 2 weeks of the operation, another significant bone formation period was found between 4 and 8 weeks after the implantation. By contrast, the control group that received the implant without rhBMP2 did not show similar bone formation tendencies. The results of CT analyses were confirmed by histological studies. This study suggests that a critical size bone defect of the anterior VB can be developed in a rat model. Characterization of this model demonstrated that 4 to 6 weeks after creation of the defect was a significant bone growth period for VB bone repair in rats. This animal model has further utility for the study of different biomaterials for VB bone repair. Implantation of a bioactive PLGA scaffold carrying rhBMP2 allowed more successful repair of the VB defect. Although further characterization studies are needed, the bioactive PLGA scaffold developed in this study will likely adapt easily to other in vivo osteogenesis applications.
Collapse
Affiliation(s)
- Haixiang Liang
- Department of Orthopaedic Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | | | | | | | | |
Collapse
|
31
|
Shi X, Wang Y, Varshney RR, Ren L, Gong Y, Wang DA. Microsphere-based drug releasing scaffolds for inducing osteogenesis of human mesenchymal stem cells in vitro. Eur J Pharm Sci 2009; 39:59-67. [PMID: 19895885 DOI: 10.1016/j.ejps.2009.10.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2009] [Revised: 10/15/2009] [Accepted: 10/25/2009] [Indexed: 01/09/2023]
Abstract
In this study, in vitro osteogenesis was successfully achieved in human mesenchymal stem cells (hMSCs) by controlled release of the osteogenesis-inducing drugs dexamethasone, ascorbic acid (AA) and beta-glycerophosphate (GP) from poly(lactic-co-glycolic acid) (PLGA) sintered microsphere scaffolds (SMS). We investigated the osteogenesis of human MSCs (hMSCs) on dexamethasone laden PLGA-SMS (PLGA-Dex-SMS), and dexamethasone, AA and GP laden PLGA-SMS (PLGA-Com-SMS). hMSCs cultured on the microsphere systems, which act as drug release vehicles and also promote cell growth/tissue formation-displayed a strong osteogenic commitment locally. The osteogenic commitment of hMSCs on the scaffolds were verified by alkaline phosphatase (ALP) activity assay, calcium secretion assay, real-time PCR and immunohistochemistry analysis. The results indicated hMSCs cultured on PLGA-Com-SMS exhibited superior osteogenic differentiation owing to significantly high phenotypic expression of typical osteogenic genes-osteocalcin (OC), type I collagen, alkaline phosphatase (ALP), and Runx-2/Cbfa-1, and protein secretion of bone-relevant markers such as osteoclast and type I collagen when compared with PLGA-Dex-SMS. In conclusion, by promoting osteogenic development of hMSCs in vitro, this newly designed controlled release system opens a new door to bone reparation and regeneration.
Collapse
Affiliation(s)
- Xuetao Shi
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | | | | | | | | | | |
Collapse
|
32
|
Zhao Y, Lin H, Zhang J, Chen B, Sun W, Wang X, Zhao W, Xiao Z, Dai J. Crosslinked three-dimensional demineralized bone matrix for the adipose-derived stromal cell proliferation and differentiation. Tissue Eng Part A 2009; 15:13-21. [PMID: 18652539 DOI: 10.1089/ten.tea.2008.0039] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Stem cell-based therapy has been a promising method for tissue regeneration and wound repair. Adult adipose-derived stromal cells (ADSCs) are often used for adipose and bone tissue reconstruction because of their abundant sources and multipotential differentiation ability. When combined with carriers, ADSCs could be useful for constructing tissue substitutes in vitro or facilitating tissue regeneration in vivo. Demineralized bone matrix (DBM) has been used for tissue reconstruction because collagen presents good cell compatibility. However, DBM degrades rapidly when used for three-dimensional ADSC culture. Here DBM was crosslinked with 1-ethyl-3-(3-dimethyl aminopropyl) carbodiimide and N-hydroxysulfosuccinimide to investigate whether crosslinked DBM (CRL-DBM) could be used as ADSC carrier. CRL-DBM showed not only improved mechanical property and enhanced stability, but also sustained ADSC proliferation and effective differentiation into adipocytes and bone lineage cells. The results indicated that CRL-DBM may be a suitable ADSC carrier for adipose and bone tissue regeneration.
Collapse
Affiliation(s)
- Yannan Zhao
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Boschetti F, Tomei AA, Turri S, Swartz MA, Levi M. Design, fabrication, and characterization of a composite scaffold for bone tissue engineering. Int J Artif Organs 2009; 31:697-707. [PMID: 18825642 DOI: 10.1177/039139880803100803] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Poly(lactide-co-glycolide) (PLGA) scaffolds have been successfully used in bone tissue engineering, with or without hydroxyapatite (HA) and with a macroporosity given either by simple PLGA sphere packaging and/or by leaching out NaCl. The objective of this work was the optimization of the design parameters for bone tissue engineering scaffolds made by sintering microspheres of PLGA, HA nanocrystals for matrix reinforcement and osteoconduction, and salt crystals for macroporosity and control of matrix pore size. Microsphere fabrication by a single-emulsion and solvent evaporation technique was first optimized to obtain a high yield of PLGA microspheres with a diameter between 80 and 300 microm. The influence of the sintering process and matrix composition on the scaffold structure was then evaluated morphologically and mechanically. Three scaffold types were tested for biocompatibility by culturing with human fibroblasts for up to 14 days. The most important parameters to obtain microspheres with the selected diameter range were the viscosity ratio of the dispersed phase to the continuous phase and the relative volume fraction of the 2 phases. The Young's modulus and the ultimate strength of the sintered matrices ranged between 168-265 MPa and 6-17 MPa, respectively, within the range for trabecular bone. Biocompatibility was demonstrated by fibroblast adhesion, proliferation, and spreading throughout the matrix. This work builds upon previous work of the PLGA/HA sintering technique to give design criteria for fabricating a bone tissue engineered matrix with optimized morphological, functional, and biological properties to fit the requirements of bone replacements.
Collapse
Affiliation(s)
- F Boschetti
- LaBS, Department of Structural Engineering, Politecnico di Milano, Milan and IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.
| | | | | | | | | |
Collapse
|
34
|
Gruber RM, Ludwig A, Merten HA, Pippig S, Kramer FJ, Schliephake H. Sinus floor augmentation with recombinant human growth and differentiation factor-5 (rhGDF-5): a pilot study in the Goettingen miniature pig comparing autogenous bone and rhGDF-5. Clin Oral Implants Res 2008; 20:175-82. [PMID: 19077151 DOI: 10.1111/j.1600-0501.2008.01628.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
AIM The aim of this study was to test the hypothesis that recombinant human growth and differentiation factor-5 (rhGDF-5) in combination with a beta-tricalcium phosphate (beta-TCP) scaffold material results in superior bone formation in sinus floor augmentations in miniature pigs compared with a particulated autogenous bone graft combined with the scaffold material. MATERIAL AND METHODS Six adult female Goettingen minipigs underwent a maxillary sinus floor augmentation procedure. In a split-mouth design, the sinus floors were augmented with beta-TCP mixed with autogenous cortical bone chips, in a ratio of approximately 1 : 1, on one side. The contralateral test site was augmented using beta-TCP coated with two concentrations of rhGDF-5 (400 microg rhGDF-5/g beta-TCP or 800 microg rhGDF-5/g beta-TCP; three animals in each case). Simultaneously, one dental implant was inserted into each sinus floor augmentation. After 12 weeks, a histological and histomorphometric assessment of non-decalcified histological specimens was made. RESULTS There were significantly higher mean values of volume density of newly formed bone using beta-TCP coated with two concentrations of rhGDF-5 (400 microg: 32.9%; 800 microg: 23.9%) than with the corresponding control (autogenous bone/beta-TCP) (14.6%, 12.9%) (P=0.012, P=0.049). The bone-to-implant contact rates (BIC) were significantly enhanced in test sites (400 microg: 84.2%; 800 microg: 69.8%) compared with the corresponding control sites (24.8%, 40.8%) (P=.027, P=.045). CONCLUSION rhGDF-5 delivered on beta-TCP significantly enhanced bone formation compared with beta-TCP combined with autogenous bone in sinus lift procedures in miniature pigs.
Collapse
Affiliation(s)
- Rudolf Matthias Gruber
- Department of Oral and Maxillofacial Surgery, Georgia Augusta University, Goettingen, Germany.
| | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Arpornmaeklong P, Pripatnanont P, Suwatwirote N. Properties of chitosan–collagen sponges and osteogenic differentiation of rat-bone-marrow stromal cells. Int J Oral Maxillofac Surg 2008; 37:357-66. [DOI: 10.1016/j.ijom.2007.11.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Revised: 09/16/2007] [Accepted: 11/27/2007] [Indexed: 10/22/2022]
|
37
|
Shen FH, Marks I, Shaffrey C, Ouellet J, Arlet V. The use of an expandable cage for corpectomy reconstruction of vertebral body tumors through a posterior extracavitary approach: a multicenter consecutive case series of prospectively followed patients. Spine J 2008; 8:329-39. [PMID: 17923442 DOI: 10.1016/j.spinee.2007.05.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2007] [Revised: 03/24/2007] [Accepted: 05/02/2007] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Posterior decompressions in the form of laminectomies for vertebral body tumors have poor outcomes. Surgical management typically requires anterior decompression and reconstruction; however, these procedures can be associated with significant morbidity and mortality. PURPOSE To evaluate the feasibility of anterior spinal column reconstruction using an expandable cage through a posterior approach. STUDY DESIGN/SETTING Multicenter consecutive case series of 21 prospectively followed patients. PATIENT SAMPLE Twenty-one patients with vertebral body tumors treated with anterior and posterior resection and reconstruction from a single posterior approach were followed prospectively. OUTCOME MEASURES Pre- and postoperative neurologic status, number of levels instrumented and fused, length of surgery, length of stay after surgery, and complications related directly or indirectly to surgery were analyzed. In addition, pre- and postoperative radiographs, computed tomography (CT) scans, and magnetic resonance imaging (MRI) scans were evaluated for involvement of the vertebral body and associated posterior elements. Particular attention was paid to the presence of either unilateral or bilateral pedicle and/or middle column involvement. METHODS Patients were placed in a prone position on a rotating radiolucent table. Corpectomy was performed from an extracavitary approach, and anterior column reconstruction was completed with an expandable cage. The posterior tension band and spinal fusion was completed with segmental pedicle screw fixation and performed through the same posterior exposure. No patient required a separate anterior procedure. RESULTS Patients' average age was 60.3 years (range, 17-78); there were 12 women and 9 men. Eighteen underwent single-level corpectomies (11 thoracic and 7 lumbar), and 3 underwent two-level corpectomies (T4-T5, T11-T12, and T12-L1). Average estimated blood loss (EBL) and length of surgery per level were 1,360 cc (range, 200-2,500) and 5.3 hours (range, 2.7-8.6), respectively. Average postoperative stay was 4.7 days. Nine patients had at least one partial motor grade improvement. One patient had postoperative left lower extremity weakness after surgical decompression and reconstruction secondary to iatrogenic nerve root traction but remained ambulatory. No chest tubes or postoperative bracing was required. At the most recent follow-up, six patients were alive at an average of 16.1 months (range, 3-33). For the 15 patients who died, the average life span after surgery was 6.8 months (range, 1-16). In addition to the iatrogenic nerve root injury, one cage required repositioning on postoperative Day 2 and one cage demonstrated radiographic evidence of settling but did not require surgical intervention; there were no deep venous thromboses (DVTs), pneumothoraces, pneumonias, ileus, or other complications, with a total complication rate of 14.3%. CONCLUSIONS This is the largest study that specifically examines the use of an expandable cage through a posterior extracavitary approach for reconstruction after vertebral body tumor resection. The use of an expandable cage combined with an extracavitary approach is feasible and allows the surgeon to address both the anterior and posterior columns through a single incision. Although technically challenging, both one- and two-level corpectomies in the thoracic and/or lumbar spine can be performed with this technique. Furthermore, insertion of the expandable cage in the collapsed position and then expansion in situ after implantation allowed for all lumbar reconstructions to be completed without sacrificing any of the lumbar nerve roots. Our 14.3% complication rate is similar to those reported in anterior-alone and circumferential spinal procedures.
Collapse
Affiliation(s)
- Francis H Shen
- Department of Orthopaedic Surgery, University of Virginia, PO Box 800159, Charlottesville, VA 22908-0159, USA.
| | | | | | | | | |
Collapse
|
38
|
Abstract
Biologic therapies to promote fracture-healing such as use of bone morphogenetic proteins (BMPs) are being increasingly employed in multiple clinical scenarios. However, it has been challenging to design therapies that deliver sufficient quantities of protein over a sustained time period. A potential solution is the application of gene therapy that transfers genetic information to host cells at the fracture site, resulting in the continuous and localized production of the desired proteins. This approach has demonstrated tremendous potential in preclinical animal models of fracture-healing. This article will review the current state of gene therapy approaches to fracture-healing with an emphasis on potential clinical applications.
Collapse
|
39
|
Abstract
The umbilical cord is a noncontroversial source of mesenchymal-like stem cells. Mesenchymal-like cells are found in several tissue compartments of the umbilical cord, placenta, and decidua. Here, we confine ourselves to discussing mesenchymal-like cells derived from Wharton's Jelly, called umbilical cord matrix stem cells (UCMSCs). Work from several laboratories shows that these cells have therapeutic potential, possibly as a substitute cell for bone marrow-derived mesenchymal stem cells for cellular therapy. There have been no head-to-head comparisons between mesenchymal cells derived from different sources for therapy; therefore, their relative utility is not understood. In this chapter, the isolation protocols of the Wharton's Jelly-derived mesenchymal cells are provided as are protocols for their in vitro culturing and storage. The cell culture methods provided will enable basic scientific research on the UCMSCs. Our vision is that both umbilical cord blood and UCMSCs will be commercially collected and stored in the future for preclinical work, public and private banking services, etc. While umbilical cord blood banking standard operating procedures exist, the scenario mentioned above requires clinical-grade UCMSCs. The hurdles that have been identified for the generation of clinical-grade umbilical cord-derived mesenchymal cells are discussed.
Collapse
|
40
|
Egusa H, Iida K, Kobayashi M, Lin TY, Zhu M, Zuk PA, Wang CJ, Thakor DK, Hedrick MH, Nishimura I. Downregulation of extracellular matrix-related gene clusters during osteogenic differentiation of human bone marrow- and adipose tissue-derived stromal cells. ACTA ACUST UNITED AC 2007; 13:2589-600. [PMID: 17666000 DOI: 10.1089/ten.2007.0080] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Bone marrow- and adipose tissue-derived stromal cells (BMSCs and ASCs, respectively) exhibit a similar capacity for osteogenic differentiation in vitro, but it is unclear whether they share a common differentiation process, because they originate from different tissues. The aim of this study was to explore BMSC and ASC osteogenic differentiation by focusing on the expression of extracellular matrix-related genes (ECMGs), which play a crucial role in osteogenesis and bone tissue regeneration in vivo. We characterized the gene expression profiles of BMSCs and ASCs using a custom complementary deoxyribonucleic acid microarray containing 55 ECMGs. Undifferentiated BMSCs and ASCs actively expressed a wide range of ECMGs. Once BMSCs and ASCs were placed in an osteogenic differentiation medium, 24 and 17 ECMGs, respectively, underwent considerable downregulation over the course of the culture period. The remaining genes were maintained at a similar expression level to corresponding uninduced cell cultures. Although the suppression phenomenon was consistent irrespective of stromal cell origin, collagen (COL)2A1, COL6A1, COL9A1, parathyroid hormone receptor, integrin (INT)-beta3, and TenascinX genes were only downregulated in osteogenic BMSCs, whereas COL1A2, COL3A1, COL4A1, COL5A2, COL15A1, osteopontin, osteonectin, and INT-beta1 genes were only downregulated in osteogenic ASCs. During this time period, cell viability was sustained, suggesting that the observed downregulation did not occur by selection and elimination of unfit cells from the whole cell population. These data suggest that osteogenically differentiating BMSCs and ASCs transition away from a diverse gene expression pattern, reflecting their multipotency toward a configuration specifically meeting the requirements of the target lineage. This change may serve to normalize gene expression in mixed populations of stem cells derived from different tissues.
Collapse
Affiliation(s)
- Hiroshi Egusa
- Jane and Jerry Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, Biomaterials and Hospital Dentistry, UCLA School of Dentistry, Los Angeles, CA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kimelman N, Pelled G, Helm GA, Huard J, Schwarz EM, Gazit D. Review: gene- and stem cell-based therapeutics for bone regeneration and repair. ACTA ACUST UNITED AC 2007; 13:1135-50. [PMID: 17516852 DOI: 10.1089/ten.2007.0096] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Many clinical conditions require regeneration or implantation of bone. This is one focus shared by neurosurgery and orthopedics. Current therapeutic options (bone grafting and protein-based therapy) do not provide satisfying solutions to the problem of massive bone defects. In the past few years, gene- and stem cell-based therapy has been extensively studied to achieve a viable alternative to current solutions offered by modern medicine for bone-loss repair. The use of adult stem cells for bone regeneration has gained much focus. This unique population of multipotential cells has been isolated from various sources, including bone marrow, adipose, and muscle tissues. Genetic engineering of adult stem cells with potent osteogenic genes has led to fracture repair and rapid bone formation in vivo. It is hypothesized that these genetically modified cells exert both an autocrine and a paracrine effects on host stem cells, leading to an enhanced osteogenic effect. The use of direct gene delivery has also shown much promise for in vivo bone repair. Several viral and nonviral methods have been used to achieve substantial bone tissue formation in various sites in animal models. To advance these platforms to the clinical setting, it will be mandatory to overcome specific hurdles, such as control over transgene expression, viral vector toxicity, and prolonged culture periods of therapeutic stem cells. This review covers a prospect of cell and gene therapy for bone repair as well as some very recent advancements in stem cell isolation, genetic engineering, and exogenous control of transgene expression.
Collapse
Affiliation(s)
- Nadav Kimelman
- Skeletal Biotech Lab, The Hebrew University of Jerusalem-Hadassah Medical Campus, Ein Kerem, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
42
|
Graziano A, d'Aquino R, Cusella-De Angelis MG, Laino G, Piattelli A, Pacifici M, De Rosa A, Papaccio G. Concave pit-containing scaffold surfaces improve stem cell-derived osteoblast performance and lead to significant bone tissue formation. PLoS One 2007; 2:e496. [PMID: 17551577 PMCID: PMC1876259 DOI: 10.1371/journal.pone.0000496] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Accepted: 05/09/2007] [Indexed: 01/02/2023] Open
Abstract
Background Scaffold surface features are thought to be important regulators of stem cell performance and endurance in tissue engineering applications, but details about these fundamental aspects of stem cell biology remain largely unclear. Methodology and Findings In the present study, smooth clinical-grade lactide-coglyolic acid 85:15 (PLGA) scaffolds were carved as membranes and treated with NMP (N-metil-pyrrolidone) to create controlled subtractive pits or microcavities. Scanning electron and confocal microscopy revealed that the NMP-treated membranes contained: (i) large microcavities of 80–120 µm in diameter and 40–100 µm in depth, which we termed primary; and (ii) smaller microcavities of 10–20 µm in diameter and 3–10 µm in depth located within the primary cavities, which we termed secondary. We asked whether a microcavity-rich scaffold had distinct bone-forming capabilities compared to a smooth one. To do so, mesenchymal stem cells derived from human dental pulp were seeded onto the two types of scaffold and monitored over time for cytoarchitectural characteristics, differentiation status and production of important factors, including bone morphogenetic protein-2 (BMP-2) and vascular endothelial growth factor (VEGF). We found that the microcavity-rich scaffold enhanced cell adhesion: the cells created intimate contact with secondary microcavities and were polarized. These cytological responses were not seen with the smooth-surface scaffold. Moreover, cells on the microcavity-rich scaffold released larger amounts of BMP-2 and VEGF into the culture medium and expressed higher alkaline phosphatase activity. When this type of scaffold was transplanted into rats, superior bone formation was elicited compared to cells seeded on the smooth scaffold. Conclusion In conclusion, surface microcavities appear to support a more vigorous osteogenic response of stem cells and should be used in the design of therapeutic substrates to improve bone repair and bioengineering applications in the future.
Collapse
Affiliation(s)
- Antonio Graziano
- Dipartimento di Medicina Sperimentale, Sezione di Istologia ed Embriologia, Secondo Ateneo di Napoli, Naples, Italy
- * To whom correspondence should be addressed. E-mail:
| | - Riccardo d'Aquino
- Dipartimento di Medicina Sperimentale, Sezione di Istologia ed Embriologia, Secondo Ateneo di Napoli, Naples, Italy
- Dipartimento di Discipline Odontostomatologiche, Ortodontiche e Chirurgiche, Secondo Ateneo di Napoli, Naples, Italy
| | | | - Gregorio Laino
- Dipartimento di Discipline Odontostomatologiche, Ortodontiche e Chirurgiche, Secondo Ateneo di Napoli, Naples, Italy
| | - Adriano Piattelli
- Dipartimento di Scienze Odontostomatologiche, Università degli Studi “G. d'Annunzio”, Chieti, Italy
| | - Maurizio Pacifici
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Alfredo De Rosa
- Dipartimento di Discipline Odontostomatologiche, Ortodontiche e Chirurgiche, Secondo Ateneo di Napoli, Naples, Italy
| | - Gianpaolo Papaccio
- Dipartimento di Medicina Sperimentale, Sezione di Istologia ed Embriologia, Secondo Ateneo di Napoli, Naples, Italy
| |
Collapse
|
43
|
Graziano A, d'Aquino R, Cusella-De Angelis MG, De Francesco F, Giordano A, Laino G, Piattelli A, Traini T, De Rosa A, Papaccio G. Scaffold's surface geometry significantly affects human stem cell bone tissue engineering. J Cell Physiol 2007; 214:166-72. [PMID: 17565721 DOI: 10.1002/jcp.21175] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In this study, we have observed dental pulp stem cells (SBP-DPSCs) performances on different scaffolds, such as PLGA 85:15, hydroxyapatite chips (HA) and titanium. Stem cells were challenged with each engineered surface, either in plane cultures or in a rotating apparatus, for a month. Gingival fibroblasts were used as controls. Results showed that stem cells exerted a different response, depending on the different type of textured surface: in fact, microconcavities significantly affected SBP-DPSC differentiation into osteoblasts, both temporally and quantitatively, with respect to the other textured surfaces. Actually, stem cells challenged with concave surfaces differentiated quicker and showed nuclear polarity, an index of secretion, cellular activity and matrix formation. Moreover, bone-specific proteins were significantly expressed and the obtained bone tissue was of significant thickness. Thus, cells cultured on the concave textured surface had better cell-scaffold interactions and were induced to secrete factors that, due to their autocrine effects, quickly lead to osteodifferentiation, bone tissue formation, and vascularization. The worst cell performance was obtained using convex surfaces, due to the scarce cell proliferation on to the scaffold and the poor matrix secretion. In conclusion, this study stresses that for a suitable and successful bone tissue reconstruction the surface texture is of paramount importance.
Collapse
Affiliation(s)
- Antonio Graziano
- Dipartimento di Medicina Sperimentale, Sezione di Istologia ed Embriologia, Secondo Ateneo di Napoli, Napoli, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|