151
|
Shimozono Y, Fansa AM, Kennedy JG. The Role of Biologics in the Treatment of Flatfoot. TECHNIQUES IN FOOT & ANKLE SURGERY 2019; 18:160-165. [DOI: 10.1097/btf.0000000000000242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Symptomatic adult-acquired flatfoot deformity (AAFD) is traditionally treated by realignment osteotomy and a tendon transfer. Despite high success rates for this procedure, prolonged recovery time and associated morbidities may lead many patients to shy away from having this type of surgery performed on them. Over the past decade, the use of biologics such as platelet-rich plasma and concentrated bone marrow aspirate concentrate has been gaining much popularity. The efficacy of these biologics to treat tendon pathologies is currently well supported in the literature. Therefore, when treating early AAFD with a functioning posterior tibial tendon, biological agents have the potential to enhance tendon healing and functional recovery. In this review we will be discussing the treatment algorithm which we currently use in our practice to manage AAFD. This will include the use of biologics with minimally invasive procedures, such as posterior tibial tendon tendoscopy and subtalar arthroereisis screws, which have the potential to address the biological and mechanical aspects of this common pathology.
Level of Evidence: Diagnostic Level IV. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
| | - Ashraf M. Fansa
- Department of Foot and Ankle, Hospital for Special Surgery, New York, NY
| | | |
Collapse
|
152
|
Zhang H, Wang Y, Yang G, Yu H, Zhou Z, Tang M. MicroRNA-30a regulates chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells through targeting Sox9. Exp Ther Med 2019; 18:4689-4697. [PMID: 31807153 PMCID: PMC6878886 DOI: 10.3892/etm.2019.8148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 07/05/2019] [Indexed: 12/20/2022] Open
Abstract
Cartilage injury is difficult to repair since the cartilage tissue lacks self-restoration ability. Improved formation of chondrocytes differentiated from the mesenchymal stem cells (MSC) by genetic regulation is a potentially promising therapeutic option. SOX9 is a critical transcription factor for mesenchymal condensation prior to chondrogenesis. Previous studies demonstrated that several microRNAs (miRNAs or miRs) play a critical role in the chondrogenic differentiation of MSCs. However, the interactional relations between miR-30a and SOX9 during chondrogenic differentiation of MSCs need to be further elucidated. In the present study, human bone marrow-derived mesenchymal stem cells have been isolated and induced into chondrogenic differentiation to imitate the cartilage formation in vitro. Additionally, the expression levels of several miRNAs that were reported to interact with the SOX9 3'untranslated region (UTR) were examined by using reverse transcription-quantitative PCR. The interactional relations between candidate miRNAs and SOX9 were verified with the transfection of a miRNA mimic or inhibitor and a luciferase reporter gene assay. The results indicate that miR-30a and miR-195 were consistently increased during MSC chondrogenic differentiation. Additionally, the binding of miR-30a to the SOX9 3UTR was verified. Then, the authors upregulated the expression of miR-30a and found that MSC chondrogenic differentiation was inhibited. Taken together, the results of the present study demonstrate that miR-30a has a negative regulatory effect on MSC chondrogenic differentiation by targeting SOX9. Advances in epigenetic regulating methods will likely be the future of systemic treatment of cartilage injury.
Collapse
Affiliation(s)
- Hongqi Zhang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yunjia Wang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Guanteng Yang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Honggui Yu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhenhai Zhou
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Mingxing Tang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
153
|
Mesenchymal stem cells in the treatment of articular cartilage degeneration: New biological insights for an old-timer cell. Cytotherapy 2019; 21:1179-1197. [DOI: 10.1016/j.jcyt.2019.10.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/10/2019] [Accepted: 10/13/2019] [Indexed: 01/15/2023]
|
154
|
Uzieliene I, Bernotiene E, Rakauskiene G, Denkovskij J, Bagdonas E, Mackiewicz Z, Porvaneckas N, Kvederas G, Mobasheri A. The Antihypertensive Drug Nifedipine Modulates the Metabolism of Chondrocytes and Human Bone Marrow-Derived Mesenchymal Stem Cells. Front Endocrinol (Lausanne) 2019; 10:756. [PMID: 31781032 PMCID: PMC6857108 DOI: 10.3389/fendo.2019.00756] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/18/2019] [Indexed: 11/30/2022] Open
Abstract
Aging is associated with the development of various chronic diseases, in which both cardiovascular disorders and osteoarthritis are dominant. Currently, there is no effective treatment for osteoarthritis, whereas hypertension is often treated with L-type voltage-operated calcium channel blocking drugs, nifedipine being among the most classical ones. Although nifedipine together with other L-type voltage-operated calcium channel inhibitors plays an important role in controlling hypertension, there are unresolved questions concerning its possible effect on cartilage tissue homeostasis and the development of osteoarthritis. The aim of this study was to analyse the effects of nifedipine on metabolic processes in human chondrocytes and bone marrow mesenchymal stem cells. To better understand whether the metabolic effects are mediated specifically through L-type voltage-operated calcium channel, effects of the agonist BayK8644 were analyzed in parallel. Nifedipine downregulated and mitochondrial respiration and ATP production in both cell types. Analysis of cartilage explants by electron microscopy also suggested that a small number of chondrocyte mitochondria's lose their activity in response to nifedipine. Conversely, nifedipine enhanced glycolytic capacity in chondrocytes, suggesting that these cells have the capacity to switch from oxidative phosphorylation to glycolysis and alter their metabolic activity in response to L-type voltage-operated calcium channel inhibition. Such a metabolic switch was not observed in bone marrow mesenchymal stem cells. Nitric oxide activity was upregulated by nifedipine in bone marrow mesenchymal stem cells and particularly in chondrocytes, implying its involvement in the effects of nifedipine on metabolism in both tested cell types. Furthermore, stimulation with nifedipine resulted in elevated production of collagen type II and glycosaminoglycans in micromass cultures under chondrogenic conditions. Taken together, we conclude that the antihypertensive drug nifedipine inhibits mitochondrial respiration in both chondrocytes and bone marrow mesenchymal stem cells and that these effects may be associated with the increased nitric oxide accumulation and pro-inflammatory activity. Nifedipine had positive effects on the production of collagen type II and proteoglycans in both cell types, implying potentially beneficial anabolic responses in articular cartilage. These results highlight a potential link between antihypertensive drugs and cartilage health.
Collapse
Affiliation(s)
- Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Greta Rakauskiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Jaroslav Denkovskij
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Edvardas Bagdonas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Zygmunt Mackiewicz
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | | | | | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, Queen's Medical Centre, Nottingham, United Kingdom
- Sheik Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis With Stem Cells, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
155
|
Scalzone A, Ferreira AM, Tonda-Turo C, Ciardelli G, Dalgarno K, Gentile P. The interplay between chondrocyte spheroids and mesenchymal stem cells boosts cartilage regeneration within a 3D natural-based hydrogel. Sci Rep 2019; 9:14630. [PMID: 31601910 PMCID: PMC6787336 DOI: 10.1038/s41598-019-51070-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/25/2019] [Indexed: 12/30/2022] Open
Abstract
Articular cartilage (AC) lacks the ability to self-repair and cell-based approaches, primarily based on using chondrocytes and mesenchymal stem cells (MSCs), are emerging as effective technology to restore cartilage functionality, because cells synergic functionality may support the maintenance of chondrogenic phenotype and promote extracellular matrix regeneration. This work aims to develop a more physiologically representative co-culture system to investigate the influence of MSCs on the activity of chondrocytes. A thermo-sensitive chitosan-based hydrogel, ionically crosslinked with β-glycerophosphate, is optimised to obtain sol/gel transition at physiological conditions within 5 minutes, high porosity with pores diameter <30 µm, and in vitro mechanical integrity with compressive and equilibrium Young's moduli of 37 kPa and 17 kPa, respectively. Live/dead staining showed that after 1 and 3 days in culture, the encapsulated MSCs into the hydrogels are viable and characterised by round-like morphology. Furthermore chondrocyte spheroids, seeded on top of gels that contained either MSCs or no cells, show that the encapsulated MSCs stimulate chondrocyte activity within a gel co-culture, both in terms of maintaining the coherence of chondrocyte spheroids, leading to a larger quantity of CD44 (by immunofluorescence) and a higher production of collagen and glycosaminoglycans (by histology) compared with the mono-culture.
Collapse
Affiliation(s)
- Annachiara Scalzone
- School of Engineering, Newcastle University, Claremont Road, Newcastle upon Tyne, NE1 7RU, United Kingdom
| | - Ana M Ferreira
- School of Engineering, Newcastle University, Claremont Road, Newcastle upon Tyne, NE1 7RU, United Kingdom
| | - Chiara Tonda-Turo
- Department of Mechanical and Aerospace Engineering (DIMEAS), Politecnico di Torino Corso Duca degli Abruzzi 29, Turin, 10129, Italy
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering (DIMEAS), Politecnico di Torino Corso Duca degli Abruzzi 29, Turin, 10129, Italy
| | - Kenny Dalgarno
- School of Engineering, Newcastle University, Claremont Road, Newcastle upon Tyne, NE1 7RU, United Kingdom
| | - Piergiorgio Gentile
- School of Engineering, Newcastle University, Claremont Road, Newcastle upon Tyne, NE1 7RU, United Kingdom.
| |
Collapse
|
156
|
Salonius E, Kontturi L, Laitinen A, Haaparanta AM, Korhonen M, Nystedt J, Kiviranta I, Muhonen V. Chondrogenic differentiation of human bone marrow-derived mesenchymal stromal cells in a three-dimensional environment. J Cell Physiol 2019; 235:3497-3507. [PMID: 31552691 DOI: 10.1002/jcp.29238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
Cell therapy combined with biomaterial scaffolds is used to treat cartilage defects. We hypothesized that chondrogenic differentiation bone marrow-derived mesenchymal stem cells (BM-MSCs) in three-dimensional biomaterial scaffolds would initiate cartilaginous matrix deposition and prepare the construct for cartilage regeneration in situ. The chondrogenic capability of human BM-MSCs was first verified in a pellet culture. The BM-MSCs were then either seeded onto a composite scaffold rhCo-PLA combining polylactide and collagen type II (C2) or type III (C3), or commercial collagen type I/III membrane (CG). The BM-MSCs were either cultured in a proliferation medium or chondrogenic culture medium. Adult human chondrocytes (ACs) served as controls. After 3, 14, and 28 days, the constructs were analyzed with quantitative polymerase chain reaction and confocal microscopy and sulfated glycosaminoglycans (GAGs) were measured. The differentiated BM-MSCs entered a hypertrophic state by Day 14 of culture. The ACs showed dedifferentiation with no expression of chondrogenic genes and low amount of GAG. The CG membrane induced the highest expression levels of hypertrophic genes. The two different collagen types in composite scaffolds yielded similar results. Regardless of the biomaterial scaffold, culturing BM-MSCs in chondrogenic differentiation medium resulted in chondrocyte hypertrophy. Thus, caution for cell fate is required when designing cell-biomaterial constructs for cartilage regeneration.
Collapse
Affiliation(s)
- Eve Salonius
- Department of Orthopaedics and Traumatology, Clinicum, University of Helsinki, Helsinki, Finland
| | - Leena Kontturi
- Drug Research Program, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Anita Laitinen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Anne-Marie Haaparanta
- Department of Electronics and Communications Engineering, Tampere University of Technology and BioMediTech, Tampere, Finland
| | - Matti Korhonen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Johanna Nystedt
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Ilkka Kiviranta
- Department of Orthopaedics and Traumatology, Clinicum, University of Helsinki, Helsinki, Finland.,Department of Orthopaedics and Traumatology, Helsinki University Hospital, Helsinki, Finland
| | - Virpi Muhonen
- Department of Orthopaedics and Traumatology, Clinicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
157
|
Huang MJ, Zhao JY, Xu JJ, Li J, Zhuang YF, Zhang XL. lncRNA ADAMTS9-AS2 Controls Human Mesenchymal Stem Cell Chondrogenic Differentiation and Functions as a ceRNA. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 18:533-545. [PMID: 31671346 PMCID: PMC6838486 DOI: 10.1016/j.omtn.2019.08.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/17/2019] [Accepted: 08/23/2019] [Indexed: 12/17/2022]
Abstract
Long noncoding RNAs (lncRNAs) have emerged as key regulators of cell differentiation and development. However, potential roles for lncRNAs in chondrogenic differentiation have remained poorly understood. Here we identify lncRNA ADAMTS9 antisense RNA 2, ADAMTS9-AS2, which controls the chondrogenic differentiation by acting as a competing endogenous RNA (ceRNA) in human mesenchymal stem cells (hMSCs). We screen out ADAMTS9-AS2 of undifferentiated and differentiated cells during chondrogenic differentiation by microarrays. Suppression or overexpression of lncRNA ADAMTS9-AS2 correlates with inhibition and promotion of hMSC chondrogenic differentiation, respectively. We find that ADAMTS9-AS2 can sponge miR-942-5p to regulate the expression of Scrg1, a transcription factor promoting chondrogenic gene expression. Finally, we confirm the function of ADAMTS9-AS2 to cartilage repair in the absence of transforming growth factor β (TGF-β) in vivo. In conclusion, ADAMTS9-AS2 plays an important role in chondrogenic differentiation as a ceRNA, so that it can be regarded as a therapy target for cartilage repair.
Collapse
Affiliation(s)
- Ming-Jian Huang
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Jing-Yu Zhao
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Jia-Jia Xu
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Jing Li
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Yi-Fu Zhuang
- Department of Orthopedic Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 201999, China
| | - Xiao-Ling Zhang
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| |
Collapse
|
158
|
Vail DJ, Somoza RA, Caplan AI, Khalil AM. Transcriptome dynamics of long noncoding RNAs and transcription factors demarcate human neonatal, adult, and human mesenchymal stem cell-derived engineered cartilage. J Tissue Eng Regen Med 2019; 14:29-44. [PMID: 31503387 DOI: 10.1002/term.2961] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 08/02/2019] [Accepted: 09/03/2019] [Indexed: 11/08/2022]
Abstract
The engineering of a native-like articular cartilage (AC) is a long-standing objective that could serve the clinical needs of millions of patients suffering from osteoarthritis and cartilage injury. An incomplete understanding of the developmental stages of AC has contributed to limited success in this endeavor. Using next generation RNA sequencing, we have transcriptionally characterized two critical stages of AC development in humans-that is, immature neonatal and mature adult, as well as tissue-engineered cartilage derived from culture expanded human mesenchymal stem cells. We identified key transcription factors (TFs) and long noncoding RNAs (lncRNAs) as candidate drivers of the distinct phenotypes of these tissues. AGTR2, SCGB3A1, TFCP2L1, RORC, and TBX4 stand out as key TFs, whose expression may be capable of reprogramming engineered cartilage into a more expandable and neonatal-like cartilage primed for maturation into biomechanically competent cartilage. We also identified that the transcriptional profiles of many annotated but poorly studied lncRNAs were dramatically different between these cartilages, indicating that lncRNAs may also be playing significant roles in cartilage biology. Key neonatal-specific lncRNAs identified include AC092818.1, AC099560.1, and KC877982. Collectively, our results suggest that tissue-engineered cartilage can be optimized for future clinical applications by the specific expression of TFs and lncRNAs.
Collapse
Affiliation(s)
- Daniel J Vail
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Rodrigo A Somoza
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, OH
| | - Arnold I Caplan
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, OH
| | - Ahmad M Khalil
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
159
|
Razmara E, Bitaraf A, Yousefi H, Nguyen TH, Garshasbi M, Cho WCS, Babashah S. Non-Coding RNAs in Cartilage Development: An Updated Review. Int J Mol Sci 2019; 20:4475. [PMID: 31514268 PMCID: PMC6769748 DOI: 10.3390/ijms20184475] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/02/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023] Open
Abstract
In the development of the skeleton, the long bones are arising from the process of endochondral ossification (EO) in which cartilage is replaced by bone. This complex process is regulated by various factors including genetic, epigenetic, and environmental elements. It is recognized that DNA methylation, higher-order chromatin structure, and post-translational modifications of histones regulate the EO. With emerging understanding, non-coding RNAs (ncRNAs) have been identified as another mode of EO regulation, which is consist of microRNAs (miRNAs or miRs) and long non-coding RNAs (lncRNAs). There is expanding experimental evidence to unlock the role of ncRNAs in the differentiation of cartilage cells, as well as the pathogenesis of several skeletal disorders including osteoarthritis. Cutting-edge technologies such as epigenome-wide association studies have been employed to reveal disease-specific patterns regarding ncRNAs. This opens a new avenue of our understanding of skeletal cell biology, and may also identify potential epigenetic-based biomarkers. In this review, we provide an updated overview of recent advances in the role of ncRNAs especially focus on miRNA and lncRNA in the development of bone from cartilage, as well as their roles in skeletal pathophysiology.
Collapse
Affiliation(s)
- Ehsan Razmara
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran P.O. Box 14115-111, Iran
| | - Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran P.O. Box 14115-111, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, LA 70112, USA
| | - Tina H Nguyen
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, LA 70112, USA
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran P.O. Box 14115-111, Iran
| | | | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran P.O. Box 14115-111, Iran.
| |
Collapse
|
160
|
Kwon H, Brown WE, Lee CA, Wang D, Paschos N, Hu JC, Athanasiou KA. Surgical and tissue engineering strategies for articular cartilage and meniscus repair. Nat Rev Rheumatol 2019; 15:550-570. [PMID: 31296933 PMCID: PMC7192556 DOI: 10.1038/s41584-019-0255-1] [Citation(s) in RCA: 413] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2019] [Indexed: 12/30/2022]
Abstract
Injuries to articular cartilage and menisci can lead to cartilage degeneration that ultimately results in arthritis. Different forms of arthritis affect ~50 million people in the USA alone, and it is therefore crucial to identify methods that will halt or slow the progression to arthritis, starting with the initiating events of cartilage and meniscus defects. The surgical approaches in current use have a limited capacity for tissue regeneration and yield only short-term relief of symptoms. Tissue engineering approaches are emerging as alternatives to current surgical methods for cartilage and meniscus repair. Several cell-based and tissue-engineered products are currently in clinical trials for cartilage lesions and meniscal tears, opening new avenues for cartilage and meniscus regeneration. This Review provides a summary of surgical techniques, including tissue-engineered products, that are currently in clinical use, as well as a discussion of state-of-the-art tissue engineering strategies and technologies that are being developed for use in articular cartilage and meniscus repair and regeneration. The obstacles to clinical translation of these strategies are also included to inform the development of innovative tissue engineering approaches.
Collapse
Affiliation(s)
- Heenam Kwon
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Wendy E Brown
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Cassandra A Lee
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA, USA
| | - Dean Wang
- Department of Orthopaedic Surgery, University of California Irvine Medical Center, Orange, CA, USA
| | - Nikolaos Paschos
- Division of Sports Medicine, Department of Orthopaedic Surgery, New England Baptist Hospital, Tufts University School of Medicine, Boston, MA, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
161
|
The Combination of TGF-β3 and BMP-6 Synergistically Promotes the Chondrogenic Differentiation of Equine Bone Marrow-Derived Mesenchymal Stem Cells. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09880-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
162
|
Yang J, Li Y, Liu Y, Li D, Zhang L, Wang Q, Xiao Y, Zhang X. Influence of hydrogel network microstructures on mesenchymal stem cell chondrogenesis in vitro and in vivo. Acta Biomater 2019; 91:159-172. [PMID: 31055122 DOI: 10.1016/j.actbio.2019.04.054] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/21/2019] [Accepted: 04/24/2019] [Indexed: 12/27/2022]
Abstract
Hydrogels, which provide three-dimensional (3D) niches for encapsulating bone marrow mesenchymal stem cells (BMSCs), are becoming a promising tissue engineering solution for chondrogenic differentiation of BMSCs. However, it remains a challenge to design a hydrogel material for effective chondrogenesis of BMSCs because of the complexity of cartilage ECM and cell-matrix interactions. Thus far, various studies have shown the physical-chemical cues of hydrogel materials to impact BMSCs chondrogenesis, but the design of the 3D network microstructure of the hydrogel to induce BMSCs chondrogenesis is still far from optimized. In this study, we successfully prepared two types of collagen hydrogels, namely, the fibrous network and porous network, with the same chemical composition and similar mechanical strength but with two distinct network microstructures. The two different network microstructures significantly influenced mass transfer, protein adsorption, degradability, and contraction of the collagen hydrogels. Moreover, the cells presented distinct proliferation and morphology in the two hydrogels, which consequently modulated chondrogenic differentiation of BMSCs derived from rat. Collagen hydrogels with a fibrous network promoted more chondrogenic differentiation of BMSCs without additional growth factors in vitro and subcutaneous implantation in vivo than those with a porous network. Moreover, fibrous network resulted in less ECM calcification than porous network. However, the fibrous network could not prevent hypertrophy of the chondrogenic cells induced by BMSCs. Overall, these results revealed that the 3D network microstructure of a hydrogel was a key design parameter for the chondrogenic differentiation of BMSCs. STATEMENT OF SIGNIFICANCE: Hydrogels had been used to induce the chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in cartilage tissue engineering, but the key design parameters remain unoptimized. This was mainly due to the different material properties including composition, strength, and microstructure, which would interplay with each other and result in difficulties to investigate the effects for one factor. In this study, we fabricated two collagen hydrogels with the same chemical composition and mechanical strength, but two distinct network microstructures. The effects of the two network microstructures on the chondrogenic differentiation of BMSCs were investigated by in vitro and in vivo assays. The results highlight the effects of network microstructures and provide important information about optimizing the design of future hydrogels in cartilage tissue engineering.
Collapse
Affiliation(s)
- Jirong Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| | - Yuanqi Li
- Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yanbo Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| | - Dongxiao Li
- Sichuan Academy of Chinese Medicine Science, Chengdu 61004, Sichuan, China
| | - Lei Zhang
- Sichuan Academy of Chinese Medicine Science, Chengdu 61004, Sichuan, China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China.
| | - Yumei Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| |
Collapse
|
163
|
Atkinson SP. A Preview of Selected Articles. Stem Cells Transl Med 2019. [PMCID: PMC6525554 DOI: 10.1002/sctm.19-0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
164
|
Dissecting the Pharmacodynamics and Pharmacokinetics of MSCs to Overcome Limitations in Their Clinical Translation. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:1-15. [PMID: 31236426 PMCID: PMC6581775 DOI: 10.1016/j.omtm.2019.05.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recently, mesenchymal stromal stem cells (MSCs) have been proposed as therapeutic agents because of their promising preclinical features and good safety profile. However, their introduction into clinical practice has been associated with a suboptimal therapeutic profile. In this review, we address the biodistribution of MSCs in preclinical studies with a focus on the current understanding of the pharmacodynamics (PD) and pharmacokinetics (PK) of MSCs as key aspects to overcome unsatisfactory clinical benefits of MSC application. Beginning with evidence of MSC biodistribution and highlighting PK and PD factors, a new PK-PD model is also proposed. According to this theory, MSCs and their released factors are key players in PK, and the efficacy biomarkers are considered relevant for PD in more predictive preclinical investigations. Accounting for the PK-PD relationship in MSC translational research and proposing new models combined with better biodistribution studies could allow realization of the promise of more robust MSC clinical translation.
Collapse
|
165
|
Kavand H, Rahaie M, Koohsorkhi J, Haghighipour N, Bonakdar S. A conductive cell-imprinted substrate based on CNT-PDMS composite. Biotechnol Appl Biochem 2019; 66:445-453. [PMID: 30817028 DOI: 10.1002/bab.1741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 02/24/2019] [Indexed: 11/08/2022]
Abstract
Cell function regulation is influenced by continuous biochemical and biophysical signal exchange within the body. Substrates with nano/micro-scaled topographies that mimic the physiological niche are widely applied for tissue engineering applications. As the cartilage niche is composed of several stimulating factors, a multifunctional substrate providing topographical features while having the capability of electrical stimulation is presented. Herein, we demonstrate a biocompatible and conductive chondrocyte cell-imprinted substrate using polydimethylsiloxane (PDMS) and carbon nanotubes (CNTs) as conductive fillers. Unlike the conventional silicon wafers or structural photoresist masters used for molding, cell surface topographical replication is challenging as biological cells showed extremely sensitive to chemical solvent residues during molding. The composite showed no significant difference compared with PDMS with regard to cytotoxicity, whereas an enhanced cell adhesion was observed on the conductive composite's surface. Integration of nanomaterials into the cell seeding scaffolds can make tissue regeneration process more efficient.
Collapse
Affiliation(s)
- Hanie Kavand
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Mahdi Rahaie
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Javad Koohsorkhi
- Advanced Micro and Nano Devices Lab, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | | | - Shahin Bonakdar
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
166
|
Kavand H, van Lintel H, Renaud P. Efficacy of pulsed electromagnetic fields and electromagnetic fields tuned to the ion cyclotron resonance frequency of Ca 2+ on chondrogenic differentiation. J Tissue Eng Regen Med 2019; 13:799-811. [PMID: 30793837 DOI: 10.1002/term.2829] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/05/2019] [Accepted: 02/21/2019] [Indexed: 12/17/2022]
Abstract
Previous studies provide strong evidence for the therapeutic effect of electromagnetic fields (EMFs) on different tissues including cartilage. Diverse exposure parameters applied in scientific reports and the unknown interacting mechanism of EMF with biological systems make EMF studies challenging. In 1985, Liboff proposed that when magnetic fields are tuned to the cyclotron resonance frequencies of critical ions, the motion of ions through cell membranes is enhanced, and thus biological effects appear. Such exposure system consists of a weak alternating magnetic field (B1 ) in the presence of a static magnetic field (B0 ) and depends on the relationship between the magnitudes of B0 and B1 and the angular frequency Ω. The purpose of the present study is to determine the chondrogenic potential of EMF with regards to pulsed EMF (PEMF) and the ion cyclotron resonance (ICR) theory. We used different stimulating systems to generate EMFs in which cells are either stimulated with ubiquitous PEMF parameters, frequently reported, or parameters tuned to satisfy the ICR for Ca2+ (including negative and positive control groups). Chondrogenesis was analysed after 3 weeks of treatment. Cell stimulation under the ICR condition showed positive results in the context of glycosaminoglycans and type II collagen synthesis. In contrast, the other electromagnetically stimulated groups showed no changes compared with the control groups. Furthermore, gene expression assays revealed an increase in the expression of chondrogenic markers (COL2A1, SOX9, and ACAN) in the ICR group. These results suggest that the Ca2+ ICR condition can be an effective factor in inducing chondrogenesis.
Collapse
Affiliation(s)
- Hanie Kavand
- Microsystems Laboratory, Institute of Microengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Harald van Lintel
- Microsystems Laboratory, Institute of Microengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Philippe Renaud
- Microsystems Laboratory, Institute of Microengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
167
|
Human Diseased Articular Cartilage Contains a Mesenchymal Stem Cell-Like Population of Chondroprogenitors with Strong Immunomodulatory Responses. J Clin Med 2019; 8:jcm8040423. [PMID: 30925656 PMCID: PMC6517884 DOI: 10.3390/jcm8040423] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 02/07/2023] Open
Abstract
Background: osteoarthritic human articular cartilage (AC)-derived cartilage cells (CCs) with same-donor bone marrow (BMSCs) and adipose tissue (ASCs)-derived mesenchymal stem cells were compared, in terms of stemness features, and secretory and immunomodulatory responses to inflammation. Methods: proteoglycan 4 (PRG4) presence was evaluated in AC and CCs. MSCs and CCs (n = 8) were cultured (P1 to P4) and characterized for clonogenicity, nanog homeobox (NANOG), and POU class 5 homeobox 1 (POU5F1) expression, immunotypification, and tri-lineage differentiation. Their basal and interleukin-1β (IL-1β)-stimulated expression of matrix metalloproteases (MMPs), tissue inhibitors (TIMPs), release of growth factors, and cytokines were analyzed, along with the immunomodulatory ability of CCs. Results: PRG4 was mainly expressed in the intact AC surface, whereas shifted to the intermediate zone in damaged cartilage and increased its expression in CCs upon culture. All cells exhibited a similar phenotype and stemness maintenance over passages. CCs showed highest chondrogenic ability, no adipogenic potential, a superior basal secretion of growth factors and cytokines, the latter further increased after inflammatory stimulation, and an immunomodulatory behavior. All stimulated cells shared an increased MMP expression without a corresponding TIMP production. Conclusion: based on the observed features, CCs obtained from pathological joints may constitute a potential tissue-specific therapeutic target or agent to improve damaged cartilage healing, especially damage caused by inflammatory/immune mediated conditions.
Collapse
|
168
|
Rodriguez-Merchan EC, Valentino LA. The Role of Gene Therapy in Cartilage Repair. THE ARCHIVES OF BONE AND JOINT SURGERY 2019; 7:79-90. [PMID: 31211186 PMCID: PMC6510927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 09/06/2018] [Indexed: 06/09/2023]
Abstract
The key principle of gene delivery to articulations by direct intra-articular injection is to release complementary DNA (cDNA)-encoding medical products that will lead to maintained, endogenous production of the gene products within the articulation. In fact, this has been accomplished for both in vivo and ex vivo gene delivery, using several vectors, genes, and cells in some animal models. Some clinical trials for rheumatoid arthritis and osteoarthritis (OA) using retrovirus vectors for ex vivo gene delivery and adeno-associated virus (AAV) for in vivo delivery have been reported. AAV is of special attention because, contrary to other viral vectors, it can enter deep within joint cartilage and transduce chondrocytes in situ. This quality is of special significance in OA, in which modifications in chondrocyte metabolism are believed to be crucial to the pathophysiology of the disease. The clinical effectiveness of TissueGene-C (TG-C), a cell and gene therapy for OA consisting of nontransformed and transduced chondrocytes (3:1) retrovirally transduced to overexpress TGF-β1 has been reported in patients with knee OA. The most common complications of TG-C were peripheral edema (9%), arthralgia (8%), articular swelling (6%), and injection site pain (5%). TG-C was associated with relevant ameliorations in function and pain. Gene therapy appears to be a viable method for the management of articular cartilage defects and OA.
Collapse
Affiliation(s)
- E Carlos Rodriguez-Merchan
- Department of Orthopaedic Surgery and La Paz Research Institute ("Instituto de Investigación La Paz - IdiPaz"), "La Paz" University Hospital, Madrid, Spain
- Rush University, Chicago, Illinois, USA
- Research performed at Department of Orthopaedic Surgery, La Paz University Hospital, Madrid, Spain
| | - Leonard A Valentino
- Department of Orthopaedic Surgery and La Paz Research Institute ("Instituto de Investigación La Paz - IdiPaz"), "La Paz" University Hospital, Madrid, Spain
- Rush University, Chicago, Illinois, USA
- Research performed at Department of Orthopaedic Surgery, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
169
|
Fu R, Han F, Liu L, Yu F, Gui Z, Wang X, Li B, Fang B, Xia L. The Effects of Leptin on the Proliferation and Differentiation of Primary Chondrocytes in Vitro and Cartilage Regeneration in Vivo. ACS Biomater Sci Eng 2019; 5:1907-1919. [PMID: 33405564 DOI: 10.1021/acsbiomaterials.8b01168] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Runqing Fu
- Department of Orthodontics, Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Fengxuan Han
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lu Liu
- Department of Orthodontics, Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Fei Yu
- Department of Orthodontics, Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zhipeng Gui
- Department of Oral Surgery, Shanghai Ninth People’s Hospital Affiliated with Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xiaoting Wang
- Department of Orthodontics, Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Bin Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bing Fang
- Department of Orthodontics, Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lunguo Xia
- Department of Orthodontics, Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
170
|
Kuznetsov SA, Hailu-Lazmi A, Cherman N, de Castro LF, Robey PG, Gorodetsky R. In Vivo Formation of Stable Hyaline Cartilage by Naïve Human Bone Marrow Stromal Cells with Modified Fibrin Microbeads. Stem Cells Transl Med 2019; 8:586-592. [PMID: 30767420 PMCID: PMC6525579 DOI: 10.1002/sctm.18-0129] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 12/20/2018] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritic and other types of articular cartilage defects never heal on their own. Medicinal and surgical approaches are often ineffective, and the supply of autologous chondrocytes for tissue engineering is very limited. Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) have been suggested as an adequate cell source for cartilage reconstruction. However, the majority of studies employing BMSCs for cartilage tissue engineering have used BMSCs predifferentiated into cartilage prior to implantation. This strategy has failed to achieve formation of stable, hyaline-like cartilage, resistant to hypertrophy in vivo. We hypothesized that in vitro predifferentiation of BMSCs is not necessary when cells are combined with an adequate scaffold that supports the formation of stable cartilage in vivo. In this study, naïve (undifferentiated) human BMSCs were attached to dehydrothermally crosslinked stable fibrin microbeads (FMBs) without and with other scaffolds and implanted subcutaneously into immunocompromised mice. Optimal formation of abundant, hypertrophy-resistant, ectopic hyaline-like cartilage was achieved when BMSCs were attached to FMBs covalently coated with hyaluronic acid. The cartilage that was formed was of human origin and was stable for at least 28 weeks in vivo. Stem Cells Translational Medicine 2019;8:586-592.
Collapse
Affiliation(s)
- Sergei A Kuznetsov
- Department of Health and Human Services, Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Astar Hailu-Lazmi
- Biotechnology and Radiobiology Laboratory, Hadassah - Hebrew University Medical Center, Sharett Institute of Oncology, Jerusalem, Israel
| | - Natasha Cherman
- Department of Health and Human Services, Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Luis F de Castro
- Department of Health and Human Services, Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Pamela G Robey
- Department of Health and Human Services, Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Raphael Gorodetsky
- Biotechnology and Radiobiology Laboratory, Hadassah - Hebrew University Medical Center, Sharett Institute of Oncology, Jerusalem, Israel
| |
Collapse
|
171
|
Tangtrongsup S, Kisiday JD. Differential Effects of the Antioxidants N-Acetylcysteine and Pyrrolidine Dithiocarbamate on Mesenchymal Stem Cell Chondrogenesis. Cell Mol Bioeng 2019; 12:153-163. [PMID: 31719906 DOI: 10.1007/s12195-019-00566-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 01/03/2019] [Indexed: 01/07/2023] Open
Abstract
Introduction Mesenchymal stem cell (MSC) chondrogenesis is associated with increases in intracellular reactive oxygen species (ROS), which may result in oxidative stress that is detrimental to cartilage regeneration. This study evaluated the ability of the antioxidants N-acetylcysteine (NAC) or pyrrolidine dithiocarbamate (PDTC) to reduce intracellular ROS, and their effect on MSC chondrogenesis and maturation of cartilage-like extracellular matrix. Methods Equine bone marrow MSCs were cultured in serum-supplemented chondrogenic medium with or without NAC or PDTC. ROS was quantified in monolayer after 8 and 72 h of culture. MSCs were seeded into agarose, cultured for 15 days, and analyzed for viable cell density, glycosaminoglycan (GAG) and hydroxyproline accumulation, and collagen gene expression. PDTC cultures were evaluated for oxidative damage by protein carbonylation, and mechanical properties via compressive testing. Results NAC significantly lowered levels of ROS after 8 but not 72 h, and suppressed GAG accumulation (70%). In secondary experiments using serum-free medium, NAC significantly increased levels of ROS at 72 h, and lowered cell viability and extracellular matrix accumulation. PDTC significantly reduced levels of ROS (~ 30%) and protein carbonylation (27%), and enhanced GAG accumulation (20%). However, the compressive modulus for PDTC-treated samples was significantly lower (40%) than controls. Gene expression was largely unaffected by the antioxidants. Conclusions NAC demonstrated a limited ability to reduce intracellular ROS in chondrogenic culture, and generally suppressed accumulation of extracellular matrix. Conversely, PDTC was an effective antioxidant that enhanced GAG accumulation, although the concomitant reduction in compressive properties is a significant limitation for cartilage repair.
Collapse
Affiliation(s)
- Suwimol Tangtrongsup
- Department of Clinical Sciences, Orthopaedic Research Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 300W. Drake Road, Fort Collins, CO 80523 USA
| | - John D Kisiday
- Department of Clinical Sciences, Orthopaedic Research Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, 300W. Drake Road, Fort Collins, CO 80523 USA
| |
Collapse
|
172
|
Zimta AA, Baru O, Badea M, Buduru SD, Berindan-Neagoe I. The Role of Angiogenesis and Pro-Angiogenic Exosomes in Regenerative Dentistry. Int J Mol Sci 2019; 20:ijms20020406. [PMID: 30669338 PMCID: PMC6359271 DOI: 10.3390/ijms20020406] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/04/2019] [Accepted: 01/15/2019] [Indexed: 02/07/2023] Open
Abstract
Dental surgeries can result in traumatic wounds that provoke major discomfort and have a high risk of infection. In recent years, density research has taken a keen interest in finding answers to this problem by looking at the latest results made in regenerative medicine and adapting them to the specificities of oral tissue. One of the undertaken directions is the study of angiogenesis as an integrative part of oral tissue regeneration. The stimulation of this process is intended to enhance the local availability of stem cells, oxygen levels, nutrient supply, and evacuation of toxic waste. For a successful stimulation of local angiogenesis, two major cellular components must be considered: the stem cells and the vascular endothelial cells. The exosomes are extracellular vesicles, which mediate the communication between two cell types. In regenerative dentistry, the analysis of exosome miRNA content taps into the extended communication between these cell types with the purpose of improving the regenerative potential of oral tissue. This review analyzes the stem cells available for the dentistry, the molecular cargo of their exosomes, and the possible implications these may have for a future therapeutic induction of angiogenesis in the oral wounds.
Collapse
Affiliation(s)
- Alina-Andreea Zimta
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania.
| | - Oana Baru
- Department of Preventive Dentistry, Faculty of Dental Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400083 Cluj-Napoca, Romania.
| | - Mandra Badea
- Department of Preventive Dentistry, Faculty of Dental Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400083 Cluj-Napoca, Romania.
| | - Smaranda Dana Buduru
- Prosthetics and Dental materials, Faculty of Dental Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, 32 Clinicilor Street, 400006 Cluj-Napoca, Romania.
- Stomestet Stomatology Clinic, Calea Manastur 68A Street, 400658 Cluj-Napoca, Romania.
| | - Ioana Berindan-Neagoe
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania.
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania.
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", Republicii 34th street, 400015 Cluj-Napoca, Romania.
| |
Collapse
|
173
|
Sun H, Peng G, Ning X, Wang J, Yang H, Deng J. Emerging roles of long noncoding RNA in chondrogenesis, osteogenesis, and osteoarthritis. Am J Transl Res 2019; 11:16-30. [PMID: 30787967 PMCID: PMC6357308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/18/2018] [Indexed: 06/09/2023]
Abstract
Osteoarthritis (OA) is the most prevalent age-related debilitating joint disease, and is characterized primarily by articular cartilage degradation and subchondral bone lesions. It is also the leading cause of chronic morbidity in older populations. The etiology of OA is multifactorial, with the underlying regulatory mechanisms remaining largely unknown. Long noncoding RNA (lncRNA) is a group of noncoding RNAs defined as being >200 nucleotides in length. Increasing evidence demonstrates that many lncRNAs serve as critical regulators of chondrogenesis and bone and cartilage homeostasis, thereby influencing OA development. In this review, we highlight the current understanding concerning lncRNAs, including their physical features, biological functions, and potential roles in chondrogenesis, osteogenesis, and OA. This information may shed new light on the epigenetic regulation of cartilage and substantiate lncRNAs as novel therapeutic targets in OA.
Collapse
Affiliation(s)
- Hong Sun
- Department of Orthopaedics, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, China
| | - Guoxuan Peng
- Department of Emergence Medicine, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, China
| | - Xu Ning
- Department of Orthopaedics, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, China
| | - Jian Wang
- Department of Orthopaedics, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, China
| | - Hua Yang
- Department of Orthopaedics, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, China
| | - Jin Deng
- Department of Emergence Medicine, Affiliated Hospital of Guizhou Medical UniversityGuiyang 550004, China
| |
Collapse
|
174
|
Di Matteo B, El Araby MM, D'Angelo A, Iacono F, Nannini A, Vitale ND, Marcacci M, Respizzi S, Kon E. Adipose-Derived Stem Cell Treatments and Formulations. Clin Sports Med 2018; 38:61-78. [PMID: 30466723 DOI: 10.1016/j.csm.2018.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This article analyzes the current literature on the use of adipose-derived stem cells (ASCs) to evaluate the available evidence regarding their therapeutic potential in the treatment of cartilage pathology. Seventeen articles were included and analyzed, showing that there is overall a lack of high-quality evidence concerning the use of ASCs. Most trials are case series with short-term evaluation. The most adopted approach consists of an intra-articular injection of the stromal vascular fraction (SVF) rather than the expanded cells. Based on the available data, no specific preparation method or formulation could be considered as the preferred choice in clinical practice.
Collapse
Affiliation(s)
- Berardo Di Matteo
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, Milan 20089, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan 20089, Italy.
| | - Mohamed Marzouk El Araby
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, Milan 20089, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Alessandro D'Angelo
- Department of Orthopaedic, Traumatology and Rehabilitation, Azienda Ospedaliero Universitaria Città della Salute e della Scienza, CTO Hospital, Via Zuretti 29, Turin 10126, Italy
| | - Francesco Iacono
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, Milan 20089, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Alessandra Nannini
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, Milan 20089, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Nicolò Danilo Vitale
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, Milan 20089, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Maurilio Marcacci
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, Milan 20089, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Stefano Respizzi
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, Milan 20089, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Elizaveta Kon
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, Milan 20089, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan 20089, Italy
| |
Collapse
|
175
|
The Potential of Menstrual Blood-Derived Mesenchymal Stem Cells for Cartilage Repair and Regeneration: Novel Aspects. Stem Cells Int 2018; 2018:5748126. [PMID: 30627174 PMCID: PMC6304826 DOI: 10.1155/2018/5748126] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/21/2018] [Indexed: 12/16/2022] Open
Abstract
Menstrual blood is a unique body fluid that contains mesenchymal stem cells (MSCs). These cells have attracted a great deal of attention due to their exceptional advantages including easy access and frequently accessible sample source and no need for complex ethical and surgical interventions, as compared to other tissues. Menstrual blood-derived MSCs possess all the major stem cell properties and even have a greater proliferation and differentiation potential as compared to bone marrow-derived MSCs, making them a perspective tool in a further clinical practice. Although the potential of menstrual blood stem cells to differentiate into a large variety of tissue cells has been studied in many studies, their chondrogenic properties have not been extensively explored and investigated. Articular cartilage is susceptible to traumas and degenerative diseases, such as osteoarthritis, and has poor self-regeneration capacity and therefore requires more effective therapeutic technique. MSCs seem promising candidates for cartilage regeneration; however, no clinically effective stem cell-based repair method has yet emerged. This chapter focuses on studies in the field of menstrual blood-derived MSCs and their chondrogenic differentiation potential and suitability for application in cartilage regeneration. Although a very limited number of studies have been made in this field thus far, these cells might emerge as an efficient and easily accessible source of multipotent cells for cartilage engineering and cell-based chondroprotective therapy.
Collapse
|
176
|
Irawan V, Sung TC, Higuchi A, Ikoma T. Collagen Scaffolds in Cartilage Tissue Engineering and Relevant Approaches for Future Development. Tissue Eng Regen Med 2018; 15:673-697. [PMID: 30603588 PMCID: PMC6250655 DOI: 10.1007/s13770-018-0135-9] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/30/2018] [Accepted: 06/15/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cartilage tissue engineering (CTE) aims to obtain a structure mimicking native cartilage tissue through the combination of relevant cells, three-dimensional scaffolds, and extraneous signals. Implantation of 'matured' constructs is thus expected to provide solution for treating large injury of articular cartilage. Type I collagen is widely used as scaffolds for CTE products undergoing clinical trial, owing to its ubiquitous biocompatibility and vast clinical approval. However, the long-term performance of pure type I collagen scaffolds would suffer from its limited chondrogenic capacity and inferior mechanical properties. This paper aims to provide insights necessary for advancing type I collagen scaffolds in the CTE applications. METHODS Initially, the interactions of type I/II collagen with CTE-relevant cells [i.e., articular chondrocytes (ACs) and mesenchymal stem cells (MSCs)] are discussed. Next, the physical features and chemical composition of the scaffolds crucial to support chondrogenic activities of AC and MSC are highlighted. Attempts to optimize the collagen scaffolds by blending with natural/synthetic polymers are described. Hybrid strategy in which collagen and structural polymers are combined in non-blending manner is detailed. RESULTS Type I collagen is sufficient to support cellular activities of ACs and MSCs; however it shows limited chondrogenic performance than type II collagen. Nonetheless, type I collagen is the clinically feasible option since type II collagen shows arthritogenic potency. Physical features of scaffolds such as internal structure, pore size, stiffness, etc. are shown to be crucial in influencing the differentiation fate and secreting extracellular matrixes from ACs and MSCs. Collagen can be blended with native or synthetic polymer to improve the mechanical and bioactivities of final composites. However, the versatility of blending strategy is limited due to denaturation of type I collagen at harsh processing condition. Hybrid strategy is successful in maximizing bioactivity of collagen scaffolds and mechanical robustness of structural polymer. CONCLUSION Considering the previous improvements of physical and compositional properties of collagen scaffolds and recent manufacturing developments of structural polymer, it is concluded that hybrid strategy is a promising approach to advance further collagen-based scaffolds in CTE.
Collapse
Affiliation(s)
- Vincent Irawan
- Department of Materials Science and Engineering, Tokyo Institute of Technology, 2 Chome-12-1, Meguro-ku, Tokyo, 152-8550 Japan
| | - Tzu-Cheng Sung
- Department of Chemical and Materials Engineering, National Central University, No. 300 Jung Da Rd., Chung-Li, Taoyuan, 320 Taiwan
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, No. 300 Jung Da Rd., Chung-Li, Taoyuan, 320 Taiwan
| | - Toshiyuki Ikoma
- Department of Materials Science and Engineering, Tokyo Institute of Technology, 2 Chome-12-1, Meguro-ku, Tokyo, 152-8550 Japan
| |
Collapse
|
177
|
Deng Y, Lei G, Lin Z, Yang Y, Lin H, Tuan RS. Engineering hyaline cartilage from mesenchymal stem cells with low hypertrophy potential via modulation of culture conditions and Wnt/β-catenin pathway. Biomaterials 2018; 192:569-578. [PMID: 30544046 DOI: 10.1016/j.biomaterials.2018.11.036] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 11/24/2018] [Accepted: 11/30/2018] [Indexed: 12/27/2022]
Abstract
Mesenchymal stem cells (MSCs) represent a promising cell source to regenerate articular cartilage, but current chondroinduction protocols, commonly using transforming growth factor-β (TGFβ), lead to concomitant chondrocytic hypertrophy with ossification risk. Here, we showed that a 14-day culture of MSC-laden hyaluronic acid hydrogel in the presence of TGFβ, followed by 7 days culture in TGFβ-free medium, with the supplement of Wnt/β-catenin inhibitor XAV939 from day 10-21, resulted in significantly reduced hypertrophy phenotype. The stability of the hyaline phenotype of the MSC-derived cartilage, generated with a standard protocol (Control) or the optimized (Optimized) method developed in this study, was further examined through intramuscular implantation in nude mice. After 4 weeks, constructs from the Control group showed obvious mineralization; in contrast, the Optimized group displayed no signs of mineralization, and maintained cartilaginous histology. Further analysis showed that TGFβ treatment time affected p38 expression, while exposure to XAV939 significantly inhibited P-Smad 1/5 level, which together resulted in decreased level of Runx2. These findings suggest a novel treatment regimen to generate hyaline cartilage from human MSCs-loaded scaffolds, which have a minimal risk of eliciting endochondral ossification.
Collapse
Affiliation(s)
- Yuhao Deng
- Department of Orthopaedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Xiangya Third Hospital, Central South University, Changsha, Hunan, China
| | - Guanghua Lei
- Department of Orthopaedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zixuan Lin
- Department of Orthopaedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Yuanheng Yang
- Department of Orthopaedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Xiangya Third Hospital, Central South University, Changsha, Hunan, China
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, PA, 15261, USA; The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
178
|
Glucose metabolism induced by Bmp signaling is essential for murine skeletal development. Nat Commun 2018; 9:4831. [PMID: 30446646 PMCID: PMC6240091 DOI: 10.1038/s41467-018-07316-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/24/2018] [Indexed: 02/08/2023] Open
Abstract
Much of the mammalian skeleton originates from a cartilage template eventually replaced by bone via endochondral ossification. Despite much knowledge about growth factors and nuclear proteins in skeletal development, little is understood about the role of metabolic regulation. Here we report that genetic deletion of the glucose transporter Glut1 (Slc2a1), either before or after the onset of chondrogenesis in the limb, severely impairs chondrocyte proliferation and hypertrophy, resulting in dramatic shortening of the limbs. The cartilage defects are reminiscent to those caused by deficiency in Bmp signaling. Importantly, deletion of Bmpr1a in chondrocytes markedly reduces Glut1 levels in vivo, whereas recombinant BMP2 increases Glut1 mRNA and protein levels, boosting glucose metabolism in primary chondrocytes. Biochemical studies identify a Bmp-mTORC1-Hif1a signaling cascade resulting in upregulation of Glut1 in chondrocytes. The results therefore uncover a hitherto unknown connection between Bmp signaling and glucose metabolism in the regulation of cartilage development. It is unclear how metabolic regulation affects development of the skeleton. Here, the authors show that deletion of the glucose transporter Glut1 (Slc2a1) both prior to and following chondrogenesis in the mouse limb impairs chondrocyte proliferation and shortening of the limbs, modulated by BMP signaling.
Collapse
|
179
|
Hyaluronan microenvironment enhances cartilage regeneration of human adipose-derived stem cells in a chondral defect model. Int J Biol Macromol 2018; 119:726-740. [DOI: 10.1016/j.ijbiomac.2018.07.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 06/28/2018] [Accepted: 07/11/2018] [Indexed: 12/22/2022]
|
180
|
Laowanitwattana T, Aungsuchawan S, Narakornsak S, Markmee R, Tancharoen W, Keawdee J, Boonma N, Tasuya W, Peerapapong L, Pangjaidee N, Pothacharoen P. Osteoblastic differentiation potential of human amniotic fluid-derived mesenchymal stem cells in different culture conditions. Acta Histochem 2018; 120:701-712. [PMID: 30078494 DOI: 10.1016/j.acthis.2018.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 02/07/2023]
Abstract
Osteoporosis is a bone degenerative disease characterized by a decrease in bone strength and an alteration in the osseous micro-architecture causing an increase in the risk of fractures. These diseases usually happen in post-menopausal women and elderly men. The most common treatment involves anti-resorptive agent drugs. However, the inhibition of bone resorption alone is not adequate for recovery in patients at the severe stage of osteoporosis who already have a fracture. Therefore, the combination of utilizing osteoblast micro mimetic scaffold in cultivation with the stimulation of osteoblastic differentiations to regain bone formation is a treatment strategy of considerable interest. The aims of this current study are to investigate the osteoblastic differentiation potential of mesenchymal stem cells derived from human amniotic fluid and to compare the monolayer culture and scaffold culture conditions. The results showed the morphology of cells in human amniotic fluid as f-type, which is a typical cell shape of mesenchymal stem cells. In addition, the proliferation rate of cells in human amniotic fluid reached the highest peak after 14 days of culturing. After which time, the growth rate slowly decreased. Moreover, the positive expression of specific mesenchymal cell surface markers including CD44, CD73, CD90, and also HLA-ABC (MHC class I) were recorded. On the other hand, the negative expressions of the endothelial stem cells markers (CD31), the hematopoietic stem cells markers (CD34, 45), the amniotic stem cells markers (CD117), and also the HLA-DR (MHC class II) were also recorded. The expressions of osteoblastogenic related genes including OCN, COL1A1, and ALP were higher in the osteogenic-induced group when compared to the control group. Interestingly, the osteoblastogenic related gene expressions that occurred under scaffold culture conditions were superior to the monolayer culture conditions. Additionally, higher ALP activity and greater calcium deposition were recorded in the extracellular matrix in the osteogenic-induced group than in the culture in the scaffold group. In summary, the mesenchymal stem cells derived from human amniotic fluid can be induced to be differentiated into osteoblastic-like cells and can promote osteoblastic differentiation using the applied scaffold.
Collapse
|
181
|
Lu J, Shen X, Sun X, Yin H, Yang S, Lu C, Wang Y, Liu Y, Huang Y, Yang Z, Dong X, Wang C, Guo Q, Zhao L, Sun X, Lu S, Mikos AG, Peng J, Wang X. Increased recruitment of endogenous stem cells and chondrogenic differentiation by a composite scaffold containing bone marrow homing peptide for cartilage regeneration. Theranostics 2018; 8:5039-5058. [PMID: 30429885 PMCID: PMC6217070 DOI: 10.7150/thno.26981] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/27/2018] [Indexed: 01/17/2023] Open
Abstract
Even small cartilage defects could finally degenerate to osteoarthritis if left untreated, owing to the poor self-healing ability of articular cartilage. Stem cell transplantation has been well implemented as a common approach in cartilage tissue engineering but has technical complexity and safety concerns. The stem cell homing-based technique emerged as an alternative promising therapy for cartilage repair to overcome traditional limitations. In this study, we constructed a composite hydrogel scaffold by combining an oriented acellular cartilage matrix (ACM) with a bone marrow homing peptide (BMHP)-functionalized self-assembling peptide (SAP). We hypothesized that increased recruitment of endogenous stem cells by the composite scaffold could enhance cartilage regeneration. Methods: To test our hypothesis, in vitro proliferation, attachment and chondrogenic differentiation of rabbit mesenchymal stem cells (MSCs) were tested to confirm the bioactivities of the functionalized peptide hydrogel. The composite scaffold was then implanted into full-thickness cartilage defects on rabbit knee joints for cartilage repair, in comparison with microfracture or other sample groups. Stem cell recruitment was monitored by dual labeling with CD29 and CD90 under confocal microcopy at 1 week after implantation, followed by chondrogenic differentiation examined by qRT-PCR. Repaired tissue of the cartilage defects was evaluated by histological and immunohistochemistry staining, microcomputed tomography (micro-CT) and magnetic resonance imaging (MRI) at 3 and 6 months post-surgery. Macroscopic and histological scoring was done to evaluate the optimal in vivo repair outcomes of this composite scaffold. Results: The functionalized SAP hydrogels could stimulate rabbit MSC proliferation, attachment and chondrogenic differentiation during in vitro culture. At 7 days after implantation, increased recruitment of MSCs based on CD29+ /CD90+ double-positive cells was found in vivo in the composite hydrogel scaffold, as well as upregulation of cartilage-associated genes (aggrecan, Sox9 and type II collagen). After 3 and 6 months post-surgery, the articular cartilage defect in the composite scaffold-treated group was fully covered with cartilage-like tissue with a smooth surface, which was similar to the surrounding native cartilage, according to the results of histological and immunohistochemistry staining, micro-CT and MRI analysis. Macroscopic and histological scoring confirmed that the quality of cartilage repair was significantly improved with implantation of the composite scaffold at each timepoint, in comparison with microfracture or other sample groups. Conclusion: Our findings demonstrated that the composite scaffold could enhance endogenous stem cell homing and chondrogenic differentiation and significantly improve the therapeutic outcome of chondral defects. The present study provides a promising approach for in vivo cartilage repair without cell transplantation. Optimization of this strategy may offer great potential and benefits for clinical application in the future.
Collapse
Affiliation(s)
- Jiaju Lu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xuezhen Shen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Xun Sun
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Heyong Yin
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, Munich 80336, Germany
| | - Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Changfeng Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Yifan Liu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Yingqi Huang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Zijin Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xianqi Dong
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Chenhao Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Quanyi Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Shibi Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Antonios G. Mikos
- Department of Bioengineering, Bioscience Research Collaborative, Rice University, Texas 77030, USA
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
182
|
Bae HC, Park HJ, Wang SY, Yang HR, Lee MC, Han HS. Hypoxic condition enhances chondrogenesis in synovium-derived mesenchymal stem cells. Biomater Res 2018; 22:28. [PMID: 30275971 PMCID: PMC6158840 DOI: 10.1186/s40824-018-0134-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/29/2018] [Indexed: 12/21/2022] Open
Abstract
Background The chondrogenic differentiation of mesenchymal stem cells (MSCs) is regulated by many factors, including oxygen tensions, growth factors, and cytokines. Evidences have suggested that low oxygen tension seems to be an important regulatory factor in the proliferation and chondrogenic differentiation in various MSCs. Recent studies report that synovium-derived mesenchymal stem cells (SDSCs) are a potential source of stem cells for the repair of articular cartilage defects. But, the effect of low oxygen tension on the proliferation and chondrogenic differentiation in SDSCs has not characterized. In this study, we investigated the effects of hypoxia on proliferation and chondrogenesis in SDSCs. Method SDSCs were isolated from patients with osteoarthritis at total knee replacement. To determine the effect of oxygen tension on proliferation and colony-forming characteristics of SDSCs, A colony-forming unit (CFU) assay and cell counting-based proliferation assay were performed under normoxic (21% oxygen) or hypoxic (5% oxygen). For in vitro chondrogenic differentiation, SDSCs were concentrated to form pellets and subjected to conditions appropriate for chondrogenic differentiation under normoxia and hypoxia, followed by the analysis for the expression of genes and proteins of chondrogenesis. qRT-PCR, histological assay, and glycosoaminoglycan assays were determined to assess chondrogenesis. Results Low oxygen condition significantly increased proliferation and colony-forming characteristics of SDSCs compared to that of SDSCs under normoxic culture. Similar pellet size and weight were found for chondrogensis period under hypoxia and normoxia condition. The mRNA expression of types II collagen, aggrecan, and the transcription factor SOX9 was increased under hypoxia condition. Histological sections stained with Safranin-O demonstrated that hypoxic conditions had increased proteoglycan synthesis. Immunohistochemistry for types II collagen demonstrated that hypoxic culture of SDSCs increased type II collagen expression. In addition, GAG deposition was significantly higher in hypoxia compared with normoxia at 21 days of differentiation. Conclusion These findings show that hypoxia condition has an important role in regulating the synthesis ECM matrix by SDSCs as they undergo chondrogenesis. This has important implications for cartilage tissue engineering applications of SDSCs.
Collapse
Affiliation(s)
- Hyun Cheol Bae
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| | - Hee Jung Park
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| | - Sun Young Wang
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| | - Ha Ru Yang
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| | - Myung Chul Lee
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| | - Hyuk-Soo Han
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744 Republic of Korea
| |
Collapse
|
183
|
Shin YS, Yoon JR, Kim HS, Lee SH. Intra-Articular Injection of Bone Marrow-Derived Mesenchymal Stem Cells Leading to Better Clinical Outcomes without Difference in MRI Outcomes from Baseline in Patients with Knee Osteoarthritis. Knee Surg Relat Res 2018; 30:206-214. [PMID: 29983008 PMCID: PMC6122947 DOI: 10.5792/ksrr.17.201] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/06/2018] [Accepted: 04/24/2018] [Indexed: 12/21/2022] Open
Abstract
Purpose Bone marrow (BM) is frequently used as a source of mesenchymal stem cells (MSCs) because they have a high potential for differentiation. However, it is unclear whether BM-derived MSCs lead to better clinical and magnetic resonance imaging (MRI) outcomes postoperatively. Materials and Methods This meta-analysis compared the clinical and MRI outcomes in patients with knee osteoarthritis (OA) treated with BM-derived MSCs. Eight studies comparing the clinical and MRI outcomes assessed with various measurement tools in patients with knee OA treated with BM-derived MSCs were included. Results The range of motion (95% confidence interval [CI], −13.05 to 4.24; p=0.32) and MRI outcomes (95% CI, −0.16 to 1.40; p=0.12) did not differ significantly between the baseline and final follow-up. In contrast, pain (95% CI, 0.89 to 1.87; p<0.001) and functional outcomes (95% CI, 0.70 to 2.07; p<0.001) were significantly improved at the final follow-up when compared to the baseline. Conclusions This meta-analysis found no significant difference in the tested range of motion and MRI outcomes between the baseline and the final follow-up in patients treated with BM-derived MSCs, whereas significant functional improvement and pain relief were noted when compared with the baseline. Thus, BM-derived MSCs appear to be a viable alternative for patients with knee OA, although long-term and high-quality randomized controlled trials are needed to confirm the clinical benefits.
Collapse
Affiliation(s)
- Young-Soo Shin
- Department of Orthopedic Surgery, Veterans Health Service Medical Center, Seoul, Korea
| | - Jung-Ro Yoon
- Department of Orthopedic Surgery, Veterans Health Service Medical Center, Seoul, Korea
| | - Hee-Sun Kim
- Chonbuk Research Institute of Nursing Science, College of Nursing, Chonbuk National University, Jeonju, Korea
| | - Seon-Heui Lee
- Department of Nursing Science, College of Nursing, Gachon University, Incheon, Korea
| |
Collapse
|
184
|
Huri PY, Hamsici S, Ergene E, Huri G, Doral MN. Infrapatellar Fat Pad-Derived Stem Cell-Based Regenerative Strategies in Orthopedic Surgery. Knee Surg Relat Res 2018; 30:179-186. [PMID: 29554720 PMCID: PMC6122943 DOI: 10.5792/ksrr.17.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 12/22/2017] [Accepted: 01/05/2018] [Indexed: 01/10/2023] Open
Abstract
Infrapatellar fat pad is a densely vascularized and innervated extrasynovial tissue that fills the anterior knee compartment. It plays a role in knee biomechanics as well as constitutes a source of stem cells for regeneration after knee injury. Infrapatellar fat pad-derived stem cells (IPFP-ASCs) possess enhanced and age-independent differentiation capacity as compared to other stem cells, which makes them a very promising candidate in stem cell-based regenerative therapy. The aims of this review are to outline the latest advances and potential trends in using IPFP-ASCs and to emphasize the advantages over other sources of stem cells for applications in orthopedic surgery.
Collapse
Affiliation(s)
- Pinar Yilgor Huri
- Department of Biomedical Engineering, Ankara University Faculty of Engineering, Ankara, Turkey
| | - Seren Hamsici
- Department of Biomedical Engineering, Ankara University Faculty of Engineering, Ankara, Turkey
| | - Emre Ergene
- Department of Biomedical Engineering, Ankara University Faculty of Engineering, Ankara, Turkey.,Ankara University Biotechnology Institute, Ankara, Turkey
| | - Gazi Huri
- Department of Orthopedics and Traumatology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Mahmut Nedim Doral
- Department of Orthopedics and Traumatology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
185
|
Aisenbrey EA, Bryant SJ. A MMP7-sensitive photoclickable biomimetic hydrogel for MSC encapsulation towards engineering human cartilage. J Biomed Mater Res A 2018; 106:2344-2355. [PMID: 29577606 PMCID: PMC6030485 DOI: 10.1002/jbm.a.36412] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/30/2018] [Accepted: 03/15/2018] [Indexed: 12/28/2022]
Abstract
Cartilage tissue engineering strategies that use in situ forming degradable hydrogels for mesenchymal stem cell (MSC) delivery are promising for treating chondral defects. Hydrogels that recapitulate aspects of the native tissue have the potential to encourage chondrogenesis, permit cellular mediated degradation, and facilitate tissue growth. This study investigated photoclickable poly(ethylene glycol) hydrogels, which were tailored to mimic the cartilage microenvironment by incorporating extracellular matrix analogs, chondroitin sulfate and RGD, and crosslinks sensitive to matrix metalloproteinase 7 (MMP7). Human MSCs were encapsulated in the hydrogel, cultured up to nine weeks, and assessed by mRNA expression, protein production and biochemical analysis. Chondrogenic genes, SOX9, ACAN, and COL2A1, significantly increased with culture time, and the ratios of COL2A1:COL10A1 and SOX9:RUNX2 reached values of ∼20-100 by week 6. The encapsulated MSCs degraded the hydrogel, which was nearly undetectable by week 9. There was substantial deposition of aggrecan and collagen II, which correlated with degradation of the hydrogel. Minimal collagen X was detectable, but collagen I was prevalent. After week 1, extracellular matrix elaboration was accompanied by a ∼twofold increase in compressive modulus with culture time. The MMP7-sensitive cartilage mimetic hydrogel supported MSC chondrogenesis and promoted macroscopic neocartilaginous matrix elaboration representative of fibrocartilage. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2344-2355, 2018.
Collapse
Affiliation(s)
- Elizabeth A Aisenbrey
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309
| | - Stephanie J. Bryant
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309
- Material Science and Engineering Program, University of Colorado, Boulder, CO 80309
| |
Collapse
|
186
|
Lee JY, Matthias N, Pothiawala A, Ang BK, Lee M, Li J, Sun D, Pigeot S, Martin I, Huard J, Huang Y, Nakayama N. Pre-transplantational Control of the Post-transplantational Fate of Human Pluripotent Stem Cell-Derived Cartilage. Stem Cell Reports 2018; 11:440-453. [PMID: 30057264 PMCID: PMC6092881 DOI: 10.1016/j.stemcr.2018.06.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 01/24/2023] Open
Abstract
Cartilage pellets generated from ectomesenchymal progeny of human pluripotent stem cells (hPSCs) in vitro eventually show signs of commitment of chondrocytes to hypertrophic differentiation. When transplanted subcutaneously, most of the surviving pellets were fully mineralized by 8 weeks. In contrast, treatment with the adenylyl cyclase activator, forskolin, in vitro resulted in slightly enlarged cartilage pellets containing an increased proportion of proliferating immature chondrocytes that expressed very low levels of hypertrophic/terminally matured chondrocyte-specific genes. Forskolin treatment also enhanced hyaline cartilage formation by reducing type I collagen gene expression and increasing sulfated glycosaminoglycan accumulation in the developed cartilage. Chondrogenic mesoderm from hPSCs and dedifferentiated nasal chondrocytes responded similarly to forskolin. Furthermore, forskolin treatment in vitro increased the frequency at which the cartilage pellets maintained unmineralized chondrocytes after subcutaneous transplantation. Thus, the post-transplantational fate of chondrocytes originating from hPSC-derived chondroprogenitors can be controlled during their genesis in vitro. Forskolin/cAMP suppresses/delays BMP-induced chondrocyte maturation in vitro Forskolin supports chondrocyte proliferation and hyaline chondrogenesis in vitro Forskolin suppresses osteogenesis and BMP signaling gene expression in cartilage In vitro forskolin treatment improves in vivo maintenance of uncalcified cartilage
Collapse
Affiliation(s)
- John Y Lee
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston (UTHealth) Medical School, 1825 Pressler St., Houston, TX 77030, USA
| | - Nadine Matthias
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston (UTHealth) Medical School, 1825 Pressler St., Houston, TX 77030, USA
| | - Azim Pothiawala
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston (UTHealth) Medical School, 1825 Pressler St., Houston, TX 77030, USA
| | - Bryan K Ang
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston (UTHealth) Medical School, 1825 Pressler St., Houston, TX 77030, USA
| | - Minjung Lee
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Jia Li
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Deqiang Sun
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Sebastien Pigeot
- Department of Biomedicine, University Hospital Basel, Basel CH-4031, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, Basel CH-4031, Switzerland
| | - Johnny Huard
- Department of Orthopaedic Surgery, UTHealth Medical School, Houston, TX 77030, USA
| | - Yun Huang
- Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Naoki Nakayama
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston (UTHealth) Medical School, 1825 Pressler St., Houston, TX 77030, USA; Department of Orthopaedic Surgery, UTHealth Medical School, Houston, TX 77030, USA.
| |
Collapse
|
187
|
Huynh NPT, Brunger JM, Gloss CC, Moutos FT, Gersbach CA, Guilak F. Genetic Engineering of Mesenchymal Stem Cells for Differential Matrix Deposition on 3D Woven Scaffolds. Tissue Eng Part A 2018; 24:1531-1544. [PMID: 29756533 DOI: 10.1089/ten.tea.2017.0510] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Tissue engineering approaches for the repair of osteochondral defects using biomaterial scaffolds and stem cells have remained challenging due to the inherent complexities of inducing cartilage-like matrix and bone-like matrix within the same local environment. Members of the transforming growth factor β (TGFβ) family have been extensively utilized in the engineering of skeletal tissues, but have distinct effects on chondrogenic and osteogenic differentiation of progenitor cells. The goal of this study was to develop a method to direct human bone marrow-derived mesenchymal stem cells (MSCs) to deposit either mineralized matrix or a cartilaginous matrix rich in glycosaminoglycan and type II collagen within the same biochemical environment. This differential induction was performed by culturing cells on engineered three-dimensionally woven poly(ɛ-caprolactone) (PCL) scaffolds in a chondrogenic environment for cartilage-like matrix production while inhibiting TGFβ3 signaling through Mothers against DPP homolog 3 (SMAD3) knockdown, in combination with overexpressing RUNX2, to achieve mineralization. The highest levels of mineral deposition and alkaline phosphatase activity were observed on scaffolds with genetically engineered MSCs and exhibited a synergistic effect in response to SMAD3 knockdown and RUNX2 expression. Meanwhile, unmodified MSCs on PCL scaffolds exhibited accumulation of an extracellular matrix rich in glycosaminoglycan and type II collagen in the same biochemical environment. This ability to derive differential matrix deposition in a single culture condition opens new avenues for developing complex tissue replacements for chondral or osteochondral defects.
Collapse
Affiliation(s)
- Nguyen P T Huynh
- 1 Department of Orthopaedic Surgery, Washington University in Saint Louis , Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis , St. Louis, Missouri.,3 Department of Cell Biology, Duke University , Durham, North Carolina
| | | | - Catherine C Gloss
- 1 Department of Orthopaedic Surgery, Washington University in Saint Louis , Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis , St. Louis, Missouri
| | | | - Charles A Gersbach
- 6 Department of Biomedical Engineering, Duke University , Durham, North Carolina
| | - Farshid Guilak
- 1 Department of Orthopaedic Surgery, Washington University in Saint Louis , Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis , St. Louis, Missouri.,5 Cytex Therapeutics, Inc. , Durham, North Carolina
| |
Collapse
|
188
|
Andia I, Maffulli N. How far have biological therapies come in regenerative sports medicine? Expert Opin Biol Ther 2018; 18:785-793. [PMID: 29939773 DOI: 10.1080/14712598.2018.1492541] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Regular engagement in sports produces many health benefits, but also exposes to increased injury risk. The quality of medical care available is crucial not only for sports trauma but also to avoid overuse syndromes and post-traumatic degenerative conditions. AREAS COVERED We provide background information on some clinical needs in sport injuries and describe the main families of biological products used in clinical practice. We also discuss limitations of the current clinical experience. EXPERT OPINION Sport and exercise impairment affects different segments of the population with different needs. The exceptional demands of elite athletes and subsequent media coverage have created hype around regenerative therapies. Statistical evidence, whether weak (cell products) or moderate (PRPs), is not enough to drive medical decisions because of the heterogeneity of the biological products available and their application procedures. Moreover, the specific needs of the different segments of the population along with the available clinical evidence for each musculoskeletal condition should be considered in the decision-making process. There is urgent need to develop regenerative protocols combined with post-intervention rehabilitation, and gather meaningful clinical data on the safety and efficacy of these interventions in the different populations segments.
Collapse
Affiliation(s)
- Isabel Andia
- a Regenerative Medicine Laboratory, BioCruces Health Research Institute, Cruces University Hospital , Barakaldo , Spain
| | - Nicola Maffulli
- b Department of Musculoskeletal Disorders , University of Salerno School of Medicine and Dentristry , Salerno , Italy.,c Centre for Sport and Exercise Medicine , Queen Mary University of London, Barts and the London School of Medicine and Dentistry , London , England
| |
Collapse
|
189
|
El Qashty RMN, Mohamed NN, Radwan LRS, Ibrahim FMM. Effect of bone marrow mesenchymal stem cells on healing of temporomandibular joints in rats with induced rheumatoid arthritis. Eur J Oral Sci 2018; 126:272-281. [PMID: 29952027 DOI: 10.1111/eos.12533] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2018] [Indexed: 12/15/2022]
Abstract
The healing capacity of bone marrow mesenchymal stem cells (BMMSCs) has been evaluated in various studies. This study aimed to evaluate the effect of BMMSCs on the healing of temporomandibular joints (TMJs) with induced rheumatoid arthritis. Fifty healthy male Sprague Dawley rats were divided into three groups: group I (n = 10), negative control; group II (n = 20), positive control (induction of arthritis by adjuvant followed by intravenous injection of 0.1 ml of PBS); and group III (n = 20), intervention (as for group II but injected intravenously with 1 × 106 cells ml-1 of BMMSCs suspended in PBS). Half of the rats in each group were euthanized 3 wk after the start of the experiment and the other half was euthanized after 5 wk. Group I revealed normal TMJ features. Group II showed thickening of disc, thinning of cartilage, disordered bone trabeculae, and decreased in mean % area staining positive of collagen fibers at 3 wk, while at 5 wk these effects were more aggravated. Group III showed nearly normal thickness of disc and condylar cartilage, nearly normal arrangement of bone trabeculae and regenerated collagen fibers at 3 wk, while after 5 wk the TMJ features were almost normal. Two-way anova revealed statistically significant differences between groups. Thus, treatment of induced rheumatoid arthritis with BMMSCs shows promising results that need to be further investigated in humans.
Collapse
Affiliation(s)
- Rana M N El Qashty
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| | - Nesreen N Mohamed
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| | - Lobna R S Radwan
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt.,Department of Oral Biology, Faculty of Oral and Dental Medicine, Delta University for Science and Technology, Gamasa, Egypt
| | - Fatma M M Ibrahim
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| |
Collapse
|
190
|
Bousnaki M, Bakopoulou A, Papadogianni D, Barkoula NM, Alpantaki K, Kritis A, Chatzinikolaidou M, Koidis P. Fibro/chondrogenic differentiation of dental stem cells into chitosan/alginate scaffolds towards temporomandibular joint disc regeneration. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2018; 29:97. [PMID: 29946796 DOI: 10.1007/s10856-018-6109-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 06/15/2018] [Indexed: 06/08/2023]
Abstract
Tissue engineering (TE) may provide effective alternative treatment for challenging temporomandibular joint (TMJ) pathologies associated with disc malpositioning or degeneration and leading to severe masticatory dysfunction. Aim of this study was to evaluate the potential of chitosan/alginate (Ch/Alg) scaffolds to promote fibro/chondrogenic differentiation of dental pulp stem cells (DPSCs) and production of fibrocartilage tissue, serving as a replacement of the natural TMJ disc. Ch/Alg scaffolds were fabricated by crosslinking with CaCl2 combined or not with glutaraldehyde, resulting in two scaffold types that were physicochemically characterized, seeded with DPSCs or human nucleus pulposus cells (hNPCs) used as control and evaluated for cell attachment, viability, and proliferation. The DPSCs/scaffold constructs were incubated for up to 8 weeks and assessed for extracellular matrix production by means of histology, immunofluorescence, and thermomechanical analysis. Both Ch/Alg scaffold types with a mass ratio of 1:1 presented a gel-like structure with interconnected pores. Scaffolds supported cell adhesion and long-term viability/proliferation of DPSCs and hNPCs. DPSCs cultured into Ch/Alg scaffolds demonstrated a significant increase of gene expression of fibrocartilaginous markers (COLI, COL X, SOX9, COM, ACAN) after up to 3 weeks in culture. Dynamic thermomechanical analysis revealed that scaffolds loaded with DPSCs significantly increased storage modulus and elastic response compared to cell-free scaffolds, obtaining values similar to those of native TMJ disc. Histological data and immunochemical staining for aggrecan after 4 to 8 weeks indicated that the scaffolds support abundant fibrocartilaginous tissue formation, thus providing a promising strategy for TMJ disc TE-based replacement.
Collapse
Affiliation(s)
- Maria Bousnaki
- Department of Prosthodontics, Faculty of Dentistry, Aristotle University of Thessaloniki, University Campus, Dentistry Building, 54124, Thessaloniki, Greece
| | - Athina Bakopoulou
- Department of Prosthodontics, Faculty of Dentistry, Aristotle University of Thessaloniki, University Campus, Dentistry Building, 54124, Thessaloniki, Greece
| | - Danai Papadogianni
- Department of Materials Science and Technology, University of Crete, Voutes Campus, Heraklion, 71003, Crete, Greece
| | - Nektaria-Marianthi Barkoula
- Department of Materials Science and Engineering, University of Ioannina, University Campus, 45500, Ioannina, Greece
| | - Kalliopi Alpantaki
- Department of Materials Science and Technology, University of Crete, Voutes Campus, Heraklion, 71003, Crete, Greece
| | - Aristidis Kritis
- Department of Physiology and Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, University Campus, 54006, Thessaloniki, Greece
- cGMP Regenerative Medicine facility, Department of Physiology and Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 54006, Greece
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, Voutes Campus, Heraklion, 71003, Crete, Greece
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, Nikolaou Plastira 100, Vassilika Vouton, Heraklion, 70013, Crete, Greece
| | - Petros Koidis
- Department of Prosthodontics, Faculty of Dentistry, Aristotle University of Thessaloniki, University Campus, Dentistry Building, 54124, Thessaloniki, Greece.
| |
Collapse
|
191
|
Abstract
Regenerative medicine seeks to harness the potential of cell biology for tissue replacement therapies, which will restore lost tissue functionality. Controlling and enhancing tissue healing is not just a matter of cells, but also of molecules and mechanical forces. We first describe the main biological technologies to boost musculoskeletal healing, including bone marrow and subcutaneous fat-derived regenerative products, as well as platelet-rich plasma and conditioned media. We provide some information describing possible mechanisms of action. We performed a literature search up to January 2016 searching for clinical outcomes following the use of cell therapies for sports conditions, tendons, and joints. The safety and efficacy of cell therapies for tendon conditions was examined in nine studies involving undifferentiated and differentiated (skin fibroblasts, tenocytes) cells. A total of 54 studies investigated the effects of mesenchymal stem-cell (MSC) products for joint conditions including anterior cruciate ligament, meniscus, and chondral lesions as well as osteoarthritis. In 22 studies, cellular products were injected intra-articularly, whereas in 32 studies MSC products were implanted during surgical/arthroscopic procedures. The heterogeneity of clinical conditions, cellular products, and approaches for delivery/implantation make comparability difficult. MSC products appear safe in the short- and mid-term, but studies with a long follow-up are scarce. Although the current number of randomized clinical studies is low, stem-cell products may have therapeutic potential. However, these regenerative technologies still need to be optimized.
Collapse
Affiliation(s)
- Isabel Andia
- Regenerative Medicine Laboratory, BioCruces Health Research Institute, Cruces University Hospital, Pza Cruces 12, 48903, Barakaldo, Spain.
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, University of Salerno School of Medicine and Dentistry, Salerno, Italy.,Queen Mary University of London, Barts and the London School of Medicine and Dentistry Centre for Sports and Exercise Medicine, Mile End Hospital, 275 Bancroft Road, London, E1 4DG, England
| |
Collapse
|
192
|
Making Them Commit: Strategies to Influence Phenotypic Differentiation in Mesenchymal Stem Cells. Sports Med Arthrosc Rev 2018; 26:64-69. [DOI: 10.1097/jsa.0000000000000187] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
193
|
Music E, Futrega K, Doran MR. Sheep as a model for evaluating mesenchymal stem/stromal cell (MSC)-based chondral defect repair. Osteoarthritis Cartilage 2018; 26:730-740. [PMID: 29580978 DOI: 10.1016/j.joca.2018.03.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/05/2018] [Accepted: 03/19/2018] [Indexed: 02/02/2023]
Abstract
Osteoarthritis results from the degradation of articular cartilage and is one of the leading global causes of pain and immobility. Cartilage has a limited capacity for self-repair. While repair can be enhanced through surgical intervention, current methods often generate inferior fibrocartilage and repair is transient. The development of tissue engineering strategies to improve repair outcomes is an active area of research. While small animal models such as rodents and rabbits are often used in early pre-clinical work, larger animals that better recapitulate the anatomy and loading of the human joint are required for late-stage preclinical evaluation. Because of their physiological similarities to humans, and low cost relative to other large animals, sheep are routinely used in orthopedic research, including cartilage repair studies. In recent years, there has been considerable research investment into the development of cartilage repair strategies that utilize mesenchymal stem/stromal cells (MSC). In contrast to autologous chondrocytes derived from biopsies of articular cartilage, MSC offer some benefits including greater expansion capacity and elimination of the risk of morbidity at the cartilage biopsy site. The disadvantages of MSC are related to the challenges of inducing and maintaining a stable chondrocyte-like cell population capable of generating hyaline cartilage. Ovine MSC (oMSC) biology and their utility in sheep cartilage repair models have not been reviewed. Herein, we review the biological properties of MSC derived from sheep tissues, and the use of these cells to study articular cartilage repair in this large animal model.
Collapse
Affiliation(s)
- E Music
- Queensland University of Technology, Institute of Health and Biomedical Innovation, Brisbane, QLD, Australia; Translational Research Institute, Brisbane, QLD, Australia.
| | - K Futrega
- Queensland University of Technology, Institute of Health and Biomedical Innovation, Brisbane, QLD, Australia; Translational Research Institute, Brisbane, QLD, Australia.
| | - M R Doran
- Queensland University of Technology, Institute of Health and Biomedical Innovation, Brisbane, QLD, Australia; Translational Research Institute, Brisbane, QLD, Australia; Mater Research Institute - University of Queensland, Brisbane, QLD, Australia; Australian National Centre for the Public Awareness of Science, Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
194
|
Bilgen B, Jayasuriya CT, Owens BD. Current Concepts in Meniscus Tissue Engineering and Repair. Adv Healthc Mater 2018; 7:e1701407. [PMID: 29542287 PMCID: PMC6176857 DOI: 10.1002/adhm.201701407] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/22/2018] [Indexed: 12/13/2022]
Abstract
The meniscus is the most commonly injured structure in the human knee. Meniscus deficiency has been shown to lead to advanced osteoarthritis (OA) due to abnormal mechanical forces, and replacement strategies for this structure have lagged behind other tissue engineering endeavors. The challenges include the complex 3D structure with individualized size parameters, the significant compressive, tensile and shear loads encountered, and the poor blood supply. In this progress report, a review of the current clinical treatments for different types of meniscal injury is provided. The state-of-the-art research in cellular therapies and novel cell sources for these therapies is discussed. The clinically available cell-free biomaterial implants and the current progress on cell-free biomaterial implants are reviewed. Cell-based tissue engineering strategies for the repair and replacement of meniscus are presented, and the current challenges are identified. Tissue-engineered meniscal biocomposite implants may provide an alternative solution for the treatment of meniscal injury to prevent OA in the long run, because of the limitations of the existing therapies.
Collapse
Affiliation(s)
- Bahar Bilgen
- Department of Orthopaedics, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 1 Hoppin St, Providence, RI, 02903, USA
- Providence VA Medical Center, Providence, RI, 02908, USA
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 1 Hoppin St, Providence, RI, 02903, USA
| | - Brett D Owens
- Department of Orthopaedics, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 1 Hoppin St, Providence, RI, 02903, USA
| |
Collapse
|
195
|
Goudarzi F, Mohammadalipour A, Bahabadi M, Goodarzi MT, Sarveazad A, Khodadadi I. Hydrogen peroxide: a potent inducer of differentiation of human adipose-derived stem cells into chondrocytes. Free Radic Res 2018; 52:763-774. [DOI: 10.1080/10715762.2018.1466121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Farjam Goudarzi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Adel Mohammadalipour
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Majid Bahabadi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Taghi Goodarzi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Arash Sarveazad
- Colorectal Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Iraj Khodadadi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
196
|
Abstract
"Although there is ample evidence that beneficial results can be obtained from the use of mesenchymal stem cells, several questions regarding their use remain to be answered. For many of these questions, preclinical models will be helpful, but the task of evaluating and implementing these findings for orthopaedic patients falls onto the shoulders of clinical researchers. Evaluation of these questions is a daunting, but such a challenge fits the concept of personalized medicine in today's medicine."
Collapse
|
197
|
Gabner S, Ertl R, Velde K, Renner M, Jenner F, Egerbacher M, Hlavaty J. Cytokine-induced interleukin-1 receptor antagonist protein expression in genetically engineered equine mesenchymal stem cells for osteoarthritis treatment. J Gene Med 2018; 20:e3021. [PMID: 29608232 PMCID: PMC6001542 DOI: 10.1002/jgm.3021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/23/2018] [Accepted: 03/24/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND A combination of tissue engineering methods employing mesenchymal stem cells (MSCs) together with gene transfer takes advantage of innovative strategies and highlights a new approach for targeting osteoarthritis (OA) and other cartilage defects. Furthermore, the development of systems allowing tunable transgene expression as regulated by natural disease-induced substances is highly desirable. METHODS Bone marrow-derived equine MSCs were transduced with a lentiviral vector expressing interleukin-1 receptor antagonist (IL-1Ra) gene under the control of an inducible nuclear factor-kappa B-responsive promoter and IL-1Ra production upon pro-inflammatory cytokine stimulation [tumor necrosis factor (TNF)α, interleukin (IL)-1β] was analysed. To assess the biological activity of the IL-1Ra protein that was produced and the therapeutic effect of IL-1Ra-expressing MSCs (MSC/IL-1Ra), cytokine-based two- and three-dimensional in vitro models of osteoarthritis using equine chondrocytes were established and quantitative real-time polymerase chain reaction (PCR) analysis was used to measure the gene expression of aggrecan, collagen IIA1, interleukin-1β, interleukin-6, interleukin-8, matrix metalloproteinase-1 and matrix metalloproteinase-13. RESULTS A dose-dependent increase in IL-1Ra expression was found in MSC/IL-1Ra cells upon TNFα administration, whereas stimulation using IL-1β did not lead to IL-1Ra production above the basal level observed in nonstimulated cells as a result of the existing feedback loop. Repeated cycles of induction allowed on/off modulation of transgene expression. In vitro analyses revealed that IL-1Ra protein present in the conditioned medium from MSC/IL-1Ra cells blocks OA onset in cytokine-treated equine chondrocytes and co-cultivation of MSC/IL-1Ra cells with osteoarthritic spheroids alleviates the severity of the osteoarthritic changes. CONCLUSIONS Thus, pro-inflammatory cytokine induced IL-1Ra protein expression from genetically modified MSCs might represent a promising strategy for osteoarthritis treatment.
Collapse
Affiliation(s)
- Simone Gabner
- Institute of Pathology and Forensic Veterinary Medicine, Working Group Histology and EmbryologyUniversity of Veterinary Medicine ViennaViennaAustria
| | - Reinhard Ertl
- VetCORE, Facility for ResearchUniversity of Veterinary MedicineViennaAustria
| | - Karsten Velde
- Equine University HospitalUniversity of Veterinary Medicine ViennaViennaAustria
| | - Matthias Renner
- Division of Medical BiotechnologyPaul‐Ehrlich‐InstitutLangenGermany
| | - Florien Jenner
- Equine University HospitalUniversity of Veterinary Medicine ViennaViennaAustria
| | - Monika Egerbacher
- Institute of Pathology and Forensic Veterinary Medicine, Working Group Histology and EmbryologyUniversity of Veterinary Medicine ViennaViennaAustria
| | - Juraj Hlavaty
- Institute of Pathology and Forensic Veterinary Medicine, Working Group Histology and EmbryologyUniversity of Veterinary Medicine ViennaViennaAustria
| |
Collapse
|
198
|
Assis-Ribas T, Forni MF, Winnischofer SMB, Sogayar MC, Trombetta-Lima M. Extracellular matrix dynamics during mesenchymal stem cells differentiation. Dev Biol 2018; 437:63-74. [PMID: 29544769 DOI: 10.1016/j.ydbio.2018.03.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/05/2018] [Accepted: 03/02/2018] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are stromal cells that display self-renewal and multipotent differentiation capacity. The repertoire of mature cells generated ranges but is not restricted to: fat, bone and cartilage. Their potential importance for both cell therapy and maintenance of in vivo homeostasis is indisputable. Nonetheless, both their in vivo identity and use in cell therapy remain elusive. A drawback generated by this fact is that little is known about the MSC niche and how it impacts differentiation and homeostasis maintenance. Hence, the roles played by the extracellular matrix (ECM) and its main regulators namely: the Matrix Metalloproteinases (MMPs) and their counteracting inhibitors (TIMPs and RECK) upon stem cells differentiation are only now beginning to be unveiled. Here, we will focus on mesenchymal stem cells and review the main mechanisms involved in adipo, chondro and osteogenesis, discussing how the extracellular matrix can impact not only lineage commitment, but, also, their survival and potentiality. This review critically analyzes recent work in the field in an effort towards a better understanding of the roles of Matrix Metalloproteinases and their inhibitors in the above-cited events.
Collapse
Affiliation(s)
- Thais Assis-Ribas
- NUCEL-NETCEM-Faculdade de Medicina, Departamento de Clínica Médica, Universidade de São Paulo, São Paulo, SP 05360-120, Brazil
| | - Maria Fernanda Forni
- Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | | | - Mari Cleide Sogayar
- NUCEL-NETCEM-Faculdade de Medicina, Departamento de Clínica Médica, Universidade de São Paulo, São Paulo, SP 05360-120, Brazil; Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Marina Trombetta-Lima
- NUCEL-NETCEM-Faculdade de Medicina, Departamento de Clínica Médica, Universidade de São Paulo, São Paulo, SP 05360-120, Brazil.
| |
Collapse
|
199
|
Rinker TE, Philbrick BD, Hettiaratchi MH, Smalley DM, McDevitt TC, Temenoff JS. Microparticle-mediated sequestration of cell-secreted proteins to modulate chondrocytic differentiation. Acta Biomater 2018; 68:125-136. [PMID: 29292168 DOI: 10.1016/j.actbio.2017.12.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/05/2017] [Accepted: 12/22/2017] [Indexed: 10/18/2022]
Abstract
Protein delivery is often used in tissue engineering applications to control differentiation processes, but is limited by protein instability and cost. An alternative approach is to control the cellular microenvironment through biomaterial-mediated sequestration of cell-secreted proteins important to differentiation. Thus, we utilized heparin-based microparticles to modulate cellular differentiation via protein sequestration in an in vitro model system of endochondral ossification. Heparin and poly(ethylene-glycol) (PEG; a low-binding material control)-based microparticles were incorporated into ATDC5 cell spheroids or incubated with ATDC5 cells in transwell culture. Reduced differentiation was observed in the heparin microparticle group as compared to PEG and no microparticle-containing groups. To determine if observed changes were due to sequestration of cell-secreted protein, the proteins sequestered by heparin microparticles were analyzed using SDS-PAGE and mass spectrometry. It was found that heparin microparticles bound insulin-like growth factor binding proteins (IGFBP)-3 and 5. When incubated with a small-molecule inhibitor of IGFBPs, NBI 31772, a similar delay in differentiation of ATDC5 cells was observed. These results indicate that heparin microparticles modulated chondrocytic differentiation in this system via sequestration of cell-secreted protein, a technique that could be beneficial in the future as a means to control cellular differentiation processes. STATEMENT OF SIGNIFICANCE In this work, we present a proof-of-principle set of experiments in which heparin-based microparticles are shown to modulate cellular differentiation through binding of cell-secreted protein. Unlike existing systems that rely on expensive protein with limited half-lives to elicit changes in cellular behavior, this technique focuses on temporal modulation of cell-generated proteins. This technique also provides a biomaterials-based method that can be used to further identify sequestered proteins of interest. Thus, this work indicates that glycosaminoglycan-based biomaterial approaches could be used as substitutes or additions to traditional methods for modulating and identifying the cell-secreted proteins involved in directing cellular behavior.
Collapse
|
200
|
Bellavia D, Veronesi F, Carina V, Costa V, Raimondi L, De Luca A, Alessandro R, Fini M, Giavaresi G. Gene therapy for chondral and osteochondral regeneration: is the future now? Cell Mol Life Sci 2018; 75:649-667. [PMID: 28864934 PMCID: PMC11105387 DOI: 10.1007/s00018-017-2637-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/28/2017] [Indexed: 12/26/2022]
Abstract
Gene therapy might represent a promising strategy for chondral and osteochondral defects repair by balancing the management of temporary joint mechanical incompetence with altered metabolic and inflammatory homeostasis. This review analysed preclinical and clinical studies on gene therapy for the repair of articular cartilage defects performed over the last 10 years, focussing on expression vectors (non-viral and viral), type of genes delivered and gene therapy procedures (direct or indirect). Plasmids (non-viral expression vectors) and adenovirus (viral vectors) were the most employed vectors in preclinical studies. Genes delivered encoded mainly for growth factors, followed by transcription factors, anti-inflammatory cytokines and, less frequently, by cell signalling proteins, matrix proteins and receptors. Direct injection of the expression vector was used less than indirect injection of cells, with or without scaffolds, transduced with genes of interest and then implanted into the lesion site. Clinical trials (phases I, II or III) on safety, biological activity, efficacy, toxicity or bio-distribution employed adenovirus viral vectors to deliver growth factors or anti-inflammatory cytokines, for the treatment of osteoarthritis or degenerative arthritis, and tumour necrosis factor receptor or interferon for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Daniele Bellavia
- Rizzoli Orthopedic Institute, Bologna, Italy.
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy.
| | - F Veronesi
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - V Carina
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - V Costa
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - L Raimondi
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - A De Luca
- Rizzoli Orthopedic Institute, Bologna, Italy
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
| | - R Alessandro
- Biology and Genetics Unit, Department of Biopathology and Medical Biotechnology, University of Palermo, Palermo, Italy
| | - M Fini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - G Giavaresi
- Innovative Technology Platforms for Tissue Engineering, Theranostic and Oncology, Rizzoli Orthopaedic Institute, Via Divisi 83, 90133, Palermo, Italy
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| |
Collapse
|