1
|
Cieśla J, Tomsia M. Differentiation of stem cells into chondrocytes and their potential clinical application in cartilage regeneration. Histochem Cell Biol 2025; 163:27. [PMID: 39863760 DOI: 10.1007/s00418-025-02356-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2025] [Indexed: 01/27/2025]
Abstract
Cartilage diseases and injuries are considered difficult to treat owing to the low regenerative capacity of this tissue. Using stem cells (SCs) is one of the potential methods of treating cartilage defects and creating functional cartilage models for transplants. Their ability to proliferate and to generate functional chondrocytes, a natural tissue environment, and extracellular cartilage matrix, makes SCs a new opportunity for patients with articular injuries or incurable diseases, such as osteoarthritis (OA). The review summarizes the most important scientific reports on biology and mechanisms of SC-derived chondrogenesis and sources of SCs for chondrogenic purposes. Additionally, it focuses on the genetic mechanisms, microRNA (miRNA) regulation, and epigenetic processes steering the chondrogenic differentiation of SCs. It also describes the attempts to create functional cartilage with tissue engineering using growth factors and scaffolds. Finally, it presents the challenges that researchers will have to face in the future to effectuate SC differentiation methods into clinical practice for treating cartilage diseases.
Collapse
Affiliation(s)
- Julia Cieśla
- School of Medicine in Katowice, Medical University of Silesia, 18 Medyków Street, 40-752, Katowice, Poland
| | - Marcin Tomsia
- Department of Forensic Medicine and Forensic Toxicology, Medical University of Silesia, 18 Medyków Street, 40-752, Katowice, Poland.
| |
Collapse
|
2
|
Xu X, Yu H, Yang M, Xie J, Xu K, Li E, Wan X, Wang J, Wang G, Pan Y, Xu P, Guo J. Unraveling the relationship between obstructive sleep apnea and osteoarthritis: A multivariate mendelian randomization highlighting the role of BMI as a confounding factor. Exp Gerontol 2025; 199:112657. [PMID: 39672284 DOI: 10.1016/j.exger.2024.112657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Osteoarthritis (OA) and obstructive sleep apnea (OSA) are prevalent chronic conditions with emerging evidence suggesting a potential link. However, the causality of this association remains unclear, possibly influenced by confounders like high body mass index (BMI). This study aimed to explore causal relationships between OA and OSA using Mendelian randomization (MR). METHODS MR analysis was performed to assess causality between OA and OSA. Inverse variance weighting (IVW) was the primary MR method, complemented by sensitivity analyses, including MR steiger, MR-Egger, MR-PRESSO, weighted median, heterogeneity tests, and leave-one-out approaches to evaluate pleiotropy and confirm the robustness of the causal estimates. To exclude confounding effects of BMI, we also used a multivariate MR (MVMR). RESULTS After adjusting for BMI through MVMR, no significant causal relationship was identified between genetically predicted OSA and OA phenotypes, including knee (KOA) and hip osteoarthritis (HOA), suggesting that obesity largely drives the observed relationship between these conditions. Similarly, MR steiger doesn't support a causal effect from OA on OSA. Sensitivity analyses confirmed the robustness of these results, with no significant evidence of horizontal pleiotropy or heterogeneity affecting outcomes. The findings indicate that BMI acts as a critical confounder in the relationship between OSA and OA, rather than OSA directly contributing to OA development. CONCLUSIONS Our findings indicate that there is no significant causal relationship between genetically predicted OSA and OA after adjusting for BMI. These findings underscore obesity as the primary shared risk factor, highlighting the importance of weight management as a key strategy for mitigating the risks of both conditions. Future research should aim to validate these findings in diverse populations and explore other metabolic pathways that may contribute to these complex associations.
Collapse
Affiliation(s)
- Xin Xu
- Xi'an Jiaotong University, Xi'an 710054, China; Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710054, China
| | - Hui Yu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710054, China
| | - Mingyi Yang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Jiale Xie
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710054, China
| | - Ke Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Erliang Li
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710054, China
| | - Xianjie Wan
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Jiachen Wang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710054, China
| | - Guoqiang Wang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710054, China
| | - Ying Pan
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Peng Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710054, China.
| | - Junfei Guo
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China; Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an 710054, China.
| |
Collapse
|
3
|
Yi J, Byun Y, Kang SS, Shim KM, Jang K, Lee JY. Enhanced Chondrogenic Differentiation of Electrically Primed Human Mesenchymal Stem Cells for the Regeneration of Osteochondral Defects. Biomater Res 2024; 28:0109. [PMID: 39697183 PMCID: PMC11654951 DOI: 10.34133/bmr.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/12/2024] [Accepted: 10/26/2024] [Indexed: 12/20/2024] Open
Abstract
Background: Mesenchymal stem cells (MSCs) offer a promising avenue for cartilage regeneration; however, their therapeutic efficacy requires substantial improvement. Cell priming using electrical stimulation (ES) is a promising approach to augmenting the therapeutic potential of MSCs and has shown potential for various regenerative applications. This study aimed to promote the ES-mediated chondrogenic differentiation of human MSCs and facilitate the repair of injured articular cartilage. Methods: MSCs were subjected to ES under various conditions (e.g., voltage, frequency, and number of repetitions) to enhance their capability of chondrogenesis and cartilage regeneration. Chondrogenic differentiation of electrically primed MSCs (epMSCs) was assessed based on gene expression and sulfated glycosaminoglycan production, and epMSCs with hyaluronic acid were transplanted into a rat osteochondral defect model. Transcriptomic analysis was performed to determine changes in gene expression by ES. Results: epMSCs exhibited significantly increased chondrogenic gene expression and sulfated glycosaminoglycan production compared with those in unstimulated controls. Macroscopic and histological results showed that in vivo epMSC transplantation considerably enhanced cartilage regeneration. Furthermore, ES markedly altered the expression of numerous genes of MSCs, including those associated with the extracellular matrix, the Wnt signaling pathway, and cartilage development. Conclusion: ES can effectively prime MSCs to improve articular cartilage repair, offering a promising strategy for enhancing the efficacy of various MSC-based therapies.
Collapse
Affiliation(s)
- Jongdarm Yi
- School of Materials Science and Engineering,
Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Yujin Byun
- Department of Veterinary Surgery, College of Veterinary Medicine and BK21 FOUR Program,
Chonnam National University, Gwangju 61186, Republic of Korea
- Biomaterial R&BD Center,
Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seong Soo Kang
- Department of Veterinary Surgery, College of Veterinary Medicine and BK21 FOUR Program,
Chonnam National University, Gwangju 61186, Republic of Korea
- Biomaterial R&BD Center,
Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kyung Mi Shim
- Department of Veterinary Surgery, College of Veterinary Medicine and BK21 FOUR Program,
Chonnam National University, Gwangju 61186, Republic of Korea
- Biomaterial R&BD Center,
Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kwangsik Jang
- Department of Veterinary Surgery, College of Veterinary Medicine and BK21 FOUR Program,
Chonnam National University, Gwangju 61186, Republic of Korea
- Biomaterial R&BD Center,
Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering,
Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| |
Collapse
|
4
|
Al-Sherief S, El-Hadidy A, Hamed S, El-Hawwary A, Mazroa S. Bone marrow mesenchymal stem cells (BM-MSCs) modulate MMP9 expression and promote articular cartilage regeneration in knee joint of a model of arthritis induced in adult rat: histological and immunohistochemical study. J Mol Histol 2024; 56:38. [PMID: 39661261 DOI: 10.1007/s10735-024-10284-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 10/31/2024] [Indexed: 12/12/2024]
Abstract
Arthritis is characterized by the progressive degeneration of articular cartilage, and the avascular nature of cartilage limits its capacity for self-repair. Stem cells are considered a promising treatment option due to their multipotent differentiation potential. The aim of this work was to investigate the structural changes in the hyaline articular cartilage of the knee joint in a model of arthritis induced by complete Freund's adjuvant, and to assess intra-articular injection of bone marrow mesenchymal stem cells (BM-MSCs) through both histological and immunohistochemical study. Adult male albino rats were divided into four groups: group 0 (donor group), group I (control group), group II (arthritis group) and group III (BM-MSCs treated arthritis group). Samples were collected 2, 6 and 10 weeks after the onset of the experiment. Sections were stained with; hematoxylin and eosin, Safranin O fast green stain, Masson's trichrome stain and anti-MMP9 antibody. In Group II (arthritis group), the articular cartilage showed signs of degeneration, including chondrocyte extensive proliferation, fibrillations, fissuring, and denudation, with fibrous tissue covering the exposed surface. There was a significant decrease in cartilage thickness, collagen content, and proteoglycan levels. The integrated density of MMP9 in the cartilage was significantly increased compared to Group I (control group). In contrast, Group III (BM-MSCs-treated arthritis group) exhibited a continuous cartilage surface with no cracks or fissures. There was a significant increase in cartilage thickness, collagen content, and proteoglycan levels, while the integrated density of MMP9 was significantly decreased compared to Group II (arthritis group).
Collapse
Affiliation(s)
- Sara Al-Sherief
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Azza El-Hadidy
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Shereen Hamed
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amany El-Hawwary
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Shireen Mazroa
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
5
|
Rao J, Xie H, Liang Z, Yang Z, Chen P, Zhou M, Xu X, Lin Y, Lin F, Wang R, Wang C, Chen C. Hypoxic-preconditioned mesenchymal stem cell-derived small extracellular vesicles inhibit neuronal death after spinal cord injury by regulating the SIRT1/Nrf2/HO-1 pathway. Front Pharmacol 2024; 15:1419390. [PMID: 39246654 PMCID: PMC11377843 DOI: 10.3389/fphar.2024.1419390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024] Open
Abstract
Background Oxidative stress and apoptosis of neurons significantly contribute to the pathophysiological cascade of spinal cord injury (SCI). However, the role of hypoxic-preconditioned mesenchymal stem cell-derived small extracellular vesicles (H-sEVs) in promoting SCI repair remains unclear. Hence, the present study aims to investigate the regulatory effects of H-sEVs on neuronal oxidative stress and apoptotic responses following SCI. Methods The administration of H-sEVs of SCI rats was assessed using behavioral evaluations such as Basso-Beattie-Bresnahan (BBB) scores, neuroelectrophysiological monitoring, and Catwalk gait analysis. Indices of oxidative stress (including superoxide dismutase [SOD], total antioxidant capacity [T-AOC], and malondialdehyde [MDA]) were measured. Neuronal survival was evaluated through Nissl staining, while the expression level of sirtuin 1 (SIRT1) was examined using immunohistochemical staining. Additionally, histological evaluation of lesion size was performed using hematoxylin-eosin (HE) staining. Tunel cell apoptosis staining and analysis of apoptosis-associated proteins (B-cell lymphoma-2 [Bcl2] and BCL2-Associated X [Bax]) were conducted through immunofluorescence staining and western blot, respectively. Furthermore, the model of oxidative stress was established using PC12 cells, and apoptosis levels were assessed via flow cytometry and western blot analysis. Importantly, to ascertain the critical role of SIRT1, we performed SIRT1 knockout experiments in PC12 cells using lentivirus transfection, followed by western blot. Results Using those behavioral evaluations, we observed significant functional improvement after H-sEVs treatment. Nissl staining revealed that H-sEVs treatment promoted neuronal survival. Moreover, we found that H-sEVs effectively reduced oxidative stress levels after SCI. HE staining demonstrated that H-sEVs could reduce lesion area. Immunohistochemical analysis revealed that H-sEVs enhanced SIRT1 expression. Furthermore, Tunel cell apoptosis staining and western blot analysis of apoptosis-related proteins confirmed the anti-apoptotic effects of H-sEVs. The PC12 cells were used to further substantiate the neuroprotective properties of H-sEVs by significantly inhibiting neuronal death and attenuating oxidative stress. Remarkably, SIRT1 knockout in PC12 cells reversed the antioxidant stress effects induced by H-sEVs treatment. Additionally, we elucidated the involvement of the downstream Nrf2/HO-1 signaling pathway. Conclusion Our study provides valuable insights into the effects of H-sEVs on neuronal oxidative stress and apoptosis after SCI. These findings underscore the potential clinical significance of H-sEVs-based therapies for SCI.
Collapse
Affiliation(s)
- Jian Rao
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| | - Haishu Xie
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| | - Zeyan Liang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| | - Zhelun Yang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| | - Pingping Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| | - Maochao Zhou
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| | - Xiongjie Xu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| | - Yike Lin
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| | - Fabin Lin
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| | - Rui Wang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| | - Chunhua Wang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| | - Chunmei Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Neurosurgical Institute, Fuzhou, Fujian, China
| |
Collapse
|
6
|
Khajeh S, Razban V, Naeimzadeh Y, Nadimi E, Asadi-Golshan R, Heidari Z, Talaei-Khozani T, Dehghani F, Mostafavi-Pour Z, Shirali M. Plasticity Comparison of Two Stem Cell Sources with Different Hox Gene Expression Profiles in Response to Cobalt Chloride Treatment during Chondrogenic Differentiation. BIOLOGY 2024; 13:560. [PMID: 39194498 DOI: 10.3390/biology13080560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024]
Abstract
The limited self-repair capacity of articular cartilage is a challenge for healing injuries. While mesenchymal stem/stromal cells (MSCs) are a promising approach for tissue regeneration, the criteria for selecting a suitable cell source remain undefined. To propose a molecular criterion, dental pulp stem cells (DPSCs) with a Hox-negative expression pattern and bone marrow mesenchymal stromal cells (BMSCs), which actively express Hox genes, were differentiated towards chondrocytes in 3D pellets, employing a two-step protocol. The MSCs' response to preconditioning by cobalt chloride (CoCl2), a hypoxia-mimicking agent, was explored in an assessment of the chondrogenic differentiation's efficiency using morphological, histochemical, immunohistochemical, and biochemical experiments. The preconditioned DPSC pellets exhibited significantly elevated levels of collagen II and glycosaminoglycans (GAGs) and reduced levels of the hypertrophic marker collagen X. No significant effect on GAGs production was observed in the preconditioned BMSC pellets, but collagen II and collagen X levels were elevated. While preconditioning did not modify the ALP specific activity in either cell type, it was notably lower in the DPSCs differentiated pellets compared to their BMSCs counterparts. These results could be interpreted as demonstrating the higher plasticity of DPSCs compared to BMSCs, suggesting the contribution of their unique molecular characteristics, including their negative Hox expression pattern, to promote a chondrogenic differentiation potential. Consequently, DPSCs could be considered compelling candidates for future cartilage cell therapy.
Collapse
Affiliation(s)
- Sahar Khajeh
- Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Elham Nadimi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Reza Asadi-Golshan
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran 14166-34793, Iran
| | - Zahra Heidari
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Tahereh Talaei-Khozani
- Tissue Engineering Laboratory, Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Farzaneh Dehghani
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Zohreh Mostafavi-Pour
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Masoud Shirali
- School of Biological Sciences, Queen's University Belfast, Belfast BT9 5AJ, UK
- Agri-Food and Biosciences Institute, Hillsborough BT26 6DR, UK
| |
Collapse
|
7
|
Yang Z, Liang Z, Rao J, Xie H, Zhou M, Xu X, Lin Y, Lin F, Wang C, Chen C. Hypoxic-preconditioned mesenchymal stem cell-derived small extracellular vesicles promote the recovery of spinal cord injury by affecting the phenotype of astrocytes through the miR-21/JAK2/STAT3 pathway. CNS Neurosci Ther 2024; 30:e14428. [PMID: 37641874 PMCID: PMC10915983 DOI: 10.1111/cns.14428] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/14/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Secondary injury after spinal cord injury (SCI) is a major obstacle to their neurological recovery. Among them, changes in astrocyte phenotype regulate secondary injury dominated by neuroinflammation. Hypoxia-preconditioned mesenchymal stem cells (MSCs)-derived extracellular vesicle (H-EV) plays a multifaceted role in secondary injury by interacting with cellular components and signaling pathways. They possess anti-inflammatory properties, regulate oxidative stress, and modulate apoptotic pathways, promoting cell survival and reducing neuronal loss. Given the unique aspects of secondary injury, H-EV shows promise as a therapeutic approach to mitigate its devastating consequences. Our study aimed to determine whether H-EV could promote SCI repair by altering the phenotype of astrocytes. METHODS Rat bone marrow MSCs (BMSCs) and EVs secreted by them were extracted and characterized. After the SCI model was successfully constructed, EV and H-EV were administered into the tail vein of the rats, respectively, and then their motor function was evaluated by the Basso-Beattie-Bresnahan (BBB) score, Catwalk footprint analysis, and electrophysiological monitoring. The lesion size of the spinal cord was evaluated by hematoxylin-eosin (HE) staining. The key point was to use glial fibrillary acidic protein (GFAP) as a marker of reactive astrocytes to co-localize with A1-type marker complement C3 and A2-type marker S100A10, respectively, to observe phenotypic changes in astrocytes within tissues. The western blot (WB) of the spinal cord was also used to verify the results. We also compared the efficacy differences in apoptosis and inflammatory responses using terminal deoxynucleotidyl transferase dUTP terminal labeling (TUNEL) assay, WB, and enzyme-linked immunosorbent assay (ELISA). Experiments in vitro were also performed to verify the results. Subsequently, we performed microRNA (miRNA) sequencing analysis of EV and H-EV and carried out a series of knockdown and overexpression experiments to further validate the mechanism by which miRNA in H-EV plays a role in promoting astrocyte phenotypic changes, as well as the regulated signaling pathways, using WB both in vivo and in vitro. RESULTS Our findings suggest that H-EV is more effective than EV in the recovery of motor function, anti-apoptosis, and anti-inflammatory effects after SCI, both in vivo and in vitro. More importantly, H-EV promoted the conversion of A1 astrocytes into A2 astrocytes more than EV. Moreover, miR-21, which was found to be highly expressed in H-EV by miRNA sequencing results, was also demonstrated to influence changes in astrocyte phenotype through a series of knockdown and overexpression experiments. At the same time, we also found that H-EV might affect astrocyte phenotypic alterations by delivering miR-21 targeting the JAK2/STAT3 signaling pathway. CONCLUSION H-EV exerts neuroprotective effects by delivering miR-21 to promote astrocyte transformation from the A1 phenotype to the A2 phenotype, providing new targets and ideas for the treatment of SCI.
Collapse
Affiliation(s)
- Zhelun Yang
- Department of NeurosurgeryFujian Medical University Union HospitalFuzhouFujianChina
| | - Zeyan Liang
- Department of NeurosurgeryFujian Medical University Union HospitalFuzhouFujianChina
| | - Jian Rao
- Department of NeurosurgeryFujian Medical University Union HospitalFuzhouFujianChina
| | - Haishu Xie
- Department of NeurosurgeryFujian Medical University Union HospitalFuzhouFujianChina
| | - Maochao Zhou
- Department of NeurosurgeryFujian Medical University Union HospitalFuzhouFujianChina
| | - Xiongjie Xu
- Department of NeurosurgeryFujian Medical University Union HospitalFuzhouFujianChina
| | - Yike Lin
- Department of NeurosurgeryFujian Medical University Union HospitalFuzhouFujianChina
| | - Fabin Lin
- Department of NeurosurgeryFujian Medical University Union HospitalFuzhouFujianChina
| | - Chunhua Wang
- Department of NeurosurgeryFujian Medical University Union HospitalFuzhouFujianChina
| | - Chunmei Chen
- Department of NeurosurgeryFujian Medical University Union HospitalFuzhouFujianChina
| |
Collapse
|
8
|
Reina-Mahecha A, Beers MJ, van der Veen HC, Zuhorn IS, van Kooten TG, Sharma PK. A Review of the Role of Bioreactors for iPSCs-Based Tissue-Engineered Articular Cartilage. Tissue Eng Regen Med 2023; 20:1041-1052. [PMID: 37861960 PMCID: PMC10645985 DOI: 10.1007/s13770-023-00573-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most common degenerative joint disease without an ultimate treatment. In a search for novel approaches, tissue engineering (TE) has shown great potential to be an effective way for hyaline cartilage regeneration and repair in advanced stages of OA. Recently, induced pluripotent stem cells (iPSCs) have been appointed to be essential stem cells for degenerative disease treatment because they allow a personalized medicine approach. For clinical translation, bioreactors in combination with iPSCs-engineerd cartilage could match patients needs, serve as platform for large-scale patient specific cartilage production, and be a tool for patient OA modelling and drug screening. Furthermore, to minimize in vivo experiments and improve cell differentiation and cartilage extracellular matrix (ECM) deposition, TE combines existing approaches with bioreactors. METHODS This review summarizes the current understanding of bioreactors and the necessary parameters when they are intended for cartilage TE, focusing on the potential use of iPSCs. RESULTS Bioreactors intended for cartilage TE must resemble the joint cavity niche. However, recreating human synovial joints is not trivial because the interactions between various stimuli are not entirely understood. CONCLUSION The use of mechanical and electrical stimulation to differentiate iPSCs, and maintain and test chondrocytes are key stimuli influencing hyaline cartilage homeostasis. Incorporating these stimuli to bioreactors can positively impact cartilage TE approaches and their possibility for posterior translation into the clinics.
Collapse
Affiliation(s)
- Alejandro Reina-Mahecha
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Martine J Beers
- Department of Orthopedics, University Medical Center Groningen, Groningen, The Netherlands
| | - Hugo C van der Veen
- Department of Orthopedics, University Medical Center Groningen, Groningen, The Netherlands
| | - Inge S Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Theo G van Kooten
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands
| | - Prashant K Sharma
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, FB40, Antonius Deusinglaan -1, 9713AV, Groningen, The Netherlands.
| |
Collapse
|
9
|
Zecca PA, Reguzzoni M, Borgese M, Protasoni M, Filibian M, Raspanti M. Investigating the interfaces of the epiphyseal plate: An integrated approach of histochemistry, microtomography and SEM. J Anat 2023; 243:870-877. [PMID: 37391907 PMCID: PMC10557393 DOI: 10.1111/joa.13924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/24/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023] Open
Abstract
We investigated the interfaces of the epiphyseal plate with over- and underlying bone segments using an integrated approach of histochemistry, microtomography and scanning electron microscopy (SEM) to overcome the inherent limitations of sections-based techniques. Microtomography was able to provide an unobstructed, frontal view of large expanses of the two bone surfaces facing the growth plate, while SEM observation after removal of the soft matrix granted an equally unhindered access with a higher resolution. The two interfaces appeared widely dissimilar. On the diaphyseal side the hypertrophic chondrocytes were arranged in tall columns packed in a sort of compact palisade; the interposed extracellular matrix was actively calcifying into a thick mineralized crust growing towards the epiphysis. Behind the mineralization front, histochemical data revealed a number of surviving cartilage islets which were being slowly remodelled into bone. In contrast, the epiphyseal side of the cartilage consisted of a relatively quiescent reserve zone whose mineralization was marginal in amount and discontinuous in extension; the epiphyseal bone consisted of a loose trabecular meshwork, with ample vascular spaces opening directly into the non-mineralized cartilage. On both sides the calcification process took place through the formation of spheroidal bodies 1-2 μm wide which gradually grew by apposition and coalesced into a solid mass, in a way distinctly different from that of bone and other calcified tissues.
Collapse
Affiliation(s)
| | | | - Marina Borgese
- Department of Medicine & SurgeryInsubria UniversityVareseItaly
| | | | - Marta Filibian
- Centro Grandi StrumentiUniversity of PaviaPaviaItaly
- Istituto Nazionale di Fisica Nucleare, Pavia UnitPaviaItaly
| | - Mario Raspanti
- Department of Medicine & SurgeryInsubria UniversityVareseItaly
| |
Collapse
|
10
|
Er E, Vahabi A, Kaya Biçer E, Yılmaz Ö, Aydoğdu S. Use of polyglycolic acid-hyaluronic acid/β-tricalcium phosphate scaffold with or without mesenchymal stem cells found ineffective in treating osteochondral lesions in rabbit knees. ACTA ORTHOPAEDICA ET TRAUMATOLOGICA TURCICA 2023; 57:209-214. [PMID: 37828858 PMCID: PMC10724785 DOI: 10.5152/j.aott.2023.23054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 09/11/2023] [Indexed: 10/14/2023]
Abstract
OBJECTIVE In this experimental animal study, a novel bilayered scaffold used in the treatment of osteochondral defects in rabbit knees was evaluated. This novel scaffold's upper (cartilage) layer consists of polyglycolic acid and hyaluronic acid, and the lower (bone) layer consists of β-tricalcium phosphate. The purpose of this study was to evaluate the efficacy of this novel scaffold, combined with or without mesenchymal stem cells (MSCs), in the treatment of osteochondral defects in rabbit knees. METHODS Osteochondral defects were created in the left femoral trochlea of 30 rabbits. In group A, defects were treated with scaffold combined with MSCs; in group B, defects were treated with cell-free scaffolds; and group C was a control group with defects left untreated. In the 12th week, animals were sacrificed for macroscopic evaluation. RESULTS The mean International Cartilage Repair Society (ICRS) macroscopic scores were 4.95 for group A, 6.16 for group B, and 8.25 for group C. The mean Oswestry Arthroscopic Scores (OAS) were 1.65 for group A, 3.39 for group B, and 6.05 for group C. The macroscopic scores were significantly higher in group C than group A for ICRS scores and group A and group B for OAS (P < .001, P < .000, P < .022). CONCLUSION In essence, our findings indicate that the newly developed osteochondral scaffold, when tested in a rabbit model, is not as effective as expected in repairing full-thickness osteochondral defects, with or without the supplementation of MSCs. Further investigation is required to enhance the effectiveness of this novel combination.
Collapse
Affiliation(s)
- Erdem Er
- Department of Orthopedics and Traumatology, Ege University School of Medicine, İzmir, Turkey
| | - Arman Vahabi
- Department of Orthopedics and Traumatology, Ege University School of Medicine, İzmir, Turkey
| | - Elcil Kaya Biçer
- Department of Orthopedics and Traumatology, Ege University School of Medicine, İzmir, Turkey
| | - Özlem Yılmaz
- Department of Histology and Embryology, Ege University School of Medicine, İzmir, Turkey
| | - Semih Aydoğdu
- Department of Orthopedics and Traumatology, Ege University School of Medicine, İzmir, Turkey
| |
Collapse
|
11
|
Hammad M, Veyssiere A, Leclercq S, Patron V, Baugé C, Boumédiene K. Hypoxia Differentially Affects Chondrogenic Differentiation of Progenitor Cells from Different Origins. Int J Stem Cells 2023; 16:304-314. [PMID: 37105555 PMCID: PMC10465331 DOI: 10.15283/ijsc21242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 04/29/2023] Open
Abstract
Background and Objectives Ear cartilage malformations are commonly encountered problems in reconstructive surgery, since cartilage has low self-regenerating capacity. Malformations that impose psychological and social burden on one's life are currently treated using ear prosthesis, synthetic implants or autologous flaps from rib cartilage. These approaches are challenging because not only they request high surgical expertise, but also they lack flexibility and induce severe donor-site morbidity. Through the last decade, tissue engineering gained attention where it aims at regenerating human tissues or organs in order to restore normal functions. This technique consists of three main elements, cells, growth factors, and above all, a scaffold that supports cells and guides their behavior. Several studies have investigated different scaffolds prepared from both synthetic or natural materials and their effects on cellular differentiation and behavior. Methods and Results In this study, we investigated a natural scaffold (alginate) as tridimensional hydrogel seeded with progenitors from different origins such as bone marrow, perichondrium and dental pulp. In contact with the scaffold, these cells remained viable and were able to differentiate into chondrocytes when cultured in vitro. Quantitative and qualitative results show the presence of different chondrogenic markers as well as elastic ones for the purpose of ear cartilage, upon different culture conditions. Conclusions We confirmed that auricular perichondrial cells outperform other cells to produce chondrogenic tissue in normal oxygen levels and we report for the first time the effect of hypoxia on these cells. Our results provide updates for cartilage engineering for future clinical applications.
Collapse
Affiliation(s)
- Mira Hammad
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
| | - Alexis Veyssiere
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
- Service de chirurgie Maxillo-faciale, CHU de Caen, Caen, France
| | - Sylvain Leclercq
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Clinique Saint Martin, Service de Chirurgie Orthopédique, Caen, France
| | - Vincent Patron
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Service ORL et chirurgie cervico-faciale, CHU de Caen, Caen, France
| | - Catherine Baugé
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
| | - Karim Boumédiene
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
| |
Collapse
|
12
|
Ouzin M, Kogler G. Mesenchymal Stromal Cells: Heterogeneity and Therapeutical Applications. Cells 2023; 12:2039. [PMID: 37626848 PMCID: PMC10453316 DOI: 10.3390/cells12162039] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Mesenchymal stromal cells nowadays emerge as a major player in the field of regenerative medicine and translational research. They constitute, with their derived products, the most frequently used cell type in different therapies. However, their heterogeneity, including different subpopulations, the anatomic source of isolation, and high donor-to-donor variability, constitutes a major controversial issue that affects their use in clinical applications. Furthermore, the intrinsic and extrinsic molecular mechanisms underlying their self-renewal and fate specification are still not completely elucidated. This review dissects the different heterogeneity aspects of the tissue source associated with a distinct developmental origin that need to be considered when generating homogenous products before their usage for clinical applications.
Collapse
Affiliation(s)
- Meryem Ouzin
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Hospital Düsseldorf, 40225 Düsseldorf, Germany;
| | | |
Collapse
|
13
|
Peng Y, Jiang H, Zuo HD. Factors affecting osteogenesis and chondrogenic differentiation of mesenchymal stem cells in osteoarthritis. World J Stem Cells 2023; 15:548-560. [PMID: 37424946 PMCID: PMC10324504 DOI: 10.4252/wjsc.v15.i6.548] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 06/26/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that often involves progressive cartilage degeneration and bone destruction of subchondral bone. At present, clinical treatment is mainly for pain relief, and there are no effective methods to delay the progression of the disease. When this disease progresses to the advanced stage, the only treatment option for most patients is total knee replacement surgery, which causes patients great pain and anxiety. As a type of stem cell, mesenchymal stem cells (MSCs) have multidirectional differentiation potential. The osteogenic differentiation and chondrogenic differentiation of MSCs can play vital roles in the treatment of OA, as they can relieve pain in patients and improve joint function. The differentiation direction of MSCs is accurately controlled by a variety of signaling pathways, so there are many factors that can affect the differentiation direction of MSCs by acting on these signaling pathways. When MSCs are applied to OA treatment, the microenvironment of the joints, injected drugs, scaffold materials, source of MSCs and other factors exert specific impacts on the differentiation direction of MSCs. This review aims to summarize the mechanisms by which these factors influence MSC differentiation to produce better curative effects when MSCs are applied clinically in the future.
Collapse
Affiliation(s)
- Yi Peng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Hai Jiang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Hou-Dong Zuo
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
- Department of Radiology, Chengdu Xinhua Hospital, Chengdu 610067, Sichuan Province, China
| |
Collapse
|
14
|
Cho GH, Bae HC, Cho WY, Jeong EM, Park HJ, Yang HR, Wang SY, Kim YJ, Shin DM, Chung HM, Kim IG, Han HS. High-glutathione mesenchymal stem cells isolated using the FreSHtracer probe enhance cartilage regeneration in a rabbit chondral defect model. Biomater Res 2023; 27:54. [PMID: 37259149 PMCID: PMC10233867 DOI: 10.1186/s40824-023-00398-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/20/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are a promising cell source for cartilage regeneration. However, the function of MSC can vary according to cell culture conditions, donor age, and heterogeneity of the MSC population, resulting in unregulated MSC quality control. To overcome these limitations, we previously developed a fluorescent real-time thiol tracer (FreSHtracer) that monitors cellular levels of glutathione (GSH), which are known to be closely associated with stem cell function. In this study, we investigated whether using FreSHtracer could selectively separate high-functioning MSCs based on GSH levels and evaluated the chondrogenic potential of MSCs with high GSH levels to repair cartilage defects in vivo. METHODS Flow cytometry was conducted on FreSHtracer-loaded MSCs to select cells according to their GSH levels. To determine the function of FreSHtracer-isolated MSCs, mRNA expression, migration, and CFU assays were conducted. The MSCs underwent chondrogenic differentiation, followed by analysis of chondrogenic-related gene expression. For in vivo assessment, MSCs with different cellular GSH levels or cell culture densities were injected in a rabbit chondral defect model, followed by histological analysis of cartilage-regenerated defect sites. RESULTS FreSHtracer successfully isolated MSCs according to GSH levels. MSCs with high cellular GSH levels showed enhanced MSC function, including stem cell marker mRNA expression, migration, CFU, and oxidant resistance. Regardless of the stem cell tissue source, FreSHtracer selectively isolated MSCs with high GSH levels and high functionality. The in vitro chondrogenic potential was the highest in pellets generated by MSCs with high GSH levels, with increased ECM formation and chondrogenic marker expression. Furthermore, the MSCs' function was dependent on cell culture conditions, with relatively higher cell culture densities resulting in higher GSH levels. In vivo, improved cartilage repair was achieved by articular injection of MSCs with high levels of cellular GSH and MSCs cultured under high-density conditions, as confirmed by Collagen type 2 IHC, Safranin-O staining and O'Driscoll scores showing that more hyaline cartilage was formed on the defects. CONCLUSION FreSHtracer selectively isolates highly functional MSCs that have enhanced in vitro chondrogenesis and in vivo hyaline cartilage regeneration, which can ultimately overcome the current limitations of MSC therapy.
Collapse
Affiliation(s)
- Gun Hee Cho
- Department of Orthopedic Surgery, College of Medicine, Seoul National University, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - Hyun Cheol Bae
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - Won Young Cho
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - Eui Man Jeong
- Department of Pharmacy, College of Pharmacy, Jeju National University, Jeju Special Self-Governing Province, Jeju-do, Republic of Korea
| | - Hee Jung Park
- 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
| | - Sun Young Wang
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - You Jung Kim
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea
| | - Dong Myung Shin
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88 Olymic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - In Gyu Kim
- Laboratory for Cellular Response to Oxidative Stress, Cell2in, Inc, Seoul, 03127, Republic of Korea
| | - Hyuk-Soo Han
- Department of Orthopedic Surgery, College of Medicine, Seoul National University, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea.
- Department of Orthopedic Surgery, Seoul National University Hospital, Yongondong Chongnogu, Seoul, 110-744, Republic of Korea.
| |
Collapse
|
15
|
Zecca PA, Reguzzoni M, Protasoni M, Raspanti M. The chondro-osseous junction of articular cartilage. Tissue Cell 2023; 80:101993. [PMID: 36516570 DOI: 10.1016/j.tice.2022.101993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022]
Abstract
In the synovial joints the transition between the soft articular cartilage and the subchondral bone is mediated by a layer of calcified cartilage of structural and mechanical characteristics closer to those of bone. This layer, buried in the depth of articular cartilage, is not directly accessible and is mostly visualized in histological sections of decalcified tissue, where it appears as a darker strip in contact with the subchondral bone. In this study conventional histology and scanning electron microscopy (SEM) with secondary electron imaging (SE) or backscattered electron imaging (BSE) were used to discriminate the calcified and the uncalcified cartilage in high resolution on native, untreated tissue as well as in deproteinated or demineralized tissue. This approach evidenced a high heterogeneity of the calcified layer of articular cartilage. High resolution pictures revealed that the mineralization process originates by progressive accretion and confluence of individual, small mineral clusters, in a very different way from other hard tissues such as bone, dentin and mineralized tendons. Finally, selective removal of the soft matrix by thermal treatment allowed for the first time a face-on, unrestricted 3D view of the mineralization front.
Collapse
Affiliation(s)
| | | | - Marina Protasoni
- Department of Medicine & Surgery, Insubria University, Varese, Italy
| | - Mario Raspanti
- Department of Medicine & Surgery, Insubria University, Varese, Italy.
| |
Collapse
|
16
|
Abstract
Cartilage resides under a low oxygen tension within articulating joints. The oxygen tension within cartilage of the knee joint has been measured to be between 2% and 5% oxygen. Although the literature has historically termed this level of oxygen as hypoxia, particularly when doing experiments in vitro in this range, this is actually the physiological oxygen tension experienced in vivo and is more accurately termed physioxia. In general, culture of chondrogenic cells under physioxia has demonstrated a donor-dependent beneficial effect on chondrogenesis, with an upregulation in cartilage genes (SOX9, COL2A1, ACAN) and matrix deposition (sulfated glycosaminoglycans (sGAGs), collagen II). Physioxia also reduces the expression of hypertrophic markers (COL10A1, MMP13). This chapter will outline the methods for the expansion and differentiation of chondrogenic cells under physioxia using oxygen-controlled incubators and glove box environments, with the typical assays used for qualitative and quantitative assessment of chondrogenesis.
Collapse
Affiliation(s)
- Girish Pattappa
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center of Regensburg, Regensburg, Germany
| | - Brandon D Markway
- Department of Orthopaedics & Rehabilitation, Oregon Health & Science University, Portland, OR, USA
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center of Regensburg, Regensburg, Germany
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Wurzburg, Wurzburg, Germany
| | - Brian Johnstone
- Department of Orthopaedics & Rehabilitation, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
17
|
Xie M, Zhang Y, Xiong Z, Hines S, Shangjiang Y, Clark KL, Tan S, Alexander PG, Lin H. Generation of hyaline-like cartilage tissue from human mesenchymal stromal cells within the self-generated extracellular matrix. Acta Biomater 2022; 149:150-166. [PMID: 35779770 DOI: 10.1016/j.actbio.2022.06.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 12/20/2022]
Abstract
Chondrocytic hypertrophy, a phenotype not observed in healthy hyaline cartilage, is often concomitant with the chondrogenesis of human mesenchymal stromal cells (hMSCs). This undesired feature represents one of the major obstacles in applying hMSCs for hyaline cartilage repair. Previously, we developed a method to induce hMSC chondrogenesis within self-generated extracellular matrix (mECM), which formed a cartilage tissue with a lower hypertrophy level than conventional hMSC pellets. In this study, we aimed to test the utility of hypoxia and insulin-like growth factor-1 (IGF1) on further reducing hypertrophy. MSC-mECM constructs were first subjected to chondrogenic culture in normoxic or hypoxic (5%) conditions. The results indicated that hMSC-derived cartilage formed in hypoxic culture displayed a significantly reduced hypertrophy level than normoxic culture. However, hMSC chondrogenesis was also suppressed under hypoxic culture, partially due to the reduced activity of the IGF1 pathway. IGF1 was then supplemented in the chondrogenic medium, which promoted remarkable hMSC chondrogenesis under hypoxic culture. Interestingly, the IGF1-enhanced hMSC chondrogenesis, under hypoxic culture, was not at the expense of promoting significantly increased hypertrophy. Lastly, the cartilage tissues created by hMSCs with different conditions were implanted into osteochondral defect in rats. The results indicated that the tissue formed under hypoxic condition and induced with IGF1-supplemented chondrogenic medium displayed the best reparative results with minimal hypertrophy level. Our results demonstrate a new method to generate hyaline cartilage-like tissue from hMSCs without using exogenous scaffolds, which further pave the road for the clinical application of hMSC-based cartilage tissue engineering. STATEMENT OF SIGNIFICANCE: In this study, hyaline cartilage-like tissues were generated from human mesenchymal stromal cells (hMSCs), which displayed robust capacity in repairing the osteochondral defect in rats. In particular, the extracellular matrix created by hMSCs was used, so no exogenous scaffold was needed. Through a series of optimization, we defined that hypoxic culture and supplementation of insulin-like growth factor-1 (IGF-1) in chondrogenic medium resulted in robust cartilage formation with minimal hypertrophy. We also demonstrated that hypoxic culture suppressed chondrogenesis and hypertrophy through modulating the Wnt/β-catenin and IGF1 pathways, respectively. Our results demonstrate a new method to generate hyaline cartilage-like tissue from hMSCs without using exogenous scaffolds, which will further pave the road for the clinical application of hMSCs-based cartilage tissue engineering.
Collapse
Affiliation(s)
- Mingsheng Xie
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA; Department of Orthopaedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yiqian Zhang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA; Department of Orthopaedic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zixuan Xiong
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410008, China
| | - Sophie Hines
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA
| | - Yingzi Shangjiang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA
| | - Karen L Clark
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA
| | - Susheng Tan
- Department of Electrical and Computer Engineering, Swanson School of Engineering, and Petersen Institute of NanoScience and Engineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Peter G Alexander
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 217, Pittsburgh, PA 15217, 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, Pittsburgh, PA 15219, USA.
| |
Collapse
|
18
|
Abdelrahman SA, Abdelrahman AA, Samy W, Dessouky AA, Ahmed SM. Hypoxia pretreatment enhances the therapeutic potential of mesenchymal stem cells (BMSCs) on ozone-induced lung injury in rats. Cell Tissue Res 2022; 389:201-217. [PMID: 35551479 PMCID: PMC9287250 DOI: 10.1007/s00441-022-03627-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 04/16/2022] [Indexed: 11/26/2022]
Abstract
Ozone (O3) gas is a double-sided weapon. It provides a shield that protects life on earth from the harmful ultraviolet (UV) rays, but ground-level O3 is considered an urban air pollutant. So, a rat model of chronic O3 inhalation was established to assess the biochemical and morphological alterations in the lung tissue and to investigate the ameliorative effects of bone marrow-derived mesenchymal stem cells (BMSCs) with or without hypoxia pre-treatment. Forty-two adult male albino rats were divided into four groups: control, ozone-exposed, normoxic BMSC-treated, and hypoxic BMSC-treated groups. Lung tissue sections were processed for light and electron microscope examination, immunohistochemical staining for caspase 3, and iNOS. Quantitative real-time PCR for IL-1α, IL-17, TNF-α, and Nrf2 mRNA gene expression were also performed. Chronic O3 exposure caused elevated inflammatory cytokines and decreased antioxidant Nrf2 mRNA expression. Marked morphological alterations with increased collagen deposition and elevated apoptotic markers and iNOS were evident. BMSC treatment showed immunomodulatory (decreased inflammatory cytokine gene expression), antioxidant (increased Nrf2 expression and decreased iNOS), and anti-apoptotic (decreased caspase3 expression) effects. Consequently, ameliorated lung morphology with diminished collagen deposition was observed. Hypoxia pretreatment enhanced BMSC survival by MTT assay. It also augmented the previously mentioned effects of BMSCs on the lung tissue as proved by statistical analysis. Lung morphology was similar to that of control group. In conclusion, hypoxia pretreatment represents a valuable intervention to enhance the effects of MSCs on chronic lung injury.
Collapse
Affiliation(s)
- Shaimaa A Abdelrahman
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Abeer A Abdelrahman
- Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Walaa Samy
- Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Arigue A Dessouky
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Samah M Ahmed
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
19
|
Mohd Yunus MH, Lee Y, Nordin A, Chua KH, Bt Hj Idrus R. Remodeling Osteoarthritic Articular Cartilage under Hypoxic Conditions. Int J Mol Sci 2022; 23:ijms23105356. [PMID: 35628163 PMCID: PMC9141680 DOI: 10.3390/ijms23105356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/07/2022] [Accepted: 05/08/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is one of the leading joint diseases induced by abnormalities or inflammation in the synovial membrane and articular cartilage, causing severe pain and disability. Along with the cartilage malfunction, imbalanced oxygen uptake occurs, changing chondrocytes into type I collagen- and type X collagen-producing dedifferentiated cells, contributing to OA progression. However, mounting evidence suggests treating OA by inducing a hypoxic environment in the articular cartilage, targeting the inhibition of several OA-related pathways to bring chondrocytes into a normal state. This review discusses the implications of OA-diseased articular cartilage on chondrocyte phenotypes and turnover and debates the hypoxic mechanism of action. Furthermore, this review highlights the new understanding of OA, provided by tissue engineering and a regenerative medicine experimental design, modeling the disease into diverse 2D and 3D structures and investigating hypoxia and hypoxia-inducing biomolecules and potential cell therapies. This review also reports the mechanism of hypoxic regulation and highlights the importance of activating and stabilizing the hypoxia-inducible factor and related molecules to protect chondrocytes from mitochondrial dysfunction and apoptosis occurring under the influence of OA.
Collapse
Affiliation(s)
- Mohd Heikal Mohd Yunus
- Department of Physiology, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (K.H.C.); (R.B.H.I.)
- Correspondence: ; Tel.: +603-9145-8624
| | - Yemin Lee
- MedCentral Consulting, Jalan 27/117A, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (A.N.)
| | - Abid Nordin
- MedCentral Consulting, Jalan 27/117A, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (A.N.)
| | - Kien Hui Chua
- Department of Physiology, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (K.H.C.); (R.B.H.I.)
| | - Ruszymah Bt Hj Idrus
- Department of Physiology, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (K.H.C.); (R.B.H.I.)
| |
Collapse
|
20
|
Falcon JM, Chirman D, Veneziale A, Morman J, Bolten K, Kandel S, Querido W, Freeman T, Pleshko N. DMOG Negatively Impacts Tissue Engineered Cartilage Development. Cartilage 2021; 13:722S-733S. [PMID: 33100027 PMCID: PMC8804718 DOI: 10.1177/1947603520967060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Articular cartilage exists in a hypoxic environment, which motivates the use of hypoxia-simulating chemical agents to improve matrix production in cartilage tissue engineering. The aim of this study was to investigate whether dimethyloxalylglycine (DMOG), a HIF-1α stabilizer, would improve matrix production in 3-dimensional (3D) porcine synovial-derived mesenchymal stem cell (SYN-MSC) co-culture with chondrocytes. DESIGN Pellet cultures and scaffold-based engineered cartilage were grown in vitro to determine the impact of chemically simulated hypoxia on 2 types of 3D cell culture. DMOG-treated groups were exposed to DMOG from day 14 to day 21 and grown up to 6 weeks with n = 3 per condition and time point. RESULTS The addition of DMOG resulted in HIF-1α stabilization in the exterior of the engineered constructs, which resulted in increased regional type II collagen deposition, but the stabilization did not translate to overall increased extracellular matrix deposition. There was no increase in HIF-1α stabilization in the pellet cultures. DMOG treatment also negatively affected the mechanical competency of the engineered cartilage. CONCLUSIONS Despite previous studies that demonstrated the efficacy of DMOG, here, short-term treatment with DMOG did not have a uniformly positive impact on the chondrogenic capacity of SYN-MSCs in either pellet culture or in scaffold-based engineered cartilage, as evidenced by reduced matrix production. Such 3D constructs generally have a naturally occurring hypoxic center, which allows for the stabilization of HIF-1α in the interior tissue. Thus, short-term addition of DMOG may not further improve this in cartilage tissue engineered constructs.
Collapse
Affiliation(s)
- Jessica M. Falcon
- Tissue Imaging and Spectroscopy Lab,
Department of Bioengineering, Temple University, Philadelphia, PA, USA
| | - Dylan Chirman
- Tissue Imaging and Spectroscopy Lab,
Department of Bioengineering, Temple University, Philadelphia, PA, USA
| | - Alyssa Veneziale
- Tissue Imaging and Spectroscopy Lab,
Department of Bioengineering, Temple University, Philadelphia, PA, USA
| | - Justin Morman
- Tissue Imaging and Spectroscopy Lab,
Department of Bioengineering, Temple University, Philadelphia, PA, USA
| | - Katherine Bolten
- Tissue Imaging and Spectroscopy Lab,
Department of Bioengineering, Temple University, Philadelphia, PA, USA
| | - Shital Kandel
- Tissue Imaging and Spectroscopy Lab,
Department of Bioengineering, Temple University, Philadelphia, PA, USA
| | - William Querido
- Tissue Imaging and Spectroscopy Lab,
Department of Bioengineering, Temple University, Philadelphia, PA, USA
| | - Theresa Freeman
- Division of Orthopaedic Research, Thomas
Jefferson University, Philadelphia, PA, USA
| | - Nancy Pleshko
- Tissue Imaging and Spectroscopy Lab,
Department of Bioengineering, Temple University, Philadelphia, PA, USA,Nancy Pleshko, Department of Bioengineering,
Temple University, 1947 N. 12th St., Philadelphia, PA 19122, USA.
| |
Collapse
|
21
|
Ge D, O'Brien MJ, Savoie FH, Gimble JM, Wu X, Gilbert MH, Clark-Patterson GL, Schuster JD, Miller KS, Wang A, Myers L, You Z. Human adipose-derived stromal/stem cells expressing doublecortin improve cartilage repair in rabbits and monkeys. NPJ Regen Med 2021; 6:82. [PMID: 34848747 PMCID: PMC8633050 DOI: 10.1038/s41536-021-00192-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 10/29/2021] [Indexed: 11/25/2022] Open
Abstract
Localized cartilage lesions in early osteoarthritis and acute joint injuries are usually treated surgically to restore function and relieve pain. However, a persistent clinical challenge remains in how to repair the cartilage lesions. We expressed doublecortin (DCX) in human adipose-derived stromal/stem cells (hASCs) and engineered hASCs into cartilage tissues using an in vitro 96-well pellet culture system. The cartilage tissue constructs with and without DCX expression were implanted in the knee cartilage defects of rabbits (n = 42) and monkeys (n = 12). Cohorts of animals were euthanized at 6, 12, and 24 months after surgery to evaluate the cartilage repair outcomes. We found that DCX expression in hASCs increased expression of growth differentiation factor 5 (GDF5) and matrilin 2 in the engineered cartilage tissues. The cartilage tissues with DCX expression significantly enhanced cartilage repair as assessed macroscopically and histologically at 6, 12, and 24 months after implantation in the rabbits and 24 months after implantation in the monkeys, compared to the cartilage tissues without DCX expression. These findings suggest that hASCs expressing DCX may be engineered into cartilage tissues that can be used to treat localized cartilage lesions.
Collapse
Affiliation(s)
- Dongxia Ge
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Michael J O'Brien
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Felix H Savoie
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jeffrey M Gimble
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- LaCell LLC and Obatala Sciences Inc., New Orleans, LA, USA
- Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Xiying Wu
- LaCell LLC and Obatala Sciences Inc., New Orleans, LA, USA
| | - Margaret H Gilbert
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | | | - Jason D Schuster
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Kristin S Miller
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Alun Wang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Leann Myers
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropic Medicine, New Orleans, LA, USA
| | - Zongbing You
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA.
- Department of Orthopaedic Surgery, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Cancer Center and Louisiana Cancer Research Consortium, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA, USA.
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA.
| |
Collapse
|
22
|
Shestovskaya MV, Bozhkova SA, Sopova JV, Khotin MG, Bozhokin MS. Methods of Modification of Mesenchymal Stem Cells and Conditions of Their Culturing for Hyaline Cartilage Tissue Engineering. Biomedicines 2021; 9:biomedicines9111666. [PMID: 34829895 PMCID: PMC8615732 DOI: 10.3390/biomedicines9111666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
The use of mesenchymal stromal cells (MSCs) for tissue engineering of hyaline cartilage is a topical area of regenerative medicine that has already entered clinical practice. The key stage of this procedure is to create conditions for chondrogenic differentiation of MSCs, increase the synthesis of hyaline cartilage extracellular matrix proteins by these cells and activate their proliferation. The first such works consisted in the indirect modification of cells, namely, in changing the conditions in which they are located, including microfracturing of the subchondral bone and the use of 3D biodegradable scaffolds. The most effective methods for modifying the cell culture of MSCs are protein and physical, which have already been partially introduced into clinical practice. Genetic methods for modifying MSCs, despite their effectiveness, have significant limitations. Techniques have not yet been developed that allow studying the effectiveness of their application even in limited groups of patients. The use of MSC modification methods allows precise regulation of cell culture proliferation, and in combination with the use of a 3D biodegradable scaffold, it allows obtaining a hyaline-like regenerate in the damaged area. This review is devoted to the consideration and comparison of various methods used to modify the cell culture of MSCs for their use in regenerative medicine of cartilage tissue.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Svetlana A. Bozhkova
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
| | - Julia V. Sopova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Mikhail G. Khotin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Mikhail S. Bozhokin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
- Correspondence:
| |
Collapse
|
23
|
Foo JB, Looi QH, How CW, Lee SH, Al-Masawa ME, Chong PP, Law JX. Mesenchymal Stem Cell-Derived Exosomes and MicroRNAs in Cartilage Regeneration: Biogenesis, Efficacy, miRNA Enrichment and Delivery. Pharmaceuticals (Basel) 2021; 14:1093. [PMID: 34832875 PMCID: PMC8618513 DOI: 10.3390/ph14111093] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes are the small extracellular vesicles secreted by cells for intercellular communication. Exosomes are rich in therapeutic cargos such as microRNA (miRNA), long non-coding RNA (lncRNA), small interfering RNA (siRNA), DNA, protein, and lipids. Recently, many studies have focused on miRNAs as a promising therapeutic factor to support cartilage regeneration. Exosomes are known to contain a substantial amount of a variety of miRNAs. miRNAs regulate the post-transcriptional gene expression by base-pairing with the target messenger RNA (mRNA), leading to gene silencing. Several exosomal miRNAs have been found to play a role in cartilage regeneration by promoting chondrocyte proliferation and matrix secretion, reducing scar tissue formation, and subsiding inflammation. The exosomal miRNA cargo can be modulated using techniques such as cell transfection and priming as well as post-secretion modifications to upregulate specific miRNAs to enhance the therapeutic effect. Exosomes are delivered to the joints through direct injection or via encapsulation within a scaffold for sustained release. To date, exosome therapy for cartilage injuries has yet to be optimized as the ideal cell source for exosomes, and the dose and method of delivery have yet to be identified. More importantly, a deeper understanding of the role of exosomal miRNAs in cartilage repair is paramount for the development of more effective exosome therapy.
Collapse
Affiliation(s)
- Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
- Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Qi Hao Looi
- My Cytohealth Sdn. Bhd., D353a, Menara Suezcap 1, KL Gateway, no. 2, Jalan Kerinchi, Gerbang Kerinchi Lestari, Kuala Lumpur 59200, Malaysia;
- National Orthopaedic Centre of Excellence in Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Bandar Sunway 47500, Malaysia;
| | - Sau Har Lee
- Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Maimonah Eissa Al-Masawa
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia;
| | - Pei Pei Chong
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
24
|
Rhatomy S, Setyawan R, Romulo MA. Enhancement of Chondrogenesis in Hypoxic Precondition Culture: A Systematic Review. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.5850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Cartilage tear has begun to be treated with stem cells. However, stem cell oxygen level culture has not been evaluated for the best environment to enhance chondrogenesis.
AIM: The purpose of this review is to focus on the hypoxic oxygen level of stem cells culture as a treatment for cartilage tear.
METHODS: A literature search was systemically conducted on PubMed (MEDLINE), OVID, EMBASE, the Cochrane Library, Scopus, Web of Science, Science Direct, Wiley Online Library, Google Scholar, and bibliography of selected articles with the terms (“culture”) AND (“stem cell” OR “mesenchymal stem cell” OR “MSC”) AND (“hypoxic” OR “hypoxia”) AND (“cartilage” OR “chondro*”) as the main keywords. A total of 438 articles were reviewed. Thirty-six articles were considered relevant for this systematic review.
RESULTS: The result of this review supports stimulation effects of hypoxic oxygen level stem cell culture in chondrogenesis process. Most studies used 5% oxygen concentration for culture, both of in vivo and in vitro studies. Due to the heterogeneity nature of the included studies, meta-analysis was unable to be conducted.
CONCLUSION: Hypoxia state seems to play an important role in chondrocytes proliferation, differentiation, and matrix production.
Collapse
|
25
|
Monaco G, Ladner YD, El Haj AJ, Forsyth NR, Alini M, Stoddart MJ. Mesenchymal Stromal Cell Differentiation for Generating Cartilage and Bone-Like Tissues In Vitro. Cells 2021; 10:cells10082165. [PMID: 34440934 PMCID: PMC8391162 DOI: 10.3390/cells10082165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/12/2021] [Accepted: 08/18/2021] [Indexed: 01/22/2023] Open
Abstract
In the field of tissue engineering, progress has been made towards the development of new treatments for cartilage and bone defects. However, in vitro culture conditions for human bone marrow mesenchymal stromal cells (hBMSCs) have not yet been fully defined. To improve our understanding of cartilage and bone in vitro differentiation, we investigated the effect of culture conditions on hBMSC differentiation. We hypothesized that the use of two different culture media including specific growth factors, TGFβ1 or BMP2, as well as low (2% O2) or high (20% O2) oxygen tension, would improve the chondrogenic and osteogenic potential, respectively. Chondrogenic and osteogenic differentiation of hBMSCs isolated from multiple donors and expanded under the same conditions were directly compared. Chondrogenic groups showed a notable upregulation of chondrogenic markers compared with osteogenic groups. Greater sGAG production and deposition, and collagen type II and I accumulation occurred for chondrogenic groups. Chondrogenesis at 2% O2 significantly reduced ALP gene expression and reduced type I collagen deposition, producing a more stable and less hypertrophic chondrogenic phenotype. An O2 tension of 2% did not inhibit osteogenic differentiation at the protein level but reduced ALP and OC gene expression. An upregulation of ALP and OC occurred during osteogenesis in BMP2 containing media under 20% O2; BMP2 free osteogenic media downregulated ALP and also led to higher sGAG release. A higher mineralization was observed in the presence of BMP2 during osteogenesis. This study demonstrates how the modulation of O2 tension, combined with tissue-specific growth factors and media composition can be tailored in vitro to promote chondral or endochondral differentiation while using the same donor cell population.
Collapse
Affiliation(s)
- Graziana Monaco
- AO Research Institute Davos, Regenerative Orthopaedics Program, 7270 Davos Platz, Switzerland; (G.M.); (Y.D.L.); (M.A.)
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire ST4 7QB, UK;
| | - Yann D. Ladner
- AO Research Institute Davos, Regenerative Orthopaedics Program, 7270 Davos Platz, Switzerland; (G.M.); (Y.D.L.); (M.A.)
- Institute for Biomechanics, ETH Zurich, Lengghalde 5, CH-8008 Zurich, Switzerland
| | - Alicia J. El Haj
- Healthcare Technology Institute, Institute of Translational Medicine, University of Birmingham, Birmingham B15 2TT, UK;
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire ST4 7QB, UK;
| | - Mauro Alini
- AO Research Institute Davos, Regenerative Orthopaedics Program, 7270 Davos Platz, Switzerland; (G.M.); (Y.D.L.); (M.A.)
| | - Martin J. Stoddart
- AO Research Institute Davos, Regenerative Orthopaedics Program, 7270 Davos Platz, Switzerland; (G.M.); (Y.D.L.); (M.A.)
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire ST4 7QB, UK;
- Correspondence:
| |
Collapse
|
26
|
Characterization of Scleraxis and SRY-Box 9 from Adipose-Derived Stem Cells Culture Seeded with Enthesis Scaffold in Hypoxic Condition. JOURNAL OF BIOMIMETICS BIOMATERIALS AND BIOMEDICAL ENGINEERING 2021. [DOI: 10.4028/www.scientific.net/jbbbe.52.76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The use of mesenchymal stem cells can add local improvements potential to enthesis tissue regeneration based on tropical activity through secretions of growth factors, cytokines, and vesicles (e.g. exosomes), collectively known as secretomes. This study aims to analyze secretomes characterization from adipose-derived mesenchymal stem cells seeded with enthesis tissue scaffold in hypoxic conditions and to analyze the influence of hypoxic environment to the characterization of secretomes. This is an in-vitro study using a Randomized Control Group Post-Test Only design. This study using Adipose Stem Cells (ASCs) were cultured in hypoxia (Oxygen 5%) and Normoxia (21%) condition. The scaffolds are fresh-frozen enthesis tissue and was seeded in the treatment group and compared to control. The evaluation of Scleraxis (Scx) and SRY-box (Sox9) was measured using ELISA on the 2nd, 4th, and 6th days. Comparison of Scx levels between each evaluation time showed a positive trend in a group with scaffold in hypoxia condition although it has no significant differences (p=0.085), with the highest level on day 6, that is 13,568 ng/ml. Conversely, the comparison of Sox9 showed significant differences (p=0.02) in a group with scaffold in hypoxia condition, with the highest level on day 4, that is 28,250 ng/ml. The use of enthesis scaffold seeded in adipose-derived mesenchymal stem cells in hypoxic conditions shows a positive trend as regenerative effort of injured enthesis tissue through Scleraxis and Sox9 secretomes induction.
Collapse
|
27
|
Huang Y, Fan H, Gong X, Yang L, Wang F. Scaffold With Natural Calcified Cartilage Zone for Osteochondral Defect Repair in Minipigs. Am J Sports Med 2021; 49:1883-1891. [PMID: 33961510 DOI: 10.1177/03635465211007139] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Long-term outcomes of current clinical interventions for osteochondral defect are less than satisfactory. One possible reason is an ignorance of the interface structure between cartilage and subchondral bone, the calcified cartilage zone (CCZ). However, the importance of natural CCZ in osteochondral defects has not been directly described. PURPOSE To explore the feasibility of fabricating trilayer scaffold containing natural CCZ for osteochondral defects and the role of CCZ in the repair process. STUDY DESIGN Controlled laboratory study. METHODS The scaffold was prepared by cross-linking lyophilized type II collagen sponge and acellular normal pig subchondral bone with or without natural CCZ. Autologous bone marrow stem cells (BMSCs) of minipig were mixed with type II collagen gel and injected into the cartilage layer of the scaffold before operation. Thirty minipigs were randomly divided into CCZ (n = 10), non-CCZ (n = 10), and blank control (n = 10) groups. An 8 mm-diameter full-thickness osteochondral defect was created on the trochlear surface, and scaffold containing BMSCs was transplanted into the defect according to grouping requirements. At 12 and 24 weeks postoperatively, specimens were assessed by macroscopic observation, magnetic resonance imaging examination, and histological observations (hematoxylin and eosin, Safranin O-fast green, type II collagen immunohistochemical, and Sirius red staining). Semiquantitative cartilage repair scoring was conducted using the MOCART (Magnetic Resonance Observation of Cartilage Repair Tissue) system and the O'Driscoll repaired cartilage value system. RESULTS The defects in the blank control and non-CCZ groups were filled with fibrous tissue, while the cartilage layer of the CCZ group was mainly repaired by hyaline cartilage at 24 weeks postoperatively. The superior repair outcome of the CCZ group was confirmed by MOCART and O'Driscoll score. CONCLUSION The trilayer scaffold containing natural CCZ obtained the best repair effect compared with the non-CCZ scaffold and the blank control, indicating the importance of the CCZ in osteochondral tissue engineering. CLINICAL RELEVANCE This study demonstrates the necessity to reconstruct CCZ in clinical osteochondral defect repair and provides a possible strategy for osteochondral tissue engineering.
Collapse
Affiliation(s)
- Yang Huang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Huaquan Fan
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaoyuan Gong
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Liu Yang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fuyou Wang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
28
|
Zhou X, Xu W, Wang Y, Zhang H, Zhang L, Li C, Yao S, Huang Z, Huang L, Luo D. LncRNA DNM3OS regulates GREM2 via miR-127-5p to suppress early chondrogenic differentiation of rat mesenchymal stem cells under hypoxic conditions. Cell Mol Biol Lett 2021; 26:22. [PMID: 34049478 PMCID: PMC8161583 DOI: 10.1186/s11658-021-00269-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background Improved chondrogenic differentiation of mesenchymal stem cells (MSCs) by genetic regulation is a potential method for regenerating articular cartilage. MiR-127-5p has been reported to promote cartilage differentiation of rat bone marrow MSCs (rMSCs); however, the regulatory mechanisms underlying hypoxia-stimulated chondrogenic differentiation remain unknown. Methods rMSCs were induced to undergo chondrogenic differentiation under normoxic or hypoxic conditions. Expression of lncRNA DNM3OS, miR-127-5p, and GREM2 was detected by quantitative real-time PCR. Proteoglycans were detected by Alcian blue staining. Western blot assays were performed to examine the relative levels of GREM2 and chondrogenic differentiation related proteins. Luciferase reporter assays were performed to assess the association among DNM3OS, miR-127-5p, and GREM2. Results MiR-127-5p levels were upregulated, while DNM3OS and GREM2 levels were downregulated in rMSCs induced to undergo chondrogenic differentiation, and those changes were attenuated by hypoxic conditions (1% O2). Further in vitro experiments revealed that downregulation of miR-127-5p reduced the production of proteoglycans and expression of chondrogenic differentiation markers (COL1A1, COL2A1, SOX9, and ACAN) and osteo/chondrogenic markers (BMP-2, p-SMAD1/2). MiR-127-5p overexpression produced the opposite results in rMSCs induced to undergo chondrogenic differentiation under hypoxic conditions. GREM2 was found to be a direct target of miR-127-5p, which was suppressed in rMSCs undergoing chondrogenic differentiation. Moreover, DNM3OS could directly bind to miR-127-5p and inhibit chondrogenic differentiation of rMSCs via regulating GREM2. Conclusions Our study revealed a novel molecular pathway (DNM3OS/miR-127-5p/GREM2) that may be involved in hypoxic chondrogenic differentiation.
Collapse
Affiliation(s)
- Xiaozhong Zhou
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wangyang Xu
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Yeyang Wang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Hui Zhang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Li Zhang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Chao Li
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Shun Yao
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Zixiang Huang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Lishan Huang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Dixin Luo
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.
| |
Collapse
|
29
|
Extracellular Vesicles from Mesenchymal Stem Cells as Potential Treatments for Osteoarthritis. Cells 2021; 10:cells10061287. [PMID: 34067325 PMCID: PMC8224601 DOI: 10.3390/cells10061287] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/12/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disorder of the joint and its prevalence and severity is increasing owing to ageing of the population. Osteoarthritis is characterized by the degradation of articular cartilage and remodeling of the underlying bone. There is little understanding of the cellular and molecular processes involved in pathophysiology of OA. Currently the treatment for OA is limited to painkillers and anti-inflammatory drugs, which only treat the symptoms. Some patients may also undergo surgical procedures to replace the damaged joints. Extracellular vesicles (EV) play an important role in intercellular communications and their concentration is elevated in the joints of OA patients, although their mechanism is unclear. Extracellular vesicles are naturally released by cells and they carry their origin cell information to be delivered to target cells. On the other hand, mesenchymal stem cells (MSCs) are highly proliferative and have a great potential in cartilage regeneration. In this review, we provide an overview of the current OA treatments and their limitations. We also discuss the role of EV in OA pathophysiology. Finally, we highlight the therapeutic potential of MSC-derived EV in OA and their challenges.
Collapse
|
30
|
Tomecka E, Lech W, Zychowicz M, Sarnowska A, Murzyn M, Oldak T, Domanska-Janik K, Buzanska L, Rozwadowska N. Assessment of the Neuroprotective and Stemness Properties of Human Wharton's Jelly-Derived Mesenchymal Stem Cells under Variable (5% vs. 21%) Aerobic Conditions. Cells 2021; 10:717. [PMID: 33804841 PMCID: PMC8063843 DOI: 10.3390/cells10040717] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/20/2021] [Accepted: 03/21/2021] [Indexed: 12/20/2022] Open
Abstract
To optimise the culture conditions for human Wharton's jelly-derived mesenchymal stem cells (hWJ-MSCs) intended for clinical use, we investigated ten different properties of these cells cultured under 21% (atmospheric) and 5% (physiological normoxia) oxygen concentrations. The obtained results indicate that 5% O2 has beneficial effects on the proliferation rate, clonogenicity, and slowdown of senescence of hWJ-MSCs; however, the oxygen level did not have an influence on the cell morphology, immunophenotype, or neuroprotective effect of the hWJ-MSCs. Nonetheless, the potential to differentiate into adipocytes, osteocytes, and chondrocytes was comparable under both oxygen conditions. However, spontaneous differentiation of hWJ-MSCs into neuronal lineages was observed and enhanced under atmospheric oxygen conditions. The cells relied more on mitochondrial respiration than glycolysis, regardless of the oxygen conditions. Based on these results, we can conclude that hWJ-MSCs could be effectively cultured and prepared under both oxygen conditions for cell-based therapy. However, the 5% oxygen level seemed to create a more balanced and appropriate environment for hWJ-MSCs.
Collapse
Affiliation(s)
- Ewelina Tomecka
- Polish Stem Cell Bank, FamiCord Group, 00-867 Warsaw, Poland; (E.T.); (M.M.); (T.O.)
| | - Wioletta Lech
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.L.); (M.Z.); (A.S.); (K.D.-J.)
| | - Marzena Zychowicz
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.L.); (M.Z.); (A.S.); (K.D.-J.)
| | - Anna Sarnowska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.L.); (M.Z.); (A.S.); (K.D.-J.)
| | - Magdalena Murzyn
- Polish Stem Cell Bank, FamiCord Group, 00-867 Warsaw, Poland; (E.T.); (M.M.); (T.O.)
| | - Tomasz Oldak
- Polish Stem Cell Bank, FamiCord Group, 00-867 Warsaw, Poland; (E.T.); (M.M.); (T.O.)
| | - Krystyna Domanska-Janik
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.L.); (M.Z.); (A.S.); (K.D.-J.)
| | - Leonora Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (W.L.); (M.Z.); (A.S.); (K.D.-J.)
| | - Natalia Rozwadowska
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland;
| |
Collapse
|
31
|
Thorp H, Kim K, Kondo M, Maak T, Grainger DW, Okano T. Trends in Articular Cartilage Tissue Engineering: 3D Mesenchymal Stem Cell Sheets as Candidates for Engineered Hyaline-Like Cartilage. Cells 2021; 10:cells10030643. [PMID: 33805764 PMCID: PMC7998529 DOI: 10.3390/cells10030643] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Articular cartilage defects represent an inciting factor for future osteoarthritis (OA) and degenerative joint disease progression. Despite multiple clinically available therapies that succeed in providing short term pain reduction and restoration of limited mobility, current treatments do not reliably regenerate native hyaline cartilage or halt cartilage degeneration at these defect sites. Novel therapeutics aimed at addressing limitations of current clinical cartilage regeneration therapies increasingly focus on allogeneic cells, specifically mesenchymal stem cells (MSCs), as potent, banked, and available cell sources that express chondrogenic lineage commitment capabilities. Innovative tissue engineering approaches employing allogeneic MSCs aim to develop three-dimensional (3D), chondrogenically differentiated constructs for direct and immediate replacement of hyaline cartilage, improve local site tissue integration, and optimize treatment outcomes. Among emerging tissue engineering technologies, advancements in cell sheet tissue engineering offer promising capabilities for achieving both in vitro hyaline-like differentiation and effective transplantation, based on controlled 3D cellular interactions and retained cellular adhesion molecules. This review focuses on 3D MSC-based tissue engineering approaches for fabricating “ready-to-use” hyaline-like cartilage constructs for future rapid in vivo regenerative cartilage therapies. We highlight current approaches and future directions regarding development of MSC-derived cartilage therapies, emphasizing cell sheet tissue engineering, with specific focus on regulating 3D cellular interactions for controlled chondrogenic differentiation and post-differentiation transplantation capabilities.
Collapse
Affiliation(s)
- Hallie Thorp
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Department of Biomedical Engineering, University of Utah, 36 S Wasatch Dr, Salt Lake City, UT 84112, USA
| | - Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Correspondence: (K.K.); (T.O.); Tel.: +1-801-585-0070 (K.K. & T.O.); Fax: +1-801-581-3674 (K.K. & T.O.)
| | - Makoto Kondo
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
| | - Travis Maak
- Department of Orthopaedic Surgery, University of Utah, 590 Wakara Way, Salt Lake City, UT 84108, USA;
| | - David W. Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Department of Biomedical Engineering, University of Utah, 36 S Wasatch Dr, Salt Lake City, UT 84112, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Wakamatsucho, 2−2, Shinjuku-ku, Tokyo 162-8480, Japan
- Correspondence: (K.K.); (T.O.); Tel.: +1-801-585-0070 (K.K. & T.O.); Fax: +1-801-581-3674 (K.K. & T.O.)
| |
Collapse
|
32
|
Abstract
BACKGROUND In spite of advances in the treatment of cartilage defects using cell and scaffold-based therapeutic strategies, the long-term outcome is still not satisfying since clinical scores decline years after treatment. Scaffold materials currently used in clinical settings have shown limitations in providing suitable biomechanical properties and an authentic and protective environment for regenerative cells. To tackle this problem, we developed a scaffold material based on decellularised human articular cartilage. METHODS Human articular cartilage matrix was engraved using a CO2 laser and treated for decellularisation and glycosaminoglycan removal. Characterisation of the resulting scaffold was performed via mechanical testing, DNA and GAG quantification and in vitro cultivation with adipose-derived stromal cells (ASC). Cell vitality, adhesion and chondrogenic differentiation were assessed. An ectopic, unloaded mouse model was used for the assessment of the in vivo performance of the scaffold in combination with ASC and human as well as bovine chondrocytes. The novel scaffold was compared to a commercial collagen type I/III scaffold. FINDINGS Crossed line engravings of the matrix allowed for a most regular and ubiquitous distribution of cells and chemical as well as enzymatic matrix treatment was performed to increase cell adhesion. The biomechanical characteristics of this novel scaffold that we term CartiScaff were found to be superior to those of commercially available materials. Neo-tissue was integrated excellently into the scaffold matrix and new collagen fibres were guided by the laser incisions towards a vertical alignment, a typical feature of native cartilage important for nutrition and biomechanics. In an ectopic, unloaded in vivo model, chondrocytes and mesenchymal stromal cells differentiated within the incisions despite the lack of growth factors and load, indicating a strong chondrogenic microenvironment within the scaffold incisions. Cells, most noticeably bone marrow-derived cells, were able to repopulate the empty chondrocyte lacunae inside the scaffold matrix. INTERPRETATION Due to the better load-bearing, its chondrogenic effect and the ability to guide matrix-deposition, CartiScaff is a promising biomaterial to accelerate rehabilitation and to improve long term clinical success of cartilage defect treatment. FUNDING Austrian Research Promotion Agency FFG ("CartiScaff" #842455), Lorenz Böhler Fonds (16/13), City of Vienna Competence Team Project Signaltissue (MA23, #18-08).
Collapse
|
33
|
Repopulation of decellularised articular cartilage by laser-based matrix engraving. EBioMedicine 2021; 64:103196. [PMID: 33483297 PMCID: PMC7910698 DOI: 10.1016/j.ebiom.2020.103196] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/25/2020] [Accepted: 12/15/2020] [Indexed: 12/28/2022] Open
Abstract
Background In spite of advances in the treatment of cartilage defects using cell and scaffold-based therapeutic strategies, the long-term outcome is still not satisfying since clinical scores decline years after treatment. Scaffold materials currently used in clinical settings have shown limitations in providing suitable biomechanical properties and an authentic and protective environment for regenerative cells. To tackle this problem, we developed a scaffold material based on decellularised human articular cartilage. Methods Human articular cartilage matrix was engraved using a CO2 laser and treated for decellularisation and glycosaminoglycan removal. Characterisation of the resulting scaffold was performed via mechanical testing, DNA and GAG quantification and in vitro cultivation with adipose-derived stromal cells (ASC). Cell vitality, adhesion and chondrogenic differentiation were assessed. An ectopic, unloaded mouse model was used for the assessment of the in vivo performance of the scaffold in combination with ASC and human as well as bovine chondrocytes. The novel scaffold was compared to a commercial collagen type I/III scaffold. Findings Crossed line engravings of the matrix allowed for a most regular and ubiquitous distribution of cells and chemical as well as enzymatic matrix treatment was performed to increase cell adhesion. The biomechanical characteristics of this novel scaffold that we term CartiScaff were found to be superior to those of commercially available materials. Neo-tissue was integrated excellently into the scaffold matrix and new collagen fibres were guided by the laser incisions towards a vertical alignment, a typical feature of native cartilage important for nutrition and biomechanics. In an ectopic, unloaded in vivo model, chondrocytes and mesenchymal stromal cells differentiated within the incisions despite the lack of growth factors and load, indicating a strong chondrogenic microenvironment within the scaffold incisions. Cells, most noticeably bone marrow-derived cells, were able to repopulate the empty chondrocyte lacunae inside the scaffold matrix. Interpretation Due to the better load-bearing, its chondrogenic effect and the ability to guide matrix-deposition, CartiScaff is a promising biomaterial to accelerate rehabilitation and to improve long term clinical success of cartilage defect treatment. Funding Austrian Research Promotion Agency FFG (“CartiScaff” #842455), Lorenz Böhler Fonds (16/13), City of Vienna Competence Team Project Signaltissue (MA23, #18-08)
Collapse
|
34
|
Schmidt S, Abinzano F, Mensinga A, Teßmar J, Groll J, Malda J, Levato R, Blunk T. Differential Production of Cartilage ECM in 3D Agarose Constructs by Equine Articular Cartilage Progenitor Cells and Mesenchymal Stromal Cells. Int J Mol Sci 2020; 21:ijms21197071. [PMID: 32992847 PMCID: PMC7582568 DOI: 10.3390/ijms21197071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/23/2022] Open
Abstract
Identification of articular cartilage progenitor cells (ACPCs) has opened up new opportunities for cartilage repair. These cells may be used as alternatives for or in combination with mesenchymal stromal cells (MSCs) in cartilage engineering. However, their potential needs to be further investigated, since only a few studies have compared ACPCs and MSCs when cultured in hydrogels. Therefore, in this study, we compared chondrogenic differentiation of equine ACPCs and MSCs in agarose constructs as monocultures and as zonally layered co-cultures under both normoxic and hypoxic conditions. ACPCs and MSCs exhibited distinctly differential production of the cartilaginous extracellular matrix (ECM). For ACPC constructs, markedly higher glycosaminoglycan (GAG) contents were determined by histological and quantitative biochemical evaluation, both in normoxia and hypoxia. Differential GAG production was also reflected in layered co-culture constructs. For both cell types, similar staining for type II collagen was detected. However, distinctly weaker staining for undesired type I collagen was observed in the ACPC constructs. For ACPCs, only very low alkaline phosphatase (ALP) activity, a marker of terminal differentiation, was determined, in stark contrast to what was found for MSCs. This study underscores the potential of ACPCs as a promising cell source for cartilage engineering.
Collapse
Affiliation(s)
- Stefanie Schmidt
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Germany;
| | - Florencia Abinzano
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
| | - Anneloes Mensinga
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
| | - Jörg Teßmar
- Department for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany; (J.T.); (J.G.)
| | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany; (J.T.); (J.G.)
| | - Jos Malda
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
| | - Riccardo Levato
- Department of Orthopedics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (F.A.); (A.M.); (J.M.)
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
- Correspondence: (R.L.); (T.B.)
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Germany;
- Correspondence: (R.L.); (T.B.)
| |
Collapse
|
35
|
Bahir B, S. Choudhery M, Hussain I. Hypoxic Preconditioning as a Strategy to Maintain the Regenerative Potential of Mesenchymal Stem Cells. Regen Med 2020. [DOI: 10.5772/intechopen.93217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
36
|
Chen W, Zhuo Y, Duan D, Lu M. Effects of Hypoxia on Differentiation of Mesenchymal Stem Cells. Curr Stem Cell Res Ther 2020; 15:332-339. [PMID: 31441734 DOI: 10.2174/1574888x14666190823144928] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/25/2019] [Accepted: 07/15/2019] [Indexed: 12/20/2022]
Abstract
Mesenchymal Stem Cells (MSCs) are distributed in many parts of the human body, including
the bone marrow, placenta, umbilical cord, fat, and nasal mucosa. One of the unique features of
MSCs is their multidirectional differentiation potential, including the ability to undergo osteogenesis,
adipogenesis, and chondrogenesis, and to produce neurons, endothelial cells, Schwann cells, medullary
nucleus cells, cardiomyocytes, and alveolar epithelial cells. MSCs have thus become a hot research
topic in recent years. Numerous studies have investigated the differentiation of MSCs into various
types of cells in vitro and their application to numerous fields. However, most studies have cultured
MSCs under atmospheric oxygen tension with an oxygen concentration of 21%, which does not reflect
a normal physiological state, given that the oxygen concentration generally used in vitro is four to ten
times that to which MSCs would be exposed in the body. We therefore review the growing number of
studies exploring the effect of hypoxic preconditioning on the differentiation of MSCs.
Collapse
Affiliation(s)
- Wei Chen
- Hunan Provincial Key Laboratory of Neurorestoratology, The Second Affiliated Hospital (the 921st Hospital of PLA), Hunan Normal University, Changsha, Hunan 410003, China
| | - Yi Zhuo
- Hunan Provincial Key Laboratory of Neurorestoratology, The Second Affiliated Hospital (the 921st Hospital of PLA), Hunan Normal University, Changsha, Hunan 410003, China
| | - Da Duan
- Hunan Provincial Key Laboratory of Neurorestoratology, The Second Affiliated Hospital (the 921st Hospital of PLA), Hunan Normal University, Changsha, Hunan 410003, China
| | - Ming Lu
- Hunan Provincial Key Laboratory of Neurorestoratology, The Second Affiliated Hospital (the 921st Hospital of PLA), Hunan Normal University, Changsha, Hunan 410003, China
| |
Collapse
|
37
|
Silva JC, Han X, Silva TP, Xia K, Mikael PE, Cabral JMS, Ferreira FC, Linhardt RJ. Glycosaminoglycan remodeling during chondrogenic differentiation of human bone marrow-/synovial-derived mesenchymal stem/stromal cells under normoxia and hypoxia. Glycoconj J 2020; 37:345-360. [PMID: 32086666 DOI: 10.1007/s10719-020-09911-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/20/2020] [Accepted: 01/23/2020] [Indexed: 12/19/2022]
Abstract
Glycosaminoglycans (GAGs) are major components of cartilage extracellular matrix (ECM), which play an important role in tissue homeostasis not only by providing mechanical load resistance, but also as signaling mediators of key cellular processes such as adhesion, migration, proliferation and differentiation. Specific GAG types as well as their disaccharide sulfation patterns can be predictive of the tissue maturation level but also of disease states such as osteoarthritis. In this work, we used a highly sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) method to perform a comparative study in terms of temporal changes in GAG and disaccharide composition between tissues generated from human bone marrow- and synovial-derived mesenchymal stem/stromal cells (hBMSC/hSMSC) after chondrogenic differentiation under normoxic (21% O2) and hypoxic (5% O2) micromass cultures. The chondrogenic differentiation of hBMSC/hSMSC cultured under different oxygen tensions was assessed through aggregate size measurement, chondrogenic gene expression analysis and histological/immunofluorescence staining in comparison to human chondrocytes. For all the studied conditions, the compositional analysis demonstrated a notable increase in the average relative percentage of chondroitin sulfate (CS), the main GAG in cartilage composition, throughout MSC chondrogenic differentiation. Additionally, hypoxic culture conditions resulted in significantly different average GAG and CS disaccharide percentage compositions compared to the normoxic ones. However, such effect was considerably more evident for hBMSC-derived chondrogenic aggregates. In summary, the GAG profiles described here may provide new insights for the prediction of cartilage tissue differentiation/disease states and to characterize the quality of MSC-generated chondrocytes obtained under different oxygen tension culture conditions.
Collapse
Affiliation(s)
- João C Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal.,Department of Chemistry and Chemical Biology, Biological Sciences, Biomedical Engineering and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180-3590, USA
| | - Xiaorui Han
- Department of Chemistry and Chemical Biology, Biological Sciences, Biomedical Engineering and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180-3590, USA
| | - Teresa P Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal
| | - Ke Xia
- Department of Chemistry and Chemical Biology, Biological Sciences, Biomedical Engineering and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180-3590, USA
| | - Paiyz E Mikael
- Department of Chemistry and Chemical Biology, Biological Sciences, Biomedical Engineering and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180-3590, USA
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology, Biological Sciences, Biomedical Engineering and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180-3590, USA.
| |
Collapse
|
38
|
Improved therapeutics of modified mesenchymal stem cells: an update. J Transl Med 2020; 18:42. [PMID: 32000804 PMCID: PMC6993499 DOI: 10.1186/s12967-020-02234-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/22/2020] [Indexed: 12/15/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) have attracted intense interest due to their powerful intrinsic properties of self-regeneration, immunomodulation and multi-potency, as well as being readily available and easy to isolate and culture. Notwithstanding, MSC based therapy suffers reduced efficacy due to several challenges which include unfavorable microenvironmental factors in vitro and in vivo. Body In the quest to circumvent these challenges, several modification techniques have been applied to the naïve MSC to improve its inherent therapeutic properties. These modification approaches can be broadly divided into two groups to include genetic modification and preconditioning modification (using drugs, growth factors and other molecules). This field has witnessed great progress and continues to gather interest and novelty. We review these innovative approaches in not only maintaining, but also enhancing the inherent biological activities and therapeutics of MSCs with respect to migration, homing to target site, adhesion, survival and reduced premature senescence. We discuss the application of the improved modified MSC in some selected human diseases. Possible ways of yet better enhancing the therapeutic outcome and overcoming challenges of MSC modification in the future are also elaborated. Conclusion The importance of prosurvival and promigratory abilities of MSCs in their therapeutic applications can never be overemphasized. These abilities are maintained and even further enhanced via MSC modifications against the inhospitable microenvironment during culture and transplantation. This is a turning point in MSC-based therapy with promising preclinical studies and higher future prospect.
Collapse
|
39
|
Silva JC, Moura CS, Borrecho G, Alves de Matos AP, Cabral JMS, Linhardt RJ, Ferreira FC. Effects of glycosaminoglycan supplementation in the chondrogenic differentiation of bone marrow- and synovial- derived mesenchymal stem/stromal cells on 3D-extruded poly (ε-caprolactone) scaffolds. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2019.1706511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- João C. Silva
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Department of Chemistry and Chemical Biology, Biological Sciences, Biomedical Engineering and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Carla S. Moura
- CDRSP – Centre for Rapid and Sustainable Product Development, Polytechnic Institute of Leiria, Rua de Portugal-Zona Industrial, Marinha Grande, Portugal
| | - Gonçalo Borrecho
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Quinta da Granja, Caparica, Portugal
| | | | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Robert J. Linhardt
- Department of Chemistry and Chemical Biology, Biological Sciences, Biomedical Engineering and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
40
|
Arjmand B, Tayanloo-Beik A, Foroughi Heravani N, Alaei S, Payab M, Alavi-Moghadam S, Goodarzi P, Gholami M, Larijani B. Zebrafish for Personalized Regenerative Medicine; A More Predictive Humanized Model of Endocrine Disease. Front Endocrinol (Lausanne) 2020; 11:396. [PMID: 32765420 PMCID: PMC7379230 DOI: 10.3389/fendo.2020.00396] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 05/18/2020] [Indexed: 12/18/2022] Open
Abstract
Regenerative medicine is a multidisciplinary field that aims to determine different factors and develop various methods to regenerate impaired tissues, organs, and cells in the disease and impairment conditions. When treatment procedures are specified according to the individual's information, the leading role of personalized regenerative medicine will be revealed in developing more effective therapies. In this concept, endocrine disorders can be considered as potential candidates for regenerative medicine application. Diabetes mellitus as a worldwide prevalent endocrine disease causes different damages such as blood vessel damages, pancreatic damages, and impaired wound healing. Therefore, a global effort has been devoted to diabetes mellitus investigations. Hereupon, the preclinical study is a fundamental step. Up to now, several species of animals have been modeled to identify the mechanism of multiple diseases. However, more recent researches have been demonstrated that animal models with the ability of tissue regeneration are more suitable choices for regenerative medicine studies in endocrine disorders, typically diabetes mellitus. Accordingly, zebrafish has been introduced as a model that possesses the capacity to regenerate different organs and tissues. Especially, fine regeneration in zebrafish has been broadly investigated in the regenerative medicine field. In addition, zebrafish is a suitable model for studying a variety of different situations. For instance, it has been used for developmental studies because of the special characteristics of its larva. In this review, we discuss the features of zebrafish that make it a desirable animal model, the advantages of zebrafish and recent research that shows zebrafish is a promising animal model for personalized regenerative diseases. Ultimately, we conclude that as a newly introduced model, zebrafish can have a leading role in regeneration studies of endocrine diseases and provide a good perception of underlying mechanisms.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmeh Foroughi Heravani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Setareh Alaei
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology and Pharmacology, Toxicology and Poisoning Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Bagher Larijani
| |
Collapse
|
41
|
Gale AL, Mammone RM, Dodson ME, Linardi RL, Ortved KF. The effect of hypoxia on chondrogenesis of equine synovial membrane-derived and bone marrow-derived mesenchymal stem cells. BMC Vet Res 2019; 15:201. [PMID: 31200719 PMCID: PMC6567476 DOI: 10.1186/s12917-019-1954-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Joint injury is extremely common in equine athletes and post-traumatic osteoarthritis (PTOA), a progressive and debilitating disease, is estimated to affect 60% of horses in the USA. The limited potential for intrinsic healing of articular cartilage has prompted intense efforts to identify a cell-based repair strategy to prevent progression of PTOA. Mesenchymal stem cells (MSCs) have the potential to become an ideal source for cell-based treatment of cartilage lesions; however, full chondrogenic differentiation remains elusive. Due to the relatively low oxygen tension in articular cartilage, hypoxia has been proposed as a method of increasing MSC chondrogenesis. The objective of this study was to investigate the effect of hypoxic culture conditions on chondrogenesis in equine synovial membrane-derived MSCs (SM-MSCs) and bone marrow-derived MSCs (BM-MSCs). MSCs were isolated from synovial membrane and bone marrow collected from 5 horses. Flow cytometric analysis was used to assess cell surface marker expression including CD29, CD44, CD90, CD105, CD45, CD-79α, MHCI and MHCII. MSC pellets were cultured in normoxic (21% O2) or in hypoxic (5% O2) culture conditions for 28 days. Following the culture period, chondrogenesis was assessed by histology, biochemical analyses and gene expression of chondrogenic-related genes including ACAN, COL2b, SOX9, and COL10A1. RESULTS Both cell types expressed markers consistent with stemness including CD29, CD44, CD90, CD105, and MHCI and were negative for exclusion markers (CD45, CD79α, and MHCII). Although the majority of outcome variables of chondrogenic differentiation were not significantly different between cell types or culture conditions, COL10A1 expression, a marker of chondrocyte hypertrophy, was lowest in hypoxic SM-MSCs and was significantly lower in hypoxic SM-MSCs compared to hypoxic BM-MSCs. CONCLUSIONS Hypoxic culture conditions do not appear to increase chondrogenesis of equine SM-MSCs or BM-MSCs; however, hypoxia may downregulate the hypertrophic marker COL10A1 in SM-MSCs.
Collapse
Affiliation(s)
- Alexis L Gale
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Renata M Mammone
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Michael E Dodson
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Renata L Linardi
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Kyla F Ortved
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA.
| |
Collapse
|
42
|
Correction to: Hypoxic condition enhances chondrogenesis in synovium-derived mesenchymal stem cells. Biomater Res 2019; 23:7. [PMID: 30873291 PMCID: PMC6399916 DOI: 10.1186/s40824-019-0158-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 11/25/2022] Open
|
43
|
Pattappa G, Johnstone B, Zellner J, Docheva D, Angele P. The Importance of Physioxia in Mesenchymal Stem Cell Chondrogenesis and the Mechanisms Controlling Its Response. Int J Mol Sci 2019; 20:E484. [PMID: 30678074 PMCID: PMC6387316 DOI: 10.3390/ijms20030484] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 12/12/2022] Open
Abstract
Articular cartilage covers the surface of synovial joints and enables joint movement. However, it is susceptible to progressive degeneration with age that can be accelerated by either previous joint injury or meniscectomy. This degenerative disease is known as osteoarthritis (OA) and it greatly affects the adult population. Cell-based tissue engineering provides a possible solution for treating OA at its earliest stages, particularly focal cartilage lesions. A candidate cell type for treating these focal defects are Mesenchymal Stem Cells (MSCs). However, present methods for differentiating these cells towards the chondrogenic lineage lead to hypertrophic chondrocytes and bone formation in vivo. Environmental stimuli that can stabilise the articular chondrocyte phenotype without compromising tissue formation have been extensively investigated. One factor that has generated intensive investigation in MSC chondrogenesis is low oxygen tension or physioxia (2⁻5% oxygen). In vivo articular cartilage resides at oxygen tensions between 1⁻4%, and in vitro results suggest that these conditions are beneficial for MSC expansion and chondrogenesis, particularly in suppressing the cartilage hypertrophy. This review will summarise the current literature regarding the effects of physioxia on MSC chondrogenesis with an emphasis on the pathways that control tissue formation and cartilage hypertrophy.
Collapse
Affiliation(s)
- Girish Pattappa
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany.
| | - Brian Johnstone
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.
| | - Johannes Zellner
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany.
| | - Denitsa Docheva
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany.
| | - Peter Angele
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany.
- Sporthopaedicum Regensburg, Hildegard von Bingen Strasse 1, 93053 Regensburg, Germany.
| |
Collapse
|
44
|
Mesenchymal stem cell-based therapy of osteoarthritis: Current knowledge and future perspectives. Biomed Pharmacother 2018; 109:2318-2326. [PMID: 30551490 DOI: 10.1016/j.biopha.2018.11.099] [Citation(s) in RCA: 195] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/30/2018] [Accepted: 11/25/2018] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is a chronic, prevalent, debilitating joint disease characterized by progressive cartilage degradation, subchondral bone remodeling, bone marrow lesions, meniscal damage, and synovitis. Innate immune cells (natural killer cells, macrophages, and mast cells) play the most important pathogenic role in the early inflammatory response, while cells of adaptive immunity (CD4 + Th1 lymphocytes and antibody producing B cells) significantly contribute to the development of chronic, relapsing course of inflammation in OA patients. Conventional therapy for OA is directed toward symptomatic treatment, mainly pain management, and is not able to promote regeneration of degenerated cartilage or to attenuate joint inflammation. Since articular cartilage, intra-articular ligaments, and menisci have no ability to heal, regeneration of these tissues remains one of the most important goals of new therapeutic approaches used for OA treatment. Due to their capacity for differentiation into chondrocytes and due to their immunomodulatory properties, mesenchymal stem cells (MSCs) have been the most extensively explored as new therapeutic agents in the cell-based therapy of OA. Simple acquisition, rapid proliferation, maintenance of differentiation potential after repeated passages in vitro, minor immunological rejection due to the low surface expression of major histocompatibility complex antigens, efficient engraftment and long-term coexistence in the host are the main characteristics of MSCs that enable their therapeutic use in OA. In this review article, we emphasized current knowledge and future perspectives regarding molecular and cellular mechanisms responsible for beneficial effects of autologous and allogeneic MSCs in the treatment of OA.
Collapse
|