101
|
Wang Y, Hu G, Hill RC, Dzieciatkowska M, Hansen KC, Zhang XB, Yan Z, Pei M. Matrix reverses immortalization-mediated stem cell fate determination. Biomaterials 2021; 265:120387. [PMID: 32987274 PMCID: PMC7944411 DOI: 10.1016/j.biomaterials.2020.120387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/24/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Primary cell culture in vitro suffers from cellular senescence. We hypothesized that expansion on decellularized extracellular matrix (dECM) deposited by simian virus 40 large T antigen (SV40LT) transduced autologous infrapatellar fat pad stem cells (IPFSCs) could rejuvenate high-passage IPFSCs in both proliferation and chondrogenic differentiation. In the study, we found that SV40LT transduced IPFSCs exhibited increased proliferation and adipogenic potential but decreased chondrogenic potential. Expansion on dECMs deposited by passage 5 IPFSCs yielded IPFSCs with dramatically increased proliferation and chondrogenic differentiation capacity; however, this enhanced capacity diminished if IPFSCs were grown on dECM deposited by passage 15 IPFSCs. Interestingly, expansion on dECM deposited by SV40LT transduced IPFSCs yielded IPFSCs with enhanced proliferation and chondrogenic capacity but decreased adipogenic potential, particularly for the dECM group derived from SV40LT transduced passage 15 cells. Our immunofluorescence staining and proteomics data identify matrix components such as basement membrane proteins as top candidates for matrix mediated IPFSC rejuvenation. Both cell proliferation and differentiation were endorsed by transcripts measured by RNASeq during the process. This study provides a promising model for in-depth investigation of the matrix protein influence on surrounding stem cell differentiation.
Collapse
Affiliation(s)
- Yiming Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Bioinformatics Core, West Virginia University, Morgantown, WV, USA
| | - Ryan C Hill
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Kirk C Hansen
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Medicine, Loma Linda University, Loma Linda, CA, USA.
| | - Zuoqin Yan
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
102
|
Kim YS, Suh DS, Tak DH, Chung PK, Koh YG. Mesenchymal Stem Cell Implantation in Knee Osteoarthritis: Midterm Outcomes and Survival Analysis in 467 Patients. Orthop J Sports Med 2020; 8:2325967120969189. [PMID: 33415176 PMCID: PMC7750771 DOI: 10.1177/2325967120969189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022] Open
Abstract
Background A cell-based tissue engineering approach that uses mesenchymal stem cells (MSCs) has addressed the issue of articular cartilage repair in knees with osteoarthritis (OA). Purpose To evaluate the midterm outcomes, analyze the survival rates, and identify the factors affecting the survival rate of MSC implantation to treat knee OA. Study Design Case series; Level of evidence, 4. Methods We retrospectively evaluated 467 patients (483 knees) who underwent MSC implantation on a fibrin glue scaffold for knee OA with a minimum 5-year follow-up. Clinical outcomes were determined based on the International Knee Documentation Committee (IKDC) and Tegner activity scale results measured preoperatively and during follow-up. Standard radiographs were evaluated using Kellgren-Lawrence grading. Statistical analyses were performed to determine the survival rate and the effect of different factors on the clinical outcomes. Results The mean IKDC scores (baseline, 39.2 ± 7.2; 1 year, 66.6 ± 9.6; 3 years, 67.2 ± 9.9; 5 years, 66.1 ± 9.7; 9 years, 62.8 ± 8.5) and Tegner scores (baseline, 2.3 ± 1.0; 1 year, 3.4 ± 0.9; 3 years, 3.5 ± 0.9; 5 years, 3.4 ± 0.9; 9 years, 3.2 ± 0.9) were significantly improved until 3 years postoperatively and gradually decreased from 3- to 9-year follow-up (P < .05 for all, except for Tegner score at 5 years vs 1 year [P = .237]). Gradual deterioration of radiological outcomes according to the Kellgren-Lawrence grade was found during follow-up. Survival rates based on either a decrease in IKDC or an advancement of radiographic OA with Kellgren-Lawrence scores were 99.8%, 94.5%, and 74.5% at 5, 7, and 9 years, respectively. Based on multivariate analyses, older age and the presence of bipolar kissing lesion were associated with significantly worse outcomes (P = .002 and .013, respectively), and a larger number of MSCs was associated with significantly better outcomes (P < .001) after MSC implantation. Conclusion MSC implantation provided encouraging outcomes with acceptable duration of symptom relief at midterm follow-up in patients with early knee OA. Patient age, presence of bipolar kissing lesion, and number of MSCs were independent factors associated with failure of MSC implantation.
Collapse
Affiliation(s)
- Yong Sang Kim
- Center for Stem Cell & Arthritis Research, Department of Orthopaedic Surgery, Yonsei Sarang Hospital, Seoul, Republic of Korea
| | - Dong Suk Suh
- Center for Stem Cell & Arthritis Research, Department of Orthopaedic Surgery, Yonsei Sarang Hospital, Seoul, Republic of Korea
| | - Dae Hyun Tak
- Center for Stem Cell & Arthritis Research, Department of Orthopaedic Surgery, Yonsei Sarang Hospital, Seoul, Republic of Korea
| | - Pill Ku Chung
- Center for Stem Cell & Arthritis Research, Department of Orthopaedic Surgery, Yonsei Sarang Hospital, Seoul, Republic of Korea
| | - Yong Gon Koh
- Center for Stem Cell & Arthritis Research, Department of Orthopaedic Surgery, Yonsei Sarang Hospital, Seoul, Republic of Korea
| |
Collapse
|
103
|
Wei J, Wang K, Hettinghouse A, Liu C. Atsttrin Promotes Cartilage Repair Primarily Through TNFR2-Akt Pathway. Front Cell Dev Biol 2020; 8:577572. [PMID: 33195216 PMCID: PMC7658268 DOI: 10.3389/fcell.2020.577572] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/12/2020] [Indexed: 01/16/2023] Open
Abstract
Background Cartilage defects account for substantial economic and humanistic burdens and pose a significant clinical problem. The efficacy of clinical approaches to cartilage repair is often inadequate, in part, owing to the restricted proliferative capacity of chondrocytes. Molecules have the capacity to promote the differentiation of multipotent mesenchymal stem cells into chondrocytes and may also gain the ability to repair the damaged cartilage. Objective This study aimed to investigate the role of Atsttrin (progranulin-derived engineered protein) in cartilage repair as well as the signaling pathway involved. Methods Primary and mesenchymal stem cell lines were used for the micromass culture. A murine cartilage defect model was used to determine the role of Atsttrin in cartilage repair in vivo. Real-time polymerase chain reaction and Western blot analysis were used to monitor the effect of Atsttrin on the transcriptional and protein levels, respectively, of key anabolic and catabolic signaling molecules. Results Atsttrin stimulated chondrogenesis in vitro and accelerated cartilage repair in vivo. In addition, Atsttrin-mediated cartilage repair occurred primarily through tumor necrosis factor receptor 2-initiated Akt signaling and downstream JunB transcription factor. Conclusion Atsttrin might serve as a promising therapeutic modality for cartilage regeneration.
Collapse
Affiliation(s)
- Jianlu Wei
- Department of Orthopaedic Surgery, Qilu Hospital of Shandong University, Jinan, China.,Department of Orthopaedic Surgery, New York University Langone Medical Center, New York, NY, United States
| | - Kaidi Wang
- Department of Orthopaedic Surgery, Qilu Hospital of Shandong University, Jinan, China
| | | | - Chuanju Liu
- Department of Orthopaedic Surgery, Qilu Hospital of Shandong University, Jinan, China.,Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
104
|
Kim YS, Suh DS, Tak DH, Chung PK, Kwon YB, Kim TY, Koh YG. Comparative matched-pair cohort analysis of the short-term clinical outcomes of mesenchymal stem cells versus hyaluronic acid treatments through intra-articular injections for knee osteoarthritis. J Exp Orthop 2020; 7:90. [PMID: 33188474 PMCID: PMC7666263 DOI: 10.1186/s40634-020-00310-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/09/2020] [Indexed: 02/08/2023] Open
Abstract
Purpose Intra-articular injection of hyaluronic acid (HA) has shown promises in reducing pain and improving physical function in knee osteoarthritis (OA). Recently, cell-based therapies using mesenchymal stem cells (MSCs) have emerged as potential treatments. However, few studies have compared the treatment outcomes between MSCs and HA. This study aimed to compare the clinical and radiological outcomes of intra-articular injections of MSCs versus HA in patients with knee OA. Methods A cohort of 209 patients with knee OA were retrospectively screened for those who underwent intra-articular injections using MSCs or HA. Thirty MSC-treated patients (MSC group) were pair-matched with thirty HA-treated patients (HA group) based on gender and age. Clinical outcomes were evaluated using the visual analog scale (VAS), International Knee Documentation Committee (IKDC) rating system, and Lysholm scoring system. Radiological evaluation was assessed using the Kellgren-Lawrence (K-L) grading system. Results MSC treatment yielded consistent significant improvements in VAS, IKDC and Lysholm scores. In the HA group, VAS scores significantly decreased at 1 month, slightly increased at 3 months, and increased significantly from 3 months to 1 year after injection. The IKDC and Lysholm scores improved significantly until 3 months, but gradually worsened thereafter. Significantly greater improvements in VAS (P = 0.041), IKDC (P = 0.014), and Lysholm (P = 0.020) scores were observed in the MSC group compared to those in the HA group at 1-year post-treatment. The K-L grade worsened in a few patients, especially those in the HA group, albeit no significant difference. Conclusions MSC group showed better VAS, IKDC, and Lysholm scores at 1-year post-treatment, compared to the HA group, although earlier clinical improvements were superior in the HA group for the initial 3 months. Level of Evidence Therapeutic study, Level III.
Collapse
Affiliation(s)
- Yong Sang Kim
- Department of Orthopaedic Surgery, Center for Stem Cell & Arthritis Research, Yonsei Sarang Hospital, 10, Hyoryeong-ro, Seocho-gu, Seoul, Republic of Korea
| | - Dong Suk Suh
- Department of Orthopaedic Surgery, Center for Stem Cell & Arthritis Research, Yonsei Sarang Hospital, 10, Hyoryeong-ro, Seocho-gu, Seoul, Republic of Korea
| | - Dae Hyun Tak
- Department of Orthopaedic Surgery, Center for Stem Cell & Arthritis Research, Yonsei Sarang Hospital, 10, Hyoryeong-ro, Seocho-gu, Seoul, Republic of Korea
| | - Pill Ku Chung
- Department of Orthopaedic Surgery, Center for Stem Cell & Arthritis Research, Yonsei Sarang Hospital, 10, Hyoryeong-ro, Seocho-gu, Seoul, Republic of Korea
| | - Yoo Beom Kwon
- Department of Orthopaedic Surgery, Center for Stem Cell & Arthritis Research, Yonsei Sarang Hospital, 10, Hyoryeong-ro, Seocho-gu, Seoul, Republic of Korea
| | - Tae Yong Kim
- Department of Orthopaedic Surgery, Center for Stem Cell & Arthritis Research, Yonsei Sarang Hospital, 10, Hyoryeong-ro, Seocho-gu, Seoul, Republic of Korea
| | - Yong Gon Koh
- Department of Orthopaedic Surgery, Center for Stem Cell & Arthritis Research, Yonsei Sarang Hospital, 10, Hyoryeong-ro, Seocho-gu, Seoul, Republic of Korea.
| |
Collapse
|
105
|
Ge M, Sheng Y, Qi S, Cao L, Zhang Y, Yang J. PLGA/chitosan-heparin composite microparticles prepared with microfluidics for the construction of hMSC aggregates. J Mater Chem B 2020; 8:9921-9932. [PMID: 33034328 DOI: 10.1039/d0tb01593h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Incorporating poly(lactic-co-glycolic) acid (PLGA) microparticles into human mesenchymal stem cells (hMSC) aggregates has shown promising application prospects. However, the acidic degradation products and burst release of PLGA microparticles still need to be ameliorated. In this study, the PLGA/chitosan-heparin (P/C-h) composite microparticles were successfully fabricated by integrating the double emulsion and microfluidic technology through the precise manipulation of the emulsion composition and flow rate of the two-phase in a flow-focusing chip. The P/C-h microparticles were highly monodispersed with a diameter of 23.45 ± 0.25 μm and shell-core structure of the PLGA encapsulated C-h complex, which were suitable for the fabrication of hMSC aggregates. When the mass ratio of PLGA to the C-h complex was optimized to 2 : 1, the pH of the leach liquor of P/C-h microparticles remained neutral. Compared with those of PLGA microparticles, the cytotoxicity and the initial burst release (loaded FGF-2 and VEGF) were both significantly reduced in P/C-h microparticles. Furthermore, the survival, stemness, as well as secretion and migration abilities of cells in hMSC aggregates incorporating P/C-h microparticles were also enhanced. In summary, the P/C-h composite microparticles prepared by the droplet microfluidic technique support the optimal biological and functional profile of the hMSC aggregates, which may facilitate the clinical applications of MSC-based therapy.
Collapse
Affiliation(s)
- Min Ge
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China.
| | - Yaqi Sheng
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China.
| | - Shuyue Qi
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China.
| | - Lei Cao
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China.
| | - Yan Zhang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China. and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China.
| | - Jun Yang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
106
|
Jacob G, Shimomura K, Nakamura N. Osteochondral Injury, Management and Tissue Engineering Approaches. Front Cell Dev Biol 2020; 8:580868. [PMID: 33251212 PMCID: PMC7673409 DOI: 10.3389/fcell.2020.580868] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Osteochondral lesions (OL) are a common clinical problem for orthopedic surgeons worldwide and are associated with multiple clinical scenarios ranging from trauma to osteonecrosis. OL vary from chondral lesions in that they involve the subchondral bone and chondral surface, making their management more complex than an isolated chondral injury. Subchondral bone involvement allows for a natural healing response from the body as marrow elements are able to come into contact with the defect site. However, this repair is inadequate resulting in fibrous scar tissue. The second differentiating feature of OL is that damage to the subchondral bone has deleterious effects on the mechanical strength and nutritive capabilities to the chondral joint surface. The clinical solution must, therefore, address both the articular cartilage as well as the subchondral bone beneath it to restore and preserve joint health. Both cartilage and subchondral bone have distinctive functional requirements and therefore their physical and biological characteristics are very much dissimilar, yet they must work together as one unit for ideal joint functioning. In the past, the obvious solution was autologous graft transfer, where an osteochondral bone plug was harvested from a non-weight bearing portion of the joint and implanted into the defect site. Allografts have been utilized similarly to eliminate the donor site morbidity associated with autologous techniques and overall results have been good but both techniques have their drawbacks and limitations. Tissue engineering has thus been an attractive option to create multiphasic scaffolds and implants. Biphasic and triphasic implants have been under explored and have both a chondral and subchondral component with an interface between the two to deliver an implant which is biocompatible and emulates the osteochondral unit as a whole. It has been a challenge to develop such implants and many manufacturing techniques have been utilized to bring together two unalike materials and combine them with cellular therapies. We summarize the functions of the osteochondral unit and describe the currently available management techniques under study.
Collapse
Affiliation(s)
- George Jacob
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Orthopedics, Tejasvini Hospital, Mangalore, India
| | - Kazunori Shimomura
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Norimasa Nakamura
- Institute for Medical Science in Sports, Osaka Health Science University, Osaka, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| |
Collapse
|
107
|
Zhao X, Zhao Y, Sun X, Xing Y, Wang X, Yang Q. Immunomodulation of MSCs and MSC-Derived Extracellular Vesicles in Osteoarthritis. Front Bioeng Biotechnol 2020; 8:575057. [PMID: 33251195 PMCID: PMC7673418 DOI: 10.3389/fbioe.2020.575057] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) has become recognized as a low-grade inflammatory state. Inflammatory infiltration of the synovium by macrophages, T cells, B cells, and other immune cells is often observed in OA patients and plays a key role in the pathogenesis of OA. Hence, orchestrating the local inflammatory microenvironment and tissue regeneration microenvironment is important for the treatment of OA. Mesenchymal stem cells (MSCs) offer the potential for cartilage regeneration owing to their effective immunomodulatory properties and anti-inflammatory abilities. The paracrine effect, mediated by MSC-derived extracellular vehicles (EVs), has recently been suggested as a mechanism for their therapeutic properties. In this review, we summarize the interactions between MSCs or MSC-derived EVs and OA-related immune cells and discuss their therapeutic effects in OA. Additionally, we discuss the potential of MSC-derived EVs as a novel cell-free therapy approach for the clinical treatment of OA.
Collapse
Affiliation(s)
- Xige Zhao
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
| | - Yanhong Zhao
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Yi Xing
- Stomatological Hospital of Tianjin Medical University, Tianjin, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, China
| |
Collapse
|
108
|
Madry H, Venkatesan JK, Carballo-Pedrares N, Rey-Rico A, Cucchiarini M. Scaffold-Mediated Gene Delivery for Osteochondral Repair. Pharmaceutics 2020; 12:pharmaceutics12100930. [PMID: 33003607 PMCID: PMC7601511 DOI: 10.3390/pharmaceutics12100930] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022] Open
Abstract
Osteochondral defects involve both the articular cartilage and the underlying subchondral bone. If left untreated, they may lead to osteoarthritis. Advanced biomaterial-guided delivery of gene vectors has recently emerged as an attractive therapeutic concept for osteochondral repair. The goal of this review is to provide an overview of the variety of biomaterials employed as nonviral or viral gene carriers for osteochondral repair approaches both in vitro and in vivo, including hydrogels, solid scaffolds, and hybrid materials. The data show that a site-specific delivery of therapeutic gene vectors in the context of acellular or cellular strategies allows for a spatial and temporal control of osteochondral neotissue composition in vitro. In vivo, implantation of acellular hydrogels loaded with nonviral or viral vectors has been reported to significantly improve osteochondral repair in translational defect models. These advances support the concept of scaffold-mediated gene delivery for osteochondral repair.
Collapse
Affiliation(s)
- Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany; (H.M.); (J.K.V.)
| | - Jagadeesh Kumar Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany; (H.M.); (J.K.V.)
| | - Natalia Carballo-Pedrares
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, S-15071 A Coruña, Spain; (N.C.-P.); (A.R.-R.)
| | - Ana Rey-Rico
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, S-15071 A Coruña, Spain; (N.C.-P.); (A.R.-R.)
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany; (H.M.); (J.K.V.)
- Correspondence: ; Tel.: +49-684-1162-4987; Fax: +49-684-1162-4988
| |
Collapse
|
109
|
Li F, Chen J, Gong M, Bi Y, Hu C, Zhang Y, Li M. Isolation and Characterization of Human Synovial Fluid-Derived Mesenchymal Stromal Cells from Popliteal Cyst. Stem Cells Int 2020; 2020:7416493. [PMID: 33014069 PMCID: PMC7519976 DOI: 10.1155/2020/7416493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/03/2020] [Accepted: 08/21/2020] [Indexed: 12/02/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitor cells in adult tissues. The aim of this study is to isolate and identify synovial fluid-derived mesenchymal stromal cells (SF-MSCs) from the popliteal cyst fluid of pediatric patients. SF-MSCs were collected from the popliteal cyst fluid of pediatric patients during cystectomy surgery. After cyst fluid extraction and adherent culturing, in vitro morphology, growth curve, and cell cycle were observed. The expression of stem cell surface markers was analyzed by flow cytometry, and expression of cell marker protein was detected by immunofluorescence. SF-MSCs were cultured in osteogenic, adipogenic, and chondrogenic differentiation medium. The differentiation potential of SF-MSCs was analyzed by alkaline phosphatase (Alizarin Red), Oil Red O, and Alcian blue. Antibody detection of human angiogenesis-related proteins was performed compared with bone marrow mesenchymal stem cells (BM-MSCs). The results show that SF-MSCs from the popliteal cyst fluid of pediatric patients showed a shuttle appearance and logarithmic growth. Flow cytometry analysis revealed that SF-MSCs were negative for hematopoietic lineage markers (CD34, CD45) and positive for MSC markers (CD44, CD73, CD90, and CD105). Interstitial cell marker (vimentin) and myofibroblast-like cell marker alpha-smooth muscle actin (α-SMA) were positive. These cells could differentiate into osteogenic, adipogenic, and chondrogenic lineages, respectively. Several types of human angiogenesis-related proteins were detected in the cell secretory fluid. These results show that we successfully obtained SF-MSCs from the popliteal cyst fluid of pediatric patients, which have the potential to be a valuable source of MSCs.
Collapse
Affiliation(s)
- Fang Li
- Department of Orthopedics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jianglin Chen
- Department of Orthopedics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Mengjia Gong
- Department of Orthopedics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Bi
- Department of Orthopedics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Chengchen Hu
- Department of Pediatric Research Institute, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanyuan Zhang
- Department of Pediatric Research Institute, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Li
- Department of Orthopedics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
110
|
Wang K, Li F, Yuan Y, Shan L, Cui Y, Qu J, Lian F. Synovial Mesenchymal Stem Cell-Derived EV-Packaged miR-31 Downregulates Histone Demethylase KDM2A to Prevent Knee Osteoarthritis. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:1078-1091. [PMID: 33294294 PMCID: PMC7691165 DOI: 10.1016/j.omtn.2020.09.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 09/11/2020] [Indexed: 12/26/2022]
Abstract
Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have emerged as important mediators of intercellular communication in response to cartilage damage. In this study, we sought to characterize the inhibitory role of microRNA (miR)-31 encapsulated in synovial MSC (SMSC)-derived EVs in knee osteoarthritis (OA). The expression of miR-31, lysine demethylase 2A (KDM2A), E2F transcription factor 1 (E2F1), and pituitary tumor transforming gene 1 (PTTG1) was validated in cartilage tissues of knee OA patients. Following SMSC-EV extraction and identification, chondrocytes with the miR-31 inhibitor were added with SMSC-EVs, whereupon the effects of miR-31 on proliferation and migration of chondrocytes were assessed. The interaction among miR-31, KDM2A, E2F1, and PTTG1 in chondrocyte activities was probed in vitro, along with an in vivo mouse knee OA model. We identified downregulated miR-31, E2F1, and PTTG1 and upregulated KDM2A in cartilage tissues of knee OA patients. SMSC-EV-packaged miR-31 potentiated chondrocyte proliferation and migration as well as cartilage formation by targeting KDM2A. Mechanistically, KDM2A bound to the transcription factor E2F1 and inhibited its transcriptional activity. Enrichment of E2F1 in the PTTG1 promoter region activated PTTG1 transcription, accelerating chondrocyte proliferation and migration. SMSC-EVs and EVs from miR-31-overexpressed SMSCs alleviated cartilage damage and inflammation in knee joints in vivo. SMSC-EV-encapsulated miR-31 ameliorates knee OA via the KDM2A/E2F1/PTTG1 axis.
Collapse
Affiliation(s)
- Kunpeng Wang
- Department of Orthopaedics, The Fourth Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Feng Li
- Department of Orthopaedics, the Second Hospital of Harbin Medical University, Harbin 150001, P.R. P. China
| | - Yuan Yuan
- Department of Obstetrics, The Fourth Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Liang Shan
- Department of Outpatient, The Fourth Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Yong Cui
- Department of Orthopaedics, The Fourth Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Jing Qu
- Department of Orthopaedics, The Fourth Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Feng Lian
- Department of Orthopaedics, The Fourth Hospital of Harbin Medical University, Harbin 150001, P.R. China
| |
Collapse
|
111
|
Song Y, Zhang J, Xu H, Lin Z, Chang H, Liu W, Kong L. Mesenchymal stem cells in knee osteoarthritis treatment: A systematic review and meta-analysis. J Orthop Translat 2020; 24:121-130. [PMID: 32913710 PMCID: PMC7452318 DOI: 10.1016/j.jot.2020.03.015] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Stem cells are considered to be one of the greatest potential treatments to cure degenerative diseases. Stem cells injection for knee osteoarthritis (OA) is still a relatively new treatment and has not yet gained popularity. So, the effectiveness, safety and potential of mesenchymal stem cells (MSCs) for knee OA treatment is worthy to be explored. Explore the effectiveness and safety of mesenchymal stem cells (MSCs) in the treatment of knee osteoarthritis. We collected clinical trials using MSCs as treatment for knee OA (before April 2019), including randomized controlled trials (RCTs), retrospective studies and cohort studies. We searched PubMed, EMBASE, Cochrane Library, Web of Science and the ClinicalTrials.gov with keywords (Mesenchymal stem cells [MSCs], Knee osteoarthritis, Effectiveness and Safety), and then performed a systematic review and cumulative metaanalysis of all RCTs and retrospective comparative studies. To evaluate the effectiveness and safety of MSC in knee OA treatment, we applied visual analog scale score, Western Ontario and McMaster Universities Osteo-arthritis Index and adverse events. We included 15 RCTs, two retrospective studies and two cohort studies including a total of 584 knee OA patients in this study. We demonstrated that MSC treatment could significantly decrease visual analog scale in a 12-month follow-up study compared with controls (p < 0.001). MSC therapy also showed significant decreases in Western Ontario and McMaster Universities Osteoarthritis Index scores after the 6-month follow-up (p < 0.001). MSC therapy showed no difference compared with controls (p > 0.05) in adverse events. We suggest that MSC therapy could serve as an effective and safe therapy for clinical application in OA treatment. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE This study provided the best available evidence and a wider perspective to MSCs application in the management of knee OA. MSCs therapy will have great translational potential in the clinical treatment of various degenerative diseases once optimum formula and explicit target population are identified.
Collapse
Affiliation(s)
- Yancheng Song
- Department of Orthopedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Corresponding author. Department of Orthopedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Rd., Yuexiu District, 510000, Guangzhou, China.
| | - Junhui Zhang
- Department of Orthopedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Hualiang Xu
- Department of Orthopedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhujian Lin
- Department of Orthopedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Hong Chang
- Department of Orthopedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Wei Liu
- Department of Orthopedics, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Ling Kong
- Department of Basic Research & International Cooperation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- Corresponding author. Department of Basic Research & International Cooperation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China.
| |
Collapse
|
112
|
Maki CB, Beck A, Wallis CBCC, Choo J, Ramos T, Tong R, Borjesson DL, Izadyar F. Intra-articular Administration of Allogeneic Adipose Derived MSCs Reduces Pain and Lameness in Dogs With Hip Osteoarthritis: A Double Blinded, Randomized, Placebo Controlled Pilot Study. Front Vet Sci 2020; 7:570. [PMID: 33110913 PMCID: PMC7489271 DOI: 10.3389/fvets.2020.00570] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/16/2020] [Indexed: 01/07/2023] Open
Abstract
This study was conducted to investigate the therapeutic effect of allogeneic adipose-derived MSCs on dogs with hip osteoarthritis (OA). Twenty dogs with bilateral osteoarthritis of the coxofemoral (hip) joint, diagnosed by a veterinarian through physical examination and radiographs were randomly allocated into four groups. Group 1 served as a placebo control and were injected with 0.9% sodium chloride (saline) (n = 4). Group 2 were injected with a single dose of 5 million MSCs (n = 5). Group 3 received a single dose of 25 million MSCs (n = 6) and Group 4 received a single dose of 50 million MSCs (n = 5). Intra-articular administration of allogeneic MSCs into multiple joints did not result in any serious adverse events. The average lameness score of the dogs in the placebo control group (−0.31) did not show improvement after 90 days of intra-articular saline administration. However, the average lameness score of the all MSC-treated dogs was improved 2.11 grade at this time point (P < 0.001). Overall, sixty five percent (65%) of the dogs that received various doses of MSCs showed improvement in lameness scores 90 days after intra-articular MSC administration. Our results showed that intra-articular administration of allogeneic adipose derived MSCs was well-tolerated and improved lameness scores and reduced pain in dogs associated with hip OA. All doses of MSCs were effective. Subsequent studies with more animals per group are needed to make a conclusion about the dose response. The improved lameness effect was present up to 90 days post-injection. Serum interleukin 10 was increased in a majority of the dogs that received MSCs and that also had improved lameness.
Collapse
Affiliation(s)
- Chad B Maki
- VetCell Therapeutics USA, Santa Ana, CA, United States
| | - Anthony Beck
- Doctors Beck and Stone Clinic, Discovery Bay, Hong Kong
| | | | - Justin Choo
- Doctors Beck and Stone Clinic, Discovery Bay, Hong Kong
| | - Thomas Ramos
- VetCell Therapeutics USA, Santa Ana, CA, United States
| | | | - Dori L Borjesson
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, Veterinary Institute for Regenerative Cures, University of California, Davis, Davis, CA, United States
| | | |
Collapse
|
113
|
Kim GB, Kim JD, Choi Y, Choi CH, Lee GW. Intra-Articular Bone Marrow Aspirate Concentrate Injection in Patients with Knee Osteoarthritis. APPLIED SCIENCES 2020; 10:5945. [DOI: 10.3390/app10175945] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
We aimed to evaluate the 5-year follow-up outcomes of an intra-articular bone marrow aspirate concentrate (BMAC) injection in patients with knee osteoarthritis. This is the first study to report the outcomes following BMAC injections over a 5-year follow-up period. Seventy knees of 37 patients, including 33 bilateral knees, were investigated. The primary outcome was the visual analogue scale (VAS) score for pain in the knee joint, and the secondary outcomes were the International Knee Documentation Committee score, the 36-Item Short Form Health Survey score, the Knee injury Osteoarthritis Outcome Score, Lysholm Knee Questionnaire/Tegner activity scale, BMAC injection-induced complications, and 5-year treatment success rate. The 5-year post-injection VAS scores (4.7 ± 0.5) were significantly lower than the preoperative scores (8.3 ± 1.2) (p = 0.01). Improvement in VAS scores was significantly greater in patients with Kellgren–Lawrence (K-L) Grade I or II than those in those with K-L Grade III or IV. Improvement in other clinical parameters and success rates were significantly low and the rates of secondary operation and failure were significantly higher in patients with K-L Grades III or IV. Intra-articular BMAC injections could be useful for managing patients with K-L Grades I or II osteoarthritis.
Collapse
Affiliation(s)
- Gi Beom Kim
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyeonchung-ro, Namgu, Daegu 42415, Korea
| | - Jae-Do Kim
- Department of Orthopedic Surgery, Kosin University College of Medicine, Kosin University Gospel Hospital, 34 Amnam-dong, Seogu, Busan 602-702, Korea
| | - Young Choi
- Department of Orthopedic Surgery, Kosin University College of Medicine, Kosin University Gospel Hospital, 34 Amnam-dong, Seogu, Busan 602-702, Korea
| | - Chang Hyun Choi
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyeonchung-ro, Namgu, Daegu 42415, Korea
| | - Gun Woo Lee
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyeonchung-ro, Namgu, Daegu 42415, Korea
| |
Collapse
|
114
|
Yohimbine Ameliorates Temporomandibular Joint Chondrocyte Inflammation with Suppression of NF-κB Pathway. Inflammation 2020; 44:80-90. [PMID: 32789555 DOI: 10.1007/s10753-020-01310-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Local joint inflammation plays an important role in the pathogenesis of temporomandibular joint (TMJ) osteoarthrosis (TMJOA). Yohimbine, an alpha-2 adrenergic receptor antagonist, possesses anti-inflammatory properties; however, the ability of Yohimbine to protect against TMJOA-associated chondrocyte inflammation remains unclear. We conducted in vitro and in vivo analyses to investigate whether Yohimbine could ameliorate TMJOA-induced chondrocyte inflammation and to elucidate the mechanisms involved. Chondrocytes of TMJOA mice were stimulated with interleukin (IL)-1β or noradrenaline (NE), and the resulting production of inflammation-related factors was evaluated in the presence or absence of Yohimbine. Furthermore, two TMJOA mouse models were treated with Yohimbine and the therapeutic effect was quantified. NE (10-6 M) triggered inflammatory cytokine secretion by TMJ chondrocytes, and Yohimbine suppressed IL-1β- or NE-induced IL-6 upregulation in TMJ chondrocytes with the nuclear factor (NF)-κB pathway inhibition. Yohimbine also ameliorated cartilage destruction in the TMJOA models. Interestingly, αmpT, a tyrosine hydroxylase inhibitor, reversed the effects of Yohimbine by activating the NF-κB pathway. Collectively, these findings show that Yohimbine ameliorated TMJ chondrocyte inflammation and the suppression of NF-κB pathway contributes to this effect.
Collapse
|
115
|
Gong W, Wang F, He Y, Zeng X, Zhang D, Chen Q. Mesenchymal Stem Cell Therapy for Oral Inflammatory Diseases: Research Progress and Future Perspectives. Curr Stem Cell Res Ther 2020; 16:165-174. [PMID: 32713335 DOI: 10.2174/1574888x15666200726224132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/21/2020] [Accepted: 06/03/2020] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cell (MSC) therapy for clinical diseases associated with inflammation and tissue damage has become a progressive treatment strategy. MSCs have unique biological functions, such as homing, immune regulation, and differentiation capabilities, which provide the prerequisites for the treatment of clinical diseases. Oral diseases are often associated with abnormal immune regulation and epithelial tissue damage. In this review, we summarize previous studies that use MSC therapy to treat various oral inflammatory diseases, including oral ulceration, allergic diseases, chemo/radiotherapy-induced oral mucositis, periodontitis, osteonecrosis of the jaw, Sjögren's syndrome (SS), among other similar diseases. We highlight MSC treatment as a promising approach in the management of oral inflammatory diseases, and discuss the obstacles that remain and must be overcome for MSC treatment to thrive in the future.
Collapse
Affiliation(s)
- Wang Gong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Fei Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuqing He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Dunfang Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
116
|
Keenan CM, Ramos-Mucci L, Kanakis I, Milner PI, Leask A, Abraham D, Bou-Gharios G, Poulet B. Post-traumatic osteoarthritis development is not modified by postnatal chondrocyte deletion of Ccn2. Dis Model Mech 2020; 13:dmm044719. [PMID: 32616521 PMCID: PMC7375478 DOI: 10.1242/dmm.044719] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/29/2020] [Indexed: 01/20/2023] Open
Abstract
CCN2 is a matricellular protein involved in several crucial biological processes. In particular, CCN2 is involved in cartilage development and in osteoarthritis. Ccn2 null mice exhibit a range of skeletal dysmorphisms, highlighting its importance in regulating matrix formation during development; however, its role in adult cartilage remains unclear. The aim of this study was to determine the role of CCN2 in postnatal chondrocytes in models of post-traumatic osteoarthritis (PTOA). Ccn2 deletion was induced in articular chondrocytes of male transgenic mice at 8 weeks of age. PTOA was induced in knees either surgically or non-invasively by repetitive mechanical loading at 10 weeks of age. Knee joints were harvested, scanned with micro-computed tomography and processed for histology. Sections were stained with Toluidine Blue and scored using the Osteoarthritis Research Society International (OARSI) grading system. In the non-invasive model, cartilage lesions were present in the lateral femur, but no significant differences were observed between wild-type (WT) and Ccn2 knockout (KO) mice 6 weeks post-loading. In the surgical model, severe cartilage degeneration was observed in the medial compartments, but no significant differences were observed between WT and Ccn2 KO mice at 2, 4 and 8 weeks post-surgery. We conclude that Ccn2 deletion in chondrocytes does not modify the development of PTOA in mice, suggesting that chondrocyte expression of CCN2 in adults is not a crucial factor in protecting cartilage from the degeneration associated with PTOA.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Craig M Keenan
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, West Derby Street, Liverpool L7 8TX, UK
| | - Lorenzo Ramos-Mucci
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, West Derby Street, Liverpool L7 8TX, UK
| | - Ioannis Kanakis
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, West Derby Street, Liverpool L7 8TX, UK
| | - Peter I Milner
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, West Derby Street, Liverpool L7 8TX, UK
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, SK S7N 5E4, Canada
| | - David Abraham
- Centre for Rheumatology and Connective Tissue Diseases, University College London, London NW3 2PF, UK
| | - George Bou-Gharios
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, West Derby Street, Liverpool L7 8TX, UK
| | - Blandine Poulet
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, West Derby Street, Liverpool L7 8TX, UK
| |
Collapse
|
117
|
The osteogenic commitment of CD271+CD56+ bone marrow stromal cells (BMSCs) in osteoarthritic femoral head bone. Sci Rep 2020; 10:11145. [PMID: 32636407 PMCID: PMC7341749 DOI: 10.1038/s41598-020-67998-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA), the most common joint disorder, is characterised by progressive structural changes in both the cartilage and the underlying subchondral bone. In late disease stages, subchondral bone sclerosis has been linked to heightened osteogenic commitment of bone marrow stromal cells (BMSCs). This study utilised cell sorting and immunohistochemistry to identify a phenotypically-distinct, osteogenically-committed BMSC subset in human OA trabecular bone. Femoral head trabecular bone tissue digests were sorted into CD45-CD271+CD56+CD146-, CD45-CD271+CD56-CD146+ and CD45-CD271+CD56-CD146-(termed double-negative, DN) subsets, and CD45+CD271-hematopoietic-lineage cells served as control. Compared to the CD146+ subset, the CD56+ subset possessed a lower-level expression of adipocyte-associated genes and significantly over 100-fold higher-level expression of many osteoblast-related genes including osteopontin and osteocalcin, whilst the DN subset presented a transcriptionally ‘intermediate’ BMSC population. All subsets were tri-potential following culture-expansion and were present in control non-OA trabecular bone. However, while in non-OA bone CD56+ cells only localised on the bone surface, in OA bone they were additionally present in the areas of new bone formation rich in osteoblasts and newly-embedded osteocytes. In summary, this study reveals a distinct osteogenically-committed CD271+CD56+ BMSC subset and implicates it in subchondral bone sclerosis in hip OA. CD271+CD56+ subset may represent a future therapeutic target for OA and other bone-associated pathologies.
Collapse
|
118
|
Masson AO, Krawetz RJ. Understanding cartilage protection in OA and injury: a spectrum of possibilities. BMC Musculoskelet Disord 2020; 21:432. [PMID: 32620156 PMCID: PMC7334861 DOI: 10.1186/s12891-020-03363-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/25/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent musculoskeletal disease resulting in progressive degeneration of the hyaline articular cartilage within synovial joints. Current repair treatments for OA often result in poor quality tissue that is functionally ineffective compared to the hyaline cartilage and demonstrates increased failure rates post-treatment. Complicating efforts to improve clinical outcomes, animal models used in pre-clinical research show significant heterogeneity in their regenerative and degenerative responses associated with their species, age, genetic/epigenetic traits, and context of cartilage injury or disease. These can lead to variable outcomes when testing and validating novel therapeutic approaches for OA. Furthermore, it remains unclear whether protection against OA among different model systems is driven by inhibition of cartilage degeneration, enhancement of cartilage regeneration, or any combination thereof. MAIN TEXT Understanding the mechanistic basis underlying this context-dependent duality is essential for the rational design of targeted cartilage repair and OA therapies. Here, we discuss some of the critical variables related to the cross-species paradigm of degenerative and regenerative abilities found in pre-clinical animal models, to highlight that a gradient of regenerative competence within cartilage may exist across species and even in the greater human population, and likely influences clinical outcomes. CONCLUSIONS A more complete understanding of the endogenous regenerative potential of cartilage in a species specific context may facilitate the development of effective therapeutic approaches for cartilage injury and/or OA.
Collapse
Affiliation(s)
- Anand O Masson
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Roman J Krawetz
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, Canada. .,Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada. .,Department Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada. .,Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
119
|
Nakayama N, Pothiawala A, Lee JY, Matthias N, Umeda K, Ang BK, Huard J, Huang Y, Sun D. Human pluripotent stem cell-derived chondroprogenitors for cartilage tissue engineering. Cell Mol Life Sci 2020; 77:2543-2563. [PMID: 31915836 PMCID: PMC11104892 DOI: 10.1007/s00018-019-03445-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/24/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
The cartilage of joints, such as meniscus and articular cartilage, is normally long lasting (i.e., permanent). However, once damaged, especially in large animals and humans, joint cartilage is not spontaneously repaired. Compensating the lack of repair activity by supplying cartilage-(re)forming cells, such as chondrocytes or mesenchymal stromal cells, or by transplanting a piece of normal cartilage, has been the basis of therapy for biological restoration of damaged joint cartilage. Unfortunately, current biological therapies face problems on a number of fronts. The joint cartilage is generated de novo from a specialized cell type, termed a 'joint progenitor' or 'interzone cell' during embryogenesis. Therefore, embryonic chondroprogenitors that mimic the property of joint progenitors might be the best type of cell for regenerating joint cartilage in the adult. Pluripotent stem cells (PSCs) are expected to differentiate in culture into any somatic cell type through processes that mimic embryogenesis, making human (h)PSCs a promising source of embryonic chondroprogenitors. The major research goals toward the clinical application of PSCs in joint cartilage regeneration are to (1) efficiently generate lineage-specific chondroprogenitors from hPSCs, (2) expand the chondroprogenitors to the number needed for therapy without loss of their chondrogenic activity, and (3) direct the in vivo or in vitro differentiation of the chondroprogenitors to articular or meniscal (i.e., permanent) chondrocytes rather than growth plate (i.e., transient) chondrocytes. This review is aimed at providing the current state of research toward meeting these goals. We also include our recent achievement of successful generation of "permanent-like" cartilage from long-term expandable, hPSC-derived ectomesenchymal chondroprogenitors.
Collapse
Affiliation(s)
- Naoki Nakayama
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA.
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA.
| | - Azim Pothiawala
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - John Y Lee
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Nadine Matthias
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - Katsutsugu Umeda
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Department of Pediatrics, Kyoto University School of Medicine, Kyoto, Japan
| | - Bryan K Ang
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Weil Cornell Medicine, New York, NY, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA
- Steadman Philippon Research Institute, Vail, CO, USA
| | - Yun Huang
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Deqiang Sun
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| |
Collapse
|
120
|
Maleitzke T, Elazaly H, Festbaum C, Eder C, Karczewski D, Perka C, Duda GN, Winkler T. Mesenchymal Stromal Cell-Based Therapy-An Alternative to Arthroplasty for the Treatment of Osteoarthritis? A State of the Art Review of Clinical Trials. J Clin Med 2020; 9:jcm9072062. [PMID: 32630066 PMCID: PMC7409016 DOI: 10.3390/jcm9072062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 01/06/2023] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disorder worldwide and to date no regenerative treatment has been established in clinical practice. This review evaluates the current literature on the clinical translation of mesenchymal stromal cell (MSC)-based therapy in OA management with a focus on safety, outcomes and procedural specifics. PubMed, Cochrane Library and clinicaltrials.gov were searched for clinical studies using MSCs for OA treatment. 290 articles were initially identified and 42 articles of interest, including a total of 1325 patients, remained for further examination. Most of the included studies used adipose tissue-derived MSCs or bone-marrow-derived MSCs to treat patients suffering from knee OA. MSC-based therapy for knee OA appears to be safe and presumably effective in selected parameters. Yet, a direct comparison between studies was difficult due to a pronounced variance regarding methodology, assessed outcomes and evidence levels. Intensive scientific engagement is needed to identify the most effective source and dosage of MSCs for OA treatment in the future. Consent on outcome measures has to be reached and eventually patient sub-populations need to be identified that will profit most from MSC-based treatment for OA.
Collapse
Affiliation(s)
- Tazio Maleitzke
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Hisham Elazaly
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Christian Festbaum
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
| | - Christian Eder
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
| | - Daniel Karczewski
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
| | - Carsten Perka
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Tobias Winkler
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-559084
| |
Collapse
|
121
|
Zheng G, Xie ZY, Wang P, Wu YF, Shen HY. Recent advances of single-cell RNA sequencing technology in mesenchymal stem cell research. World J Stem Cells 2020; 12:438-447. [PMID: 32742561 PMCID: PMC7360991 DOI: 10.4252/wjsc.v12.i6.438] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/13/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells with great potential for clinical applications. However, little is known about their cell heterogeneity at a single-cell resolution, which severely impedes the development of MSC therapy. In this review, we focus on advances in the identification of novel surface markers and functional subpopulations of MSCs made by single-cell RNA sequencing and discuss their participation in the pathophysiology of stem cells and related diseases. The challenges and future directions of single-cell RNA sequencing in MSCs are also addressed in this review.
Collapse
Affiliation(s)
- Guan Zheng
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Zhong-Yu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Yan-Feng Wu
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong Province, China
| | - Hui-Yong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| |
Collapse
|
122
|
The Efficacy of Stem Cells Secretome Application in Osteoarthritis: A Systematic Review of In Vivo Studies. Stem Cell Rev Rep 2020; 16:1222-1241. [DOI: 10.1007/s12015-020-09980-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
123
|
Dykstra JA, Blue ED, Negrão de Assis PL, Weimer JM, Kota DJ. Human adipose-derived stromal vascular fraction: characterization, safety and therapeutic potential in an experimental mouse model of articular injury. J Stem Cells Regen Med 2020; 16:16-25. [PMID: 32536767 DOI: 10.46582/jsrm.1601004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 01/09/2020] [Indexed: 12/16/2022]
Abstract
Due to their capacity to self-renew, proliferate and generate multi-lineage cells, adult-derived stem cells offer great potential in regenerative therapies to treat maladies such as diabetes, cardiac disease, neurological disorders and orthopedic injuries. Commonly derived from adipose tissue, the stromal vascular fraction (SVF), a heterogeneous cell population enriched with mesenchymal stem cells (MSCs), has garnered interest as a cellular therapy due to ease of accessibility as an autologous, point-of-care application. However, the heterogeneous cell population within SVF is not historically taken into consideration when injecting into patients. Here, we characterized SVF, determined its safety and verify its therapeutic effects in a NOD/scid mouse model of articular injury. SVF were isolated from lipoaspirates utilizing a commercially available system (InGeneron Inc.), while MSCs were isolated from SVF via cell culture. Flow cytometry showed that neither age nor BMI affects the frequency of progenitor cells-like (CD31+CD34+), immune cells-like (CD4+) T cells, (CD14+) monocytes and total number of cells obtained. However, there was a negative correlation between donor BMI and MSC frequency within the SVF. ELISAs showed that following LPS activation in SVF, there were low levels of TNF-α and high levels of IL-10 secreted. However, T cell activation with anti-CD3 or anti-CD3+ anti-CD28, while leading to expected high levels of IFN-γ, did not lead to significant levels of TGF-β. PCR analysis showed no significant numbers of cells outside the joint 1-hour post injection, moreover, no engraftment or abnormal growth in other organs 60-days post injection. Finally, both cell populations were able to ameliorate disease progression, as confirmed by the increase in movement of treated groups compared to injured groups. Noteworthy, the histological analysis indicated that there was no cartilage growth, suggesting an alternative therapeutic mechanism to cartilage regeneration.
Collapse
Affiliation(s)
- Jordan A Dykstra
- Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA.,Sanford Research, Sioux Falls, SD 57104, USA: These authors contributed equally
| | - Elliot D Blue
- Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA.,Sanford Research, Sioux Falls, SD 57104, USA: These authors contributed equally
| | | | - Jill M Weimer
- Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA.,Sanford Research, Sioux Falls, SD 57104, USA: These authors contributed equally
| | - Daniel Jiro Kota
- Sanford Research, Sioux Falls, SD 57104, USA: These authors contributed equally.,Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
124
|
Li W, Alahdal M, Deng Z, Liu J, Zhao Z, Cheng X, Chen X, Li J, Yin J, Li Y, Wang G, Wang D, Tang K, Zhang J. Molecular functions of FSTL1 in the osteoarthritis. Int Immunopharmacol 2020; 83:106465. [PMID: 32259701 DOI: 10.1016/j.intimp.2020.106465] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/11/2020] [Accepted: 03/29/2020] [Indexed: 12/18/2022]
|
125
|
Fan W, Yuan L, Li J, Wang Z, Chen J, Guo C, Mo X, Yan Z. Injectable double-crosslinked hydrogels with kartogenin-conjugated polyurethane nano-particles and transforming growth factor β3 for in-situ cartilage regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110705. [PMID: 32204019 DOI: 10.1016/j.msec.2020.110705] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/28/2019] [Accepted: 01/28/2020] [Indexed: 01/10/2023]
Abstract
Articular cartilage has a limited ability for self-repair after injury. Implantation of scaffolds functionalized with bioactive molecules that could induce the migration and chondrogenesis of endogenous mesenchymal stem cells (MSCs) provides a convenient alternative for in-situ cartilage regeneration. In this study, we found the synergistic effects of kartogenin (KGN) and transforming growth factor β3 (TGF-β3) on chondrogenesis of MSCs in vitro, indicating that KGN and TGF-β3 are a good match for cartilage regeneration. Furthermore, we confirmed that KGN promoted the chondrogenesis of MSCs through attenuating the degradation of Runx1, which physically interacted with p-Smad3 in nuclei of MSCs. Meanwhile, we designed an injectable double-crosslinked hydrogel with superior mechanical property and longer support for cartilage regeneration by modifying sodium alginate and gelatin. When loaded with KGN conjugated polyurethane nanoparticles (PN-KGN) and TGF-β3, this hydrogel showed biological functions by the release of KGN and TGF-β3, which promoted the MSC migration and cartilage regeneration in one system. In conclusion, the cell-free hydrogel, along with PN-KGN and TGF-β3, provides a promising strategy for cartilage repair by attracting endogenous MSCs and inducing chondrogenesis of recruited cells in a single-step procedure.
Collapse
Affiliation(s)
- Wenshuai Fan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liu Yuan
- Biomaterials and Tissue Engineering Lab, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Jinghuan Li
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhe Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jifei Chen
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Changan Guo
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiumei Mo
- Biomaterials and Tissue Engineering Lab, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China.
| | - Zuoqin Yan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
126
|
Geng Y, Chen J, Alahdal M, Chang C, Duan L, Zhu W, Mou L, Xiong J, Wang M, Wang D. Intra-articular injection of hUC-MSCs expressing miR-140-5p induces cartilage self-repairing in the rat osteoarthritis. J Bone Miner Metab 2020; 38:277-288. [PMID: 31760502 DOI: 10.1007/s00774-019-01055-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 10/10/2019] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Currently, osteoarthritis (OA) receives global increasing attention because it associates severe joint pain and serious disability. Stem cells intra-articular injection therapy showed a potential therapeutic superiority to reduce OA development and to improve treating outputs. However, the long-term effect of stem cells intra-articular injection on the cartilage regeneration remains unclear. Recently, miR-140-5p was confirmed as a critical positive regulator in chondrogenesis. We hypothesized that hUC-MSCs overexpressing miR-140-5p have better therapeutic effect on osteoarthritis. MATERIALS AND METHODS To enhance stem cell chondrogenic differentiation, we have transfected human umbilical cord mesenchymal stem cells (hUC-MSCs) with miR-140-5p mimics and miR-140-5p lentivirus to overexpress miR-140-5p in a short term or a long term accordingly. Thereafter, MSCs proliferation, chondrogenic genes expression and extracellular matrix were assessed. Destabilization of the medial meniscus (DMM) surgery was performed on the knee joints of SD rats as an OA model, and then intra-articular injection of hUC-MSCs or hUC-MSCs transfected with miR-140-5p lentivirus was carried to evaluate the cartilage healing effect with histological staining and OARSI scores. The localization of hUC-MSCs after intra-articular injection was further confirmed by immunohistochemical staining. RESULTS Significant induction of chondrogenic differentiation in the miR-140-5p-hUC-MSCs (140-MSCs), while its proliferation was not influenced. Interestingly, intra-articular injection of 140-MSCs significantly enhanced articular cartilage self-repairing in comparison to normal hUC-MSCs. Moreover, we noticed that intra-articular injection of high 140-MSCs numbers reinforces cells assembling on the impaired cartilage surface and subsequently differentiated into chondrocytes. CONCLUSIONS In conclusion, these results indicate therapeutic superiority of hUC-MSCs overexpressing miR-140-5p to treat OA using intra-articular injection.
Collapse
Affiliation(s)
- Yiyun Geng
- The First Affiliated Hospital to Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen, 518035, Guangdong Province, China
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518035, Guangdong Province, China
| | - Jinfu Chen
- The First Affiliated Hospital to Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
| | - Murad Alahdal
- The First Affiliated Hospital to Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen, 518035, Guangdong Province, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Chongfei Chang
- The First Affiliated Hospital to Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
| | - Li Duan
- The First Affiliated Hospital to Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen, 518035, Guangdong Province, China
| | - Weimin Zhu
- The First Affiliated Hospital to Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
| | - Lisha Mou
- The First Affiliated Hospital to Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
| | - Jianyi Xiong
- The First Affiliated Hospital to Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen, 518035, Guangdong Province, China
| | - Manyi Wang
- The First Affiliated Hospital to Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China
| | - Daping Wang
- The First Affiliated Hospital to Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong Province, China.
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen, 518035, Guangdong Province, China.
| |
Collapse
|
127
|
War AR, Dang K, Jiang S, Xiao Z, Miao Z, Yang T, Li Y, Qian A. Role of cancer stem cells in the development of giant cell tumor of bone. Cancer Cell Int 2020; 20:135. [PMID: 32351329 PMCID: PMC7183664 DOI: 10.1186/s12935-020-01218-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 04/17/2020] [Indexed: 02/06/2023] Open
Abstract
The primary bone tumor is usually observed in adolescence age group which has been shown to be part of nearly 20% of the sarcomas known today. Giant cell tumor of bone (GCTB) can be benign as well as malignant tumor which exhibits localized dynamism and is usually associated with the end point of a long bone. Giant cell tumor (GCT) involves mononuclear stromal cells which proliferate at a high rate, multinucleated giant cells and stromal cells are equally present in this type of tumor. Cancer stem cells (CSCs) have been confirmed to play a potential role in the development of GCT. Cancer stem cell-based microRNAs have been shown to contribute to a greater extent in giant cell tumor of bone. CSCs and microRNAs present in the tumors specifically are a great concern today which need in-depth knowledge as well as advanced techniques to treat the bone cancer effectively. In this review, we attempted to summarize the role played by cancer stem cells involving certain important molecules/factors such as; Mesenchymal Stem Cells (MSCs), miRNAs and signaling mechanism such as; mTOR/PI3K-AKT, towards the formation of giant cell tumor of bone, in order to get an insight regarding various effective strategies and research advancements to obtain adequate knowledge related to CSCs which may help to focus on highly effective treatment procedures for bone tumors.
Collapse
Affiliation(s)
- Abdul Rouf War
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
| | - Kai Dang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
| | - Shanfen Jiang
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
| | - Zhongwei Xiao
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399 People’s Republic of China
| | - Zhiping Miao
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
| | - Tuanmin Yang
- Honghui Hospital, Xi’an, Jiaotong University College of Medicine, Xi’an, Shaanxi China
| | - Yu Li
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
| | - Airong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
| |
Collapse
|
128
|
Kim GB, Shon OJ. Current perspectives in stem cell therapies for osteoarthritis of the knee. Yeungnam Univ J Med 2020; 37:149-158. [PMID: 32279478 PMCID: PMC7384917 DOI: 10.12701/yujm.2020.00157] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are emerging as an attractive option for osteoarthritis (OA) of the knee joint, due to their marked disease-modifying ability and chondrogenic potential. MSCs can be isolated from various organ tissues, such as bone marrow, adipose tissue, synovium, umbilical cord blood, and articular cartilage with similar phenotypic characteristics but different proliferation and differentiation potentials. They can be differentiated into a variety of connective tissues such as bone, adipose tissue, cartilage, intervertebral discs, ligaments, and muscles. Although several studies have reported on the clinical efficacy of MSCs in knee OA, the results lack consistency. Furthermore, there is no consensus regarding the proper cell dosage and application method to achieve the optimal effect of stem cells. Therefore, the purpose of this study is to review the characteristics of various type of stem cells in knee OA, especially MSCs. Moreover, we summarize the clinical issues faced during the application of MSCs.
Collapse
Affiliation(s)
- Gi Beom Kim
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Daegu, Korea
| | - Oog-Jin Shon
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
129
|
Song M, Zhang Z, Yan L, Liu D. [Effectiveness of arthroscopic microfracture combined with osteochondral autologous transplantation for large area cartilage injury of femoral condyle of knee]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2020; 34:330-334. [PMID: 32174078 DOI: 10.7507/1002-1892.201907048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To explore the effectiveness of arthroscopic microfracture combined with osteochondral autologous transplantation (OAT) in treatment of large area (4-6 cm 2) cartilage injury of the femoral condyle of knee. Methods Between March 2016 and June 2017, 22 patients of large area cartilage injury of the femoral condyle of knee were treated with arthroscopic microfracture combined with OAT. There were 16 males and 6 females with an average age of 22-60 years (mean, 38.6 years). The cause of injury was traffic accident in 8 cases and sports injuries in 14 cases. The disease duration was 1-6 months (mean, 3.4 months). There were 15 cases of medial femoral condyle injuries and 7 cases of lateral condyle injuries. The area of cartilage defect was 4-6 cm 2 (mean, 4.98 cm 2). According to the International Cartilage Repair Society (ICRS) classification, 9 cases were rated as grade Ⅲ and 13 cases as grade Ⅳ. Eighteen cases were combined with meniscus injuries. Preoperative visual analogue scale (VAS) score was 6.36±1.25 and Lysholm score was 36.00±7.77. Results All incisions healed by first intention. All patients were followed up 2-3 years with an average of 2.3 years. At 2 years after operation, the VAS score was 1.27±0.94 and the Lysholm score was 77.82±6.21, which were significantly improved when compared with those before operation ( t=16.595, P=0.000; t=21.895, P=0.000). At 2 years after operation, MRI showed that the cartilage defect was repaired well. Conclusion Arthroscopic microfracture combined with OAT can be used to treat large area cartilage injury of the femoral condyle of knee, and the good early effectiveness can be obtained.
Collapse
Affiliation(s)
- Meng Song
- Department of Orthopedic Surgery and Sports Medicine, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, 121001, P.R.China
| | - Zheng Zhang
- Department of Orthopedic Surgery and Sports Medicine, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, 121001, P.R.China
| | - Lixiang Yan
- Department of Orthopedic Surgery and Sports Medicine, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, 121001, P.R.China
| | - Danping Liu
- Department of Orthopedic Surgery and Sports Medicine, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou Liaoning, 121001, P.R.China
| |
Collapse
|
130
|
Shu CC, Zaki S, Ravi V, Schiavinato A, Smith MM, Little CB. The relationship between synovial inflammation, structural pathology, and pain in post-traumatic osteoarthritis: differential effect of stem cell and hyaluronan treatment. Arthritis Res Ther 2020; 22:29. [PMID: 32059749 PMCID: PMC7023816 DOI: 10.1186/s13075-020-2117-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/06/2020] [Indexed: 01/02/2023] Open
Abstract
Background Synovitis is implicated in the severity and progression of pain and structural pathology of osteoarthritis (OA). Increases in inflammatory or immune cell subpopulations including macrophages and lymphocytes have been reported in OA synovium, but how the particular subpopulations influence symptomatic or structural OA disease progression is unclear. Two therapies, hyaluronan (HA) and mesenchymal stem cells (MSCs), have demonstrated efficacy in some clinical settings: HA acting as device to improve joint function and provide pain relief, while MSCs may have immunomodulatory and disease-modifying effects. We used these agents to investigate whether changes in pain sensitization or structural damage were linked to modulation of the synovial inflammatory response in post-traumatic OA. Methods Skeletally mature C57BL6 male mice underwent medial-meniscal destabilisation (DMM) surgery followed by intra-articular injection of saline, a hyaluronan hexadecylamide derivative (Hymovis), bone marrow-derived stem cells (MSCs), or MSC + Hymovis. We quantified the progression of OA-related cartilage, subchondral bone and synovial histopathology, and associated pain sensitization (tactile allodynia). Synovial lymphocytes, monocyte/macrophages and their subpopulations were quantified by fluorescent-activated cell sorting (FACS), and the expression of key inflammatory mediators and catabolic enzyme genes quantified by real-time polymerase chain reaction (PCR). Results MSC but not Hymovis significantly reduced late-stage (12-week post-DMM) cartilage proteoglycan loss and structural damage. Allodynia was initially reduced by both treatments but significantly better at 8 and 12 weeks by Hymovis. Chondroprotection by MSCs was not associated with specific changes in synovial inflammatory cell populations but rather regulation of post-injury synovial Adamts4, Adamts5, Mmp3, and Mmp9 expression. Reduced acute post-injury allodynia with all treatments coincided with decreased synovial macrophage and T cell numbers, while longer-term effect on pain sensitization with Hymovis was associated with increased M2c macrophages. Conclusions This therapeutic study in mice demonstrated a poor correlation between cartilage, bone or synovium (histo)pathology, and pain sensitization. Changes in the specific synovial inflammatory cell subpopulations may be associated with chronic OA pain sensitization, and a novel target for symptomatic treatment.
Collapse
Affiliation(s)
- Cindy C Shu
- Raymond Purves Bone and Joint Laboratory, Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney, Level 10 Kolling Building - B6, Royal North Shore Hospital, St. Leonards, NSW, 2065, Australia
| | - Sanaa Zaki
- Raymond Purves Bone and Joint Laboratory, Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney, Level 10 Kolling Building - B6, Royal North Shore Hospital, St. Leonards, NSW, 2065, Australia
| | - Varshini Ravi
- Raymond Purves Bone and Joint Laboratory, Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney, Level 10 Kolling Building - B6, Royal North Shore Hospital, St. Leonards, NSW, 2065, Australia
| | | | - Margaret M Smith
- Raymond Purves Bone and Joint Laboratory, Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney, Level 10 Kolling Building - B6, Royal North Shore Hospital, St. Leonards, NSW, 2065, Australia
| | - Christopher B Little
- Raymond Purves Bone and Joint Laboratory, Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney, Level 10 Kolling Building - B6, Royal North Shore Hospital, St. Leonards, NSW, 2065, Australia.
| |
Collapse
|
131
|
Barrachina L, Cequier A, Romero A, Vitoria A, Zaragoza P, Vázquez FJ, Rodellar C. Allo-antibody production after intraarticular administration of mesenchymal stem cells (MSCs) in an equine osteoarthritis model: effect of repeated administration, MSC inflammatory stimulation, and equine leukocyte antigen (ELA) compatibility. Stem Cell Res Ther 2020; 11:52. [PMID: 32028995 PMCID: PMC7006079 DOI: 10.1186/s13287-020-1571-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/11/2019] [Accepted: 01/26/2020] [Indexed: 02/07/2023] Open
Abstract
Background Antibody production after allogeneic administration of mesenchymal stem cells (MSCs) could impact their clinical application. Proinflammatory priming of MSCs can potentiate their regulatory ability in vivo but increased expression of major histocompatibility complex (MHC) might augment their immunogenicity, potentially leading to immune memory thus limiting repeated allogeneic administration. This study aimed at evaluating the production of cytotoxic allo-antibodies directed against donor’s ELA (equine leukocyte antigen) in mismatched and halfmatched horses receiving repeated intraarticular administration of stimulated MSCs (MSC-primed) and unstimulated MSCs (MSC-naïve) in pathologic joints. Methods From available stored samples from a previous in vivo study, cells from one donor and serially collected sera (five time-points) from three groups of recipients were used based on their ELA haplotypes to perform microcytotoxicity assays: Group 1 recipients mismatched with the donor that received MSC-naïve (naïve-mismatched recipients); Group 2 recipients mismatched with the donor that received MSC-primed (primed-mismatched recipients); Group 3 recipients halfmatched with the donor (sharing 1/2 haplotypes) that received MSC-primed (primed-halfmatched recipients). Sera from recipients (neat, 1:2 and 1:16 dilution) were tested against target cells from the donor (cryopreserved and expanded MSC-naïve and MSC-primed) or from one animal presenting the same ELA haplotypes than the donor (fresh peripheral blood lymphocytes as control). Results One to three weeks after first MSC administration, all recipient groups produced allo-antibodies regardless of MSC received (naïve or primed) and matching degree with donor. However, secondary response after MSC re-exposure was less evident in halfmatched recipients (MSC-primed) than in mismatched ones (both MSC-naïve and MSC-primed). Recipients of MSC-primed (both mismatched and halfmatched) tended towards developing lower antibody response than MSC-naïve recipients in vivo, but MSC-primed were targeted to death in higher percentage in vitro in the microcytoxicity assay. Conclusions After first intraarticular allogeneic administration, the immunomodulatory profile of MSC-primed would have led to lower antibody production, but these antibodies would target more easily MSC-primed after second injection (re-exposure), likely because of their higher MHC expression. Electronic supplementary material The online version of this article (10.1186/s13287-020-1571-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura Barrachina
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza - CITA, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Servicio de Cirugía y Medicina Equina, Hospital Veterinario, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Alina Cequier
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza - CITA, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Antonio Romero
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Servicio de Cirugía y Medicina Equina, Hospital Veterinario, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Arantza Vitoria
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Servicio de Cirugía y Medicina Equina, Hospital Veterinario, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Pilar Zaragoza
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza - CITA, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Francisco José Vázquez
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain.,Servicio de Cirugía y Medicina Equina, Hospital Veterinario, Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain
| | - Clementina Rodellar
- Laboratorio de Genética Bioquímica LAGENBIO - Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza, C/Miguel Servet, 177, 50013, Zaragoza, Spain. .,Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza - CITA, C/Miguel Servet, 177, 50013, Zaragoza, Spain.
| |
Collapse
|
132
|
Dilogo IH, Canintika AF, Hanitya AL, Pawitan JA, Liem IK, Pandelaki J. Umbilical cord-derived mesenchymal stem cells for treating osteoarthritis of the knee: a single-arm, open-label study. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY AND TRAUMATOLOGY 2020; 30:799-807. [PMID: 31989258 DOI: 10.1007/s00590-020-02630-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Despite being a common cause of quality-of-life impairment, there are no efficacious therapies that could prevent the progression of knee osteoarthritis (KOA). We conducted an open-label trial of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) and hyaluronic acid (HA) for treating KOA. METHODS This open-label study was conducted from July 2015 to December 2018 at Cipto Mangunkusumo Hospital, Jakarta, Indonesia. Patients diagnosed with KOA were injected three times, comprising of 10 × 106 units of hUC-MSCs in 2-ml secretome implantation and 2-ml hyaluronic acid (HA) injection in the first week, followed with 2-ml HA injection twice in the second and third week. RESULTS Twenty-nine subjects (57 knees) were recruited. Seventeen (58.6%) subjects were male, and the mean age was 58.3 ± 9.6 years. Thirty-three (57.9%) knees were classified into Kellgren-Lawrence grade I-II KOA (mild OA). hUC-MSCs significantly decreased pain measured by visual analogue scale in severe KOA from initial to 6th month follow-up [5 ± 2.97 to 3.38 ± 2.44 (p = 0.035)]. The International Knee Documentation Committee score significantly increased at 6th month follow-up (53.26 ± 16.66 to 65.49 ± 13.01, p < 0.001, in subjects with grade I-II and 48.84 ± 18.41 to 61.83 ± 18.83, p = 0.008, in subjects with severe KOA). The Western Ontario and McMaster Universities Osteoarthritis decreased significantly in both groups from initial to 6th month follow-up (from 22.55 ± 15.94 to 13.23 ± 10.29, p = 0.003, and from 27.57 ± 15.99 to 17.92 ± 19.1, p = 0.003, in those with mild and severe KOA, respectively). CONCLUSIONS hUC-MSCs could be a potentially new regenerative treatment for KOA. The maximum effect of hUC-MSCs was achieved after 6 months of injection. LEVEL OF EVIDENCE Therapeutic level II.
Collapse
Affiliation(s)
- Ismail Hadisoebroto Dilogo
- Department of Orthopaedics and Traumatology, Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia. .,Stem Cell Medical Technology Integrated Medical Service Unit, Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia. .,Stem Cell and Tissue Engineering Research Center, IMERI, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
| | - Anissa Feby Canintika
- Department of Orthopaedics and Traumatology, Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Alberto Lastiko Hanitya
- Department of Orthopaedics and Traumatology, Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Jeanne Adiwinata Pawitan
- Stem Cell Medical Technology Integrated Medical Service Unit, Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Stem Cell and Tissue Engineering Research Center, IMERI, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Isabella Kurnia Liem
- Stem Cell Medical Technology Integrated Medical Service Unit, Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Stem Cell and Tissue Engineering Research Center, IMERI, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Jacub Pandelaki
- Department of Radiology, Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
133
|
Klontzas ME, Karantanas AH. Considerations on the Use of Ferumoxytol-enhanced MRI for Tracking Stem Cell Implants in Cartilage Defects. Radiology 2020; 294:238-239. [DOI: 10.1148/radiol.2019192103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Michail E. Klontzas
- Department of Medical Imaging, University Hospital, Voutes, 71110, Heraklion, Crete, Greece
- Department of Radiology, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Apostolos H. Karantanas
- Department of Medical Imaging, University Hospital, Voutes, 71110, Heraklion, Crete, Greece
- Department of Radiology, Medical School, University of Crete, Heraklion, Crete, Greece
- Advanced Hybrid Imaging Systems, Institute of Computer Science-FORTH, Crete, Greece
| |
Collapse
|
134
|
Oral administration of EP4-selective agonist KAG-308 suppresses mouse knee osteoarthritis development through reduction of chondrocyte hypertrophy and TNF secretion. Sci Rep 2019; 9:20329. [PMID: 31889132 PMCID: PMC6937271 DOI: 10.1038/s41598-019-56861-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/17/2019] [Indexed: 11/17/2022] Open
Abstract
Osteoarthritis (OA) is one of the world’s most common degenerative diseases, but there is no disease-modifying treatment available. Previous studies have shown that prostaglandin E2 (PGE2) and PGE2 receptor 4 (EP4) are involved in OA pathogenesis; however, their roles are not fully understood. Here, we examined the efficacy of oral administration of KAG-308, an EP4-selective agonist, in surgically induced mouse knee OA. Cartilage degeneration and synovitis were significantly inhibited by the KAG-308 treatment. Chondrocyte hypertrophy and expression of tumor necrosis factor alpha (TNF) and matrix metalloproteinase 13 (Mmp13) in the synovium were suppressed in the KAG-308-treated mice. In cultured chondrocytes, hypertrophic differentiation was inhibited by KAG-308 and intranuclear translocation of histone deacetylase 4 (Hdac4) was enhanced. In cultured synoviocytes, lipopolysaccharide (LPS)-induced expression of TNF and Mmp13 was also suppressed by KAG-308. KAG-308 was detected in the synovium and cartilage of orally treated mice. TNF secretion from the synovia of KAG-308-treated mice was significantly lower than control mice. Thus, we conclude that oral administration of KAG-308 suppresses OA development through suppression of chondrocyte hypertrophy and synovitis. KAG-308 may be a potent candidate for OA drug development.
Collapse
|
135
|
The shift in the balance between osteoblastogenesis and adipogenesis of mesenchymal stem cells mediated by glucocorticoid receptor. Stem Cell Res Ther 2019; 10:377. [PMID: 31805987 PMCID: PMC6896503 DOI: 10.1186/s13287-019-1498-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/09/2019] [Accepted: 11/18/2019] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells capable of differentiating into several tissues, such as bone, cartilage, and fat. Glucocorticoids affect a variety of biological processes such as proliferation, differentiation, and apoptosis of various cell types, including osteoblasts, adipocytes, or chondrocytes. Glucocorticoids exert their function by binding to the glucocorticoid receptor (GR). Physiological concentrations of glucocorticoids stimulate osteoblast proliferation and promote osteogenic differentiation of MSCs. However, pharmacological concentrations of glucocorticoids can not only induce apoptosis of osteoblasts and osteocytes but can also reduce proliferation and inhibit the differentiation of osteoprogenitor cells. Several signaling pathways, including the Wnt, TGFβ/BMP superfamily and Notch signaling pathways, transcription factors, post-transcriptional regulators, and other regulators, regulate osteoblastogenesis and adipogenesis of MSCs mediated by GR. These signaling pathways target key transcription factors, such as Runx2 and TAZ for osteogenesis and PPARγ and C/EBPs for adipogenesis. Glucocorticoid-induced osteonecrosis and osteoporosis are caused by various factors including dysfunction of bone marrow MSCs. Transplantation of MSCs is valuable in regenerative medicine for the treatment of osteonecrosis of the femoral head, osteoporosis, osteogenesis imperfecta, and other skeletal disorders. However, the mechanism of inducing MSCs to differentiate toward the osteogenic lineage is the key to an efficient treatment. Thus, a better understanding of the molecular mechanisms behind the imbalance between GR-mediated osteoblastogenesis and adipogenesis of MSCs would not only help us to identify the pathogenic causes of glucocorticoid-induced osteonecrosis and osteoporosis but also promote future clinical applications for stem cell-based tissue engineering and regenerative medicine. Here, we primarily review the signaling mechanisms involved in adipogenesis and osteogenesis mediated by GR and discuss the factors that control the adipo-osteogenic balance.
Collapse
|
136
|
Almeida B, Wang Y, Shukla A. Effects of Nanoparticle Properties on Kartogenin Delivery and Interactions with Mesenchymal Stem Cells. Ann Biomed Eng 2019; 48:2090-2102. [DOI: 10.1007/s10439-019-02430-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022]
|
137
|
Zhang H, Wang Y, Yang G, Yu H, Zhou Z, Tang M. MicroRNA-30a regulates chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells through targeting Sox9. Exp Ther Med 2019; 18:4689-4697. [PMID: 31807153 PMCID: PMC6878886 DOI: 10.3892/etm.2019.8148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 07/05/2019] [Indexed: 12/20/2022] Open
Abstract
Cartilage injury is difficult to repair since the cartilage tissue lacks self-restoration ability. Improved formation of chondrocytes differentiated from the mesenchymal stem cells (MSC) by genetic regulation is a potentially promising therapeutic option. SOX9 is a critical transcription factor for mesenchymal condensation prior to chondrogenesis. Previous studies demonstrated that several microRNAs (miRNAs or miRs) play a critical role in the chondrogenic differentiation of MSCs. However, the interactional relations between miR-30a and SOX9 during chondrogenic differentiation of MSCs need to be further elucidated. In the present study, human bone marrow-derived mesenchymal stem cells have been isolated and induced into chondrogenic differentiation to imitate the cartilage formation in vitro. Additionally, the expression levels of several miRNAs that were reported to interact with the SOX9 3'untranslated region (UTR) were examined by using reverse transcription-quantitative PCR. The interactional relations between candidate miRNAs and SOX9 were verified with the transfection of a miRNA mimic or inhibitor and a luciferase reporter gene assay. The results indicate that miR-30a and miR-195 were consistently increased during MSC chondrogenic differentiation. Additionally, the binding of miR-30a to the SOX9 3UTR was verified. Then, the authors upregulated the expression of miR-30a and found that MSC chondrogenic differentiation was inhibited. Taken together, the results of the present study demonstrate that miR-30a has a negative regulatory effect on MSC chondrogenic differentiation by targeting SOX9. Advances in epigenetic regulating methods will likely be the future of systemic treatment of cartilage injury.
Collapse
Affiliation(s)
- Hongqi Zhang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yunjia Wang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Guanteng Yang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Honggui Yu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhenhai Zhou
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Mingxing Tang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
138
|
Zhao Q, Zhang L, Wei Y, Yu H, Zou L, Huo J, Yang H, Song B, Wei T, Wu D, Zhang W, Zhang L, Liu D, Li Z, Chi Y, Han Z, Han Z. Systematic comparison of hUC-MSCs at various passages reveals the variations of signatures and therapeutic effect on acute graft-versus-host disease. Stem Cell Res Ther 2019; 10:354. [PMID: 31779707 PMCID: PMC6883552 DOI: 10.1186/s13287-019-1478-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/24/2019] [Accepted: 10/30/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells are heterogenous populations with hematopoietic supporting and immunomodulating capacities. Enormous studies have focused on their preclinical or clinical therapeutic effects, yet the systematic study of continuous in vitro passages on signatures and functions of UC-MSCs at both the cellular and molecular levels is still lacking. METHODS In this study, to systematically evaluate the biological properties of MSCs at various passages, we analyzed biomarker expression, cell proliferation and apoptosis, chromosome karyotype, and tri-lineage differentiation potential. Subsequently, we took advantage of whole-exome sequencing to compare the somatic hypermutation of hUC-MSCs at P3, P6, and P15 including SNV and INDEL mutations. In addition, to explore the safety of the abovementioned hUC-MSCs, we performed metabolic pathway enrichment analysis and in vivo transplantation analysis. Furthermore, we cocultured the abovementioned hUC-MSCs with UCB-CD34+ HSCs to evaluate their hematopoietic supporting capacity in vitro. Finally, we transplanted the cells into acute graft-versus-host disease (aGVHD) mice to further evaluate their therapeutic effect in vivo. RESULTS The hUC-MSCs at P3, P6, and P15 showed similar morphology, biomarker expression, and cytokine secretion. hUC-MSCs at P15 had advantages on adipogenic differentiation and some cytokine secretion such as IL-6 and VEGF, with disadvantages on cell proliferation, apoptosis, and osteogenic and chondrogenic differentiation potential. Based on the SNP data of 334,378 exons and bioinformatic analyses, we found the somatic point mutations could be divided into 96 subsets and formed 30 kinds of signatures but did not show correlation with risk of tumorigenesis, which was confirmed by the in vivo transplantation experiments. However, hUC-MSCs at P15 showed impaired hematologic supporting effect in vitro and declined therapeutic effect on aGVHD in vivo. CONCLUSIONS In this study, we systematically evaluated the biological and genetic properties of hUC-MSCs at various passages. Our findings have provided new references for safety and effectiveness assessments, which will provide overwhelming evidence for the safety of hUC-MSCs after continuous in vitro passages both at the cellular and molecular levels for the first time. Taken together, our studies could help understand the controversial effects of disease treatment and benefit the clinical research of UC-MSCs.
Collapse
Affiliation(s)
- Qinjun Zhao
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China.,National Stem Cell Engineering Research Center, Tianjin Ang-sai Stem Cell and Gene Technology Co., Ltd., Tianjin, 300450, China
| | - Leisheng Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China. .,National Stem Cell Engineering Research Center, Tianjin Ang-sai Stem Cell and Gene Technology Co., Ltd., Tianjin, 300450, China. .,The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China. .,The Enterprise Postdoctoral Working Station, Tianjin Chase Sun Pharmaceutical Co., Ltd., Tianjin, 301700, China. .,Precision Medicine Division, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd., Tianjin, 301700, China. .,Jiangxi Research Center of Stem Cell Engineering, Jiangxi Health-Biotech Stem Cell Technology Co., Ltd., Shangrao, 334000, China.
| | - Yimeng Wei
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Hao Yu
- National Stem Cell Engineering Research Center, Tianjin Ang-sai Stem Cell and Gene Technology Co., Ltd., Tianjin, 300450, China
| | - Linglin Zou
- Division of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Jiali Huo
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Hongju Yang
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Baoquan Song
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Teng Wei
- Cytotherapy Laboratory, Shenzhen People's Hospital & The second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Dan Wu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Wenxia Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Lei Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Dengke Liu
- The Enterprise Postdoctoral Working Station, Tianjin Chase Sun Pharmaceutical Co., Ltd., Tianjin, 301700, China
| | - Zongjin Li
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Ying Chi
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
| | - Zhibo Han
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China. .,National Stem Cell Engineering Research Center, Tianjin Ang-sai Stem Cell and Gene Technology Co., Ltd., Tianjin, 300450, China.
| | - Zhongchao Han
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China. .,National Stem Cell Engineering Research Center, Tianjin Ang-sai Stem Cell and Gene Technology Co., Ltd., Tianjin, 300450, China. .,Precision Medicine Division, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd., Tianjin, 301700, China.
| |
Collapse
|
139
|
Cheng BF, Lian JJ, Yang HJ, Wang L, Yu HH, Bi JJ, Gao YX, Chen SJ, Wang M, Feng ZW. Neural cell adhesion molecule regulates chondrocyte hypertrophy in chondrogenic differentiation and experimental osteoarthritis. Stem Cells Transl Med 2019; 9:273-283. [PMID: 31742919 PMCID: PMC6988767 DOI: 10.1002/sctm.19-0190] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/25/2019] [Indexed: 12/28/2022] Open
Abstract
Chondrocyte hypertrophy-like change is an important pathological process of osteoarthritis (OA), but the mechanism remains largely unknown. Neural cell adhesion molecule (NCAM) is highly expressed and involved in the chondrocyte differentiation of mesenchymal stem cells (MSCs). In this study, we found that NCAM deficiency accelerates chondrocyte hypertrophy in articular cartilage and growth plate of OA mice. NCAM deficiency leads to hypertrophic chondrocyte differentiation in both murine MSCs and chondrogenic cells, in which extracellular signal-regulated kinase (ERK) signaling plays an important role. Moreover, NCAM expression is downregulated in an interleukin-1β-stimulated OA cellular model and monosodium iodoacetate-induced OA rats. Overexpression of NCAM substantially inhibits hypertrophic differentiation in the OA cellular model. In conclusion, NCAM could inhibit hypertrophic chondrocyte differentiation of MSCs by inhibiting ERK signaling and reduce chondrocyte hypertrophy in experimental OA model, suggesting the potential utility of NCAM as a novel therapeutic target for alleviating chondrocyte hypertrophy of OA.
Collapse
Affiliation(s)
- Bin-Feng Cheng
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Jun-Jiang Lian
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China.,Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| | - Hai-Jie Yang
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Lei Wang
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Hao-Heng Yu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Jia-Jia Bi
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Yao-Xin Gao
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Su-Juan Chen
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Mian Wang
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Zhi-Wei Feng
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| |
Collapse
|
140
|
Abdelrazik H, Giordano E, Barbanti Brodano G, Griffoni C, De Falco E, Pelagalli A. Substantial Overview on Mesenchymal Stem Cell Biological and Physical Properties as an Opportunity in Translational Medicine. Int J Mol Sci 2019; 20:5386. [PMID: 31671788 PMCID: PMC6862078 DOI: 10.3390/ijms20215386] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 10/25/2019] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSC) have piqued worldwide interest for their extensive potential to treat a large array of clinical indications, their unique and controversial immunogenic and immune modulatory properties allowing ample discussions and debates for their possible applications. Emerging data demonstrating that the interaction of biomaterials and physical cues with MSC can guide their differentiation into specific cell lineages also provide new interesting insights for further MSC manipulation in different clinical applications. Moreover, recent discoveries of some regulatory molecules and signaling pathways in MSC niche that may regulate cell fate to distinct lineage herald breakthroughs in regenerative medicine. Although the advancement and success in the MSC field had led to an enormous increase in the amount of ongoing clinical trials, we still lack defined clinical therapeutic protocols. This review will explore the exciting opportunities offered by human and animal MSC, describing relevant biological properties of these cells in the light of the novel emerging evidence mentioned above while addressing the limitations and challenges MSC are still facing.
Collapse
Affiliation(s)
- Heba Abdelrazik
- Department of Clinical Pathology, Cairo University, Cairo 1137, Egypt.
- Department of Diagnosis, central laboratory department, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, 16131 Genoa, Italy.
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, 47522 Cesena, Italy.
| | - Giovanni Barbanti Brodano
- Department of Oncological and Degenerative Spine Surgery, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Cristiana Griffoni
- Department of Oncological and Degenerative Spine Surgery, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Elena De Falco
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy.
- Mediterranea Cardiocentro, 80122 Napoli, Italy.
| | - Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy.
- Institute of Biostructures and Bioimages (IBB), National Research Council (CNR), 80131 Naples, Italy.
| |
Collapse
|
141
|
Hayes AJ, Melrose J. Glycosaminoglycan and Proteoglycan Biotherapeutics in Articular Cartilage Protection and Repair Strategies: Novel Approaches to Visco‐supplementation in Orthobiologics. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Anthony J. Hayes
- Bioimaging Research HubCardiff School of BiosciencesCardiff University Cardiff CF10 3AX Wales UK
| | - James Melrose
- Graduate School of Biomedical EngineeringUNSW Sydney Sydney NSW 2052 Australia
- Raymond Purves Bone and Joint Research LaboratoriesKolling Institute of Medical ResearchRoyal North Shore Hospital and The Faculty of Medicine and HealthUniversity of Sydney St. Leonards NSW 2065 Australia
- Sydney Medical SchoolNorthernRoyal North Shore HospitalSydney University St. Leonards NSW 2065 Australia
| |
Collapse
|
142
|
Palomares Cabeza V, Hoogduijn MJ, Kraaijeveld R, Franquesa M, Witte-Bouma J, Wolvius EB, Farrell E, Brama PAJ. Pediatric Mesenchymal Stem Cells Exhibit Immunomodulatory Properties Toward Allogeneic T and B Cells Under Inflammatory Conditions. Front Bioeng Biotechnol 2019; 7:142. [PMID: 31245368 PMCID: PMC6581756 DOI: 10.3389/fbioe.2019.00142] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/28/2019] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells from pediatric patients (pMSCs) are an attractive cell source in regenerative medicine, due to their higher proliferation rates and better differentiation abilities compared to adult MSCs (aMSCs). We have previously characterized the immunomodulatory abilities of pMSCs on T cells under co-culture. It has also been reported that aMSCs can inhibit B cell proliferation and maturation under inflammatory conditions. In this study, we therefore aimed to clarify the immunomodulatory effect of pMSCs toward T and B cells in an inflammatory microenvironment. Bone marrow derived pMSCs were primed to simulate inflammatory conditions by exposure with 50 ng/mL of IFN-γ for 3 days. To analyze the interaction between pMSCs and T cells, CD3/CD28 stimulated peripheral blood mononuclear cells (PBMCs) were co-cultured with primed or unprimed pMSCs. To investigate B cell responses, quiescent B cells obtained from spleens by CD43 negative selection were stimulated with anti-IgM, anti-CD40, IL-2, and co-cultured with either IFN-γ primed or unprimed pMSC. pMSC phenotype, B and T cell proliferation, and B cell functionality were analyzed. Gene expression of indoleamine 2,3-dioxygenease (IDO), as well as the expression of HLA-ABC, HLA-DR and the co-stimulatory molecules CD80 and CD86 was upregulated on pMSCs upon IFN-γ priming. IFN-γ did not alter the immunomodulatory abilities of pMSCs upon CD4+ nor CD8+ stimulated T cells compared to unprimed pMSCs. IFN-γ primed pMSCs but not unprimed pMSCs strongly inhibited naïve (CD19+CD27−), memory (CD19+CD27+), and total B cell proliferation. Antibody-producing plasmablast (CD19+CD27highCD38high) formation and IgG production were also significantly inhibited by IFN-γ primed pMSCs compared to unprimed pMSCs. Collectively, these results show that pMSCs have immunomodulatory effects upon the adaptive immune response which can be potentiated by inflammatory stimuli. This knowledge is useful in regenerative medicine and allogeneic transplantation applications toward tailoring pMSCs function to best modulate the immune response for a successful implant engraftment and avoidance of a strong immune reaction.
Collapse
Affiliation(s)
- Virginia Palomares Cabeza
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, Netherlands.,Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands.,School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Martin Johannes Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Rens Kraaijeveld
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Marcella Franquesa
- REMAR Group and Nephrology Service, Germans Trias i Pujol Health Science Institute and University Hospital, Badalona, Spain
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eppo B Wolvius
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Pieter A J Brama
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
143
|
Pereira H, Fatih Cengiz I, Gomes S, Espregueira-Mendes J, Ripoll PL, Monllau JC, Reis RL, Oliveira JM. Meniscal allograft transplants and new scaffolding techniques. EFORT Open Rev 2019; 4:279-295. [PMID: 31210969 PMCID: PMC6549113 DOI: 10.1302/2058-5241.4.180103] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Clinical management of meniscal injuries has changed radically in recent years. We have moved from the model of systematic tissue removal (meniscectomy) to understanding the need to preserve the tissue.Based on the increased knowledge of the basic science of meniscal functions and their role in joint homeostasis, meniscus preservation and/or repair, whenever indicated and possible, are currently the guidelines for management.However, when repair is no longer possible or when facing the fact of the previous partial, subtotal or total loss of the meniscus, meniscus replacement has proved its clinical value. Nevertheless, meniscectomy remains amongst the most frequent orthopaedic procedures.Meniscus replacement is currently possible by means of meniscal allograft transplantation (MAT) which provides replacement of the whole meniscus with or without bone plugs/slots. Partial replacement has been achieved by means of meniscal scaffolds (mainly collagen or polyurethane-based). Despite the favourable clinical outcomes, it is still debatable whether MAT is capable of preventing progression to osteoarthritis. Moreover, current scaffolds have shown some fundamental limitations, such as the fact that the newly formed tissue may be different from the native fibrocartilage of the meniscus.Regenerative tissue engineering strategies have been used in an attempt to provide a new generation of meniscal implants, either for partial or total replacement. The goal is to provide biomaterials (acellular or cell-seeded constructs) which provide the biomechanical properties but also the biological features to replace the loss of native tissue. Moreover, these approaches include possibilities for patient-specific implants of correct size and shape, as well as advanced strategies combining cells, bioactive agents, hydrogels or gene therapy.Herein, the clinical evidence and tips concerning MAT, currently available meniscus scaffolds and future perspectives are discussed. Cite this article: EFORT Open Rev 2019;4 DOI: 10.1302/2058-5241.4.180103.
Collapse
Affiliation(s)
- Hélder Pereira
- Orthopedic Department of Póvoa de Varzim - Vila do Conde Hospital Centre, Vila do Conde, Portugal
- Ripoll y De Prado Sports Clinic, Murcia-Madrid, FIFA Medical Centre of Excellence, Madrid, Spain
- International Centre of Sports Traumatology of the Ave, Vila do Conde, Portugal
- 3Bs Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal
- ICVS/3Bs, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ibrahim Fatih Cengiz
- 3Bs Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal
- ICVS/3Bs, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sérgio Gomes
- International Centre of Sports Traumatology of the Ave, Vila do Conde, Portugal
| | - João Espregueira-Mendes
- 3Bs Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal
- ICVS/3Bs, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Clínica do Dragão, Espregueira-Mendes Sports Centre, FIFA Medical Centre of Excellence, Porto, Portugal
- Orthopedic Department, University of Minho, Braga, Portugal
| | - Pedro L. Ripoll
- Ripoll y De Prado Sports Clinic, Murcia-Madrid, FIFA Medical Centre of Excellence, Madrid, Spain
| | - Joan C. Monllau
- Orthopaedic Department, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Rui L. Reis
- 3Bs Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal
- ICVS/3Bs, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Barco, Guimarães, Portugal
| | - J. Miguel Oliveira
- 3Bs Research Group, I3Bs, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal
- ICVS/3Bs, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Orthopaedic Department, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, Spain
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Barco, Guimarães, Portugal
| |
Collapse
|
144
|
Wang AT, Feng Y, Jia HH, Zhao M, Yu H. Application of mesenchymal stem cell therapy for the treatment of osteoarthritis of the knee: A concise review. World J Stem Cells 2019; 11:222-235. [PMID: 31110603 PMCID: PMC6503460 DOI: 10.4252/wjsc.v11.i4.222] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/07/2019] [Accepted: 03/16/2019] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) refers to a chronic joint disease characterized by degenerative changes of articular cartilage and secondary bone hyperplasia. Since articular cartilage has a special structure, namely the absence of blood vessels as well as the low conversion rate of chondrocytes in the cartilage matrix, the treatment faces numerous clinical challenges. Traditional OA treatment (e.g., arthroscopic debridement, microfracture, autologous or allogeneic cartilage transplantation, chondrocyte transplantation) is primarily symptomatic treatment and pain management, which cannot contribute to regenerating degenerated cartilage or reducing joint inflammation. Also, the generated mixed fibrous cartilage tissue is not the same as natural hyaline cartilage. Mesenchymal stem cells (MSCs) have turned into the most extensively explored new therapeutic drugs in cell-based OA treatment as a result of their ability to differentiate into chondrocytes and their immunomodulatory properties. In this study, the preliminary results of preclinical (OA animal model)/clinical trials regarding the effects of MSCs on cartilage repair of knee joints are briefly summarized, which lay a solid application basis for more and deeper clinical studies on cell-based OA treatment.
Collapse
Affiliation(s)
- Ai-Tong Wang
- Cell Products of National Engineering Research Center, National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Ying Feng
- Cell Products of National Engineering Research Center, National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Hong-Hong Jia
- Cell Products of National Engineering Research Center, National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Meng Zhao
- Cell Products of National Engineering Research Center, National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Hao Yu
- Cell Products of National Engineering Research Center, National Stem Cell Engineering Research Center, Tianjin 300457, China
| |
Collapse
|
145
|
Chen Y, Wu T, Huang S, Suen CWW, Cheng X, Li J, Hou H, She G, Zhang H, Wang H, Zheng X, Zha Z. Sustained Release SDF-1α/TGF-β1-Loaded Silk Fibroin-Porous Gelatin Scaffold Promotes Cartilage Repair. ACS APPLIED MATERIALS & INTERFACES 2019; 11:14608-14618. [PMID: 30938503 DOI: 10.1021/acsami.9b01532] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Continuous delivery of growth factors to the injury site is crucial to creating a favorable microenvironment for cartilage injury repair. In the present study, we fabricated a novel sustained-release scaffold, stromal-derived factor-1α (SDF-1α)/transforming growth factor-β1 (TGF-β1)-loaded silk fibroin-porous gelatin scaffold (GSTS). GSTS persistently releases SDF-1α and TGF-β1, which enhance cartilage repair by facilitating cell homing and chondrogenic differentiation. Scanning electron microscopy showed that GSTS is a porous microstructure and the protein release assay demonstrated the sustainable release of SDF-1α and TGF-β1 from GSTS. Bone marrow-derived mesenchymal stem cells (MSCs) maintain high in vitro cell activity and excellent cell distribution and phenotype after seeding into GSTS. Furthermore, MSCs acquired enhanced chondrogenic differentiation capability in the TGF-β1-loaded scaffolds (GSTS or GST: loading TGF-β1 only) and the conditioned medium from SDF-1α-loaded scaffolds (GSTS or GSS: loading SDF-1α only) effectively promoted MSCs migration. GSTS was transplanted into the osteochondral defects in the knee joint of rats, and it could promote cartilage regeneration and repair the cartilage defects at 12 weeks after transplantation. Our study shows that GSTS can facilitate in vitro MSCs homing, migration, chondrogenic differentiation and SDF-1α and TGF-β1 have a synergistic effect on the promotion of in vivo cartilage forming. This SDF-1α and TGF-β1 releasing GSTS have promising therapeutic potential in cartilage repair.
Collapse
Affiliation(s)
- Yuanfeng Chen
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital , Jinan University , Guangzhou 510630 , P. R. China
| | - Tingting Wu
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital , Jinan University , Guangzhou 510630 , P. R. China
| | - Shusen Huang
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital , Jinan University , Guangzhou 510630 , P. R. China
| | - Chun-Wai Wade Suen
- Department of Genetics , University of Cambridge , Cambridge CB2 3EH , United Kingdom
| | - Xin Cheng
- Department of Histology and Embryology, Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College , Jinan University , Guangzhou 510632 , Guangdong , P. R. China
| | - Jieruo Li
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital , Jinan University , Guangzhou 510630 , P. R. China
| | - Huige Hou
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital , Jinan University , Guangzhou 510630 , P. R. China
| | - Guorong She
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital , Jinan University , Guangzhou 510630 , P. R. China
| | - Huantian Zhang
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital , Jinan University , Guangzhou 510630 , P. R. China
| | - Huajun Wang
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital , Jinan University , Guangzhou 510630 , P. R. China
| | - Xiaofei Zheng
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital , Jinan University , Guangzhou 510630 , P. R. China
| | - Zhengang Zha
- Institute of Orthopedic Diseases and Center for Joint Surgery and Sports Medicine, The First Affiliated Hospital , Jinan University , Guangzhou 510630 , P. R. China
| |
Collapse
|
146
|
Chahal J, Gómez-Aristizábal A, Shestopaloff K, Bhatt S, Chaboureau A, Fazio A, Chisholm J, Weston A, Chiovitti J, Keating A, Kapoor M, Ogilvie-Harris DJ, Syed KA, Gandhi R, Mahomed NN, Marshall KW, Sussman MS, Naraghi AM, Viswanathan S. Bone Marrow Mesenchymal Stromal Cell Treatment in Patients with Osteoarthritis Results in Overall Improvement in Pain and Symptoms and Reduces Synovial Inflammation. Stem Cells Transl Med 2019; 8:746-757. [PMID: 30964245 PMCID: PMC6646697 DOI: 10.1002/sctm.18-0183] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 02/13/2019] [Indexed: 12/14/2022] Open
Abstract
Patients with late‐stage Kellgren‐Lawrence knee osteoarthritis received a single intra‐articular injection of 1, 10, or 50 million bone marrow mesenchymal stromal cells (BM‐MSCs) in a phase I/IIa trial to assess safety and efficacy using a broad toolset of analytical methods. Besides safety, outcomes included patient‐reported outcome measures (PROMs): Knee Injury and Osteoarthritis Outcome Score (KOOS) and Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC); contrast‐enhanced magnetic resonance imaging (MRI) for cartilage morphology (Whole Organ MRI Scores [WORMS]), collagen content (T2 scores), and synovitis; and inflammation and cartilage turnover biomarkers, all over 12 months. BM‐MSCs were characterized by a panel of anti‐inflammatory markers to predict clinical efficacy. There were no serious adverse events, although four patients had minor, transient adverse events. There were significant overall improvements in KOOS pain, symptoms, quality of life, and WOMAC stiffness relative to baseline; the 50 million dose achieved clinically relevant improvements across most PROMs. WORMS and T2 scores did not change relative to baseline. However, cartilage catabolic biomarkers and MRI synovitis were significantly lower at higher doses. Pro‐inflammatory monocytes/macrophages and interleukin 12 levels decreased in the synovial fluid after MSC injection. The panel of BM‐MSC anti‐inflammatory markers was strongly predictive of PROMs over 12 months. Autologous BM‐MSCs are safe and result in significant improvements in PROMs at 12 months. Our analytical tools provide important insights into BM‐MSC dosing and BM‐MSC reduction of synovial inflammation and cartilage degradation and provide a highly predictive donor selection criterion that will be critical in translating MSC therapy for osteoarthritis. stem cells translational medicine2019;8:746&757
Collapse
Affiliation(s)
- Jaskarndip Chahal
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Alejandro Gómez-Aristizábal
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Cell Therapy Program, University Health Network, Toronto, Ontario, Canada
| | - Konstantin Shestopaloff
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Shashank Bhatt
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Cell Therapy Program, University Health Network, Toronto, Ontario, Canada
| | - Amélie Chaboureau
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Cell Therapy Program, University Health Network, Toronto, Ontario, Canada
| | - Antonietta Fazio
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jolene Chisholm
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Cell Therapy Program, University Health Network, Toronto, Ontario, Canada
| | - Amanda Weston
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Julia Chiovitti
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Armand Keating
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Cell Therapy Program, University Health Network, Toronto, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Mohit Kapoor
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Darrell J Ogilvie-Harris
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Khalid A Syed
- Arthritis Program, University Health Network, Toronto, Ontario, Canada
| | - Rajiv Gandhi
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Nizar N Mahomed
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kenneth W Marshall
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Marshall S Sussman
- Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Ali M Naraghi
- Joint Department of Medical Imaging, University Health Network, Toronto, Ontario, Canada
| | - Sowmya Viswanathan
- Arthritis Program, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Cell Therapy Program, University Health Network, Toronto, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
147
|
Chen Z, Luo X, Zhao X, Yang M, Wen C. Label-free cell sorting strategies via biophysical and biochemical gradients. J Orthop Translat 2019; 17:55-63. [PMID: 31194093 PMCID: PMC6551360 DOI: 10.1016/j.jot.2019.01.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/23/2019] [Accepted: 01/28/2019] [Indexed: 01/08/2023] Open
Abstract
Isolating active mesenchymal stem cells from a heterogeneous population is an essential step that determines the efficacy of stem cell therapy such as for osteoarthritis. Nowadays, the gold standard of cell sorting, fluorescence-activated cell sorting, relies on labelling surface markers via antibody-antigen reaction. However, sorting stem cells with high stemness usually requires the labelling of multiple biomarkers. Moreover, the labelling process is costly, and the high operating pressure is harmful to cell functionality and viability. Although label-free cell sorting, based on physical characteristics, has gained increasing interest in the past decades, it has not shown the ability to eliminate stem cells with low stemness. Cell motility, as a novel sorting marker, is hence proposed for label-free sorting active stem cells. Accumulating evidence has demonstrated the feasibility in manipulating directional cell migration through patterning the biophysical, biochemical or both gradients of the extracellular matrix. However, applying those findings to label-free cell sorting has not been well discussed and studied. This review thus first provides a brief overview about the effect of biophysical and biochemical gradients of the extracellular matrix on cell migration. State-of-the-art fabrication techniques for generating such gradients of hydrogels are then introduced. Among current research, the authors suggest that hydrogels with dual-gradients of biochemistry and biophysics are potential tools for accurate label-free cell sorting with satisfactory selectivity and efficiency. TRANSLATIONAL POTENTIAL OF THIS ARTICLE The reviewed label-free cell sorting approaches enable us to isolate active cell for cytotherapy. The proposed system can be further modified for single-cell analysis and drug screening.
Collapse
Affiliation(s)
| | | | | | | | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
148
|
Xie ZY, Wang P, Wu YF, Shen HY. Long non-coding RNA: The functional regulator of mesenchymal stem cells. World J Stem Cells 2019; 11:167-179. [PMID: 30949295 PMCID: PMC6441937 DOI: 10.4252/wjsc.v11.i3.167] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/07/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a subset of multipotent stroma cells residing in various tissues of the body. Apart from supporting the hematopoietic stem cell niche, MSCs possess strong immunoregulatory ability and multiple differentiation potentials. These powerful capacities allow the extensive application of MSCs in clinical practice as an effective treatment for diseases. Therefore, illuminating the functional mechanism of MSCs will help to improve their curative effect and promote their clinical use. Long noncoding RNA (LncRNA) is a novel class of noncoding RNA longer than 200 nt. Recently, multiple studies have demonstrated that LncRNA is widely involved in growth and development through controlling the fate of cells, including MSCs. In this review, we highlight the role of LncRNA in regulating the functions of MSCs and discuss their participation in the pathogenesis of diseases and clinical use in diagnosis and treatment.
Collapse
Affiliation(s)
- Zhong-Yu Xie
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Peng Wang
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| | - Yan-Feng Wu
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong Province, China
| | - Hui-Yong Shen
- Department of Orthopedics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, Guangdong Province, China
| |
Collapse
|
149
|
Foppiani EM, Candini O, Mastrolia I, Murgia A, Grisendi G, Samarelli AV, Boscaini G, Pacchioni L, Pinelli M, De Santis G, Horwitz EM, Veronesi E, Dominici M. Impact of HOXB7 overexpression on human adipose-derived mesenchymal progenitors. Stem Cell Res Ther 2019; 10:101. [PMID: 30890185 PMCID: PMC6423808 DOI: 10.1186/s13287-019-1200-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 02/04/2019] [Accepted: 03/01/2019] [Indexed: 01/02/2023] Open
Abstract
Background The ex vivo expansion potential of mesenchymal stromal/stem cells (MSC) together with their differentiation and secretion properties makes these cells an attractive tool for transplantation and tissue engineering. Although the use of MSC is currently being tested in a growing number of clinical trials, it is still desirable to identify molecular markers that may help improve their performance both in vitro and after transplantation. Methods Recently, HOXB7 was identified as a master player driving the proliferation and differentiation of bone marrow mesenchymal progenitors. In this study, we investigated the effect of HOXB7 overexpression on the ex vivo features of adipose mesenchymal progenitors (AD-MSC). Results HOXB7 increased AD-MSC proliferation potential, reduced senescence, and improved chondrogenesis together with a significant increase of basic fibroblast growth factor (bFGF) secretion. Conclusion While further investigations and in vivo models shall be applied for better understanding, these data suggest that modulation of HOXB7 may be a strategy for innovative tissue regeneration applications.
Collapse
Affiliation(s)
- Elisabetta Manuela Foppiani
- Division of Oncology, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41100, Modena, Italy.,Rigenerand srl, Modena, Medolla, Italy
| | - Olivia Candini
- Division of Oncology, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41100, Modena, Italy.,Rigenerand srl, Modena, Medolla, Italy
| | - Ilenia Mastrolia
- Division of Oncology, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41100, Modena, Italy
| | - Alba Murgia
- Division of Oncology, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41100, Modena, Italy
| | - Giulia Grisendi
- Division of Oncology, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41100, Modena, Italy.,Rigenerand srl, Modena, Medolla, Italy
| | - Anna Valeria Samarelli
- Division of Oncology, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41100, Modena, Italy
| | - Giulia Boscaini
- Division of Plastic Surgery, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Lucrezia Pacchioni
- Division of Plastic Surgery, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Pinelli
- Division of Plastic Surgery, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Giorgio De Santis
- Division of Plastic Surgery, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Edwin M Horwitz
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University Department of Pediatrics, Atlanta, GA, USA
| | | | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Via del Pozzo, 71, 41100, Modena, Italy. .,Rigenerand srl, Modena, Medolla, Italy.
| |
Collapse
|
150
|
Mehranfar S, Abdi Rad I, Mostafavi E, Akbarzadeh A. The use of stromal vascular fraction (SVF), platelet-rich plasma (PRP) and stem cells in the treatment of osteoarthritis: an overview of clinical trials. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:882-890. [DOI: 10.1080/21691401.2019.1576710] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Sahar Mehranfar
- Department of Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Isa Abdi Rad
- Department of Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Ebrahim Mostafavi
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Abolfazl Akbarzadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|