1
|
Li HZ, Zhang JL, Yuan DL, Xie WQ, Ladel CH, Mobasheri A, Li YS. Role of signaling pathways in age-related orthopedic diseases: focus on the fibroblast growth factor family. Mil Med Res 2024; 11:40. [PMID: 38902808 PMCID: PMC11191355 DOI: 10.1186/s40779-024-00544-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Fibroblast growth factor (FGF) signaling encompasses a multitude of functions, including regulation of cell proliferation, differentiation, morphogenesis, and patterning. FGFs and their receptors (FGFR) are crucial for adult tissue repair processes. Aberrant FGF signal transduction is associated with various pathological conditions such as cartilage damage, bone loss, muscle reduction, and other core pathological changes observed in orthopedic degenerative diseases like osteoarthritis (OA), intervertebral disc degeneration (IVDD), osteoporosis (OP), and sarcopenia. In OA and IVDD pathologies specifically, FGF1, FGF2, FGF8, FGF9, FGF18, FGF21, and FGF23 regulate the synthesis, catabolism, and ossification of cartilage tissue. Additionally, the dysregulation of FGFR expression (FGFR1 and FGFR3) promotes the pathological process of cartilage degradation. In OP and sarcopenia, endocrine-derived FGFs (FGF19, FGF21, and FGF23) modulate bone mineral synthesis and decomposition as well as muscle tissues. FGF2 and other FGFs also exert regulatory roles. A growing body of research has focused on understanding the implications of FGF signaling in orthopedic degeneration. Moreover, an increasing number of potential targets within the FGF signaling have been identified, such as FGF9, FGF18, and FGF23. However, it should be noted that most of these discoveries are still in the experimental stage, and further studies are needed before clinical application can be considered. Presently, this review aims to document the association between the FGF signaling pathway and the development and progression of orthopedic diseases. Besides, current therapeutic strategies targeting the FGF signaling pathway to prevent and treat orthopedic degeneration will be evaluated.
Collapse
Affiliation(s)
- Heng-Zhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing-Lve Zhang
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Dong-Liang Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Wen-Qing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | | | - Ali Mobasheri
- Faculty of Medicine, Research Unit of Health Sciences and Technology, University of Oulu, 90014, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406, Vilnius, Lithuania.
- Department of Rheumatology and Clinical Immunology, Universitair Medisch Centrum Utrecht, Utrecht, 3508, GA, the Netherlands.
- Department of Joint Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000, Liège, Belgium.
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
2
|
Esmaeili A, Hosseini S, Baghaban Eslaminejad M. Co-culture engineering: a promising strategy for production of engineered extracellular vesicle for osteoarthritis treatment. Cell Commun Signal 2024; 22:29. [PMID: 38200606 PMCID: PMC10782541 DOI: 10.1186/s12964-023-01467-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
The therapeutic effects of extracellular vesicles (EVs) have been identified as a significant factor in intercellular communication in different disease treatments, including osteoarthritis (OA). Compared to the conventional approaches in treating OA, EV therapy is a non-invasive and cell-free method. However, improving the yield of EVs and their therapeutic effects are the main challenges for clinical applications. In this regard, researchers are using the EV engineering potential to overcome these challenges. New findings suggest that the co-culture strategy as an indirect EV engineering method efficiently increases EV production and quality. The co-culture of mesenchymal stem cells (MSCs) and chondrocytes has improved their chondrogenesis, anti-inflammatory effects, and regenerative properties which are mediated by EVs. Hence, co-culture engineering by considerable systems could be useful in producing engineered EVs for different therapeutic applications. Here, we review various co-culture approaches, including diverse direct and indirect, 2D and 3D cell cultures, as well as static and dynamic systems. Meanwhile, we suggest and discuss the advantages of combined strategies to achieve engineered EVs for OA treatment.
Collapse
Affiliation(s)
- Abazar Esmaeili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
3
|
Ehioghae M, Vippa TK, Askins D, Slusarczyk S, Bobo E, Montoya A, Anderson D, Robinson CL, Kaye AD, Urits I. Exploring Orthopedic Stem-Cell Approaches for Osteoarthritis Management: Current Trends and Future Horizons. Curr Pain Headache Rep 2024; 28:27-35. [PMID: 38010488 DOI: 10.1007/s11916-023-01191-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE OF REVIEW Osteoarthritis (OA) is a prevalent and debilitating condition characterized by joint degeneration and pain. Current treatment options aim to alleviate symptoms and slow disease progression but lack curative potential. Stem cell therapies have emerged as a promising alternative. This article explores the epidemiology, pathophysiology, clinical manifestations of hip and knee OA, and the evolving role of stem cell therapies in their treatment. RECENT FINDINGS The global prevalence of OA, with knee OA being the most common form, has fueled the demand for stem cell therapies. Despite limited robust evidence supporting their efficacy, clinical trials investigating stem-cell treatments for OA have reported encouraging radiological and clinical improvements. Stem cell therapies offer potential disease-modifying benefits through immunomodulatory actions, growth factor secretion, and chondrogenic capabilities. Adipose-derived mesenchymal stem cells (ADMSCs) have shown promise in clinical trials for OA treatment, offering potential pain relief and functional improvement. ADMSCs possess advantages such as accessibility and a favorable safety profile, making them a viable option for OA management. Although other stem-cell types, including human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs), have been used in OA treatment, ADMSCs have demonstrated superior outcomes. By providing a comprehensive overview of the evolving landscape of stem cell therapies for hip and knee OA, this article highlights the potential of stem-cell treatments to address the limitations of current therapies. However, further research is required to establish their long-term efficacy, identify optimal stem-cell types, and develop standardized protocols.
Collapse
Affiliation(s)
| | | | | | | | - Elena Bobo
- Tulane University School of Medicine, New Orleans, USA
| | - Alexis Montoya
- University of Arizona College of Medicine-Phoenix, Phoenix, USA
| | | | | | - Alan D Kaye
- Louisiana State University Health Shreveport, Shreveport, USA
| | | |
Collapse
|
4
|
Wan Z, Chen YF, Pan Q, Wang Y, Yuan S, Chin HY, Wu HH, Lin WT, Cheng PY, Yang YJ, Wang YF, Kumta SM, Lee CW, Lee OKS. Single-cell transcriptome analysis reveals the effectiveness of cytokine priming irrespective of heterogeneity in mesenchymal stromal cells. Cytotherapy 2023; 25:1155-1166. [PMID: 37715776 DOI: 10.1016/j.jcyt.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/10/2023] [Accepted: 08/19/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) are recognized as a potential cell-based therapy for regenerative medicine. Short-term inflammatory cytokine pre-stimulation (cytokine priming) is a promising approach to enhance regenerative efficacy of MSCs. However, it is unclear whether their intrinsic heterogenic nature causes an unequal response to cytokine priming, which might blunt the accessibility of clinical applications. METHODS In this study, by analyzing the single-cell transcriptomic landscape of human bone marrow MSCs from a naïve to cytokine-primed state, we elucidated the potential mechanism of superior therapeutic potential in cytokine-primed MSCs. RESULTS We found that cytokine-primed MSCs had a distinct transcriptome landscape. Although substantial heterogeneity was identified within the population in both naïve and primed states, cytokine priming enhanced the several characteristics of MSCs associated with therapeutic efficacy irrespective of heterogeneity. After cytokine-priming, all sub-clusters of MSCs possessed high levels of immunoregulatory molecules, trophic factors, stemness-related genes, anti-apoptosis markers and low levels of multi-lineage and senescence signatures, which are critical for their therapeutic potency. CONCLUSIONS In conclusion, our results provide new insights into MSC heterogeneity under cytokine stimulation and suggest that cytokine priming reprogrammed MSCs independent of heterogeneity.
Collapse
Affiliation(s)
- Zihao Wan
- Department of Orthopaedics and Limb Reconstruction/Paediatric Orthopaedics, South China Hospital of Shenzhen University, Shenzhen, China; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China; Hospital Authority, Hong Kong SAR, China
| | - Yu-Fan Chen
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung, Taiwan; Department of Biomedical Engineering, China Medical University, Taichung, Taiwan
| | - Qi Pan
- Department of Orthopaedics and Limb Reconstruction/Paediatric Orthopaedics, South China Hospital of Shenzhen University, Shenzhen, China
| | - Yiwei Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Hui Yen Chin
- Hong Kong Hub of Paediatric Excellence, Hong Kong Children's Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hao-Hsiang Wu
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Ting Lin
- Doctoral Degree Program of Translational Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Po-Yu Cheng
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung, Taiwan
| | - Yun-Jung Yang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Fan Wang
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shekhar Madhukar Kumta
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chien-Wei Lee
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung, Taiwan; Department of Biomedical Engineering, China Medical University, Taichung, Taiwan.
| | - Oscar Kuang-Sheng Lee
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung, Taiwan; Doctoral Degree Program of Translational Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Orthopedics, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
5
|
Joshi JM, Muttigi MS, Upadhya R, Seetharam RN. An overview of the current advances in the treatment of inflammatory diseases using mesenchymal stromal cell secretome. Immunopharmacol Immunotoxicol 2023:1-11. [PMID: 36786742 DOI: 10.1080/08923973.2023.2180388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
The growing interest in mesenchymal stromal cell (MSC) therapy has been leading to the utilization of its therapeutic properties in a variety of inflammatory diseases. The clinical translation of the related research from bench to bedside is cumbersome due to some obvious limitations of cell therapy. It is evident from the literature that the MSC secretome components mediate their wide range of functions. Cell-free therapy using MSC secretome is being considered as an emerging and promising area of biotherapeutics. The secretome mainly consists of bioactive factors, free nucleic acids, and extracellular vesicles. Constituents of the secretome are greatly influenced by the cell's microenvironment. The broad array of immunomodulatory properties of MSCs are now being employed to target inflammatory diseases. This review focuses on the emerging MSC secretome therapies for various inflammatory diseases. The mechanism of action of the various anti-inflammatory factors is discussed. The potential of MSC secretome as a viable anti-inflammatory therapy is deliberated.
Collapse
Affiliation(s)
- Jahnavy Madhukar Joshi
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Manjunatha S Muttigi
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Raghavendra Upadhya
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Raviraja N Seetharam
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
6
|
Vonk LA. Potency Assay Considerations for Cartilage Repair, Osteoarthritis and Use of Extracellular Vesicles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:59-80. [PMID: 37258784 DOI: 10.1007/978-3-031-30040-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Articular cartilage covers the ends of bones in synovial joints acting as a shock absorber that helps movement of bones. Damage of the articular cartilage needs treatment as it does not repair itself and the damage can progress to osteoarthritis. In osteoarthritis all the joint tissues are involved with characteristic progressive cartilage degradation and inflammation. Autologous chondrocyte implantation is a well-proven cell-based treatment for cartilage defects, but a main downside it that it requires two surgeries. Multipotent, aka mesenchymal stromal cell (MSC)-based cartilage repair has gained attention as it can be used as a one-step treatment. It is proposed that a combination of immunomodulatory and regenerative capacities make MSC attractive for the treatment of osteoarthritis. Furthermore, since part of the paracrine effects of MSCs are attributed to extracellular vesicles (EVs), small membrane enclosed particles secreted by cells, EVs are currently being widely investigated for their potential therapeutic effects. Although MSCs have entered clinical cartilage treatments and EVs are used in in vivo efficacy studies, not much attention has been given to determine their potency and to the development of potency assays. This chapter provides considerations and suggestions for the development of potency assays for the use of MSCs and MSC-EVs for the treatment of cartilage defects and osteoarthritis.
Collapse
Affiliation(s)
- Lucienne A Vonk
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
7
|
Rojas-Murillo JA, Simental-Mendía MA, Moncada-Saucedo NK, Delgado-Gonzalez P, Islas JF, Roacho-Pérez JA, Garza-Treviño EN. Physical, Mechanical, and Biological Properties of Fibrin Scaffolds for Cartilage Repair. Int J Mol Sci 2022; 23:ijms23179879. [PMID: 36077276 PMCID: PMC9456199 DOI: 10.3390/ijms23179879] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Articular cartilage is a highly organized tissue that provides remarkable load-bearing and low friction properties, allowing for smooth movement of diarthrodial joints; however, due to the avascular, aneural, and non-lymphatic characteristics of cartilage, joint cartilage has self-regeneration and repair limitations. Cartilage tissue engineering is a promising alternative for chondral defect repair. It proposes models that mimic natural tissue structure through the use of cells, scaffolds, and signaling factors to repair, replace, maintain, or improve the specific function of the tissue. In chondral tissue engineering, fibrin is a biocompatible biomaterial suitable for cell growth and differentiation with adequate properties to regenerate damaged cartilage. Additionally, its mechanical, biological, and physical properties can be enhanced by combining it with other materials or biological components. This review addresses the biological, physical, and mechanical properties of fibrin as a biomaterial for cartilage tissue engineering and as an element to enhance the regeneration or repair of chondral lesions.
Collapse
Affiliation(s)
- Juan Antonio Rojas-Murillo
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, NL, Mexico
| | - Mario A. Simental-Mendía
- Servicio de Ortopedia y Traumatología, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, NL, Mexico
| | - Nidia K. Moncada-Saucedo
- Departamento de Hematología, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo León, Monterrey 64460, NL, Mexico
| | - Paulina Delgado-Gonzalez
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, NL, Mexico
| | - José Francisco Islas
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, NL, Mexico
| | - Jorge A. Roacho-Pérez
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, NL, Mexico
| | - Elsa N. Garza-Treviño
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, NL, Mexico
- Correspondence: ; Tel.: +52-81-83294173
| |
Collapse
|
8
|
Mechanically Derived Tissue Stromal Vascular Fraction Acts Anti-inflammatory on TNF Alpha-Stimulated Chondrocytes In Vitro. Bioengineering (Basel) 2022; 9:bioengineering9080345. [PMID: 35892757 PMCID: PMC9332748 DOI: 10.3390/bioengineering9080345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Enzymatically isolated stromal vascular fraction (SVF) has already shown to be effective as a treatment for osteoarthritis (OA). Yet, the use of enzymes for clinical purpose is highly regulated in many countries. Mechanical preparation of SVF results in a tissue-like SVF (tSVF) containing intact cell−cell connections including extracellular matrix (ECM) and is therefore less regulated. The purpose of this study was to investigate the immunomodulatory and pro-regenerative effect of tSVF on TNFα-stimulated chondrocytes in vitro. tSVF was mechanically derived using the Fractionation of Adipose Tissue (FAT) procedure. Characterization of tSVF was performed, e.g., cellular composition based on CD marker expression, colony forming unit and differentiation capacity after enzymatic dissociation (from heron referred to as tSVF-derived cells). Different co-cultures of tSVF-derived cells and TNFα-stimulated chondrocytes were analysed based on the production of sulphated glycosaminoglycans and the anti-inflammatory response of chondrocytes. Characterization of tSVF-derived cells mainly contained ASCs, endothelial cells, leukocytes and supra-adventitial cells. tSVF-derived cells were able to form colonies and differentiate into multiple cell lineages. Co-cultures with chondrocytes resulted in a shift of the ratio between tSVF cells: chondrocytes, in favor of chondrocytes alone (p < 0.05), and IL-1β and COX2 gene expression was upregulated in TNFα-treated chondrocytes. After treatment with (a conditioned medium of) tSVF-derived cells, IL-1β and COX2 gene expression was significantly reduced (p < 0.01). These results suggest mechanically derived tSVF stimulates chondrocyte proliferation while preserving the function of chondrocytes. Moreover, tSVF suppresses TNFα-stimulated chondrocyte inflammation in vitro. This pro-regenerative and anti-inflammatory effect shows the potential of tSVF as a treatment for osteoarthritis.
Collapse
|
9
|
Anti-hypertrophic effect of synovium-derived stromal cells on costal chondrocytes promotes cartilage repairs. J Orthop Translat 2021; 32:59-68. [PMID: 34934627 PMCID: PMC8645424 DOI: 10.1016/j.jot.2021.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 01/08/2023] Open
Abstract
Background Costal chondrocytes (CCs), as a promising donor cell source for cell-based therapy for cartilage repair, have strong tendency of hypertrophy and calcification, which limited CCs from further application in cartilage regenerative medicine. Synovium-derived stromal cells (SDSCs), have shown their beneficial effect for chondrocytes to maintain phenotype. This study aims to investigate whether SDSCs could help CCs to maintain chondrogenic phenotype and suppress hypertrophic differentiation in cartilage repairs. Methods CCs were directly cocultured with SDSCs in pellet or indirectly cocultured using a conditioned medium in vitro for 3 weeks. Cartilage matrix formation and hypertrophic differentiation of CCs were analyzed by RT-PCR, biochemical assays, and histological staining. Cocultured pellets were implanted into the osteochondral defects made on the femoral groove of the rats. Then, macroscopic and histological evaluations were performed. Results Pellets formed by CCs alone and CCs cocultured with SDSCs reveal equal cartilage matrix deposition. However, the gene expression of type X collagen was significantly downregulated in cocultured pellets. Immunohistochemistry analysis revealed suppressed expression of type X collagen in cocultured pellets, indicating SDSCs may suppress hypertrophic differentiation of chondrocytes. Further in indirect coculture experiment, SDSCs suppressed type X collagen expression as well and promoted the proliferation of CCs, indicating SDSCs may influence CCs by paracrine mechanism. The pellets implanted in the osteochondral defects showed good restoration effects, whereas the grafts constructed with CCs and SDSCs showed lower type X expression levels. Conclusion These results suggest that SDSCs may maintain the phenotype of CCs and prevent the hypertrophic differentiation of CCs in cartilage repair. The Translational Potential of this Article: CCs is a promising donor cell source for cell-based therapy for cartilage repair. Based on our study, cocultured with SDSCs weakened the tendency of hypertrophy and calcification of CCs, which provide a potential usage of SDSCs in CCs-based cartilage repair therapy to suppress newly formed cartilage calcification and improve clinical outcomes.
Collapse
|
10
|
Owida HA, Kuiper NL, Yang Y. Maintenance and Acceleration of Pericellular Matrix Formation within 3D Cartilage Cell Culture Models. Cartilage 2021; 13:847S-861S. [PMID: 31455088 PMCID: PMC8804781 DOI: 10.1177/1947603519870839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE In native articular cartilage, chondrocytes are surrounded by a thin pericellular matrix (PCM) forming chondrons. The PCM is exclusively rich in type VI collagen. The retention of the PCM has a significant influence on the metabolic activity of the chondrocytes. DESIGN This study investigated the influence of 2 hydrogels (hyaluronic acid [HA] and agarose) and 2 media compositions (basal and chondrogenic) on the preservation/maintenance and acceleration of PCM formation over a 21-day time course. Different combinations of chondrocytes, chondrons, and mesenchymal stem cells (MSCs) were studied. RESULTS Both hydrogels preserved chondrons PCM from day 1 up to 21-day culture regardless of media composition. Type VI collagen immunostaining of the cultured chondrons appeared both dense and homogenous. The presence of MSCs did not influence this outcome. At day 1, type VI collagen was not present around chondrocytes alone or their co-culture with MSCs. In the HA hydrogel, type VI collagen was located within the PCM after 7 days in both mono- and co-cultures. In the agarose hydrogel, collagen VI was located within the PCM at 7 days (co-cultures) and 14 days (monocultures). In both hydrogel systems, chondrogenic media enhanced the production of key extracellular matrix components in both mono- and co-cultures in comparison to basal media (11.5% and 14% more in glycosaminoglycans and type II collagen for chondrocytes samples at day 21 culture samples, respectively). However, the media types did not enhance type VI collagen synthesis. CONCLUSION Altogether, a 3D chondrogenic hydrogel environment is the primary condition for maintenance and acceleration of PCM formation.
Collapse
Affiliation(s)
- Hamza A. Owida
- Institute of Science & Technology in
Medicine, School of Medicine, University of Keele, Stoke-on-Trent, UK
| | - Nicola L. Kuiper
- Institute of Science & Technology in
Medicine, School of Medicine, University of Keele, Stoke-on-Trent, UK,Arthritis Research Centre, Robert Jones
& Agnes Hunt Orthopaedic Hospital, Oswestry, UK
| | - Ying Yang
- Institute of Science & Technology in
Medicine, School of Medicine, University of Keele, Stoke-on-Trent, UK,Ying Yang, Institute of Science &
Technology in Medicine, School of Medicine, University of Keele, Stoke-on-Trent
ST4 7QB, UK.
| |
Collapse
|
11
|
Paggi CA, Dudakovic A, Fu Y, Garces CG, Hevesi M, Galeano Garces D, Dietz AB, van Wijnen AJ, Karperien M. Autophagy Is Involved in Mesenchymal Stem Cell Death in Coculture with Chondrocytes. Cartilage 2021; 13:969S-979S. [PMID: 32693629 PMCID: PMC8721613 DOI: 10.1177/1947603520941227] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Cartilage formation is stimulated in mixtures of chondrocytes and human adipose-derived mesenchymal stromal cells (MSCs) both in vitro and in vivo. During coculture, human MSCs perish. The goal of this study is to elucidate the mechanism by which adipose tissue-derived MSC cell death occurs in the presence of chondrocytes. METHODS Human primary chondrocytes were cocultured with human MSCs derived from 3 donors. The cells were cultured in monoculture or coculture (20% chondrocytes and 80% MSCs) in pellets (200,000 cells/pellet) for 7 days in chondrocyte proliferation media in hypoxia (2% O2). RNA sequencing was performed to assess for differences in gene expression between monocultures or coculture. Immune fluorescence assays were performed to determine the presence of caspase-3, LC3B, and P62. RESULTS RNA sequencing revealed significant upregulation of >90 genes in the 3 cocultures when compared with monocultures. STRING analysis showed interconnections between >50 of these genes. Remarkably, 75% of these genes play a role in cell death pathways such as apoptosis and autophagy. Immunofluorescence shows a clear upregulation of the autophagic machinery with no substantial activation of the apoptotic pathway. CONCLUSION In cocultures of human MSCs with primary chondrocytes, autophagy is involved in the disappearance of MSCs. We propose that this sacrificial cell death may contribute to the trophic effects of MSCs on cartilage formation.
Collapse
Affiliation(s)
- Carlo Alberto Paggi
- Department of Developmental
BioEngineering, University of Twente, Enschede, Netherlands,Department of Orthopedic Surgery, Mayo
Clinic, Rochester, MN, USA,Department of Biochemistry and Molecular
Biology, Mayo Clinic, Rochester, MN, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo
Clinic, Rochester, MN, USA,Department of Biochemistry and Molecular
Biology, Mayo Clinic, Rochester, MN, USA
| | - Yao Fu
- Department of Developmental
BioEngineering, University of Twente, Enschede, Netherlands
| | | | - Mario Hevesi
- Department of Orthopedic Surgery, Mayo
Clinic, Rochester, MN, USA
| | | | - Allan B. Dietz
- Department of Laboratory Medicine and
Pathology, Mayo Clinic, Rochester, MN, USA
| | - Andre J. van Wijnen
- Department of Orthopedic Surgery, Mayo
Clinic, Rochester, MN, USA,Department of Biochemistry and Molecular
Biology, Mayo Clinic, Rochester, MN, USA,Andre J. van Wijnen, Department of
Orthopedic Surgery, Mayo Clinic, 200 First Street SW, MedSci 3-69, Rochester, MN
5590, USA.
| | - Marcel Karperien
- Department of Developmental
BioEngineering, University of Twente, Enschede, Netherlands,Marcel Karperien, Department of
Developmental BioEngineering, University of Twente, 7522 NB, Enschede,
Netherlands.
| |
Collapse
|
12
|
Matheakakis A, Batsali A, Papadaki HA, Pontikoglou CG. Therapeutic Implications of Mesenchymal Stromal Cells and Their Extracellular Vesicles in Autoimmune Diseases: From Biology to Clinical Applications. Int J Mol Sci 2021; 22:10132. [PMID: 34576296 PMCID: PMC8468750 DOI: 10.3390/ijms221810132] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are perivascular multipotent stem cells originally identified in the bone marrow (BM) stroma and subsequently in virtually all vascularized tissues. Because of their ability to differentiate into various mesodermal lineages, their trophic properties, homing capacity, and immunomodulatory functions, MSCs have emerged as attractive candidates in tissue repair and treatment of autoimmune disorders. Accumulating evidence suggests that the beneficial effects of MSCs may be primarily mediated via a number of paracrine-acting soluble factors and extracellular vesicles (EVs). EVs are membrane-coated vesicles that are increasingly being acknowledged as playing a key role in intercellular communication via their capacity to carry and deliver their cargo, consisting of proteins, nucleic acids, and lipids to recipient cells. MSC-EVs recapitulate the functions of the cells they originate, including immunoregulatory effects but do not seem to be associated with the limitations and concerns of cell-based therapies, thereby emerging as an appealing alternative therapeutic option in immune-mediated disorders. In the present review, the biology of MSCs will be outlined and an overview of their immunomodulatory functions will be provided. In addition, current knowledge on the features of MSC-EVs and their immunoregulatory potential will be summarized. Finally, therapeutic applications of MSCs and MSC-EVs in autoimmune disorders will be discussed.
Collapse
Affiliation(s)
- Angelos Matheakakis
- Department of Hematology, School of Medicine, University of Crete, 71500 Heraklion, Greece; (A.M.); (H.A.P.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Aristea Batsali
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Helen A. Papadaki
- Department of Hematology, School of Medicine, University of Crete, 71500 Heraklion, Greece; (A.M.); (H.A.P.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Charalampos G. Pontikoglou
- Department of Hematology, School of Medicine, University of Crete, 71500 Heraklion, Greece; (A.M.); (H.A.P.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| |
Collapse
|
13
|
Bartolotti I, Roseti L, Petretta M, Grigolo B, Desando G. A Roadmap of In Vitro Models in Osteoarthritis: A Focus on Their Biological Relevance in Regenerative Medicine. J Clin Med 2021; 10:1920. [PMID: 33925222 PMCID: PMC8124812 DOI: 10.3390/jcm10091920] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a multifaceted musculoskeletal disorder, with a high prevalence worldwide. Articular cartilage and synovial membrane are among the main biological targets in the OA microenvironment. Gaining more knowledge on the accuracy of preclinical in vitro OA models could open innovative avenues in regenerative medicine to bridge major gaps, especially in translation from animals to humans. Our methodological approach entailed searches on Scopus, the Web of Science Core Collection, and EMBASE databases to select the most relevant preclinical in vitro models for studying OA. Predicting the biological response of regenerative strategies requires developing relevant preclinical models able to mimic the OA milieu influencing tissue responses and organ complexity. In this light, standard 2D culture models lack critical properties beyond cell biology, while animal models suffer from several limitations due to species differences. In the literature, most of the in vitro models only recapitulate a tissue compartment, by providing fragmented results. Biotechnological advances may enable scientists to generate new in vitro models that combine easy manipulation and organ complexity. Here, we review the state-of-the-art of preclinical in vitro models in OA and outline how the different preclinical systems (inflammatory/biomechanical/microfluidic models) may be valid tools in regenerative medicine, describing their pros and cons. We then discuss the prospects of specific and combinatorial models to predict biological responses following regenerative approaches focusing on mesenchymal stromal cells (MSCs)-based therapies to reduce animal testing.
Collapse
Affiliation(s)
- Isabella Bartolotti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Livia Roseti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Mauro Petretta
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
- RegenHu Company, Z.I Du Vivier 22, 1690 Villaz-St-Pierre, Switzerland
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Giovanna Desando
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| |
Collapse
|
14
|
Zhuang WZ, Lin YH, Su LJ, Wu MS, Jeng HY, Chang HC, Huang YH, Ling TY. Mesenchymal stem/stromal cell-based therapy: mechanism, systemic safety and biodistribution for precision clinical applications. J Biomed Sci 2021; 28:28. [PMID: 33849537 PMCID: PMC8043779 DOI: 10.1186/s12929-021-00725-7] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are a promising resource for cell-based therapy because of their high immunomodulation ability, tropism towards inflamed and injured tissues, and their easy access and isolation. Currently, there are more than 1200 registered MSC clinical trials globally. However, a lack of standardized methods to characterize cell safety, efficacy, and biodistribution dramatically hinders the progress of MSC utility in clinical practice. In this review, we summarize the current state of MSC-based cell therapy, focusing on the systemic safety and biodistribution of MSCs. MSC-associated risks of tumor initiation and promotion and the underlying mechanisms of these risks are discussed. In addition, MSC biodistribution methodology and the pharmacokinetics and pharmacodynamics of cell therapies are addressed. Better understanding of the systemic safety and biodistribution of MSCs will facilitate future clinical applications of precision medicine using stem cells.
Collapse
Affiliation(s)
- Wei-Zhan Zhuang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.,TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Yi-Heng Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.,Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, 10041, Taiwan.,Department of Obstetrics and Gynecology, National Taiwan University Hospital Yunlin Branch, Yunlin, 64041, Taiwan
| | - Long-Jyun Su
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106, Taiwan
| | - Meng-Shiue Wu
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Han-Yin Jeng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.,TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Huan-Cheng Chang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106, Taiwan.,Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 106, Taiwan
| | - Yen-Hua Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan. .,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan. .,TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan. .,International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan. .,Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11031, Taiwan. .,Comprehensive Cancer Center of Taipei Medical University, Taipei, 11031, Taiwan. .,The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10617, Taiwan. .,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 100, Taiwan.
| |
Collapse
|
15
|
Mukhamedshina Y, Zhuravleva M, Sergeev M, Zakirova E, Gracheva O, Mukhutdinova D, Rizvanov A. Improving Culture Conditions, Proliferation, and Migration of Porcine Mesenchymal Stem Cells on Spinal Cord Contusion Injury Model in vitro. Cells Tissues Organs 2021; 209:236-247. [PMID: 33508824 DOI: 10.1159/000511865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/22/2020] [Indexed: 11/19/2022] Open
Abstract
Adipose tissue-derived mesenchymal stem cells (AD-MSCs) are promising for cell therapy in spinal cord injury (SCI). The pig is one of the most approximate models of many human diseases, including SCI. In our study, we selected the optimal conditions for the culture of porcine AD-MSCs and developed an in vitro SCI model based on the culture of cells in injured spinal cord extracts (SCE) 3 days and 6 weeks after SCI. We show that Dulbecco's Modified Eagle Medium (DMEM) with 20% serum content, supplemented with a combination of 5 mM L-ascorbate-2-phosphate and nonessential amino acids, stimulated a typical fibroblast-like morphology and high proliferation of porcine AD-MSCs. SCE caused a higher proliferation of porcine AD-MSCs compared with extracts from an intact spinal cord. The optimal proliferating effect was achieved using rostral 3 days SCE, and proliferation was lower in caudal and central SCE. Porcine AD-MSCs migration to the 3 days and 6 weeks SCE was higher than to an intact one and preferred the rostral SCE, avoiding central and caudal SCE. We also studied 13 cytokines contained in SCE but did not observe any definite relationship between some analyte concentrations and a change in the behavior of AD-MSCs.
Collapse
Affiliation(s)
- Yana Mukhamedshina
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation, .,Department of Histology, Cytology, and Embryology, Kazan State Medical University, Kazan, Russian Federation,
| | - Margarita Zhuravleva
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Mikhail Sergeev
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation.,Department of Veterinary Surgery, Obstetrics and Small Animal Pathology, Kazan State Academy of Veterinary, Kazan, Russian Federation
| | - Elena Zakirova
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Olga Gracheva
- Department of Therapy and Clinical Diagnostics with Radiology, Kazan State Academy of Veterinary, Kazan, Russian Federation
| | - Dina Mukhutdinova
- Department of Therapy and Clinical Diagnostics with Radiology, Kazan State Academy of Veterinary, Kazan, Russian Federation
| | - Albert Rizvanov
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| |
Collapse
|
16
|
Apelgren P, Amoroso M, Säljö K, Lindahl A, Brantsing C, Stridh Orrhult L, Markstedt K, Gatenholm P, Kölby L. Long-term in vivo integrity and safety of 3D-bioprinted cartilaginous constructs. J Biomed Mater Res B Appl Biomater 2021; 109:126-136. [PMID: 32633102 DOI: 10.1002/jbm.b.34687] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/31/2020] [Accepted: 06/23/2020] [Indexed: 11/07/2022]
Abstract
Long-term stability and biological safety are crucial for translation of 3D-bioprinting technology into clinical applications. Here, we addressed the long-term safety and stability issues associated with 3D-bioprinted constructs comprising a cellulose scaffold and human cells (chondrocytes and stem cells) over a period of 10 months in nude mice. Our findings showed that increasing unconfined compression strength over time significantly improved the mechanical stability of the cell-containing constructs relative to cell-free scaffolds. Additionally, the cell-free constructs exhibited a mean compressive stress and stiffness (compressive modulus) of 0.04 ± 0.05 MPa and 0.14 ± 0.18 MPa, respectively, whereas these values for the cell-containing constructs were 0.11 ± 0.08 MPa (p = .019) and 0.53 ± 0.59 MPa (p = .012), respectively. Moreover, histomorphologic analysis revealed that cartilage formed from the cell-containing constructs harbored an abundance of proliferating chondrocytes in clusters, and after 10 months, resembled native cartilage. Furthermore, extension of the experiment over the complete lifecycle of the animal model revealed no signs of ossification, fibrosis, necrosis, or implant-related tumor development in the 3D-bioprinted constructs. These findings confirm the in vivo biological safety and mechanical stability of 3D-bioprinted cartilaginous tissues and support their potential translation into clinical applications.
Collapse
Affiliation(s)
- Peter Apelgren
- Department of Plastic Surgery, Sahlgrenska University Hospital, University of Gothenburg, The Sahlgrenska Academy, Institute of Clinical Sciences, Göteborg, Sweden
| | - Matteo Amoroso
- Department of Plastic Surgery, Sahlgrenska University Hospital, University of Gothenburg, The Sahlgrenska Academy, Institute of Clinical Sciences, Göteborg, Sweden
| | - Karin Säljö
- Department of Plastic Surgery, Sahlgrenska University Hospital, University of Gothenburg, The Sahlgrenska Academy, Institute of Clinical Sciences, Göteborg, Sweden
| | - Anders Lindahl
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Camilla Brantsing
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Linnéa Stridh Orrhult
- 3D Bioprinting Centre, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Göteborg, Sweden
| | - Kajsa Markstedt
- 3D Bioprinting Centre, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Göteborg, Sweden
| | - Paul Gatenholm
- 3D Bioprinting Centre, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Göteborg, Sweden
| | - Lars Kölby
- Department of Plastic Surgery, Sahlgrenska University Hospital, University of Gothenburg, The Sahlgrenska Academy, Institute of Clinical Sciences, Göteborg, Sweden
| |
Collapse
|
17
|
Fu Y, Paggi CA, Dudakovic A, van Wijnen AJ, Post JN, Karperien M. Engineering Cartilage Tissue by Co-culturing of Chondrocytes and Mesenchymal Stromal Cells. Methods Mol Biol 2021; 2221:53-70. [PMID: 32979198 DOI: 10.1007/978-1-0716-0989-7_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Co-culture of chondrocytes and mesenchymal stromal cells (MSCs) has been shown to be beneficial in engineering cartilage tissue in vitro. In these co-cultures, MSCs increase the proliferation and matrix deposition of chondrocytes. The MSCs accomplish this beneficial effect by so-called trophic actions. Thus, large cartilage constructs can be made with a relatively small number of chondrocytes. In this chapter, we describe different methods for making co-cultures of MSCs and chondrocytes. We also provide detailed protocols for analyzing MSC-chondrocyte co-cultures with cell tracking, proliferation assays, species-specific polymerase chain reactions (PCR), rheological analysis, compression analysis, RNA-sequencing analysis, short tandem repeats analysis, and biochemical examination.
Collapse
Affiliation(s)
- Yao Fu
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, Enschede, The Netherlands
| | - Carlo A Paggi
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, Enschede, The Netherlands
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Janine N Post
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, Enschede, The Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
18
|
Chen Y, Ouyang X, Wu Y, Guo S, Xie Y, Wang G. Co-culture and Mechanical Stimulation on Mesenchymal Stem Cells and Chondrocytes for Cartilage Tissue Engineering. Curr Stem Cell Res Ther 2020; 15:54-60. [PMID: 31660820 DOI: 10.2174/1574888x14666191029104249] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/09/2019] [Accepted: 09/18/2019] [Indexed: 02/08/2023]
Abstract
Defects in articular cartilage injury and chronic osteoarthritis are very widespread and common, and the ability of injured cartilage to repair itself is limited. Stem cell-based cartilage tissue engineering provides a promising therapeutic option for articular cartilage damage. However, the application of the technique is limited by the number, source, proliferation, and differentiation of stem cells. The co-culture of mesenchymal stem cells and chondrocytes is available for cartilage tissue engineering, and mechanical stimulation is an important factor that should not be ignored. A combination of these two approaches, i.e., co-culture of mesenchymal stem cells and chondrocytes under mechanical stimulation, can provide sufficient quantity and quality of cells for cartilage tissue engineering, and when combined with scaffold materials and cytokines, this approach ultimately achieves the purpose of cartilage repair and reconstruction. In this review, we focus on the effects of co-culture and mechanical stimulation on mesenchymal stem cells and chondrocytes for articular cartilage tissue engineering. An in-depth understanding of the impact of co-culture and mechanical stimulation of mesenchymal stem cells and chondrocytes can facilitate the development of additional strategies for articular cartilage tissue engineering.
Collapse
Affiliation(s)
- Yawen Chen
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Xinli Ouyang
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Yide Wu
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Shaojia Guo
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Yongfang Xie
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Guohui Wang
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| |
Collapse
|
19
|
Xiao S, Chen L. The emerging landscape of nanotheranostic-based diagnosis and therapy for osteoarthritis. J Control Release 2020; 328:817-833. [PMID: 33176171 DOI: 10.1016/j.jconrel.2020.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a common degenerative disease involving numerous joint tissues and cells, with a growing rate in prevalence that ultimately results in a negative social impact. Early diagnosis, OA progression monitoring and effective treatment are of significant importance in halting OA process. However, traditional imaging techniques lack sensitivity and specificity, which lead to a delay in timely clinical intervention. Additionally, current treatments only slow the progression of OA but have not meet the largely medical need for disease-modifying therapy. In order to overcome the above-mentioned problems and improve clinical efficacy, nanotheranostics has been proposed on OA remedy, which has confirmed success in animal models. In this review, different imaging targets-based nanoprobe for early and timely OA diagnosis is first discussed. Second, therapeutic strategies delivered by nanosystem are summarized as much as possible. Their advantages and the potential for clinical translation are detailed discussed. Third, nanomedicine simultaneously combined with the imaging for OA treatment is introduced. Nanotheranostics dynamically tracked the OA treatment outcomes to timely and individually adjust therapy. Finally, future prospects and challenges of nanotechnology-based OA diagnosis, imaging and treatment are concluded and predicted. It is believed that nanoprobe and nanomedicine will become prospective in OA therapeutic revolution.
Collapse
Affiliation(s)
- Shuyi Xiao
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Liang Chen
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China.
| |
Collapse
|
20
|
Schivo S, Khurana S, Govindaraj K, Scholma J, Kerkhofs J, Zhong L, Huang X, van de Pol J, Langerak R, van Wijnen AJ, Geris L, Karperien M, Post JN. ECHO, the executable CHOndrocyte: A computational model to study articular chondrocytes in health and disease. Cell Signal 2020; 68:109471. [DOI: 10.1016/j.cellsig.2019.109471] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/01/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
|
21
|
You Q, Liu Z, Zhang J, Shen M, Li Y, Jin Y, Liu Y. Human Amniotic Mesenchymal Stem Cell Sheets Encapsulating Cartilage Particles Facilitate Repair of Rabbit Osteochondral Defects. Am J Sports Med 2020; 48:599-611. [PMID: 31940211 DOI: 10.1177/0363546519897912] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Human amniotic mesenchymal stem cells (hAMSCs) are being widely applied in various fields. Therefore, hAMSCs represent a promising candidate to facilitate cartilage regeneration. Nonetheless, no studies have investigated the application of hAMSC sheets to repair cartilage defects in vivo. PURPOSE To evaluate hAMSC sheets encapsulating cartilage particles to promote repair of rabbit osteochondral defects. STUDY DESIGN Controlled laboratory study. METHODS hAMSC sheets were constructed with passage 3 hAMSCs. The phenotypic and structural characteristics of hAMSC sheets were evaluated by flow cytometry and scanning electron microscopy, respectively. The potential for chondrogenic differentiation of hAMSC sheets was assessed by cartilage-specific marker staining, immunohistochemistry, and mRNA and protein expression (SOX9, COLII, and ACAN). Osteochondral defects (diameter, 3.5 mm; depth, 3 mm) were created in the left patellar grooves of 20 New Zealand White rabbits (female or male). The defects were treated with hAMSC sheet/cartilage particles (n = 5), cartilage particles (n = 5), hAMSC sheets (n = 5), or fibrin glue (n = 5). Macroscopic and histological evaluations of the regenerated tissue were conducted after 3 months. The survival time and differentiation of transplanted hAMSCs in the defect area were evaluated by immunofluorescence. RESULTS hAMSC sheets had a multilayered structure, with cells stacked layer by layer. Importantly, hAMSC sheets highly expressed phenotypic markers of mesenchymal stem cells. Cartilage-specific marker staining and immunohistochemistry were positive, and mRNA and protein expression was higher in the chondrogenically induced hAMSC sheet group than in the hAMSC sheet group (P < .05). hAMSC sheet/cartilage particles formed a large amount of hyaline-like cartilage in the defect area. In addition, macroscopic and histological scores were significantly higher than those in the other groups. Integration with surrounding normal cartilage and subchondral bone regeneration in the hAMSC sheet/cartilage particles group were better when compared with the other groups. A large number of human nuclear-specific antigen-positive cells were observed in the defect area of hAMSC sheet/cartilage particles and hAMSC sheet groups. Moreover, some positive cells expressed SOX9. CONCLUSION hAMSC sheets encapsulating cartilage particles facilitate osteochondral defect repair. CLINICAL RELEVANCE Delivery of cells in the form of a cell sheet in conjunction with cartilage particles provides a novel approach for cell-based cartilage regeneration.
Collapse
Affiliation(s)
- Qi You
- First Department of Orthopedics, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Ziming Liu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Jun Zhang
- First Department of Orthopedics, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Mengjie Shen
- Hospital of Stomatology, Zunyi Medical University, Zunyi, China
| | - Yuwan Li
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Jin
- First Department of Orthopedics, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Yi Liu
- First Department of Orthopedics, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
22
|
Implantation of mesenchymal stem cells in combination with allogenic cartilage improves cartilage regeneration and clinical outcomes in patients with concomitant high tibial osteotomy. Knee Surg Sports Traumatol Arthrosc 2020; 28:544-554. [PMID: 31549208 DOI: 10.1007/s00167-019-05729-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/16/2019] [Indexed: 01/22/2023]
Abstract
PURPOSE This study aimed to compare the clinical, radiological, and second-look arthroscopic outcomes of implanting mesenchymal stem cells (MSCs) alone and together with allogenic cartilage in patients treated with concomitant high tibial oteotomy (HTO) for varus knee osteoarthritis. METHODS Eighty patients treated with cartilage repair procedures and concomitant HTO were prospectively randomized into two groups: MSC implantation (MSC group), and MSC implantation with allogenic cartilage (MSC-AC group). Clinical outcomes were evaluated using the Lysholm Score and the Knee Injury and Osteoarthritis Outcome Score (KOOS) at preoperative and every follow-up visit. Radiological outcomes were evaluated by measuring the femorotibial angle and posterior tibial slope. During second-look arthroscopy, cartilage regeneration was evaluated according to the Kanamiya grade. RESULTS Clinical outcomes at the second-look arthroscopy (mean 12.5 months [MSC group] and 12.4 months [MSC-AC group]) improved significantly in both groups (P < 0.001 for all). Clinical outcomes from the second-look arthroscopy to the final follow-up (mean 27.3 months [MSC group] and 27.8 months [MSC-AC group]) improved further only in the MSC-AC group (P < 0.05 for all). Overall, the Kanamiya grades, which were significantly correlated with clinical outcomes, were significantly higher in the MSC-AC group than in the MSC group. Radiological outcomes at final follow-up revealed improved knee joint alignments relative to preoperative conditions but without significant correlation between clinical outcomes and Kanamiya grade in either group (n.s. for all). CONCLUSION Implantation of MSCs with allogenic cartilage is superior to implantation of MSCs alone in cartilage regeneration accompanied with better clinical outcomes. LEVEL OF EVIDENCE Therapeutic study, level II.
Collapse
|
23
|
Chu DT, Phuong TNT, Tien NLB, Tran DK, Thanh VV, Quang TL, Truong DT, Pham VH, Ngoc VTN, Chu-Dinh T, Kushekhar K. An Update on the Progress of Isolation, Culture, Storage, and Clinical Application of Human Bone Marrow Mesenchymal Stem/Stromal Cells. Int J Mol Sci 2020; 21:E708. [PMID: 31973182 PMCID: PMC7037097 DOI: 10.3390/ijms21030708] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Bone marrow mesenchymal stem/stromal cells (BMSCs), which are known as multipotent cells, are widely used in the treatment of various diseases via their self-renewable, differentiation, and immunomodulatory properties. In-vitro and in-vivo studies have supported the understanding mechanisms, safety, and efficacy of BMSCs therapy in clinical applications. The number of clinical trials in phase I/II is accelerating; however, they are limited in the size of subjects, regulations, and standards for the preparation and transportation and administration of BMSCs, leading to inconsistency in the input and outcome of the therapy. Based on the International Society for Cellular Therapy guidelines, the characterization, isolation, cultivation, differentiation, and applications can be optimized and standardized, which are compliant with good manufacturing practice requirements to produce clinical-grade preparation of BMSCs. This review highlights and updates on the progress of production, as well as provides further challenges in the studies of BMSCs, for the approval of BMSCs widely in clinical application.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam;
| | - Thuy Nguyen Thi Phuong
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju 61186, Korea
| | - Nguyen Le Bao Tien
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam; (N.L.B.T.); (V.V.T.)
| | - Dang Khoa Tran
- Department of Anatomy, University of Medicine Pham Ngoc Thach, Ho Chi Minh City 700000, Vietnam;
| | - Vo Van Thanh
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam; (N.L.B.T.); (V.V.T.)
- Department of Surgery, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Thuy Luu Quang
- Center for Anesthesia and Surgical Intensive Care, Viet Duc Hospital, Hanoi 100000, Vietnam;
| | | | - Van Huy Pham
- AI Lab, Faculty of Information Technology, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | - Vo Truong Nhu Ngoc
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam;
| | - Thien Chu-Dinh
- Institute for Research and Development, Duy Tan University, Danang 550000, Vietnam
| | - Kushi Kushekhar
- Institute of Cancer Research, Oslo University Hospital, 0310 Oslo, Norway;
| |
Collapse
|
24
|
Naji A, Eitoku M, Favier B, Deschaseaux F, Rouas-Freiss N, Suganuma N. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci 2019; 76:3323-3348. [PMID: 31055643 PMCID: PMC11105258 DOI: 10.1007/s00018-019-03125-1] [Citation(s) in RCA: 324] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/19/2019] [Accepted: 04/30/2019] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are isolated from multiple biological tissues-adult bone marrow and adipose tissues and neonatal tissues such as umbilical cord and placenta. In vitro, MSCs show biological features of extensive proliferation ability and multipotency. Moreover, MSCs have trophic, homing/migration and immunosuppression functions that have been demonstrated both in vitro and in vivo. A number of clinical trials are using MSCs for therapeutic interventions in severe degenerative and/or inflammatory diseases, including Crohn's disease and graft-versus-host disease, alone or in combination with other drugs. MSCs are promising for therapeutic applications given the ease in obtaining them, their genetic stability, their poor immunogenicity and their curative properties for tissue repair and immunomodulation. The success of MSC therapy in degenerative and/or inflammatory diseases might depend on the robustness of the biological functions of MSCs, which should be linked to their therapeutic potency. Here, we outline the fundamental and advanced concepts of MSC biological features and underline the biological functions of MSCs in their basic and translational aspects in therapy for degenerative and/or inflammatory diseases.
Collapse
Affiliation(s)
- Abderrahim Naji
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan.
| | - Masamitsu Eitoku
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| | - Benoit Favier
- CEA, DRF-IBFJ, IDMIT, INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Paris-Sud University, Fontenay-aux-Roses, France
| | - Frédéric Deschaseaux
- STROMALab, Etablissement Français du Sang Occitanie, UMR 5273 CNRS, INSERM U1031, Université de Toulouse, Toulouse, France
| | - Nathalie Rouas-Freiss
- CEA, DRF-Francois Jacob Institute, Research Division in Hematology and Immunology (SRHI), Saint-Louis Hospital, IRSL, UMRS 976, Paris, France
| | - Narufumi Suganuma
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School, Kochi University, Kohasu, Oko-Cho, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|
25
|
Bhardwaj N, Singh YP, Mandal BB. Silk Fibroin Scaffold-Based 3D Co-Culture Model for Modulation of Chondrogenesis without Hypertrophy via Reciprocal Cross-talk and Paracrine Signaling. ACS Biomater Sci Eng 2019; 5:5240-5254. [DOI: 10.1021/acsbiomaterials.9b00573] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Nandana Bhardwaj
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati 781125, India
| | - Yogendra Pratap Singh
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Biman B. Mandal
- Biomaterial and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| |
Collapse
|
26
|
Mechanical stimulation promotes the proliferation and the cartilage phenotype of mesenchymal stem cells and chondrocytes co-cultured in vitro. Biomed Pharmacother 2019; 117:109146. [PMID: 31387186 DOI: 10.1016/j.biopha.2019.109146] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 01/29/2023] Open
Abstract
Mesenchymal stem cells and chondrocytes are an important source of the cells for cartilage tissue engineering. Therefore, the culture and expansion methods of these cells need to be improved to overcome the aging of chondrocytes and induced chondrogenic differentiation of mesenchymal stem cells. The aim of this study was to expand the cells for cartilage tissue engineering by combining the advantages of growing cells in co-culture and under a mechanically-stimulated environment. Rabbit chondrocytes and co-cultured cells (bone mesenchymal stem cells and chondrocytes) were subjected to cyclic sinusoidal dynamic tensile mechanical stimulationusing the FX-4000 tension system. Chondrocyte proliferation was assayed by flow cytometry and CFSE labeling. The cell cartilage phenotype was determined by detecting GAG, collagen II and TGF-β1 protein expression by ELISA and the Col2α1, TGF-β1 and Sox9 gene expression by RT-PCR. The results show that the co-culture improved both the proliferation ability of chondrocytes and the cartilage phenotype of co-cultured cells. A proper cyclic sinusoidal dynamic tensile mechanical stimulation improved the proliferation ability and cartilage phenotype of chondrocytes and co-cultured cells. These results suggest that the co-culture of mesenchymal stem cells with chondrocytes and proper mechanical stimulation may be an appropriate way to rapidly expand the cells that have an improved cartilage phenotype for cartilage tissue engineering.
Collapse
|
27
|
Armiento AR, Alini M, Stoddart MJ. Articular fibrocartilage - Why does hyaline cartilage fail to repair? Adv Drug Deliv Rev 2019; 146:289-305. [PMID: 30605736 DOI: 10.1016/j.addr.2018.12.015] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/07/2018] [Accepted: 12/27/2018] [Indexed: 12/12/2022]
Abstract
Once damaged, articular cartilage has a limited potential to repair. Clinically, a repair tissue is formed, yet, it is often mechanically inferior fibrocartilage. The use of monolayer expanded versus naïve cells may explain one of the biggest discrepancies in mesenchymal stromal/stem cell (MSC) based cartilage regeneration. Namely, studies utilizing monolayer expanded MSCs, as indicated by numerous in vitro studies, report as a main limitation the induction of type X collagen and hypertrophy, a phenotype associated with endochondral bone formation. However, marrow stimulation and transfer studies report a mechanically inferior collagen I/II fibrocartilage as the main outcome. Therefore, this review will highlight the collagen species produced during the different therapeutic approaches. New developments in scaffold design and delivery of therapeutic molecules will be described. Potential future directions towards clinical translation will be discussed. New delivery mechanisms are being developed and they offer new hope in targeted therapeutic delivery.
Collapse
Affiliation(s)
| | - Mauro Alini
- AO Research Institute Davos, 7270 Davos Platz, Switzerland.
| | | |
Collapse
|
28
|
Apelgren P, Karabulut E, Amoroso M, Mantas A, Martínez Ávila H, Kölby L, Kondo T, Toriz G, Gatenholm P. In Vivo Human Cartilage Formation in Three-Dimensional Bioprinted Constructs with a Novel Bacterial Nanocellulose Bioink. ACS Biomater Sci Eng 2019; 5:2482-2490. [DOI: 10.1021/acsbiomaterials.9b00157] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Peter Apelgren
- Sahlgrenska Academy, Institute of Clinical Sciences, Department of Plastic Surgery, Sahlgrenska University Hospital, Gothenburg University, Gothenburg 41685, Sweden
| | - Erdem Karabulut
- 3D Bioprinting Center, BBV at Biotech Center, Gothenburg 41346, Sweden
- Wallenberg Wood Science Center, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg 41296, Sweden
| | - Matteo Amoroso
- Sahlgrenska Academy, Institute of Clinical Sciences, Department of Plastic Surgery, Sahlgrenska University Hospital, Gothenburg University, Gothenburg 41685, Sweden
| | - Athanasios Mantas
- 3D Bioprinting Center, BBV at Biotech Center, Gothenburg 41346, Sweden
| | | | - Lars Kölby
- Sahlgrenska Academy, Institute of Clinical Sciences, Department of Plastic Surgery, Sahlgrenska University Hospital, Gothenburg University, Gothenburg 41685, Sweden
| | - Tetsuo Kondo
- Biomacromolecular Materials Lab, and Biomaterial Design Lab, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 6-10-1, Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan
| | - Guillermo Toriz
- Wood, Cellulose and Paper Research, University of Guadalajara, Guadalajara 44100, Mexico
| | - Paul Gatenholm
- 3D Bioprinting Center, BBV at Biotech Center, Gothenburg 41346, Sweden
- Wallenberg Wood Science Center, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg 41296, Sweden
| |
Collapse
|
29
|
Kim HJ, Li Q, Song WJ, Yang HM, Kim SY, Park SC, Ahn JO, Youn HY. Fibroblast growth factor-1 as a mediator of paracrine effects of canine adipose tissue-derived mesenchymal stem cells on in vitro-induced insulin resistance models. BMC Vet Res 2018; 14:351. [PMID: 30445954 PMCID: PMC6240186 DOI: 10.1186/s12917-018-1671-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/25/2018] [Indexed: 12/13/2022] Open
Abstract
Background In the field of diabetes research, many studies on cell therapy have been conducted using mesenchymal stem cells. This research was intended to shed light on the influence of canine adipose-tissue-derived mesenchymal stem cell conditioned medium (cAT-MSC CM) on in vitro insulin resistance models that were induced in differentiated 3T3-L1 adipocytes and the possible mechanisms involved in the phenomenon. Results Gene expression levels of insulin receptor substrate-1 (IRS-1) and glucose transporter type 4 (GLUT4) were used as indicators of insulin resistance. Relative protein expression levels of IRS-1 and GLUT4 were augmented in the cAT-MSC CM treatment group compared to insulin resistance models, indicating beneficial effects of cAT-MSC to DM, probably by actions of secreting factors. With reference to previous studies on fibroblast growth factor-1 (FGF1), we proposed FGF1 as a key contributing factor to the mechanism of action. We added anti-FGF1 neutralizing antibody to the CM-treated insulin resistance models. As a result, significantly diminished protein levels of IRS-1 and GLUT4 were observed, supporting our assumption. Similar results were observed in glucose uptake assay. Conclusions Accordingly, this study advocated the potential of FGF-1 from cAT-MSC CM as an alternative insulin sensitizer and discovered a signalling factor associated with the paracrine effects of cAT-MSC. Electronic supplementary material The online version of this article (10.1186/s12917-018-1671-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyeon-Jin Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Qiang Li
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Woo-Jin Song
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hye-Mi Yang
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Su-Yeon Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang-Chul Park
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jin-Ok Ahn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.,Current Address: Department of Veterinary Internal Medicine, College of Veterinary Medicine, Kangwon National University, Chuncheon, 24341, Gangwondo, Republic of Korea
| | - Hwa-Young Youn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
30
|
Abstract
INTRODUCTION Cartilage tissue engineering has rapidly developed in recent decades, exhibiting promising potential to regenerate and repair cartilage. However, the origin of a large amount of a suitable seed cell source is the major bottleneck for the further clinical application of cartilage tissue engineering. The use of a monoculture of passaged chondrocytes or mesenchymal stem cells results in undesired outcomes, such as fibrocartilage formation and hypertrophy. In the last two decades, co-cultures of chondrocytes and a variety of mesenchymal stem cells have been intensively investigated in vitro and in vivo, shedding light on the perspective of co-culture in cartilage tissue engineering. AREAS COVERED We summarize the recent literature on the application of heterologous cell co-culture systems in cartilage tissue engineering and compare the differences between direct and indirect co-culture systems as well as discuss the underlying mechanisms. EXPERT OPINION Co-culture system is proven to address many issues encountered by monocultures in cartilage tissue engineering, including reducing the number of chondrocytes needed and alleviating the dedifferentiation of chondrocytes. With the further development and knowledge of biomaterials, cartilage tissue engineering that combines the co-culture system and advanced biomaterials is expected to solve the difficult problem regarding the regeneration of functional cartilage.
Collapse
Affiliation(s)
- Jianyu Zou
- a Department of Joint Surgery , The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China.,b Guangdong key laboratory of orthopaedic technology and implant materials , The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| | - Bo Bai
- a Department of Joint Surgery , The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China.,b Guangdong key laboratory of orthopaedic technology and implant materials , The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| | - Yongchang Yao
- a Department of Joint Surgery , The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China.,b Guangdong key laboratory of orthopaedic technology and implant materials , The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| |
Collapse
|
31
|
Fu Y, Karbaat L, Wu L, Leijten J, Both SK, Karperien M. Trophic Effects of Mesenchymal Stem Cells in Tissue Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2018; 23:515-528. [PMID: 28490258 DOI: 10.1089/ten.teb.2016.0365] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are considered to hold great therapeutic value for cell-based therapy and for tissue regeneration in particular. Recent evidence indicates that the main underlying mechanism for MSCs' beneficial effects in tissue regeneration is based on their capability to produce a large variety of bioactive trophic factors that stimulate neighboring parenchymal cells to start repairing damaged tissues. These new findings could potentially replace the classical paradigm of MSC differentiation and cell replacement. These bioactive factors have diverse actions like modulating the local immune system, enhancing angiogenesis, preventing cell apoptosis, and stimulating survival, proliferation, and differentiation of resident tissue specific cells. Therefore, MSCs are referred to as conductors of tissue repair and regeneration by secreting trophic mediators. In this review article, we have summarized the studies that focused on the trophic effects of MSC within the context of tissue regeneration. We will also highlight the various underlying mechanisms used by MSCs to act as trophic mediators. Besides the secretion of growth factors, we discuss two additional mechanisms that are likely to mediate MSC's beneficial effects in tissue regeneration, namely the production of extracellular vesicles and the formation of membrane nanotubes, which can both connect different cells and transfer a variety of trophic factors varying from proteins to mRNAs and miRNAs. Furthermore, we postulate that apoptosis of the MSCs is an integral part of the trophic effect during tissue repair.
Collapse
Affiliation(s)
- Yao Fu
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Lisanne Karbaat
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Ling Wu
- 2 Center for Craniofacial Molecular Biology, University of Southern California , Los Angeles, Los Angeles, California
| | - Jeroen Leijten
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Sanne K Both
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Marcel Karperien
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| |
Collapse
|
32
|
Engineered Co-culture Strategies Using Stem Cells for Facilitated Chondrogenic Differentiation and Cartilage Repair. BIOTECHNOL BIOPROC E 2018. [DOI: 10.1007/s12257-018-0149-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
33
|
Critchley SE, Eswaramoorthy R, Kelly DJ. Low‐oxygen conditions promote synergistic increases in chondrogenesis during co‐culture of human osteoarthritic stem cells and chondrocytes. J Tissue Eng Regen Med 2018; 12:1074-1084. [DOI: 10.1002/term.2608] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 10/11/2017] [Accepted: 10/23/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Susan E. Critchley
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Rajalakshmanan Eswaramoorthy
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Daniel J. Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Department of AnatomyRoyal College of Surgeons in Ireland Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| |
Collapse
|
34
|
Vonk LA, van Dooremalen SFJ, Liv N, Klumperman J, Coffer PJ, Saris DB, Lorenowicz MJ. Mesenchymal Stromal/stem Cell-derived Extracellular Vesicles Promote Human Cartilage Regeneration In Vitro. Am J Cancer Res 2018; 8:906-920. [PMID: 29463990 PMCID: PMC5817101 DOI: 10.7150/thno.20746] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 10/08/2017] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA) is a rheumatic disease leading to chronic pain and disability with no effective treatment available. Recently, allogeneic human mesenchymal stromal/stem cells (MSC) entered clinical trials as a novel therapy for OA. Increasing evidence suggests that therapeutic efficacy of MSC depends on paracrine signalling. Here we investigated the role of extracellular vesicles (EVs) secreted by human bone marrow derived MSC (BMMSC) in human OA cartilage repair. Methods: To test the effect of BMMSC-EVs on OA cartilage inflammation, TNF-alpha-stimulated OA chondrocyte monolayer cultures were treated with BMMSC-EVs and pro-inflammatory gene expression was measured by qRT-PCR after 48 h. To assess the impact of BMMSC-EVs on cartilage regeneration, BMMSC-EVs were added to the regeneration cultures of human OA chondrocytes, which were analyzed after 4 weeks for glycosaminoglycan content by 1,9-dimethylmethylene blue (DMMB) assay. Furthermore, paraffin sections of the regenerated tissue were stained for proteoglycans (safranin-O) and type II collagen (immunostaining). Results: We show that BMMSC-EVs inhibit the adverse effects of inflammatory mediators on cartilage homeostasis. When co-cultured with OA chondrocytes, BMMSC-EVs abrogated the TNF-alpha-mediated upregulation of COX2 and pro-inflammatory interleukins and inhibited TNF-alpha-induced collagenase activity. BMMSC-EVs also promoted cartilage regeneration in vitro. Addition of BMMSC-EVs to cultures of chondrocytes isolated from OA patients stimulated production of proteoglycans and type II collagen by these cells. Conclusion: Our data demonstrate that BMMSC-EVs can be important mediators of cartilage repair and hold great promise as a novel therapeutic for cartilage regeneration and osteoarthritis.
Collapse
|
35
|
Apelgren P, Amoroso M, Lindahl A, Brantsing C, Rotter N, Gatenholm P, Kölby L. Chondrocytes and stem cells in 3D-bioprinted structures create human cartilage in vivo. PLoS One 2017; 12:e0189428. [PMID: 29236765 PMCID: PMC5728520 DOI: 10.1371/journal.pone.0189428] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/25/2017] [Indexed: 12/21/2022] Open
Abstract
Cartilage repair and replacement is a major challenge in plastic reconstructive surgery. The development of a process capable of creating a patient-specific cartilage framework would be a major breakthrough. Here, we described methods for creating human cartilage in vivo and quantitatively assessing the proliferative capacity and cartilage-formation ability in mono- and co-cultures of human chondrocytes and human mesenchymal stem cells in a three-dimensional (3D)-bioprinted hydrogel scaffold. The 3D-bioprinted constructs (5 × 5 × 1.2 mm) were produced using nanofibrillated cellulose and alginate in combination with human chondrocytes and human mesenchymal stem cells using a 3D-extrusion bioprinter. Immediately following bioprinting, the constructs were implanted subcutaneously on the back of 48 nude mice and explanted after 30 and 60 days, respectively, for morphological and immunohistochemical examination. During explantation, the constructs were easy to handle, and the majority had retained their macroscopic grid appearance. Constructs consisting of human nasal chondrocytes showed good proliferation ability, with 17.2% of the surface areas covered with proliferating chondrocytes after 60 days. In constructs comprising a mixture of chondrocytes and stem cells, an additional proliferative effect was observed involving chondrocyte production of glycosaminoglycans and type 2 collagen. This clinically highly relevant study revealed 3D bioprinting as a promising technology for the creation of human cartilage.
Collapse
Affiliation(s)
- Peter Apelgren
- Department of Plastic Surgery, Institute of Clinical Sciences, Sahlgrenska University Hospital, Sahlgrenska Academy, Göteborg, Sweden
| | - Matteo Amoroso
- Department of Plastic Surgery, Institute of Clinical Sciences, Sahlgrenska University Hospital, Sahlgrenska Academy, Göteborg, Sweden
| | - Anders Lindahl
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Sahlgrenska Academy, Göteborg, Sweden
| | - Camilla Brantsing
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Sahlgrenska Academy, Göteborg, Sweden
| | - Nicole Rotter
- Department of Otorhinolaryngology, University Medical Centre Ulm, Ulm, Germany
| | - Paul Gatenholm
- 3D Bioprinting Centre, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Göteborg, Sweden
| | - Lars Kölby
- Department of Plastic Surgery, Institute of Clinical Sciences, Sahlgrenska University Hospital, Sahlgrenska Academy, Göteborg, Sweden
| |
Collapse
|
36
|
Ouyang A, Cerchiari AE, Tang X, Liebenberg E, Alliston T, Gartner ZJ, Lotz JC. Effects of cell type and configuration on anabolic and catabolic activity in 3D co-culture of mesenchymal stem cells and nucleus pulposus cells. J Orthop Res 2017; 35:61-73. [PMID: 27699833 PMCID: PMC5258652 DOI: 10.1002/jor.23452] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 09/24/2016] [Indexed: 02/04/2023]
Abstract
Tissue engineering constructs to treat intervertebral disc degeneration must adapt to the hypoxic and inflammatory degenerative disc microenvironment. The objective of this study was to determine the effects of two key design factors, cell type and cell configuration, on the regenerative potential of nucleus pulposus cell (NPC) and mesenchymal stem cell (MSC) constructs. Anabolic and catabolic activity was quantified in constructs of varying cell type (NPCs, MSCs, and a 50:50 co-culture) and varying configuration (individual cells and micropellets). Anabolic and catabolic outcomes were both dependent on cell type. Gene expression of Agg and Col2A1, glycosaminoglycan (GAG) content, and aggrecan immunohistochemistry (IHC), were significantly higher in NPC-only and co-culture groups than in MSC-only groups, with NPC-only groups exhibiting the highest anabolic gene expression levels. However, NPC-only constructs also responded to inflammation and hypoxia with significant upregulation of catabolic genes (MMP-1, MMP-9, MMP-13, and ADAMTS-5). MSC-only groups were unaffected by degenerative media conditions, and co-culture with MSCs modulated catabolic induction of the NPCs. Culturing cells in a micropellet configuration dramatically reduced catabolic induction in co-culture and NPC-only groups. Co-culture micropellets, which take advantage of both cell type and configuration effects, had the most immunomodulatory response, with a significant decrease in MMP-13 and ADAMTS-5 expression in hypoxic and inflammatory media conditions. Co-culture micropellets were also found to self-organize into bilaminar formations with an MSC core and NPC outer layer. Further understanding of these cell type and configuration effects can improve tissue engineering designs. © 2016 The Authors. Journal of Orthopaedic Research published by Wiley Periodicals, Inc. on behalf of the Orthopaedic Research Society. J Orthop Res 35:61-73, 2017.
Collapse
Affiliation(s)
- Ann Ouyang
- University of California; San Francisco California
| | | | - Xinyan Tang
- University of California; San Francisco California
| | | | | | | | | |
Collapse
|
37
|
Bioinspired seeding of biomaterials using three dimensional microtissues induces chondrogenic stem cell differentiation and cartilage formation under growth factor free conditions. Sci Rep 2016; 6:36011. [PMID: 27808102 PMCID: PMC5093556 DOI: 10.1038/srep36011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 10/10/2016] [Indexed: 11/30/2022] Open
Abstract
Cell laden biomaterials are archetypically seeded with individual cells and steered into the desired behavior using exogenous stimuli to control growth and differentiation. In contrast, direct cell-cell contact is instructive and even essential for natural tissue formation. Namely, microaggregation and condensation of mesenchymal progenitor cells triggers chondrogenesis and thereby drives limb formation. Yet a biomimetic strategy translating this approach into a cell laden biomaterial-based therapy has remained largely unexplored. Here, we integrate the microenvironment of cellular condensation into biomaterials by encapsulating microaggregates of a hundred human periosteum-derived stem cells. This resulted in decreased stemness-related markers, up regulation of chondrogenic genes and improved in vivo cartilage tissue formation, as compared to single cell seeded biomaterials. Importantly, even in the absence of exogenous growth factors, the microaggregate laden hydrogels outperformed conventional single cell laden hydrogels containing supraphysiological levels of the chondrogenic growth factor TGFB. Overall, the bioinspired seeding strategy described herein represents an efficient and growth factor-free approach to efficiently steer cell fate and drive tissue formation for biomaterial-based tissue engineering strategies.
Collapse
|
38
|
Zhong L, Huang X, Rodrigues ED, Leijten JCH, Verrips T, El Khattabi M, Karperien M, Post JN. Endogenous DKK1 and FRZB Regulate Chondrogenesis and Hypertrophy in Three-Dimensional Cultures of Human Chondrocytes and Human Mesenchymal Stem Cells. Stem Cells Dev 2016; 25:1808-1817. [PMID: 27733096 PMCID: PMC5124737 DOI: 10.1089/scd.2016.0222] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hypertrophic differentiation occurs during in vitro chondrogenesis of mesenchymal stem cells (MSCs), decreasing the quality of the cartilage construct. Previously we identified WNT pathway antagonists Dickkopf 1 homolog (DKK1) and frizzled-related protein (FRZB) as key factors in blocking hypertrophic differentiation of human MSCs (hMSCs). In this study, we investigated the role of endogenously expressed DKK1 and FRZB in chondrogenesis of hMSC and chondrocyte redifferentiation and in preventing cell hypertrophy using three relevant human cell based systems, isolated hMSCs, isolated primary human chondrocytes (hChs), and cocultures of hMSCs with hChs for which we specifically designed neutralizing nano-antibodies. We selected and tested variable domain of single chain heavy chain only antibodies (VHH) for their ability to neutralize the function of DKK1 or FRZB. In the presence of DKK1 and FRZB neutralizing VHH, glycosaminoglycan and collagen type II staining were significantly reduced in monocultured hMSCs and monocultured chondrocytes. Furthermore, in cocultures, cells in pellets showed hypertrophic differentiation. In conclusion, endogenous expression of the WNT antagonists DKK1 and FRZB is necessary for multiple steps during chondrogenesis: first DKK1 and FRZB are indispensable for the initial steps of chondrogenic differentiation of hMSCs, second they are necessary for chondrocyte redifferentiation, and finally in preventing hypertrophic differentiation of articular chondrocytes.
Collapse
Affiliation(s)
- Leilei Zhong
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Xiaobin Huang
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Emilie Dooms Rodrigues
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Jeroen C H Leijten
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | | | | | - Marcel Karperien
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Janine N Post
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| |
Collapse
|
39
|
Hofer HR, Tuan RS. Secreted trophic factors of mesenchymal stem cells support neurovascular and musculoskeletal therapies. Stem Cell Res Ther 2016; 7:131. [PMID: 27612948 PMCID: PMC5016979 DOI: 10.1186/s13287-016-0394-0] [Citation(s) in RCA: 240] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Adult mesenchymal stem cells (MSCs) represent a subject of intense experimental and biomedical interest. Recently, trophic activities of MSCs have become the topic of a number of revealing studies that span both basic and clinical fields. In this review, we focus on recent investigations that have elucidated trophic mechanisms and shed light on MSC clinical efficacy relevant to musculoskeletal applications. Innate differences due to MSC sourcing may play a role in the clinical utility of isolated MSCs. Pain management, osteochondral, nerve, or blood vessel support by MSCs derived from both autologous and allogeneic sources have been examined. Recent mechanistic insights into the trophic activities of these cells point to ultimate regulation by nitric oxide, nuclear factor-kB, and indoleamine, among other signaling pathways. Classic growth factors and cytokines-such as VEGF, CNTF, GDNF, TGF-β, interleukins (IL-1β, IL-6, and IL-8), and C-C ligands (CCL-2, CCL-5, and CCL-23)-serve as paracrine control molecules secreted or packaged into extracellular vesicles, or exosomes, by MSCs. Recent studies have also implicated signaling by microRNAs contained in MSC-derived exosomes. The response of target cells is further regulated by their microenvironment, involving the extracellular matrix, which may be modified by MSC-produced matrix metalloproteinases (MMPs) and tissue inhibitor of MMPs. Trophic activities of MSCs, either resident or introduced exogenously, are thus intricately controlled, and may be further fine-tuned via implant material modifications. MSCs are actively being investigated for the repair and regeneration of both osteochondral and other musculoskeletal tissues, such as tendon/ligament and meniscus. Future rational and effective MSC-based musculoskeletal therapies will benefit from better mechanistic understanding of MSC trophic activities, for example using analytical "-omics" profiling approaches.
Collapse
Affiliation(s)
- Heidi R Hofer
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA, 15219, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Room 221, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
40
|
Xu L, Wu Y, Xiong Z, Zhou Y, Ye Z, Tan WS. Mesenchymal Stem Cells Reshape and Provoke Proliferation of Articular Chondrocytes by Paracrine Secretion. Sci Rep 2016; 6:32705. [PMID: 27596239 PMCID: PMC5011711 DOI: 10.1038/srep32705] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 08/08/2016] [Indexed: 01/03/2023] Open
Abstract
Coculture between mesenchymal stem cells (MSCs) and articular chondrocytes (ACs) represents a promising strategy for cartilage regeneration. This study aimed at elaborating how ACs were regulated by MSCs. Rabbit ACs (rACs) and rabbit MSCs (rMSCs) were seeded separately in a Transwell system to initiate non-contact coculture in growth medium without chondrogenic factors. Cell morphology, cell proliferation, production of extracellular matrix (ECM), and gene expression of rACs were characterized. Upon coculture, rACs underwent a morphological transition from a rounded or polygonal shape into a fibroblast-like one and proliferation was provoked simultaneously. Such effects were dependent on the amount of rMSCs. Along with these changes, ECM production and gene expression of rACs were also perturbed. Importantly, when a ROCK inhibitor (Y27632) was supplemented to coculture, the effects except that on cell proliferation were inhibited, suggesting the involvement of RhoA/ROCK signaling. By applying an inhibitor (BIBF1120) of VEGFR1/2/3, FGFR1/2/3 and PDGFRα/β in coculture, or supplementing FGF-1, VEGF-A and PDGFbb in monoculture, it was confirmed that the paracrine factors by rMSCs mediated the compounding effects on rACs. These findings shed light on MSCs-ACs interactions and might confer an insight view on cell-based cartilage regeneration.
Collapse
Affiliation(s)
- Lei Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yuxi Wu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Zhimiao Xiong
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yan Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Zhaoyang Ye
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
41
|
Zhou Y, Tao H, Li Y, Deng M, He B, Xia S, Zhang C, Liu S. Berberine promotes proliferation of sodium nitroprusside-stimulated rat chondrocytes and osteoarthritic rat cartilage via Wnt/β-catenin pathway. Eur J Pharmacol 2016; 789:109-118. [PMID: 27445236 DOI: 10.1016/j.ejphar.2016.07.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 07/05/2016] [Accepted: 07/16/2016] [Indexed: 01/07/2023]
Abstract
Berberine chloride (BBR) is an isoquinoline derivative alkaloid isolated from medicinal herbs, including Coptis chinensis and Berberis aristate. This compound plays significant roles in the treatment of osteoarthritis (OA). The purpose of this study was to investigate the effects of BBR on the proliferation of sodium nitroprusside (SNP)-stimulated chondrocytes in vitro, the articular cartilage in a rat OA model, as well as to discuss the molecular mechanisms underlying these effects. In vitro, we demonstrated that BBR led to cell proliferation, increased the cell population in S-phase and decreased that in G0/G1-phase; moreover, the F-actin remodeling in SNP-stimulated chondrocytes were prevented. In addition, BBR markedly up-regulated β-catenin, c-Myc, and cyclin D1 expression of genes and proteins, and down-regulated glycogen synthase kinase-3β (GSK-3β) and matrix metalloproteinase-7 (MMP-7) expression. Notably, inhibition of the Wnt/β-catenin pathway by XAV939 partially blocked these effects. The in vivo results suggested that BBR promoted β-catenin protein level and enhanced proliferating cell nuclear antigen (PCNA) expression in osteoarthritic rat cartilage. In conclusion, these findings indicate that BBR promotes SNP-stimulated chondrocyte proliferation by promoting G1/S phase transition and synthesis of PCNA in cartilage through activation of Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Department of Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Laboratory of Clinical Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | - Haiying Tao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Laboratory of Clinical Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | - Yaming Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Laboratory of Clinical Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | - Ming Deng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Laboratory of Clinical Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | - Bin He
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | - Shaoqiang Xia
- Department of Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Laboratory of Clinical Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | - Chun Zhang
- Department of Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Laboratory of Clinical Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China
| | - Shiqing Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China; Department of Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, PR China.
| |
Collapse
|
42
|
Park GC, Song JS, Park HY, Shin SC, Jang JY, Lee JC, Wang SG, Lee BJ, Jung JS. Role of Fibroblast Growth Factor-5 on the Proliferation of Human Tonsil-Derived Mesenchymal Stem Cells. Stem Cells Dev 2016; 25:1149-60. [PMID: 27224250 DOI: 10.1089/scd.2016.0061] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Human mesenchymal stem cells (MSCs) are a promising tool for therapeutic applications in cell-based therapy and regenerative medicine, and MSCs from the human palatine tonsils have recently been used as a new tissue source. However, the understanding of the proliferation and differentiation capacity of tonsil-derived MSCs (T-MSCs) is limited. In this study, we compared the proliferative potential of T-MSCs with those of bone marrow MSCs (BM-MSCs) and adipose tissue-derived MSCs (A-MSCs). Additionally, we investigated the underlying mechanism of T-MSC function. We showed that T-MSCs proliferated faster than A-MSCs and BM-MSCs in methylthiazolyl diphenyl-tetrazolium (MTT) assays, cell count assays, and cell cycle distribution analyses. DNA microarray and real-time PCR analyses revealed that the expression of fibroblast growth factor-5 (FGF5) was significantly elevated in T-MSCs compared with those in A-MSCs and BM-MSCs. Cell growth curves showed a difference in cell growth between untreated cells and siFGF5-treated T-MSCs. The administration of recombinant human FGF5 (rhFGF5) to the cells transfected with siFGF5 led to a significant increase in the proliferation rates. The administration of rhFGF5 to T-MSCs led to an increase in the levels of phosphorylated ERK1/2. However, treatment with siFGF5 resulted in an overall decrease in the level of phosphorylated ERK1/2. The osteogenic differentiation of T-MSCs was reduced following siFGF5 transfection, and it recovered to near-normal levels when rhFGF5 was added. These findings indicate that T-MSCs show significantly higher proliferative potential compared with those of BM-MSCs and A-MSCs. FGF5 facilitates cell proliferation through ERK1/2 activation, and it influences the osteogenic differentiation of T-MSCs.
Collapse
Affiliation(s)
- Gi Cheol Park
- 1 Department of Otolaryngology-Head and Neck Surgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine , Changwon, Korea
| | - Ji Sun Song
- 2 Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University , Busan, Korea
| | - Hee-Young Park
- 2 Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University , Busan, Korea
| | - Sung-Chan Shin
- 2 Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University , Busan, Korea
| | - Jeon Yeob Jang
- 2 Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University , Busan, Korea
| | - Jin-Choon Lee
- 3 Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University , Yangsan Hospital, Yangsan, Korea
| | - Soo-Geun Wang
- 2 Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University , Busan, Korea
| | - Byung-Joo Lee
- 2 Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University , Busan, Korea
| | - Jin-Sup Jung
- 4 Department of Physiology, School of Medicine, Pusan National University , Yangsan, Korea
| |
Collapse
|
43
|
Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells Contribute to Chondrogenesis in Coculture with Chondrocytes. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3827057. [PMID: 27446948 PMCID: PMC4944057 DOI: 10.1155/2016/3827057] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/28/2016] [Accepted: 05/22/2016] [Indexed: 12/21/2022]
Abstract
Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) have been shown as the most potential stem cell source for articular cartilage repair. In this study, we aimed to develop a method for long-term coculture of human articular chondrocytes (hACs) and hUCB-MSCs at low density in vitro to determine if the low density of hACs could enhance the hUCB-MSC chondrogenic differentiation as well as to determine the optimal ratio of the two cell types. Also, we compared the difference between direct coculture and indirect coculture at low density. Monolayer cultures of hUCB-MSCs and hACs were investigated at different ratios, at direct cell-cell contact groups for 21 days. Compared to direct coculture, hUCB-MSCs and hACs indirect contact culture significantly increased type II collagen (COL2) and decreased type I collagen (COL1) protein expression levels. SRY-box 9 (SOX9) mRNA levels and protein expression were highest in indirect coculture. Overall, these results indicate that low density direct coculture induces fibrocartilage. However, indirect coculture in conditioned chondrocyte cell culture medium can increase expression of chondrogenic markers and induce hUCB-MSCs differentiation into mature chondrocytes. This work demonstrates that it is possible to promote chondrogenesis of hUCB-MSCs in combination with hACs, further supporting the concept of novel coculture strategies for tissue engineering.
Collapse
|
44
|
Leijten J, Rouwkema J, Zhang YS, Nasajpour A, Dokmeci MR, Khademhosseini A. Advancing Tissue Engineering: A Tale of Nano-, Micro-, and Macroscale Integration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:2130-45. [PMID: 27101419 PMCID: PMC4895865 DOI: 10.1002/smll.201501798] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 08/16/2015] [Indexed: 05/19/2023]
Abstract
Tissue engineering has the potential to revolutionize the health care industry. Delivering on this promise requires the generation of efficient, controllable and predictable implants. The integration of nano- and microtechnologies into macroscale regenerative biomaterials plays an essential role in the generation of such implants, by enabling spatiotemporal control of the cellular microenvironment. Here we review the role, function and progress of a wide range of nano- and microtechnologies that are driving the advancements in the field of tissue engineering.
Collapse
Affiliation(s)
- Jeroen Leijten
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medicine, Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Jeroen Rouwkema
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medicine, Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Department of Biomechanical Engineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Yu Shrike Zhang
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medicine, Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Amir Nasajpour
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medicine, Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Mehmet Remzi Dokmeci
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medicine, Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Ali Khademhosseini
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medicine, Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul, 143-701, Republic of Korea
- Department of Physics, King Abdulaziz University, Jeddah, 21569, Saudi Arabia
| |
Collapse
|
45
|
Pers YM, Ruiz M, Noël D, Jorgensen C. Mesenchymal stem cells for the management of inflammation in osteoarthritis: state of the art and perspectives. Osteoarthritis Cartilage 2015; 23:2027-35. [PMID: 26521749 DOI: 10.1016/j.joca.2015.07.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 06/29/2015] [Accepted: 07/07/2015] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is the most common form of degenerative arthritis, mainly characterized by the degradation of articular cartilage and associated with subchondral bone lesions. Novel therapeutic approaches for OA include cell-based therapies that have become thriving areas of research and development. In this context, mesenchymal stem or stromal cells (MSCs) have gained much interest based on their trophic and immunomodulatory properties that can help tissue repair/regeneration. The present review article discusses the interest of using MSCs in cell-therapy approaches with a focus on the mechanisms by which MSCs might exhibit a therapeutic potential in OA. Special attention is given to the anti-inflammatory function of MSCs and on miRNA modulation in OA for possible future innovative strategies. The paper also presents the current data on the undergoing MSCs-based clinical trials in OA.
Collapse
Affiliation(s)
- Y-M Pers
- Inserm U1183, Hôpital Saint-Eloi, Montpellier, F-34295, France; Université Montpellier, UFR de Médecine, Montpellier, F-34000, France; Service d'immuno-Rhumatologie, Hôpital Lapeyronie, Montpellier, F-34295, France
| | - M Ruiz
- Inserm U1183, Hôpital Saint-Eloi, Montpellier, F-34295, France; Université Montpellier, UFR de Médecine, Montpellier, F-34000, France
| | - D Noël
- Inserm U1183, Hôpital Saint-Eloi, Montpellier, F-34295, France; Université Montpellier, UFR de Médecine, Montpellier, F-34000, France.
| | - C Jorgensen
- Inserm U1183, Hôpital Saint-Eloi, Montpellier, F-34295, France; Université Montpellier, UFR de Médecine, Montpellier, F-34000, France; Service d'immuno-Rhumatologie, Hôpital Lapeyronie, Montpellier, F-34295, France
| |
Collapse
|
46
|
de Windt TS, Saris DBF, Slaper-Cortenbach ICM, van Rijen MHP, Gawlitta D, Creemers LB, de Weger RA, Dhert WJA, Vonk LA. Direct Cell-Cell Contact with Chondrocytes Is a Key Mechanism in Multipotent Mesenchymal Stromal Cell-Mediated Chondrogenesis. Tissue Eng Part A 2015; 21:2536-47. [PMID: 26166387 DOI: 10.1089/ten.tea.2014.0673] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Using a combination of articular chondrocytes (ACs) and mesenchymal stromal cells (MSCs) has shown to be a viable option for a single-stage cell-based treatment of focal cartilage defects. However, there is still considerable debate whether MSCs differentiate or have a chondroinductive role through trophic factors. In addition, it remains unclear whether direct cell-cell contact is necessary for chondrogenesis. Therefore, the aim of this study was to investigate whether direct or indirect cell-cell contact between ACs and MSCs is essential for increased cartilage production in different cellular environments and elucidate the mechanisms behind these cellular interactions. Human ACs and MSCs were cultured in a 10:90 ratio in alginate beads, fibrin scaffolds, and pellets. Cells were mixed in direct cocultures, separated by a Transwell filter (indirect cocultures), or cultured with conditioned medium. Short tandem repeat analysis revealed that the percentages of ACs increased during culture, while those of MSCs decreased, with the biggest change in fibrin glue scaffolds. For alginate, where the lack of cell-cell contact could be confirmed by histological analysis, no difference was found in matrix production between direct and indirect cocultures. For fibrin scaffolds and pellet cultures, an increased glycosaminoglycan production and type II collagen deposition were found in direct cocultures compared with indirect cocultures and conditioned medium. Positive connexin 43 staining and transfer of cytosolic calcein indicated communication through gap junctions in direct cocultures. Taken together, these results suggest that MSCs stimulate cartilage formation when placed in close proximity to chondrocytes and that direct cell-cell contact and communication through gap junctions are essential in this chondroinductive interplay.
Collapse
Affiliation(s)
- Tommy S de Windt
- 1 Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Daniel B F Saris
- 1 Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands .,2 MIRA Institute for Biotechnology and Technical Medicine, University Twente , Enschede, The Netherlands
| | - Ineke C M Slaper-Cortenbach
- 3 Cell Therapy Facility, Department of Clinical Pharmacy, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Mattie H P van Rijen
- 1 Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Debby Gawlitta
- 1 Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Laura B Creemers
- 1 Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Roel A de Weger
- 4 Department of Pathology, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Wouter J A Dhert
- 5 Faculty of Veterinary Medicine, University of Utrecht , Utrecht, The Netherlands
| | - Lucienne A Vonk
- 1 Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| |
Collapse
|
47
|
Mesallati T, Buckley CT, Kelly DJ. Engineering cartilaginous grafts using chondrocyte-laden hydrogels supported by a superficial layer of stem cells. J Tissue Eng Regen Med 2015; 11:1343-1353. [PMID: 26010516 DOI: 10.1002/term.2033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 04/03/2015] [Accepted: 04/21/2015] [Indexed: 11/07/2022]
Abstract
During postnatal joint development, progenitor cells that reside in the superficial region of articular cartilage first drive the rapid growth of the tissue and later help direct the formation of mature hyaline cartilage. These developmental processes may provide directions for the optimal structuring of co-cultured chondrocytes (CCs) and multipotent stromal/stem cells (MSCs) required for engineering cartilaginous tissues. The objective of this study was to engineer cartilage grafts by recapitulating aspects of joint development where a population of superficial progenitor cells drives the development of the tissue. To this end, MSCs were either self-assembled on top of CC-laden agarose gels (structured co-culture) or were mixed with CCs before being embedded in an agarose hydrogel (mixed co-culture). Porcine infrapatellar fat pad-derived stem cells (FPSCs) and bone marrow-derived MSCs (BMSCs) were used as sources of progenitor cells. The DNA, sGAG and collagen content of a mixed co-culture of FPSCs and CCs was found to be lower than the combined content of two control hydrogels seeded with CCs and FPSCs only. In contrast, a mixed co-culture of BMSCs and CCs led to increased proliferation and sGAG and collagen accumulation. Of note was the finding that a structured co-culture, at the appropriate cell density, led to greater sGAG accumulation than a mixed co-culture for both MSC sources. In conclusion, assembling MSCs onto CC-laden hydrogels dramatically enhances the development of the engineered tissue, with the superficial layer of progenitor cells driving CC proliferation and cartilage ECM production, mimicking certain aspects of developing cartilage. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Tariq Mesallati
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Conor T Buckley
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and the Royal College of Surgeons in Ireland, Ireland
| |
Collapse
|
48
|
Leijten J, Chai Y, Papantoniou I, Geris L, Schrooten J, Luyten F. Cell based advanced therapeutic medicinal products for bone repair: Keep it simple? Adv Drug Deliv Rev 2015; 84:30-44. [PMID: 25451134 DOI: 10.1016/j.addr.2014.10.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 09/18/2014] [Accepted: 10/20/2014] [Indexed: 02/08/2023]
Abstract
The development of cell based advanced therapeutic medicinal products (ATMPs) for bone repair has been expected to revolutionize the health care system for the clinical treatment of bone defects. Despite this great promise, the clinical outcomes of the few cell based ATMPs that have been translated into clinical treatments have been far from impressive. In part, the clinical outcomes have been hampered because of the simplicity of the first wave of products. In response the field has set-out and amassed a plethora of complexities to alleviate the simplicity induced limitations. Many of these potential second wave products have remained "stuck" in the development pipeline. This is due to a number of reasons including the lack of a regulatory framework that has been evolving in the last years and the shortage of enabling technologies for industrial manufacturing to deal with these novel complexities. In this review, we reflect on the current ATMPs and give special attention to novel approaches that are able to provide complexity to ATMPs in a straightforward manner. Moreover, we discuss the potential tools able to produce or predict 'goldilocks' ATMPs, which are neither too simple nor too complex.
Collapse
|
49
|
Georgi N, Cillero-Pastor B, Eijkel GB, Periyasamy PC, Kiss A, van Blitterswijk C, Post JN, Heeren RMA, Karperien M. Differentiation of Mesenchymal Stem Cells under Hypoxia and Normoxia: Lipid Profiles Revealed by Time-of-Flight Secondary Ion Mass Spectrometry and Multivariate Analysis. Anal Chem 2015; 87:3981-8. [DOI: 10.1021/acs.analchem.5b00114] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Nicole Georgi
- Developmental
BioEngineering, MIRA Institute for Biomedical Technology
and Technical Medicine, Faculty of Science and Technology, University of Twente, 7522
NB Enschede, The Netherlands
| | - Berta Cillero-Pastor
- Biomolecular
Imaging
Mass Spectrometry, FOM Institute AMOLF, 1098 XG Amsterdam, The Netherlands
- The Maastricht Multimodal
Molecular Imaging Institute, M4I, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Gert B. Eijkel
- Biomolecular
Imaging
Mass Spectrometry, FOM Institute AMOLF, 1098 XG Amsterdam, The Netherlands
| | - Parthiban C. Periyasamy
- Developmental
BioEngineering, MIRA Institute for Biomedical Technology
and Technical Medicine, Faculty of Science and Technology, University of Twente, 7522
NB Enschede, The Netherlands
| | - Andras Kiss
- Biomolecular
Imaging
Mass Spectrometry, FOM Institute AMOLF, 1098 XG Amsterdam, The Netherlands
| | - Clemens van Blitterswijk
- Department
of Tissue Regeneration, MIRA Institute for Biomedical
Technology and Technical Medicine, Faculty of Science and Technology, University of Twente, 7522
NB Enschede, The Netherlands
| | - Janine N. Post
- Developmental
BioEngineering, MIRA Institute for Biomedical Technology
and Technical Medicine, Faculty of Science and Technology, University of Twente, 7522
NB Enschede, The Netherlands
| | - Ron M. A. Heeren
- Biomolecular
Imaging
Mass Spectrometry, FOM Institute AMOLF, 1098 XG Amsterdam, The Netherlands
- The Maastricht Multimodal
Molecular Imaging Institute, M4I, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Marcel Karperien
- Developmental
BioEngineering, MIRA Institute for Biomedical Technology
and Technical Medicine, Faculty of Science and Technology, University of Twente, 7522
NB Enschede, The Netherlands
| |
Collapse
|
50
|
Beneficial effects of coculturing synovial derived mesenchymal stem cells with meniscus fibrochondrocytes are mediated by fibroblast growth factor 1: increased proliferation and collagen synthesis. Stem Cells Int 2015; 2015:926325. [PMID: 25852755 PMCID: PMC4379431 DOI: 10.1155/2015/926325] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 12/16/2014] [Accepted: 01/01/2015] [Indexed: 01/06/2023] Open
Abstract
Meniscus reconstruction is in great need for orthopedic surgeons. Meniscal fibrochondrocytes transplantation was proposed to regenerate functional meniscus, with limited donor supply. We hypothesized that coculture of synovial mesenchymal stem cells (SSC) with meniscal fibrochondrocytes (me-CH) can support matrix production of me-CH, thus reducing the number of me-CH needed for meniscus reconstruction. A pellet coculture system of human SSC and me-CH was used in this study. Enhanced glycosaminoglycans (GAG) in coculture pellets were demonstrated by Alcian blue staining and GAG quantification, when compared to monoculture. More collagen synthesis was shown in coculture pellets by hydroxyproline assay. Increased proliferation of me-CH was observed in coculture. Data from BrdU staining and ELISA demonstrated that conditioned medium of SSCs enhanced the proliferation and collagen synthesis of me-CH, and this effect was blocked by neutralizing antibody against fibroblast growth factor 1 (FGF1). Western blot showed that conditioned medium of SSCs can activate mitogen-activated protein kinase (MAPK) signaling pathways by increasing the phosphorylation of mitogen-activated regulated protein kinase 1/2 (MEK) and extracellular-signal-regulated kinases 1/2 (ERK). Overall, this study provided evidence that synovial MSCs can support proliferation and collagen synthesis of fibrochondrocytes, by secreting FGF1. Coimplantation of SSC and me-CH could be a useful strategy for reconstructing meniscus.
Collapse
|