1
|
Dupuy S, Salvador J, Morille M, Noël D, Belamie E. Control and interplay of scaffold-biomolecule interactions applied to cartilage tissue engineering. Biomater Sci 2025; 13:1871-1900. [PMID: 40052975 DOI: 10.1039/d5bm00049a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Cartilage tissue engineering based on the combination of biomaterials, adult or stem cells and bioactive factors is a challenging approach for regenerative medicine with the aim of achieving the formation of a functional neotissue stable in the long term. Various 3D scaffolds have been developed to mimic the extracellular matrix environment and promote cartilage repair. In addition, bioactive factors have been extensively employed to induce and maintain the cartilage phenotype. However, the spatiotemporal control of bioactive factor release remains critical for maximizing the regenerative potential of multipotent cells, such as mesenchymal stromal cells (MSCs), and achieving efficient chondrogenesis and sustained tissue homeostasis, which are essential for the repair of hyaline cartilage. Despite advances, the effective delivery of bioactive factors is limited by challenges such as insufficient retention at the site of injury and the loss of therapeutic efficacy due to uncontrolled drug release. These limitations have prompted research on biomolecule-scaffold interactions to develop advanced delivery systems that provide sustained release and controlled bioavailability of biological factors, thereby improving therapeutic outcomes. This review focuses specifically on biomaterials (natural, hybrid and synthetic) and biomolecules (molecules, proteins, nucleic acids) of interest for cartilage engineering. Herein, we review in detail the approaches developed to maintain the biomolecules in scaffolds and control their release, based on their chemical nature and structure, through steric, non-covalent and/or covalent interactions, with a view to their application in cartilage repair.
Collapse
Affiliation(s)
- Silouane Dupuy
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Jérémy Salvador
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
- EPHE, PSL Research University, 75014 Paris, France
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Marie Morille
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Emmanuel Belamie
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
- EPHE, PSL Research University, 75014 Paris, France
| |
Collapse
|
2
|
Kronemberger GS, Spagnuolo FD, Karam AS, Chattahy K, Storey KJ, Kelly DJ. Growth Factor Stimulation Regimes to Support the Development and Fusion of Cartilage Microtissues. Tissue Eng Part C Methods 2025; 31:36-48. [PMID: 39813639 DOI: 10.1089/ten.tec.2024.0309] [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] [Indexed: 01/18/2025] Open
Abstract
Scaffold-free tissue engineering strategies using cellular aggregates, microtissues, or organoids as "biological building blocks" could potentially be used for the engineering of scaled-up articular cartilage or endochondral bone-forming grafts. Such approaches require large numbers of cells; however, little is known about how different chondrogenic growth factor stimulation regimes during cellular expansion and differentiation influence the capacity of cellular aggregates or microtissues to fuse and generate hyaline cartilage. In this study, human bone marrow mesenchymal stem/stromal cells (MSCs) were additionally stimulated with bone morphogenetic protein 2 (BMP-2) and/or transforming growth factor (TGF)-β1 during both monolayer expansion and subsequent chondrogenic differentiation in a microtissue format. MSCs displayed a higher proliferative potential when expanded in the presence of TGF-β1 or TGF-β1 and BMP-2. Next, the chondrogenic potential of these human MSCs was explored in a medium-high throughput microtissue system. After 3 weeks of culture, MSCs stimulated with BMP-2 during expansion and differentiation deposited higher levels of glycosaminoglycans (GAGs) and collagen, while staining negative for calcium deposits. The fusion capacity of the microtissues was not impacted by these different growth factor stimulation regimes. After 3 weeks of fusion, it was observed that MSCs stimulated with TGF-β1 during expansion and additionally with BMP-2 during chondrogenic differentiation deposited the highest levels of sulfated GAGs. No increase in type X collagen deposition was observed with additional growth factor stimulation. This study demonstrates the importance of carefully optimizing MSC expansion and differentiation conditions when developing modular tissue engineering strategies (e.g., cellular aggregates and microtissues) for cartilage tissue engineering applications.
Collapse
Affiliation(s)
- Gabriela S Kronemberger
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Francesca D Spagnuolo
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Aliaa S Karam
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kaoutar Chattahy
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kyle J Storey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Royal 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
|
3
|
Tymińska A, Karska N, Skoniecka A, Zawrzykraj M, Banach-Kopeć A, Mania S, Zieliński J, Kondej K, Gurzawska-Comis K, Skowron PM, Tylingo R, Rodziewicz-Motowidło S, Pikuła M. A novel chitosan-peptide system for cartilage tissue engineering with adipose-derived stromal cells. Biomed Pharmacother 2024; 181:117683. [PMID: 39561590 DOI: 10.1016/j.biopha.2024.117683] [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: 08/23/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
The natural healing process of cartilage injuries often fails to fully restore the tissue's biological and mechanical functions. Cartilage grafts are costly and require surgical intervention, often associated with complications such as intraoperative infection and rejection by the recipient due to ischemia. Novel tissue engineering technologies aim to ideally fill the cartilage defect to prevent disease progression or regenerate damaged tissue. Despite many studies on designing biocompatible composites to stimulate chondrogenesis, only few focus on peptides and carriers that promote stem cell proliferation or differentiation to promote healing. Our research aimed to design a carbohydrate chitosan-based biomaterial to stimulate stem cells into the chondrogenesis pathway. Our strategy was to combine chitosan with a novel peptide (UG28) that sequence was based on the copin protein. The construct stimulated human adipose-derived stem cells (AD-SCs) cells to undergo chondrogenic differentiation. Chitosan 75/500 allows AD-SCs to grow and has no harmful effects on the cells. The combination of UG28 peptide with the chitosan composite offers promising properties for cell differentiation, indicating its potential for clinical applications in cartilage regeneration.
Collapse
Affiliation(s)
- Agata Tymińska
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Department of Anatomy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk 80-211, Poland.
| | - Natalia Karska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk 80-308, Poland
| | - Aneta Skoniecka
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Department of Anatomy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk 80-211, Poland
| | - Małgorzata Zawrzykraj
- Division of Clinical Anatomy, Department of Anatomy, Medical University of Gdańsk, 80-211, Poland
| | - Adrianna Banach-Kopeć
- Department of Chemistry, Technology and Biotechnology of Food Gdańsk University of Technology, Gdańsk 80-233, Poland
| | - Szymon Mania
- Department of Chemistry, Technology and Biotechnology of Food Gdańsk University of Technology, Gdańsk 80-233, Poland
| | - Jacek Zieliński
- Department of Oncologic Surgery, Medical University of Gdańsk, Gdańsk 80-214, Poland
| | - Karolina Kondej
- Department of Plastic Surgery, Medical University of Gdańsk, Gdańsk 80-214, Poland
| | - Katarzyna Gurzawska-Comis
- Department of Dentistry and Oral Health, Aarhus University, Vennelyst Boulevard 9, Aarhus C DK-8000, Denmark
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, 80-308, Poland
| | - Robert Tylingo
- Department of Chemistry, Technology and Biotechnology of Food Gdańsk University of Technology, Gdańsk 80-233, Poland
| | | | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Department of Anatomy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk 80-211, Poland.
| |
Collapse
|
4
|
Wang M, Wang J, Xu X, Li E, Xu P. Engineering gene-activated bioprinted scaffolds for enhancing articular cartilage repair. Mater Today Bio 2024; 29:101351. [PMID: 39649247 PMCID: PMC11621797 DOI: 10.1016/j.mtbio.2024.101351] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/10/2024] Open
Abstract
Untreated articular cartilage injuries often result in severe chronic pain and dyskinesia. Current repair strategies have limitations in effectively promoting articular cartilage repair, underscoring the need for innovative therapeutic approaches. A gene-activated matrix (GAM) is a promising and comprehensive therapeutic strategy that integrates tissue-engineered scaffold-guided gene therapy to promote long-term articular cartilage repair by enhancing gene retention, reducing gene loss, and regulating gene release. However, for effective articular cartilage repair, the GAM scaffold must mimic the complex gradient structure of natural articular cartilage. Three-dimensional (3D) bioprinting technology has emerged as a compelling solution, offering the ability to precisely create complex microstructures that mimic the natural articular cartilage. In this review, we summarize the recent research progress on GAM and 3D bioprinted scaffolds in articular cartilage tissue engineering (CTE), while also exploring future challenges and development directions. This review aims to provide new ideas and concepts for the development of gene-activated bioprinted scaffolds with specific properties tailored to meet the stringent requirements of articular cartilage repair.
Collapse
Affiliation(s)
- Min Wang
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, 710000, China
| | - Jiachen Wang
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, 710000, China
| | - Xin Xu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, 710000, China
| | - Erliang Li
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, 710000, China
| | - Peng Xu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, 710000, China
| |
Collapse
|
5
|
Bandyopadhyay A, Ghibhela B, Mandal BB. Current advances in engineering meniscal tissues: insights into 3D printing, injectable hydrogels and physical stimulation based strategies. Biofabrication 2024; 16:022006. [PMID: 38277686 DOI: 10.1088/1758-5090/ad22f0] [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: 09/15/2023] [Accepted: 01/26/2024] [Indexed: 01/28/2024]
Abstract
The knee meniscus is the cushioning fibro-cartilage tissue present in between the femoral condyles and tibial plateau of the knee joint. It is largely avascular in nature and suffers from a wide range of tears and injuries caused by accidents, trauma, active lifestyle of the populace and old age of individuals. Healing of the meniscus is especially difficult due to its avascularity and hence requires invasive arthroscopic approaches such as surgical resection, suturing or implantation. Though various tissue engineering approaches are proposed for the treatment of meniscus tears, three-dimensional (3D) printing/bioprinting, injectable hydrogels and physical stimulation involving modalities are gaining forefront in the past decade. A plethora of new printing approaches such as direct light photopolymerization and volumetric printing, injectable biomaterials loaded with growth factors and physical stimulation such as low-intensity ultrasound approaches are being added to the treatment portfolio along with the contemporary tear mitigation measures. This review discusses on the necessary design considerations, approaches for 3D modeling and design practices for meniscal tear treatments within the scope of tissue engineering and regeneration. Also, the suitable materials, cell sources, growth factors, fixation and lubrication strategies, mechanical stimulation approaches, 3D printing strategies and injectable hydrogels for meniscal tear management have been elaborated. We have also summarized potential technologies and the potential framework that could be the herald of the future of meniscus tissue engineering and repair approaches.
Collapse
Affiliation(s)
- Ashutosh Bandyopadhyay
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Baishali Ghibhela
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Biman B Mandal
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| |
Collapse
|
6
|
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
|
7
|
He S, Deng H, Li P, Hu J, Yang Y, Xu Z, Liu S, Guo W, Guo Q. Arthritic Microenvironment-Dictated Fate Decisions for Stem Cells in Cartilage Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207715. [PMID: 37518822 PMCID: PMC10520688 DOI: 10.1002/advs.202207715] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 06/05/2023] [Indexed: 08/01/2023]
Abstract
The microenvironment and stem cell fate guidance of post-traumatic articular cartilage regeneration is primarily the focus of cartilage tissue engineering. In articular cartilage, stem cells are characterized by overlapping lineages and uneven effectiveness. Within the first 12 weeks after trauma, the articular inflammatory microenvironment (AIME) plays a decisive role in determining the fate of stem cells and cartilage. The development of fibrocartilage and osteophyte hyperplasia is an adverse outcome of chronic inflammation, which results from an imbalance in the AIME during the cartilage tissue repair process. In this review, the sources for the different types of stem cells and their fate are summarized. The main pathophysiological events that occur within the AIME as well as their protagonists are also discussed. Additionally, regulatory strategies that may guide the fate of stem cells within the AIME are proposed. Finally, strategies that provide insight into AIME pathophysiology are discussed and the design of new materials that match the post-traumatic progress of AIME pathophysiology in a spatial and temporal manner is guided. Thus, by regulating an appropriately modified inflammatory microenvironment, efficient stem cell-mediated tissue repair may be achieved.
Collapse
Affiliation(s)
- Songlin He
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Haotian Deng
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Peiqi Li
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Jingjing Hu
- Department of GastroenterologyInstitute of GeriatricsChinese PLA General HospitalBeijing100853China
| | - Yongkang Yang
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Ziheng Xu
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Shuyun Liu
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| | - Weimin Guo
- Department of Orthopaedic SurgeryGuangdong Provincial Key Laboratory of Orthopedics and TraumatologyFirst Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510080China
| | - Quanyi Guo
- School of MedicineNankai UniversityTianjin300071China
- Institute of Orthopedicsthe First Medical CenterChinese PLA General HospitalBeijing Key Lab of Regenerative Medicine in OrthopedicsKey Laboratory of Musculoskeletal Trauma & War Injuries PLABeijing100853China
| |
Collapse
|
8
|
Hammad M, Veyssiere A, Leclercq S, Patron V, Baugé C, Boumédiene K. Hypoxia Differentially Affects Chondrogenic Differentiation of Progenitor Cells from Different Origins. Int J Stem Cells 2023; 16:304-314. [PMID: 37105555 PMCID: PMC10465331 DOI: 10.15283/ijsc21242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 04/29/2023] Open
Abstract
Background and Objectives Ear cartilage malformations are commonly encountered problems in reconstructive surgery, since cartilage has low self-regenerating capacity. Malformations that impose psychological and social burden on one's life are currently treated using ear prosthesis, synthetic implants or autologous flaps from rib cartilage. These approaches are challenging because not only they request high surgical expertise, but also they lack flexibility and induce severe donor-site morbidity. Through the last decade, tissue engineering gained attention where it aims at regenerating human tissues or organs in order to restore normal functions. This technique consists of three main elements, cells, growth factors, and above all, a scaffold that supports cells and guides their behavior. Several studies have investigated different scaffolds prepared from both synthetic or natural materials and their effects on cellular differentiation and behavior. Methods and Results In this study, we investigated a natural scaffold (alginate) as tridimensional hydrogel seeded with progenitors from different origins such as bone marrow, perichondrium and dental pulp. In contact with the scaffold, these cells remained viable and were able to differentiate into chondrocytes when cultured in vitro. Quantitative and qualitative results show the presence of different chondrogenic markers as well as elastic ones for the purpose of ear cartilage, upon different culture conditions. Conclusions We confirmed that auricular perichondrial cells outperform other cells to produce chondrogenic tissue in normal oxygen levels and we report for the first time the effect of hypoxia on these cells. Our results provide updates for cartilage engineering for future clinical applications.
Collapse
Affiliation(s)
- Mira Hammad
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
| | - Alexis Veyssiere
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
- Service de chirurgie Maxillo-faciale, CHU de Caen, Caen, France
| | - Sylvain Leclercq
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Clinique Saint Martin, Service de Chirurgie Orthopédique, Caen, France
| | - Vincent Patron
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Service ORL et chirurgie cervico-faciale, CHU de Caen, Caen, France
| | - Catherine Baugé
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
| | - Karim Boumédiene
- Normandy University, UNICAEN, EA 7451 BioConnecT, Caen, France
- Fédération Hospitalo Universitaire SURFACE, Amiens, Caen, France
| |
Collapse
|
9
|
Jiang Y, Tuan RS. Bioactivity of human adult stem cells and functional relevance of stem cell-derived extracellular matrix in chondrogenesis. Stem Cell Res Ther 2023; 14:160. [PMID: 37316923 DOI: 10.1186/s13287-023-03392-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 05/31/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Autologous chondrocyte implantation (ACI) has been used to treat articular cartilage defects for over two decades. Adult stem cells have been proposed as a solution to inadequate donor cell numbers often encountered in ACI. Multipotent stem/progenitor cells isolated from adipose, bone marrow, and cartilage are the most promising cell therapy candidates. However, different essential growth factors are required to induce these tissue-specific stem cells to initiate chondrogenic differentiation and subsequent deposition of extracellular matrix (ECM) to form cartilage-like tissue. Upon transplantation into cartilage defects in vivo, the levels of growth factors in the host tissue are likely to be inadequate to support chondrogenesis of these cells in situ. The contribution of stem/progenitor cells to cartilage repair and the quality of ECM produced by the implanted cells required for cartilage repair remain largely unknown. Here, we evaluated the bioactivity and chondrogenic induction ability of the ECM produced by different adult stem cells. METHODS Adult stem/progenitor cells were isolated from human adipose (hADSCs), bone marrow (hBMSCs), and articular cartilage (hCDPCs) and cultured for 14 days in monolayer in mesenchymal stromal cell (MSC)-ECM induction medium to allow matrix deposition and cell sheet formation. The cell sheets were then decellularized, and the protein composition of the decellularized ECM (dECM) was analyzed by BCA assay, SDS-PAGE, and immunoblotting for fibronectin (FN), collagen types I (COL1) and III (COL3). The chondrogenic induction ability of the dECM was examined by seeding undifferentiated hBMSCs onto the respective freeze-dried solid dECM followed by culturing in serum-free medium for 7 days. The expression levels of chondrogenic genes SOX9, COL2, AGN, and CD44 were analyzed by q-PCR. RESULTS hADSCs, hBMSCs, and hCDPCs generated different ECM protein profiles and exhibited significantly different chondrogenic effects. hADSCs produced 20-60% more proteins than hBMSCs and hCDPCs and showed a fibrillar-like ECM pattern (FNhigh, COL1high). hCDPCs produced more COL3 and deposited less FN and COL1 than the other cell types. The dECM derived from hBMSCs and hCDPCs induced spontaneous chondrogenic gene expression in hBMSCs. CONCLUSIONS These findings provide new insights on application of adult stem cells and stem cell-derived ECM to enhance cartilage regeneration.
Collapse
Affiliation(s)
- Yangzi Jiang
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China.
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China.
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
10
|
Zhang C, Wang G, Lin H, Shang Y, Liu N, Zhen Y, An Y. Cartilage 3D bioprinting for rhinoplasty using adipose-derived stem cells as seed cells: Review and recent advances. Cell Prolif 2023; 56:e13417. [PMID: 36775884 PMCID: PMC10068946 DOI: 10.1111/cpr.13417] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/10/2023] [Accepted: 01/18/2023] [Indexed: 02/14/2023] Open
Abstract
Nasal deformities due to various causes affect the aesthetics and use of the nose, in which case rhinoplasty is necessary. However, the lack of cartilage for grafting has been a major problem and tissue engineering seems to be a promising solution. 3D bioprinting has become one of the most advanced tissue engineering methods. To construct ideal cartilage, bio-ink, seed cells, growth factors and other methods to promote chondrogenesis should be considered and weighed carefully. With continuous progress in the field, bio-ink choices are becoming increasingly abundant, from a single hydrogel to a combination of hydrogels with various characteristics, and more 3D bioprinting methods are also emerging. Adipose-derived stem cells (ADSCs) have become one of the most popular seed cells in cartilage 3D bioprinting, owing to their abundance, excellent proliferative potential, minimal morbidity during harvest and lack of ethical considerations limitations. In addition, the co-culture of ADSCs and chondrocytes is commonly used to achieve better chondrogenesis. To promote chondrogenic differentiation of ADSCs and construct ideal highly bionic tissue-engineered cartilage, researchers have used a variety of methods, including adding appropriate growth factors, applying biomechanical stimuli and reducing oxygen tension. According to the process and sequence of cartilage 3D bioprinting, this review summarizes and discusses the selection of hydrogel and seed cells (centered on ADSCs), the design of printing, and methods for inducing the chondrogenesis of ADSCs.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Guanhuier Wang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Hongying Lin
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Yujia Shang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China.,Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Na Liu
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China.,Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yonghuan Zhen
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| |
Collapse
|
11
|
Katiyar S, Singh D, Kumari S, Srivastava P, Mishra A. Novel strategies for designing regenerative skin products for accelerated wound healing. 3 Biotech 2022; 12:316. [PMID: 36276437 PMCID: PMC9547767 DOI: 10.1007/s13205-022-03331-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/23/2022] [Indexed: 11/01/2022] Open
Abstract
Healthy skin protects from pathogens, water loss, ultraviolet rays, and also maintains homeostasis conditions along with sensory perceptions in normal circumstances. Skin wound healing mechanism is a multi-phased biodynamic process that ultimately triggers intercellular and intracellular mechanisms. Failure to implement the normal and effective healing process may result in chronic injuries and aberrant scarring. Chronic wounds lead to substantial rising healthcare expenditure, and innovative methods to diagnose and control severe consequences are urgently needed. Skin tissue engineering (STE) has achieved several therapeutic accomplishments during the last few decades, demonstrating tremendous development. The engineered skin substitutes provide instant coverage for extensive wounds and facilitate the prevention of microbial infections and fluid loss; furthermore, they help in fighting inflammation and allow rapid neo-tissue formation. The current review primarily focused on the wound recovery and restoration process and the current conditions of STE with various advancements and complexities associated with different strategies such as cell sources, biopolymers, innovative fabrication techniques, and growth factors delivery systems.
Collapse
Affiliation(s)
- Soumya Katiyar
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005 India
| | - Divakar Singh
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005 India
| | - Shikha Kumari
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005 India
| | - Pradeep Srivastava
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005 India
| | - Abha Mishra
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005 India
| |
Collapse
|
12
|
|
13
|
He SK, Ning LJ, Hu RN, Yao X, Cui J, Ding W, Luo JC, Qin TW. Segmentally Demineralized Cortical Bone With Stem Cell-Derived Matrix Promotes Proliferation, Migration and Differentiation of Stem Cells in vitro. Front Cell Dev Biol 2022; 9:776884. [PMID: 35155445 PMCID: PMC8826562 DOI: 10.3389/fcell.2021.776884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/31/2021] [Indexed: 11/13/2022] Open
Abstract
A recent study has shown that demineralized cortical bone (DCB) did not improve the healing of tendon-bone interface. Considering that there is a gradient of mineral content in the tendon-bone interface, we designed a segmentally demineralized cortical bone (sDCB) scaffold with two different regions: undemineralized cortical bone section within the scaffold (sDCB-B) and complete demineralized cortical bone section within the scaffold (sDCB-D), to mimic the natural structure of the tendon-bone interface. Furthermore, the extracellular matrix (ECM) from tendon-derived stem cells (TDSCs) was used to modify the sDCB-D region of sDCB to construct a novel scaffold (sDCB-ECM) for enhancing the bioactivity of the sDCB-D. The surface topography, elemental distribution, histological structure, and surface elastic modulus of the scaffold were observed using scanning electron microscopy, energy-dispersive X-ray spectroscopy, Fourier transform infrared spectroscopy, histological staining and atomic force microscopy. Cell proliferation of bone marrow mesenchymal stem cells (BMSCs) and TDSCs cultured on scaffolds was evaluated using the Cell Counting kit-8, and cell viability was assessed by Live/Dead cell staining. Cell morphology was detected by fluorescent staining. The ability of the scaffolds to recruit stem cells was tested using transwell migration assay. The expression levels of bone-, cartilage- and tendon-related genes and proteins in stem cells were assessed by the polymerase chain reaction and western blotting. Our results demonstrated that there was a gradient of Ca and P elements in sDCB, and TDSC-derived ECM existed on the surface of the sDCB-D region of sDCB. The sDCB-ECM could promote stem cell proliferation and migration. Moreover, the sDCB-B region of sDCB-ECM could stimulate osteogenic and chondrogenic differentiation of BMSCs, and the sDCB-D-ECM region of sDCB-ECM could stimulate chondrogenic and tenogenic differentiation of TDSCs when compared to DCB. Our study indicated that sDCB-ECM might be a potential bioscaffold to enhance the tendon-bone interface regeneration.
Collapse
Affiliation(s)
- Shu-Kun He
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Orthopedic Research Institute, Sichuan University, Chengdu, China
- Department of Orthopedics, West China Hospital, Orthopedic Research Institute, Sichuan University, Chengdu, China
- Department of Orthopedics, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Liang-Ju Ning
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Orthopedic Research Institute, Sichuan University, Chengdu, China
| | - Ruo-Nan Hu
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Orthopedic Research Institute, Sichuan University, Chengdu, China
| | - Xuan Yao
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Orthopedic Research Institute, Sichuan University, Chengdu, China
- Department of Clinical Hematology, Faculty of Laboratory Medicine, Army Medical University, Chongqing, China
| | - Jing Cui
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Orthopedic Research Institute, Sichuan University, Chengdu, China
| | - Wei Ding
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Orthopedic Research Institute, Sichuan University, Chengdu, China
| | - Jing-Cong Luo
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Orthopedic Research Institute, Sichuan University, Chengdu, China
| | - Ting-Wu Qin
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Orthopedic Research Institute, Sichuan University, Chengdu, China
- *Correspondence: Ting-Wu Qin,
| |
Collapse
|
14
|
Walters B, Turner PA, Rolauffs B, Hart ML, Stegemann JP. Controlled Growth Factor Delivery and Cyclic Stretch Induces a Smooth Muscle Cell-like Phenotype in Adipose-Derived Stem Cells. Cells 2021; 10:cells10113123. [PMID: 34831345 PMCID: PMC8624888 DOI: 10.3390/cells10113123] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 01/02/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are an abundant and easily accessible multipotent stem cell source with potential application in smooth muscle regeneration strategies. In 3D collagen hydrogels, we investigated whether sustained release of growth factors (GF) PDGF-AB and TGF-β1 from GF-loaded microspheres could induce a smooth muscle cell (SMC) phenotype in ASCs, and if the addition of uniaxial cyclic stretch could enhance the differentiation level. This study demonstrated that the combination of cyclic stretch and GF release over time from loaded microspheres potentiated the differentiation of ASCs, as quantified by protein expression of early to late SMC differentiation markers (SMA, TGLN and smooth muscle MHC). The delivery of GFs via microspheres produced large ASCs with a spindle-shaped, elongated SMC-like morphology. Cyclic strain produced the largest, longest, and most spindle-shaped cells regardless of the presence or absence of growth factors or the growth factor delivery method. Protein expression and cell morphology data confirmed that the sustained release of GFs from GF-loaded microspheres can be used to promote the differentiation of ASCs into SMCs and that the addition of uniaxial cyclic stretch significantly enhances the differentiation level, as quantified by intermediate and late SMC markers and a SMC-like elongated cell morphology.
Collapse
Affiliation(s)
- Brandan Walters
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Paul A. Turner
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Bernd Rolauffs
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
| | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| |
Collapse
|
15
|
He X, Ao H, Qiao Y, Li Z. 3D-printed porous scaffold promotes osteogenic differentiation of hADMSCs. Open Med (Wars) 2021. [PMID: 33521318 PMCID: PMC7811365 DOI: 10.1515/med-2021-0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Objective
To explore the role of a three-dimensional (3D)-printed porous titanium alloy scaffold (3D scaffold) in the osteogenic differentiation of human adipose-derived mesenchymal stem cells (hADMSCs) and the underlying mechanism.
Methods
hADMSCs were divided into control and 3D scaffold groups. The osteogenic differentiation of hADMSCs and expression of osteogenic makers were estimated. Based on the information from published articles, five candidate circular RNAs were selected, and among them, hsa_circ_0019142 showed the most promising results. Finally, control group cells were overexpressed or silenced with the hsa_circ_0019142. Then, Alizarin red S (ARS) staining, calcium content analysis and estimation of alkaline phosphatase (ALP), osteocalcin (OCN), runt-related transcription factor 2 (RUNX2), and collagen-1 (COL1) were performed to evaluate the role of hsa_circ_0019142 on osteogenic differentiation.
Results
Osteogenic differentiation of the hADMSCs was significantly higher in the 3D scaffold group than in the control group, as evidenced by ARS staining, increased calcium concentration, and elevated expression of above four osteogenic factors. qPCR revealed that the expression of hsa_circ_0019142 was significantly higher in the 3D scaffold group. Overexpression of hsa_circ_0019142 promoted the osteogenic differentiation of hADMSCs, while knockdown of hsa_circ_0019142 caused the opposite results.
Conclusion
The 3D-printed scaffold promoted osteogenic differentiation of hADMSCs by upregulating hsa_circ_0019142.
Collapse
Affiliation(s)
- Xuebin He
- Ear-Nose-Throat Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Huafei Ao
- Ear-Nose-Throat Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Ying Qiao
- Ear-Nose-Throat Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Zhengwen Li
- Ear-Nose-Throat Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| |
Collapse
|
16
|
Rahman S, Szojka ARA, Liang Y, Kunze M, Goncalves V, Mulet-Sierra A, Jomha NM, Adesida AB. Inability of Low Oxygen Tension to Induce Chondrogenesis in Human Infrapatellar Fat Pad Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:703038. [PMID: 34381784 PMCID: PMC8350173 DOI: 10.3389/fcell.2021.703038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Articular cartilage of the knee joint is avascular, exists under a low oxygen tension microenvironment, and does not self-heal when injured. Human infrapatellar fat pad-sourced mesenchymal stem cells (IFP-MSC) are an arthroscopically accessible source of mesenchymal stem cells (MSC) for the repair of articular cartilage defects. Human IFP-MSC exists physiologically under a low oxygen tension (i.e., 1-5%) microenvironment. Human bone marrow mesenchymal stem cells (BM-MSC) exist physiologically within a similar range of oxygen tension. A low oxygen tension of 2% spontaneously induced chondrogenesis in micromass pellets of human BM-MSC. However, this is yet to be demonstrated in human IFP-MSC or other adipose tissue-sourced MSC. In this study, we explored the potential of low oxygen tension at 2% to drive the in vitro chondrogenesis of IFP-MSC. We hypothesized that 2% O2 will induce stable chondrogenesis in human IFP-MSC without the risk of undergoing endochondral ossification at ectopic sites of implantation. METHODS Micromass pellets of human IFP-MSC were cultured under 2% O2 or 21% O2 (normal atmosphere O2) in the presence or absence of chondrogenic medium with transforming growth factor-β3 (TGFβ3) for 3 weeks. Following in vitro chondrogenesis, the resulting pellets were implanted in immunodeficient athymic nude mice for 3 weeks. RESULTS A low oxygen tension of 2% was unable to induce chondrogenesis in human IFP-MSC. In contrast, chondrogenic medium with TGFβ3 induced in vitro chondrogenesis. All pellets were devoid of any evidence of undergoing endochondral ossification after subcutaneous implantation in athymic mice.
Collapse
Affiliation(s)
- Samia Rahman
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Alexander R. A. Szojka
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Yan Liang
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Victoria Goncalves
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Nadr M. Jomha
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Adetola B. Adesida
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of Alberta Hospital, Edmonton, AB, Canada
| |
Collapse
|
17
|
Rauch A, Mandrup S. Transcriptional networks controlling stromal cell differentiation. Nat Rev Mol Cell Biol 2021; 22:465-482. [PMID: 33837369 DOI: 10.1038/s41580-021-00357-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 02/02/2023]
Abstract
Stromal progenitors are found in many different tissues, where they play an important role in the maintenance of tissue homeostasis owing to their ability to differentiate into parenchymal cells. These progenitor cells are differentially pre-programmed by their tissue microenvironment but, when cultured and stimulated in vitro, these cells - commonly referred to as mesenchymal stromal cells (MSCs) - exhibit a marked plasticity to differentiate into many different cell lineages. Loss-of-function studies in vitro and in vivo have uncovered the involvement of specific signalling pathways and key transcriptional regulators that work in a sequential and coordinated fashion to activate lineage-selective gene programmes. Recent advances in omics and single-cell technologies have made it possible to obtain system-wide insights into the gene regulatory networks that drive lineage determination and cell differentiation. These insights have important implications for the understanding of cell differentiation, the contribution of stromal cells to human disease and for the development of cell-based therapeutic applications.
Collapse
Affiliation(s)
- Alexander Rauch
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark. .,Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark.
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity, Functional Genomics & Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
18
|
Zhang Y, Grant RA, Shivakumar MK, Zaleski M, Sofoluke N, Slotkin JR, Williams MS, Lee MTM. Genome-wide Association Analysis Across 16,956 Patients Identifies a Novel Genetic Association Between BMP6, NIPAL1, CNGA1 and Spondylosis. Spine (Phila Pa 1976) 2021; 46:E625-E631. [PMID: 33332786 DOI: 10.1097/brs.0000000000003880] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN A case-control genome-wide association study (GWAS) on spondylosis. OBJECTIVE Leveraging Geisinger's MyCode initiative's multimodal dataset, we aimed to identify genetic associations with degenerative spine disease. SUMMARY OF BACKGROUND DATA Degenerative spine conditions are a leading cause of global disability; however, the genetic underpinnings of these conditions remain under-investigated. Previous studies using candidate-gene approach suggest a genetic risk for degenerative spine conditions, but large-scale GWASs are lacking. METHODS We identified 4434 patients with a diagnosis of spondylosis using ICD diagnosis codes with genotype data available. We identified a population-based control of 12,522 patients who did not have any diagnosis for osteoarthritis. A linear-mix, additive genetic model was employed to perform the genetic association tests adjusting for age, sex, and genetic principal components to account for the population structure and relatedness. Gene-based association tests were performed and heritability and genetic correlations with other traits were investigated. RESULTS We identified a genome-wide significant locus at rs12190551 (odds ratio = 1.034, 95% confidence interval 1.022-1.046, P = 8.5 × 10-9, minor allele frequency = 36.9%) located in the intron of BMP6. Additionally, NIPAL1 and CNGA1 achieved Bonferroni significance in the gene-based association tests. The estimated heritability was 7.19%. Furthermore, significant genetic correlations with pain, depression, lumbar spine bone mineral density, and osteoarthritis were identified. CONCLUSION We demonstrated the use of a massive database of genotypes combined with electronic health record data to identify a novel and significant association spondylosis. We also identified significant genetic correlations with pain, depression, bone mineral density, and osteoarthritis, suggesting shared genetic etiology and molecular pathways with these phenotypes.Level of Evidence: N/A.
Collapse
Affiliation(s)
- Yanfei Zhang
- Genomic Medicine Institute, Geisinger, Danville, PA
- Genomic Research, Musculoskeletal Institute, Geisinger, Danville, PA
| | - Ryan A Grant
- Department of Neurosurgery, Geisinger, Danville, PA
| | - Manu K Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA
| | | | | | | | | | - Ming Ta Michael Lee
- Genomic Medicine Institute, Geisinger, Danville, PA
- Genomic Research, Musculoskeletal Institute, Geisinger, Danville, PA
| |
Collapse
|
19
|
Wu KC, Weng HK, Hsu YS, Huang PJ, Wang YK. Aqueous extract of Arctium lappa L. root (burdock) enhances chondrogenesis in human bone marrow-derived mesenchymal stem cells. BMC Complement Med Ther 2020; 20:364. [PMID: 33228629 PMCID: PMC7686739 DOI: 10.1186/s12906-020-03158-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 11/13/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Arctium lappa L. root (burdock root) has long been recommended for the treatment of different diseases in traditional Chinese medicine. Burdock root possesses anti-oxidative, anti-inflammatory, anti-cancer, and anti-microbial activities. The aim of the study was to elucidate whether aqueous extract of burdock root regulates mesenchymal stem cell proliferation and differentiation. METHODS Human bone marrow-derived mesenchymal stem cells in 2D high density culture and in 3D micromass pellets were treated with chondrogenic induction medium and chondral basal medium in the absence or presence of aqueous extract of burdock root. The chondrogenic differentiation was accessed by staining glucosaminoglycans, immunostaining SOX9 and type II collagen and immuonblotting of SOX9, aggrecan and type II collagen. RESULTS Treatment of aqueous extract of burdock root increased the cell proliferation of hMSCs. It did not have significant effect on osteogenic and adipogenic differentiation, but significantly enhanced chondrogenic induction medium-induced chondrogenesis. The increment was dose dependent, as examined by staining glucosaminoglycans, SOX9, and type II collagen and immunobloting of SOX9, aggrecan and type II collagen in 2D and 3D cultures. In the presence of supplemental materials, burdock root aqueous extract showed equivalent chondrogenic induction capability to that of TGF-β. CONCLUSIONS The results demonstrate that aqueous extract of Arctium lappa L. root promotes chondrogenic medium-induced chondrogenic differentiation. The aqueous extract of burdock root can even be used alone to stimulate chondrogenic differentiation. The study suggests that the aqueous extract of burdock root can be used as an alternative strategy for treatment purposes.
Collapse
Affiliation(s)
- King-Chuen Wu
- Department of Nursing, Chang Gung University of Science and Technology, Chia-Yi County, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Chiayi County, Taiwan
| | - Hung-Kai Weng
- Department of Orthopaedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Yun-Shang Hsu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Pin-Jia Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Yang-Kao Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan. .,Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan City, Taiwan.
| |
Collapse
|
20
|
Cartilage repair using stem cells & biomaterials: advancement from bench to bedside. Mol Biol Rep 2020; 47:8007-8021. [PMID: 32888123 DOI: 10.1007/s11033-020-05748-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 08/28/2020] [Indexed: 10/23/2022]
Abstract
Osteoarthritis (OA) involves gradual destruction of articular cartilagemanifested by pain, stiffness of joints, and impaired movement especially in knees and hips. Non-vascularity of this tissue hinders its self-regenerative capacity and thus, the application of reparative or restorative modalities becomes imperative in OA treatment. In recent years, stem cell-based therapies have been explored as potential modalities for addressing OA complications. While mesenchymal stem cells (MSCs) hold immense promise, the recapitulation of native articular cartilage usingMSCs remains elusive. In this review, we have highlighted the chondrogenic potential of MSCs, factors guiding in vitro chondrogenic differentiation, biomaterials available for cartilage repair, their current market status, and the outcomes of major clinical trials. Our search on ClinicalTrials.gov using terms "stem cell" and "osteoarthritis" yielded 83 results. An analysis of the 29 trials that have been completed revealed differences in source of MSCs (bone marrow, adipose tissue, umbilical cord etc.), cell type (autologous or allogenic), and dose administered. Moreover, only 02 out of 29 studies have reported the use of matrix for cartilage repair. From future perspective, aconsensus on choice of cells, differentiation inducers, biomaterials, and clinical settings might pave a way for concocting robust strategies to improve the clinical applicability of biomimetic neocartilage constructs.
Collapse
|
21
|
Degenerative osteoarthritis a reversible chronic disease. Regen Ther 2020; 15:149-160. [PMID: 33426213 PMCID: PMC7770340 DOI: 10.1016/j.reth.2020.07.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/09/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is the most common chronic musculoskeletal disorder. It can affect any joint and is the most frequent single cause of disability in older adults. OA is a progressive degenerative disease involving the entire joint structure in a vicious circle that includes the capsule-bursa tissue inflammation, synovial fluid modifications, cartilage breakdown and erosions, osteochondral inflammatory damage leading to bone erosion and distortion. Research has identified the initial inflammatory-immunologic process that starts this vicious cycle leading to so-called early OA. Research has also identified the role played in the disease advancement by synoviocytes type A and B, chondrocytes, extracellular matrix, local immune-inflammatory mediators and proteases. This article investigates the joint-resident MSCs that play an essential local homeostatic role and regulate cell turn over and tissue repair. Resident MSCs establish and maintain a local regenerative microenvironment. The understanding of OA physiopathology clarifies the core mechanisms by which minimally invasive interventions might be able to halt and reverse the course of early stage OA. Interventions employing PRP, MSCs and exosomes are considered in this article.
Collapse
|
22
|
An immortalized human adipose-derived stem cell line with highly enhanced chondrogenic properties. Biochem Biophys Res Commun 2020; 530:252-258. [PMID: 32828295 DOI: 10.1016/j.bbrc.2020.07.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/04/2020] [Indexed: 12/28/2022]
Abstract
Human adipose-derived stem cells (ASCs) are a commonly used cell type for cartilage tissue engineering. However, donor-to-donor variability, cell heterogeneity, inconsistent chondrogenic potential, and limited expansion potential can hinder the use of these cells for modeling chondrogenesis, in vitro screening of drugs and treatments for joint diseases, or translational applications for tissue engineered cartilage repair. The goal of this study was to create an immortalized ASC line that showed enhanced and consistent chondrogenic potential for applications in cartilage tissue engineering as well as to provide a platform for investigation of biological and mechanobiological pathways involved in cartilage homeostasis and disease. Starting with the ASC52telo cell line, a hTERT-immortalized ASC line, we used lentivirus to overexpress SOX9, a master regulator of chondrogenesis, and screened several clonal populations of SOX9 overexpressing cells to form a new stable cell line with high chondrogenic potential. One clonal line, named ASC52telo-SOX9, displayed increased GAG and type II collagen synthesis and was found to be responsive to both mechanical and inflammatory stimuli in a manner similar to native chondrocytes. The development of a clonal line such as ASC52telo-SOX9 has the potential to be a powerful tool for studying cartilage homeostasis and disease mechanisms in vitro, and potentially as a platform for in vitro drug screening for diseases that affect articular cartilage. Our findings provide an approach for the development of other immortalized cell lines with improved chondrogenic capabilities in ASCs or other adult stem cells.
Collapse
|
23
|
Dual Network Hydrogels Incorporated with Bone Morphogenic Protein-7-Loaded Hyaluronic Acid Complex Nanoparticles for Inducing Chondrogenic Differentiation of Synovium-Derived Mesenchymal Stem Cells. Pharmaceutics 2020; 12:pharmaceutics12070613. [PMID: 32630047 PMCID: PMC7407334 DOI: 10.3390/pharmaceutics12070613] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/19/2020] [Accepted: 06/26/2020] [Indexed: 01/30/2023] Open
Abstract
Alginate-poloxamer (ALG-POL) copolymer with optimal POL content was synthesized, and it was combined with silk fibroin (SF) for building ALG-POL/SF dual network hydrogels. Hyaluronic acid(HA)/chitosan-poly(dioxanone)(CH-PDO) complex nanoparticles (NPs) with optimized composition and high encapsulation efficiency were employed as a vehicle for loading bone morphogenic protein-7 (BMP-7). BMP-7-loaded HA/CH-PDO NPs were incorporated into ALG-POL/SF hydrogel for constructing composite gels to achieve controlled release of BMP-7. These gels showed thermosensitive sol-gel transitions near physiological temperature and pH; and they were tested to be elastic, tough and strong. Some gels exhibited abilities to administer the BMP-7 release in nearly linear manners for a few weeks. Synovium-derived mesenchymal stem cells (SMSCs) were seeded into optimally fabricated gels for assessing their chondrogenic differentiation potency. Real-time PCR analyses showed that the blank ALG-POL/SF gels were not able to induce the chondrogenic differentiation of SMSCs, whereas SMSCs were detected to significantly express cartilage-related genes once they were seeded in the BMP-7-loaded ALG-POL/SF gel for two weeks. The synthesis of cartilaginous matrix components further confirmed that SMSCs seeded in the BMP-7-loaded ALG-POL/SF gel differentiated toward chondrogenesis. Results suggest that BMP-7-loaded ALG-POL/SF composite gels can function as a promising biomaterial for cartilage tissue engineering applications.
Collapse
|
24
|
Tseng SJ, Huang ST, Wu CC, Cheng CH, Lin JC. Studies of proliferation and chondrogenic differentiation of rat adipose stem cells using an anti-oxidative polyurethane scaffold combined with cyclic compression culture. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 112:110964. [PMID: 32409092 DOI: 10.1016/j.msec.2020.110964] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/21/2020] [Accepted: 04/11/2020] [Indexed: 02/08/2023]
Abstract
The adipose stem cell is a potential candidate for the autologous chondrocytes repairing approach because of the abundance of fat in the animal body and its versatile differentiation capability. In this study, rat adipose stem cells (rASCs) were seeded into anti-oxidative N-acetylcysteine (NAC) grafted polyurethane (PU) scaffold and then combined with short dynamic compressive stimulation (24 h) to induce rASCs chondrogenesis differentiation in vitro. The inner pore surface of the PU scaffold was first modified via alginate and type I collagen to promote rASCs adherence. The modified layers crosslinked by genipin showed outstanding stability after ultrasonic treatment, indicating the modified layers were stable and can keep the cells adhesion well during dynamic compressive stimulation. After inner pore surface modification and 10 mM NAC grafting, the PU scaffold-A-C-G (graft 10 mM NAC) has shown the best proliferation efficiency with homogeneous cell distribution after 72hr static culture. After short term dynamic compressive stimulation, significant gene expression in chondrogenic markers, Sox-9, and Aggrecan, were noted in both PU scaffold-A-C-G and PU scaffold-A-C-G (graft 10 mM NAC). Considering the cell proliferation efficiency and gene expression, the anti-oxidative NAC grafted PU scaffold combined with short term dynamic compressive stimulation could be useful for cell culturing in stem cell therapy.
Collapse
Affiliation(s)
- Shen-Jui Tseng
- Department of Chemical Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - Shih-Ting Huang
- Department of Chemical Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Hui Cheng
- Department of Pediatrics, College of Medicine, Chang Gung University, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
| | - Jui-Che Lin
- Department of Chemical Engineering, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
25
|
Lin Z, Li Z, Li EN, Li X, Del Duke CJ, Shen H, Hao T, O'Donnell B, Bunnell BA, Goodman SB, Alexander PG, Tuan RS, Lin H. Osteochondral Tissue Chip Derived From iPSCs: Modeling OA Pathologies and Testing Drugs. Front Bioeng Biotechnol 2019; 7:411. [PMID: 31921815 PMCID: PMC6930794 DOI: 10.3389/fbioe.2019.00411] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/27/2019] [Indexed: 01/17/2023] Open
Abstract
Osteoarthritis (OA) is a chronic disease mainly characterized by degenerative changes in cartilage, but other joint elements such as bone are also affected. To date, there are no disease-modifying OA drugs (DMOADs), owing in part to a deficiency of current models in simulating OA pathologies and etiologies in humans. In this study, we aimed to develop microphysiological osteochondral (OC) tissue chips derived from human induced pluripotent stem cells (iPSCs) to model the pathologies of OA. We first induced iPSCs into mesenchymal progenitor cells (iMPCs) and optimized the chondro- and osteo-inductive conditions for iMPCs. Then iMPCs were encapsulated into photocrosslinked gelatin scaffolds and cultured within a dual-flow bioreactor, in which the top stream was chondrogenic medium and the bottom stream was osteogenic medium. After 28 days of differentiation, OC tissue chips were successfully generated and phenotypes were confirmed by real time RT-PCR and histology. To create an OA model, interleukin-1β (IL-1β) was used to challenge the cartilage component for 7 days. While under control conditions, the bone tissue promoted chondrogenesis and suppressed chondrocyte terminal differentiation of the overlying chondral tissue. Under conditions modeling OA, the bone tissue accelerated the degradation of chondral tissue which is likely via the production of catabolic and inflammatory cytokines. These findings suggest active functional crosstalk between the bone and cartilage tissue components in the OC tissue chip under both normal and pathologic conditions. Finally, a selective COX-2 inhibitor commonly prescribed drug for OA, Celecoxib, was shown to downregulate the expression of catabolic and proinflammatory cytokines in the OA model, demonstrating the utility of the OC tissue chip model for drug screening. In summary, the iPSC-derived OC tissue chip developed in this study represents a high-throughput platform applicable for modeling OA and for the screening and testing of candidate DMOADs.
Collapse
Affiliation(s)
- Zixuan Lin
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhong Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Eileen N. Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
| | - Xinyu Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
| | - Colin J. Del Duke
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - He Shen
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Tingjun Hao
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
| | - Benjamen O'Donnell
- Department of Pharmacology, Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA, United States
| | - Bruce A. Bunnell
- Department of Pharmacology, Center for Stem Cell Research, Tulane University School of Medicine, New Orleans, LA, United States
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery and Bioengineering, Stanford University, Stanford, CA, United States
| | - Peter G. Alexander
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Rocky S. Tuan
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Hang Lin
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
26
|
Sivasubramaniyan K, Koevoet WJLM, Hakimiyan AA, Sande M, Farrell E, Hoogduijn MJ, Verhaar JAN, Chubinskaya S, Bühring HJ, van Osch GJVM. Cell-surface markers identify tissue resident multipotential stem/stromal cell subsets in synovial intimal and sub-intimal compartments with distinct chondrogenic properties. Osteoarthritis Cartilage 2019; 27:1831-1840. [PMID: 31536814 DOI: 10.1016/j.joca.2019.08.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 08/24/2019] [Accepted: 08/29/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Synovium contains multipotent progenitor/stromal cells (MPCs) with potential to participate in cartilage repair. Understanding the identity of these MPCs will allow their therapeutic potential to be fully exploited. Hence this study aimed to identify primary synovial MPCs and characterize them in the context of cartilage regeneration. METHODS Primary MPC/MPC-subset specific markers in synovium were identified by FACS analysis of uncultured cells. MPC-subsets from human synovium obtained from patients undergoing total knee arthroplasty were FACS sorted, cultured, immunophenotyped and chondrogenically differentiated. The anatomical localization of MPCs in synovium was examined using immunohistochemistry. Finally, the presence of these MPC subsets in healthy synovium obtained from human organ donors was examined. RESULTS A combination of CD45, CD31, CD73 and CD90 can isolate two distinct MPC-subsets in synovium. These MPC-subsets, freshly isolated from synovium, did not express CD45 or CD31, but expressed CD73. Additionally, a sub-population of CD73+ cells also expressed CD90. CD45-CD31-CD73+CD90- cells were significantly more chondrogenic than CD45-CD31-CD73+CD90+ cells in the presence of TGFβ1. Interestingly, reduced chondrogenic ability of CD73+CD90+ cells could be reversed by the addition of BMP2, showing discrete chondrogenic factor requirements by distinct cell-subsets. In addition, these MPCs had distinct anatomical localization; CD73 was expressed both in intimal and sub-intimal region while CD90 was enriched in the sub-intimal region. We further demonstrated that these subsets are also present in healthy synovium. CONCLUSIONS We provide indications that primary MPCs in synovial intima and sub-intima are phenotypically and functionally distinct with different chondrogenic properties.
Collapse
Affiliation(s)
- K Sivasubramaniyan
- Department of Orthopaedics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - W J L M Koevoet
- Department of Otorhinolaryngology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - A A Hakimiyan
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA
| | - M Sande
- Department of Orthopaedics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - E Farrell
- Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - M J Hoogduijn
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - J A N Verhaar
- Department of Orthopaedics, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - S Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA
| | - H-J Bühring
- Department of Internal Medicine II, Division of Hematology, University Clinic of Tübingen, Tübingen, Germany
| | - G J V M van Osch
- Department of Orthopaedics, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Otorhinolaryngology, Erasmus MC University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
27
|
Chen L, Shi Y, Zhang X, Hu X, Shao Z, Dai L, Ju X, Ao Y, Wang J. CaAlg hydrogel containing bone morphogenetic protein 4-enhanced adipose-derived stem cells combined with osteochondral mosaicplasty facilitated the repair of large osteochondral defects. Knee Surg Sports Traumatol Arthrosc 2019; 27:3668-3678. [PMID: 30923857 DOI: 10.1007/s00167-019-05418-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 02/15/2019] [Indexed: 11/30/2022]
Abstract
PURPOSE Cartilage repair presents a challenge to clinicians and researchers. A more effective procedure that can produce hyaline-like cartilage is needed for articular cartilage repair. Mosaic osteochondral grafts for large osteochondral defects often show poor integration between the grafts and the surrounding normal cartilage, leading to defective cracks filled with fibrous tissue instead of hyaline-like cartilage. In the present study, we aimed to repair the defective cracks with a calcium alginate (CaAlg) hydrogel containing bone morphogenetic protein 4 (BMP4)-enhanced adipose-derived stem cells (ADSCs). METHODS ADSCs were transduced with BMP4 (B-ADSCs). The expression of BMP4 and type II collagen was confirmed using an enzyme-linked immunosorbent assay (ELISA). Swine models of large cartilage defects of the knee were constructed and received one of the four treatments: mosaicplasty only, mosaicplasty with the CaAlg hydrogel, mosaicplasty with the CaAlg hydrogel containing ADSCs, or mosaicplasty with the CaAlg hydrogel containing B-ADSCs injected into the defective cracks. Outcomes were evaluated at 12 and 24 weeks after surgery. RESULTS The in vitro study showed that the osteogenic and chondrogenic activities of the B-ADSCs were enhanced compared with those of the control. In vivo, in the group that received mosaicplasty-containing B-ADSCs, osteochondral tissue was completely integrated with an intact surface. Additionally, the histological scores of the mosaicplasty-containing B-ADSCs group were significantly higher than those of the other groups. Biomechanical examination confirmed that the neocartilage possessed properties similar to those of normal cartilage. CONCLUSIONS Mosaicplasty and hydrogel containing B-ADSCs promoted the repair of large cartilage defects by regenerating hyaline cartilage and repairing dead spaces between osteochondral grafts and donor-site defects, thus improving the feasibility and success rate of one-stage complete repair surgery for large osteochondral defects. This proposed method provides a novel and effective means for the repair of large articular osteochondral defects.
Collapse
Affiliation(s)
- Linxin Chen
- Institute of Sports Medicine, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, People's Republic of China
| | - Yuanyuan Shi
- Institute of Sports Medicine, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, People's Republic of China
| | - Xin Zhang
- Institute of Sports Medicine, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, People's Republic of China
| | - Xiaoqing Hu
- Institute of Sports Medicine, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, People's Republic of China
| | - Zhenxing Shao
- Institute of Sports Medicine, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, People's Republic of China
| | - Linghui Dai
- Institute of Sports Medicine, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, People's Republic of China
| | - Xiaodong Ju
- Institute of Sports Medicine, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, People's Republic of China
| | - Yingfang Ao
- Institute of Sports Medicine, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, People's Republic of China.
| | - Jianquan Wang
- Institute of Sports Medicine, Peking University Third Hospital, 49 North Garden Rd, Haidian District, Beijing, 100191, People's Republic of China.
| |
Collapse
|
28
|
Ye F, Xu H, Yin H, Zhao X, Li D, Zhu Q, Wang Y. The role of BMP6 in the proliferation and differentiation of chicken cartilage cells. PLoS One 2019; 14:e0204384. [PMID: 31260450 PMCID: PMC6602178 DOI: 10.1371/journal.pone.0204384] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 05/28/2019] [Indexed: 11/18/2022] Open
Abstract
Previous studies have indicated that bone morphogenetic protein (BMP) 6 may play an important role in skeletal system development and progression. However, the mechanism underlying the effects of BMP6 in cartilage cell proliferation and differentiation remains unknown. In this study, cartilage cells were isolated from shanks of chicken embryos and treated with different concentrations of Growth Hormone. Cell proliferation potential was assessed using real-time polymerase chain reaction (RT-PCR), western blotting and CCK-8 assays in vitro. The results showed that at 48 h, the Collagen II and BMP6 expression levels in 50 ng/μl GH-treated cartilage cells were significantly higher than in groups treated with 100 ng/μl or 200 ng/μl GH. We further observed that knockdown of BMP6 in cartilage cells led to significantly decreased expression mRNAs and proteins of Collagen II and Collagen X. Moreover, the suppression of BMP6 expression by a specific siRNA led to significantly decreased expression mRNA levels of IGF1R, JAK2, PKC, PTH, IHH and PTHrP and decreased protein levels of PKC, IHH and PTHrP. Taken together, our data suggest that BMP6 may play a critical role in chicken cartilage cell proliferation and differentiation through the regulation of IGF1, JAK2, PKC, PTH, and IHH-PTHrP signaling pathways.
Collapse
Affiliation(s)
- Fei Ye
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Hengyong Xu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Huadong Yin
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Xiaoling Zhao
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Diyan Li
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Qing Zhu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| | - Yan Wang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
29
|
Kuroda Y, Matsumoto T, Hayashi S, Hashimoto S, Takayama K, Kirizuki S, Tsubosaka M, Kamenaga T, Takashima Y, Matsushita T, Niikura T, Kuroda R. Intra-articular autologous uncultured adipose-derived stromal cell transplantation inhibited the progression of cartilage degeneration. J Orthop Res 2019; 37:1376-1386. [PMID: 30378173 DOI: 10.1002/jor.24174] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/22/2018] [Indexed: 02/04/2023]
Abstract
The role of uncultured adipose-derived stromal cells for osteoarthritis treatment remains unclear despite sporadic reports supporting their use in clinical settings. This study aimed to evaluate the therapeutic effects of autologous uncultured adipose-derived stromal cell transplantation in a rabbit osteoarthritis model. Uncultured adipose-derived stromal cells isolated from rabbits were administered via intra-articular injection into the knees after osteoarthritis onset. Animals were sacrificed at 8 and 12 weeks after osteoarthritis onset to compare the macroscopic, histological, and immunohistochemical characteristics between the uncultured adipose-derived stromal cell and control groups. Co-culture assay was also performed. The chondrocytes isolated from the model were co-cultured with adipose-derived stromal cells. The cell viability of chondrocytes and expression of chondrocyte-specific genes in the co-culture (uncultured adipose-derived stromal cell) group were compared with the mono-culture (control; chondrocytes only) group. In macroscopic and histological analyses, the uncultured adipose-derived stromal cell group showed less damage to the cartilage surface than the control group at 8 and 12 weeks after osteoarthritis onset. In immunohistochemical and co-culture assay, the uncultured adipose-derived stromal cell group showed higher expression of collagen type II and SRY box-9 and lower expression of matrix metalloproteinase-13 than the control group. The cell viability of chondrocytes in the uncultured adipose-derived stromal cell group was higher than that in the control group. Intra-articular autologous uncultured adipose-derived stromal cell transplantation inhibited the progression of cartilage degeneration in a rabbit osteoarthritis model by regulating chondrocyte viability and secreting chondrocyte-protecting cytokines or growth factors, which promote anabolic factors and inhibit catabolic factors. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1376-1386, 2019.
Collapse
Affiliation(s)
- Yuichi Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shingo Hashimoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koji Takayama
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinsuke Kirizuki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Kamenaga
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshinori Takashima
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takehiko Matsushita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takahiro Niikura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
30
|
Guilak F, Sandell LJ, Huard J. Journal of Orthopaedic Research: Special Issue on Stem Cells. J Orthop Res 2019; 37:1209-1211. [PMID: 31050013 DOI: 10.1002/jor.24338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri.,Shriners Hospitals for Children-St. Louis, St. Louis, Missouri.,Center of Regenerative Medicine, Washington University, St. Louis, Missouri
| | - Linda J Sandell
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri.,Center of Regenerative Medicine, Washington University, St. Louis, Missouri
| | - Johnny Huard
- Steadman Philippon Research Institute, Vail, Colorado
| |
Collapse
|
31
|
Guilak F, Pferdehirt L, Ross AK, Choi YR, Collins KH, Nims RJ, Katz DB, Klimak M, Tabbaa S, Pham CT. Designer Stem Cells: Genome Engineering and the Next Generation of Cell-Based Therapies. J Orthop Res 2019; 37:1287-1293. [PMID: 30977548 PMCID: PMC6546536 DOI: 10.1002/jor.24304] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 02/04/2023]
Abstract
Stem cells provide tremendous promise for the development of new therapeutic approaches for musculoskeletal conditions. In addition to their multipotency, certain types of stem cells exhibit immunomodulatory effects that can mitigate inflammation and enhance tissue repair. However, the translation of stem cell therapies to clinical practice has proven difficult due to challenges in intradonor and interdonor variability, engraftment, variability in recipient microenvironment and patient indications, and limited therapeutic biological activity. In this regard, the success of stem cell-based therapies may benefit from cellular engineering approaches to enhance factors such as purification, homing and cell survival, trophic effects, or immunomodulatory signaling. By combining recent advances in gene editing, synthetic biology, and tissue engineering, the potential exists to create new classes of "designer" cells that have prescribed cell-surface molecules and receptors as well as synthetic gene circuits that provide for autoregulated drug delivery or enhanced tissue repair. Published by Wiley Periodicals, Inc. J Orthop Res 37:1287-1293, 2019.
Collapse
Affiliation(s)
- Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110,Correspondence: Farshid Guilak, Ph.D. Center of Regenerative Medicine, Washington University, St. Louis, Campus Box 8233, McKinley Research Bldg, Room 3121, St. Louis, MO 63110-1624.
| | - Lara Pferdehirt
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Alison K. Ross
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Yun-Rak Choi
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110
| | - Robert J. Nims
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110
| | - Dakota B. Katz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Molly Klimak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | | | - Christine T.N. Pham
- Division of Rheumatology, Department of Medicine, Washington University in St. Louis, MO, 63110
| |
Collapse
|
32
|
Long Noncoding RNA H19 Participates in the Regulation of Adipose-Derived Stem Cells Cartilage Differentiation. Stem Cells Int 2019; 2019:2139814. [PMID: 31191668 PMCID: PMC6525810 DOI: 10.1155/2019/2139814] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/28/2019] [Accepted: 02/13/2019] [Indexed: 12/22/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) are multipotent and have received increasing attention for their applications in medicine. Cell-based therapies are optimal for diseases with loss or damage to tissues or organs. ADSCs and bone marrow mesenchymal stem cells (BMSCs) can differentiate into many cell lineages. Because of their advantages in accessibility and volume, ADSCs are regarded as a desirable alternative to BMSCs. In this study, we focused on the chondrocytic differentiation potential of ADSCs and the underlying mechanism. We found that the long noncoding RNA H19 plays an important role in this process. Overexpression of H19 in ADSCs induced differentiation towards chondrocytes. H19 is abundantly expressed during embryonic development and downregulated after birth, implying its regulatory role in determining cell fate. However, in our experiments, H19 exerted its regulatory function during cartilage differentiation of ADSCs through competing miRNA regulation of STAT2.
Collapse
|
33
|
Decellularized biological scaffold and stem cells from autologous human adipose tissue for cartilage tissue engineering. Methods 2019; 171:97-107. [PMID: 31051252 DOI: 10.1016/j.ymeth.2019.04.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 04/05/2019] [Accepted: 04/27/2019] [Indexed: 02/08/2023] Open
Abstract
Here, the in vitro engineering of a cartilage-like tissue by using decellularized extracellular matrix scaffold (hECM) seeded with human adipose stem cells (hASCs) which can both be isolated from the human waste adipose tissue is described. Cell-free, highly fibrous and porous hECM was produced using a protocol containing physical (homogenization, centrifugation, molding) and chemical (crosslinking) treatments, characterized by SEM, histochemistry, immunohistochemistry and in vitro cell interaction study. A construct of hECM seeded with hASCs was cultured in chondrogenic medium (with TGF-β3 and BMP-6) for 42 days. SEM and histology showed that the biological scaffold was highly porous and had a compact structure suitable for handling and subsequent cell culture stages. Cells successfully integrated into the scaffold and had good cellular viability and continuity to proliferate. Constructs showed the formation of cartilage-like tissue with the synthesis of cartilage-specific proteins, Collagen type II and Aggrecan. Dimethylmethylene blue dye binding assay demonstrated that the GAG content of the constructs was in tendency to increase with time confirming chondrogenic differentiation of hASCs. The results support that human waste adipose tissue is an important source for decellularized hECM as well as stem cells, and adipose hECM scaffold provides a suitable environment for chondrogenic differentiation of hASCs.
Collapse
|
34
|
Human Diseased Articular Cartilage Contains a Mesenchymal Stem Cell-Like Population of Chondroprogenitors with Strong Immunomodulatory Responses. J Clin Med 2019; 8:jcm8040423. [PMID: 30925656 PMCID: PMC6517884 DOI: 10.3390/jcm8040423] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 02/07/2023] Open
Abstract
Background: osteoarthritic human articular cartilage (AC)-derived cartilage cells (CCs) with same-donor bone marrow (BMSCs) and adipose tissue (ASCs)-derived mesenchymal stem cells were compared, in terms of stemness features, and secretory and immunomodulatory responses to inflammation. Methods: proteoglycan 4 (PRG4) presence was evaluated in AC and CCs. MSCs and CCs (n = 8) were cultured (P1 to P4) and characterized for clonogenicity, nanog homeobox (NANOG), and POU class 5 homeobox 1 (POU5F1) expression, immunotypification, and tri-lineage differentiation. Their basal and interleukin-1β (IL-1β)-stimulated expression of matrix metalloproteases (MMPs), tissue inhibitors (TIMPs), release of growth factors, and cytokines were analyzed, along with the immunomodulatory ability of CCs. Results: PRG4 was mainly expressed in the intact AC surface, whereas shifted to the intermediate zone in damaged cartilage and increased its expression in CCs upon culture. All cells exhibited a similar phenotype and stemness maintenance over passages. CCs showed highest chondrogenic ability, no adipogenic potential, a superior basal secretion of growth factors and cytokines, the latter further increased after inflammatory stimulation, and an immunomodulatory behavior. All stimulated cells shared an increased MMP expression without a corresponding TIMP production. Conclusion: based on the observed features, CCs obtained from pathological joints may constitute a potential tissue-specific therapeutic target or agent to improve damaged cartilage healing, especially damage caused by inflammatory/immune mediated conditions.
Collapse
|
35
|
Hyaluronan microenvironment enhances cartilage regeneration of human adipose-derived stem cells in a chondral defect model. Int J Biol Macromol 2018; 119:726-740. [DOI: 10.1016/j.ijbiomac.2018.07.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 06/28/2018] [Accepted: 07/11/2018] [Indexed: 12/22/2022]
|
36
|
Gugjoo MB, Amarpal, Makhdoomi DM, Sharma GT. Equine Mesenchymal Stem Cells: Properties, Sources, Characterization, and Potential Therapeutic Applications. J Equine Vet Sci 2018; 72:16-27. [PMID: 30929778 DOI: 10.1016/j.jevs.2018.10.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 09/06/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023]
Abstract
Properties like sustained multiplication and self-renewal, and homing and multilineage differentiation to undertake repair of the damaged tissues make stem cells the lifeline for any living system. Therefore, stem cell therapy is regarded to carry immense therapeutic potential. Though the dearth of understanding about the basic biological properties and pathways involved in therapeutic benefits currently limit the application of stem cells in humans as well as animals, there are innumerable reports that suggest clinical benefits of stem cell therapy in equine. Among various stem cell sources, currently adult mesenchymal stem cells (MSCs) are preferred for therapeutic application in horse owing to their easy availability, capacity to modulate inflammation, and promote healing. Also the cells carry very limited teratogenic risk compared to the pluripotent stem cells. Mesenchymal stem cells were earlier considered mainly for musculoskeletal tissues, but now may also be utilized in other diverse clinical problems in horse, and the results may be extrapolated even for human medicine. The current review highlights biological properties, sources, mechanisms, and potential therapeutic applications of stem cells in equine practice.
Collapse
Affiliation(s)
- Mudasir Bashir Gugjoo
- Division of Surgery, Indian Veterinary Research Institute-Izatnagar, Bareilly, UP, India.
| | - Amarpal
- Division of Surgery, Indian Veterinary Research Institute-Izatnagar, Bareilly, UP, India
| | - Dil Mohammad Makhdoomi
- Division of Surgery, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-Kashmir, Srinagar, J&K, India
| | - Gutulla Taru Sharma
- Division of Physiology and Climatology, Indian Veterinary Research Institute-Izatnagar, Bareilly, UP, India
| |
Collapse
|
37
|
Martin-Pena A, Porter R, Plumton G, McCarrel T, Morton A, Guijarro M, Ghivizzani S, Sharma B, Palmer G. Lentiviral-based reporter constructs for profiling chondrogenic activity in primary equine cell populations. Eur Cell Mater 2018; 36:156-170. [PMID: 30311630 PMCID: PMC6788286 DOI: 10.22203/ecm.v036a12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Successful clinical translation of mesenchymal stem cell (MSC)-based therapies for cartilage repair will likely require the implementation of standardised protocols and broadly applicable tools to facilitate the comparisons among cell types and chondroinduction methods. The present study investigated the utility of recombinant lentiviral reporter vectors as reliable tools for comparing chondrogenic potential among primary cell populations and distinguishing cellular-level variations of chondrogenic activity in widely used three-dimensional (3D) culture systems. Primary equine MSCs and chondrocytes were transduced with vectors containing combinations of fluorescent and luciferase reporter genes under constitutive cytomeglavirus (CMV) or chondrocyte-lineage (Col2) promoters. Reporter activity was measured by fluorescence imaging and luciferase assay. In 3D cultures of MSC aggregates and polyethylene glycol-hyaluronic acid (PEG-HA) hydrogels, transforming growth factor beta 3 (TGF-β3)-mediated chondroinduction increased Col2 reporter activity, demonstrating close correlation with histology and mRNA expression levels of COL2A1 and SOX9. Comparison of chondrogenic activities among MSC populations using a secretable luciferase reporter revealed enhanced chondrogenesis in bone-marrow-derived MSCs relative to MSC populations from synovium and adipose tissues. A dual fluorescence reporter - enabling discrimination of highly chondrogenic (Col2-GFP) cells within an MSC population (CMV-tdTomato) - revealed marked heterogeneity in differentiating aggregate cultures and identified chondrogenic cells in chondrocyte-seeded PEG-HA hydrogels after 6 weeks in a subcutaneous implant model - indicating stable, long-term reporter expression in vivo. These results suggested that lentiviral reporter vectors may be used to address fundamental questions regarding chondrogenic activity in chondroprogenitor cell populations and accelerate clinical translation of cell-based cartilage repair strategies.
Collapse
Affiliation(s)
- A. Martin-Pena
- Department of Orthopaedics and Rehabilitation, University
of Florida, Gainesville, Florida USA
| | - R.M. Porter
- Department of Orthopaedics, University of Arkansas, Little
Rock, Arkansas, USA
| | - G Plumton
- Department of Biomedical Engineering, University of
Florida, Gainesville, Florida USA
| | - T.M. McCarrel
- College of Veterinary Sciences, University of Florida,
Gainesville, Florida USA
| | - A.J. Morton
- College of Veterinary Sciences, University of Florida,
Gainesville, Florida USA
| | - M.V. Guijarro
- Department of Anatomy and Cell Biology, University of
Florida, Gainesville, Florida USA
| | - S.C. Ghivizzani
- Department of Orthopaedics and Rehabilitation, University
of Florida, Gainesville, Florida USA
| | - B. Sharma
- Department of Orthopaedics, University of Arkansas, Little
Rock, Arkansas, USA
| | - G.D. Palmer
- Department of Orthopaedics and Rehabilitation, University
of Florida, Gainesville, Florida USA,Address for correspondence: Glyn Palmer, Ph.D,
Dept of Orthopaedics and Rehabilitation, University of Florida, 1600 SW Archer
Rd, MSB, M2-235, Gainesville, FL 32610, Telephone: +1 352 273 7087, Fax: +1 352
273 7427,
| |
Collapse
|
38
|
Neshati V, Mollazadeh S, Fazly Bazzaz BS, Iranshahi M, Mojarrad M, Naderi-Meshkin H, Kerachian MA. Cardiogenic effects of characterized Geum urbanum extracts on adipose-derived human mesenchymal stem cells. Biochem Cell Biol 2018; 96:610-618. [DOI: 10.1139/bcb-2017-0313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Stem cell therapy is considered as a promising treatment for cardiovascular diseases. Adipose-derived mesenchymal stem cells (ADMSCs) have the ability to undergo cardiomyogenesis. Medicinal plants are effective and safe candidates for cell differentiation. Therefore, the aim of our study was to investigate cardiogenic effects of characterized (HPLC–UV) extracts of Geum urbanum on ADMSCs of adipose tissue. The methanolic extracts of the root and aerial parts of G. urbanum were obtained and MTT assay was used for studying their cytotoxic effects. Then, cells were treated with 50 or 100 μg/mL of the extracts from root and aerial parts of G. urbanum. MTT assay showed that the extracts of G. urbanum did not have any toxic effects on ADMSCs. Immunostaining results showed increase in the expression of α-actinin and cardiac troponin I (cTnI), and quantitative real-time reverse-transcription PCR data confirmed the upregulation of ACTN, ACTC1, and TNNI3 genes in ADMSCs after treatment. According to HPLC fingerprinting, some cardiogenic effects of G. urbanum extracts are probably due to ellagic and gallic acid derivatives. Our findings indicated that G. urbanum extracts effectively upregulated some essential cardiogenic markers, which confirmed the therapeutic role of this plant as a traditional cardiac medicine.
Collapse
Affiliation(s)
- Vajiheh Neshati
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Mollazadeh
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Sedigheh Fazly Bazzaz
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hojjat Naderi-Meshkin
- Stem Cell and Regenerative Medicine Research Group, Academic Center for Education, Culture, Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
39
|
Deng ZH, Li YS, Gao X, Lei GH, Huard J. Bone morphogenetic proteins for articular cartilage regeneration. Osteoarthritis Cartilage 2018; 26:1153-1161. [PMID: 29580979 DOI: 10.1016/j.joca.2018.03.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 02/18/2018] [Accepted: 03/19/2018] [Indexed: 02/02/2023]
Abstract
Degeneration of articular cartilage (AC) tissue is the most common cause of osteoarthritis (OA) and rheumatoid arthritis. Bone morphogenetic proteins (BMPs) play important roles in bone and cartilage formation. This article reviews the experimental and clinical applications of BMPs in cartilage regeneration. Experimental evidence indicates that BMPs play an important role in protection against cartilage damage caused by inflammation or trauma, by binding to different receptor combinations and, consequently, activating different intracellular signaling pathways. Loss of function of BMP-related receptors contributes to the decreased intrinsic repair capacity of damaged cartilage and, thus, the multifunctional effects of BMPs make them attractive tools for the treatment of cartilage damage in patients with degenerative diseases. However, the development of BMP therapy as a treatment modality for cartilage regeneration has been hampered by certain factors, such as the eligibility of participants in clinical trials, financial support, drug delivery carrier safety, availabilities of effective scaffolds, appropriate selection of optimal dose and timing of administration, and side effects. Further research is needed to overcome these issues for future routine clinical applications. Research and development leading to the successful application of BMPs can initiate a new era in the treatment of cartilage degenerative diseases like OA.
Collapse
Affiliation(s)
- Z H Deng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China; Department of Orthopaedic Surgery, Center for Tissue Engineering and Aging Research, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Orthopedics, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, Guangdong Province, China
| | - Y S Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - X Gao
- Department of Orthopaedic Surgery, Center for Tissue Engineering and Aging Research, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA; The Steadman Philippon Research Institute, Vail, CO, USA
| | - G H Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| | - J Huard
- Department of Orthopaedic Surgery, Center for Tissue Engineering and Aging Research, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA; The Steadman Philippon Research Institute, Vail, CO, USA.
| |
Collapse
|
40
|
Yin H, Wang Y, Sun X, Cui G, Sun Z, Chen P, Xu Y, Yuan X, Meng H, Xu W, Wang A, Guo Q, Lu S, Peng J. Functional tissue-engineered microtissue derived from cartilage extracellular matrix for articular cartilage regeneration. Acta Biomater 2018; 77:127-141. [PMID: 30030172 DOI: 10.1016/j.actbio.2018.07.031] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/21/2022]
Abstract
We developed a promising cell carrier prepared from articular cartilage slices, designated cartilage extracellular matrix (ECM)-derived particles (CEDPs), through processes involving physical pulverization, size screening, and chemical decellularization. Rabbit articular chondrocytes (ACs) or adipose-derived stem cells (ASCs) rapidly attached to the surface of the CEDPs and proliferated with high cell viability under microgravity (MG) condition in a rotary cell culture system (RCCS) or static condition. Gene profiling results demonstrated that ACs expanded on CEDPs exhibited significantly enhanced chondrogenic phenotypes compared with monolayer culture, and that ASCs differentiated into a chondrogenic phenotype without the use of exogenous growth factors. Moreover, MG culture conditions in a RCCS bioreactor were superior to static culture conditions in terms of maintaining the chondrogenic phenotype of ACs and inducing ACS chondrogenesis. With prolonged expansion, functional microtissue aggregates of AC- or ASC-laden CEDPs were formed. Further, AC- or ASC-based microtissues were directly implanted in vivo to repair articular osteochondral defects in a rabbit model. Histological results, biomechanical evaluations, and radiographic assessments indicated that AC- and ASC-based microtissues displayed equal levels of superior hyaline cartilage repair, whereas the other two treatment groups, in which osteochondral defects were treated with CEDPs alone or fibrin glue, exhibited primarily fibrous tissue repair. These findings provide an alternative method for cell culture and stem cell differentiation and a promising strategy for constructing tissue-engineered cartilage microtissues for cartilage regeneration. STATEMENT OF SIGNIFICANCE Despite the remarkable progress in cartilage tissue engineering, cartilage repair still remains elusive. In the present study, we developed a cell carrier, namely cartilage extracellular matrix-derived particles (CEDPs), for cell proliferation of articular chondrocytes (ACs) and adipose-derived stem cells (ASCs), which improved the maintenance of chondrogenic phenotype of ACs, and induced chondrogenesis of ASCs. Moreover, the functional microtissue aggregates of AC- or ASC-laden CEDPs induced equal levels of superior hyaline cartilage repair in a rabbit model. Therefore, our study demonstrated an alternative method for chondrocyte culture and stem cell differentiation, and a promising strategy for constructing tissue-engineered cartilage microtissues for in vivo articular cartilage repair and regeneration.
Collapse
Affiliation(s)
- Heyong Yin
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China; Department of Surgery, Ludwig-Maximilians-University (LMU), Nussbaumstr. 20, D-80336 Munich, Germany
| | - Yu Wang
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China
| | - Xun Sun
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China; Department of Orthopaedics, Tianjin Hospital, No. 406 Jiefang Nan Road, Tianjin 300211, PR China
| | - Ganghua Cui
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China
| | - Zhen Sun
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China
| | - Peng Chen
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China
| | - Yichi Xu
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China
| | - Xueling Yuan
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China
| | - Haoye Meng
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China
| | - Wenjing Xu
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China
| | - Aiyuan Wang
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China
| | - Quanyi Guo
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China
| | - Shibi Lu
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China
| | - Jiang Peng
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Beijing 100853, PR China.
| |
Collapse
|
41
|
Park YB, Ha CW, Rhim JH, Lee HJ. Stem Cell Therapy for Articular Cartilage Repair: Review of the Entity of Cell Populations Used and the Result of the Clinical Application of Each Entity. Am J Sports Med 2018; 46:2540-2552. [PMID: 29023156 DOI: 10.1177/0363546517729152] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Following successful preclinical studies, stem cell therapy is emerging as a candidate for the treatment of articular cartilage lesions. Because stem cell therapy for cartilage repair in humans is at an early phase, confusion and errors are found in the literature regarding use of the term stem cell therapy in this field. PURPOSE To provide an overview of the outcomes of cartilage repair, elucidating the various cell populations used, and thus reduce confusion with regard to using the term stem cell therapy. STUDY DESIGN Systematic review. METHODS The authors systematically reviewed any studies on clinical application of mesenchymal stem cells (MSCs) in human subjects. A comprehensive search was performed in MEDLINE, EMBASE, the Cochrane Library, CINAHL, Web of Science, and Scopus for human studies that evaluated articular cartilage repair with cell populations containing MSCs. These studies were classified as using bone marrow-derived MSCs, adipose tissue-derived MSCs, peripheral blood-derived MSCs, synovium-derived MSCs, and umbilical cord blood-derived MSCs according to the entity of cell population used. RESULTS Forty-six clinical studies were identified to focus on cartilage repair with MSCs: 20 studies with bone marrow-derived MSCs, 21 studies with adipose tissue-derived MSCs, 3 studies with peripheral blood-derived MSCs, 1 study with synovium-derived MSCs, and 1 study with umbilical cord blood-derived MSCs. All clinical studies reported that cartilage treated with MSCs showed favorable clinical outcomes in terms of clinical scores or cartilage repair evaluated by MRI. However, most studies were limited to case reports and case series. Among these 46 clinical studies, 18 studies erroneously referred to adipose tissue-derived stromal vascular fractions as "adipose-derived MSCs," 2 studies referred to peripheral blood-derived progenitor cells as "peripheral blood-derived MSCs," and 1 study referred to bone marrow aspirate concentrate as "bone marrow-derived MSCs." CONCLUSION Limited evidence is available regarding clinical benefit of stem cell therapy for articular cartilage repair. Because the literature contains substantial errors in describing the therapeutic cells used, researchers need to be alert and observant of proper terms, especially regarding whether the cells used were stem cells or cell populations containing a small portion of stem cells, to prevent confusion in understanding the results of a given stem cell-based therapy.
Collapse
Affiliation(s)
- Yong-Beom Park
- Department of Orthopedic Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Dongjak-gu, Seoul, Republic of Korea
| | - Chul-Won Ha
- Department of Orthopedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Republic of Korea.,Stem Cell & Regenerative Medicine Research Institute, Samsung Medical Center, Gangnam-gu, Seoul, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Gangnam-gu, Seoul, Republic of Korea
| | - Ji Heon Rhim
- Department of Orthopedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Republic of Korea
| | - Han-Jun Lee
- Department of Orthopedic Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Dongjak-gu, Seoul, Republic of Korea
| |
Collapse
|
42
|
Dubey NK, Mishra VK, Dubey R, Deng YH, Tsai FC, Deng WP. Revisiting the Advances in Isolation, Characterization and Secretome of Adipose-Derived Stromal/Stem Cells. Int J Mol Sci 2018; 19:ijms19082200. [PMID: 30060511 PMCID: PMC6121360 DOI: 10.3390/ijms19082200] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/08/2018] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stromal/stem cells (ASCs) seems to be a promising regenerative therapeutic agent due to the minimally invasive approach of their harvest and multi-lineage differentiation potential. The harvested adipose tissues are further digested to extract stromal vascular fraction (SVF), which is cultured, and the anchorage-dependent cells are isolated in order to characterize their stemness, surface markers, and multi-differentiation potential. The differentiation potential of ASCs is directed through manipulating culture medium composition with an introduction of growth factors to obtain the desired cell type. ASCs have been widely studied for its regenerative therapeutic solution to neurologic, skin, wound, muscle, bone, and other disorders. These therapeutic outcomes of ASCs are achieved possibly via autocrine and paracrine effects of their secretome comprising of cytokines, extracellular proteins and RNAs. Therefore, secretome-derivatives might offer huge advantages over cells through their synthesis and storage for long-term use. When considering the therapeutic significance and future prospects of ASCs, this review summarizes the recent developments made in harvesting, isolation, and characterization. Furthermore, this article also provides a deeper insight into secretome of ASCs mediating regenerative efficacy.
Collapse
Affiliation(s)
- Navneet Kumar Dubey
- Ceramics and Biomaterials Research Group, Advanced Institute of Materials Science, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
| | - Viraj Krishna Mishra
- Applied Biotech Engineering Centre (ABEC), Department of Biotechnology, Ambala College of Engineering and Applied Research, Ambala 133101, India.
| | - Rajni Dubey
- Graduate Institute Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan.
| | - Yue-Hua Deng
- Stem Cell Research Center, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Life Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan.
| | - Feng-Chou Tsai
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Win-Ping Deng
- Stem Cell Research Center, Taipei Medical University, Taipei 11031, Taiwan.
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Basic medicine, Fu-Jen Catholic University, New Taipei City 24205, Taiwan.
| |
Collapse
|
43
|
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
|
44
|
Choi B, Kim D, Han I, Lee SH. Microenvironmental Regulation of Stem Cell Behavior Through Biochemical and Biophysical Stimulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:147-160. [PMID: 30471031 DOI: 10.1007/978-981-13-0445-3_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Stem cells proliferate by undergoing self-renewal and differentiate into multiple cell lineages in response to biochemical and biophysical stimuli. Various biochemical cues such as growth factors, nucleic acids, chemical reagents, and small molecules have been used to induce stem cell differentiation or reprogramming or to maintain their pluripotency. Moreover, biophysical cues such as matrix stiffness, substrate topography, and external stress and strain play a major role in modulating stem cell behavior. In this chapter, we have summarized microenvironmental regulation of stem cell behavior through biochemical and biophysical stimulation.
Collapse
Affiliation(s)
- Bogyu Choi
- Department of Biomedical Science, CHA University, Seongnam-si, South Korea
| | - Deogil Kim
- Department of Biomedical Science, CHA University, Seongnam-si, South Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University, CHA Bundang Medical Center, Seongnam-si, South Korea
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University, Goyang-si, Gyeonggi-do, South Korea.
| |
Collapse
|
45
|
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
|
46
|
Abstract
Adipose-derived stem/stromal cells (ASCs), together with adipocytes, vascular endothelial cells, and vascular smooth muscle cells, are contained in fat tissue. ASCs, like the human bone marrow stromal/stem cells (BMSCs), can differentiate into several lineages (adipose cells, fibroblast, chondrocytes, osteoblasts, neuronal cells, endothelial cells, myocytes, and cardiomyocytes). They have also been shown to be immunoprivileged, and genetically stable in long-term cultures. Nevertheless, unlike the BMSCs, ASCs can be easily harvested in large amounts with minimal invasive procedures. The combination of these properties suggests that these cells may be a useful tool in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Simone Ciuffi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Roberto Zonefrati
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Maria Luisa Brandi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| |
Collapse
|
47
|
Intra-articular Injection of Mesenchymal Stem Cells and Platelet-Rich Plasma to Treat Patellofemoral Osteoarthritis: Preliminary Results of a Long-Term Pilot Study. J Vasc Interv Radiol 2017; 28:1708-1713. [PMID: 29031987 DOI: 10.1016/j.jvir.2017.08.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 12/27/2022] Open
Abstract
PURPOSE To assess the feasibility and safety of concomitant intra-articular (IA) knee injection of mesenchymal stem cells (MSCs) and platelet-rich plasma (PRP) under fluoroscopic guidance to treat patellofemoral osteoarthritis (OA). MATERIALS AND METHODS This prospective study included 19 consecutive patients referred for fluoroscopically guided IA MSC and PRP injection for symptomatic patellofemoral chondropathy in which conservative treatment had failed. Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score and magnetic resonance (MR) data, including T2 mapping sequence, were prospectively collected before and 6 months after treatment. Clinical data without MR imaging were collected until 12 months after the procedure. RESULTS WOMAC scores were significantly lower after IA injection of MSCs and PRP at 6 months and during 12-months follow-up compared with baseline (mean score decreased from 34.3 to 14.2; P < .0018). Patients reported no complications. Concerning MR imaging follow-up, there were no significant differences in grade, surface, or T2 value of the chondral lesions (P > .375). CONCLUSIONS IA injection of MSCs and PRP in early patellofemoral OA appears to allow functional improvement.
Collapse
|
48
|
Narayanan G, Bhattacharjee M, Nair LS, Laurencin CT. Musculoskeletal Tissue Regeneration: the Role of the Stem Cells. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2017. [DOI: 10.1007/s40883-017-0036-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
49
|
Farhang N, Brunger JM, Stover JD, Thakore PI, Lawrence B, Guilak F, Gersbach CA, Setton LA, Bowles RD. * CRISPR-Based Epigenome Editing of Cytokine Receptors for the Promotion of Cell Survival and Tissue Deposition in Inflammatory Environments. Tissue Eng Part A 2017; 23:738-749. [PMID: 28095751 PMCID: PMC5568019 DOI: 10.1089/ten.tea.2016.0441] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/11/2017] [Indexed: 01/08/2023] Open
Abstract
Musculoskeletal diseases have been associated with inflammatory cytokine action, particularly action by TNF-α and IL-1β. These inflammatory cytokines promote apoptosis and senescence of cells in diseased tissue and extracellular matrix breakdown. Stem cell-based therapies are being considered for the treatment of musculoskeletal diseases, but the presence of these inflammatory cytokines will have similar deleterious action on therapeutic cells delivered to these environments. Methods that prevent inflammatory-induced apoptosis and proinflammatory signaling, in cell and pathway-specific manners are needed. In this study we demonstrate the use of clustered regularly interspaced short palindromic repeats (CRISPR)-based epigenome editing to alter cell response to inflammatory environments by repressing inflammatory cytokine cell receptors, specifically TNFR1 and IL1R1. We targeted CRISPR/Cas9-based repressors to TNFR1 and IL1R1 gene regulatory elements in human adipose-derived stem cells (hADSCs) and investigated the functional outcomes of repression of these genes. Efficient signaling regulation was demonstrated in engineered hADSCs, as activity of the downstream transcription factor NF-κB was significantly reduced or maintained at baseline levels in the presence of TNF-α or IL-1β. Pellet culture of undifferentiated hADSCs demonstrated improved survival in engineered hADSCs treated with TNF-α or IL-1β, while having little effect on their immunomodulatory properties. Furthermore, engineered hADSCs demonstrated improved chondrogenic differentiation capacity in the presence of TNF-α or IL-1β, as shown by superior production of glycosaminglycans in this inflammatory environment. Overall this work demonstrates a novel method for modulating cell response to inflammatory signaling that has applications in engineering cells delivered to inflammatory environments, and as a direct gene therapy to protect endogenous cells exposed to chronic inflammation, as observed in a broad spectrum of degenerative musculoskeletal pathology.
Collapse
Affiliation(s)
- Niloofar Farhang
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Jonathan M. Brunger
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Joshua D. Stover
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | | | - Brandon Lawrence
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah
| | - Farshid Guilak
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri
- Department of Orthopaedic Surgery, Washington University in St. Louis and Shriners Hospitals for Children–St. Louis, Saint Louis, Missouri
| | - Charles A. Gersbach
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Lori A. Setton
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri
- Department of Orthopaedic Surgery, Washington University in St. Louis and Shriners Hospitals for Children–St. Louis, Saint Louis, Missouri
| | - Robby D. Bowles
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah
| |
Collapse
|
50
|
Scioli MG, Bielli A, Gentile P, Cervelli V, Orlandi A. Combined treatment with platelet-rich plasma and insulin favours chondrogenic and osteogenic differentiation of human adipose-derived stem cells in three-dimensional collagen scaffolds. J Tissue Eng Regen Med 2017; 11:2398-2410. [PMID: 27074878 DOI: 10.1002/term.2139] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 11/18/2015] [Accepted: 12/10/2015] [Indexed: 02/06/2023]
Abstract
Osteochondral lesions due to injury or other pathology commonly result in the development of osteoarthritis and progressive joint destruction. Bioengineered scaffolds are widely studied for regenerative surgery strategies in osteochondral defect management, also combining the use of stem cells, growth factors and hormones. The utility in tissue engineering of human adipose-derived stem cells (ASCs) isolated from adipose tissue has been widely noted. Autologous platelet-rich plasma (PRP) represents an alternative strategy in regenerative medicine for the local release of endogenous growth factors and hormones. Here we compared the effects of three-dimensional (3D) collagen type I scaffold culture and combined treatment with PRP and human recombinant insulin on the chondro-/osteogenic differentiation of ASCs. Histochemical and biomolecular analyses demonstrated that chondro-/osteogenic differentiation was increased in ASC-populated 3D collagen scaffolds compared with two-dimensional (2D) plastic dish culture. Chondro-/osteogenic differentiation was further enhanced in the presence of combined PRP (5% v/v) and insulin (100 nm) treatment. In addition, chondro-/osteogenic differentiation associated with the contraction of ASC-populated 3D collagen scaffold and increased β1/β3-integrin expression. Inhibition studies demonstrated that PRP/insulin-induced chondro-/osteogenic differentiation is independent of insulin-like growth factor 1 receptor (IGF-1R) and mammalian target of rapamycin (mTOR) signalling; IGF-R1/mTOR inhibition even enhanced ASC chondro-/osteogenic differentiation. Our findings underline that 3D collagen scaffold culture in association with platelet-derived growth factors and insulin favour the chondro-/osteogenic differentiation of ASCs, suggesting new translational applications in regenerative medicine for the management of osteochondral defects. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Maria Giovanna Scioli
- Institute of Anatomical Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Alessandra Bielli
- Institute of Anatomical Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Pietro Gentile
- Plastic and Reconstructive Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Augusto Orlandi
- Institute of Anatomical Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| |
Collapse
|