1
|
Ferrao Blanco MN, Lesage R, Kops N, Fahy N, Bekedam FT, Chavli A, Bastiaansen-Jenniskens YM, Geris L, Chambers MG, Pitsillides AA, Narcisi R, van Osch GJ. A multi-model approach identifies ALW-II-41-27 as a promising therapy for osteoarthritis-associated inflammation and endochondral ossification. Heliyon 2024; 10:e40871. [PMID: 39717596 PMCID: PMC11664402 DOI: 10.1016/j.heliyon.2024.e40871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024] Open
Abstract
Low-grade inflammation and pathological endochondral ossification are key processes underlying the progression of osteoarthritis, the most prevalent joint disease worldwide. In this study, we employed a multi-faceted approach, integrating publicly available datasets, in silico analyses, in vitro experiments and in vivo models to identify new therapeutic candidates targeting these processes. Data mining of transcriptomic datasets identified EPHA2, a receptor tyrosine kinase associated with cancer, as being linked to both inflammation and endochondral ossification in osteoarthritis. A computational model of cellular signaling networks in chondrocytes predicted that in silico activation of EPHA2 in healthy chondrocytes increases inflammatory mediators and induces hypertrophic differentiation, a hallmark of endochondral ossification. We then evaluated the effect of EPHA2 inhibition using the tyrosine kinase inhibitor ALW-II-41-27 in cultured human chondrocytes from individuals with osteoarthritis, demonstrating significant reductions in both inflammation and hypertrophy. Additionally, systemic subcutaneous administration of ALW-II-41-27 in a mouse osteoarthritic model attenuated joint degeneration by reducing local inflammation and pathological endochondral ossification. Collectively, this study demonstrates a novel drug discovery pipeline that integrates computational, experimental, and animal models, paving the way for the development of disease-modifying treatments for osteoarthritis.
Collapse
Affiliation(s)
- Mauricio N. Ferrao Blanco
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Raphaelle Lesage
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Belgium
- Biomechanics Section, KU Leuven, Belgium
| | - Nicole Kops
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Niamh Fahy
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Applied Science, Technological University of the Shannon: Midlands Midwest, Limerick, Ireland
| | - Fjodor T. Bekedam
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Athina Chavli
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Belgium
- Biomechanics Section, KU Leuven, Belgium
- GIGA In Silico Medicine, University of Liège, Belgium
| | - Mark G. Chambers
- Lilly Research Laboratories, Eli Lilly Pharmaceuticals, Indianapolis, USA
| | | | - Roberto Narcisi
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, University of Technology Delft, Delft, the Netherlands
| |
Collapse
|
2
|
Chen M, Jiang Z, Zou X, You X, Cai Z, Huang J. Advancements in tissue engineering for articular cartilage regeneration. Heliyon 2024; 10:e25400. [PMID: 38352769 PMCID: PMC10862692 DOI: 10.1016/j.heliyon.2024.e25400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2024] Open
Abstract
Articular cartilage injury is a prevalent clinical condition resulting from trauma, tumors, infection, osteoarthritis, and other factors. The intrinsic lack of blood vessels, nerves, and lymphatic vessels within cartilage tissue severely limits its self-regenerative capacity after injury. Current treatment options, such as conservative drug therapy and joint replacement, have inherent limitations. Achieving perfect regeneration and repair of articular cartilage remains an ongoing challenge in the field of regenerative medicine. Tissue engineering has emerged as a key focus in articular cartilage injury research, aiming to utilize cultured and expanded tissue cells combined with suitable scaffold materials to create viable, functional tissues. This review article encompasses the latest advancements in seed cells, scaffolds, and cytokines. Additionally, the role of stimulatory factors including cytokines and growth factors, genetic engineering techniques, biophysical stimulation, and bioreactor systems, as well as the role of scaffolding materials including natural scaffolds, synthetic scaffolds, and nanostructured scaffolds in the regeneration of cartilage tissues are discussed. Finally, we also outline the signaling pathways involved in cartilage regeneration. Our review provides valuable insights for scholars to address the complex problem of cartilage regeneration and repair.
Collapse
Affiliation(s)
- Maohua Chen
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhiyuan Jiang
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiuyuan Zou
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiaobo You
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhen Cai
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jinming Huang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Stage-Dependent Activity and Pro-Chondrogenic Function of PI3K/AKT during Cartilage Neogenesis from Mesenchymal Stromal Cells. Cells 2022; 11:cells11192965. [PMID: 36230927 PMCID: PMC9563299 DOI: 10.3390/cells11192965] [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: 08/30/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Differentiating mesenchymal stromal cells (MSCs) into articular chondrocytes (ACs) for application in clinical cartilage regeneration requires a profound understanding of signaling pathways regulating stem cell chondrogenesis and hypertrophic degeneration. Classifying endochondral signals into drivers of chondrogenic speed versus hypertrophy, we here focused on insulin/insulin-like growth factor 1 (IGF1)-induced phosphoinositide 3-kinase (PI3K)/AKT signaling. Aware of its proliferative function during early but not late MSC chondrogenesis, we aimed to unravel the late pro-chondrogenic versus pro-hypertrophic PI3K/AKT role. PI3K/AKT activity in human MSC and AC chondrogenic 3D cultures was assessed via Western blot detection of phosphorylated AKT. The effects of PI3K inhibition with LY294002 on chondrogenesis and hypertrophy were assessed via histology, qPCR, the quantification of proteoglycans, and alkaline phosphatase activity. Being repressed by ACs, PI3K/AKT activity transiently rose in differentiating MSCs independent of TGFβ or endogenous BMP/WNT activity and climaxed around day 21. PI3K/AKT inhibition from day 21 on equally reduced chondrocyte and hypertrophy markers. Proving important for TGFβ-induced SMAD2 phosphorylation and SOX9 accumulation, PI3K/AKT activity was here identified as a required stage-dependent driver of chondrogenic speed but not of hypertrophy. Thus, future attempts to improve MSC chondrogenesis will depend on the adequate stimulation and upregulation of PI3K/AKT activity to generate high-quality cartilage from human MSCs.
Collapse
|
4
|
Tangyuenyong S, Kongdang P, Sirikaew N, Ongchai S. First study on the effect of transforming growth factor beta 1 and insulin-like growth factor 1 on the chondrogenesis of elephant articular chondrocytes in a scaffold-based 3D culture model. Vet World 2022; 15:1869-1879. [PMID: 36185520 PMCID: PMC9394124 DOI: 10.14202/vetworld.2022.1869-1879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: Osteoarthritis (OA) is recognized as a degenerative joint disease that leads to chronic pain and low quality of life in animals. Captive elephants, the largest land mammals with a long lifespan, are more prone to develop OA due to restricted spaces and insufficient physical activity. This study aimed to investigate the effect of transforming growth factor-β1 (TGF-β1) and insulin-like growth factor 1 (IGF-1) on elephant chondrogenesis in a scaffold culture of articular chondrocytes.
Materials and Methods: Elephant chondrocytes-seeded gelatin scaffolds were cultured in chondrogenic media with or without 10 ng/mL of TGF-β1 or IGF-1 alone or 5–10 ng/mL of their combination for up to 21 days. The mRNA expression of cartilage-specific anabolic genes, ACAN and COL2A1, was analyzed using a real-time reverse transcription-polymerase chain reaction. The amounts of sulfated glycosaminoglycans (sGAGs) in conditioned media and contents in cultured scaffolds were determined through dimethylmethylene blue assay. Cell morphology, accumulation of proteoglycans, and details of the cultured scaffolds were determined using hematoxylin-eosin staining, safranin O staining, and scanning electron microscopy (SEM), respectively.
Results: TGF-β1 alone significantly upregulated ACAN gene expression but not COL2A1, while IGF-1 alone did not enhance both ACAN and COL2A1 genes. The combination significantly upregulated both mRNA expression levels of ACAN and COL2A1 gene at day 14. The sGAGs accumulation and contents in the treatment groups, except IGF-1 tended to be higher than the controls, concomitantly with the production of the extracellular matrix, showed the formation of a cartilage-like tissue through histological and SEM analyses.
Conclusion: Together, our results suggest that the single treatment of TGF-β1 has a selective effect on ACAN gene, while the combined growth factors seem to be an advantage on elephant chondrogenesis. This three-dimensional culture model is probably helpful for developing cartilage regeneration in vitro and is further applied in tissue engineering for OA treatment in vivo.
Collapse
Affiliation(s)
- Siriwan Tangyuenyong
- Equine Clinic, Department of Companion Animal and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Patiwat Kongdang
- Center of Multidisciplinary Technology for Advanced Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nutnicha Sirikaew
- Musculoskeletal Science and Translational Research Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriwan Ongchai
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
5
|
Mao S, Wang S, Niu Y, Wu J, Jia P, Zheng J, Dong Y. Induction of Cartilage Regeneration by Nanoparticles Loaded with Dentin Matrix Extracted Proteins. Tissue Eng Part A 2022; 28:807-817. [PMID: 35473319 DOI: 10.1089/ten.tea.2022.0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Due to the limited self-repair capacity of articular cartilage, tissue engineering has good application prospects for cartilage regeneration. Dentin contains several key growth factors involved in cartilage regeneration. However, it remains unknown whether dentin matrix extracted proteins (DMEP) can be utilized as a complex growth-factor mixture to induce cartilage regeneration. In this work, we extracted DMEP from human dentin and improved the content and activity of chondrogenic-related growth factors in DMEP by alkaline conditioning. Afterwards, mesoporous silica nanoparticles (MSNs) with particular physical and chemical properties were composed to selectively load and sustain the release of proteins in DMEP. MSN-DMEP promoted chondrogenic differentiation of rat bone marrow-derived mesenchymal stem cells with fewer growth factors than exogenously added transforming growth factor-β1 (TGF-β1). Therefore, MSN-DMEP may serve as a promising candidate for cartilage regeneration as an alternative to expensive synthetic growth factors.
Collapse
Affiliation(s)
- Sicong Mao
- Peking University School and Hospital of Stomatology, Department of Cariology and Endodontology, Beijing, China;
| | - Sainan Wang
- Peking University School and Hospital of Stomatology, Department of Cariology and Endodontology, 22 Zhongguancun Nandajie, Haidian District, Beijing, China, 100081;
| | - Yuting Niu
- Peking University School and Hospital of Stomatology, Department of Prosthodontics, Beijing, China;
| | - Jilin Wu
- Peking University School and Hospital of Stomatology, Department of Cariology and Endodontology, Beijing, China;
| | - Peipei Jia
- Peking University School and Hospital of Stomatology, Department of Cariology and Endodontology, Beijing, China;
| | - Jinxuan Zheng
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, GUangdong, China;
| | - Yanmei Dong
- Peking University School and Hospital of Stomatology, Department of Cariology and Endodontology, Beijing, China;
| |
Collapse
|
6
|
Liu S, Deng Z, Chen K, Jian S, Zhou F, Yang Y, Fu Z, Xie H, Xiong J, Zhu W. Cartilage tissue engineering: From proinflammatory and anti‑inflammatory cytokines to osteoarthritis treatments (Review). Mol Med Rep 2022; 25:99. [PMID: 35088882 PMCID: PMC8809050 DOI: 10.3892/mmr.2022.12615] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA), one of the most common joint diseases, is characterized by fibrosis, rhagadia, ulcers and attrition of articular cartilage due to a number of factors. The etiology of OA remains unclear, but its occurrence has been associated with age, obesity, inflammation, trauma and genetic factors. Inflammatory cytokines are crucial for the occurrence and progression of OA. The intra-articular proinflammatory and anti-inflammatory cytokines jointly maintain a dynamic balance, in accordance with the physiological metabolism of articular cartilage. However, dynamic imbalance between proinflammatory and anti-inflammatory cytokines can cause abnormal metabolism in knee articular cartilage, which leads to deformation, loss and abnormal regeneration, and ultimately destroys the normal structure of the knee joint. The ability of articular cartilage to self-repair once damaged is limited, due to its inability to obtain nutrients from blood vessels, nerves and lymphatic vessels, as well as limitations in the extracellular matrix. There are several disadvantages inherent to conventional repair methods, while cartilage tissue engineering (CTE), which combines proinflammatory and anti-inflammatory cytokines, offers a new therapeutic approach for OA. The aim of the present review was to examine the proinflammatory factors implicated in OA, including IL-1β, TNF-α, IL-6, IL-15, IL-17 and IL-18, as well as the key anti-inflammatory factors reducing OA-related articular damage, including IL-4, insulin-like growth factor and TGF-β. The predominance of proinflammatory over anti-inflammatory cytokine effects ultimately leads to the development of OA. CTE, which employs mesenchymal stem cells and scaffolding technology, may prevent OA by maintaining the homeostasis of pro- and anti-inflammatory factors.
Collapse
Affiliation(s)
- Shuyu Liu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Kang Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Shengsheng Jian
- Department of Orthopedics, Luo Hu Hospital, Shenzhen, Guangdong 518001, P.R. China
| | - Feifei Zhou
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Yuan Yang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Zicai Fu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Huanyu Xie
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Jianyi Xiong
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Weimin Zhu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| |
Collapse
|
7
|
Ferrao Blanco MN, Bastiaansen-Jenniskens YM, Chambers MG, Pitsillides AA, Narcisi R, van Osch GJ. Effect of Inflammatory Signaling on Human Articular Chondrocyte Hypertrophy: Potential Involvement of Tissue Repair Macrophages. Cartilage 2021; 13:168S-174S. [PMID: 34165367 PMCID: PMC8739598 DOI: 10.1177/19476035211021907] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE In osteoarthritis, chondrocytes tend to acquire a hypertrophic phenotype, which contributes to the modification of the extracellular matrix, resulting in permanent cartilage changes. In mouse chondrocytes, pro-inflammatory macrophages and pro-inflammatory cytokines have been shown to stimulate hypertrophy via the activation of the nuclear factor kappa B (NF-κB) pathway. Whether or not this also occurs in human chondrocytes remains unclear. We therefore aimed to investigate whether hypertrophy-like responses in human cartilage are driven mainly by intrinsic inflammatory signaling or shaped by specific macrophage populations. DESIGN Human articular chondrocytes were cultured with pro-inflammatory cytokines or medium conditioned by defined macrophage subsets. Furthermore, the effect of inhibition of NF-κB-dependent gene expression was evaluated using the NF-κB inhibitor SC-514. Hypertrophy was assessed by measuring the transcription level of alkaline phosphatase (ALPL), type X collagen (COL10A1), Indian hedgehog (IHH), and runt-related transcription factor 2 (RUNX2). RESULTS The expression of hypertrophic genes was not promoted in human chondrocytes by pro-inflammatory cytokines neither pro-inflammatory M(IFNγ + TNFα) macrophages. Inhibition of the NF-κB-dependent gene expression did not affect human articular chondrocyte hypertrophy. However, tissue repair M(IL4) macrophages induced hypertrophy by promoting the expression of COL10A1, RUNX2, and IHH. CONCLUSION Intrinsic inflammatory signaling activation is not involved in the hypertrophic shift observed in human articular chondrocytes cultured in vitro. However, tissue repair macrophages may contribute to the onset of this detrimental phenotype in human osteoarthritic cartilage, given the effect observed in our experimental models.
Collapse
Affiliation(s)
- Mauricio N. Ferrao Blanco
- Department of Orthopaedics and
Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam,
The Netherlands
| | | | - Mark G. Chambers
- Lilly Research Laboratories, Eli
Lilly Pharmaceuticals, Indianapolis, IN, USA
| | | | - Roberto Narcisi
- Department of Orthopaedics and
Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam,
The Netherlands
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics and
Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam,
The Netherlands,Department of
Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam,
Rotterdam, The Netherlands,Department of Biomechanical
Engineering, TU Delft, Delft, The Netherlands,Gerjo J.V.M. van Osch, Erasmus MC,
University Medical Center Rotterdam, Wytemaweg 80, Room Ee 16.51b,
Rotterdam, 3015 CN, The Netherlands.
| |
Collapse
|
8
|
Subedar OD, Chiu LLY, Waldman SD. Cell Cycle Synchronization of Primary Articular Chondrocytes Enhances Chondrogenesis. Cartilage 2021; 12:526-535. [PMID: 30971093 PMCID: PMC8461165 DOI: 10.1177/1947603519841677] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Although tissue engineering is a promising option for articular cartilage repair, it has been challenging to generate functional cartilaginous tissue. While the synthetic response of chondrocytes can be influenced by various means, most approaches treat chondrocytes as a homogeneous population that would respond similarly. However, isolated cells heterogeneously progress through the cell cycle, which can affect macromolecular biosynthesis. As it is possible to synchronize cells within discrete cell cycle phases, the purpose of this study was to investigate the effects of cell cycle synchronization on the chondrogenic potential of primary articular chondrocytes. DESIGN Different methods of cell synchronization (serum starvation, thymidine, nocodazole, aphidicolin, and RO-3306) were tested for their ability to synchronize primary articular chondrocytes during the process of cell isolation. Cells (unsynchronized and synchronized) were then encapsulated in alginate gels, cultured for 4 weeks, and analyzed for their structural and biochemical properties. RESULTS The double-thymidine method yielded the highest level of cell purity, with cells synchronized in S phase. While the cells started to lose synchronization after 24 hours, tissue constructs developed from initially S phase synchronized cells had significantly higher glycosaminoglycan and collagen II amounts than those developed using unsynchronized cells. CONCLUSIONS Initial synchronization led to long-term changes in cartilaginous tissue formation. This effect was postulated to be due to the rapid auto-induction of TGF-βs by actively dividing S phase cells, thereby stimulating chondrogenesis. Cell synchronization methods may also be applied in conjunction with redifferentiation methods to improve the chondrogenic potential of dedifferentiated or diseased chondrocytes.
Collapse
Affiliation(s)
- Omar D. Subedar
- Department of Chemical Engineering, Ryerson University, Toronto, Ontario, Canada,Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Loraine L. Y. Chiu
- Department of Chemical Engineering, Ryerson University, Toronto, Ontario, Canada,Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Stephen D. Waldman
- Department of Chemical Engineering, Ryerson University, Toronto, Ontario, Canada,Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada,Stephen D. Waldman, Department of Chemical Engineering, Faculty of Engineering & Architectural Science, Ryerson University, Kerr Hall South, KHS 241N, Toronto, Ontario, Canada M5B 2K3.
| |
Collapse
|
9
|
Lee S, Chae DS, Song BW, Lim S, Kim SW, Kim IK, Hwang KC. ADSC-Based Cell Therapies for Musculoskeletal Disorders: A Review of Recent Clinical Trials. Int J Mol Sci 2021; 22:ijms221910586. [PMID: 34638927 PMCID: PMC8508846 DOI: 10.3390/ijms221910586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 01/04/2023] Open
Abstract
Recently published clinical trials involving the use of adipose-derived stem cells (ADSCs) indicated that approximately one-third of the studies were conducted on musculoskeletal disorders (MSD). MSD refers to a wide range of degenerative conditions of joints, bones, and muscles, and these conditions are the most common causes of chronic disability worldwide, being a major burden to the society. Conventional treatment modalities for MSD are not sufficient to correct the underlying structural abnormalities. Hence, ADSC-based cell therapies are being tested as a form of alternative, yet more effective, therapies in the management of MSDs. Therefore, in this review, MSDs subjected to the ADSC-based therapy were further categorized as arthritis, craniomaxillofacial defects, tendon/ligament related disorders, and spine disorders, and their brief characterization as well as the corresponding conventional therapeutic approaches with possible mechanisms with which ADSCs produce regenerative effects in disease-specific microenvironments were discussed to provide an overview of under which circumstances and on what bases the ADSC-based cell therapy was implemented. Providing an overview of the current status of ADSC-based cell therapy on MSDs can help to develop better and optimized strategies of ADSC-based therapeutics for MSDs as well as help to find novel clinical applications of ADSCs in the near future.
Collapse
Affiliation(s)
- Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Dong-Sik Chae
- Department of Orthopedic Surgery, International St. Mary’s Hospital, Catholic Kwandong University, Gangneung 210-701, Korea;
| | - Byeong-Wook Song
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Il-Kwon Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
- Correspondence: (I.-K.K.); (K.-C.H.); Fax: +82-32-290-2774 (K.-C.H.)
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
- Correspondence: (I.-K.K.); (K.-C.H.); Fax: +82-32-290-2774 (K.-C.H.)
| |
Collapse
|
10
|
Urlić I, Ivković A. Cell Sources for Cartilage Repair-Biological and Clinical Perspective. Cells 2021; 10:cells10092496. [PMID: 34572145 PMCID: PMC8468484 DOI: 10.3390/cells10092496] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 01/04/2023] Open
Abstract
Cell-based therapy represents a promising treatment strategy for cartilage defects. Alone or in combination with scaffolds/biological signals, these strategies open many new avenues for cartilage tissue engineering. However, the choice of the optimal cell source is not that straightforward. Currently, various types of differentiated cells (articular and nasal chondrocytes) and stem cells (mesenchymal stem cells, induced pluripotent stem cells) are being researched to objectively assess their merits and disadvantages with respect to the ability to repair damaged articular cartilage. In this paper, we focus on the different cell types used in cartilage treatment, first from a biological scientist’s perspective and then from a clinician’s standpoint. We compare and analyze the advantages and disadvantages of these cell types and offer a potential outlook for future research and clinical application.
Collapse
Affiliation(s)
- Inga Urlić
- Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Correspondence: (I.U.); (A.I.)
| | - Alan Ivković
- Department of Orthopaedic Surgery, University Hospital Sveti Duh, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Clinical Medicine, University of Applied Health Sciences, 10000 Zagreb, Croatia
- Correspondence: (I.U.); (A.I.)
| |
Collapse
|
11
|
Stampoultzis T, Karami P, Pioletti DP. Thoughts on cartilage tissue engineering: A 21st century perspective. Curr Res Transl Med 2021; 69:103299. [PMID: 34192658 DOI: 10.1016/j.retram.2021.103299] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 04/11/2021] [Accepted: 05/26/2021] [Indexed: 12/15/2022]
Abstract
In mature individuals, hyaline cartilage demonstrates a poor intrinsic capacity for repair, thus even minor defects could result in progressive degeneration, impeding quality of life. Although numerous attempts have been made over the past years for the advancement of effective treatments, significant challenges still remain regarding the translation of in vitro cartilage engineering strategies from bench to bedside. This paper reviews the latest concepts on engineering cartilage tissue in view of biomaterial scaffolds, tissue biofabrication, mechanobiology, as well as preclinical studies in different animal models. The current work is not meant to provide a methodical review, rather a perspective of where the field is currently focusing and what are the requirements for bridging the gap between laboratory-based research and clinical applications, in light of the current state-of-the-art literature. While remarkable progress has been accomplished over the last 20 years, the current sophisticated strategies have reached their limit to further enhance healthcare outcomes. Considering a clinical aspect together with expertise in mechanobiology, biomaterial science and biofabrication methods, will aid to deal with the current challenges and will present a milestone for the furtherance of functional cartilage engineering.
Collapse
Affiliation(s)
| | - Peyman Karami
- Laboratory of Biomechanical Orthopedics, EPFL, Lausanne, Switzerland.
| | | |
Collapse
|
12
|
Ping SH, Tian FM, Liu H, Sun Q, Shao LT, Lian QQ, Zhang L. Raloxifene inhibits the overexpression of TGF-β1 in cartilage and regulates the metabolism of subchondral bone in rats with osteoporotic osteoarthritis. Bosn J Basic Med Sci 2021; 21:284-293. [PMID: 33259777 PMCID: PMC8112563 DOI: 10.17305/bjbms.2020.5142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/16/2020] [Indexed: 01/18/2023] Open
Abstract
Overexpression of transforming growth factor-beta 1 (TGF-β1) and subchondral bone remodelling play key roles in osteoarthritis (OA). Raloxifene (RAL) reduces the serum level of TGF-β1 in postmenopausal women. However, the effect of RAL on TGF-β1 expression in articular cartilage is still unclear. Therefore, we aimed to investigate the protective effect of RAL on osteoporotic osteoarthritis via affecting TGF-β1 expression in cartilage and the metabolism of subchondral bone. Osteoporotic osteoarthritis was induced by a combination of anterior cruciate transection (ACLT) and ovariectomy (OVX). Rats were divided into five groups (n = 12): The sham group, the ACLT group, the OVX group, the ACLT + OVX group, and the RAL group (ACLT + OVX + RAL, 6.25 mg/kg/day for 12 weeks). Assessment was performed by histomorphology, microcomputed tomography (micro-CT) scan, immunohistochemistry, and tartrate-resistant acid phosphatase (TRAP) staining. We found that severe cartilage degeneration was shown in the ACLT + OVX group. The histomorphological scores, the levels of TGF-β1, and its related catabolic enzymes and osteoclasts numbers in the ACLT + OVX group were higher than those in other groups (p < 0.05). Furthermore, structure model index (SMI) and trabecular spacing (Tb.Sp) were decreased (p < 0.05), while bone mineral density (BMD), bone volume fraction (BV/TV), and trabecular number (Tb.N) were increased by RAL compared with the ACLT + OVX group (p < 0.05). Our findings demonstrated that RAL in clinical doses retards the development of osteoporotic osteoarthritis by inhibiting the overexpression of TGF-β1 in cartilage and regulating the metabolism of subchondral bone. These results provide support for RAL in the expansion of clinical indication for prevention and treatment in postmenopausal osteoarthritis.
Collapse
Affiliation(s)
- Shao-Hua Ping
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, China
| | - Fa-Ming Tian
- Medical Research Center, North China University of Science and Technology, Tangshan, China
| | - Hao Liu
- Department of Orthopedic Surgery, Affiliated Hospital of North China University of Science and Technology, Tangshan, China
| | - Qi Sun
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, China
| | - Li-Tao Shao
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, China
| | - Qiang-Qiang Lian
- Department of Orthopedic Surgery, the Affiliated Hospital of North China University of Science and Technology, Tangshan, China
| | - Liu Zhang
- Department of Orthopedic Surgery, Hebei Medical University, Shijiazhuang, China; Department of Orthopedic Surgery, Emergency General Hospital, Beijing, China
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW The decreased contact area, edge loading, and increased stress in the adjacent area cartilage resulting from chondral defects are believed to predispose this tissue to degenerative changes that have significant economic implications, especially when considering its progression to osteoarthritis of the knee. Growth factors are considered therapeutic possibilities to enhance healing of chondral injuries and modify the progression to degenerative arthritis. Thus, the purposes of this review are to first to summarize important points for defect preparation and recent advances in techniques for marrow stimulation and second, and to identify specific growth factors and cytokines that have the capacity to advance cartilage regeneration and the treatment of osteoarthritis in light of recent laboratory and clinical studies. RECENT FINDINGS TGF-β, BMP-2, BMP-7, IGF-1, as IL-1 receptor antagonist, and recombinant human FGF-18 are some of the promising growth factor/cytokine treatments with pioneering and evolving clinical developments. The bulk of the review describes and discusses these developments in light of fundamental basic science. It is crucial to also understand the other underlying advances made in the surgical management of cartilage defects prior to onset of OA. These advances are in techniques for defect preparation and marrow stimulation, a common cartilage repair procedure used in combination with growth factor/cytokine augmentation. Multiple growth factor/cytokine modulation therapies are currently undergoing clinical trial investigation including Invossa (currently in phase III study), Kineret (currently in phase I study), and Sprifermin (currently in phase II study) for the treatment of symptomatic osteoarthritis.
Collapse
|
14
|
Satake H, Sakata T. Estimation of Extracellular Matrix Production Using a Cultured-Chondrocyte-Based Gate Ion-Sensitive Field-Effect Transistor. Anal Chem 2019; 91:16017-16022. [DOI: 10.1021/acs.analchem.9b04789] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hiroto Satake
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Toshiya Sakata
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
15
|
Pourakbari R, Khodadadi M, Aghebati-Maleki A, Aghebati-Maleki L, Yousefi M. The potential of exosomes in the therapy of the cartilage and bone complications; emphasis on osteoarthritis. Life Sci 2019; 236:116861. [PMID: 31513815 DOI: 10.1016/j.lfs.2019.116861] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/04/2019] [Accepted: 09/08/2019] [Indexed: 12/13/2022]
Abstract
Osteoarthritis is a prevalent worldwide joint disease, which demonstrates a remarkable adverse effect on the patients' life modality. Medicinal agents, exclusively nonsteroidal anti-inflammatory drugs (NSAIDs), have been routinely applied in the clinic. But, their effects are restricted to pain control with insignificant effects on cartilage renovation, which would finally lead to cartilage destruction. In the field of regenerative medicine, many researchers have tried to use stem cells to repair tissues and other human organs. However, in recent years, with the discovery of extracellular microvesicles, especially exosomes, researchers have been able to offer more exciting alternatives on the subject. Exosomes and microvesicles are derived from different types of bone cells such as mesenchymal stem cells, osteoblasts, and osteoclasts. They are also recognized to play substantial roles in bone remodeling processes including osteogenesis, osteoclastogenesis, and angiogenesis. Specifically, exosomes derived from a mesenchymal stem cell have shown a great potential for the desired purpose. Exosomal products include miRNA, DNA, proteins, and other factors. At present, if it is possible to extract exosomes from various stem cells effectively and load certain products or drugs into them, they can be used in diseases, such as rheumatoid arthritis, osteoarthritis, bone fractures, and other diseases. Of course, to achieve proper clinical use, advances have to be made to establish a promising regenerative ability for microvesicles for treatment purposes in the orthopedic disorders. In this review, we describe the exosomes biogenesis and bone cell derived exosomes in the regenerate process of bone and cartilage remodeling.
Collapse
Affiliation(s)
- Ramin Pourakbari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Meysam Khodadadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Schneider MC, Chu S, Randolph MA, Bryant SJ. An in vitro and in vivo comparison of cartilage growth in chondrocyte-laden matrix metalloproteinase-sensitive poly(ethylene glycol) hydrogels with localized transforming growth factor β3. Acta Biomater 2019; 93:97-110. [PMID: 30914256 DOI: 10.1016/j.actbio.2019.03.046] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 12/25/2022]
Abstract
While matrix-assisted autologous chondrocyte implantation has emerged as a promising therapy to treat focal chondral defects, matrices that support regeneration of hyaline cartilage remain challenging. The goal of this work was to investigate the potential of a matrix metalloproteinase (MMP)-sensitive poly(ethylene glycol) (PEG) hydrogel containing the tethered growth factor, transforming growth factor β3 (TGF-β3), and compare cartilage regeneration in vitro and in vivo. The in vitro environment comprised chemically-defined medium while the in vivo environment utilized the subcutaneous implant model in athymic mice. Porcine chondrocytes were isolated and expanded in 2D culture for 10 days prior to encapsulation. The presence of tethered TGF-β3 reduced cell spreading. Chondrocyte-laden hydrogels were analyzed for total sulfated glycosaminoglycan and collagen contents, MMP activity, and spatial deposition of aggrecan, decorin, biglycan, and collagens type II and I. The total amount of extracellular matrix (ECM) deposited in the hydrogel constructs was similar in vitro and in vivo. However, the in vitro environment was not able to support long-term culture up to 64 days of the engineered cartilage leading to the eventual breakdown of aggrecan. The in vivo environment, on the other hand, led to more elaborate ECM, which correlated with higher MMP activity, and an overall higher quality of engineered tissue that was rich in aggrecan, decorin, biglycan and collagen type II with minimal collagen type I. Overall, the MMP-sensitive PEG hydrogel containing tethered TGF-β3 is a promising matrix for hyaline cartilage regeneration in vivo. STATEMENT OF SIGNIFICANCE: Regenerating hyaline cartilage remains a significant clinical challenge. The resultant repair tissue is often fibrocartilage, which long-term cannot be sustained. The goal of this study was to investigate the potential of a synthetic hydrogel matrix containing peptide crosslinks that can be degraded by enzymes secreted by encapsulated cartilage cells (i.e., chondrocytes) and tethered growth factors, specifically TGF-β3, to provide localized chondrogenic cues to the cells. This hydrogel led to hyaline cartilage-like tissue growth in vitro and in vivo, with minimal formation of fibrocartilage. However, the tissue formed in vitro, could not be maintained long-term. In vivo this hydrogel shows great promise as a potential matrix for use in regenerating hyaline cartilage.
Collapse
|
17
|
Charlier E, Deroyer C, Ciregia F, Malaise O, Neuville S, Plener Z, Malaise M, de Seny D. Chondrocyte dedifferentiation and osteoarthritis (OA). Biochem Pharmacol 2019; 165:49-65. [DOI: 10.1016/j.bcp.2019.02.036] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/28/2019] [Indexed: 02/08/2023]
|
18
|
Vermeulen S, Vasilevich A, Tsiapalis D, Roumans N, Vroemen P, Beijer NRM, Dede Eren A, Zeugolis D, de Boer J. Identification of topographical architectures supporting the phenotype of rat tenocytes. Acta Biomater 2019; 83:277-290. [PMID: 30394345 DOI: 10.1016/j.actbio.2018.10.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 09/22/2018] [Accepted: 10/25/2018] [Indexed: 01/01/2023]
Abstract
Tenocytes, the main cell type of the tendon, require mechanical stimuli for their proper function. When the tenocyte environment changes due to tissue damage or by transferring tenocytes from their native environment into cell culture, the signals from the tenocyte niche are lost, leading towards a decline of phenotypic markers. It is known that micro-topographies can influence cell fate by the physical cues they provide. To identify the optimal topography-induced biomechanical niche in vitro, we seeded tenocytes on the TopoChip, a micro-topographical screening platform, and measured expression of the tendon transcription factor Scleraxis. Through machine learning algorithms, we associated elevated Scleraxis levels with topological design parameters. Fabricating micro-topographies with optimal surface characteristics on larger surfaces allowed finding an improved expression of multiple tenogenic markers. However, long-term confluent culture conditions coincided with osteogenic marker expression and the loss of morphological characteristics. In contrast, passaging tenocytes which migrated from the tendon directly on the topography resulted in prolonged elongated morphology and elevated Scleraxis levels. This research provides new insights into how micro-topographies influence tenocyte cell fate, and supports the notion that micro-topographical design can be implemented in a new generation of tissue culture platforms for supporting the phenotype of tenocytes. STATEMENT OF SIGNIFICANCE: The challenge in controlling in vitro cell behavior lies in controlling the complex culture environment. Here, we present for the first time the use of micro-topographies as a biomechanical niche to support the phenotype of tenocytes. For this, we applied the TopoChip platform, a screening tool with 2176 unique micro-topographies for identifying feature characteristics associated with elevated Scleraxis expression, a tendon related marker. Large area fabrication of micro-topographies with favorable characteristics allowed us to find a beneficial influence on other tenogenic markers as well. Furthermore, passaging cells is more beneficial for Scleraxis marker expression and tenocyte morphology compared to confluent conditions. This study presents important insights for the understanding of tenocyte behavior in vitro, a necessary step towards tendon engineering.
Collapse
Affiliation(s)
- Steven Vermeulen
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, The Netherlands
| | - Aliaksei Vasilevich
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, The Netherlands
| | - Dimitrios Tsiapalis
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland; Science Foundation Ireland, Centre for Research in Medical Device, National University of Ireland Galway, Galway, Ireland
| | - Nadia Roumans
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, The Netherlands
| | - Pascal Vroemen
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, The Netherlands; University Eye Clinic Maastricht UMC+, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Nick R M Beijer
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, The Netherlands
| | - Aysegul Dede Eren
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, The Netherlands
| | - Dimitrios Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory, National University of Ireland Galway, Galway, Ireland; Science Foundation Ireland, Centre for Research in Medical Device, National University of Ireland Galway, Galway, Ireland
| | - Jan de Boer
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, The Netherlands; Dept. of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
19
|
Onset and Progression of Human Osteoarthritis-Can Growth Factors, Inflammatory Cytokines, or Differential miRNA Expression Concomitantly Induce Proliferation, ECM Degradation, and Inflammation in Articular Cartilage? Int J Mol Sci 2018; 19:ijms19082282. [PMID: 30081513 PMCID: PMC6121276 DOI: 10.3390/ijms19082282] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 07/22/2018] [Accepted: 08/01/2018] [Indexed: 12/30/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative whole joint disease, for which no preventative or therapeutic biological interventions are available. This is likely due to the fact that OA pathogenesis includes several signaling pathways, whose interactions remain unclear, especially at disease onset. Early OA is characterized by three key events: a rarely considered early phase of proliferation of cartilage-resident cells, in contrast to well-established increased synthesis, and degradation of extracellular matrix components and inflammation, associated with OA progression. We focused on the question, which of these key events are regulated by growth factors, inflammatory cytokines, and/or miRNA abundance. Collectively, we elucidated a specific sequence of the OA key events that are described best as a very early phase of proliferation of human articular cartilage (AC) cells and concomitant anabolic/catabolic effects that are accompanied by incipient pro-inflammatory effects. Many of the reviewed factors appeared able to induce one or two key events. Only one factor, fibroblast growth factor 2 (FGF2), is capable of concomitantly inducing all key events. Moreover, AC cell proliferation cannot be induced and, in fact, is suppressed by inflammatory signaling, suggesting that inflammatory signaling cannot be the sole inductor of all early OA key events, especially at disease onset.
Collapse
|
20
|
Lee J, Lee JY, Chae BC, Jang J, Lee E, Son Y. Fully Dedifferentiated Chondrocytes Expanded in Specific Mesenchymal Stem Cell Growth Medium with FGF2 Obtains Mesenchymal Stem Cell Phenotype In Vitro but Retains Chondrocyte Phenotype In Vivo. Cell Transplant 2018; 26:1673-1687. [PMID: 29251111 PMCID: PMC5753982 DOI: 10.1177/0963689717724794] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Given recent progress in regenerative medicine, we need a means to expand chondrocytes in quantity without losing their regenerative capability. Although many reports have shown that growth factor supplementation can have beneficial effects, the use of growth factor-supplemented basal media has widespread effect on the characteristics of chondrocytes. Chondrocytes were in vitro cultured in the 2 most widely used chondrocyte growth media, conventional chondrocyte culture medium and mesenchymal stem cell (MSC) culture medium, both with and without fibroblast growth factor-2 (FGF2) supplementation. Their expansion rates, expressions of extracellular matrix-related factors, senescence, and differentiation potentials were examined in vitro and in vivo. Our results revealed that chondrocytes quickly dedifferentiated during expansion in all tested media, as assessed by the loss of type II collagen expression. The 2 basal media (chondrocyte culture medium vs. MSC culture medium) were associated with distinct differences in cell senescence. Consistent with the literature, FGF2 was associated with accelerated dedifferentiation during expansion culture and superior redifferentiation upon induction. However, chondrocytes expanded in FGF2-containing conventional chondrocyte culture medium showed MSC-like features, as indicated by their ability to direct ectopic bone formation and cartilage formation. In contrast, chondrocytes cultured in FGF2-supplemented MSC culture medium showed potent chondrogenesis and almost no bone formation. The present findings show that the chosen basal medium can exert profound effects on the characteristics and activity of in vitro-expanded chondrocytes and indicate that right growth factor/medium combination can help chondrocytes retain a high-level chondrogenic potential without undergoing hypertrophic transition.
Collapse
Affiliation(s)
- Jungsun Lee
- 1 R&D Institute, Biosolution Inc., Seoul, South Korea
| | - Jin-Yeon Lee
- 1 R&D Institute, Biosolution Inc., Seoul, South Korea
| | | | - Jeongho Jang
- 2 Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin, South Korea
| | - EunAh Lee
- 2 Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin, South Korea.,3 Impedance Imaging Research Center, Kyung Hee University, Seoul, South Korea
| | - Youngsook Son
- 2 Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin, South Korea
| |
Collapse
|
21
|
Satake H, Saito A, Sakata T. Elucidation of interfacial pH behaviour at the cell/substrate nanogap for in situ monitoring of cellular respiration. NANOSCALE 2018; 10:10130-10136. [PMID: 29781490 DOI: 10.1039/c8nr02950d] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In situ monitoring of cellular metabolism is useful for elucidating dynamic functions of living cells. In our previous studies, cellular respiration was continuously monitored as a change in pH at the cell/electrode nanoscale interface (i.e., interfacial pH) using an ion-sensitive field-effect transistor (ISFET). However, such interfacial pH behaviour on the nanoscale has not been confirmed using other methods such as fluorescence imaging. In this study, we have clarified the interfacial pH behaviour at a cell/substrate nanogap using a laser scanning confocal fluorescence microscope. The phospholipid fluorescein used as a pH indicator was fixed to the plasma membrane on the external side of a cell by inserting its lipophilic alkyl chain into the membrane, and used to observe the change in interfacial pH. As a result, hydrogen ions generated by cellular respiration were gradually accumulated at the cell/substrate nanogap, resulting in a decrease in pH. Moreover, the interfacial pH between the plasma membrane and the substrate became lower than the pH near the surface of cells not in contact with the substrate. The data obtained in this study support the idea that potentiometric ion sensors such as ISFETs can detect a cellular-metabolism-induced change in pH at a cell/electrode nanogap in real time.
Collapse
Affiliation(s)
- Hiroto Satake
- Department of Materials Engineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan 113-8656.
| | | | | |
Collapse
|
22
|
Goepfert C, Lutz V, Lünse S, Kittel S, Wiegandt K, Kammal M, Püschel K, Pörtner R. Evaluation of Cartilage Specific Matrix Synthesis of Human Articular Chondrocytes after Extended Propagation on Microcarriers by Image Analysis. Int J Artif Organs 2018. [DOI: 10.1177/039139881003300405] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Cell-based technologies for the repair of cartilage defects usually rely on the expansion of low numbers of chondrocytes isolated from biopsies of healthy cartilage. Proliferating chondrocytes are known to undergo dedifferentiation characterized by downregulation of collagen type II and proteoglycan production, and by upregulation of collagen type I synthesis. Re-expression of cartilage specific matrix components by expanded chondrocytes is therefore critical for successful cartilage repair. Methods Human articular chondrocytes were expanded on microcarriers Cytodex 3. The growth area was increased by adding empty microcarriers. Added microcarriers were colonized by bead-to-bead transfer of the cells. The chondrocytes were harvested from the microcarriers and characterized by their ability to synthesize collagen type II when cultivated in alginate beads using chondrogenic growth factors. A semi-automatic image analysis technique was developed to determine the fractions of collagen type II and type I positive cells. Results The expansion of human articular chondrocytes on microcarriers yielded high cell numbers and propagation rates compared to chondrocytes expanded in flask culture for one passage. The proportion of collagen type II positive cells compared to collagen type I synthesizing cells was increased compared to chondrocytes expanded using conventional methods. The matrix synthesis upon treatment with chondrogenic factors IGF-I and BMP-7 was enhanced whereas TGF-β had an inhibitory effect on microcarrier expanded chondrocytes. Conclusions Expanding human articular chondrocytes on microcarriers omitting subcultivation steps leads to superior ratios of collagen type II to type I forming cells compared to the expansion in conventional monolayer culture.
Collapse
Affiliation(s)
- Christiane Goepfert
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| | - Vivien Lutz
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| | - Svenja Lünse
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| | - Sabrina Kittel
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| | - Katharina Wiegandt
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| | - Michael Kammal
- University Medical Center Hamburg-Eppendorf, Department of Legal Medicine, Hamburg - Germany
| | - Klaus Püschel
- University Medical Center Hamburg-Eppendorf, Department of Legal Medicine, Hamburg - Germany
| | - Ralf Pörtner
- Hamburg University of Technology, Institute of Bioprocess and Biosystems Engineering, Hamburg - Germany
| |
Collapse
|
23
|
Effect of Cyclic Dynamic Compressive Loading on Chondrocytes and Adipose-Derived Stem Cells Co-Cultured in Highly Elastic Cryogel Scaffolds. Int J Mol Sci 2018; 19:ijms19020370. [PMID: 29373507 PMCID: PMC5855592 DOI: 10.3390/ijms19020370] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 12/23/2022] Open
Abstract
In this study, we first used gelatin/chondroitin-6-sulfate/hyaluronan/chitosan highly elastic cryogels, which showed total recovery from large strains during repeated compression cycles, as 3D scaffolds to study the effects of cyclic dynamic compressive loading on chondrocyte gene expression and extracellular matrix (ECM) production. Dynamic culture of porcine chondrocytes was studied at 1 Hz, 10% to 40% strain and 1 to 9 h/day stimulation duration, in a mechanical-driven multi-chamber bioreactor for 14 days. From the experimental results, we could identify the optimum dynamic culture condition (20% and 3 h/day) to enhance the chondrocytic phenotype of chondrocytes from the expression of marker (Col I, Col II, Col X, TNF-α, TGF-β1 and IGF-1) genes by quantitative real-time polymerase chain reactions (qRT-PCR) and production of ECM (GAGs and Col II) by biochemical analysis and immunofluorescence staining. With up-regulated growth factor (TGF-β1 and IGF-1) genes, co-culture of chondrocytes with porcine adipose-derived stem cells (ASCs) was employed to facilitate chondrogenic differentiation of ASCs during dynamic culture in cryogel scaffolds. By replacing half of the chondrocytes with ASCs during co-culture, we could obtain similar production of ECM (GAGs and Col II) and expression of Col II, but reduced expression of Col I, Col X and TNF-α. Subcutaneous implantation of cells/scaffold constructs in nude mice after mono-culture (chondrocytes or ASCs) or co-culture (chondrocytes + ASCs) and subject to static or dynamic culture condition in vitro for 14 days was tested for tissue-engineering applications. The constructs were retrieved 8 weeks post-implantation for histological analysis by Alcian blue, Safranin O and Col II immunohistochemical staining. The most abundant ectopic cartilage tissue was found for the chondrocytes and chondrocytes + ASCs groups using dynamic culture, which showed similar neo-cartilage formation capability with half of the chondrocytes replaced by ASCs for co-culture. This combined co-culture/dynamic culture strategy is expected to cut down the amount of donor chondrocytes needed for cartilage-tissue engineering.
Collapse
|
24
|
Chen M, Guo W, Gao S, Hao C, Shen S, Zhang Z, Wang Z, Wang Z, Li X, Jing X, Zhang X, Yuan Z, Wang M, Zhang Y, Peng J, Wang A, Wang Y, Sui X, Liu S, Guo Q. Biochemical Stimulus-Based Strategies for Meniscus Tissue Engineering and Regeneration. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8472309. [PMID: 29581987 PMCID: PMC5822894 DOI: 10.1155/2018/8472309] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/19/2017] [Indexed: 12/18/2022]
Abstract
Meniscus injuries are very common and still pose a challenge for the orthopedic surgeon. Meniscus injuries in the inner two-thirds of the meniscus remain incurable. Tissue-engineered meniscus strategies seem to offer a new approach for treating meniscus injuries with a combination of seed cells, scaffolds, and biochemical or biomechanical stimulation. Cell- or scaffold-based strategies play a pivotal role in meniscus regeneration. Similarly, biochemical and biomechanical stimulation are also important. Seed cells and scaffolds can be used to construct a tissue-engineered tissue; however, stimulation to enhance tissue maturation and remodeling is still needed. Such stimulation can be biomechanical or biochemical, but this review focuses only on biochemical stimulation. Growth factors (GFs) are one of the most important forms of biochemical stimulation. Frequently used GFs always play a critical role in normal limb development and growth. Further understanding of the functional mechanism of GFs will help scientists to design the best therapy strategies. In this review, we summarize some of the most important GFs in tissue-engineered menisci, as well as other types of biological stimulation.
Collapse
Affiliation(s)
- Mingxue Chen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Weimin Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Shunag Gao
- Center for Biomaterial and Tissue Engineering, Academy for Advanced Interdisciplinary Studies, No. 5 Yiheyuan Road, Haidian District, Peking University, Beijing 100871, China
| | - Chunxiang Hao
- Institute of Anesthesiology, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Shi Shen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
- Department of Bone and Joint Surgery, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Road, Luzhou 646000, China
| | - Zengzeng Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
- First Department of Orthopedics, First Affiliated Hospital of Jiamusi University, No. 348 Dexiang Road, Xiangyang District, Jiamusi 154002, China
| | - Zhenyong Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
- First Department of Orthopedics, First Affiliated Hospital of Jiamusi University, No. 348 Dexiang Road, Xiangyang District, Jiamusi 154002, China
| | - Zehao Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xu Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiaoguang Jing
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
- First Department of Orthopedics, First Affiliated Hospital of Jiamusi University, No. 348 Dexiang Road, Xiangyang District, Jiamusi 154002, China
| | - Xueliang Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
- Shanxi Traditional Chinese Hospital, No. 46 Binzhou West Street, Yingze District, Taiyuan 030001, China
| | - Zhiguo Yuan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Mingjie Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Yu Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Aiyuan Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xiang Sui
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Shuyun Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Quanyi Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries, PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| |
Collapse
|
25
|
Nowicki M, Castro NJ, Rao R, Plesniak M, Zhang LG. Integrating three-dimensional printing and nanotechnology for musculoskeletal regeneration. NANOTECHNOLOGY 2017; 28:382001. [PMID: 28762957 PMCID: PMC5612478 DOI: 10.1088/1361-6528/aa8351] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The field of tissue engineering is advancing steadily, partly due to advancements in rapid prototyping technology. Even with increasing focus, successful complex tissue regeneration of vascularized bone, cartilage and the osteochondral interface remains largely illusive. This review examines current three-dimensional printing techniques and their application towards bone, cartilage and osteochondral regeneration. The importance of, and benefit to, nanomaterial integration is also highlighted with recent published examples. Early-stage successes and challenges of recent studies are discussed, with an outlook to future research in the related areas.
Collapse
Affiliation(s)
- Margaret Nowicki
- Department of Mechanical and Aerospace Engineering, The George Washington University, 800 22nd Street, NW, Washington DC 20052, United States of America
| | | | | | | | | |
Collapse
|
26
|
Martin F, Lehmann M, Sack U, Anderer U. Featured Article: In vitro development of personalized cartilage microtissues uncovers an individualized differentiation capacity of human chondrocytes. Exp Biol Med (Maywood) 2017; 242:1746-1756. [PMID: 28853609 DOI: 10.1177/1535370217728498] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Personalized features in the treatment of knee injuries and articular replacement therapies play an important role in modern life with increasing demand. Therefore, cell-based therapeutic approaches for the regeneration of traumatic defects of cartilage tissue were developed. However, great variations in the quality of repair tissue or therapeutic outcome were observed. The aim of the study was to capture and visualize individual differentiation capacities of chondrocytes derived from different donors with regard to a possible personal regeneration capacity using a cell-based therapy. The redifferentiation potential of monolayer cultured cells was analyzed in a scaffold-free three-dimensional tissue model. Furthermore, stimulating options using cartilage maturation factors such as L-ascorbic acid and transforming growth factor beta 2 (TGF-β2) on this process were of special interest. Cells and tissues were analyzed via histological and immunohistochemical methods. Gene expression was measured by quantitative real-time polymerase chain reaction. In monolayer culture, cells from all donors showed an almost identical differentiation profile. In contrast, the differentiation state of cartilage-like three-dimensional microtissues revealed clear differences with respect to individual donors. Analyses at the protein and mRNA levels showed high variations regarding cartilage-typical matrix components (e.g. proteoglycans, collagen type II) and intracellular proteins (e.g. S100). Interestingly, only donor chondrocytes with a basic tendency to re-differentiate in a three-dimensional environment were able to increase this tissue-specific maturation when exposed to L-ascorbic acid and/or TGF-β2. Our approach revealed clear-cut possibilities for classification of individual donors into responders or non-responders. On the basis of these results an in vitro platform could be designed to discriminate responders from non-responders. This in vitro three-dimensional test system may be a suitable basis to establish a "personalized diagnostic tool" with the opportunity to assess the capacity of expanded chondrocytes to respond to an autologous cell-based therapy. Impact statement A challenge in cell-based cartilage regeneration therapies is the identification of a "personalized diagnostic tool" to predict the chondrogenic potency of cells from patients who are going to be treated with autologous cells. Comparing the phenotype of isolated chondrocytes from different donors in vitro revealed an individual cartilage-specific differentiation capacity. These personalized features are not detectable in vitro until the monolayer cells have the possibility to rearrange in 3D tissues. Cells from articular cartilage in monolayer culture may not be a suitable basis to discriminate responders from non-responders with respect to a personalized cell-based therapy to treat cartilage defects. A more physiological 3D (micro-)environment enable the cells to present their individual differentiation capacity. The here described microtissue model might be the basis for an in vitro platform to predict the therapeutic outcome of autologous cell-based cartilage repair and/or a suitable tool to identify early biomarkers to classify the patients.
Collapse
Affiliation(s)
- Frank Martin
- 1 Department of Cell Biology and Tissue Engineering, Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg D-01968, Germany.,2 Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig D-04103, Germany
| | - Mario Lehmann
- 1 Department of Cell Biology and Tissue Engineering, Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg D-01968, Germany.,2 Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig D-04103, Germany
| | - Ulrich Sack
- 2 Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig D-04103, Germany.,3 Translational Centre for Regenerative Medicine (TRM), University of Leipzig, Leipzig D-04103, Germany
| | - Ursula Anderer
- 1 Department of Cell Biology and Tissue Engineering, Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg D-01968, Germany
| |
Collapse
|
27
|
Lee JK, Huwe LW, Paschos N, Aryaei A, Gegg CA, Hu JC, Athanasiou KA. Tension stimulation drives tissue formation in scaffold-free systems. NATURE MATERIALS 2017; 16:864-873. [PMID: 28604717 PMCID: PMC5532069 DOI: 10.1038/nmat4917] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/04/2017] [Indexed: 05/04/2023]
Abstract
Scaffold-free systems have emerged as viable approaches for engineering load-bearing tissues. However, the tensile properties of engineered tissues have remained far below the values for native tissue. Here, by using self-assembled articular cartilage as a model to examine the effects of intermittent and continuous tension stimulation on tissue formation, we show that the application of tension alone, or in combination with matrix remodelling and synthesis agents, leads to neocartilage with tensile properties approaching those of native tissue. Implantation of tension-stimulated tissues results in neotissues that are morphologically reminiscent of native cartilage. We also show that tension stimulation can be translated to a human cell source to generate anisotropic human neocartilage with enhanced tensile properties. Tension stimulation, which results in nearly sixfold improvements in tensile properties over unstimulated controls, may allow the engineering of mechanically robust biological replacements of native tissue.
Collapse
Affiliation(s)
- Jennifer K. Lee
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Le W. Huwe
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Nikolaos Paschos
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Ashkan Aryaei
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Courtney A. Gegg
- Department of Bioengineering, Stanford University, 443 Via Ortega, Stanford, CA, 94305
| | - Jerry C. Hu
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
- Department of Orthopaedic Surgery, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
- Correspondence and reprint requests should be addressed to: KA Athanasiou, Tel.: (530) 754-6645, Fax: (530) 754-5739, , Department of Biomedical Engineering, University of California, Davis, One Shields Ave, Davis, CA 95616, USA
| |
Collapse
|
28
|
Koh S, Purser M, Wysk R, Piedrahita JA. Improved Chondrogenic Potential and Proteomic Phenotype of Porcine Chondrocytes Grown in Optimized Culture Conditions. Cell Reprogram 2017; 19:232-244. [PMID: 28749737 DOI: 10.1089/cell.2017.0005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
For successful cartilage tissue engineering, the ability to generate a high number of chondrocytes in vitro while avoiding terminal differentiation or de-differentiation is critical. The ability to accomplish this by using the abundant and easily sampled costal cartilage could provide a practical alternative to the use of articular cartilage and mesenchymal stem cells. Chondrocytes isolated from pig costal cartilage were expanded in either serum-free medium with FGF2 (SFM) or fetal bovine serum-containing medium (SCM), under either high (21%) or low (5%) oxygen conditions. Overall, chondrocytes cultured in SFM and low oxygen (Low-SFM) demonstrated the highest cell growth rate (p < 0.05). The effect of passage number on the differentiation status of the chondrocytes was analyzed by alkaline phosphatase (AP) staining and real-time PCR for known chondrocyte quality markers. AP staining indicated that chondrocytes grown in SCM had a higher proportion of terminally differentiated (hypertrophic) chondrocytes (p < 0.05). At the mRNA level, expression ratios of ACAN/VCAN and COL2/COL1 were significantly higher (p < 0.05) in cells expanded in Low-SFM, indicating reduced de-differentiation. In vitro re-differentiation capacity was assessed after a 6-week induction, and chondrocytes grown in Low-SFM showed similar expression ratios of COL2/COL1 and ACAN/VCAN to native cartilage. Proteomic analysis of in vitro produced cartilage indicated that the Low-SFM condition most closely matched the proteomic profile of native costal and articular cartilage. In conclusion, Low-SFM culture conditions resulted in improved cell growth rates, reduced levels of de-differentiation during expansion, greater ability to re-differentiate into cartilage on induction, and an improved proteomic profile that resembles that of in vivo cartilage.
Collapse
Affiliation(s)
- Sehwon Koh
- 1 Genomics Program, North Carolina State University , Raleigh, North Carolina.,2 Comparative Medicine Institute, North Carolina State University , Raleigh, North Carolina.,3 Department of Cell Biology, Duke University , Durham, North Carolina
| | - Molly Purser
- 4 Department of Industrial and Systems Engineering, North Carolina state University , Raleigh, North Carolina.,5 RTI Health Solutions, Research Triangle International , Raleigh, North Carolina
| | - Richard Wysk
- 2 Comparative Medicine Institute, North Carolina State University , Raleigh, North Carolina.,4 Department of Industrial and Systems Engineering, North Carolina state University , Raleigh, North Carolina
| | - Jorge A Piedrahita
- 1 Genomics Program, North Carolina State University , Raleigh, North Carolina.,2 Comparative Medicine Institute, North Carolina State University , Raleigh, North Carolina.,6 Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University , Raleigh, North Carolina
| |
Collapse
|
29
|
Tallheden T, van der Lee J, Brantsing C, Månsson JE, Sjögren-Jansson E, Lindahl A. Human Serum for Culture of Articular Chondrocytes. Cell Transplant 2017; 14:469-79. [PMID: 16285255 DOI: 10.3727/000000005783982909] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In the field of cell and tissue engineering, culture expansion of human cells in monolayer plays an important part. Traditionally, cell cultures have been supplemented with serum to support attachment and proliferation, but serum is a potential source of foreign protein contamination and viral protein transmission. In this study, we evaluated the use of human serum for experimental human articular chondrocyte expansion and to develop a method for preparation of large volumes of high-quality human serum from healthy blood donors. Human autologous serum contained high levels of epidermal-derived growth factor and platelet-derived growth factor-AB and supported proliferation up to 7 times higher than FCS in primary chondrocyte cultures. By letting the coagulation take place in a commercially available transfusion bag overnight, up to 250 ml of growth factor-rich human serum could be obtained from one donor. The allogenic human serum supported high proliferation rate without loosing expression of cartilage-specific genes. The expanded chondrocytes were able to redifferentiate and form cartilage matrix in comparable amounts to autologous serums. In conclusion, the transfusion bags allow preparation of large volumes of growth factor-rich human serum with the capacity to support in vitro cell expansion. The data further indicate that by controlling the coagulation process there are possibilities of optimizing the release of growth factors for other emerging cell therapies.
Collapse
Affiliation(s)
- Tommi Tallheden
- Department of Clinical Chemistry and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg University, Sweden.
| | | | | | | | | | | |
Collapse
|
30
|
Witt A, Salamon A, Boy D, Hansmann D, Büttner A, Wree A, Bader R, Jonitz-Heincke A. Gene expression analysis of growth factor receptors in human chondrocytes in monolayer and 3D pellet cultures. Int J Mol Med 2017; 40:10-20. [PMID: 28534942 PMCID: PMC5466384 DOI: 10.3892/ijmm.2017.2994] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 04/10/2017] [Indexed: 11/06/2022] Open
Abstract
The main goal of cartilage repair is to create functional tissue by enhancing the in vitro conditions to more physiological in vivo conditions. Chondrogenic growth factors play an important role in influencing cartilage homeostasis. Insulin‑like growth factor (IGF)‑1 and transforming growth factor (TGF)‑β1 affect the expression of collagen type II (Col2) and glycosaminoglycans (GAGs) and, therefore, the targeted use of growth factors could make chondrogenic redifferentiation more efficient. In the present study, human chondrocytes were postmortally isolated from healthy articular cartilage and cultivated as monolayer or 3D pellet cultures either under normoxia or hypoxia and stimulated with IGF‑1 and/or TGF‑β1 to compare the impact of the different growth factors. The mRNA levels of the specific receptors (IGF1R, TGFBR1, TGFBR2) were analyzed at different time points. Moreover, gene expression rates of collagen type 1 and 2 in pellet cultures were observed over a period of 5 weeks. Additionally, hyaline‑like Col2 protein and sulphated GAG (sGAG) levels were quantified. Stimulation with IGF‑1 resulted in an enhanced expression of IGF1R and TGFBR2 whereas TGF‑β1 stimulated TGFBR1 in the monolayer and pellet cultures. In monolayer, the differences reached levels of significance. This effect was more pronounced under hypoxic culture conditions. In pellet cultures, increased amounts of Col2 protein and sGAGs after incubation with TGF‑β1 and/or IGF‑1 were validated. In summary, constructing a gene expression profile regarding mRNA levels of specific growth factor receptors in monolayer cultures could be helpful for a targeted application of growth factors in cartilage tissue engineering.
Collapse
Affiliation(s)
- Anika Witt
- Department of Orthopaedics, Biomechanics and Implant Technology Research Laboratory, University Medical Centre Rostock, D‑18057 Rostock, Germany
| | - Achim Salamon
- Department of Cell Biology, University Medical Centre Rostock, D‑18057 Rostock, Germany
| | - Diana Boy
- Institute of Forensic Medicine, University Medical Centre Rostock, D‑18057 Rostock, Germany
| | - Doris Hansmann
- Department of Orthopaedics, Biomechanics and Implant Technology Research Laboratory, University Medical Centre Rostock, D‑18057 Rostock, Germany
| | - Andreas Büttner
- Institute of Forensic Medicine, University Medical Centre Rostock, D‑18057 Rostock, Germany
| | - Andreas Wree
- Institute of Anatomy, University Medical Centre Rostock, D‑18057 Rostock, Germany
| | - Rainer Bader
- Department of Orthopaedics, Biomechanics and Implant Technology Research Laboratory, University Medical Centre Rostock, D‑18057 Rostock, Germany
| | - Anika Jonitz-Heincke
- Department of Orthopaedics, Biomechanics and Implant Technology Research Laboratory, University Medical Centre Rostock, D‑18057 Rostock, Germany
| |
Collapse
|
31
|
Yin W, Xu H, Sheng J, Xu Z, Xie X, Zhang C. Comparative evaluation of the effects of platelet‑rich plasma formulations on extracellular matrix formation and the NF‑κB signaling pathway in human articular chondrocytes. Mol Med Rep 2017; 15:2940-2948. [PMID: 28339078 PMCID: PMC5428536 DOI: 10.3892/mmr.2017.6365] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 01/16/2017] [Indexed: 12/23/2022] Open
Abstract
Concentrated leukocytes in leukocyte and platelet-rich plasma (L-PRP) may deliver increased levels of pro-inflammatory cytokines to activate the nuclear factor (NF)-κB signaling pathway, to counter or overwhelm the beneficial effects of growth factors on cartilage regeneration. However, to date, no relevant studies have substantiated this. In the present study, L-PRP and pure platelet-rich plasma (P-PRP) were prepared, and leukocytes, platelets, pro-inflammatory cytokines and growth factor concentrations were quantified; they were then used to treat human articular chondrocytes (HACs). Pyrrolidine dithiocarbamate (PDTC; 50 µM) was used to inhibit the activation of NF-κB. The nuclear translocation of NF-κB p65 and the protein expression of cartilaginous markers (collagen II, aggrecan and sex-determining region Y-box 9) were determined using western blot analysis. The mRNA expression of NF-κB-dependent inflammatory mediators, including inducible nitric oxide synthase and cyclooxygenase-2, and cartilaginous markers were determined using reverse transcription-quantitative polymerase chain reaction analysis. The production of prostaglandin E2, nitric oxide and glycosaminoglycan (GAG) were quantified using enzyme-linked immunosorbent assays, the Griess reaction and a 1,9-dimethylmethylene blue assay, respectively. The results demonstrated that L-PRP induced the nuclear translocation of NF-κB p65, upregulated the mRNA expression of NF-κB-dependent inflammatory mediators and upregulated the production of their products, whereas P-PRP, which had similar growth factor concentrations but significantly lower pro-inflammatory cytokine concentrations than L-PRP, did not. P-PRP promoted the mRNA and protein expression levels of cartilaginous markers and the production of GAG more effectively, compared with L-PRP. Furthermore, inhibition of the activation of NF-κB by PDTC enhanced the effects of L-PRP on extracellular matrix formation in the HACs to a level similar to that of P-PRP. These findings suggested that leukocytes in L-PRP activated the NF-κB signaling pathway via the delivery of interleukin-1β and tumor necrosis factor-α to counter the beneficial effects of growth factors on extracellular matrix formation in HACs. Therefore, P-PRP may be more suitable for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Wenjing Yin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Haitao Xu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Jiagen Sheng
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Zhengliang Xu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Xuetao Xie
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Changqing Zhang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
32
|
Hyc A, Moskalewski S, Osiecka-Iwan A. Influence of cartilage interstitial fluid on the mRNA levels of matrix proteins, cytokines, metalloproteases and their inhibitors in synovial membrane. Int J Mol Med 2016; 38:937-42. [PMID: 27430724 DOI: 10.3892/ijmm.2016.2684] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/16/2016] [Indexed: 11/06/2022] Open
Abstract
Articular cartilage and the synovial membrane both ensure the smooth action of synovial joints; however, the influence of chondrocytes on synovial metabolism remains unclear. The secretory activity of chondrocytes is usually studied in cell cultures and may differ from that in intact cartilage. According to McCutchen's theory of 'weeping' joint lubrication, loading of the articular cartilage during motion squeezes the fluid with lubricating properties from the cartilage. The purpose of the study was to obtain cartilage interstitial fluid (CIF) from intact cartilage and to evaluate its influence on gene expression in the synovial membrane cells. CIF was rinsed out from the cartilage of newborn rats at a pressure of three bar. The chondrocytes survived rinsing and grew in culture. Cytokines in CIF were detected using the enzyme-linked immunosorbent assay (ELISA). The influence of CIF and CIF-like cocktail (all cytokines found in CIF) on gene expression in the synovial membrane cells was studied after a 4 h-incubation, by real-time PCR. Data were analyzed using the Wilcoxon matched-pair test or by the Mann‑Whitney U test. CIF contained basic fibroblast growth factor (bFGF), insulin-like growth factor (IGF)‑1, transforming growth factor β1 (TGFβ1), bone morphogenetic protein 7 (BMP7), macrophage (M)-colony-stimulating factor (CSF), granulocyte (G)-CSF and leukemia inhibitory factor (LIF). CIF stimulated the expression of hyaluronan synthase (HAS)1 and 2, lubricin, collagen I, versican, aggrecan, matrix metalloproteinases (MMPs)2 and 3, tissue inhibitors of metalloproteinases (TIMPs) 1-3, interleukin (IL)-6 and TGFβ1, and decreased the expression of tumor necrosis factor (TNF) and IL-1β. Incubation of the synovial membrane with CIF-like cocktail partially imitated the effects of CIF. Analysis of CIF composition may help to characterize the secretory activity of chondrocytes in their natural environment under various physiological and pathological conditions and to understand the interactions between articular cartilage and the synovial membrane.
Collapse
Affiliation(s)
- Anna Hyc
- Department of Histology and Embryology, Medical University of Warsaw, PL-02004 Warsaw, Poland
| | - Stanislaw Moskalewski
- Department of Histology and Embryology, Medical University of Warsaw, PL-02004 Warsaw, Poland
| | - Anna Osiecka-Iwan
- Department of Histology and Embryology, Medical University of Warsaw, PL-02004 Warsaw, Poland
| |
Collapse
|
33
|
Bioactive TGF-β1/HA Alginate-Based Scaffolds for Osteochondral Tissue Repair: Design, Realization and Multilevel Characterization. J Appl Biomater Funct Mater 2016; 14:e42-52. [DOI: 10.5301/jabfm.5000249] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2015] [Indexed: 11/20/2022] Open
Abstract
Background The design of an appropriate microenvironment for stem cell differentiation constitutes a multitask mission and a critical step toward the clinical application of tissue substitutes. With the aim of producing a bioactive material for orthopedic applications, a transforming growth factor-β (TGF- β1)/hydroxyapatite (HA) association within an alginate-based scaffold was investigated. The bioactive scaffold was carefully designed to offer specific biochemical cues for an efficient and selective cell differentiation toward the bony and chondral lineages. Methods Highly porous alginate scaffolds were fabricated from a mixture of calcium cross-linked alginates by means of a freeze-drying technique. In the chondral layer, the TGF in citric acid was mixed with an alginate/alginate-sulfate solution. In the bony layer, HA granules were added as bioactive signal, to offer an osteoinductive surface to the cells. Optical and scanning electron microscopy analyses were performed to assess the macro-micro architecture of the biphasic scaffold. Different mechanical tests were conducted to evaluate the elastic modulus of the grafts. For the biological validation of the developed prototype, mesenchymal stem cells were loaded onto the samples; cellular adhesion, proliferation and in vivo biocompatibility were evaluated. Results and conclusions The results successfully demonstrated the efficacy of the designed osteochondral graft, which combined interesting functional properties and biomechanical performances, thus becoming a promising candidate for osteochondral tissue-engineering applications.
Collapse
|
34
|
Hemphill DD, McIlwraith CW, Slayden RA, Samulski RJ, Goodrich LR. Adeno-associated virus gene therapy vector scAAVIGF-I for transduction of equine articular chondrocytes and RNA-seq analysis. Osteoarthritis Cartilage 2016; 24:902-11. [PMID: 26706703 DOI: 10.1016/j.joca.2015.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 11/08/2015] [Accepted: 12/01/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE IGF-I is one of several anabolic factors being investigated for the treatment of osteoarthritis (OA). Due to the short biological half-life, extended administration is required for more robust cartilage healing. Here we create a self-complimentary adeno-associated virus (AAV) gene therapy vector utilizing the transgene for IGF-I. DESIGN Various biochemical assays were performed to investigate the cellular response to scAAVIGF-I treatment vs an scAAVGFP positive transduction control and a negative for transduction control culture. RNA-sequencing analysis was also performed to establish a differential regulation profile of scAAVIGF-I transduced chondrocytes. RESULTS Biochemical analyses indicated an average media IGF-I concentration of 608 ng/ml in the scAAVIGF-I transduced chondrocytes. This increase in IGF-I led to increased expression of collagen type II and aggrecan and increased protein concentrations of cellular collagen type II and media glycosaminoglycan vs both controls. RNA-seq revealed a global regulatory pattern consisting of 113 differentially regulated GO categories including those for chondrocyte and cartilage development and regulation of apoptosis. CONCLUSIONS This research substantiates that scAAVIGF-I gene therapy vector increased production of IGF-I to clinically relevant levels with a biological response by chondrocytes conducive to increased cartilage healing. The RNA-seq further established a set of differentially expressed genes and gene ontologies induced by the scAAVIGF-I vector while controlling for AAV infection. This dataset provides a static representation of the cellular transcriptome that, while only consisting of one time point, will allow for further gene expression analyses to compare additional cartilage healing therapeutics or a transient cellular response.
Collapse
Affiliation(s)
- D D Hemphill
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA.
| | - C W McIlwraith
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA.
| | - R A Slayden
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| | - R J Samulski
- University of North Carolina Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - L R Goodrich
- Orthopaedic Research Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
35
|
Yin WJ, Xu HT, Sheng JG, An ZQ, Guo SC, Xie XT, Zhang CQ. Advantages of Pure Platelet-Rich Plasma Compared with Leukocyte- and Platelet-Rich Plasma in Treating Rabbit Knee Osteoarthritis. Med Sci Monit 2016; 22:1280-90. [PMID: 27086145 PMCID: PMC4837928 DOI: 10.12659/msm.898218] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Concentrated leukocytes in leukocyte- and platelet-rich plasma (L-PRP) may deliver increased levels of pro-inflammatory cytokines to activate the NF-κB signaling pathway, to counter the beneficial effects of growth factors on osteoarthritic cartilage. However, to date no relevant studies have substantiated that in vivo. Material/Methods Autologous L-PRP and pure platelet-rich plasma (P-PRP) were prepared, measured for componential composition, and injected intra-articularly after 4, 5, and 6 weeks post-anterior cruciate ligament transection. Caffeic acid phenethyl ester (CAPE) was injected intraperitoneally to inhibit NF-κB activation. All rabbits were sacrificed after 8 weeks postoperative. Enzyme-linked immunosorbent assays were performed to determine interleukin 1β (IL-1β) and prostaglandin E2 (PGE2) concentrations in the synovial fluid, Indian ink staining was performed for gross morphological assessment, and hematoxylin and eosin staining and toluidine blue staining were performed for histological assessment. Results Compared with L-PRP, P-PRP injections achieved better outcomes regarding the prevention of cartilage destruction, preservation of cartilaginous matrix, and reduction of IL-1β and PGE2 concentrations. CAPE injections reversed the increased IL-1β and PGE2 concentrations in the synovial fluid after L-PRP injections and improved the outcome of L-PRP injections to a level similar to P-PRP injections, while they had no influence on the therapeutic efficacy of P-PRP injections. Conclusions Concentrated leukocytes in L-PRP may release increased levels of pro-inflammatory cytokines to activate the NF-κB signaling pathway, to counter the beneficial effects of growth factors on osteoarthritic cartilage, and finally, result in a inferior efficacy of L-PRP to P-PRP for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Wen-Jing Yin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China (mainland)
| | - Hai-Tao Xu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China (mainland)
| | - Jia-Gen Sheng
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China (mainland)
| | - Zhi-Quan An
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China (mainland)
| | - Shang-Chun Guo
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China (mainland)
| | - Xue-Tao Xie
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China (mainland)
| | - Chang-Qing Zhang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China (mainland)
| |
Collapse
|
36
|
Kwon H, Paschos NK, Hu JC, Athanasiou K. Articular cartilage tissue engineering: the role of signaling molecules. Cell Mol Life Sci 2016; 73:1173-94. [PMID: 26811234 PMCID: PMC5435375 DOI: 10.1007/s00018-015-2115-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/23/2015] [Accepted: 12/10/2015] [Indexed: 02/08/2023]
Abstract
Effective early disease modifying options for osteoarthritis remain lacking. Tissue engineering approach to generate cartilage in vitro has emerged as a promising option for articular cartilage repair and regeneration. Signaling molecules and matrix modifying agents, derived from knowledge of cartilage development and homeostasis, have been used as biochemical stimuli toward cartilage tissue engineering and have led to improvements in the functionality of engineered cartilage. Clinical translation of neocartilage faces challenges, such as phenotypic instability of the engineered cartilage, poor integration, inflammation, and catabolic factors in the arthritic environment; these can all contribute to failure of implanted neocartilage. A comprehensive understanding of signaling molecules involved in osteoarthritis pathogenesis and their actions on engineered cartilage will be crucial. Thus, while it is important to continue deriving inspiration from cartilage development and homeostasis, it has become increasingly necessary to incorporate knowledge from osteoarthritis pathogenesis into cartilage tissue engineering.
Collapse
Affiliation(s)
- Heenam Kwon
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Nikolaos K Paschos
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Kyriacos Athanasiou
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA.
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA, USA.
| |
Collapse
|
37
|
Park JY, Choi YJ, Shim JH, Park JH, Cho DW. Development of a 3D cell printed structure as an alternative to autologs cartilage for auricular reconstruction. J Biomed Mater Res B Appl Biomater 2016; 105:1016-1028. [PMID: 26922876 DOI: 10.1002/jbm.b.33639] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/04/2016] [Accepted: 02/03/2016] [Indexed: 12/12/2022]
Abstract
Surgical technique using autologs cartilage is considered as the best treatment for cartilage tissue reconstruction, although the burdens of donor site morbidity and surgical complications still remain. The purpose of this study is to apply three-dimensional (3D) cell printing to fabricate a tissue-engineered graft, and evaluate its effects on cartilage reconstruction. A multihead tissue/organ building system is used to print cell-printed scaffold (CPS), then assessed the effect of the CPS on cartilage regeneration in a rabbit ear. The cell viability and functionality of chondrocytes were significantly higher in CPS than in cell-seeded scaffold (CSS) and cell-seeded hybrid scaffold (CSHS) in vitro. CPS was then implanted into a rabbit ear that had an 8 mm-diameter cartilage defect; at 3 months after implantation the CPS had fostered complete cartilage regeneration whereas CSS and autologs cartilage (AC) fostered only incomplete healing. This result demonstrates that cell printing technology can provide an appropriate environment in which encapsulated chondrocytes can survive and differentiate into cartilage tissue in vivo. Moreover, the effects of CPS on cartilage regeneration were even better than those of AC. Therefore, we confirmed the feasibility of CPS as an alternative to AC for auricular reconstruction. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1016-1028, 2017.
Collapse
Affiliation(s)
- Ju Young Park
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Yeong-Jin Choi
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Jin-Hyung Shim
- Department of Mechanical Engineering, Korea Polytechnic University, Siheung, Korea
| | - Jeong Hun Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| |
Collapse
|
38
|
do Amaral RJ, Matsiko A, Tomazette MR, Rocha WK, Cordeiro-Spinetti E, Levingstone TJ, Farina M, O'Brien FJ, El-Cheikh MC, Balduino A. Platelet-rich plasma releasate differently stimulates cellular commitment toward the chondrogenic lineage according to concentration. J Tissue Eng 2015; 6:2041731415594127. [PMID: 26380066 PMCID: PMC4555349 DOI: 10.1177/2041731415594127] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/03/2015] [Indexed: 01/22/2023] Open
Abstract
Platelet-rich plasma has been used to treat articular cartilage defects, with the expectations of anabolic and anti-inflammatory effects. However, its role on cellular chondrogenic or fibrogenic commitment is still a controversy. Herein, the role of platelet-rich plasma releasate, the product obtained following platelet-rich plasma activation, on cellular commitment toward the chondrogenic lineage was evaluated in vitro. Human nasoseptal chondrogenic cells and human bone marrow mesenchymal stromal cells were used as cell types already committed to the chondrogenic lineage and undifferentiated cells, respectively, as different concentrations of platelet-rich plasma releasate were tested in comparison to commonly used fetal bovine serum. Low concentration of platelet-rich plasma releasate (2.5%) presented similar effects on cellular growth compared to 10% fetal bovine serum, for both cell types. In a three-dimensional culture system, platelet-rich plasma releasate alone did not induce full nasoseptal chondrogenic cells cartilage-like pellet formation. Nonetheless, platelet-rich plasma releasate played a significant role on cell commitment as high-passage nasoseptal chondrogenic cells only originated cartilage-like pellets when expanded in the presence of platelet-rich plasma releasate rather than fetal bovine serum. Histological analyses and measurements of pellet area demonstrated that even low concentrations of platelet-rich plasma releasate were enough to prevent nasoseptal chondrogenic cells from losing their chondrogenic potential due to in vitro expansion thereby promoting their recommitment. Low concentration of platelet-rich plasma releasate supplemented in chondrogenic medium also increased the chondrogenic potential of mesenchymal stromal cells seeded on collagen-hyaluronic acid scaffolds, as observed by an increase in chondrogenic-related gene expression, sulfated glycosaminoglycan production, and compressive modulus following in vitro culture. On the contrary, higher concentration of platelet-rich plasma releasate (10%) hampered some of these features. In conclusion, platelet-rich plasma releasate was able to prevent cellular chondrogenic capacity loss, inducing regain of their phenotype, and modulate cell commitment. Our data support the hypothesis of platelet-rich plasma chondrogenic potential, allowing fetal bovine serum substitution for platelet-rich plasma releasate at specific concentrations in culture medium when chondrogenic commitment is desired on specific cell types and moments of culture.
Collapse
Affiliation(s)
- Ronaldo Jfc do Amaral
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brasil ; Excellion Serviços Biomédicos, Amil/UnitedHealth Group, Petrópolis, Brasil
| | - Amos Matsiko
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland ; Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin, Ireland ; Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Marcel Rp Tomazette
- Laboratório de Biologia e Tecnologia Celular, Universidade Veiga de Almeida, Rio de Janeiro, Brasil
| | - Wanessa Kr Rocha
- Instituto Estadual de Hematologia Arthur de Siqueira Cavalcanti, Rio de Janeiro, Brasil
| | - Eric Cordeiro-Spinetti
- Laboratório de Biologia e Tecnologia Celular, Universidade Veiga de Almeida, Rio de Janeiro, Brasil
| | - Tanya J Levingstone
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland ; Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin, Ireland ; Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Marcos Farina
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brasil
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland ; Trinity Centre for Bioengineering, Trinity College Dublin (TCD), Dublin, Ireland ; Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI & TCD, Dublin, Ireland
| | - Marcia C El-Cheikh
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brasil
| | - Alex Balduino
- Excellion Serviços Biomédicos, Amil/UnitedHealth Group, Petrópolis, Brasil ; Laboratório de Biologia e Tecnologia Celular, Universidade Veiga de Almeida, Rio de Janeiro, Brasil
| |
Collapse
|
39
|
Yoon HJ, Kim SB, Somaiya D, Noh MJ, Choi KB, Lim CL, Lee HY, Lee YJ, Yi Y, Lee KH. Type II collagen and glycosaminoglycan expression induction in primary human chondrocyte by TGF-β1. BMC Musculoskelet Disord 2015; 16:141. [PMID: 26059549 PMCID: PMC4460646 DOI: 10.1186/s12891-015-0599-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 05/27/2015] [Indexed: 12/13/2022] Open
Abstract
Background A localized non-surgical delivery of allogeneic human chondrocytes (hChonJ) with irradiated genetically modified chondrocytes (hChonJb#7) expressing transforming growth factor-β1 (TGF-β1) showed efficacy in regenerating cartilage tissue in our pre-clinical studies and human Phase I and II clinical trials. These previous observations led us to investigate the molecular mechanisms of the cartilage regeneration. Methods Genetically modified TGF-β1preprotein was evaluated by monitoring cell proliferation inhibition activity. The effect of modified TGF-β1 on chondrocytes was evaluated based on the type II collagen mRNA levels and the amount of glycosaminoclycan (GAG) formed around chondrocytes, which are indicative markers of redifferentiated chondrocytes. Among the cartilage matrix components produced by hChonJb#7 cells, type II collagen and proteoglycan, in addition to TGF-β1, were also tested to see if they could induce hChonJ redifferentiation. The ability of chondrocytes to attach to artificially induced defects in rabbit cartilage was tested using fluorescent markers. Results Throughout these experiments, the TGF-β1 produced from hChonJb#7 was shown to be equally as active as the recombinant human TGF-β1. Type II collagen and GAG production were induced in hChonJ cells by TGF-β1 secreted from the irradiated hChonJb#7 cells when the cells were co-cultured in micro-masses. Both hChonJ and hChonJb#7 cells could attach efficiently to the defect area in the rabbit cartilage. Conclusions This study suggests that the mixture (TG-C) of allogeneic human chondrocytes (hChonJ) and irradiated genetically modified human chondrocytes expressing TGF-β1 (hChonJb#7) attach to the damaged cartilage area to produce type II collagen-GAG matrices by providing a continuous supply of active TGF-β1.
Collapse
Affiliation(s)
- Hyun Joo Yoon
- TissueGene Inc., 9605 Medical Center Dr. Suite 200, Rockville, MD, 20850, USA.
| | - Suk Bum Kim
- Department of Rehabilitation and Personal training, Konyang University, 158, Gwanjeodong-ro, Daejeon, Seo-gu, Korea.
| | - Dhara Somaiya
- TissueGene Inc., 9605 Medical Center Dr. Suite 200, Rockville, MD, 20850, USA.
| | - Moon Jong Noh
- TissueGene Inc., 9605 Medical Center Dr. Suite 200, Rockville, MD, 20850, USA.
| | - Kyoung-Baek Choi
- Kolon Life Science, 13 Kolon-ro, Gwacheon-si, Gyeonggi-do, Korea.
| | - Chae-Lyul Lim
- Kolon Life Science, 13 Kolon-ro, Gwacheon-si, Gyeonggi-do, Korea.
| | - Hyeon-Youl Lee
- Kolon Life Science, 13 Kolon-ro, Gwacheon-si, Gyeonggi-do, Korea.
| | - Yeon-Ju Lee
- Kolon Life Science, 13 Kolon-ro, Gwacheon-si, Gyeonggi-do, Korea.
| | - Youngsuk Yi
- TissueGene Inc., 9605 Medical Center Dr. Suite 200, Rockville, MD, 20850, USA.
| | - Kwan Hee Lee
- TissueGene Inc., 9605 Medical Center Dr. Suite 200, Rockville, MD, 20850, USA.
| |
Collapse
|
40
|
Wardale J, Mullen L, Howard D, Ghose S, Rushton N. An ex vivo model using human osteoarthritic cartilage demonstrates the release of bioactive insulin-like growth factor-1 from a collagen-glycosaminoglycan scaffold. Cell Biochem Funct 2015; 33:277-84. [PMID: 26059711 PMCID: PMC4528234 DOI: 10.1002/cbf.3112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/01/2015] [Accepted: 04/07/2015] [Indexed: 01/04/2023]
Abstract
Biomimetic scaffolds hold great promise for therapeutic repair of cartilage, but although most scaffolds are tested with cells in vitro, there are very few ex vivo models (EVMs) where adult cartilage and scaffolds are co-cultured to optimize their interaction prior to in vivo studies. This study describes a simple, non-compressive method that is applicable to mammalian or human cartilage and provides a reasonable throughput of samples. Rings of full-depth articular cartilage slices were derived from human donors undergoing knee replacement for osteoarthritis and a 3 mm core of a collagen/glycosaminoglycan biomimetic scaffold (Tigenix, UK) inserted to create the EVM. Adult osteoarthritis chondrocytes were seeded into the scaffold and cultures maintained for up to 30 days. Ex vivo models were stable throughout experiments, and cells remained viable. Chondrocytes seeded into the EVM attached throughout the scaffold and in contact with the cartilage explants. Cell migration and deposition of extracellular matrix proteins in the scaffold was enhanced by growth factors particularly if the scaffold was preloaded with growth factors. This study demonstrates that the EVM represents a suitable model that has potential for testing a range of therapeutic parameters such as numbers/types of cell, growth factors or therapeutic drugs before progressing to costly pre-clinical trials. © 2015 The Authors. Cell Biochemistry and Function Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- J Wardale
- Orthopaedic Research Unit, University of Cambridge, Cambridge, UK
| | | | - D Howard
- Orthopaedic Research Unit, University of Cambridge, Cambridge, UK
| | | | - N Rushton
- Orthopaedic Research Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
41
|
Biologic enhancement of cartilage repair: the role of platelet-rich plasma and other commercially available growth factors. Arthroscopy 2015; 31:777-83. [PMID: 25670338 DOI: 10.1016/j.arthro.2014.11.031] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 11/17/2014] [Accepted: 11/17/2014] [Indexed: 02/02/2023]
Abstract
In part, people's quality of life depends on the "health" of their cartilage because its damage or deterioration causes pain that limits mobility and reduces autonomy. Predisposing genetic factors and modern-life environmental factors, such as diet, excessive physical exercise, or the absence of any physical exercise, in addition to injuries that can occur, all contribute to the onset and development of chronic degenerative diseases such as osteoarthritis. Regenerative medicine focuses on the repair, replacement, or regeneration of cells, tissues, or organs to restore impaired function from any cause, including congenital defects, disease, and trauma.
Collapse
|
42
|
Son MS, Levenston ME. Quantitative tracking of passage and 3D culture effects on chondrocyte and fibrochondrocyte gene expression. J Tissue Eng Regen Med 2015; 11:1185-1194. [PMID: 25824488 DOI: 10.1002/term.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 02/12/2015] [Accepted: 02/18/2015] [Indexed: 11/11/2022]
Abstract
Success in cartilage and fibrocartilage tissue engineering relies heavily on using an appropriate cell source. Many different cell sources have been identified, including primary and stem cells, along with experimental strategies to obtain the required number of cells or to induce chondrogenesis. However, no definitive method exists to quantitatively evaluate the similarity of the resulting cell phenotypes to those of the native cells between candidate strategies. In this study, we develop an integrative approach to enable such evaluations by deriving, from gene expression profiles, two quantitative metrics representing the nearest location within the range of native cell phenotypes and the deviation from it. As an example application to evaluating potential cell sources for cartilage or meniscus tissue engineering, we examine phenotypic changes of juvenile and adult articular chondrocytes and fibrochondrocytes across multiple passages and subsequent 3D culture. A substantial change was observed in cell phenotype due to the isolation process itself, followed by a clear progression toward the outer meniscal cell phenotype with passage. The new metrics also indicated that 3D culture moderately reduced the passage-induced deviation from the native meniscal phenotypes for juvenile chondrocytes and adult fibrochondrocytes, which was not obvious through examination of individual gene expressions. However, brief 3D culture alone did not move any of the cells towards an inner meniscal phenotype, the most relevant target for meniscal tissue engineering. This integrative approach of examining and combining multiple gene expressions can be used to evaluate various other tissue-engineering strategies to direct cells toward the desired phenotype. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Min-Sun Son
- Department of Bioengineering, Stanford University, CA, USA
| | - Marc E Levenston
- Department of Mechanical Engineering, Stanford University, CA, USA
| |
Collapse
|
43
|
Bleuel J, Zaucke F, Brüggemann GP, Niehoff A. Effects of cyclic tensile strain on chondrocyte metabolism: a systematic review. PLoS One 2015; 10:e0119816. [PMID: 25822615 PMCID: PMC4379081 DOI: 10.1371/journal.pone.0119816] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 01/16/2015] [Indexed: 12/25/2022] Open
Abstract
Chondrocytes reorganize the extracellular matrix of articular cartilage in response to externally applied loads. Thereby, different loading characteristics lead to different biological responses. Despite of active research in this area, it is still unclear which parts of the extracellular matrix adapt in what ways, and how specific loading characteristics affect matrix changes. This review focuses on the influence of cyclic tensile strain on chondrocyte metabolism in vitro. It also aimed to identify anabolic or catabolic chondrocyte responses to different loading protocols. The key findings show that loading cells up to 3% strain, 0.17 Hz, and 2 h, resulted in weak or no biological responses. Loading between 3–10% strain, 0.17–0.5 Hz, and 2–12 h led to anabolic responses; and above 10% strain, 0.5 Hz, and 12 h catabolic events predominated. However, this review also discusses that various other factors are involved in the remodeling of the extracellular matrix in response to loading, and that parameters like an inflammatory environment might influence the biological response.
Collapse
Affiliation(s)
- Judith Bleuel
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Köln, Germany
- * E-mail:
| | - Frank Zaucke
- Center for Biochemistry, Medical Faculty, University of Cologne, Köln, Germany
- Cologne Center for Musculoskeletal Biomechanics, Medical Faculty, University of Cologne, Köln, Germany
| | - Gert-Peter Brüggemann
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Köln, Germany
- Cologne Center for Musculoskeletal Biomechanics, Medical Faculty, University of Cologne, Köln, Germany
| | - Anja Niehoff
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Köln, Germany
- Cologne Center for Musculoskeletal Biomechanics, Medical Faculty, University of Cologne, Köln, Germany
| |
Collapse
|
44
|
Das R, Timur U, Edip S, Haak E, Wruck C, Weinans H, Jahr H. TGF-β2 is involved in the preservation of the chondrocyte phenotype under hypoxic conditions. Ann Anat 2015; 198:1-10. [DOI: 10.1016/j.aanat.2014.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 11/02/2014] [Accepted: 11/14/2014] [Indexed: 12/13/2022]
|
45
|
Ma X, Liu Y, Wang Q, Chen Y, Liu M, Li X, Xiang R, Wei Y, Duan Y, Han J. Tamoxifen induces the development of hernia in mice by activating MMP-2 and MMP-13 expression. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1038-48. [PMID: 25703139 DOI: 10.1016/j.bbadis.2015.02.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 02/01/2015] [Accepted: 02/12/2015] [Indexed: 02/05/2023]
Abstract
Hernia is a disease with defects in collagen synthesis/metabolism. However, the underlying mechanisms for hernia formation have not been fully defined. Tamoxifen is a selective estrogen receptor modulator and used for patients with breast cancer. Tamoxifen also has pleiotropic and side effects. Herein, we report that tamoxifen treatment resulted in an appearance of a large bulge in the low abdomen between the hind legs in male but not in female mice. The autopsy demonstrated that the low abdominal wall was broken and a large amount of intestine herniated out of the abdominal cavity. Histological analysis indicated that tamoxifen caused structural abnormalities in the low abdominal wall which were associated with decreased type II collagen content. Furthermore, we determined increased matrix metalloproteinase-2 (MMP-2) and MMP-13 expression in the tissue. In vitro, tamoxifen induced MMP-2 and MMP-13 expression in fibroblasts. The promoter activity analysis and ChIP assay demonstrate that induction of MMP-13 expression was associated with activation of JNK-AP-1 and ERK1/2 signaling pathways while induction of MMP-2 expression was related to activation of the ERK1/2 signaling pathway. Taken together, our study establishes a novel murine hernia model, defines a severe side effect of tamoxifen, and suggests a caution to male patients receiving tamoxifen treatment.
Collapse
Affiliation(s)
- Xingzhe Ma
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China; College of Life Sciences, Nankai University, Tianjin, China
| | - Ying Liu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Qixue Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China; College of Life Sciences, Nankai University, Tianjin, China
| | - Yuanli Chen
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China; College of Medicine, Nankai University, Tianjin, China
| | - Mengyang Liu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoju Li
- College of Life Sciences, Nankai University, Tianjin, China
| | - Rong Xiang
- College of Medicine, Nankai University, Tianjin, China; Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuquan Wei
- Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yajun Duan
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China; College of Life Sciences, Nankai University, Tianjin, China.
| | - Jihong Han
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China; College of Life Sciences, Nankai University, Tianjin, China; Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
46
|
Abstract
Chondroprogenitor cells are a subpopulation of multipotent progenitors that are primed for chondrogenesis. They are believed to have the biological repertoire to be ideal for cell-based cartilage therapy. In addition to summarizing recent advances in chondroprogenitor cell characterization, this review discusses the projected pros and cons of utilizing chondroprogenitors in regenerative medicine and compares them with that of pre-existing methods, including autologous chondrocyte implantation (ACI) and the utilization of bone marrow derived mesenchymal stem cells (MSCs) for the purpose of cartilage tissue repair.
Collapse
Affiliation(s)
- Chathuraka T Jayasuriya
- Department of Orthopedics, Warren Alpert Medical School of Brown University , Providence, RI , USA
| | | |
Collapse
|
47
|
Catherine B, Girard N, Lhuissier E, Bazille C, Boumediene K. Regulation and Role of TGFβ Signaling Pathway in Aging and Osteoarthritis Joints. Aging Dis 2014; 5:394-405. [PMID: 25489490 DOI: 10.14336/ad.2014.0500394] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/04/2013] [Accepted: 12/04/2013] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor beta (TGFβ) is a major signalling pathway in joints. This superfamilly is involved in numerous cellular processes in cartilage. Usually, they are considered to favor chondrocyte differentiation and cartilage repair. However, other studies show also deleterious effects of TGFβ which may induce hypertrophy. This may be explained at least in part by alteration of TGFβ signaling pathways in aging chondrocytes. This review focuses on the functions of TGFβ in joints and the regulation of its signaling mediators (receptors, Smads) during aging and osteoarthritis.
Collapse
Affiliation(s)
| | - Nicolas Girard
- Normandie Univ, France ; UNICAEN, EA4652 MILPAT, Caen, France
| | - Eva Lhuissier
- Normandie Univ, France ; UNICAEN, EA4652 MILPAT, Caen, France
| | - Celine Bazille
- Normandie Univ, France ; UNICAEN, EA4652 MILPAT, Caen, France ; Service d'Anatomie Pathologique, CHU, Caen, France
| | | |
Collapse
|
48
|
Maneix L, Servent A, Porée B, Ollitrault D, Branly T, Bigot N, Boujrad N, Flouriot G, Demoor M, Boumediene K, Moslemi S, Galéra P. Up-regulation of type II collagen gene by 17β-estradiol in articular chondrocytes involves Sp1/3, Sox-9, and estrogen receptor α. J Mol Med (Berl) 2014; 92:1179-200. [PMID: 25081415 DOI: 10.1007/s00109-014-1195-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 07/11/2014] [Accepted: 07/22/2014] [Indexed: 01/01/2023]
Abstract
UNLABELLED The existence of a link between estrogen deprivation and osteoarthritis (OA) in postmenopausal women suggests that 17β-estradiol (17β-E2) may be a modulator of cartilage homeostasis. Here, we demonstrate that 17β-E2 stimulates, via its receptor human estrogen receptor α 66 (hERα66), type II collagen expression in differentiated and dedifferentiated (reflecting the OA phenotype) articular chondrocytes. Transactivation of type II collagen gene (COL2A1) by ligand-independent transactivation domain (AF-1) of hERα66 was mediated by "GC" binding sites of the -266/-63-bp promoter, through physical interactions between ERα, Sp1/Sp3, Sox9, and p300, as demonstrated in chromatin immunoprecipitation (ChIP) and Re-Chromatin Immuno-Precipitation (Re-ChIP) assays in primary and dedifferentiated cells. 17β-E2 and hERα66 increased the DNA-binding activities of Sp1/Sp3 and Sox-9 to both COL2A1 promoter and enhancer regions. Besides, Sp1, Sp3, and Sox-9 small interfering RNAs (siRNAs) prevented hERα66-induced transactivation of COL2A1, suggesting that these factors and their respective cis-regions are required for hERα66-mediated COL2A1 up-regulation. Our results highlight the genomic pathway by which 17β-E2 and hERα66 modulate Sp1/Sp3 heteromer binding activity and simultaneously participate in the recruitment of the essential factors Sox-9 and p300 involved respectively in the chondrocyte-differentiated status and COL2A1 transcriptional activation. These novel findings could therefore be attractive for tissue engineering of cartilage in OA, by the fact that 17β-E2 could promote chondrocyte redifferentiation. KEY MESSAGES 17β-E2 up-regulates type II collagen gene expression in articular chondrocytes. An ERα66/Sp1/Sp3/Sox-9/p300 protein complex mediates this stimulatory effect. This heteromeric complex interacts and binds to Col2a1 promoter and enhancer in vivo. Our findings highlight a new regulatory mechanism for 17β-E2 action in chondrocytes. 17β-E2 might be an attractive candidate for cartilage engineering applications.
Collapse
Affiliation(s)
- Laure Maneix
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), EA4652, 14032, Caen, France
| | - Aurélie Servent
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), EA4652, 14032, Caen, France
| | - Benoît Porée
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), EA4652, 14032, Caen, France
| | - David Ollitrault
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), EA4652, 14032, Caen, France
| | - Thomas Branly
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), EA4652, 14032, Caen, France
| | - Nicolas Bigot
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), EA4652, 14032, Caen, France
| | - Noureddine Boujrad
- Laboratoire Endocrinologie Moléculaire de la Reproduction, Equipe Récepteurs des Oestrogènes et Destinée Cellulaire, CNRS UMR 6026, Université de Rennes I, 35042, Rennes, France
| | - Gilles Flouriot
- Laboratoire Endocrinologie Moléculaire de la Reproduction, Equipe Récepteurs des Oestrogènes et Destinée Cellulaire, CNRS UMR 6026, Université de Rennes I, 35042, Rennes, France
| | - Magali Demoor
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), EA4652, 14032, Caen, France
| | - Karim Boumediene
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), EA4652, 14032, Caen, France
| | - Safa Moslemi
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), EA4652, 14032, Caen, France
| | - Philippe Galéra
- Normandy University, Caen, France; UNICAEN, Laboratoire Microenvironnement Cellulaire et Pathologies (MILPAT), EA4652, 14032, Caen, France.
| |
Collapse
|
49
|
Evaluation of nanofiber-based polyglycolic acid scaffolds for improved chondrocyte retention and in vivo bioengineered cartilage regeneration. Plast Reconstr Surg 2014; 133:805e-813e. [PMID: 24867739 DOI: 10.1097/prs.0000000000000176] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND In conventional studies on the regeneration of auricle-shaped cartilage in autogenous models using large animals, there were problems with the cartilage regeneration induction capacity and long-term retention of geometric shape. In this study, the authors sought to improve on outcome in these regards: a nonwoven fabric of polyglycolic acid was developed through nanotechnology, and the effect of nanofiber diameter on in vitro cell-seeding efficiency and the in vivo response after implantation in an autogenous large-animal model were evaluated. METHODS Canine chondrocytes were isolated and seeded onto polyglycolic acid fabric ranging from 0.5 to 20 μm in average diameter. Cell seeding efficiency was highest for mid-range polyglycolic acid fibers (average diameter, 0.8, 3.0, and 7.0 μm). Flat and auricle-shaped scaffolds were constructed using polypropylene structural support, sandwiching a nonwoven polyglycolic acid fabric that contained autogenous chondrocytes together with basic fibroblast growth factor-laden particles and an exterior fibrin sealant. Scaffolds were then implanted autogenously and evaluated at 5 and 20 weeks. RESULTS Biomechanical strength was optimal for polyglycolic acid fiber diameters of 0.8 to 3.0 μm. Optimal cell maintenance and neocartilage response were seen with polyglycolic acid fiber diameters in the same mid-range for nanofiber constructs. CONCLUSION These findings demonstrate the potential for nanoscale modulation of auricle-shaped cartilage regeneration in a large-animal model.
Collapse
|
50
|
Plencner M, East B, Tonar Z, Otáhal M, Prosecká E, Rampichová M, Krejčí T, Litvinec A, Buzgo M, Míčková A, Nečas A, Hoch J, Amler E. Abdominal closure reinforcement by using polypropylene mesh functionalized with poly-ε-caprolactone nanofibers and growth factors for prevention of incisional hernia formation. Int J Nanomedicine 2014; 9:3263-77. [PMID: 25031534 PMCID: PMC4096451 DOI: 10.2147/ijn.s63095] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Incisional hernia affects up to 20% of patients after abdominal surgery. Unlike other types of hernia, its prognosis is poor, and patients suffer from recurrence within 10 years of the operation. Currently used hernia-repair meshes do not guarantee success, but only extend the recurrence-free period by about 5 years. Most of them are nonresorbable, and these implants can lead to many complications that are in some cases life-threatening. Electrospun nanofibers of various polymers have been used as tissue scaffolds and have been explored extensively in the last decade, due to their low cost and good biocompatibility. Their architecture mimics the natural extracellular matrix. We tested a biodegradable polyester poly-ε-caprolactone in the form of nanofibers as a scaffold for fascia healing in an abdominal closure-reinforcement model for prevention of incisional hernia formation. Both in vitro tests and an experiment on a rabbit model showed promising results.
Collapse
Affiliation(s)
- Martin Plencner
- Institute of Biophysics, Second Faculty of Medicine, Charles University in Prague, Prague, Czech Republic ; Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Barbora East
- Department of Surgery, Second Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Zbyněk Tonar
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| | - Martin Otáhal
- Department of Anatomy and Biomechanics, Faculty of Physical Education and Sport, Charles University in Prague, Prague, Czech Republic
| | - Eva Prosecká
- Institute of Biophysics, Second Faculty of Medicine, Charles University in Prague, Prague, Czech Republic ; Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Michala Rampichová
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic ; University Center for Energy Efficient Buildings, Czech Technical University in Prague, Buštěhrad, Czech Republic
| | - Tomáš Krejčí
- Department of Surgery, Second Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Andrej Litvinec
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic ; Department of Breeding and Zoohygiene of Laboratory Animals, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Matej Buzgo
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic ; University Center for Energy Efficient Buildings, Czech Technical University in Prague, Buštěhrad, Czech Republic
| | - Andrea Míčková
- Institute of Biophysics, Second Faculty of Medicine, Charles University in Prague, Prague, Czech Republic ; Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic ; University Center for Energy Efficient Buildings, Czech Technical University in Prague, Buštěhrad, Czech Republic
| | - Alois Nečas
- Department of Surgery and Orthopedics, Small Animal Clinic, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Science Brno, Central European Institute of Technology, Brno, Czech Republic
| | - Jiří Hoch
- Department of Surgery, Second Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Evžen Amler
- Institute of Biophysics, Second Faculty of Medicine, Charles University in Prague, Prague, Czech Republic ; Laboratory of Tissue Engineering, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic ; Faculty of Biomedical Engineering, Czech Technical University in Prague, Kladno, Czech Republic
| |
Collapse
|