1
|
van Mourik M, Abinzano F, Ito K. The Regulation of Pericellular Matrix Synthesis During Articular Cartilage Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40402857 DOI: 10.1089/ten.teb.2024.0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Articular cartilage, vital to the health and functioning of joints, remains challenging to regenerate. The pericellular matrix (PCM) is critical for transducing biophysical stimuli to the articular chondrocytes (ACs) that it envelops. Given the mechanobiological sensitivity of ACs, it is pivotal in maintaining the chondrogenic phenotype and the production of extracellular matrix (ECM) during articular cartilage tissue engineering. While the maintenance of the native PCM significantly improves the quality of neocartilage, current isolation methods are limited. A solution to this challenge is facilitating ACs to regenerate their PCM. However, the regulation of PCM synthesis remains poorly understood, hindering the development of effective tissue engineering strategies. This narrative review aims to provide a comprehensive analysis of the complex interplay between extracellular cues and intracellular pathways regulating PCM synthesis during articular cartilage tissue engineering. Our analysis reveals that mechanical cues, such as material stiffness and mechanical stimulation, are the primary regulators of PCM synthesis. Additionally, the use of scaffold-free techniques potentially affects the structuring of newly created PCM. Tuning these stimuli can significantly impact the quality of the formed PCM, ultimately influencing neocartilage quality. Furthermore, we highlight intracellular mechanisms involved in the transduction of these extracellular cues, including actin polymerization, yes-associated protein and transcriptional coactivator with PDZ-binding motif localization, and transforming growth factor beta-induced Smad signaling. Although the current literature suggests the involvement of these signaling pathways in regulating the synthesis of PCM components, we found that studies investigating these processes in ACs are lacking. Elucidating the relationships between extracellular stimuli, intracellular signaling, and the expression of PCM components could provide a framework for designing new cartilage tissue engineering approaches that facilitate proper PCM synthesis. Ultimately, this can improve ECM quality and advance articular cartilage regeneration.
Collapse
Affiliation(s)
- Marloes van Mourik
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Florencia Abinzano
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
2
|
Su W, Nie Y, Zheng S, Yao Y. Recent Research on Chondrocyte Dedifferentiation and Insights for Regenerative Medicine. Biotechnol Bioeng 2025; 122:749-760. [PMID: 39716991 DOI: 10.1002/bit.28915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Chondrocytes maintain the balance of the extracellular matrix by synthesizing glycoproteins, collagen, proteoglycans and hyaluronic acid. Chondrocyte dedifferentiation refers to a process in which chondrocytes lose their mature differentiated phenotype and transform into a fibroblast-like morphology with fewer differentiated stages and inferior function under external stimulation. The important mechanism of homeostasis loss in osteoarthritis (OA) is a change in the chondrocyte phenotype. The dedifferentiation markers of chondrocytes are upregulated in OA, and the pathogenic factors related to OA have also been shown to enhance chondrocyte dedifferentiation. In this review, we compile recent studies on chondrocyte dedifferentiation, with an emphasis on potential markers and the underlying mechanisms of dedifferentiation, as well as the current research progress in inhibiting dedifferentiation or achieving redifferentiation. A deep understanding of chondrocyte dedifferentiation would not only support the pathogenesis of OA theoretically but also provide insightful ideas for regenerative medicine to manipulate the functional phenotype of cells.
Collapse
Affiliation(s)
- Weixian Su
- Department of Joint Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou Medical University, Guangzhou, China
| | - Yupeng Nie
- Department of Joint Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou Medical University, Guangzhou, China
| | - Shicong Zheng
- Department of Joint Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongchang Yao
- Department of Joint Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Song Z, Freiin Grote V, Sahm F, Zimmermann J, Lutter C, Jonitz‐Heincke A, Bader R. Exploring the role of directly coupled alternating electric fields on chondrocyte morphology and redifferentiation capacity with a focus on sex differences. J Exp Orthop 2025; 12:e70261. [PMID: 40330809 PMCID: PMC12053005 DOI: 10.1002/jeo2.70261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/27/2025] [Accepted: 02/11/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose In cell-based therapies addressing articular cartilage lesions, a central challenge is to avoid the formation of fibrous cartilage resulting from dedifferentiation processes. Electrical stimulation emerges as a promising approach for promoting chondrocytic redifferentiation. This study investigated the effects of varying electric fields on morphological changes and the redifferentiation capacity of human chondrocytes with regard to alterations in sex. Methods Chondrocytes, isolated from the articular cartilage of male and female patients undergoing total knee replacement, were exposed to alternating electric fields of varying strengths ranging from 0.8 to 1.2, 15 to 20 and 100 to 140 V/m. Afterwards, cell morphology and viability, as well as the deposition of collagen (Col) 1 and 2, were evaluated. Results Following electrical stimulation, in particular at 15-20 V/m, an increase in the Col2/Col1 ratio and an elevated proportion of rounded, chondrocyte-like cell morphology were observed, indicating a promoting effect on the redifferentiation of chondrocytes. Comparative analysis between both sexes revealed that chondrocytes from female donors exhibit higher Col1 synthesis rates, a decreased Col2/Col1 ratio, and a higher proportion of elongated, fibroblast-like cells compared to chondrocytes derived from male donors. Conclusion Our in vitro study suggests that chondrocytes from male donors are more prone to re-differentiate after electrical stimulation. Level of Evidence N/A.
Collapse
Affiliation(s)
- Zezhong Song
- Department of Orthopaedics, Biomechanics and Implant Technology Research LaboratoryRostock University Medical CenterRostockGermany
| | - Vivica Freiin Grote
- Department of Orthopaedics, Biomechanics and Implant Technology Research LaboratoryRostock University Medical CenterRostockGermany
| | - Franziska Sahm
- Department of Orthopaedics, Biomechanics and Implant Technology Research LaboratoryRostock University Medical CenterRostockGermany
| | - Julius Zimmermann
- Institute of General Electrical EngineeringUniversity of RostockRostockGermany
| | - Christoph Lutter
- Department of Orthopaedics, Biomechanics and Implant Technology Research LaboratoryRostock University Medical CenterRostockGermany
| | - Anika Jonitz‐Heincke
- Department of Orthopaedics, Biomechanics and Implant Technology Research LaboratoryRostock University Medical CenterRostockGermany
| | - Rainer Bader
- Department of Orthopaedics, Biomechanics and Implant Technology Research LaboratoryRostock University Medical CenterRostockGermany
- Department Life, Light MatterUniversity of RostockRostockGermany
| |
Collapse
|
4
|
Liu Y, Chen X, Tan X, Huang Y, Zhang W, Wang Z, Yang L, Wang Y, Li Z, Zhang X. Double network hydrogels encapsulating genetically modified dedifferentiated chondrocytes for auricular cartilage regeneration. J Mater Chem B 2025; 13:1823-1844. [PMID: 39745373 DOI: 10.1039/d4tb02352h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Microtia profoundly affects patients' appearance and psychological well-being. Tissue engineering ear cartilage scaffolds have emerged as the most promising solution for ear reconstruction. However, constructing tissue engineering ear cartilage scaffolds requires multiple passaging of chondrocytes, resulting in their dedifferentiation and loss of their special phenotypes and functions. To tackle these issues, here we employ guanidinobenzoic acid (GBA) modified generation 5 polyamidoamine (PAMAM) dendrimers (PG) as a Runx1 plasmid carrier to construct PG/pRunx1 polyplex nanoparticles. The PG/pRunx1 polyplexes are transfected into human auricular chondrocytes, significantly mitigating chondrocyte dedifferentiation and enhancing cartilage regeneration during the in vitro culture. Furthermore, we develop highly porous double-network hydrogels based on methacrylate-functionalized and oxidized chondroitin sulfate and carbohydrazide-modified gelatin and the hydrogels possessed both dynamic adaptability and mechanical support characteristics by reversible dynamic covalent crosslinking and static covalent crosslinking, serving as an ideal scaffold for tissue engineering. Consequently, chondrocytes treated with PG/pRunx1 polyplex nanoparticles are incorporated into the hydrogels to construct tissue-engineered auricular cartilage scaffolds. After subcutaneous implantation in nude mice, the scaffolds containing chondrocytes treated with PG/pRunx1 nanoparticles showed more mature cartilaginous tissue, characterized by prominent ECM deposition and enhanced chondrogenesis. Therefore, this research provides a novel strategy for the development of tissue-engineered auricular cartilage scaffolds.
Collapse
Affiliation(s)
- Yang Liu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaoting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xueqin Tan
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yeqian Huang
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Wen Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | - Zhicun Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | - Zhengyong Li
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
5
|
Gavrilov NS, Ignatyeva NV, Medvedeva EV, Timashev PS. Articular cartilage tissue engineering using genetically modified induced pluripotent stem cell lines. GENES & CELLS 2024; 19:404-424. [DOI: 10.17816/gc633492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2025]
Abstract
Mature hyaline cartilage has a low regenerative potential and its repair remains a complex clinical and research issue. Articular cartilage injuries often contribute to the development of osteoarthritis, resulting in loss of joint function and patient disability. Surgical techniques for repairing articular surfaces, such as mosaic chondroplasty and microfracture, which are designed for small defects, cannot be used for degenerative and dystrophic cartilage lesions. Cell therapy using chondrocytes differentiated from induced pluripotent stem cells (iPSCs) is a promising approach to reconstruct articular cartilage tissue. iPSCs have high proliferative activity, which allows the harvesting of autologous cells in quantities necessary to repair a joint defect. CRISPR-Cas genome editing technology, based on the bacterial adaptive immune system, enables the genetic modification of iPSCs to obtain progenitor cells with specific characteristics and properties.
This review describes specific research papers on the combined use of iPSC and CRISPR-Cas technologies for the evaluation of cartilage regenerative medicine. Papers were evaluated for the last twelve years since CRISPR-Cas technology was introduced to the global community. CRISPR-Cas is currently being used to address therapeutic issues in articular cartilage regeneration by increasing the efficiency of chondrogenic differentiation of iPSC lines and harvesting a more homogeneous population of chondroprogenitor cells. Another approach is to remove the pro-inflammatory cytokine receptor sequence to produce inflammation-resistant cartilage. Finally, knocking out genes for components of the major histocompatibility complex allows harvesting chondrocytes that are invisible to the recipient's immune system. This kind of research contributes to personalized healthcare and can improve the quality of life of the world's population in the long term.
Collapse
|
6
|
Roseti L, Cavallo C, Desando G, D’Alessandro M, Grigolo B. Forty Years of the Use of Cells for Cartilage Regeneration: The Research Side. Pharmaceutics 2024; 16:1622. [PMID: 39771600 PMCID: PMC11677864 DOI: 10.3390/pharmaceutics16121622] [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: 11/18/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The treatment of articular cartilage damage has always represented a problem of considerable practical interest for orthopedics. Over the years, many surgical techniques have been proposed to induce the growth of repairing tissue and limit degeneration. In 1994, the turning point occurred: implanted autologous cells paved the way for a new treatment option based more on regeneration than repair. Objectives: This review aims to outline biological and clinical advances, from the use of mature adult chondrocytes to cell-derived products, going through progenitor cells derived from bone marrow or adipose tissue and their concentrates for articular cartilage repair. Moreover, it highlights the relevance of gene therapy as a valuable tool for successfully implementing current regenerative treatments, and overcoming the limitations of the local delivery of growth factors. Conclusions: Finally, this review concludes with an outlook on the importance of understanding the role and mechanisms of action of the different cell compounds with a view to implementing personalized treatments.
Collapse
Affiliation(s)
| | - Carola Cavallo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.); (G.D.); (M.D.); (B.G.)
| | | | | | | |
Collapse
|
7
|
Diab RG, Deeb G, Roda R, Karam M, Faraj M, Harajli M, Damiati LA, Mhanna R. Maintaining the Cartilage Phenotype of Late-Passage Chondrocytes Using Salidroside, TGF-β, and Sulfated Alginate for Cartilage Tissue Engineering Applications. Int J Mol Sci 2024; 25:13623. [PMID: 39769386 PMCID: PMC11727720 DOI: 10.3390/ijms252413623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/07/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
The limited self-repair capacity of cartilage due to its avascular and aneural nature leads to minimal regenerative ability. Autologous chondrocyte transplantation (ACT) is a popular treatment for cartilage defects but faces challenges due to chondrocyte dedifferentiation in later passages, which results in undesirable fibroblastic phenotypes. A promising treatment for cartilage injuries and diseases involves tissue engineering using cells (e.g., chondrocytes), scaffolds (e.g., Alginate Sulfate (AlgSulf)), and biochemical signals (e.g., Salidroside and TGF-β). This study focuses on investigating the effects of AlgSulf scaffolds with varying degrees of sulfation, Salidroside, and TGF-β on the proliferation, viability, and phenotype maintenance of chondrocytes. The findings demonstrate that AlgSulf films with a degree of sulfation (DS) = 2, treated with a combination of Salidroside and TGF-β, significantly enhanced chondrocyte proliferation (p < 0.001 and p < 0.0001 in P2 and P4, respectively), preserved round cell morphology, and maintained cartilage-specific gene expression (Col2, Aggrecans, and SOX9) while downregulating fibroblastic markers (Col1, MMP13, IL-1β, and IL-6). Our findings suggest the potential of this combination for enhancing cartilage regeneration in tissue engineering applications.
Collapse
Affiliation(s)
- Rita G. Diab
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - George Deeb
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - Rena Roda
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - Mia Karam
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - Marwa Faraj
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - Mohamad Harajli
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| | - Laila A. Damiati
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah 21959, Saudi Arabia
| | - Rami Mhanna
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (R.G.D.); (G.D.); (R.R.); (M.K.); (M.F.); (M.H.)
| |
Collapse
|
8
|
Pham TB, Sah RL, Masuda K, Watson D. Human Septal Cartilage Tissue Engineering: Current Methodologies and Future Directions. Bioengineering (Basel) 2024; 11:1123. [PMID: 39593783 PMCID: PMC11592323 DOI: 10.3390/bioengineering11111123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/28/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
Nasal septal cartilage tissue engineering is a promising and dynamic field with the potential to provide surgical options for patients with complex reconstruction needs and mitigate the risks incurred by other tissue sources. Developments in cell source selection, cell expansion, scaffold creation, and three-dimensional (3D) bioprinting have advanced the field in recent years. The usage of medicinal signaling cells and nasal chondroprogenitor cells can enhance chondrocyte proliferation, stimulate chondrocyte growth, and limit chondrocyte dedifferentiate. New scaffolds combined with recent innovations in 3D bioprinting have allowed for the creation of more durable and customizable constructs. Future developments may increase technical accessibility and manufacturability, and lower costs, to help incorporate these methods into pre-clinical studies and clinical applications of septal cartilage tissue engineering.
Collapse
Affiliation(s)
- Tammy B. Pham
- Department of Otolaryngology-Head and Neck Surgery, UC San Diego Health, La Jolla, CA 92093, USA;
| | - Robert L. Sah
- Shu Chien-Gene Lay Department of Bioengineering, UC San Diego Jacobs School of Engineering, La Jolla, CA 92093, USA;
| | - Koichi Masuda
- Department of Orthopedic Surgery, UC San Diego Health, La Jolla, CA 92093, USA;
| | - Deborah Watson
- Department of Otolaryngology-Head and Neck Surgery, UC San Diego Health, La Jolla, CA 92093, USA;
| |
Collapse
|
9
|
Rosochowicz MA, Lach MS, Richter M, Jagiełło I, Suchorska WM, Trzeciak T. The iPSC secretome is beneficial for in vitro propagation of primary osteoarthritic chondrocytes cell lines. Biochem Biophys Res Commun 2024; 730:150392. [PMID: 39003867 DOI: 10.1016/j.bbrc.2024.150392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND One of the obstacles to autologous chondrocyte implantation (ACI) is obtaining a large quantity of chondrocytes without depletion of their properties. The conditioned medium (CM) from different subpopulations of stem cells (mesenchymal stromal cells (MSC) or induced pluripotent stem cells (iPSC)) could be a gamechanger. MSCs' potential is related to the donor's health and age, which could be omitted when, as a source, iPSCs are used. There is a lack of data regarding their use in the chondrocyte culture expansion. Thus, we wanted to verify whether iPSC-CM could be beneficial for the cell culture of primary chondrocyte cells. METHODS We added the iPSC-CMs from GPCCi001-A and ND 41658*H cells to the culture of primary chondrocyte cell lines isolated from OA patients (n = 6) for other two passages. The composition of the CM was evaluated using Luminex technology. Then, we analysed the senescence, proliferation rate and using flow cytometry: viability, distribution of cell cycle phases, production of reactive oxygen species (ROS) and double-strand breaks. The cartilage-related markers were evaluated using Western blot and immunofluorescence. Additionally, a three-dimensional cell culture was used to determine the potential to form cartilage particles. RESULTS iPSC-CM increased proliferation and diminished cell ROS production and senescence. CM influenced the cartilage-related protein expression and promoted the growth of cartilage particles. The cell exposed to CM did not lose the ECM proteins, suggesting the chondroprotective effect for prolonged culture time. CONCLUSION Our preliminary results suggest a beneficial effect on maintaining chondrocyte biology during in vitro expansion.
Collapse
Affiliation(s)
- Monika A Rosochowicz
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland; Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland; Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland.
| | - Michał S Lach
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland; Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland
| | - Magdalena Richter
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland
| | - Inga Jagiełło
- Department of Tumour Pathology, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland
| | - Wiktoria M Suchorska
- Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland; Department of Electroradiology, Poznan University of Medical Sciences, Garbary 15 Street, 61-866, Poznan, Poland
| | - Tomasz Trzeciak
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland
| |
Collapse
|
10
|
Budde-Sagert K, Krueger S, Sehlke C, Lemcke H, Jonitz-Heincke A, David R, Bader R, Uhrmacher AM. detectCilia: An R Package for Automated Detection and Length Measurement of Primary Cilia. Bioinform Biol Insights 2024; 18:11779322241280431. [PMID: 39430098 PMCID: PMC11490958 DOI: 10.1177/11779322241280431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 07/23/2024] [Indexed: 10/22/2024] Open
Abstract
Background and objective The primary cilium is a small protrusion found on most mammalian cells. It acts as a cellular antenna, being involved in various cell signaling pathways. The length of the primary cilium affects its function. To study the impact of physical or chemical stimuli on cilia, their lengths must be determined easily and reproducibly. Methods We have developed and evaluated an open-source R package called detectCilia to detect and measure primary cilia automatically. As a case study to demonstrate the capability of our tool, we compared the influence of 4 different cell culture media compositions on the lengths of primary cilia in human chondrocytes. These media compositions include (1) insulin-transferrin-selenium (ITS); (2) ITS and dexamethasone (Dexa); (3) ITS, Dexa, insulin-like growth factor 1 (IGF-1), and transforming growth factor beta 1 (TGF-β1); and (4) fetal bovine serum (FBS). Results The assessment of detectCilia included a comparison with 2 similar tools: ACDC (Automated Cilia Detection in Cells) and CiliaQ. Several differences and advantages of our package make it a valuable addition to these tools. In the case study, we have observed variations in the ciliary lengths associated with using different media compositions. Conclusions We conclude that detectCilia can automatically and reproducibly detect and measure primary cilia in confocal microscopy images with low false-positive rates without requiring extensive user interaction.
Collapse
Affiliation(s)
- Kai Budde-Sagert
- Institute of Communications Engineering, University of Rostock, Rostock, Germany
- Institute for Visual and Analytic Computing, University of Rostock, Rostock, Germany
| | - Simone Krueger
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopedics, Rostock University Medical Center, Rostock, Germany
- Department of Life, Light, and Matter, Interdisciplinary Faculty, University of Rostock, Rostock, Germany
| | - Clemens Sehlke
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopedics, Rostock University Medical Center, Rostock, Germany
| | - Heiko Lemcke
- Department of Life, Light, and Matter, Interdisciplinary Faculty, University of Rostock, Rostock, Germany
- Reference and Translation Center for Cardiac Stem Cell Therapy, Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany
| | - Anika Jonitz-Heincke
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopedics, Rostock University Medical Center, Rostock, Germany
| | - Robert David
- Department of Life, Light, and Matter, Interdisciplinary Faculty, University of Rostock, Rostock, Germany
- Reference and Translation Center for Cardiac Stem Cell Therapy, Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany
| | - Rainer Bader
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopedics, Rostock University Medical Center, Rostock, Germany
- Department of Life, Light, and Matter, Interdisciplinary Faculty, University of Rostock, Rostock, Germany
| | - Adelinde M Uhrmacher
- Institute for Visual and Analytic Computing, University of Rostock, Rostock, Germany
- Department of Life, Light, and Matter, Interdisciplinary Faculty, University of Rostock, Rostock, Germany
| |
Collapse
|
11
|
Chu YY, Hikita A, Asawa Y, Hoshi K. Advancements in chondrocyte 3-dimensional embedded culture: Implications for tissue engineering and regenerative medicine. Biomed J 2024; 48:100786. [PMID: 39236979 PMCID: PMC12018037 DOI: 10.1016/j.bj.2024.100786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/09/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
Cartilage repair necessitates regenerative medicine because of the unreliable healing mechanism of cartilage. To yield a sufficient number of cells for transplantation, chondrocytes must be expanded in culture. However, in 2D culture, chondrocytes tend to lose their distinctive phenotypes and functionalities after serial passage, thereby limiting their efficacy for tissue engineering purposes. The mechanism of dedifferentiation in 2D culture can be attributed to various factors, including abnormal nuclear strength, stress-induced mitochondrial impairment, chromatin remodeling, ERK-1/2 and the p38/mitogen-activated protein kinase (MAPK) signaling pathway. These mechanisms collectively contribute to the loss of chondrocyte phenotype and reduced production of cartilage-specific extracellular matrix (ECM) components. Chondrocyte 3D culture methods have emerged as promising solutions to prevent dedifferentiation. Techniques, such as scaffold-based culture and scaffold-free approaches, provide chondrocytes with a more physiologically relevant environment, promoting their differentiation and matrix synthesis. These methods have been used in cartilage tissue engineering to create engineered cartilage constructs for transplantation and joint repair. However, chondrocyte 3D culture still has limitations, such as low viability and proliferation rate, and also difficulties in passage under 3D condition. These indicate challenges of obtaining a sufficient number of chondrocytes for large-scale tissue production. To address these issues, ongoing studies of many research groups have been focusing on refining culture conditions, optimizing scaffold materials, and exploring novel cell sources such as stem cells to enhance the quality and quantity of engineered cartilage tissues. Although obstacles remain, continuous endeavors to enhance culture techniques and overcome limitations offer a promising outlook for the advancement of more efficient strategies for cartilage regeneration.
Collapse
Affiliation(s)
- Yu-Ying Chu
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Plastic and Reconstructive Surgery, Craniofacial Research Centre, Chang Gung Memorial Hospital at Linko, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Atsuhiko Hikita
- Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan
| | - Yukiyo Asawa
- Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan
| | - Kazuto Hoshi
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
12
|
Li T, Liu J, Guo M, Bin FC, Duan Q, Dong XZ, Jin F, Fujita K, Zheng ML. Femtosecond Laser Maskless Optical Projection Lithography of Cartilage PCM Inspired 3D Protein Matrix to Chondrocyte Phenotype. Adv Healthc Mater 2024; 13:e2400849. [PMID: 38687974 DOI: 10.1002/adhm.202400849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/28/2024] [Indexed: 05/02/2024]
Abstract
Hydrogels containing chondrocytes have exhibited excellent potential in regenerating hyaline cartilage. However, chondrocytes are vulnerable to dedifferentiation during in vitro culture, leading to fibrosis and mechanical degradation of newly formed cartilage. It is proposed to modulate cartilage formation via the developed chondrocyte pericellular matrix (PCM) -like scaffolds for the first time, in which the S, M, and L-sized scaffolds are fabricated by femtosecond laser maskless optical projection lithography (FL-MOPL) of bovine serum albumin-glyceryl methacrylate hydrogel. Chondrocytes on the M PCM-like scaffold can maintain round morphology and synthesize extracellular matrix (ECM) to induce regeneration of hyaline cartilage microtissues by geometrical restriction. A series of M PCM-like scaffolds is fabricated with different stiffness and those with a high Young's modulus are more effective in maintaining the chondrocyte phenotype. The proposed PCM-like scaffolds are effective in modulating cartilage formation influenced by pore size, depth, and stiffness, which will pave the way for a better understanding of the geometric cues of mechanotransduction interactions in regulating cell fate and open up new avenues for tissue engineering.
Collapse
Affiliation(s)
- Teng Li
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Jie Liu
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| | - Min Guo
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Fan-Chun Bin
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Qi Duan
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Xian-Zi Dong
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| | - Feng Jin
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| | - Katsumasa Fujita
- Department of Applied Physics, Osaka University, 2-1Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mei-Ling Zheng
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| |
Collapse
|
13
|
Yammine KM, Abularach SM, Kim SY, Bikovtseva AA, Lilianty J, Butty VL, Schiavoni RP, Bateman JF, Lamandé SR, Shoulders MD. ER procollagen storage defect without coupled unfolded protein response drives precocious arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.19.562780. [PMID: 37905055 PMCID: PMC10614947 DOI: 10.1101/2023.10.19.562780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Collagenopathies are a group of clinically diverse disorders caused by defects in collagen folding and secretion. For example, mutations in the gene encoding collagen type-II, the primary collagen in cartilage, can lead to diverse chondrodysplasias. One example is the Gly1170Ser substitution in procollagen-II, which causes precocious osteoarthritis. Here, we biochemically and mechanistically characterize an induced pluripotent stem cell-based cartilage model of this disease, including both hetero- and homozygous genotypes. We show that Gly1170Ser procollagen-II is notably slow to fold and secrete. Instead, procollagen-II accumulates intracellularly, consistent with an endoplasmic reticulum (ER) storage disorder. Owing to unique features of the collagen triple helix, this accumulation is not recognized by the unfolded protein response. Gly1170Ser procollagen-II interacts to a greater extent than wild-type with specific proteostasis network components, consistent with its slow folding. These findings provide mechanistic elucidation into the etiology of this disease. Moreover, the cartilage model will enable rapid testing of therapeutic strategies to restore proteostasis in the collagenopathies.
Collapse
|
14
|
Huang YH, Chen HA, Chen CH, Liao HT, Kuo CY, Chen JP. Injectable gelatin/glucosamine cryogel microbeads as scaffolds for chondrocyte delivery in cartilage tissue engineering. Int J Biol Macromol 2023; 253:126528. [PMID: 37633562 DOI: 10.1016/j.ijbiomac.2023.126528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/16/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
In this study, we fabricate squeezable cryogel microbeads as injectable scaffolds for minimum invasive delivery of chondrocytes for cartilage tissue engineering applications. The microbeads with different glucosamine concentrations were prepared by combining the water-in-oil emulsion and cryogelation through crosslinking of gelatin with glutaraldehyde in the presence of glucosamine. The physicochemical characterization results show the successful preparation of cryogel microbeads with uniform shape and size, high porosity, large pore size, high water uptake capacity, and good injectability. In vitro analysis indicates proliferation, migration, and differentiated phenotype of rabbit chondrocytes in the cryogel scaffolds. The seeded chondrocytes in the cryogel scaffold can be delivered by injecting through an 18G needle to fully retain the cell viability. Furthermore, the incorporation of glucosamine in the cryogel promoted the differentiated phenotype of chondrocytes in a dose-dependent manner, from cartilage-specific gene expression and protein production. The in vivo study by injecting the cryogel microbeads into the subcutaneous pockets of nude mice indicates good retention ability as well as good biocompatibility and suitable biodegradability of the cryogel scaffold. Furthermore, the injected chondrocyte/cryogel microbead constructs can form ectopic functional neocartilage tissues following subcutaneous implantation in 21 days, as evidenced by histological and immunohistochemical analysis.
Collapse
Affiliation(s)
- Yen-Hsiang Huang
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Huai-An Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Chih-Hao Chen
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Keelung, Keelung 20401, Taiwan; Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan
| | - Han-Tsung Liao
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan
| | - Chang-Yi Kuo
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan; Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33305, Taiwan; Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan.
| |
Collapse
|
15
|
Bačenková D, Trebuňová M, Demeterová J, Živčák J. Human Chondrocytes, Metabolism of Articular Cartilage, and Strategies for Application to Tissue Engineering. Int J Mol Sci 2023; 24:17096. [PMID: 38069417 PMCID: PMC10707713 DOI: 10.3390/ijms242317096] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Hyaline cartilage, which is characterized by the absence of vascularization and innervation, has minimal self-repair potential in case of damage and defect formation in the chondral layer. Chondrocytes are specialized cells that ensure the synthesis of extracellular matrix components, namely type II collagen and aggregen. On their surface, they express integrins CD44, α1β1, α3β1, α5β1, α10β1, αVβ1, αVβ3, and αVβ5, which are also collagen-binding components of the extracellular matrix. This article aims to contribute to solving the problem of the possible repair of chondral defects through unique methods of tissue engineering, as well as the process of pathological events in articular cartilage. In vitro cell culture models used for hyaline cartilage repair could bring about advanced possibilities. Currently, there are several variants of the combination of natural and synthetic polymers and chondrocytes. In a three-dimensional environment, chondrocytes retain their production capacity. In the case of mesenchymal stromal cells, their favorable ability is to differentiate into a chondrogenic lineage in a three-dimensional culture.
Collapse
Affiliation(s)
- Darina Bačenková
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, Letná 9, 042 00 Košice, Slovakia; (M.T.); (J.D.); (J.Ž.)
| | | | | | | |
Collapse
|
16
|
Leal AF, Alméciga-Díaz CJ, Tomatsu S. Mucopolysaccharidosis IVA: Current Disease Models and Drawbacks. Int J Mol Sci 2023; 24:16148. [PMID: 38003337 PMCID: PMC10671113 DOI: 10.3390/ijms242216148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Mucopolysaccharidosis IVA (MPS IVA) is a rare disorder caused by mutations in the N-acetylgalactosamine-6-sulfate-sulfatase (GALNS) encoding gene. GALNS leads to the lysosomal degradation of the glycosaminoglyccreasans keratan sulfate and chondroitin 6-sulfate. Impaired GALNS enzymes result in skeletal and non-skeletal complications in patients. For years, the MPS IVA pathogenesis and the assessment of promising drugs have been evaluated using in vitro (primarily fibroblasts) and in vivo (mainly mouse) models. Even though value information has been raised from those studies, these models have several limitations. For instance, chondrocytes have been well recognized as primary cells affected in MPS IVA and responsible for displaying bone development impairment in MPS IVA patients; nonetheless, only a few investigations have used those cells to evaluate basic and applied concepts. Likewise, current animal models are extensively represented by mice lacking GALNS expression; however, it is well known that MPS IVA mice do not recapitulate the skeletal dysplasia observed in humans, making some comparisons difficult. This manuscript reviews the current in vitro and in vivo MPS IVA models and their drawbacks.
Collapse
Affiliation(s)
- Andrés Felipe Leal
- Nemours Children’s Health, Wilmington, DE 19803, USA;
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia;
| | - Carlos Javier Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia;
| | - Shunji Tomatsu
- Nemours Children’s Health, Wilmington, DE 19803, USA;
- Faculty of Arts and Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu 501-1193, Japan
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19144, USA
| |
Collapse
|
17
|
Chen CH, Kao HH, Lee YC, Chen JP. Injectable Thermosensitive Hyaluronic Acid Hydrogels for Chondrocyte Delivery in Cartilage Tissue Engineering. Pharmaceuticals (Basel) 2023; 16:1293. [PMID: 37765101 PMCID: PMC10535600 DOI: 10.3390/ph16091293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
In this study, we synthesize a hyaluronic acid-g-poly(N-isopropylacrylamide) (HPN) copolymer by grafting the amine-terminated poly(N-isopropylacrylamide) (PNIPAM-NH2) to hyaluronic acid (HA). The 5% PNIPAM-NH2 and HPN polymer solution is responsive to temperature changes with sol-to-gel phase transition temperatures around 32 °C. Compared with the PNIPAM-NH2 hydrogel, the HPN hydrogel shows higher water content and mechanical strength, as well as lower volume contraction, making it a better choice as a scaffold for chondrocyte delivery. From an in vitro cell culture, we see that cells can proliferate in an HPN hydrogel with full retention of cell viability and show the phenotypic morphology of chondrocytes. In the HPN hydrogel, chondrocytes demonstrate a differentiated phenotype with the upregulated expression of cartilage-specific genes and the enhanced secretion of extracellular matrix components, when compared with the monolayer culture on tissue culture polystyrene. In vivo studies confirm the ectopic cartilage formation when HPN was used as a cell delivery vehicle after implanting chondrocyte/HPN in nude mice subcutaneously, which is shown from a histological and gene expression analysis. Taken together, the HPN thermosensitive hydrogel will be a promising injectable scaffold with which to deliver chondrocytes in cartilage-tissue-engineering applications.
Collapse
Affiliation(s)
- Chih-Hao Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Keelung, Chang Gung University College of Medicine, Keelung 20401, Taiwan
| | - Hao-Hsi Kao
- Division of Nephrology, Chang Gung Memorial Hospital at Keelung, Chang Gung University College of Medicine, Keelung 20401, Taiwan
| | - Yen-Chen Lee
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-San, Taoyuan 33302, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
18
|
Jiang J, Altammar J, Cong X, Ramsauer L, Steinbacher V, Dornseifer U, Schilling AF, Machens HG, Moog P. Hypoxia Preconditioned Serum (HPS) Promotes Proliferation and Chondrogenic Phenotype of Chondrocytes In Vitro. Int J Mol Sci 2023; 24:10441. [PMID: 37445617 PMCID: PMC10341616 DOI: 10.3390/ijms241310441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Autologous chondrocyte implantation (ACI) for the treatment of articular cartilage defects remains challenging in terms of maintaining chondrogenic phenotype during in vitro chondrocyte expansion. Growth factor supplementation has been found supportive in improving ACI outcomes by promoting chondrocyte redifferentiation. Here, we analysed the chondrogenic growth factor concentrations in the human blood-derived secretome of Hypoxia Preconditioned Serum (HPS) and assessed the effect of HPS-10% and HPS-40% on human articular chondrocytes from osteoarthritic cartilage at different time points compared to normal fresh serum (NS-10% and NS-40%) and FCS-10% culture conditions. In HPS, the concentrations of TGF-beta1, IGF-1, bFGF, PDGF-BB and G-CSF were found to be higher than in NS. Chondrocyte proliferation was promoted with higher doses of HPS (HPS-40% vs. HPS-10%) and longer stimulation (4 vs. 2 days) compared to FCS-10%. On day 4, immunostaining of the HPS-10%-treated chondrocytes showed increased levels of collagen type II compared to the other conditions. The promotion of the chondrogenic phenotype was validated with quantitative real-time PCR for the expression of collagen type II (COL2A1), collagen type I (COL1A1), SOX9 and matrix metalloproteinase 13 (MMP13). We demonstrated the highest differentiation index (COL2A1/COL1A1) in HPS-10%-treated chondrocytes on day 4. In parallel, the expression of differentiation marker SOX9 was elevated on day 4, with HPS-10% higher than NS-10/40% and FCS-10%. The expression of the cartilage remodelling marker MMP13 was comparable across all culture conditions. These findings implicate the potential of HPS-10% to improve conventional FCS-based ACI culture protocols by promoting the proliferation and chondrogenic phenotype of chondrocytes during in vitro expansion.
Collapse
Affiliation(s)
- Jun Jiang
- Experimental Plastic Surgery, Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
| | - Jannat Altammar
- Experimental Plastic Surgery, Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
| | - Xiaobin Cong
- Experimental Plastic Surgery, Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
| | - Lukas Ramsauer
- Institute of Molecular Immunology and Experimental Oncology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
| | - Vincent Steinbacher
- Institute of Molecular Immunology and Experimental Oncology, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
| | - Ulf Dornseifer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Isar Klinikum, D-80331 Munich, Germany
| | - Arndt F. Schilling
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Göttingen, D-37075 Göttingen, Germany
| | - Hans-Günther Machens
- Experimental Plastic Surgery, Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
| | - Philipp Moog
- Experimental Plastic Surgery, Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, D-81675 Munich, Germany
| |
Collapse
|
19
|
Al-Maslamani NA, Oldershaw R, Tew S, Curran J, D’Hooghe P, Yamamoto K, Horn HF. Chondrocyte De-Differentiation: Biophysical Cues to Nuclear Alterations. Cells 2022; 11:cells11244011. [PMID: 36552775 PMCID: PMC9777101 DOI: 10.3390/cells11244011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Autologous chondrocyte implantation (ACI) is a cell therapy to repair cartilage defects. In ACI a biopsy is taken from a non-load bearing area of the knee and expanded in-vitro. The expansion process provides the benefit of generating a large number of cells required for implantation; however, during the expansion these cells de-differentiate and lose their chondrocyte phenotype. In this review we focus on examining the de-differentiation phenotype from a mechanobiology and biophysical perspective, highlighting some of the nuclear mechanics and chromatin changes in chondrocytes seen during the expansion process and how this relates to the gene expression profile. We propose that manipulating chondrocyte nuclear architecture and chromatin organization will highlight mechanisms that will help to preserve the chondrocyte phenotype.
Collapse
Affiliation(s)
- Noor A. Al-Maslamani
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Correspondence:
| | - Rachel Oldershaw
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Simon Tew
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Jude Curran
- Department of Mechanical, Materials and Aerospace Engineering, School of Engineering, University of Liverpool, Liverpool L69 3GH, UK
| | - Pieter D’Hooghe
- Department of Orthopaedic Surgery, Aspetar Orthopaedic and Sports Medicine Hospital, Doha P.O. Box 29222, Qatar
| | - Kazuhiro Yamamoto
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Henning F. Horn
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
| |
Collapse
|
20
|
Riazi Moghadam R, Keshvari H, Imani R, Nazarpak MH. A biomimetic three-layered fibrin gel/PLLA nanofibers composite as a potential scaffold for articular cartilage tissue engineering application. Biomed Mater 2022; 17. [PMID: 35973416 DOI: 10.1088/1748-605x/ac8a32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 08/16/2022] [Indexed: 11/11/2022]
Abstract
Developing an engineered scaffold inspired by structural features of healthy articular cartilage (AC) has attracted much attention. In this study, the design and fabrication of a three-layered fiber/hydrogel scaffold in which each layer replicates the organization of a pertinent layer of AC tissue is aimed. To this end, electrospun poly L-lactic acid (PLLA) nanofibers are prepared and fragmented into nano/micro cylinders via aminolysis. Three-layers of the scaffold in which continuous fibrous layer, fibrin gel incorporated by chopped fibers and fibrin gel embedded by cylindrical aligned fibrous mat perpendicular to articulating surface, respectively served as an upper, middle and bottom layers, are prepared. The layers' physicomechanical characteristics are comprehensively evaluated. Results show that optimized electrospinning set up results in the smallest fibers diameter of 367±317 nm and successful aminolysis provides amine-functionalized chopped nanofibers with a mean length of 1.65±1.2 µm. Static mechanical analysis of the layers demonstrates that Young tensile modulus of the upper layer is 152± 17 MPa while compressive moduli of the middle and bottom layers are 38±4 and 79± 6 KPa, respectively. Assessing mechanical parameters under dynamic loading also shows that adding fibrous part in the composite scaffold layers enhances viscoelastic behavior of fibrin gel. Also, incorporation of 0.25% chopped fibers into the fibrin matrix notably enhances the equilibrium water content; however, it increases in-vitro weigh loss rate from 6% to 10.5% during a seven-day period. cytocompatibility analysis confirms that all layers possess acceptable cytocompatibility. In a conclusion, the designed three-layered composite structure successfully mimics the physicomechanical as well as microstructural features of AC and could be suggested as a potential scaffold for this tissue regeneration.
Collapse
Affiliation(s)
- Roozbeh Riazi Moghadam
- Biomedical Engineering, Amirkabir University of Technology, No. 350, Hafez Ave, Valiasr Square, Tehran, Iran, Tehran, 159163-4311, Iran (the Islamic Republic of)
| | - Hamid Keshvari
- Department of Biomedical Engineering, Amirkabir University of Technology, No. 350, Hafez Ave, Valiasr Square, Tehran, Iran, IRAN, Tehran, Tehran, 159163-4311, Iran (the Islamic Republic of)
| | - Rana Imani
- Biomedical Engineering, Amirkabir University of Technology, No. 350, Hafez Ave, Valiasr Square, Tehran, Iran, Tehran, 159163-4311, Iran (the Islamic Republic of)
| | - Masoumeh Haghbin Nazarpak
- New technologies Research Center, Amirkabir University of Technology, No. 350, Hafez Ave, Valiasr Square, Tehran, Iran 1591634311, Tehran, 158754413, Iran (the Islamic Republic of)
| |
Collapse
|
21
|
Beatty LA, Mansour KL, Bryant EJ, Garcia-Godoy FJ, Pantaleon DS, Sawatari Y, Huang CYC, Garcia-Godoy F. Chondroprotective Effects of Periodontal Ligament Derived Stem Cells Conditioned Medium on Articular Cartilage After Impact Injury. Stem Cells Dev 2022; 31:498-505. [PMID: 35730119 DOI: 10.1089/scd.2022.0130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Paracrine factors secreted in the conditioned media of periodontal ligament derived stem cells (PDLSCs) have been shown to downregulate inflammatory effects of IL-1β on chondrocytes wherein milk fat globule-epidermal growth factor 8 (MFG-E8) is one of the PDLSCs highly secretory proteins. Therefore, the objective of this study was to investigate the ability of PDLSC conditioned media (CM) and MFG-E8 to reduce the inflammatory effects of impact injury on porcine talar articular cartilage (AC) and IL-1β on chondrocytes, respectively. Stem cells were isolated from human periodontal ligaments. the MFG-E8 content in CM collected at 5% and 20% oxygen was measured by ELISA assay and compared across subcultures and donors. AC samples were divided into three groups: control, impact, and impact+CM. Chondrocytes were isolated from pig knees and were divided into three groups: control, IL-1β, and IL-1β+MFG-E8. Gene expression data was analyzed by RT-PCR. It was found that impact load and IL-1β treatment upregulated IL-1β, TNF-α, ADAMTS-4, and ADAMTS-5 gene expression in AC and chondrocytes, respectively. PDLSCs-CM prevented the upregulation of all four genes due to impact whereas MFG-E8 prevented upregulation of IL-1β, ADAMTS-4, and ADAMTS-5 in chondrocytes, but it did not prevent TNF-α upregulation. There were no significant differences in MFG-E8 content in CM among oxygen levels, passage numbers, or donors. The findings suggested that MFG-E8 is an effective anti-inflammatory agent contributing to the chondroprotective effects of PDLSCs-CM on acutely injured articular cartilage. Thus, introducing PDLSCs-CM to sites of acute traumatic AC injury could prevent the development of post-traumatic osteoarthritis.
Collapse
Affiliation(s)
- Logan Albert Beatty
- University of Miami - Coral Gables Campus, 5452, Biomedical Engineering, Coral Gables, Florida, United States;
| | - Kailey L Mansour
- University of Miami Miller School of Medicine, 12235, Miami, Florida, United States;
| | - Evan J Bryant
- University of Miami Miller School of Medicine, 12235, Miami, Florida, United States;
| | | | | | - Yoh Sawatari
- University of Miami Miller School of Medicine, 12235, Division of Oral and Maxillofacial Surgery, Miami, Florida, United States;
| | - Chun-Yuh Charles Huang
- University of Miami - Coral Gables Campus, 5452, Biomedical Engineering, Coral Gables, Florida, United States;
| | - Franklin Garcia-Godoy
- University of Tennessee Health Science Center, Bioscience Research, College of Dentistry, Memphis, Tennessee, United States;
| |
Collapse
|
22
|
Kahraman E, Ribeiro R, Lamghari M, Neto E. Cutting-Edge Technologies for Inflamed Joints on Chip: How Close Are We? Front Immunol 2022; 13:802440. [PMID: 35359987 PMCID: PMC8960235 DOI: 10.3389/fimmu.2022.802440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/18/2022] [Indexed: 11/17/2022] Open
Abstract
Osteoarthritis (OA) is a painful and disabling musculoskeletal disorder, with a large impact on the global population, resulting in several limitations on daily activities. In OA, inflammation is frequent and mainly controlled through inflammatory cytokines released by immune cells. These outbalanced inflammatory cytokines cause cartilage extracellular matrix (ECM) degradation and possible growth of neuronal fibers into subchondral bone triggering pain. Even though pain is the major symptom of musculoskeletal diseases, there are still no effective treatments to counteract it and the mechanisms behind these pathologies are not fully understood. Thus, there is an urgent need to establish reliable models for assessing the molecular mechanisms and consequently new therapeutic targets. Models have been established to support this research field by providing reliable tools to replicate the joint tissue in vitro. Studies firstly started with simple 2D culture setups, followed by 3D culture focusing mainly on cell-cell interactions to mimic healthy and inflamed cartilage. Cellular approaches were improved by scaffold-based strategies to enhance cell-matrix interactions as well as contribute to developing mechanically more stable in vitro models. The progression of the cartilage tissue engineering would then profit from the integration of 3D bioprinting technologies as these provide 3D constructs with versatile structural arrangements of the 3D constructs. The upgrade of the available tools with dynamic conditions was then achieved using bioreactors and fluid systems. Finally, the organ-on-a-chip encloses all the state of the art on cartilage tissue engineering by incorporation of different microenvironments, cells and stimuli and pave the way to potentially simulate crucial biological, chemical, and mechanical features of arthritic joint. In this review, we describe the several available tools ranging from simple cartilage pellets to complex organ-on-a-chip platforms, including 3D tissue-engineered constructs and bioprinting tools. Moreover, we provide a fruitful discussion on the possible upgrades to enhance the in vitro systems making them more robust regarding the physiological and pathological modeling of the joint tissue/OA.
Collapse
Affiliation(s)
- Emine Kahraman
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Rua Dr. Roberto Frias, Porto, Portugal
| | - Ricardo Ribeiro
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Meriem Lamghari
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Estrela Neto
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| |
Collapse
|
23
|
Klimek K, Tarczynska M, Truszkiewicz W, Gaweda K, Douglas TEL, Ginalska G. Freeze-Dried Curdlan/Whey Protein Isolate-Based Biomaterial as Promising Scaffold for Matrix-Associated Autologous Chondrocyte Transplantation-A Pilot In-Vitro Study. Cells 2022; 11:282. [PMID: 35053397 PMCID: PMC8773726 DOI: 10.3390/cells11020282] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/09/2022] [Accepted: 01/12/2022] [Indexed: 01/18/2023] Open
Abstract
The purpose of this pilot study was to establish whether a novel freeze-dried curdlan/whey protein isolate-based biomaterial may be taken into consideration as a potential scaffold for matrix-associated autologous chondrocyte transplantation. For this reason, this biomaterial was initially characterized by the visualization of its micro- and macrostructures as well as evaluation of its mechanical stability, and its ability to undergo enzymatic degradation in vitro. Subsequently, the cytocompatibility of the biomaterial towards human chondrocytes (isolated from an orthopaedic patient) was assessed. It was demonstrated that the novel freeze-dried curdlan/whey protein isolate-based biomaterial possessed a porous structure and a Young's modulus close to those of the superficial and middle zones of cartilage. It also exhibited controllable degradability in collagenase II solution over nine weeks. Most importantly, this biomaterial supported the viability and proliferation of human chondrocytes, which maintained their characteristic phenotype. Moreover, quantitative reverse transcription PCR analysis and confocal microscope observations revealed that the biomaterial may protect chondrocytes from dedifferentiation towards fibroblast-like cells during 12-day culture. Thus, in conclusion, this pilot study demonstrated that novel freeze-dried curdlan/whey protein isolate-based biomaterial may be considered as a potential scaffold for matrix-associated autologous chondrocyte transplantation.
Collapse
Affiliation(s)
- Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (W.T.); (G.G.)
| | - Marta Tarczynska
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-090 Lublin, Poland; (M.T.); (K.G.)
| | - Wieslaw Truszkiewicz
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (W.T.); (G.G.)
| | - Krzysztof Gaweda
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-090 Lublin, Poland; (M.T.); (K.G.)
| | - Timothy E. L. Douglas
- Engineering Department, Lancaster University, Gillow Avenue, Lancaster LA 1 4YW, UK;
- Materials Science Institute (MSI), Lancaster University, Lancaster LA 1 4YW, UK
| | - Grazyna Ginalska
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland; (W.T.); (G.G.)
| |
Collapse
|