1
|
Kahraman E, Vasconcelos D, Ribeiro B, Monteiro AC, Mastromatteo E, Bortolin A, Couto M, Boschis L, Lamghari M, Neto E. Deciphering cartilage neuro-immune interactions and innervation profile through 3D engineered osteoarthritic micropathophysiological system. Mater Today Bio 2025; 31:101491. [PMID: 39896288 PMCID: PMC11786692 DOI: 10.1016/j.mtbio.2025.101491] [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: 04/23/2024] [Revised: 11/15/2024] [Accepted: 01/12/2025] [Indexed: 02/04/2025] Open
Abstract
Osteoarthritis (OA) is an inflammatory musculoskeletal disorder that results in cartilage breakdown and alterations in the surrounding tissue microenvironment. Imbalances caused by inflammation and catabolic processes potentiate pathological nerves and blood vessels outgrowth toward damaged areas leading to pain in the patients. Yet, the precise mechanisms leading the nerve sprouting into the aneural cartilaginous tissue remain elusive. In this work, we aim to recapitulate in vitro the hallmarks of OA pathophysiology, including the sensory innervation profile, and provide a sensitive and reliable analytical tool to monitor the in vitro disease progression at microscale. Leveraging the use of patient-derived cells and bioengineering cutting-edge technologies, we engineered cartilage-like microtissues composed of primary human chondrocytes encapsulated in gelatin methacrylate hydrogel. Engineered constructs patterned inside microfluidic devices show the expression of cartilage markers, namely collagen type II, aggrecan, SOX-9 and glycosaminoglycans. Upon pro-inflammatory triggering, using primary human pro-inflammatory macrophage secretome, hallmarks of OA are recapitulated namely catabolic processes of human chondrocytes and the sensory innervation profile, supported by gene expression and functional assays. To monitor the OA micropathological system, a highly sensitive technology - EliChip™ - is presented to quantitively assess the molecular signature of cytokines and growth factors (interleukin 6 and nerve growth factor) produced from a single microfluidic chip. Herein, we report a miniaturized pathophysiological model and analytical tool to foster the neuro-immune interactions playing a role in cartilage-related disorders.
Collapse
Affiliation(s)
- Emine Kahraman
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal
| | - Daniela Vasconcelos
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Beatriz Ribeiro
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Ana Carolina Monteiro
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Enzo Mastromatteo
- Trustech Innovation Technology, Via Baraggino, 76, 10034, Chivasso, Torino, Italy
| | - Andrea Bortolin
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal
| | - Marina Couto
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Laura Boschis
- Trustech Innovation Technology, Via Baraggino, 76, 10034, Chivasso, Torino, Italy
| | - Meriem Lamghari
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Estrela Neto
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| |
Collapse
|
2
|
Peng X, Chen X, Zhang Y, Tian Z, Wang M, Chen Z. Advances in the pathology and treatment of osteoarthritis. J Adv Res 2025:S2090-1232(25)00072-4. [PMID: 39889821 DOI: 10.1016/j.jare.2025.01.053] [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: 10/12/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Osteoarthritis (OA), a widespread degenerative joint disease, predominantly affects individuals from middle age onwards, exhibiting non-inflammatory characteristics. OA leads to the gradual deterioration of articular cartilage and subchondral bone, causing pain and reduced mobility. The risk of OA increases with age, making it a critical health concern for seniors. Despite significant research efforts and various therapeutic approaches, the precise causes of OA remain unclear. AIM OF REVIEW This paper provides a thorough examination of OA characteristics, pathogenic mechanisms at various levels, and personalized treatment strategies for different OA stages. The review aims to enhance understanding of disease mechanisms and establish a theoretical framework for developing more effective therapeutic interventions. KEY SCIENTIFIC CONCEPTS OF REVIEW This review systematically examines OA through multiple perspectives, integrating current knowledge of clinical presentation, pathological mechanisms, and associated signaling pathways. It assesses diagnostic methods and reviews both pharmacological and surgical treatments for OA, as well as emerging tissue engineering approaches to manage the disease. While therapeutic strategies such as exercise, anti-inflammatory drugs, and surgical interventions are employed to manage symptoms and modify joint structure, none have been able to effectively halt OA's advancement or achieve long-lasting symptom relief. Tissue engineering strategies, such as cell-seeded scaffolds, supportive matrices, and growth factor delivery, have emerged as promising approaches for cartilage repair and OA treatment. To combat the debilitating effects of OA, it is crucial to investigate the molecular basis of its pathogenesis and seek out innovative therapeutic targets for more potent preventive and treatment strategies.
Collapse
Affiliation(s)
- Xueliang Peng
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Xuanning Chen
- College of Stomatology, Shanghai Jiao Tong University, Shanghai 200215, China
| | - Yifan Zhang
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Zhichao Tian
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Meihua Wang
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China
| | - Zhuoyue Chen
- Provincial Key Laboratory of Biotechnology of Shaanxi, Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Faculty of Life Science, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi Province 710069, China.
| |
Collapse
|
3
|
Wagenbrenner M, Heinz T, Anderson PM, Stratos I, Arnholdt J, Mayer-Wagner S, Horas K, Docheva D, Holzapfel BM, Rudert M, Weißenberger M. Does Combined Treatment with Tranexamic Acid and Vancomycin Affect Human Chondrocytes In Vitro? Pharmaceuticals (Basel) 2024; 17:1576. [PMID: 39770418 PMCID: PMC11677089 DOI: 10.3390/ph17121576] [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: 09/28/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The aim of our study was to examine the combined effects of tranexamic acid (TXA) and vancomycin powder (VP) on chondrocytes in vitro. Despite the use of TXA and VP being linked to a reduced risk of extensive postoperative blood loss and periprosthetic joint infections (PJIs) in TKA, the possible cytotoxic side effects on periarticular cell types remain unclear. Methods: Human chondrocytes were harvested from hyaline cartilage and expanded in monolayer culture before being simultaneously exposed to different concentrations of TXA and VP for varying exposure times. Cell viability and proliferation were assessed using an ATP assay and an Annexin 5 assay, respectively, while changes in the relative expression of chondrogenic marker genes were examined using semiquantitative RT-PCR. Results: The simultaneous exposure of chondrocytes to TXA and VP for more than 48 h led to a reduction in both cell viability and proliferation rates. When exposing chondrocytes to the lowest examined concentrations of both TXA (10 mg/mL) and VP (3 mg/mL), the observed effects were delayed until 96 h. However, our study found no dependencies of the observed effects on the concentrations tested. Further, we found no effects on the expression of chondrogenic marker genes. Conclusions: Consequently, limiting the exposure time of chondrocytes to TXA and VP in an in vitro setting to 24 h may be considered safe and could help to further improve the understanding of the safe use of substances in vivo. However, further in vitro research is required to develop a comprehensive understanding of the effects of both VP and TXA on important periarticular cell types in TKA, including chondrocytes, osteocytes, and tenocytes.
Collapse
Affiliation(s)
- Mike Wagenbrenner
- Department of Orthopaedic Surgery and Musculoskeletal Tissue Regeneration, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany; (M.W.); (T.H.); (P.M.A.); , (K.H.); (D.D.); (M.R.)
| | - Tizian Heinz
- Department of Orthopaedic Surgery and Musculoskeletal Tissue Regeneration, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany; (M.W.); (T.H.); (P.M.A.); , (K.H.); (D.D.); (M.R.)
| | - Philip M. Anderson
- Department of Orthopaedic Surgery and Musculoskeletal Tissue Regeneration, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany; (M.W.); (T.H.); (P.M.A.); , (K.H.); (D.D.); (M.R.)
| | - Ioannis Stratos
- Department of Orthopaedic Surgery and Musculoskeletal Tissue Regeneration, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany; (M.W.); (T.H.); (P.M.A.); , (K.H.); (D.D.); (M.R.)
| | - Joerg Arnholdt
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, 81377 Munich, Germany; (J.A.); (S.M.-W.); (B.M.H.)
| | - Susanne Mayer-Wagner
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, 81377 Munich, Germany; (J.A.); (S.M.-W.); (B.M.H.)
| | - Konstantin Horas
- Department of Orthopaedic Surgery and Musculoskeletal Tissue Regeneration, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany; (M.W.); (T.H.); (P.M.A.); , (K.H.); (D.D.); (M.R.)
| | - Denitsa Docheva
- Department of Orthopaedic Surgery and Musculoskeletal Tissue Regeneration, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany; (M.W.); (T.H.); (P.M.A.); , (K.H.); (D.D.); (M.R.)
| | - Boris M. Holzapfel
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, 81377 Munich, Germany; (J.A.); (S.M.-W.); (B.M.H.)
| | - Maximilian Rudert
- Department of Orthopaedic Surgery and Musculoskeletal Tissue Regeneration, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany; (M.W.); (T.H.); (P.M.A.); , (K.H.); (D.D.); (M.R.)
| | - Manuel Weißenberger
- Department of Orthopaedic Surgery and Musculoskeletal Tissue Regeneration, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstr. 11, 97074 Wuerzburg, Germany; (M.W.); (T.H.); (P.M.A.); , (K.H.); (D.D.); (M.R.)
| |
Collapse
|
4
|
Kleuskens MWA, Crispim JF, van Doeselaar M, van Donkelaar CC, Janssen RPA, Ito K. Neo-cartilage formation using human nondegenerate versus osteoarthritic chondrocyte-derived cartilage organoids in a viscoelastic hydrogel. J Orthop Res 2023. [PMID: 36866819 DOI: 10.1002/jor.25540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 01/31/2023] [Accepted: 03/01/2023] [Indexed: 03/04/2023]
Abstract
Current regenerative cartilage therapies are associated with several drawbacks such as dedifferentiation of chondrocytes during expansion and the formation of fibrocartilage. Optimized chondrocyte expansion and tissue formation could lead to better clinical results of these therapies. In this study, a novel chondrocyte suspension expansion protocol that includes the addition of porcine notochordal cell-derived matrix was used to self-assemble human chondrocytes from osteoarthritic (OA) and nondegenerate (ND) origin into cartilage organoids containing collagen type II and proteoglycans. Proliferation rate and viability were similar for OA and ND chondrocytes and organoids formed had a similar histologic appearance and gene expression profile. Organoids were then encapsulated in viscoelastic alginate hydrogels to form larger tissues. Chondrocytes on the outer bounds of the organoids produced a proteoglycan-rich matrix to bridge the space between organoids. In hydrogels containing ND organoids some collagen type I was observed between the organoids. Surrounding the bulk of organoids in the center of the gels, in both OA and ND gels a continuous tissue containing cells, proteoglycans and collagen type II had been produced. No difference was observed in sulphated glycosaminoglycan and hydroxyproline content between gels containing organoids from OA or ND origin after 28 days. It was concluded that OA chondrocytes, which can be harvested from leftover surgery tissue, perform similar to ND chondrocytes in terms of human cartilage organoid formation and matrix production in alginate gels. This opens possibilities for their potential to serve as a platform for cartilage regeneration but also as an in vitro model to study pathways, pathology, or drug development.
Collapse
Affiliation(s)
- Meike W A Kleuskens
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - João F Crispim
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Marina van Doeselaar
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Corrinus C van Donkelaar
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Rob P A Janssen
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.,Department of Orthopaedic Surgery and Trauma, Máxima Medical Center, Eindhoven-Veldhoven, The Netherlands.,Department of Paramedical Sciences, Fontys University of Applied Sciences, Eindhoven, The Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
5
|
Gohari Z, Baghaei A, Mahboudi H, Hashemi J, Rahmati M, Islami M, Mansouri V. Ursolic acid incorporated nanofibers improves chondrogenic differentiation of mesenchymal stem cells. POLYM ADVAN TECHNOL 2022. [DOI: 10.1002/pat.5830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Zahra Gohari
- Department of Animal Science, School of Biology, Faculty of Science University of Tehran Tehran Iran
| | - Ahmadali Baghaei
- Faculty of Medicine, Mashhad Branch Islamic Azad University Mashhad Iran
| | - Hossein Mahboudi
- Department of Biotechnology, School of Pharmacy Alborz University of Medical Sciences karaj Iran
| | - Javad Hashemi
- Department of Pathobiology and Laboratory Sciences, Faculty of Medicine North Khorasan University of Medical Sciences Bojnurd Iran
| | - Mohammad Rahmati
- Department of Medical Biotechnology, Faculty of Paramedicine Guilan University of Medical Sciences Rasht Iran
| | - Maryam Islami
- Department of Biotechnology, School of Medicine Alborz University of Medical Science Karaj Iran
| | - Vahid Mansouri
- Proteomics Research Center, Faculty of Paramedical Sciences Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
6
|
Acevedo Rua L, Mumme M, Manferdini C, Darwiche S, Khalil A, Hilpert M, Buchner DA, Lisignoli G, Occhetta P, von Rechenberg B, Haug M, Schaefer DJ, Jakob M, Caplan A, Martin I, Barbero A, Pelttari K. Engineered nasal cartilage for the repair of osteoarthritic knee cartilage defects. Sci Transl Med 2021; 13:eaaz4499. [PMID: 34516821 DOI: 10.1126/scitranslmed.aaz4499] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Lina Acevedo Rua
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Marcus Mumme
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland.,Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Cristina Manferdini
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Salim Darwiche
- Musculoskeletal Research Unit MSRU, Equine Department, University of Zurich, 8057 Zürich, Switzerland
| | - Ahmad Khalil
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106 , USA
| | - Morgane Hilpert
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - David A Buchner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106 , USA
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Paola Occhetta
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Brigitte von Rechenberg
- Competence Center for Applied Biotechnology and Molecular Medicine CABMM, University of Zurich, 8057 Zürich, Switzerland
| | - Martin Haug
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Dirk J Schaefer
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Marcel Jakob
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Arnold Caplan
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14-16, 4123 Allschwil, Switzerland Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Karoliina Pelttari
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| |
Collapse
|
7
|
Wilken F, Slotta-Huspenina J, Laux F, Blanke F, Schauwecker J, Vogt S, Gollwitzer H. Autologous Chondrocyte Transplantation in Femoroacetabular Impingement Syndrome: Growth and Redifferentiation Potential of Chondrocytes Harvested from the Femur in Cam-Type Deformities. Cartilage 2021; 12:377-386. [PMID: 30862178 PMCID: PMC8236656 DOI: 10.1177/1947603519833138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE Cam-type femoroacetabular impingement (FAI) syndrome is one of the most frequent reasons for cartilage damage in the hip. Autologous chondrocyte transplantation has proven high success rates in the treatment of focal chondral defects; however, harvesting of chondrocytes in the hip has been reported but not specifically from the region of femoral cam lesions. Therefore, the goal of this study was to analyze the growth and redifferentiation potential of cartilage samples harvested from the cam deformities in patients with FAI. DESIGN Cartilage samples were gained from 15 patients with cam-type FAI undergoing arthroscopic femoral cam resection. Healthy (hyaline cartilage of the hip and knee joint, n = 12) and arthritic control groups (degenerative changes in cartilage of the hip joint, n = 8) were also analyzed. Chondrocytes were initially cultured under monolayer, and subsequently under pellet conditions. A comparative representation of the groups was performed by Mankin score classification, immunohistochemistry (IHC) (Col1, Col2, aggrecan), and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) (Col1, Col2, Col10, Sox9, RunX2). RESULTS Mankin score of FAI-samples (4.1±3.1, Range 0-10) showed a wide variation but was significant lower (P = 0.0244) when compared with the arthritic control (7.5 ± 2.7, range 4-12). IHC showed an increased deposition of Col2 (P = 0.0002) and aggrecan (P = 0.0261) after pellet culture compared with deposition after monolayer culture in all groups. In qRT-PCR, FAI samples showed after pellet culture increased Col2 (P = 0.0050) and Col10 expression (P = 0.0006) and also Mankin score correlated increasing gene-expression of Col10 (r = 0.8108, P = 0.0341) and RunX2 (r = 0.8829, P = 0.123). CONCLUSIONS Cartilage samples of patients with cam-type FAI showed sufficient but heterogeneous composition relating to histological quality and chondrogenic potential. However, harvesting of chondrocytes from the cam lesion might be a valid option especially if a cartilage lesion is noted in a diagnostic arthroscopy and individual preexisting stage of cartilage degeneration and appropriate pellet-culturing conditions are considered.
Collapse
Affiliation(s)
- Frauke Wilken
- Clinic of Orthopaedics and Orthopaedic Sports Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany,Department of Orthopaedic Sports Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Julia Slotta-Huspenina
- Institute of Pathology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Florian Laux
- Clinic of Orthopaedics and Orthopaedic Sports Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Fabian Blanke
- Department of Orthopaedic Sports Medicine, Hessing Stiftung, Augsburg, Germany
| | - Johannes Schauwecker
- Clinic of Orthopaedics and Orthopaedic Sports Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stephan Vogt
- Department of Orthopaedic Sports Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany,Department of Orthopaedic Sports Medicine, Hessing Stiftung, Augsburg, Germany,Stephan Vogt, Department of Orthopaedic Sports Medicine, Hessing Stiftung, Hessingstraße 17, 86199 Augsburg, Germany.
| | | |
Collapse
|
8
|
[Cartilage repair procedures for early osteoarthritis]. DER ORTHOPADE 2021; 50:356-365. [PMID: 33844031 DOI: 10.1007/s00132-021-04099-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/10/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Commonly used cartilage repair procedures have been established for focal cartilage lesions; however, degenerative lesions with accompanying changes of other intraarticular structures are much more common in clinical practice. This stage, in which classic radiological signs of osteoarthritis are absent, is called early osteoarthritis and is characterized by impaired joint homeostasis with biomechanical and biochemical changes that can have a negative effect on regenerative cartilage therapy procedures. INDICATION Cartilage repair procedures are indicated for symptomatic focal early osteoarthritis, defined as cartilage degeneration ICRS grades I or II around a focal cartilage defect ICRS grades III or IV. In more advanced osteoarthritis with significant narrowing of the joint space, cartilage repair procedures are generally contraindicated. THERAPY The most studied cartilage repair procedure for early osteoarthritis is autologous chondrocyte implantation, which has shown acceptable results in case series, although higher failure rates are to be expected compared to focal, traumatic cartilage lesions. The use of bone marrow-stimulating techniques seems to be limited in early osteoarthritis and should only be used in cases of lesion < 2 cm2 and very little surrounding cartilage degeneration. Concomitant surgical procedures, especially unloading osteotomies, are very important.
Collapse
|
9
|
Deszcz I, Lis-Nawara A, Grelewski P, Dragan S, Bar J. Utility of direct 3D co-culture model for chondrogenic differentiation of mesenchymal stem cells on hyaluronan scaffold (Hyaff-11). Regen Biomater 2020; 7:543-552. [PMID: 33365140 PMCID: PMC7748442 DOI: 10.1093/rb/rbaa026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/17/2020] [Accepted: 05/15/2020] [Indexed: 12/11/2022] Open
Abstract
This study presents direct 2D and 3D co-culture model of mesenchymal stem cells (MSCs) line with chondrocytes isolated from patients with osteoarthritis (unaffected area). MSCs differentiation into chondrocytes after 14, 17 days was checked by estimation of collagen I, II, X, aggrecan expression using immunohistochemistry. Visualization, localization of cells on Hyaff-11 was performed using enzymatic technique and THUNDER Imaging Systems. Results showed, that MSCs/chondrocytes 3D co-culture induced suitable conditions for chondrocytes grow and MSCs differentiation than 2D monoculture. Despite that differentiated cells on Hyaff-11 expressed collagen X, they showed high collagen II (80%) and aggrecan (60%) expression with simultaneous decrease of collagen I expression (10%). The high concentration of differentiated cells on Hyaff-11, indicate that this structure has an impact on cells cooperation and communication. In conclusion, we suggest that high expression of collagen II and aggrecan in 3D co-culture model, indicate that cooperation between different subpopulations may have synergistic impact on MSCs chondrogenic potential. Revealed the high concentration and localization of cells growing in deeper layers of membrane in 3D co-culture, indicate that induced microenvironmental enhance cell migration within scaffold. Additionally, we suggest that co-culture model might be useful for construction a bioactive structure for cartilage tissue regeneration.
Collapse
Affiliation(s)
- Iwona Deszcz
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland
| | - Anna Lis-Nawara
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland
| | - Piotr Grelewski
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland
| | - Szymon Dragan
- Department and Clinic of Orthopedic and Traumatologic Surgery, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland
| | - Julia Bar
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland
| |
Collapse
|
10
|
Impact of Tranexamic Acid on Chondrocytes and Osteogenically Differentiated Human Mesenchymal Stromal Cells (hMSCs) In Vitro. J Clin Med 2020; 9:jcm9123880. [PMID: 33260331 PMCID: PMC7760070 DOI: 10.3390/jcm9123880] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
The topical application of tranexamic acid (TXA) helps to prevent post-operative blood loss in total joint replacements. Despite these findings, the effects on articular and periarticular tissues remain unclear. Therefore, this in vitro study examined the effects of varying exposure times and concentrations of TXA on proliferation rates, gene expression and differentiation capacity of chondrocytes and human mesenchymal stromal cells (hMSCs), which underwent osteogenic differentiation. Chondrocytes and hMSCs were isolated and multiplied in monolayer cell cultures. Osteogenic differentiation of hMSCs was induced for 21 days using a differentiation medium containing specific growth factors. Cell proliferation was analyzed using ATP assays. Effects of TXA on cell morphology were examined via light microscopy and histological staining, while expression levels of tissue-specific genes were measured using semiquantitative RT-PCR. After treatment with 50 mg/mL of TXA, a decrease in cell proliferation rates was observed. Furthermore, treatment with concentrations of 20 mg/mL of TXA for at least 48 h led to a visible detachment of chondrocytes. TXA treatment with 50 mg/mL for at least 24 h led to a decrease in the expression of specific marker genes in chondrocytes and osteogenically differentiated hMSCs. No significant effects were observed for concentrations beyond 20 mg/mL of TXA combined with exposure times of less than 24 h. This might therefore represent a safe limit for topical application in vivo. Further research regarding in vivo conditions and effects on hMSC functionality are necessary to fully determine the effects of TXA on articular and periarticular tissues.
Collapse
|
11
|
Aizad S, Zubairi SI, Yahaya BH, Lazim AM. Centella asiatica Extract Potentiates Anticancer Activity in an Improved 3-D PHBV-Composite-CMC A549 Lung Cancer Microenvironment Scaffold. ARABIAN JOURNAL FOR SCIENCE AND ENGINEERING 2020. [DOI: 10.1007/s13369-020-05072-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
12
|
Pathophysiological Perspective of Osteoarthritis. ACTA ACUST UNITED AC 2020; 56:medicina56110614. [PMID: 33207632 PMCID: PMC7696673 DOI: 10.3390/medicina56110614] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is the most well-known degenerative disease among the geriatric and is a main cause of significant disability in daily living. It has a multifactorial etiology and is characterized by pathological changes in the knee joint structure including cartilage erosion, synovial inflammation, and subchondral sclerosis with osteophyte formation. To date, no efficient treatment is capable of altering the pathological progression of OA, and current therapy is broadly divided into pharmacological and nonpharmacological measures prior to surgical intervention. In this review, the significant risk factors and mediators, such as cytokines, proteolytic enzymes, and nitric oxide, that trigger the loss of the normal homeostasis and structural changes in the articular cartilage during the progression of OA are described. As the understanding of the mechanisms underlying OA improves, treatments are being developed that target specific mediators thought to promote the cartilage destruction that results from imbalanced catabolic and anabolic activity in the joint.
Collapse
|
13
|
Tryfonidou MA, de Vries G, Hennink WE, Creemers LB. "Old Drugs, New Tricks" - Local controlled drug release systems for treatment of degenerative joint disease. Adv Drug Deliv Rev 2020; 160:170-185. [PMID: 33122086 DOI: 10.1016/j.addr.2020.10.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022]
Abstract
Osteoarthritis (OA) and chronic low back pain (CLBP) caused by intervertebral disc (IVD) degeneration are joint diseases that have become major causes for loss of quality of life worldwide. Despite the unmet need, effective treatments other than invasive, and often ineffective, surgery are lacking. Systemic administration of drugs entails suboptimal local drug exposure in the articular joint and IVD. This review provides an overview of the potency of biomaterial-based drug delivery systems as novel treatment modality, with a focus on the biological effects of drug release systems that have reached translation at the level of in vivo models and relevant ex vivo models. These studies have shown encouraging results of biomaterial-based local delivery of several types of drugs, mostly inhibitors of inflammatory cytokines or other degenerative factors. Prevention of inflammation and degeneration and pain relief was achieved, although mainly in small animal models, with interventions applied at an early disease stage. Less convincing data were obtained with the delivery of regenerative factors. Multidisciplinary efforts towards tackling the discord between in vitro and in vivo release, combined with adaptations in the regulatory landscape may be needed to enhance safe and expeditious introduction of more and more effective controlled release-based treatments with the OA and CLBP patients.
Collapse
|
14
|
Lehoczky G, Wolf F, Mumme M, Gehmert S, Miot S, Haug M, Jakob M, Martin I, Barbero A. Intra-individual comparison of human nasal chondrocytes and debrided knee chondrocytes: Relevance for engineering autologous cartilage grafts. Clin Hemorheol Microcirc 2020; 74:67-78. [PMID: 31743993 DOI: 10.3233/ch-199236] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Implantation of autologous chondrocytes for cartilage repair requires harvesting of undamaged cartilage, implying an additional joint arthroscopy surgery and further damage to the articular surface. As alternative possible cell sources, in this study we assessed the proliferation and chondrogenic capacity of debrided Knee Chondrocytes (dKC) and Nasal Chondrocytes (NC) collected from the same patients. METHODS Matched NC and dKC pairs from 13 patients enrolled in two clinical studies (NCT01605201 and NCT026739059) were expanded in monolayer and then chondro-differentiated in 3D collagenous scaffolds in medium with or without Transforming Growth Factor beta 1 (TGFβ1). Cell proliferation and amount of cartilage matrix production by these two cell types were assessed. RESULTS dKC exhibited an inferior proliferation rate than NC, and a lower capacity to chondro-differentiate. Resulting dKC-grafts contained lower amounts of cartilage specific matrix components glycosaminoglycans and type II collagen. The cartilage forming capacity of dKC did not significantly correlate with specific clinical parameters and was only partially improved by medium supplemention with TGFβ1. CONCLUSIONS dKC exhibit a reproducibly poor capacity to engineer cartilage grafts. Our in vitro data suggest that NC would be a better suitable cell source for the generation of autologous cartilage grafts.
Collapse
Affiliation(s)
- Gyözö Lehoczky
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Francine Wolf
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Marcus Mumme
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Orthopaedics, University Children's Hospital Basel, Basel, Switzerland
| | - Sebastian Gehmert
- Department of Orthopaedics, University Children's Hospital Basel, Basel, Switzerland
| | - Sylvie Miot
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Martin Haug
- Department of Surgery, University Hospital of Basel, Basel, Switzerland
| | - Marcel Jakob
- Department of Surgery, University Hospital of Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
15
|
Lubbers R, van Schaarenburg RA, Kwekkeboom JC, Levarht EWN, Bakker AM, Mahdad R, Monteagudo S, Cherifi C, Lories RJ, Toes REM, Ioan-Facsinay A, Trouw LA. Complement component C1q is produced by isolated articular chondrocytes. Osteoarthritis Cartilage 2020; 28:675-684. [PMID: 31634584 DOI: 10.1016/j.joca.2019.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 09/10/2019] [Accepted: 09/21/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Inflammation and innate immune responses may contribute to development and progression of Osteoarthritis (OA). Chondrocytes are the sole cell type of the articular cartilage and produce extracellular-matrix molecules. How inflammatory mediators reach chondrocytes is incompletely understood. Previous studies have shown that chondrocytes express mRNA encoding complement proteins such as C1q, suggesting local protein production, which has not been demonstrated conclusively. The aim of this study is to explore C1q production at the protein level by chondrocytes. DESIGN We analysed protein expression of C1q in freshly isolated and cultured human articular chondrocytes using Western blot, ELISA and flow cytometry. We examined changes in mRNA expression of collagen, MMP-1 and various complement genes upon stimulation with pro-inflammatory cytokines or C1q. mRNA expression of C1 genes was determined in articular mouse chondrocytes. RESULTS Primary human articular chondrocytes express genes encoding C1q, C1QA, C1QB, C1QC, and secrete C1q to the extracellular medium. Stimulation of chondrocytes with pro-inflammatory cytokines upregulated C1QA, C1QB, C1QC mRNA expression, although this was not confirmed at the protein level. Extracellular C1q bound to the chondrocyte surface dose dependently. In a pilot study, binding of C1q to chondrocytes resulted in changes in the expression of collagens with a decrease in collagen type 2 and an increase in type 10. Mouse articular chondrocytes also expressed C1QA, C1QB, C1QC, C1R and C1S at the mRNA level. CONCLUSIONS C1q protein can be expressed and secreted by human articular chondrocytes and is able to bind to chondrocytes influencing the relative collagen expression.
Collapse
Affiliation(s)
- R Lubbers
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands.
| | - R A van Schaarenburg
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands; Charles River, Leiden, the Netherlands
| | - J C Kwekkeboom
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - E W N Levarht
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - A M Bakker
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - R Mahdad
- Department of Orthopedic Surgery, Alrijne Hospital, Leiderdorp, the Netherlands
| | - S Monteagudo
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - C Cherifi
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - R J Lories
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium; Division of Rheumatology, University Hospitals Leuven, Belgium
| | - R E M Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - A Ioan-Facsinay
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - L A Trouw
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands; Department of Immunohematology and Blood Transfusion, Leiden University Medical, Center, Leiden, the Netherlands.
| |
Collapse
|
16
|
Li Z, Xiang S, Li EN, Fritch MR, Alexander PG, Lin H, Tuan RS. Tissue Engineering for Musculoskeletal Regeneration and Disease Modeling. Handb Exp Pharmacol 2020; 265:235-268. [PMID: 33471201 DOI: 10.1007/164_2020_377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Musculoskeletal injuries and associated conditions are the leading cause of physical disability worldwide. The concept of tissue engineering has opened up novel approaches to repair musculoskeletal defects in a fast and/or efficient manner. Biomaterials, cells, and signaling molecules constitute the tissue engineering triad. In the past 40 years, significant progress has been made in developing and optimizing all three components, but only a very limited number of technologies have been successfully translated into clinical applications. A major limiting factor of this barrier to translation is the insufficiency of two-dimensional cell cultures and traditional animal models in informing the safety and efficacy of in-human applications. In recent years, microphysiological systems, often referred to as organ or tissue chips, generated according to tissue engineering principles, have been proposed as the next-generation drug testing models. This chapter aims to first review the current tissue engineering-based approaches that are being applied to fabricate and develop the individual critical elements involved in musculoskeletal organ/tissue chips. We next highlight the general strategy of generating musculoskeletal tissue chips and their potential in future regenerative medicine research. Exemplary microphysiological systems mimicking musculoskeletal tissues are described. With sufficient physiological accuracy and relevance, the human cell-derived, three-dimensional, multi-tissue systems have been used to model a number of orthopedic disorders and to test new treatments. We anticipate that the novel emerging tissue chip technology will continually reshape and improve our understanding of human musculoskeletal pathophysiology, ultimately accelerating the development of advanced pharmaceutics and regenerative therapies.
Collapse
Affiliation(s)
- Zhong Li
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shiqi Xiang
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eileen N Li
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
| | - Madalyn R Fritch
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter G Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA.
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
17
|
Goderecci R, Giusti I, Necozione S, Cinque B, D’Ascenzo S, Dolo V, Calvisi V. Short exposure to tranexamic acid does not affect, in vitro, the viability of human chondrocytes. Eur J Med Res 2019; 24:15. [PMID: 30795796 PMCID: PMC6385467 DOI: 10.1186/s40001-019-0373-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/14/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Only few studies have investigated the effect of topical application of tranexamic acid (TXA) on "minimally" invasive joint surgical procedures in which articular cartilage is preserved; for this reason, actually many surgeons avoid the use of topical TXA even if the disadvantage related to a blood loss can occur. The aim of this study was to evaluate the cytotoxicity, on human chondrocytes, of TXA at different concentrations and times of exposure and the mechanisms of cell death. METHODS Experiments were carried out on isolated human chondrocytes harvested from eight patients who underwent total knee replacement. Cell viability was determined using XTT assay and was assessed at 0, 24 and 48 h intervals after a 10-min-long treatment, followed by thorough washes, or at 24 and 48 h of treatment at TXA concentrations of 20, 50, 70 and 100 mg/ml. Cell cycle alterations and occurrence of cell death for apoptosis or necrosis were assessed by cytofluorimetry. Data were analyzed using Proc Mixed Procedure; LSMEANS was used to compare multiple group means with Tukey's honestly significant difference test. RESULTS A significant correlation between the controlled for factors (type of treatment, time and concentration) was found in the performed experiment. No significant effect on cell viability was observed when the TXA exposure was limited to 10 min, while for increased exposure, 24 and 48 h, a remarkable reduction was found; cell death occurred by apoptosis and was already appreciable after 24 h, reaching a statistical significance after the 48-h-long treatment. CONCLUSION A prolonged exposure to TXA may cause cartilage damage, thus its topical application can be expanded also to clinical scenarios that include retention of native cartilage chondrocytes, only if it is limited to few minutes and used at concentrations of 70 mg/ml or less.
Collapse
Affiliation(s)
- Remo Goderecci
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi 1, Blocco 11, 67100 L’Aquila, Italy
| | - Ilaria Giusti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi 1, Blocco 11, 67100 L’Aquila, Italy
| | - Stefano Necozione
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi 1, Blocco 11, 67100 L’Aquila, Italy
| | - Benedetta Cinque
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi 1, Blocco 11, 67100 L’Aquila, Italy
| | - Sandra D’Ascenzo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi 1, Blocco 11, 67100 L’Aquila, Italy
| | - Vincenza Dolo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi 1, Blocco 11, 67100 L’Aquila, Italy
| | - Vittorio Calvisi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi 1, Blocco 11, 67100 L’Aquila, Italy
| |
Collapse
|
18
|
Kim M, Erickson IE, Huang AH, Garrity ST, Mauck RL, Steinberg DR. Donor Variation and Optimization of Human Mesenchymal Stem Cell Chondrogenesis in Hyaluronic Acid. Tissue Eng Part A 2018; 24:1693-1703. [PMID: 29792383 DOI: 10.1089/ten.tea.2017.0520] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are an attractive cell type for cartilage repair that can undergo chondrogenesis in a variety of three-dimensional (3D) scaffolds. Hyaluronic acid (HA) hydrogels provide a biologically relevant interface for cell encapsulation. While previous studies have shown that MSC-laden HA constructs can mature in vitro to match native mechanical properties using cells from animal sources, clinical application will depend on the successful translation of these findings to human cells. Though numerous studies have investigated chondrogenesis of human MSC (hMSC)-laden constructs, their functional outcomes were quite inferior to those using animal sources, and donor-specific responses to 3D HA hydrogels have not been fully investigated. To that end, hMSCs were derived from seven donors, and their ability to undergo chondrogenesis in pellet culture and HA hydrogels was evaluated. Given the initial observation of overt cell aggregation and/or gel contraction for some donors, the impact of variation in cell and HA macromer concentration on functional outcomes during chondrogenesis was evaluated using one young/healthy donor. The findings show marked differences in functional chondrogenesis of hMSCs in 3D HA hydrogels based on donor. Increasing cell density resulted in increased mechanical properties, but also promoted construct contraction. Increasing the macromer density generally stabilized construct dimensions and increased extracellular matrix production, but limited the distribution of formed matrix at the center of the construct and reduced mechanical properties. Collectively, these findings suggest that the use of hMSCs may require tuning of cell density and gel mechanics on a donor-by-donor basis to provide for the most robust tissue formation for clinical application.
Collapse
Affiliation(s)
- Minwook Kim
- 1 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,2 Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania , Philadelphia, Pennsylvania.,3 Translational Musculoskeletal Research Center (TMRC), Corporal Michael J. Crescenz Veterans Affairs Medical Center , Philadelphia, Pennsylvania
| | - Isaac E Erickson
- 1 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,4 DiscGenics, Inc. , Salt Lake City, Utah
| | - Alice H Huang
- 1 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,5 Department of Orthopaedics, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Sean T Garrity
- 1 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Robert L Mauck
- 1 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,2 Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania , Philadelphia, Pennsylvania.,3 Translational Musculoskeletal Research Center (TMRC), Corporal Michael J. Crescenz Veterans Affairs Medical Center , Philadelphia, Pennsylvania
| | - David R Steinberg
- 1 McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,3 Translational Musculoskeletal Research Center (TMRC), Corporal Michael J. Crescenz Veterans Affairs Medical Center , Philadelphia, Pennsylvania
| |
Collapse
|
19
|
Galuzzi M, Perteghella S, Antonioli B, Tosca MC, Bari E, Tripodo G, Sorrenti M, Catenacci L, Mastracci L, Grillo F, Marazzi M, Torre ML. Human Engineered Cartilage and Decellularized Matrix as an Alternative to Animal Osteoarthritis Model. Polymers (Basel) 2018; 10:738. [PMID: 30960663 PMCID: PMC6403588 DOI: 10.3390/polym10070738] [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: 06/01/2018] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 01/05/2023] Open
Abstract
(1) Objective: to obtain a reproducible, robust, well-defined, and cost-affordable in vitro model of human cartilage degeneration, suitable for drug screening; (2) Methods: we proposed 3D models of engineered cartilage, considering two human chondrocyte sources (articular/nasal) and five culture methods (pellet, alginate beads, silk/alginate microcarriers, and decellularized cartilage). Engineered cartilages were treated with pro-inflammatory cytokine IL-1β to promote cartilage degradation; (3) Results: articular chondrocytes have been rejected since they exhibit low cellular doubling with respect to nasal cells, with longer culture time for cell expansion; furthermore, pellet and alginate bead cultures lead to insufficient cartilage matrix production. Decellularized cartilage resulted as good support for degeneration model, but long culture time and high cell amount are required to obtain the adequate scaffold colonization. Here, we proposed, for the first time, the combined use of decellularized cartilage, as aggrecanase substrate, with pellet, alginate beads, or silk/alginate microcarriers, as polymeric scaffolds for chondrocyte cultures. This approach enables the development of suitable models of cartilaginous pathology. The results obtained after cryopreservation also demonstrated that beads and microcarriers are able to preserve chondrocyte functionality and metabolic activity; (4) Conclusions: alginate and silk/alginate-based scaffolds can be easily produced and cryopreserved to obtain a cost-affordable and ready-to-use polymer-based product for the subsequent screening of anti-inflammatory drugs for cartilage diseases.
Collapse
Affiliation(s)
- Marta Galuzzi
- Tissue Therapy Unit, ASST Niguarda Hospital, Piazza Ospedale Maggiore 3, 20162 Milan, Italy.
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
- PharmaExceed S.r.l., 27100 Pavia, Italy.
| | - Barbara Antonioli
- Tissue Therapy Unit, ASST Niguarda Hospital, Piazza Ospedale Maggiore 3, 20162 Milan, Italy.
| | - Marta Cecilia Tosca
- Tissue Therapy Unit, ASST Niguarda Hospital, Piazza Ospedale Maggiore 3, 20162 Milan, Italy.
| | - Elia Bari
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Giuseppe Tripodo
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Milena Sorrenti
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Laura Catenacci
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Luca Mastracci
- Section of Histopathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), IRCCS San Martino IST Hospital, University of Genoa, Largo R. Benzi 8, 16121 Genoa, Italy.
| | - Federica Grillo
- Section of Histopathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), IRCCS San Martino IST Hospital, University of Genoa, Largo R. Benzi 8, 16121 Genoa, Italy.
| | - Mario Marazzi
- Tissue Therapy Unit, ASST Niguarda Hospital, Piazza Ospedale Maggiore 3, 20162 Milan, Italy.
| | - Maria Luisa Torre
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
- PharmaExceed S.r.l., 27100 Pavia, Italy.
| |
Collapse
|
20
|
Khurshid M, Mulet-Sierra A, Adesida A, Sen A. Osteoarthritic human chondrocytes proliferate in 3D co-culture with mesenchymal stem cells in suspension bioreactors. J Tissue Eng Regen Med 2017; 12:e1418-e1432. [PMID: 28752579 DOI: 10.1002/term.2531] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/20/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a painful disease, characterized by progressive surface erosion of articular cartilage. The use of human articular chondrocytes (hACs) sourced from OA patients has been proposed as a potential therapy for cartilage repair, but this approach is limited by the lack of scalable methods to produce clinically relevant quantities of cartilage-generating cells. Previous studies in static culture have shown that hACs co-cultured with human mesenchymal stem cells (hMSCs) as 3D pellets can upregulate proliferation and generate neocartilage with enhanced functional matrix formation relative to that produced from either cell type alone. However, because static culture flasks are not readily amenable to scale up, scalable suspension bioreactors were investigated to determine if they could support the co-culture of hMSCs and OA hACs under serum-free conditions to facilitate clinical translation of this approach. When hACs and hMSCs (1:3 ratio) were inoculated at 20,000 cells/ml into 125-ml suspension bioreactors and fed weekly, they spontaneously formed 3D aggregates and proliferated, resulting in a 4.75-fold increase over 16 days. Whereas the apparent growth rate was lower than that achieved during co-culture as a 2D monolayer in static culture flasks, bioreactor co-culture as 3D aggregates resulted in a significantly lower collagen I to II mRNA expression ratio and more than double the glycosaminoglycan/DNA content (5.8 vs. 2.5 μg/μg). The proliferation of hMSCs and hACs as 3D aggregates in serum-free suspension culture demonstrates that scalable bioreactors represent an accessible platform capable of supporting the generation of clinical quantities of cells for use in cell-based cartilage repair.
Collapse
Affiliation(s)
- Madiha Khurshid
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Canada.,McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Aillette Mulet-Sierra
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Adetola Adesida
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Arindom Sen
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Canada.,McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
21
|
Mayer N, Lopa S, Talò G, Lovati AB, Pasdeloup M, Riboldi SA, Moretti M, Mallein-Gerin F. Interstitial Perfusion Culture with Specific Soluble Factors Inhibits Type I Collagen Production from Human Osteoarthritic Chondrocytes in Clinical-Grade Collagen Sponges. PLoS One 2016; 11:e0161479. [PMID: 27584727 PMCID: PMC5008682 DOI: 10.1371/journal.pone.0161479] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/06/2016] [Indexed: 01/17/2023] Open
Abstract
Articular cartilage has poor healing ability and cartilage injuries often evolve to osteoarthritis. Cell-based strategies aiming to engineer cartilaginous tissue through the combination of biocompatible scaffolds and articular chondrocytes represent an alternative to standard surgical techniques. In this context, perfusion bioreactors have been introduced to enhance cellular access to oxygen and nutrients, hence overcoming the limitations of static culture and improving matrix deposition. Here, we combined an optimized cocktail of soluble factors, the BIT (BMP-2, Insulin, Thyroxin), and clinical-grade collagen sponges with a bidirectional perfusion bioreactor, namely the oscillating perfusion bioreactor (OPB), to engineer in vitro articular cartilage by human articular chondrocytes (HACs) obtained from osteoarthritic patients. After amplification, HACs were seeded and cultivated in collagen sponges either in static or dynamic conditions. Chondrocyte phenotype and the nature of the matrix synthesized by HACs were assessed using western blotting and immunohistochemistry analyses. Finally, the stability of the cartilaginous tissue produced by HACs was evaluated in vivo by subcutaneous implantation in nude mice. Our results showed that perfusion improved the distribution and quality of cartilaginous matrix deposited within the sponges, compared to static conditions. Specifically, dynamic culture in the OPB, in combination with the BIT cocktail, resulted in the homogeneous production of extracellular matrix rich in type II collagen. Remarkably, the production of type I collagen, a marker of fibrous tissues, was also inhibited, indicating that the association of the OPB with the BIT cocktail limits fibrocartilage formation, favoring the reconstruction of hyaline cartilage.
Collapse
Affiliation(s)
- Nathalie Mayer
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, Université Claude Bernard-Lyon 1 and University of Lyon, Institute for Biology and Chemistry of Proteins, Lyon, France
| | - Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Giuseppe Talò
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Arianna B. Lovati
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - Marielle Pasdeloup
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, Université Claude Bernard-Lyon 1 and University of Lyon, Institute for Biology and Chemistry of Proteins, Lyon, France
| | | | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
- Swiss Institute of Regenerative Medicine (SIRM), Lugano, Switzerland
- Fondazione Cardiocentro Ticino, Lugano, Switzerland
| | - Frédéric Mallein-Gerin
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, Université Claude Bernard-Lyon 1 and University of Lyon, Institute for Biology and Chemistry of Proteins, Lyon, France
- * E-mail:
| |
Collapse
|
22
|
Bauer C, Berger M, Baumgartner RR, Höller S, Zwickl H, Niculescu-Morzsa E, Halbwirth F, Nehrer S. A Novel Cross-Linked Hyaluronic Acid Porous Scaffold for Cartilage Repair: An In Vitro Study With Osteoarthritic Chondrocytes. Cartilage 2016; 7:265-73. [PMID: 27375842 PMCID: PMC4918062 DOI: 10.1177/1947603515611949] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
PURPOSE An important feature of biomaterials used in cartilage regeneration is their influence on the establishment and stabilization of a chondrocytic phenotype of embedded cells. The purpose of this study was to examine the effects of a porous 3-dimensional scaffold made of cross-linked hyaluronic acid on the expression and synthesis performance of human articular chondrocytes. MATERIALS AND METHODS Osteoarthritic chondrocytes from 5 patients with a mean age of 74 years were passaged twice and cultured within the cross-linked hyaluronic acid scaffolds for 2 weeks. Analyses were performed at 3 different time points. For estimation of cell content within the scaffold, DNA-content (CyQuant cell proliferation assay) was determined. The expression of chondrocyte-specific genes by embedded cells as well as the total amount of sulfated glycosaminoglycans produced during the culture period was analyzed in order to characterize the synthesis performance and differentiation status of the cells. RESULTS Cells showed a homogenous distribution within the scaffold. DNA quantification revealed a reduction of the cell number. This might be attributed to loss of cells from the scaffold during media exchange connected with a stop in cell proliferation. Indeed, the expression of cartilage-specific genes and the production of sulfated glycosaminoglycans were increased and the differentiation index was clearly improved. CONCLUSIONS These results suggest that the attachment of osteoarthritic P2 chondrocytes to the investigated material enhanced the chondrogenic phenotype as well as promoted the retention.
Collapse
Affiliation(s)
- Christoph Bauer
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria,Christoph Bauer, Center for Regenerative Medicine and Orthopedics, Danube University Krems, Dr.-Karl-Dorrek-Strasse 30, Krems, 3500, Austria.
| | - Manuela Berger
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria
| | | | | | - Hannes Zwickl
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria
| | - Eugenia Niculescu-Morzsa
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria
| | - Florian Halbwirth
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria
| | - Stefan Nehrer
- Center for Regenerative Medicine and Orthopedics, Department for Health Sciences and Biomedicine, Danube University, Krems, Austria
| |
Collapse
|
23
|
Charlier E, Malaise O, Zeddou M, Neuville S, Cobraiville G, Deroyer C, Sanchez C, Gillet P, Kurth W, de Seny D, Relic B, Malaise MG. Restriction of spontaneous and prednisolone-induced leptin production to dedifferentiated state in human hip OA chondrocytes: role of Smad1 and β-catenin activation. Osteoarthritis Cartilage 2016; 24:315-24. [PMID: 26318657 DOI: 10.1016/j.joca.2015.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aetiology of OA is not fully understood although several adipokines such as leptin are known mediators of disease progression. Since leptin levels were increased in synovial fluid compared to serum in OA patients, it was suggested that joint cells themselves could produce leptin. However, exact mechanisms underlying leptin production by chondrocytes are poorly understood. Nevertheless, prednisolone, although displaying powerful anti-inflammatory properties has been recently reported to be potent stimulator of leptin and its receptor in OA synovial fibroblasts. Therefore, we investigated, in vitro, spontaneous and prednisolone-induced leptin production in OA chondrocytes, focusing on transforming growth factor-β (TGFβ) and Wnt/β-catenin pathways. DESIGN We used an in vitro dedifferentiation model, comparing human freshly isolated hip OA chondrocytes cultivated in monolayer during 1 day (type II, COL2A1 +; type X, COL10A1 + and type I collagen, COL1A1 -) or 14 days (COL2A1 -; COL10A1 - and COL1A1+). RESULTS Leptin expression was not detected in day1 OA chondrocytes whereas day14 OA chondrocytes produced leptin, significantly increased with prednisolone. Activin receptor-like kinase 1 (ALK1)/ALK5 ratio was shifted during dedifferentiation, from high ALK5 and phospho (p)-Smad2 expression at day1 to high ALK1, endoglin and p-Smad1/5 expression at day14. Moreover, inactive glycogen synthase kinase 3 (GSK3) and active β-catenin were only found in dedifferentiated OA chondrocytes. Smad1 and β-catenin but not endoglin stable lentiviral silencing led to a significant decrease in leptin production by dedifferentiated OA chondrocytes. CONCLUSIONS Only dedifferentiated OA chondrocytes produced leptin. Prednisolone markedly enhanced leptin production, which involved Smad1 and β-catenin activation.
Collapse
Affiliation(s)
- E Charlier
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium.
| | - O Malaise
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - M Zeddou
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - S Neuville
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - G Cobraiville
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - C Deroyer
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - C Sanchez
- Bone and Cartilage Research Unit, Arthropole Liège, University of Liège, Belgium
| | - P Gillet
- Orthopedic Surgery Unit, CHU of Liège, Belgium
| | - W Kurth
- Orthopedic Surgery Unit, CHU of Liège, Belgium
| | - D de Seny
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - B Relic
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| | - M G Malaise
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, Arthropole Liège, University of Liège, Belgium
| |
Collapse
|
24
|
Yang HY, van Ee RJ, Timmer K, Craenmehr EG, Huang JH, Öner FC, Dhert WJ, Kragten AH, Willems N, Grinwis GC, Tryfonidou MA, Papen-Botterhuis NE, Creemers LB. A novel injectable thermoresponsive and cytocompatible gel of poly(N-isopropylacrylamide) with layered double hydroxides facilitates siRNA delivery into chondrocytes in 3D culture. Acta Biomater 2015; 23:214-228. [PMID: 26022968 DOI: 10.1016/j.actbio.2015.05.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 05/14/2015] [Accepted: 05/18/2015] [Indexed: 01/12/2023]
Abstract
Hybrid hydrogels composed of poly(N-isopropylacrylamide) (pNIPAAM) and layered double hydroxides (LDHs) are presented in this study as novel injectable and thermoresponsive materials for siRNA delivery, which could specifically target several negative regulators of tissue homeostasis in cartilaginous tissues. Effectiveness of siRNA transfection using pNIPAAM formulated with either MgAl-LDH or MgFe-LDH platelets was investigated using osteoarthritic chondrocytes. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as an endogenous model gene to evaluate the extent of silencing. No significant adverse effects of pNIPAAM/LDH hydrogels on cell viability were noticed. Cellular uptake of fluorescently labeled siRNA was greatly enhanced (>75%) in pNIPAAM/LDH hydrogel constructs compared to alginate, hyaluronan and fibrin gels, and was absent in pNIPAAM hydrogel without LDH platelets. When using siRNA against GAPDH, 82-98% reduction of gene expression was found in both types of pNIPAAM/LDH hydrogel constructs after 6 days of culturing. In the pNIPAAM/MgAl-LDH hybrid hydrogel, 80-95% of GAPDH enzyme activity was reduced in parallel with gene. Our findings show that the combination of a cytocompatible hydrogel and therapeutic RNA oligonucleotides is feasible. Thus it might hold promise in treating degeneration of cartilaginous tissues by providing supporting scaffolds for cells and interference with locally produced degenerative factors.
Collapse
|
25
|
Chondrogenic capability of osteoarthritic chondrocytes from the trapeziometacarpal and hip joints. Cell Tissue Bank 2015; 17:171-7. [PMID: 26150189 DOI: 10.1007/s10561-015-9519-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/22/2015] [Indexed: 10/23/2022]
Abstract
Osteoarthritis is the most common degenerative disease of joints like the hip and the trapeziometacarpal joint (rhizarthrosis). In this in vitro study, we compared the chondrogenesis of chondrocytes derived from the trapezium and the femoral head cartilage of osteoarthritic patients to have a deeper insight on trapezium chondrocyte behavior as autologous cell source for the repair of cartilage lesions in rhizarthrosis. Chondrocytes collected from trapezium and femoral head articular cartilage were cultured in pellets and analyzed for chondrogenic differentiation, cell proliferation, glycosaminoglycan production, gene expression of chondrogenic and fibrous markers, histological and immunohistochemical analyses. Our results showed a higher cartilaginous matrix deposition and a lower fibrocartilaginous phenotype of the femoral chondrocytes with respect to the trapezium chondrocytes assessed by a higher absolute glycosaminoglycan and type II collagen production, thus demonstrating a superior chondrogenic potential of the femoral with respect to the trapezium chondrocytes. The differences in chondrogenic potential between trapezium and femoral head chondrocytes confirmed a lower regenerative capability in the trapezium than in the femoral head cartilage due to the different environment and loading acting on these joints that affects the metabolism of the resident cells. This could represent a limitation to apply the cell therapy for rhizoarthrosis.
Collapse
|
26
|
Oh TI, Kim C, Karki B, Son Y, Lee E, Woo EJ. Non-destructive label-free continuous monitoring of in vitro chondrogenesis via electrical conductivity and its anisotropy. Biotechnol Bioeng 2014; 112:422-7. [DOI: 10.1002/bit.25350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/09/2014] [Accepted: 07/17/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Tong In Oh
- Department of Biomedical Engineering; Kyung Hee University; Gyeonggi-do Korea
| | - Changhwan Kim
- College of Life Science and Graduate School of Biotechnology; Kyung Hee University; 1732 Deogyeong-daero Giheung-gu Gyeonggi-do 446-701 Korea
| | - Bishal Karki
- Department of Biomedical Engineering; Kyung Hee University; Gyeonggi-do Korea
| | - Youngsook Son
- College of Life Science and Graduate School of Biotechnology; Kyung Hee University; 1732 Deogyeong-daero Giheung-gu Gyeonggi-do 446-701 Korea
| | - EunAh Lee
- College of Life Science and Graduate School of Biotechnology; Kyung Hee University; 1732 Deogyeong-daero Giheung-gu Gyeonggi-do 446-701 Korea
| | - Eung Je Woo
- Department of Biomedical Engineering; Kyung Hee University; Gyeonggi-do Korea
| |
Collapse
|
27
|
Yhee JY, Son S, Kim SH, Park K, Choi K, Kwon IC. Self-assembled glycol chitosan nanoparticles for disease-specific theranostics. J Control Release 2014; 193:202-13. [PMID: 24845129 DOI: 10.1016/j.jconrel.2014.05.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/29/2014] [Accepted: 05/07/2014] [Indexed: 01/22/2023]
Abstract
Hydrophobically modified glycol chitosan (hGC) conjugates spontaneously form self-assembled nanoparticles (NPs) in aqueous conditions, and glycol chitosan NPs (CNPs) have been extensively studied for the past few decades. For disease-specific theranostics, CNPs could be simply modified with imaging agents, and the hydrophobic domains of hGC are available for encapsulation of various drugs. Based on the excellent physiochemical and biological properties, CNPs have been investigated for multimodal imaging and target specific drug delivery. In particular, a recent application of CNPs has shown great potential as an efficient theranostic system because the CNPs could be utilized for a disease-specific theranostic delivery system of different imaging agents and therapeutics, simultaneously. Furthermore, various therapeutic agents including chemo-drugs, nucleotides, peptides, and photodynamic chemicals could be simply encapsulated into the CNPs through hydrophobic or charge-charge interactions. Under in vivo conditions, the encapsulated imaging agents and therapeutic drugs have been successfully delivered to targeted diseases. In this article, the overall research progress on CNPs is reviewed from early works. The current challenges of CNPs to overcome in theranostics are also discussed, and continuous studies would provide more opportunities for early diagnosis of diseases and personalized medicine.
Collapse
Affiliation(s)
- Ji Young Yhee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea
| | - Sohee Son
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea
| | - Kinam Park
- Purdue University, Departments of Biomedical Engineering and Pharmaceutics, West Lafayette, IN 47907, USA
| | - Kuiwon Choi
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea.
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea; KU-KIST School, Korea University, 1 Anam-dong, Seongbuk-gu, Seoul 136-701, South Korea.
| |
Collapse
|
28
|
The role of changes in extracellular matrix of cartilage in the presence of inflammation on the pathology of osteoarthritis. BIOMED RESEARCH INTERNATIONAL 2013; 2013:284873. [PMID: 24069595 PMCID: PMC3771246 DOI: 10.1155/2013/284873] [Citation(s) in RCA: 354] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/27/2013] [Accepted: 07/29/2013] [Indexed: 01/13/2023]
Abstract
Osteoarthritis (OA) is a degenerative disease that affects various tissues surrounding joints such as articular cartilage, subchondral bone, synovial membrane, and ligaments. No therapy is currently available to completely prevent the initiation or progression of the disease partly due to poor understanding of the mechanisms of the disease pathology. Cartilage is the main tissue afflicted by OA, and chondrocytes, the sole cellular component in the tissue, actively participate in the degeneration process. Multiple factors affect the development and progression of OA including inflammation that is sustained during the progression of the disease and alteration in biomechanical conditions due to wear and tear or trauma in cartilage. During the progression of OA, extracellular matrix (ECM) of cartilage is actively remodeled by chondrocytes under inflammatory conditions. This alteration of ECM, in turn, changes the biomechanical environment of chondrocytes, which further drives the progression of the disease in the presence of inflammation. The changes in ECM composition and structure also prevent participation of mesenchymal stem cells in the repair process by inhibiting their chondrogenic differentiation. This review focuses on how inflammation-induced ECM remodeling disturbs cellular activities to prevent self-regeneration of cartilage in the pathology of OA.
Collapse
|
29
|
Ono Y, Sakai T, Hiraiwa H, Hamada T, Omachi T, Nakashima M, Ishizuka S, Matsukawa T, Knudson W, Knudson CB, Ishiguro N. Chondrogenic capacity and alterations in hyaluronan synthesis of cultured human osteoarthritic chondrocytes. Biochem Biophys Res Commun 2013; 435:733-9. [PMID: 23702485 DOI: 10.1016/j.bbrc.2013.05.052] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 05/12/2013] [Indexed: 12/20/2022]
Abstract
During osteoarthritis there is a disruption and loss of the extracellular matrix of joint cartilage, composed primarily of type II collagen, aggrecan and hyaluronan. In young patients, autologous chondrocyte implantation can be used to repair cartilage defects. However, for more elderly patients with osteoarthritis, such a repair approach is contraindicated because the procedure requires a large expansion of autologous chondrocytes in vitro leading a rapid, perhaps irreversible, loss of the chondrocyte phenotype. This study investigates whether osteoarthritic chondrocytes obtained from older patients can be expanded in vitro and moreover, induced to re-activate their chondrocyte phenotype. A decrease in chondrocyte phenotype markers, collagen II, aggrecan and SOX9 mRNA was observed with successive expansion of cells in monolayer culture. However, chondrogenic induction in three-dimensional pellet culture successfully rescued the expression of all three marker genes to native levels, even with 4th passage cells-cells representing an approximate 625-fold expansion in cell number. This data supports the use of osteoarthritic cells for autologous implantation repair. In addition, another set of gene products were explored as useful markers of the chondrocyte phenotype. Differentiated primary chondrocytes exhibited a common pattern of hyaluronan synthase isoforms that changed upon cell expansion in vitro and, reverted back to the original pattern following pellet culture. Moreover, the change in isoform pattern correlated with changes in the molecular size of synthesized hyaluronan.
Collapse
Affiliation(s)
- Yohei Ono
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Tissue engineering (TE) has promise as a biological solution and a disease modifying treatment for arthritis. Although cartilage can be generated by TE, substantial inter- and intra-donor variability makes it impossible to guarantee optimal, reproducible results. TE cartilage must be able to perform the functions of native tissue, thus mechanical and biological properties approaching those of native cartilage are likely a pre-requisite for successful implantation. A quality-control assessment of these properties should be part of the implantation release criteria for TE cartilage. Release criteria should certify that selected tissue properties have reached certain target ranges, and should be predictive of the likelihood of success of an implant in vivo. Unfortunately, it is not currently known which properties are needed to establish release criteria, nor how close one has to be to the properties of native cartilage to achieve success. Achieving properties approaching those of native cartilage requires a clear understanding of the target properties and reproducible assessment methodology. Here, we review several main aspects of quality control as it applies to TE cartilage. This includes a look at known mechanical and biological properties of native cartilage, which should be the target in engineered tissues. We also present an overview of the state of the art of tissue assessment, focusing on native articular and TE cartilage. Finally, we review the arguments for developing and validating non-destructive testing methods for assessing TE products.
Collapse
Affiliation(s)
- Joseph M. Mansour
- Skeletal Research Center, Department of Biology Case Western Reserve University Cleveland, OH, 44106
| | - Jean F. Welter
- Skeletal Research Center, Department of Biology Case Western Reserve University Cleveland, OH, 44106
| |
Collapse
|
31
|
TGFβ inhibition during expansion phase increases the chondrogenic re-differentiation capacity of human articular chondrocytes. Osteoarthritis Cartilage 2012; 20:1152-60. [PMID: 22772045 DOI: 10.1016/j.joca.2012.06.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 05/30/2012] [Accepted: 06/21/2012] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Autologous chondrocyte implantation is a cell-based treatment to repair articular cartilage defects, relying on the availability of expanded (de-differentiated) chondrocytes. Unfortunately, the expansion process causes several phenotypical changes, requiring re-establishment of the native chondrogenic phenotype to sustain proper repair. Among other proteins, transforming growth factor-β (TGFβ) is known to influence the chondrogenic re-differentiation of human articular chondrocytes (HACs) and their matrix deposition. Thus we investigated the effects of TGFβ-depletion during the expansion phase. DESIGN HACs were isolated from articular cartilage and expanded in the canonical serum-supplemented medium [fetal calf serum (FCS)] or in a chemically-defined (CD) medium, with or without anti-TGFβ antibody administration. The re-differentiation potential of the cells was assessed by pellet cultures, gene expression analysis and histology. RESULTS Cell proliferation proceeded more rapidly in CD-medium than in FCS-medium; it was not affected by the use of anti-TGFβ antibody but was further increased by addition of exogenous TGFβ1, via increased p-Smad1/5/8. Conversely, in FCS-medium, addition of anti-TGFβ antibody decreased both proliferation and p-Smad1/5/8 level. Challenging either FCS- or CD-medium with anti-TGFβ antibody during expansion enhanced chondrogenesis in the subsequent pellet cultures. Moreover, TGFβ-depletion during expansion in CD-medium inhibited mRNA expression of hypertrophic markers, collagen type-X (COL10) and matrix metalloproteinase-13 (MMP-13). Interestingly, the TGFβ1 level detected by enzyme-linked immunosorbent sandwich assay (ELISA) during cell expansion was correlated with COL10 mRNA expression after re-differentiation. CONCLUSION TGFβ-depletion during expansion improves the re-differentiation capacity of chondrocytes and inhibits hypertrophy. These results indicate the importance of the expansion medium composition to improve chondrogenic re-differentiation and to inhibit hypertrophy.
Collapse
|
32
|
Caron MMJ, Emans PJ, Coolsen MME, Voss L, Surtel DAM, Cremers A, van Rhijn LW, Welting TJM. Redifferentiation of dedifferentiated human articular chondrocytes: comparison of 2D and 3D cultures. Osteoarthritis Cartilage 2012; 20:1170-8. [PMID: 22796508 DOI: 10.1016/j.joca.2012.06.016] [Citation(s) in RCA: 350] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 06/07/2012] [Accepted: 06/30/2012] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Three-dimensional (3D) cultures are widely used to redifferentiate chondrocytes. However, the rationale behind the choice for 3D above two-dimensional (2D) cultures is poorly systematically investigated and mainly based on mRNA expression and glycosaminoglycan (GAG) content. The objective was to determine the differential redifferentiation characteristics of human articular chondrocytes (HACs) in monolayer, alginate beads and pellet culture by investigating mRNA expression, protein expression, GAG content and cell proliferation. DESIGN Dedifferentiated HACs from six individuals were redifferentiated in identical medium conditions for 7 days in monolayer, alginate beads or pellet culture. Read-out parameters were expression of chondrogenic and hypertrophic mRNAs and proteins, GAG content and cell proliferation. RESULTS 3D cultures specifically expressed chondrogenic mRNAs [collagen type II (COL2A1), SRY (sex determining region Y)-box 9 (SOX9), aggrecan (ACAN)), whereas 2D cultures did not. Hypertrophic mRNAs (collagen type X (COL10A1), runt-related transcription factor 2 (RUNX2), matrix metalloproteinase 13 (MMP13), vascular endothelial growth factor A (VEGFA), osteopontin (OPN), alkaline phosphatase (ALP)) were highly increased in 2D cultures and lower in 3D cultures. Collagen type I (COL1A1) mRNA expression was highest in 3D cultures. Protein expression supports most of the mRNA data, although an important discrepancy was found between mRNA and protein expression of COL2A1 and SOX9 in monolayer culture, stressing on the importance of protein expression analysis. GAG content was highest in 3D cultures, whereas chondrocyte proliferation was almost specific for 2D cultures. CONCLUSIONS For redifferentiation of dedifferentiated HACs, 3D cultures exhibit the most potent chondrogenic potential, whereas a hypertrophic phenotype is best achieved in 2D cultures. This is the first human study that systematically evaluates the differences between proliferation, GAG content, protein expression and mRNA expression of commonly used 2D and 3D chondrocyte culture techniques.
Collapse
Affiliation(s)
- M M J Caron
- Department of Orthopaedic Surgery, CAPHRI School for Public Health and Primary Care, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Freyria AM, Mallein-Gerin F. Chondrocytes or adult stem cells for cartilage repair: the indisputable role of growth factors. Injury 2012; 43:259-65. [PMID: 21696723 DOI: 10.1016/j.injury.2011.05.035] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 05/25/2011] [Indexed: 02/02/2023]
Abstract
Articular cartilage is easily injured but difficult to repair and cell therapies are proposed as tools to regenerate the defects in the tissue. Both differentiated chondrocytes and adult mesenchymal stem cells (MSCs) are regarded as cells potentially able to restore a functional cartilage. However, it is a complex process from the cell level to the tissue end product, during which growth factors play important roles from cell proliferation, extracellular matrix synthesis, maintenance of the phenotype to induction of MSCs towards chondrogenesis. Members of the TGF-β superfamily, are especially important in fulfilling these roles. Depending on the cell type chosen to restore cartilage, the effect of growth factors will vary. In this review, the roles of these factors in the maintenance of the chondrocyte phenotype are discussed and compared with those of factors involved in the repair of cartilage defects, using chondrocytes or adult mesenchymal stem cells.
Collapse
Affiliation(s)
- Anne-Marie Freyria
- Cartilage Biology and Engineering Group, IBCP, Université Lyon 1, Univ Lyon, CNRS FRE 3310, IFR128, France.
| | | |
Collapse
|
34
|
Egli RJ, Wernike E, Grad S, Luginbühl R. Physiological cartilage tissue engineering effect of oxygen and biomechanics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 289:37-87. [PMID: 21749898 DOI: 10.1016/b978-0-12-386039-2.00002-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In vitro engineering of cartilaginous tissues has been studied for many years, and tissue-engineered constructs are sought to be used clinically for treating articular cartilage defects. Even though there is a plethora of studies and data available, no breakthroughs have been achieved yet that allow for implanting in vivo cultured articular cartilaginous tissues in patients. A review of contributions to cartilage tissue engineering over the past decades emphasizes that most of the studies were performed under environmental conditions neglecting the physiological situation. This is specifically pronounced in the use of bioreactor systems which neither allow for application of near physiomechanical stimulations nor for controlling a hypoxic environment as it is experienced in synovial joints. It is suspected that the negligence of these important parameters has slowed down progress and prevented major breakthroughs in the field. This review focuses on the main aspects of cartilage tissue engineering with emphasis on the relation and understanding of employing physiological conditions.
Collapse
|
35
|
Acharya C, Adesida A, Zajac P, Mumme M, Riesle J, Martin I, Barbero A. Enhanced chondrocyte proliferation and mesenchymal stromal cells chondrogenesis in coculture pellets mediate improved cartilage formation. J Cell Physiol 2011; 227:88-97. [PMID: 22025108 DOI: 10.1002/jcp.22706] [Citation(s) in RCA: 187] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In this study, we aimed at investigating the interactions between primary chondrocytes and mesenchymal stem/stromal cells (MSC) accounting for improved chondrogenesis in coculture systems. Expanded MSC from human bone marrow (BM-MSC) or adipose tissue (AT-MSC) were cultured in pellets alone (monoculture) or with primary human chondrocytes from articular (AC) or nasal (NC) cartilage (coculture). In order to determine the reached cell number and phenotype, selected pellets were generated by combining: (i) human BM-MSC with bovine AC, (ii) BM-MSC from HLA-A2+ with AC from HLA-A2- donors, or (iii) human green fluorescent protein transduced BM-MSC with AC. Human BM-MSC and AC were also cultured separately in transwells. Resulting tissues and/or isolated cells were assessed immunohistologically, biochemically, cytofluorimetrically, and by RT-PCR. Coculture of NC or AC (25%) with BM-MSC or AT-MSC (75%) in pellets resulted in up to 1.6-fold higher glycosaminoglycan content than what would be expected based on the relative percentages of the different cell types. This effect was not observed in the transwell model. BM-MSC decreased in number (about fivefold) over time and, if cocultured with chondrocytes, increased type II collagen and decreased type X collagen expression. Instead, AC increased in number (4.2-fold) if cocultured with BM-MSC and maintained a differentiated phenotype. Chondro-induction in MSC-chondrocyte coculture is a robust process mediated by two concomitant effects: MSC-induced chondrocyte proliferation and chondrocyte-enhanced MSC chondrogenesis. The identified interactions between progenitor and mature cell populations may lead to the efficient use of freshly harvested chondrocytes for ex vivo cartilage engineering or in situ cartilage repair.
Collapse
|
36
|
Martinez-Sanchez A, Dudek KA, Murphy CL. Regulation of human chondrocyte function through direct inhibition of cartilage master regulator SOX9 by microRNA-145 (miRNA-145). J Biol Chem 2011; 287:916-24. [PMID: 22102413 DOI: 10.1074/jbc.m111.302430] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Articular cartilage enables weight bearing and near friction-free movement in the joints. Critical to its function is the production of a specialized, mechanocompetent extracellular matrix controlled by master regulator transcription factor SOX9. Mutations in SOX9 cause campomelic dysplasia, a haploinsufficiency disorder resulting in severe skeletal defects and dwarfism. Although much is understood about how SOX9 regulates cartilage matrix synthesis and hence joint function, how this master regulator is itself regulated remains largely unknown. Here we identify a specific microRNA, miR-145, as a direct regulator of SOX9 in normal healthy human articular chondrocytes. We show that miR-145 directly represses SOX9 expression in human cells through a unique binding site in its 3'-UTR not conserved in mice. Modulation of miR-145 induced profound changes in the human chondrocyte phenotype. Specifically, increased miR-145 levels cause greatly reduced expression of critical cartilage extracellular matrix genes (COL2A1 and aggrecan) and tissue-specific microRNAs (miR-675 and miR-140) and increased levels of the hypertrophic markers RUNX2 and MMP13, characteristic of changes occurring in osteoarthritis. We propose miR-145 as an important regulator of human chondrocyte function and a new target for cartilage repair.
Collapse
Affiliation(s)
- Aida Martinez-Sanchez
- Kennedy Institute of Rheumatology, University of Oxford, 65 Aspenlea Road, London W6 8LH, United Kingdom
| | | | | |
Collapse
|
37
|
Chang HX, Yang L, Li Z, Chen G, Dai G. Age-related biological characterization of mesenchymal progenitor cells in human articular cartilage. Orthopedics 2011; 34:e382-8. [PMID: 21815581 DOI: 10.3928/01477447-20110627-06] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Adult articular cartilage has a low regeneration capacity due to lack of viable progenitor cells caused by limited blood supply to cartilage. However, recent studies have demonstrated the existence of chondroprogenitor cells in articular cartilage. A critical question is whether these mesenchymal progenitor cells are functionally viable for tissue renewal and cartilage repair to postpone cartilage degeneration. This study was designed to compare the number and function of mesenchymal progenitor cells in articular cartilage collected from human fetuses, healthy adults (aged 28-45 years), and elderly adults (aged 60-75 years) and cultured in vitro. We detected multipotent mesenchymal progenitor cells, defined as CD105+/CD166+ cells, in human articular cartilage of all ages. However, mesenchymal progenitor cells accounted for 94.69%±2.31%, 4.85%±2.62%, and 6.33%±3.05% of cells in articular cartilage obtained from fetuses, adults, and elderly patients, respectively (P<.001). Furthermore, fetal mesenchymal progenitor cells had the highest rates of proliferation measured by cell doubling times and chondrogenic differentiation as compared to those from adult and elderly patients. In contrast, alkaline phosphatase levels, which are indicative of osteogenic differentiation, did not show significant reduction with aging. However, spontaneous osteogenic differentiation was detected only in mesenchymal progenitor cells from elderly patients (with lower Markin scales). The lower chondrogenic and spontaneous osteogenic differentiation of mesenchymal progenitor cells derived from elderly patients may be associated with the development of primary osteoarthritis. These results suggest that measuring cartilage mesenchymal progenitor cells may not only identify underlying mechanisms but also offer new diagnostic and therapeutic potential for patients with osteoarthritis.
Collapse
Affiliation(s)
- Hong-Xing Chang
- Department of Joint Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
38
|
Role of polyamines in hypertrophy and terminal differentiation of osteoarthritic chondrocytes. Amino Acids 2011; 42:667-78. [PMID: 21814786 DOI: 10.1007/s00726-011-1041-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 04/28/2011] [Indexed: 01/23/2023]
Abstract
Polyamines are naturally occurring, positively charged polycations which are able to control several cellular processes in different cell types, by interacting with negatively charged compounds and structures within the living cell. Functional genomics in rodents targeting key biosynthetic or catabolic enzymes have revealed a series of phenotypic changes, many of them related to human diseases. Several pieces of evidence from the literature point at a role of polyamines in promoting chondrocyte differentiation, a process which is physiological in growth plate maturation or fracture healing, but has pathological consequences in articular chondrocytes, programmed to keep a maturational arrested state. Inappropriate differentiation of articular chondrocytes results in osteoarthritis. Thus, we have studied the effects of exogenously added spermine or spermidine in chondrocyte maturation recapitulated in 3D cultures, to tease out the effects on gene and protein expression of key chondrogenesis regulatory transcription factors, markers and effectors, as well as their posttranscriptional regulation. The results indicate that both polyamines are able to increase the rate and the extent of chondrogenesis, with enhanced collagen 2 deposition and remodeling with downstream generation of collagen 2 bioactive peptides. These were able to promote nuclear localization of RUNX-2, the pivotal transcription factor in chondrocyte hypertrophy and osteoblast generation. Indeed, samples stimulated with polyamines showed an enhanced mineralization, along with increased caspase activity, indicating increased chondrocyte terminal differentiation. In conclusion these results indicate that the polyamine pathway can represent a potential target to control and correct chondrocyte inappropriate maturation in osteoarthritis.
Collapse
|
39
|
Abstract
SOX trio (SOX-5, SOX-6, and SOX-9) maintain the chondrocytic phenotypes and are vital for chondrogenesis in embryonic development. The purpose of this study is to investigate the change in the expression of SOX trio with the advancement of osteoarthritis (OA) in human articular cartilage (AC). Human OA samples from eight patients were obtained from the distal femoral condyles during total knee arthroplasty. Minimally OA cartilage taken from areas with no obvious surface defects on lateral condyles was compared with advanced OA cartilage obtained from areas within 1 cm of overt lesion located on medial condyle surface. SOX-5, SOX-6, and SOX-9 gene expressions significantly decreased by 41% (p = 0.047), 46% (p = 0.047), and 56% (p = 0.029) in advanced OA area compared with the minimally OA area. There was a significant decrease in aggrecan and type II collagen (COL2A1) gene expressions by 73% (p = 0.029) and 65% (p = 0.029), respectively, in advanced OA area compared with the minimally OA area. From Western blotting and immunohistochemistry, SOX-5, SOX-6, SOX-9, type II collagen, and aggrecan protein expressions also significantly decreased in advanced OA cartilage compared with minimally OA cartilage. DNA methylation study of SOX-9 promoter regions revealed no difference in the epigenetic status between the two areas. It is concluded that SOX trio gene and protein decreased with advancement of OA in human articular cartilage.
Collapse
Affiliation(s)
- Jai-Sun Lee
- Department of Orthopaedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| | | |
Collapse
|
40
|
Agar G, Blumenstein S, Bar-Ziv Y, Kardosh R, Schrift-Tzadok M, Gal-Levy R, Fischler T, Goldschmid R, Yayon A. The Chondrogenic Potential of Mesenchymal Cells and Chondrocytes from Osteoarthritic Subjects: A Comparative Analysis. Cartilage 2011; 2:40-9. [PMID: 26069568 PMCID: PMC4300788 DOI: 10.1177/1947603510380899] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE The multipotential nature of stem or progenitor cells apparently makes them the ideal choice for any cell therapy, but this as yet remains to be proven. This study (30 subjects) was designed to compare the potential to repair articular cartilage of chondrocytes taken from different regions in osteoarthritic cartilage with that of mesenchymal stem cells prepared from bone marrow of the same subject. DESIGN Cartilage biopsies, bone marrow, and blood samples were taken from each of 30 individuals with chronic osteoarthritis (aged 62-85 years) undergoing total knee replacement. The chondrogenic potential of chondrocytes isolated from cartilage biopsies taken from different regions of osteoarthritic cartilage was compared with that of mesenchymal cells by quantitative analysis of several chondrocyte specific markers and an ex vivo cartilage differentiation assay. RESULTS Cartilage-derived articular chondrocytes are superior to bone marrow-derived cells when compared for their ex vivo chondrogenic potential. Interestingly, there was marked and significant difference in the expression of chondrocytic markers between chondrocytes derived from adjacent, visually distinct regions of the diseased cartilage. When cultured in the presence of a fibroblast growth factor 2 variant, all cell samples from both tissues showed a high degree of chondrogenic potential. CONCLUSIONS Although bone marrow-derived mesenchymal cells, when supplemented with the appropriate chondrogenic factors, are a suitable source for autologous cartilage implantation, adult chondroprogenitor cells, particularly those from moderately affected regions of the osteoarthritic joints, demonstrate superior chondrogenic potential.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Avner Yayon
- ProChon Biotech Ltd., Ness Ziona, Israel,Avner Yayon, PhD, MD, ProChon Biotech Ltd., 7, Golda Meir Street, Weizmann Science Park, P.O. Box 4082, Ness Ziona 70400, Israel
| |
Collapse
|
41
|
Gavenis K, Heussen N, Schmidt-Rohlfing B. Effects of Low Concentration BMP-7 on Human Osteoarthritic Chondrocytes: Comparison of Different Applications. J Biomater Appl 2010; 26:845-59. [DOI: 10.1177/0885328210388439] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
While BMP-7 (OP-1) is one of the most potent growth factors in cartilage tissue engineering, the effects of exogenous low concentration BMP-7 on osteoarthritic chondrocytes are still unknown. Human osteoarthritic chondrocytes obtained from the femoral condyles of 10 patients were grown either in monolayer or in 3D collagen type-I gel culture in vitro. The growth factor was either given as a single dose of 50 ng/mL, a repeated dose, or continuously released from PGLA microspheres. Matrix formation was monitored by immunohistochemical staining and real-time PCR. In contrast to monolayer culture, the differentiated phenotype was prevailed in 3D culture. Collagen type-II protein production in the 3D group with a continuous BMP-7 release was enhanced in comparison to all other groups. Gene expression of collagen type-II and aggrecan was elevated in all treatment groups, with the highest extent in the BMP-7 microsphere group. In summary, treatment of articular chondrocytes with a low dose of BMP-7 leads to an elevated production of extracellular matrix components. This effect is further increased when BMP-7 is given repeatedly or continuously, which proved to be the most effective form of application.
Collapse
Affiliation(s)
- Karsten Gavenis
- Department of Orthopaedic Surgery, Aachen University Hospital, Aachen, Germany
| | - Nicole Heussen
- Institute of Medical Statistics, RWTH Aachen University, Germany
| | | |
Collapse
|
42
|
Borzi RM, Olivotto E, Pagani S, Vitellozzi R, Neri S, Battistelli M, Falcieri E, Facchini A, Flamigni F, Penzo M, Platano D, Santi S, Facchini A, Marcu KB. Matrix metalloproteinase 13 loss associated with impaired extracellular matrix remodeling disrupts chondrocyte differentiation by concerted effects on multiple regulatory factors. ARTHRITIS AND RHEUMATISM 2010; 62:2370-81. [PMID: 20506238 PMCID: PMC2921033 DOI: 10.1002/art.27512] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To link matrix metalloproteinase 13 (MMP-13) activity and extracellular matrix (ECM) remodeling to alterations in regulatory factors leading to a disruption in chondrocyte homeostasis. METHODS MMP-13 expression was ablated in primary human chondrocytes by stable retrotransduction of short hairpin RNA. The effects of MMP-13 knockdown on key regulators of chondrocyte differentiation (SOX9, runt-related transcription factor 2 [RUNX-2], and beta-catenin) and angiogenesis (vascular endothelial growth factor [VEGF]) were scored at the protein level (by immunohistochemical or Western blot analysis) and RNA level (by real-time polymerase chain reaction) in high-density monolayer and micromass cultures under mineralizing conditions. Effects on cellular viability in conjunction with chondrocyte progression toward a hypertrophic-like state were assessed in micromass cultures. Alterations in SOX9 subcellular distribution were assessed using confocal microscopy in micromass cultures and also in osteoarthritic cartilage. RESULTS Differentiation of control chondrocyte micromasses progressed up to a terminal phase, with calcium deposition in conjunction with reduced cell viability and scant ECM. MMP-13 knockdown impaired ECM remodeling and suppressed differentiation in conjunction with reduced levels of RUNX-2, beta-catenin, and VEGF. MMP-13 levels in vitro and ECM remodeling in vitro and in vivo were linked to changes in SOX9 subcellular localization. SOX9 was largely excluded from the nuclei of chondrocytes with MMP-13-remodeled or -degraded ECM, and exhibited an intranuclear staining pattern in chondrocytes with impaired MMP-13 activity in vitro or with more intact ECM in vivo. CONCLUSION MMP-13 loss leads to a breakdown in primary human articular chondrocyte differentiation by altering the expression of multiple regulatory factors.
Collapse
Affiliation(s)
- Rosa Maria Borzi
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituti Ortopedici Rizzoli, Via di Barbiano 1/10, Bologna, Italy
| | - Eleonora Olivotto
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituti Ortopedici Rizzoli, Via di Barbiano 1/10, Bologna, Italy
| | - Stefania Pagani
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituti Ortopedici Rizzoli, Via di Barbiano 1/10, Bologna, Italy
| | - Roberta Vitellozzi
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituti Ortopedici Rizzoli, Via di Barbiano 1/10, Bologna, Italy
| | - Simona Neri
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituti Ortopedici Rizzoli, Via di Barbiano 1/10, Bologna, Italy
| | - Michela Battistelli
- Istituto di Scienze Morfologiche, Università degli Studi “Carlo Bo”, Urbino, Italy
| | - Elisabetta Falcieri
- Istituto di Scienze Morfologiche, Università degli Studi “Carlo Bo”, Urbino, Italy
| | - Annalisa Facchini
- Dipartimento di Biochimica, Università degli Studi di Bologna, Bologna, Italy
| | - Flavio Flamigni
- Dipartimento di Biochimica, Università degli Studi di Bologna, Bologna, Italy
| | - Marianna Penzo
- Vita Salute San Raffaele University, Via Olgettina 58, 20132 Milano, Italy
- Centro Ricerca Biomedica Applicata (CRBA), Policlinico S. Orsola-Malpighi, via Massarenti 9, Bologna, Italy
| | - Daniela Platano
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituti Ortopedici Rizzoli, Via di Barbiano 1/10, Bologna, Italy
| | | | - Andrea Facchini
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituti Ortopedici Rizzoli, Via di Barbiano 1/10, Bologna, Italy
- Dipartimento di Medicina Clinica, Università degli Studi di Bologna, Bologna, Italy
| | - Kenneth B Marcu
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Istituti Ortopedici Rizzoli, Via di Barbiano 1/10, Bologna, Italy
- Dept. of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794-5215 USA
| |
Collapse
|
43
|
Ahmed N, Taylor DW, Wunder J, Nagy A, Gross AE, Kandel RA. Passaged human chondrocytes accumulate extracellular matrix when induced by bovine chondrocytes. J Tissue Eng Regen Med 2010; 4:233-41. [DOI: 10.1002/term.235] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
44
|
Rutgers M, van Pelt MJP, Dhert WJA, Creemers LB, Saris DBF. Evaluation of histological scoring systems for tissue-engineered, repaired and osteoarthritic cartilage. Osteoarthritis Cartilage 2010; 18:12-23. [PMID: 19747584 DOI: 10.1016/j.joca.2009.08.009] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 07/21/2009] [Accepted: 08/02/2009] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Regeneration of hyaline cartilage has been the focus of an increasing number of research groups around the world. One of the most important outcome measures in evaluation of its success is the histological quality of cartilaginous tissue. Currently, a variety of histological scoring systems is used to describe the quality of osteoarthritic, in vivo repaired or in vitro engineered tissue. This review aims to provide an overview of past and currently used histological scoring systems, in an effort to aid cartilage researchers in choosing adequate and validated cartilage histological scoring systems. METHODS Histological scoring systems for analysis of osteoarthritic, tissue engineered and in vivo repaired cartilage were reviewed. The chronological development as well as the validity and practical applicability of the scoring systems is evaluated. RESULTS The Histological-Histochemical Grading System (HHGS) or a HHGS-related score is most often used for evaluation of osteoarthritic cartilage, however the Osteoarthritis Research Society International (OARSI) Osteoarthritis Cartilage Histopathology Assessment System seems a valid alternative. The O'Driscoll score and the International Cartilage Repair Society (ICRS) II score may be used for in vivo repaired cartilage. The 'Bern score' seems most adequate for evaluation of in vitro engineered cartilage. CONCLUSION A great variety of histological scoring systems exists for analysis of osteoarthritic or normal, in vivo repaired or tissue-engineered cartilage, but only few have been validated. Use of these validated scores may considerably improve exchange of information necessary for advances in the field of cartilage regeneration.
Collapse
Affiliation(s)
- M Rutgers
- Department of Orthopaedics, University Medical Center Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
45
|
Rankin KS, Lakey RL, Gerrand CH, Sprowson AP, McCaskie AW, Birch MA. A novel in vitro model to investigate behavior of articular chondrocytes in osteoarthritis. J Rheumatol 2009; 37:426-31. [PMID: 20032095 DOI: 10.3899/jrheum.080080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To investigate in vivo simulation of the microenvironment in which osteoarthritis (OA) chondrocytes are cultured in vitro. METHODS Human articular chondrocytes were cultured under normoxic and hypoxic conditions. Cells were cultured on standard culture plastic or a porous polyHEMA surface that closely resembles the in vivo cartilage microarchitecture. Morphological changes to the cells were demonstrated by fluorescent staining with DAPI and vinculin. Proteoglycan and type II collagen protein levels were assessed using established techniques. Matrix metalloproteinase-1 (MMP-1) production was assessed by ELISA. The gene expression of type II collagen and SOX9 was measured using real-time polymerase chain reaction. RESULTS Cells grown on culture plastic were seen to be flat and hexagonal. Cells cultured on the porous polyHEMA surface exhibited morphology in keeping with the in vivo microenvironment. Glycosaminoglycan release in hypoxia was high from cells cultured on standard culture plastic. Transcriptional expression of type II collagen was upregulated in hypoxia and by culture on the polyHEMA surface. Transcriptional expression of SOX9 in hypoxia was upregulated compared to normoxia; no significant effect was seen by varying the culture surface. Translational expression of type II collagen was upregulated at 20% oxygen on the polyHEMA surface compared to culture plastic and this was related to MMP-1 expression. CONCLUSION Culture of chondrocytes in hypoxia and on a porous surface simulates the in vivo microenvironment and illustrates the molecular mechanisms of OA.
Collapse
Affiliation(s)
- Kenneth S Rankin
- Musculoskeletal Research Group, University of Newcastle upon Tyne, Medical School, Newcastle upon Tyne, United Kingdom.
| | | | | | | | | | | |
Collapse
|
46
|
Hsieh-Bonassera ND, Wu I, Lin JK, Schumacher BL, Chen AC, Masuda K, Bugbee WD, Sah RL. Expansion and redifferentiation of chondrocytes from osteoarthritic cartilage: cells for human cartilage tissue engineering. Tissue Eng Part A 2009; 15:3513-23. [PMID: 19456239 PMCID: PMC2792054 DOI: 10.1089/ten.tea.2008.0628] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Accepted: 05/20/2009] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To determine if selected culture conditions enhance the expansion and redifferentiation of chondrocytes isolated from human osteoarthritic cartilage with yields appropriate for creation of constructs for treatment of joint-scale cartilage defects, damage, or osteoarthritis. METHODS Chondrocytes isolated from osteoarthritic cartilage were analyzed to determine the effects of medium supplement on cell expansion in monolayer and then cell redifferentiation in alginate beads. Expansion was assessed as cell number estimated from DNA, growth rate, and day of maximal growth. Redifferentiation was evaluated quantitatively from proteoglycan and collagen type II content, and qualitatively by histology and immunohistochemistry. RESULTS Using either serum or a growth factor cocktail (TFP: transforming growth factor beta1, fibroblast growth factor 2, and platelet-derived growth factor type bb), cell growth rate in monolayer was increased to 5.5x that of corresponding conditions without TFP, and cell number increased 100-fold within 17 days. In subsequent alginate bead culture with human serum or transforming growth factor beta1 and insulin-transferrin-selenium-linoleic acid-bovine serum albumin, redifferentiation was enhanced with increased proteoglycan and collagen type II production. Effects of human serum were dose dependent, and 5% or higher induced formation of chondron-like structures with abundant proteoglycan-rich matrix. CONCLUSION Chondrocytes from osteoarthritic cartilage can be stimulated to undergo 100-fold expansion and then redifferentiation, suggesting that they may be useful as a cell source for joint-scale cartilage tissue engineering.
Collapse
Affiliation(s)
- Nancy D. Hsieh-Bonassera
- Department of Mechanical and Aerospace Engineering, University of California–San Diego, La Jolla, California
| | - Iwen Wu
- Department of Bioengineering, University of California–San Diego, La Jolla, California
| | - Jonathan K. Lin
- Department of Bioengineering, University of California–San Diego, La Jolla, California
| | - Barbara L. Schumacher
- Department of Bioengineering, University of California–San Diego, La Jolla, California
| | - Albert C. Chen
- Department of Bioengineering, University of California–San Diego, La Jolla, California
| | - Koichi Masuda
- Department of Orthopaedic Surgery, University of California–San Diego, La Jolla, California
| | - William D. Bugbee
- Department of Orthopaedic Surgery, University of California–San Diego, La Jolla, California
- Division of Orthopaedic Surgery, Scripps Clinic, La Jolla, California
| | - Robert L. Sah
- Department of Bioengineering, University of California–San Diego, La Jolla, California
- Institute of Engineering in Medicine, University of California–San Diego, La Jolla, California
| |
Collapse
|
47
|
Dehne T, Karlsson C, Ringe J, Sittinger M, Lindahl A. Chondrogenic differentiation potential of osteoarthritic chondrocytes and their possible use in matrix-associated autologous chondrocyte transplantation. Arthritis Res Ther 2009; 11:R133. [PMID: 19723327 PMCID: PMC2787268 DOI: 10.1186/ar2800] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Revised: 07/27/2009] [Accepted: 09/02/2009] [Indexed: 02/08/2023] Open
Abstract
Introduction Autologous chondrocyte transplantation (ACT) is a routine technique to regenerate focal cartilage lesions. However, patients with osteoarthritis (OA) are lacking an appropriate long-lasting treatment alternative, partly since it is not known if chondrocytes from OA patients have the same chondrogenic differentiation potential as chondrocytes from donors not affected by OA. Methods Articular chondrocytes from patients with OA undergoing total knee replacement (Mankin Score > 3, Ahlbäck Score > 2) and from patients undergoing ACT, here referred to as normal donors (ND), were isolated applying protocols used for ACT. Their chondrogenic differentiation potential was evaluated both in high-density pellet and scaffold (Hyaff-11) cultures by histological proteoglycan assessment (Bern Score) and immunohistochemistry for collagen types I and II. Chondrocytes cultured in monolayer and scaffolds were subjected to gene expression profiling using genome-wide oligonucleotide microarrays. Expression data were verified by using real-time PCR. Results Chondrocytes from ND and OA donors demonstrated accumulation of comparable amounts of cartilage matrix components, including sulphated proteoglycans and collagen types I and II. The mRNA expression of cartilage markers (ACAN, COL2A1, COMP, CRTL1, SOX9) and genes involved in matrix synthesis (BGN, CILP2, COL9A2, COL11A1, TIMP4) was highly induced in 3D cultures of chondrocytes from both donor groups. Genes associated with hypertrophic or OA cartilage (ALPL, COL1A1, COL3A1, COL10A1, MMP13, POSTN, PTH1R, RUNX2) were not significantly regulated between the two groups of donors. The expression of 661 genes, including COMP, FN1, and SOX9, was differentially regulated between OA and ND chondrocytes cultured in monolayer. During scaffold culture, the differences diminished between the OA and ND chondrocytes, and only 184 genes were differentially regulated. Conclusions Only few genes were differentially expressed between OA and ND chondrocytes in Hyaff-11 culture. The risk of differentiation into hypertrophic cartilage does not seem to be increased for OA chondrocytes. Our findings suggest that the chondrogenic capacity is not significantly affected by OA, and OA chondrocytes fulfill the requirements for matrix-associated ACT.
Collapse
Affiliation(s)
- Tilo Dehne
- Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Tucholskystrasse 2, Berlin, 10117, Germany.
| | | | | | | | | |
Collapse
|
48
|
Blaney Davidson EN, Remst DFG, Vitters EL, van Beuningen HM, Blom AB, Goumans MJ, van den Berg WB, van der Kraan PM. Increase in ALK1/ALK5 ratio as a cause for elevated MMP-13 expression in osteoarthritis in humans and mice. THE JOURNAL OF IMMUNOLOGY 2009; 182:7937-45. [PMID: 19494318 DOI: 10.4049/jimmunol.0803991] [Citation(s) in RCA: 222] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
During osteoarthritis (OA) chondrocytes show deviant behavior resembling terminal differentiation of growth-plate chondrocytes, characterized by elevated MMP-13 expression. The latter is also a hallmark for OA. TGF-beta is generally thought to be a protective factor for cartilage, but it has also displayed deleterious effects in some studies. Recently, it was shown that besides signaling via the ALK5 (activin-like kinase 5) receptor, TGF-beta can also signal via ALK1, thereby activating Smad1/5/8 instead of Smad2/3. The Smad1/5/8 route can induce chondrocyte terminal differentiation. Murine chondrocytes stimulated with TGF-beta activated the ALK5 receptor/Smad2/3 route as well as the ALK1/Smad1/5/8 route. In cartilage of mouse models for aging and OA, ALK5 expression decreased much more than ALK1. Thus, the ALK1/ALK5 ratio increased, which was associated with changes in the respective downstream markers: an increased Id-1 (inhibitor of DNA binding-1)/PAI-1 (plasminogen activator inhibitor-1) ratio. Transfection of chondrocytes with adenovirus overexpressing constitutive active ALK1 increased MMP-13 expression, while small interfering RNA against ALK1 decreased MMP-13 expression to nondetectable levels. Adenovirus overexpressing constitutive active ALK5 transfection increased aggrecan expression, whereas small interfering RNA against ALK5 resulted in increased MMP-13 expression. Moreover, in human OA cartilage ALK1 was highly correlated with MMP-13 expression, whereas ALK5 correlated with aggrecan and collagen type II expression, important for healthy cartilage. Collectively, we show an age-related shift in ALK1/ALK5 ratio in murine cartilage and a strong correlation between ALK1 and MMP-13 expression in human cartilage. A change in balance between ALK5 and ALK1 receptors in chondrocytes caused changes in MMP-13 expression, thereby causing an OA-like phenotype. Our data suggest that dominant ALK1 signaling results in deviant chondrocyte behavior, thereby contributing to age-related cartilage destruction and OA.
Collapse
Affiliation(s)
- Esmeralda N Blaney Davidson
- Rheumatology Research and Advanced Therapeutics, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kreuz PC, Müller S, Ossendorf C, Kaps C, Erggelet C. Treatment of focal degenerative cartilage defects with polymer-based autologous chondrocyte grafts: four-year clinical results. Arthritis Res Ther 2009; 11:R33. [PMID: 19265548 PMCID: PMC2688178 DOI: 10.1186/ar2638] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 02/04/2009] [Accepted: 03/05/2009] [Indexed: 12/04/2022] Open
Abstract
Introduction Second-generation autologous chondrocyte implantation with scaffolds stabilizing the grafts is a clinically effective procedure for cartilage repair. In this ongoing prospective observational case report study, we evaluated the effectiveness of BioSeed®-C, a cell-based cartilage graft based on autologous chondrocytes embedded in fibrin and a stable resorbable polymer scaffold, for the treatment of clinical symptomatic focal degenerative defects of the knee. Methods Clinical outcome after 4-year clinical follow-up was assessed in 19 patients with preoperatively radiologically confirmed osteoarthritis and a Kellgren-Lawrence score of 2 or more. Clinical scoring was performed before implantation of the graft and 6, 12, and 48 months after implantation using the Lysholm score, the Knee injury and Osteoarthritis Outcome Score (KOOS), the International Knee Documentation Committee (IKDC) score, and the International Cartilage Repair Society (ICRS) score. Cartilage regeneration and articular resurfacing were assessed by magnetic resonance imaging (MRI) 4 years after implantation of the autologous cartilage graft. Results Significant improvement (P < 0.05) of the Lysholm and ICRS scores was observed as early as 6 months after implantation of BioSeed®-C and remained stable during follow-up. The IKDC score showed significant improvement compared with the preoperative situation at 12 and 48 months (P < 0.05). The KOOS showed significant improvement in the subclasses pain, activities of daily living, and knee-related quality of life 6 months as well as 1 and 4 years after implantation of BioSeed®-C in osteoarthritic defects (P < 0.05). MRI analysis showed moderate to complete defect filling with a normal to incidentally hyperintense signal in 16 out of 19 patients treated with BioSeed®-C. Two patients without improvement in the clinical and MRI scores received a total knee endoprosthesis after 4 years. Conclusions The results show that the good clinical outcome achieved 1 year after implantation of BioSeed®-C remains stable over the course of a period of 4 years and suggest that implanting BioSeed®-C is a promising treatment option for the repair of focal degenerative defects of the knee.
Collapse
Affiliation(s)
- Peter C Kreuz
- Department of Orthopaedic and Trauma Surgery, University Medical Center Rechts der Isar of the Technical University Munich, Munich, Germany.
| | | | | | | | | |
Collapse
|
50
|
Gavenis K, Schumacher C, Schneider U, Eisfeld J, Mollenhauer J, Schmidt-Rohlfing B. Expression of ion channels of the TRP family in articular chondrocytes from osteoarthritic patients: changes between native and in vitro propagated chondrocytes. Mol Cell Biochem 2008; 321:135-43. [PMID: 18836817 DOI: 10.1007/s11010-008-9927-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Accepted: 09/15/2008] [Indexed: 11/26/2022]
Abstract
The maintenance of a differentiated chondrocyte phenotype is influenced by several factors of which signal transduction of extracellular stimuli through the cell membrane is of major interest. One important group of membrane-bound proteins which are involved in transmembrane signal transduction are ion channels. Human articular chondrocytes were obtained from osteoarthritic femoral condyles. Cells were released from the surrounding matrix and cultivated under standard conditions. We investigated gene expression of 12 members of the TRP ion channel family of freshly prepared (passage 0; P0) and in vitro propagated human articular chondrocytes (passage 2; P2) using conventional and real-time PCR (RT-PCR). In addition, the protein appearance of four TRP channels was demonstrated by immunofluorescence and western blotting. Chondrocyte differentiation was monitored by quantification of collagen type-II, type-I, and aggrecan gene expression. By conventional PCR, 8 channels could be detected, of which some displayed a heterogeneous PCR pattern. RT-PCR quantification revealed that TRPC1 was expressed on the same level in P0 and P2 chondrocytes while gene expression of TRPC3 and TRPC6 was elevated in passage 2 cells. TRPM5, TRPM7, and TRPV1 displayed an enhanced gene expression in freshly isolated chondrocytes. Immunofluorescence signal intensity of all four investigated TRP proteins was consistent with the corresponding gene expression data. In the present study, a correlation between the appearance of some members of the TRP ion channel family and the state of de-differentiation of osteoarthritic articular chondrocytes was shown. A possible direct involvement in the process of chondrocyte de-differentiation has to be investigated in further studies.
Collapse
Affiliation(s)
- K Gavenis
- Department of Orthopaedics and Trauma Surgery, Aachen University Hospital, Aachen, Germany
| | | | | | | | | | | |
Collapse
|