1
|
Cai Z, Shu L, Wang C, Xie X, Liu X. M2 Macrophage-Derived Exosomes Promote Tendon-to-Bone Healing by Alleviating Cellular Senescence in Aged Rats. Arthroscopy 2025; 41:1731-1742.e3. [PMID: 39326562 DOI: 10.1016/j.arthro.2024.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/31/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024]
Abstract
PURPOSE To explore the potential of M2 macrophage-derived exosomes (M2-Exos) in enhancing tendon-to-bone healing in aged rats by mitigating cellular senescence of bone marrow-derived stem cells (BMSCs). METHODS In vitro, the effects of M2-Exos on alleviating cellular senescence and improving chondrogenic potential of senescent BMSCs were evaluated. Rats (24 young and 48 aged) with chronic rotator cuff tear (RCT) were repaired and assigned into 3 groups: young group (young rats injected with fibrin at the enthesis), aged group (aged rats injected with fibrin at the enthesis), and aged + M2-Exos group (aged rats injected with fibrin containing M2-Exos at the enthesis). At 6 and 12 weeks after repair, enthesis regeneration was evaluated. Proteomic analysis was conducted to explore the mechanism through which M2-Exos mitigated cellular senescence. RESULTS In senescent BMSCs treated with M2-Exos, there was a reduction in senescence biomarkers including senescence-associated β-galactosidase, p53, p21, and senescence-associated secretory phenotype (P < .001). M2-Exos also enhanced chondrogenic potential of senescent BMSCs, reflected in greater Bern score (P < .001) and increased expression of Sox9 (P = .013), Col2a1 (P < .001), and Acan (P < .001). Histologically, aged rats treated with M2-Exos demonstrated significantly greater histologic scores (P < .001 at both 6 and 12 weeks) and increased fibrocartilage regeneration at the enthesis. Biomechanically, these rats exhibited greater failure load, stiffness, and stress (all P < .001) at 12 weeks. Mechanistically, proteomic analysis suggested that M2-Exos might alleviate cellular senescence by potentially regulating DNA replication and repair. CONCLUSIONS M2-Exos can significantly alleviate BMSC senescence and thereby enhance tendon-to-bone healing in an aged rat RCT model. CLINICAL RELEVANCE This study suggests the potential utility of M2-Exos as a therapy for RCT in the older population.
Collapse
Affiliation(s)
- Zhuochang Cai
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Longqiang Shu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chongyang Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuetao Xie
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xudong Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Li Y, Xun X, Duan L, Gao E, Li J, Lin L, Li X, He A, Ao H, Xu Y, Xia H. Cartilage structure-inspired nanofiber-hydrogel composite with robust proliferation and stable chondral lineage-specific differentiation function to orchestrate cartilage regeneration for artificial tracheal construction. Bioact Mater 2025; 47:136-151. [PMID: 39897586 PMCID: PMC11787707 DOI: 10.1016/j.bioactmat.2025.01.007] [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: 09/10/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
Tissue engineering strategies hold promise for constructing biomimetic tracheal substitutes to repair circumferential tracheal defects. However, current strategies for constructing off-the-shelf cartilage analogs for artificial trachea grafts face challenges of chondrocyte scarcity and inadequate culture strategies, which require extensive cell expansion and prolonged in vitro culture to generate robust neo-cartilage. To address these issues, we developed a nanofiber-hydrogel composite with superior mechanical performance by incorporating fragment oxidized bacterial cellulose (BC) nanofibers into a gelatin methacryloyl (GelMA) hydrogel network. Additionally, a biomaterial system was developed based on this composite, featuring dual-release functionality of fibroblast growth factor (FGF) and transforming growth factor beta (TGF-β) to facilitate step-wise maturation of neo-cartilage tissue. This process includes early-stage proliferation followed by second-stage extracellular matrix (ECM) deposition, driving the transition from proliferation to chondrogenesis. By encapsulating chondrocytes within the biomaterial system, mature neo-cartilage tissues with typical cartilage lacunae structures and abundant homogeneous cartilage-specific ECM deposition were successfully regenerated in vitro and in vivo. Furthermore, with a tailor-made growth factor-releasing strategy, the biomaterial system with low cell seeding density achieved biochemically and biomechanically functional neo-cartilage tissue regeneration, comparable to that achieved with high cell seeding density in the nanofiber-hydrogel composite. Based on the current biomaterial system, mature and functional cartilage-ring analogs were successfully constructed and applied to repair tracheal defects. Overall, the biomaterial system developed in this study provides a promising strategy for engineering transplantable, high-quality cartilage substitutes, with translational potential for artificial trachea construction.
Collapse
Affiliation(s)
- Yaqiang Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200430, China
| | - Xiaowei Xun
- School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, China
| | - Liang Duan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200430, China
| | - Erji Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200430, China
| | - Jiaxin Li
- Department of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Lei Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200430, China
| | - Xinping Li
- Department of Thyroid Center, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Aijuan He
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital of Fudan University, Shanghai, 200031, China
| | - Haiyong Ao
- School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, China
| | - Yong Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200430, China
- Department of Plastic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Huitang Xia
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
- Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, Jinan, Shandong, 250014, China
| |
Collapse
|
3
|
Kutaish H, Bengtsson L, Boudabbous S, Lazeyras F, Courvoisier S, Braunersreuther V, Hammer SE, Hannouche D, Ménétrey J, Tieng V, Tscholl PM. Allogenic Bioengineered Cartilage Achieves Hyaline Cartilage Repair in a Large Animal Model: A Promising Step Forward. Am J Sports Med 2025:3635465251331224. [PMID: 40289476 DOI: 10.1177/03635465251331224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
BACKGROUND Chondrocyte-based cell therapy remains a promising method for cartilage repair, despite limitations faced during the last 30 years. PURPOSE/HYPOTHESIS This work presents hyaline-like bioengineered beads from donor chondrocytes as a novel treatment option for cartilage lesions. It was hypothesized that the implanted cartilage minigrafts would be able to treat cartilage lesions by complete fusion among themselves and by integration with surrounding tissue. No tissue rejection was expected because of cartilage's reported immunological privilege. STUDY DESIGN Controlled laboratory study. METHODS Allogenic cartilage beads with hyaline characteristics were produced from frozen chondrocytes of a minipig donor. A total of 8 Göttingen minipigs underwent the implantation of bioengineered cartilage beads into 8 to 10 mm-diameter full-thickness chondral lesions (3 lesions/knee). Animals were sacrificed at 6 weeks (n = 2) and 6 months (n = 6) after implantation. The safety and efficacy of implantation were assessed by macroscopic and histological analyses as well as by magnetic resonance imaging. RESULTS No signs of acute or chronic rejection were observed in any study animals upon implantation. For 6 minipigs at 6 months, magnetic resonance imaging results showed better coverage of the grafted lesions compared with empty (control) lesions. When the cartilage beads were maintained in the lesion, complete integration of the minigrafts with surrounding subchondral bone and native cartilage was observed. Repair tissue in grafted lesions maintained hyaline-like quality and showed evidence of a chondral zonal arrangement at 6 months' follow-up. Additionally, grafted lesions (n = 17) had better macroscopic repair scores than empty lesions (n = 7) (mean inverse Goebel score, 4.24 and 5.57, respectively). Graft-filled lesions showed only a slight superiority in histological repair scores (mean Bern score, 5.76 and 5.43, respectively). CONCLUSION Allogenic cartilage beads hold potential as an advanced therapy medicinal product for treating cartilage lesions in 1-step surgery with established safety and efficacy. CLINICAL RELEVANCE This successful preclinical study highlights allogenic cartilage beads as a promising method for cartilage repair. Moreover, using donor chondrocytes may allow reduced patient morbidity and 1-step surgery. Hence, this advanced therapy medicinal product is suitable for treating large lesions and older patients and is currently being evaluated in a phase I/IIa clinical trial.
Collapse
Affiliation(s)
- Halah Kutaish
- Division of Orthopaedic Surgery and Traumatology, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | | | - Sana Boudabbous
- Division of Radiology, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Center for Biomedical Imaging, Lausanne, Switzerland
| | - François Lazeyras
- Division of Radiology, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Center for Biomedical Imaging, Lausanne, Switzerland
| | - Sebastien Courvoisier
- Division of Radiology, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Center for Biomedical Imaging, Lausanne, Switzerland
| | - Vincent Braunersreuther
- Division of Clinical Pathology, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Sabine E Hammer
- Institute of Immunology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Didier Hannouche
- Division of Orthopaedic Surgery and Traumatology, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Jacques Ménétrey
- Centre de Médecine du Sport et de L'Exercice, Hirslanden Clinique La Colline, Geneva, Switzerland
| | | | - Philippe M Tscholl
- Division of Orthopaedic Surgery and Traumatology, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
4
|
Metzler NF, Kondo M, Matsukura K, Ford AJ, Grainger DW, Okano T. Differentiated and Untreated Juvenile Chondrocyte Sheets Regenerate Cartilage Similarly In Vivo. Tissue Eng Part A 2025; 31:184-194. [PMID: 39556329 DOI: 10.1089/ten.tea.2024.0208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Osteoarthritis, a degenerative disease of articular cartilage and the leading cause of disability, is preceded by acute cartilage injury in a significant proportion of cases. Current auto- and allograft interventions are limited by supply and variability in therapeutic efficacy, prompting interest in tissue engineering solutions. Cell sheet tissue engineering, a scaffold-free regenerative technique, has shown promise in preclinical and clinical trials across various cell types and diseases. Polydactyly-derived juvenile cartilage-derived chondrocyte (JCC) sheets from juvenile patients are a potent cell source for developing allogeneic therapies. JCC sheets have proven safe and effective in animal models and as an add-on therapy in a recent clinical cartilage repair study. However, JCC ex vivo expansion leads to de-differentiation, contributing to long healing times. This study hypothesized that in vitro differentiation of JCC sheets into hyaline-like cartilage constructs could accelerate cartilage regeneration without compromising implant integration. To this end, sheet integration, maturation, and healing of conventionally prepared vs. differentiated JCC sheets were compared in an established nude rat focal chondral defect model. Differentiated JCC sheets exhibit mature cartilage phenotypes prior to transplant. Both conventional and differentiated JCC sheets are reliably transplanted without additional fixation. Histological evaluation reveals that both transplant groups produced equivalent neocartilage regeneration, filling defects with mature hyaline cartilage at 2- and 4-weeks post-transplant. Notably, differentiated JCC sheets respond to in vivo signals, undergoing matrix remodeling and integration with adjacent and subchondral tissue. Given equivalent healing outcomes, the future utility of in vitro JCC sheet predifferentiation from other JCC donors with different healing capacities should be balanced against their increased culture costs over conventional sheets.
Collapse
Affiliation(s)
- Nicolás F Metzler
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Makoto Kondo
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
| | - Keisuke Matsukura
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Orthopaedic Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Adam J Ford
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
| | - David W Grainger
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Teruo Okano
- Department of Molecular Pharmaceutics, Health Sciences, Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
5
|
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
|
6
|
Colombini A, Lopa S, Libonati F, Talò G, Mareschi K, Marini E, Mangiavini L, Raffo V, Moretti M, de Girolamo L. Low-density cultured cartilage cells expanded in platelet lysate present distinct features to develop an innovative clinical treatment for diffuse cartilage lesions. Knee Surg Sports Traumatol Arthrosc 2024; 32:2859-2873. [PMID: 38842036 DOI: 10.1002/ksa.12305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
PURPOSE Chondrocyte-based cell therapies are effective for the treatment of chondral lesions, but remain poorly indicated for diffuse lesions in the context of early osteoarthritis (OA). The aim of this study was to develop a protocol to obtain chondroprogenitor cells suitable for the treatment of diffuse chondral lesions within early OA. METHODS Cartilage cells were expanded at low density in human platelet lysate (hPL). A test was performed to exclude senescence. The expression of surface cluster of differentiation 146, cluster of differentiation 166, major histocompatibility complex (MHC)-I and MHC-II and of genes of interest were evaluated, as well as the trophic potential of these cells, by the assessment of lubricin and matrix production. The immunomodulatory potential was assessed through their co-culture with macrophages. RESULTS Cartilage cells expanded at low density in hPL showed higher proliferation rate than standard-density cells, no replicative senescence, low immunogenicity and expression of lubricin. Moreover, they presented an increased expression of chondrogenic and antihypertrophic markers, as well as a superior matrix deposition if compared to cells cultured at standard density. Cartilage cells induced on macrophages an upregulation of CD206, although a higher increase of CD163 expression was observed in the presence of low-density cells. CONCLUSIONS These findings lay the grounds to explore the clinical usefulness of low-density cultured cartilage cells to treat diffuse lesions in early OA joints for both autologous and allogenic use. LEVEL OF EVIDENCE Not applicable.
Collapse
Affiliation(s)
| | - Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Francesca Libonati
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Giuseppe Talò
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Katia Mareschi
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Turin, Italy
| | - Elena Marini
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
| | - Laura Mangiavini
- IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Vincenzo Raffo
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Laura de Girolamo
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|
7
|
Perry AC, Adesida AB. Tissue Engineering Nasal Cartilage Grafts with Three-Dimensional Printing: A Comprehensive Review. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39311456 DOI: 10.1089/ten.teb.2024.0187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Nasal cartilage serves a crucial structural function for the nose, where rebuilding the cartilaginous framework is an essential aspect of nasal reconstruction. Conventional methods of nasal reconstruction rely on autologous cartilage harvested from patients, which contributes to donor site pain and the potential for site-specific complications. Some patients are not ideal candidates for this procedure due to a lack of adequate substitute cartilage due to age-related calcification, differences in tissue quality, or due to prior surgeries. Tissue engineering, combined with three-dimensional printing technologies, has emerged as a promising method of generating biomimetic tissues to circumvent these issues to restore normal function and aesthetics. We conducted a comprehensive literature review to examine the applications of three-dimensional printing in conjunction with tissue engineering for the generation of nasal cartilage grafts. This review aims to compare various approaches and discuss critical considerations in the design of these grafts.
Collapse
Affiliation(s)
- Alexander C Perry
- Department of Surgery, Division of Plastic Surgery, University of Alberta, Edmonton, Canada
- Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, University of Alberta, Edmonton, Canada
| | - Adetola B Adesida
- Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, University of Alberta, Edmonton, Canada
- Department of Surgery, Division of Otolaryngology, University of Alberta, Edmonton, Canada
| |
Collapse
|
8
|
Wu B, Zhang T, Chen H, Shi X, Guan C, Hu J, Lu H. Exosomes derived from bone marrow mesenchymal stem cell preconditioned by low-intensity pulsed ultrasound stimulation promote bone-tendon interface fibrocartilage regeneration and ameliorate rotator cuff fatty infiltration. J Orthop Translat 2024; 48:89-106. [PMID: 39189009 PMCID: PMC11345897 DOI: 10.1016/j.jot.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 05/28/2024] [Accepted: 07/18/2024] [Indexed: 08/28/2024] Open
Abstract
Background Fibrovascular scar healing of bone-tendon interface (BTI) instead of functional fibrocartilage regeneration is the main concern associated with unsatisfactory prognosis in rotator cuff repair. Mesenchymal stem cells (MSCs) exosomes have been reported to be a new promising cell-free approach for rotator cuff healing. Whereas, controversies abound in whether exosomes of native MSCs alone can effectively induce chondrogenesis. Purpose To explore the effect of exosomes derived from low-intensity pulsed ultrasound stimulation (LIPUS)-preconditioned bone marrow mesenchymal stem cells (LIPUS-BMSC-Exos) or un-preconditioned BMSCs (BMSC-Exos) on rotator cuff healing and the underlying mechanism. Methods C57BL/6 mice underwent unilateral supraspinatus tendon detachment and repair were randomly assigned to saline, BMSCs-Exos or LIPUS-BMSC-Exos injection therapy. Histological, immunofluorescent and biomechanical tests were detected to investigate the effect of exosomes injection on BTI healing and muscle fatty infiltration of the repaired rotator cuff. In vitro, native BMSCs were incubated with BMSC-Exos or LIPUS-BMSC-Exos and then chondrogenic/adipogenic differentiation were observed. Further, quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect the chondrogenesis/adipogenesis-related miRNA profiles of LIPUS-BMSC-Exos and BMSC-Exos. The chondrogenic/adipogenic potential of the key miRNA was verified through function recover test with its mimic and inhibitor. Results The results indicated that the biomechanical properties of the supraspinatus tendon-humeral junction were significantly improved in the LIPUS-BMSC-Exos group than that of the BMSCs-Exos group. The LIPUS-BMSC-Exos group also exhibited a higher histological score and more newly regenerated fibrocartilage at the repair site at postoperative 2 and 4 weeks and less fatty infiltration at 4 weeks than the BMSCs-Exos group. In vitro, co-culture of BMSCs with LIPUS-BMSC-Exos could significantly promote BMSCs chondrogenic differentiation and inhibit adipogenic differentiation. Subsequently, qRT-PCR revealed significantly higher enrichment of chondrogenic miRNAs and less enrichment of adipogenic miRNAs in LIPUS-BMSC-Exos compared with BMSC-Exos. Moreover, we demonstrated that this chondrogenesis-inducing potential was primarily attributed to miR-140, one of the most abundant miRNAs in LIPUS-BMSC-Exos. Conclusion LIPUS-preconditioned BMSC-Exos can effectively promote BTI fibrocartilage regeneration and ameliorate supraspinatus fatty infiltration by positive regulation of pro-chondrogenesis and anti-adipogenesis, which was primarily through delivering miR-140. The translational potential of this article These findings propose an innovative "LIPUS combined Exosomes strategy" for rotator cuff healing which combines both physiotherapeutic and biotherapeutic advantages. This strategy possesses a good translational potential as a local injection of LIPUS preconditioned BMSC-derived Exos during operation can be not only efficient for promoting fibrocartilage regeneration and ameliorating rotator cuff fatty infiltration, but also time-saving, simple and convenient for patients.
Collapse
Affiliation(s)
- Bing Wu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Tao Zhang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Huabin Chen
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Xin Shi
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Changbiao Guan
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Jianzhong Hu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Mobile Health Ministry of Education - China Mobile Joint Laboratory, Changsha, 410008, Hunan Province, China
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| |
Collapse
|
9
|
Song W, Guo Y, Liu W, Yao Y, Zhang X, Cai Z, Yuan C, Wang X, Wang Y, Jiang X, Wang H, Yu W, Li H, Zhu Y, Kong L, He Y. Circadian Rhythm-Regulated ADSC-Derived sEVs and a Triphasic Microneedle Delivery System to Enhance Tendon-to-Bone Healing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408255. [PMID: 39120049 DOI: 10.1002/adma.202408255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/23/2024] [Indexed: 08/10/2024]
Abstract
Modulating the inflammatory microenvironment to reconstruct the fibrocartilaginous layer while promoting tendon repair is crucial for enhancing tendon-to-bone healing in rotator cuff repair (RCR), a persistent challenge in orthopedics. Small extracellular vesicles (sEVs) hold significant potential to modulate inflammation, yet the efficient production of highly bioactive sEVs remains a substantial barrier to their clinical application. Moreover, achieving minimally invasive local delivery of sEVs to the tendon-to-bone interface presents significant technical difficulties. Herein, the circadian rhythm of adipose-derived stem cells is modulated to increase the yield and enhance the inflammatory regulatory capacity of sEVs. Circadian rhythm-regulated sEVs (CR-sEVs) enhance the cyclic adenosine monophosphate signaling pathway in macrophage (Mφ) via platelet factor 4 delivery, thereby inhibiting Mφ M1 polarization. Subsequently, a triphasic microneedle (MN) scaffold with a tip, stem, and base is designed for the local delivery of CR-sEVs (CR-sEVs/MN) at the tendon-to-bone junction, incorporating tendon-derived decellularized extracellular matrix in the base to facilitate tendon repair. CR-sEVs/MN mitigates inflammation, promotes fibrocartilage regeneration, and enhances tendon healing, thereby improving biomechanical strength and shoulder joint function in a rat RCR model. Combining CR-sEVs with this triphasic microneedle delivery system presents a promising strategy for enhancing tendon-to-bone healing in clinical settings.
Collapse
Affiliation(s)
- Wei Song
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Ying Guo
- Department of Cardiology, Heart Center, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Wencai Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Yijing Yao
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China
| | - Xuancheng Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Zhuochang Cai
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Chenrui Yuan
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Xin Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Yifei Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Xiping Jiang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Haoyuan Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Weilin Yu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Haiyan Li
- Chemical and Environmental Engineering Department, School of Engineering, STEM College, RMIT University, 124 La Trobe St., Melbourne, Victoria, 3000, Australia
| | - Yanlun Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Lingzhi Kong
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Yaohua He
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
- Department of Orthopedic Surgery, Jinshan District Central Hospital affiliated to Shanghai University of Medicine & Health Sciences, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, 201500, P. R. China
| |
Collapse
|
10
|
Cui M, Sun Y, Zhang X, Yang P, Jiang W. Osteochondral tissue engineering in translational practice: histological assessments and scoring systems. Front Bioeng Biotechnol 2024; 12:1434323. [PMID: 39157444 PMCID: PMC11327087 DOI: 10.3389/fbioe.2024.1434323] [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: 05/17/2024] [Accepted: 07/15/2024] [Indexed: 08/20/2024] Open
Abstract
Osteochondral lesions are common pathological alterations in synovial joints. Different techniques have been designed to achieve osteochondral repair, and tissue-engineered osteochondral grafts have shown the most promise. Histological assessments and related scoring systems are crucial for evaluating the quality of regenerated tissue, and the interpretation and comparison of various repair techniques require the establishment of a reliable and widely accepted histological method. To date, there is still no consensus on the type of histological assessment and scoring system that should be used for osteochondral repair. In this review, we summarize common osteochondral staining methods, discuss the criteria regarding high-quality histological images, and assess the current histological scoring systems for osteochondral regeneration. Safranin O/Fast green is the most widely used staining method for the cartilage layer, whereas Gomori and Van Gieson staining detect new bone formation. We suggest including the graft-host interface and more sections together with the basic histological information for images. An ideal scoring system should analyze both the cartilage and bone regions, especially for the subchondral bone plate. Furthermore, histological assessments should be performed over a longer period of time to minimize discrepancies caused by defect size and animal species.
Collapse
Affiliation(s)
- Mengying Cui
- The Second Hospital of Jilin University, Jilin, China
| | - Yang Sun
- Orthopedic Medical Center, The Second Hospital of Jilin University, Jilin, China
| | | | - Pengju Yang
- Orthopedic Medical Center, The Second Hospital of Jilin University, Jilin, China
| | - Weibo Jiang
- Orthopedic Medical Center, The Second Hospital of Jilin University, Jilin, China
| |
Collapse
|
11
|
Swain HN, Boyce PD, Bromet BA, Barozinksy K, Hance L, Shields D, Olbricht GR, Semon JA. Mesenchymal stem cells in autoimmune disease: A systematic review and meta-analysis of pre-clinical studies. Biochimie 2024; 223:54-73. [PMID: 38657832 DOI: 10.1016/j.biochi.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/08/2024] [Accepted: 04/20/2024] [Indexed: 04/26/2024]
Abstract
Mesenchymal Stem Cells (MSCs) are of interest in the clinic because of their immunomodulation capabilities, capacity to act upstream of inflammation, and ability to sense metabolic environments. In standard physiologic conditions, they play a role in maintaining the homeostasis of tissues and organs; however, there is evidence that they can contribute to some autoimmune diseases. Gaining a deeper understanding of the factors that transition MSCs from their physiological function to a pathological role in their native environment, and elucidating mechanisms that reduce their therapeutic relevance in regenerative medicine, is essential. We conducted a Systematic Review and Meta-Analysis of human MSCs in preclinical studies of autoimmune disease, evaluating 60 studies that included 845 patient samples and 571 control samples. MSCs from any tissue source were included, and the study was limited to four autoimmune diseases: multiple sclerosis, rheumatoid arthritis, systemic sclerosis, and lupus. We developed a novel Risk of Bias tool to determine study quality for in vitro studies. Using the International Society for Cell & Gene Therapy's criteria to define an MSC, most studies reported no difference in morphology, adhesion, cell surface markers, or differentiation into bone, fat, or cartilage when comparing control and autoimmune MSCs. However, there were reported differences in proliferation. Additionally, 308 biomolecules were differentially expressed, and the abilities to migrate, invade, and form capillaries were decreased. The findings from this study could help to explain the pathogenic mechanisms of autoimmune disease and potentially lead to improved MSC-based therapeutic applications.
Collapse
Affiliation(s)
- Hailey N Swain
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Parker D Boyce
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Bradley A Bromet
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Kaiden Barozinksy
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Lacy Hance
- Department of Biological Sciences, Missouri University of Science and Technology, USA
| | - Dakota Shields
- Department of Mathematics and Statistics, Missouri University of Science and Technology, USA
| | - Gayla R Olbricht
- Department of Mathematics and Statistics, Missouri University of Science and Technology, USA
| | - Julie A Semon
- Department of Biological Sciences, Missouri University of Science and Technology, USA.
| |
Collapse
|
12
|
Zupan J, Stražar K. Synovium-Derived and Bone-Derived Mesenchymal Stem/Stromal Cells from Early OA Patients Show Comparable In Vitro Properties to Those of Non-OA Patients. Cells 2024; 13:1238. [PMID: 39120270 PMCID: PMC11311703 DOI: 10.3390/cells13151238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Degenerative disorders like osteoarthritis (OA) might impair the ability of tissue-resident mesenchymal stem/stromal cells (MSCs) for tissue regeneration. As primary cells with MSC-like properties are exploited for patient-derived stem cell therapies, a detailed evaluation of their in vitro properties is needed. Here, we aimed to compare synovium-derived and bone-derived MSCs in early hip OA with those of patients without OA (non-OA). Tissues from three synovial sites of the hip (paralabral synovium, cotyloid fossa, inner surface of peripheral capsule) were collected along with peripheral trabecular bone from 16 patients undergoing hip arthroscopy (8 early OA and 8 non-OA patients). Primary cells isolated from tissues were compared using detailed in vitro analyses. Gene expression profiling was performed for the skeletal stem cell markers podoplanin (PDPN), CD73, CD164 and CD146 as well as for immune-related molecules to assess their immunomodulatory potential. Synovium-derived and bone-derived MSCs from early OA patients showed comparable clonogenicity, cumulative population doublings, osteogenic, adipogenic and chondrogenic potential, and immunophenotype to those of non-OA patients. High PDPN/low CD146 profile (reminiscent of skeletal stem cells) was identified mainly for non-OA MSCs, while low PDPN/high CD146 mainly defined early OA MSCs. These data suggest that MSCs from early OA patients are not affected by degenerative changes in the hip. Moreover, the synovium represents an alternative source of MSCs for patient-derived stem cell therapies, which is comparable to bone. The expression profile reminiscent of skeletal stem cells suggests the combination of low PDPN and high CD146 as potential biomarkers in early OA.
Collapse
Affiliation(s)
- Janja Zupan
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Askerceva 7, 1000 Ljubljana, Slovenia;
| | - Klemen Stražar
- Department of Orthopaedic Surgery, University Medical Centre Ljubljana, Zaloska 9, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| |
Collapse
|
13
|
Wu YQ, Wang J. Sequential release of transforming growth factor β1 and fibroblast growth factor 2 from nanofibrous scaffolds induces cartilage differentiation of mouse adipose-derived stem cells. Biointerphases 2024; 19:041002. [PMID: 39051723 DOI: 10.1116/6.0003687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024] Open
Abstract
Once damaged, cartilage has poor intrinsic capacity to repair itself. Current cartilage repair strategies cannot restore the damaged tissue sufficiently. It is hypothesized that biomimetic scaffolds, which can recapitulate important properties of the cartilage extracellular matrix, play a beneficial role in supporting cell behaviors such as growth, cartilage differentiation, and integration with native cartilage, ultimately facilitating tissue recovery. Adipose-derived stem cells regenerated cartilage upon the sequential release of transforming growth factor β1(TGFβ1) and fibroblast growth factor 2(FGF2) using a nanofibrous scaffold, in order to get the recovery of functional cartilage. Experiments in vitro have demonstrated that the release sequence of growth factors FGF2 to TGFβ1 is the most essential to promote adipose-derived stem cells into chondrocytes that then synthesize collagen II. Mouse subcutaneous implantation indicated that the treatment sequence of FGF2 to TGFβ1 was able to significantly induce multiple increase in cartilage regeneration in vivo. This result demonstrates that the group treated with FGF2 to TGFβ1 released from a nanofibrous scaffold provides a good strategy for cartilage regeneration by making a favorable microenvironment for cell growth and cartilage regeneration.
Collapse
Affiliation(s)
- Yun-Qi Wu
- Department of Orthopaedics, Hunan Engineering Laboratory of Advanced Artificial Osteo-Materials, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Jun Wang
- Department of Gastroenterology, Hunan Engineering Laboratory of Advanced Artificial Osteo-Materials, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
14
|
Kawabata S, Nakasa T, Nekomoto A, Yimiti D, Miyaki S, Adachi N. Osteophyte Cartilage as a Potential Source for Minced Cartilage Implantation: A Novel Approach for Articular Cartilage Repair in Osteoarthritis. Int J Mol Sci 2024; 25:5563. [PMID: 38791601 PMCID: PMC11122408 DOI: 10.3390/ijms25105563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/17/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024] Open
Abstract
Osteoarthritis (OA) is a common joint disorder characterized by cartilage degeneration, often leading to pain and functional impairment. Minced cartilage implantation (MCI) has emerged as a promising one-step alternative for large cartilage defects. However, the source of chondrocytes for MCI remains a challenge, particularly in advanced OA, as normal cartilage is scarce. We performed in vitro studies to evaluate the feasibility of MCI using osteophyte cartilage, which is present in patients with advanced OA. Osteophyte and articular cartilage samples were obtained from 22 patients who underwent total knee arthroplasty. Chondrocyte migration and proliferation were assessed using cartilage fragment/atelocollagen composites to compare the characteristics and regenerative potential of osteophytes and articular cartilage. Histological analysis revealed differences in cartilage composition between osteophytes and articular cartilage, with higher expression of type X collagen and increased chondrocyte proliferation in the osteophyte cartilage. Gene expression analysis identified distinct gene expression profiles between osteophytes and articular cartilage; the expression levels of COL2A1, ACAN, and SOX9 were not significantly different. Chondrocytes derived from osteophyte cartilage exhibit enhanced proliferation, and glycosaminoglycan production is increased in both osteophytes and articular cartilage. Osteophyte cartilage may serve as a viable alternative source of MCI for treating large cartilage defects in OA.
Collapse
Affiliation(s)
- Shingo Kawabata
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City 734-8551, Japan; (S.K.); (A.N.); (D.Y.); (N.A.)
| | - Tomoyuki Nakasa
- Department of Artificial Joints and Biomaterials, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City 734-8551, Japan
| | - Akinori Nekomoto
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City 734-8551, Japan; (S.K.); (A.N.); (D.Y.); (N.A.)
| | - Dilimulati Yimiti
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City 734-8551, Japan; (S.K.); (A.N.); (D.Y.); (N.A.)
| | - Shigeru Miyaki
- Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima City 734-8551, Japan;
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City 734-8551, Japan; (S.K.); (A.N.); (D.Y.); (N.A.)
| |
Collapse
|
15
|
He Z, Li H, Zhang Y, Gao S, Liang K, Su Y, Du Y, Wang D, Xing D, Yang Z, Lin J. Enhanced bone regeneration via endochondral ossification using Exendin-4-modified mesenchymal stem cells. Bioact Mater 2024; 34:98-111. [PMID: 38186959 PMCID: PMC10770633 DOI: 10.1016/j.bioactmat.2023.12.007] [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: 09/07/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 01/09/2024] Open
Abstract
Nonunions and delayed unions pose significant challenges in orthopedic treatment, with current therapies often proving inadequate. Bone tissue engineering (BTE), particularly through endochondral ossification (ECO), emerges as a promising strategy for addressing critical bone defects. This study introduces mesenchymal stem cells overexpressing Exendin-4 (MSC-E4), designed to modulate bone remodeling via their autocrine and paracrine functions. We established a type I collagen (Col-I) sponge-based in vitro model that effectively recapitulates the ECO pathway. MSC-E4 demonstrated superior chondrogenic and hypertrophic differentiation and enhanced the ECO cell fate in single-cell sequencing analysis. Furthermore, MSC-E4 encapsulated in microscaffold, effectively facilitated bone regeneration in a rat calvarial defect model, underscoring its potential as a therapeutic agent for bone regeneration. Our findings advocate for MSC-E4 within a BTE framework as a novel and potent approach for treating significant bone defects, leveraging the intrinsic ECO process.
Collapse
Affiliation(s)
- Zihao He
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Hui Li
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Yuanyuan Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Shuang Gao
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Yiqi Su
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Du Wang
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Zhen Yang
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Jianhao Lin
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| |
Collapse
|
16
|
Gao H, Wang L, Lin Z, Jin H, Lyu Y, Kang Y, Zhu T, Zhao J, Jiang J. Bi-lineage inducible and immunoregulatory electrospun fibers scaffolds for synchronous regeneration of tendon-to-bone interface. Mater Today Bio 2023; 22:100749. [PMID: 37545569 PMCID: PMC10400930 DOI: 10.1016/j.mtbio.2023.100749] [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: 04/27/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/08/2023] Open
Abstract
Facilitating regeneration of the tendon-to-bone interface can reduce the risk of postoperative retear after rotator cuff repair. Unfortunately, undesirable inflammatory responses following injury, difficulties in fibrocartilage regeneration, and bone loss in the surrounding area are major contributors to suboptimal tendon-bone healing. Thus, the development of biomaterials capable of regulating macrophage polarization to a favorable phenotype and promoting the synchronous regeneration of the tendon-to-bone interface is currently a top priority. Here, strontium-doped mesoporous bioglass nanoparticles (Sr-MBG) were synthesized through a modulated sol-gel method and Bi-lineage Inducible and Immunoregulatory Electrospun Fibers Scaffolds (BIIEFS) containing Sr-MBG were fabricated. The BIIEFS were biocompatible, showed sustained release of multiple types of bioactive ions, enhanced osteogenic and chondrogenic differentiation of mesenchymal stem cells (MSCs), and facilitated macrophage polarization towards the M2 phenotype in vitro. The implantation of BIIEFS at the torn rotator cuff resulted in greater numbers of M2 macrophages and the synchronous regeneration of tendon, fibrocartilage, and bone at the tendon-to-bone interface, leading to a significant improvement in the biomechanical strength of the supraspinatus tendon-humerus complexes. Our research offers a feasible strategy to fabricate immunoregulatory and multi-lineage inducible electrospun fibers scaffolds incorporating bioglass nanoparticles for the regeneration of soft-to-hard tissue interfaces.
Collapse
Affiliation(s)
- Haihan Gao
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
| | - Liren Wang
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, No. 227 South Chongqing Road, Shanghai, 200025, China
| | - Zhiqi Lin
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Haocheng Jin
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yangbao Lyu
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yuhao Kang
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Tonghe Zhu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, 333 Longteng Rd., Shanghai, 201620, PR China
| | - Jinzhong Zhao
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai, 201306, China
| | - Jia Jiang
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
17
|
Liu W, Venkatesan JK, Amini M, Oláh T, Schmitt G, Madry H, Cucchiarini M. Effects of rAAV-Mediated Overexpression of sox9 and TGF- ß via Alginate Hydrogel-Guided Vector Delivery on the Chondroreparative Activities of Human Bone Marrow-Derived Mesenchymal Stromal Cells. J Tissue Eng Regen Med 2023; 2023:4495697. [PMID: 40226403 PMCID: PMC11919174 DOI: 10.1155/2023/4495697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/28/2023] [Accepted: 07/22/2023] [Indexed: 04/15/2025]
Abstract
Recombinant adeno-associated virus (rAAV) vectors have a strong potential to promote the healing of traumatic cartilage defects and osteoarthritic lesions upon delivery and overexpression of therapeutic genes from suitable biomaterials that support a controlled release of the candidate constructs. The goal of the present work is to examine whether the administration of chondrogenic rAAV sox9 and rAAV TGF-ß gene vehicles via alginate hydrogel-guided vector delivery stimulates the biological and chondroreparative activities of human bone marrow-derived mesenchymal stromal cells (hMSCs) as a source of improved reparative cells for future implantation in sites of cartilage damage. The delivery of rAAV using an alginate (AlgPH155) hydrogel system is successfully achieved in hMSCs over time (21 days), leading to the effective overexpression of sox9 and TGF-ß that significantly increases the proliferation and chondrogenic differentiation activities of the cells relative to control (marker lacZ) gene transfer while advantageously preventing premature hypertrophy, osteogenesis, and mineralization. This study reveals the potential of alginate hydrogel-based systems to control the delivery of rAAV (sox9 and TGF-ß) gene vectors to adeptly trigger the chondroreparative activities of hMSCs for future applications that aim at improving cartilage repair.
Collapse
Affiliation(s)
- Wei Liu
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Homburg/Saar, Germany
| | - Jagadeesh K. Venkatesan
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Homburg/Saar, Germany
| | - Mahnaz Amini
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Homburg/Saar, Germany
| | - Tamás Oláh
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Homburg/Saar, Germany
| | - Gertrud Schmitt
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
18
|
Sonmez Kaplan S, Sazak Ovecoglu H, Genc D, Akkoc T. TNF-α, IL-1B and IL-6 affect the differentiation ability of dental pulp stem cells. BMC Oral Health 2023; 23:555. [PMID: 37568110 PMCID: PMC10422753 DOI: 10.1186/s12903-023-03288-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND This in vitro study examined the effect of the inflammatory cytokines (tumour necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6) on osteogenic, chondrogenic, and adipogenic differentiation of dental pulp stem cells (DPSCs) which have significant relevance in future regenerative therapies. METHODS DPSCs were isolated from the impacted third molar dental pulp and determined with flow cytometry analysis. DPSCs were divided into into 5 main groups with 3 subdivisions for each group making a total of 15 groups. Experimental groups were stimulated with TNF-α, IL-1β, IL-6, and a combination of all three to undergo osteogenic, chondrogenic, and adipogenic differentiation protocols. Next, the differentiation of each group was examined with different staining procedures under a light microscope. Histological analysis of osteogenic, chondrogenic, and adipogenic differentiated pellets was assessed using a modified Bern score. Statistical significance determined using one-way analysis of variance, and correlations were assessed using Pearson's test (two-tailed). RESULTS Stimulation with inflammatory cytokines significantly inhibited the osteogenic, chondrogenic and adipogenic differentiation of DPSCs in terms of matrix and cell formation resulting in weak staining than the unstimulated groups with inflammatory cytokines. On contrary, the unstimulated groups of MSCs have shown to be highly proliferative ability in terms of osteogenic, chondrogenic, and adipogenic differentiation. CONCLUSIONS DPSCs have high osteogenic, chondrogenic, and adipogenic differentiation capabilities. Pretreatment with inflammatory cytokines decreases the differentiation ability in vitro, thus inhibiting tissue formation.
Collapse
Affiliation(s)
- Sema Sonmez Kaplan
- Department of Endodontics, Faculty of Dentistry, Biruni University, 10. Yıl Caddesi Protokol Yolu No: 45, 34010, Topkapı, Istanbul, Turkey.
| | - Hesna Sazak Ovecoglu
- Faculty of Dentistry Department of Endodontics, Marmara University, Istanbul, Turkey
| | - Deniz Genc
- Department of Pediatric Health & Diseases Faculty of Health Sciences, Muğla Sıtkı Koçman University, Mugla, Turkey
- Research Laboratories Center, Immunology and Stem Cell Laboratory, Muğla Sıtkı Koçman University, Mugla, Turkey
| | - Tunc Akkoc
- Immunology Department, Marmara University Medical Faculty, Istanbul, Turkey
| |
Collapse
|
19
|
Bonato A, Fisch P, Ponta S, Fercher D, Manninen M, Weber D, Eklund KK, Barreto G, Zenobi‐Wong M. Engineering Inflammation-Resistant Cartilage: Bridging Gene Therapy and Tissue Engineering. Adv Healthc Mater 2023; 12:e2202271. [PMID: 36841937 PMCID: PMC11468558 DOI: 10.1002/adhm.202202271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 01/09/2023] [Indexed: 02/27/2023]
Abstract
Articular cartilage defects caused by traumatic injury rarely heal spontaneously and predispose into post-traumatic osteoarthritis. In the current autologous cell-based treatments the regenerative process is often hampered by the poor regenerative capacity of adult cells and the inflammatory state of the injured joint. The lack of ideal treatment options for cartilage injuries motivated the authors to tissue engineer a cartilage tissue which would be more resistant to inflammation. A clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 knockout of TGF-β-activated kinase 1 (TAK1) gene in polydactyly chondrocytes provides multivalent protection against the signals that activate the pro-inflammatory and catabolic NF-κB pathway. The TAK1-KO chondrocytes encapsulate into a hyaluronan hydrogel deposit copious cartilage extracellular matrix proteins and facilitate integration onto native cartilage, even under proinflammatory conditions. Furthermore, when implanted in vivo, compared to WT fewer pro-inflammatory M1 macrophages invade the cartilage, likely due to the lower levels of cytokines secreted by the TAK1-KO polydactyly chondrocytes. The engineered cartilage thus represents a new paradigm-shift for the creation of more potent and functional tissues for use in regenerative medicine.
Collapse
Affiliation(s)
- Angela Bonato
- Department of Health Sciences and TechnologyETH ZürichZürich8093Switzerland
| | - Philipp Fisch
- Department of Health Sciences and TechnologyETH ZürichZürich8093Switzerland
| | - Simone Ponta
- Department of Health Sciences and TechnologyETH ZürichZürich8093Switzerland
| | - David Fercher
- Department of Health Sciences and TechnologyETH ZürichZürich8093Switzerland
| | | | - Daniel Weber
- Division of Hand SurgeryUniversity Children's HospitalZürich8032Switzerland
| | - Kari K. Eklund
- Orton Orthopedic Hospital HelsinkiHelsinki00280Finland
- Department of RheumatologyUniversity of Helsinki and Helsinki University HospitalHelsinki00014Finland
| | - Goncalo Barreto
- Orton Orthopedic Hospital HelsinkiHelsinki00280Finland
- Translational Immunology Research ProgramFaculty of MedicineUniversity of HelsinkiHelsinki00014Finland
| | - Marcy Zenobi‐Wong
- Department of Health Sciences and TechnologyETH ZürichZürich8093Switzerland
| |
Collapse
|
20
|
Stage HJ, Trappe S, Söllig K, Trachsel DS, Kirsch K, Zieger C, Merle R, Aschenbach JR, Gehlen H. Multilineage Differentiation Potential of Equine Adipose-Derived Stromal/Stem Cells from Different Sources. Animals (Basel) 2023; 13:ani13081352. [PMID: 37106915 PMCID: PMC10135324 DOI: 10.3390/ani13081352] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
The investigation of multipotent stem/stromal cells (MSCs) in vitro represents an important basis for translational studies in large animal models. The study's aim was to examine and compare clinically relevant in vitro properties of equine MSCs, which were isolated from abdominal (abd), retrobulbar (rb) and subcutaneous (sc) adipose tissue by collagenase digestion (ASCs-SVF) and an explant technique (ASCs-EXP). Firstly, we examined proliferation and trilineage differentiation and, secondly, the cardiomyogenic differentiation potential using activin A, bone morphogenetic protein-4 and Dickkopf-1. Fibroblast-like, plastic-adherent ASCs-SVF and ASCs-EXP were obtained from all sources. The proliferation and chondrogenic differentiation potential did not differ significantly between the isolation methods and localizations. However, abd-ASCs-EXP showed the highest adipogenic differentiation potential compared to rb- and sc-ASCs-EXP on day 7 and abd-ASCs-SVF a higher adipogenic potential compared to abd-ASCs-EXP on day 14. Osteogenic differentiation potential was comparable at day 14, but by day 21, abd-ASCs-EXP demonstrated a higher osteogenic potential compared to abd-ASCs-SVF and rb-ASCs-EXP. Cardiomyogenic differentiation could not be achieved. This study provides insight into the proliferation and multilineage differentiation potential of equine ASCs and is expected to provide a basis for future preclinical and clinical studies in horses.
Collapse
Affiliation(s)
- Hannah J Stage
- Equine Clinic, Surgery and Radiology, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Susanne Trappe
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Katharina Söllig
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Dagmar S Trachsel
- Clinical Unit of Equine Internal Medicine, Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Katharina Kirsch
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Cornelia Zieger
- Institute of Veterinary Pathology Department of Veterinary Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 15, 14163 Berlin, Germany
| | - Roswitha Merle
- Institute for Veterinary Epidemiology and Biostatistics, Department of Veterinary Medicine, Freie Universität Berlin, Königsweg 67, 14163 Berlin, Germany
| | - Jörg R Aschenbach
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| | - Heidrun Gehlen
- Equine Clinic, Surgery and Radiology, Department of Veterinary Medicine, Freie Universität Berlin, Oertzenweg 19b, 14163 Berlin, Germany
| |
Collapse
|
21
|
Hochmann S, Ou K, Poupardin R, Mittermeir M, Textor M, Ali S, Wolf M, Ellinghaus A, Jacobi D, Elmiger JAJ, Donsante S, Riminucci M, Schäfer R, Kornak U, Klein O, Schallmoser K, Schmidt-Bleek K, Duda GN, Polansky JK, Geissler S, Strunk D. The enhancer landscape predetermines the skeletal regeneration capacity of stromal cells. Sci Transl Med 2023; 15:eabm7477. [PMID: 36947595 DOI: 10.1126/scitranslmed.abm7477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Multipotent stromal cells are considered attractive sources for cell therapy and tissue engineering. Despite numerous experimental and clinical studies, broad application of stromal cell therapeutics is not yet emerging. A major challenge is the functional diversity of available cell sources. Here, we investigated the regenerative potential of clinically relevant human stromal cells from bone marrow (BMSCs), white adipose tissue, and umbilical cord compared with mature chondrocytes and skin fibroblasts in vitro and in vivo. Although all stromal cell types could express transcription factors related to endochondral ossification, only BMSCs formed cartilage discs in vitro that fully regenerated critical-size femoral defects after transplantation into mice. We identified cell type-specific epigenetic landscapes as the underlying molecular mechanism controlling transcriptional stromal differentiation networks. Binding sites of commonly expressed transcription factors in the enhancer and promoter regions of ossification-related genes, including Runt and bZIP families, were accessible only in BMSCs but not in extraskeletal stromal cells. This suggests an epigenetically predetermined differentiation potential depending on cell origin that allows common transcription factors to trigger distinct organ-specific transcriptional programs, facilitating forward selection of regeneration-competent cell sources. Last, we demonstrate that viable human BMSCs initiated defect healing through the secretion of osteopontin and contributed to transient mineralized bone hard callus formation after transplantation into immunodeficient mice, which was eventually replaced by murine recipient bone during final tissue remodeling.
Collapse
Affiliation(s)
- Sarah Hochmann
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Kristy Ou
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), T Cell Epigenetics, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Rodolphe Poupardin
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Michaela Mittermeir
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Martin Textor
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Salaheddine Ali
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Martin Wolf
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Agnes Ellinghaus
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Dorit Jacobi
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Juri A J Elmiger
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohematology, Goethe University Hospital, German Red Cross Blood Service Baden-Württemberg-Hessen gGmbH, 60323 Frankfurt am Main, Germany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106 Freiburg, Germany
| | - Uwe Kornak
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Oliver Klein
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
| | | | - Katharina Schmidt-Bleek
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Georg N Duda
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Julia K Polansky
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), T Cell Epigenetics, Augustenburger Platz 1, 13353 Berlin, Germany
- German Rheumatism Research Centre (DRFZ), 10117 Berlin, Germany
| | - Sven Geissler
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Center for Advanced Therapies (BECAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Dirk Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| |
Collapse
|
22
|
Wixmerten A, Miot S, Bittorf P, Wolf F, Feliciano S, Hackenberg S, Häusner S, Krenger W, Haug M, Martin I, Pullig O, Barbero A. Good Manufacturing Practice-compliant change of raw material in the manufacturing process of a clinically used advanced therapy medicinal product-a comparability study. Cytotherapy 2023; 25:548-558. [PMID: 36894437 DOI: 10.1016/j.jcyt.2023.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/01/2022] [Accepted: 01/08/2023] [Indexed: 03/09/2023]
Abstract
The development of medicinal products often continues throughout the different phases of a clinical study and may require challenging changes in raw and starting materials at later stages. Comparability between the product properties pre- and post-change thus needs to be ensured. Here, we describe and validate the regulatory compliant change of a raw material using the example of a nasal chondrocyte tissue-engineered cartilage (N-TEC) product, initially developed for treatment of confined knee cartilage lesions. Scaling up the size of N-TEC as required for the treatment of larger osteoarthritis defects required the substitution of autologous serum with a clinical-grade human platelet lysate (hPL) to achieve greater cell numbers necessary for the manufacturing of larger size grafts. A risk-based approach was performed to fulfill regulatory requirements and demonstrate comparability of the products manufactured with the standard process (autologous serum) already applied in clinical indications and the modified process (hPL). Critical attributes with regard to quality, purity, efficacy, safety and stability of the product as well as associated test methods and acceptance criteria were defined. Results showed that hPL added during the expansion phase of nasal chondrocytes enhances proliferation rate, population doublings and cell numbers at passage 2 without promoting the overgrowth of potentially contaminant perichondrial cells. N-TEC generated with the modified versus standard process contained similar content of DNA and cartilaginous matrix proteins with even greater expression levels of chondrogenic genes. The increased risk for tumorigenicity potentially associated with the use of hPL was assessed through karyotyping of chondrocytes at passage 4, revealing no chromosomal changes. Moreover, the shelf-life of N-TEC established for the standard process could be confirmed with the modified process. In conclusion, we demonstrated the introduction of hPL in the manufacturing process of a tissue engineered product, already used in a late-stage clinical trial. Based on this study, the national competent authorities in Switzerland and Germany accepted the modified process which is now applied for ongoing clinical tests of N-TEC. The described activities can thus be taken as a paradigm for successful and regulatory compliant demonstration of comparability in advanced therapy medicinal products manufacturing.
Collapse
Affiliation(s)
- Anke Wixmerten
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Sylvie Miot
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Patrick Bittorf
- Fraunhofer ISC - Translational Center Regenerative Therapies, Würzburg, Germany
| | - Francine Wolf
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Sandra Feliciano
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Stephan Hackenberg
- Department of Otorhinolaryngology, Head and Neck Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Sebastian Häusner
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Werner Krenger
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Martin Haug
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Oliver Pullig
- Fraunhofer ISC - Translational Center Regenerative Therapies, Würzburg, Germany; Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Andrea Barbero
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
23
|
Grogan SP, Glembotski NE, D'Lima DD. ALK-5 Inhibitors for Efficient Derivation of Mesenchymal Stem Cells from Human Embryonic Stem Cells. Tissue Eng Part A 2023; 29:127-140. [PMID: 36458467 DOI: 10.1089/ten.tea.2022.0164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Objectives: Successful tissue regeneration requires a clinically viable source of mesenchymal stem cells (MSCs). We explored activin receptor-like kinase (ALK)-5 inhibitors to rapidly derive an MSC-like phenotype with high cartilage forming capacity from a xeno-free human embryonic cell line. Methods: Embryonic stem cell (ESC) lines (H9 and HADC100) were treated with the ALK-5 inhibitor SB431542; HADC100 cells were additionally treated with ALK-5 inhibitors SB525334 or GW788388. Cells were then seeded upon human fibronectin in the presence of fibroblast growth factor 2 (FGF2) in a serum-free medium. Flow cytometry was used to assess MSC markers (positive for CD73, CD90, and CD105; negative for CD34 and CD45). Differentiation status was assessed through quantitative polymerase chain reaction. Cartilage forming capacity was determined in high-density pellet cultures, in fibrin gels containing extracellular matrix (fibrin-ECM), and after implantation in ex vivo human osteoarthritic cartilage. Gene expression, histology, and immunostaining were used to assess cartilage phenotype, tissue regeneration, and integration. Results: Exposure to all three ALK-5 inhibitors lead to expression of mesodermal gene markers and differentiation into MSC-like cells (embryonic stem cell-derived mesenchymal stem cells [ES-MSCs]) based on surface marker expression. ES-MSC in pellet cultures or in fibrin-ECM gels expressed high levels of chondrogenic genes: COL2A1, ACAN, and COMP; and low levels of COL1A1 and RUNX2. Cell pellets or fibrin constructs implanted into ex vivo human osteoarthritic cartilage defects produced GAG-rich (safranin O positive) and collagen type II-positive neocartilage tissues that integrated well with native diseased tissue. Conclusions: We developed a protocol for rapid differentiation of xeno-free ESC into MSC-like cells with high cartilage forming capacity with potential for clinical applications. Impact statement Osteoarthritis (OA) is a common disease resulting in significant disability and no approved disease modifying treatment (other than total joint replacement). Embryonic stem cell-derived cell therapy has the potential to benefit patients with cartilage lesions leading to OA and may prevent or delay the need for total joint replacement.
Collapse
Affiliation(s)
- Shawn P Grogan
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, California, USA
| | - Nicholas E Glembotski
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, California, USA
| | - Darryl D D'Lima
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, California, USA
| |
Collapse
|
24
|
Kutaish H, Tscholl PM, Cosset E, Bengtsson L, Braunersreuther V, Mor FM, Laedermann J, Furfaro I, Stafylakis D, Hannouche D, Gerstel E, Krause KH, Assal M, Menetrey J, Tieng V. Articular Cartilage Repair After Implantation of Hyaline Cartilage Beads Engineered From Adult Dedifferentiated Chondrocytes: Cartibeads Preclinical Efficacy Study in a Large Animal Model. Am J Sports Med 2023; 51:237-249. [PMID: 36592016 DOI: 10.1177/03635465221138099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Chondrocyte-based cell therapy to repair cartilage has been used for >25 years despite current limitations. This work presents a new treatment option for cartilage lesions. HYPOTHESIS High-quality hyaline cartilage microtissues called Cartibeads are capable of treating focal chondral lesions once implanted in the defect, by complete fusion of Cartibeads among themselves and their integration with the surrounding native cartilage and subchondral bone. STUDY DESIGN Controlled laboratory study. METHODS Cartibeads were first produced from human donors and characterized using histology (safranin O staining of glycosaminoglycan [GAG] and immunohistochemistry of collagen I and II) and GAG dosage. Cartibeads from 6 Göttingen minipigs were engineered and implanted in an autologous condition in the knee (4 or 5 lesions per knee). One group was followed up for 3 months and the other for 6 months. Feasibility and efficacy were measured using histological analysis and macroscopic and microscopic scores. RESULTS Cartibeads revealed hyaline features with strong staining of GAG and collagen II. High GAG content was obtained: 24.6-µg/mg tissue (wet weight), 15.52-µg/mg tissue (dry weight), and 35 ± 3-µg GAG/bead (mean ± SD). Histological analysis of Göttingen minipigs showed good integration of Cartibeads grafts at 3 and 6 months after implantation. The Bern Score of the histological assay comparing grafted versus empty lesions was significant at 3 months (grafted, n = 10; nongrafted, n = 4; score, 3.3 and 5.3, respectively) and 6 months (grafted, n = 11; nongrafted, n = 3; score, 1.6 and 5.1). CONCLUSION We developed an innovative 3-step method allowing, for the first time, the use of fully dedifferentiated adult chondrocytes with a high number of cell passage (owing to the extensive amplification in culture). Cartibeads engineered from chondrocytes hold potential as an advanced therapy medicinal product for treating cartilage lesions with established efficacy. CLINICAL RELEVANCE This successful preclinical study, combined with standardized manufacturing of Cartibeads according to good manufacturing practice guidelines, led to the approval of first-in-human clinical trial by the ethics committee and local medical authority. The generated data highlighted a promising therapy to treat cartilage lesions from a small amount of starting biopsy specimen. With our innovative cell amplification technology, very large lesions can be treated, and older active patients can benefit from it.
Collapse
Affiliation(s)
- Halah Kutaish
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland. University Medical Center, University of Geneva, Geneva, Switzerland. Foot and Ankle Surgery Centre, Centre Assal, Clinique La Colline, Hirslanden Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Philippe Matthias Tscholl
- Department of Orthopaedics Surgery, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Erika Cosset
- University Medical Center, University of Geneva, Geneva, Switzerland. Laboratory of Tumor Immunology, Oncology Department, Center for Translational Research in Onco- Hematology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Laura Bengtsson
- University Medical Center, University of Geneva, Geneva, Switzerland. Vanarix SA, Lausanne, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Vincent Braunersreuther
- Service of Clinical Pathology, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Flavio Maurizio Mor
- Tissue Engineering Laboratory, HEPIA/HES-SO, University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Jeremy Laedermann
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Ivan Furfaro
- Laboratory for Soft Bioelectronic Interfaces, Institute of Microengineering, Institute of Bioengineering, Centre for Neuroprosthetics, École Polytechnique Fédeérale de Lausanne (EPFL), Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Dimitrios Stafylakis
- Department of Orthopaedics Surgery, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Didier Hannouche
- University Medical Center, University of Geneva, Geneva, Switzerland. Department of Orthopaedics Surgery, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Eric Gerstel
- University Medical Center, University of Geneva, Geneva, Switzerland. Clinique la Colline, Hirslanden, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland. University Medical Center, University of Geneva, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Mathieu Assal
- University Medical Center, University of Geneva, Geneva, Switzerland. Foot and Ankle Surgery Centre, Centre Assal, Clinique La Colline, Hirslanden Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Jacques Menetrey
- University Medical Center, University of Geneva, Geneva, Switzerland. Centre for Sports Medicine and Exercise, Clinique la Colline, Hirslanden, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Vannary Tieng
- University Medical Center, University of Geneva, Geneva, Switzerland. Vanarix SA, Lausanne, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
25
|
Zhang H, Li Q, Xu X, Zhang S, Chen Y, Yuan T, Zeng Z, Zhang Y, Mei Z, Yan S, Zhang L, Wei S. Functionalized Microscaffold-Hydrogel Composites Accelerating Osteochondral Repair through Endochondral Ossification. ACS APPLIED MATERIALS & INTERFACES 2022; 14:52599-52617. [PMID: 36394998 DOI: 10.1021/acsami.2c12694] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Osteochondral regeneration remains a key challenge because of the limited self-healing ability of the bone and its complex structure and composition. Biomaterials based on endochondral ossification (ECO) are considered an attractive candidate to promote bone repair because they can effectively address the difficulties in establishing vascularization and poor bone regeneration via intramembranous ossification (IMO). However, its clinical application is limited by the complex cellular behavior of ECO and the long time required for induction of the cell cycle. Herein, functionalized microscaffold-hydrogel composites are developed to accelerate the developmental bone growth process via recapitulating ECO. The design comprises arginine-glycine-aspartic acid (RGD)-peptide-modified microscaffolds loaded with kartogenin (KGN) and wrapped with a layer of RGD- and QK-peptide-comodified alginate hydrogel. These microscaffolds enhance the proliferation and aggregation behavior of the human bone marrow mesenchymal stem cells (hBMSCs); the controlled release of kartogenin induces the differentiation of hBMSCs into chondrocytes; and the hydrogel grafted with RGD and QK peptide facilitates chondrocyte hypertrophy, which creates a vascularized niche for osteogenesis and finally accelerates osteochondral repair in vivo. The findings provide an efficient bioengineering approach by sequentially modulating cellular ECO behavior for osteochondral defect repair.
Collapse
Affiliation(s)
- He Zhang
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Qian Li
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| | - Xiangliang Xu
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Siqi Zhang
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| | - Yang Chen
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| | - Tao Yuan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Tumor Biology, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Ziqian Zeng
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Yifei Zhang
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Zi Mei
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Shuang Yan
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Lei Zhang
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Shicheng Wei
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| |
Collapse
|
26
|
Lehoczky G, Trofin RE, Vallmajo-Martin Q, Chawla S, Pelttari K, Mumme M, Haug M, Egloff C, Jakob M, Ehrbar M, Martin I, Barbero A. In Vitro and Ectopic In Vivo Studies toward the Utilization of Rapidly Isolated Human Nasal Chondrocytes for Single-Stage Arthroscopic Cartilage Regeneration Therapy. Int J Mol Sci 2022; 23:ijms23136900. [PMID: 35805907 PMCID: PMC9267018 DOI: 10.3390/ijms23136900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/15/2022] [Accepted: 06/19/2022] [Indexed: 02/05/2023] Open
Abstract
Nasal chondrocytes (NCs) have a higher and more reproducible chondrogenic capacity than articular chondrocytes, and the engineered cartilage tissue they generate in vitro has been demonstrated to be safe in clinical applications. Here, we aimed at determining the feasibility for a single-stage application of NCs for cartilage regeneration under minimally invasive settings. In particular, we assessed whether NCs isolated using a short collagenase digestion protocol retain their potential to proliferate and chondro-differentiate within an injectable, swiftly cross-linked and matrix-metalloproteinase (MMP)-degradable polyethylene glycol (PEG) gel enriched with human platelet lysate (hPL). NC-hPL-PEG gels were additionally tested for their capacity to generate cartilage tissue in vivo and to integrate into cartilage/bone compartments of human osteochondral plugs upon ectopic subcutaneous implantation into nude mice. NCs isolated with a rapid protocol and embedded in PEG gels with hPL at low cell density were capable of efficiently proliferating and of generating tissue rich in glycosaminoglycans and collagen II. NC-hPL-PEG gels developed into hyaline-like cartilage tissues upon ectopic in vivo implantation and integrated with surrounding native cartilage and bone tissues. The delivery of NCs in PEG gels containing hPL is a feasible strategy for cartilage repair and now requires further validation in orthotopic in vivo models.
Collapse
Affiliation(s)
- Gyözö Lehoczky
- Department of Orthopaedic Surgery and Traumatology, University Hospital of Basel, 4031 Basel, Switzerland; (G.L.); (M.M.); (C.E.)
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
| | - Raluca Elena Trofin
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
| | - Queralt Vallmajo-Martin
- Department of Obstetrics, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (Q.V.-M.); (M.E.)
| | - Shikha Chawla
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
| | - Karoliina Pelttari
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
| | - Marcus Mumme
- Department of Orthopaedic Surgery and Traumatology, University Hospital of Basel, 4031 Basel, Switzerland; (G.L.); (M.M.); (C.E.)
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
- Department of Orthopaedic Surgery, University Children’s Hospital of Basel, 4056 Basel, Switzerland
| | - Martin Haug
- Department of Plastic, Reconstructive and Aesthetic Surgery and Hand Surgery, University Hospital of Basel, 4031 Basel, Switzerland;
| | - Christian Egloff
- Department of Orthopaedic Surgery and Traumatology, University Hospital of Basel, 4031 Basel, Switzerland; (G.L.); (M.M.); (C.E.)
| | | | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (Q.V.-M.); (M.E.)
| | - Ivan Martin
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
- Correspondence: ; Tel.: +41-61-2652384; Fax: +41-61-2653990
| | - Andrea Barbero
- Department of Biomedicine, Tissue Engineering Laboratory, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (R.E.T.); (S.C.); (K.P.); (A.B.)
| |
Collapse
|
27
|
Yang X, Liang X, Guo H, Ma L, Jian L, Zhao X, Wang J, Yang L, Meng Z, Jin Q. β2-Adrenergic receptor expression in subchondral bone of patients with varus knee osteoarthritis. Open Med (Wars) 2022; 17:1031-1044. [PMID: 35794997 PMCID: PMC9175016 DOI: 10.1515/med-2022-0498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
An important causative factor in osteoarthritis (OA) is the abnormal mechanical stress-induced bone remodeling of the subchondral bone. β2-adrenergic receptor (Adrb2) plays a major role in mechanical stresses that induce bone remodeling. The medial tibial plateau (MTP) and lateral tibial plateau (LTP) of patients with varus Knee osteoarthritis (KO) bear different mechanical stresses. The present study aimed to investigate the expression of Adrb2 in medial tibial plateau subchondral bone (MTPSB) and lateral tibial plateau subchondral bone (LTPSB) in patients with varus KO. A total of 30 tibial plateau samples from patients undergoing total knee arthroplasty for varus KO and MTPSB and LTPSB were studied. Statistical analysis was performed using paired sample t-tests. Safranin O-Fast Green staining and Micro-computed tomography showed significant differences in the bone structure between MTPSB and LTPSB. Tartrate-resistant acid phosphatase (TRAP)-positive cell density in MTPSB was higher than that in LTPSB. Immunohistochemistry, reverse transcription-quantitative polymerase chain reaction, and Western blot analysis revealed that compared to LTPSB, the levels of Adrb2, tyrosine hydroxylase (TH), and osteocalcin increased significantly in MTPSB. Double-labeling immunofluorescence showed Adrb2 was present in the majority of TRAP-positive multinuclear cells of the MTPSB. The expression of Adrb2 and TH was significantly higher in MTPSB than in LTPSB, confirming the involvement of these molecules in the development of OA.
Collapse
Affiliation(s)
- Xiaochun Yang
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Xuegang Liang
- Department of The General Hospital of Ningxia Medical University, Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Haohui Guo
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Long Ma
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Li Jian
- Department of Pathology, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Xin Zhao
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Jian Wang
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Lvlin Yang
- Department of The General Hospital of Ningxia Medical University, Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Zhiqiang Meng
- Department of The General Hospital of Ningxia Medical University, Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| | - Qunhua Jin
- Department of Orthopedics Ward 3, The General Hospital of Ningxia Medical University , Yinchuan , 750004, Ningxia , China
| |
Collapse
|
28
|
Shimomura S, Inoue H, Arai Y, Nakagawa S, Fujii Y, Kishida T, Shin-Ya M, Ichimaru S, Tsuchida S, Mazda O, Kubo T. Hypoxia promotes differentiation of pure cartilage from human induced pluripotent stem cells. Mol Med Rep 2022; 26:229. [PMID: 35593322 PMCID: PMC9178684 DOI: 10.3892/mmr.2022.12745] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 04/25/2022] [Indexed: 11/28/2022] Open
Abstract
While cartilage can be produced from induced pluripotent stem cells (iPSCs), challenges such as long culture periods and compromised tissue purity continue to prevail. The present study aimed to determine whether cartilaginous tissue could be produced from iPSCs under hypoxia and, if so, to evaluate its effects on cellular metabolism and purity of the produced tissue. Human iPSCs (hiPSCs) were cultured for cartilage differentiation in monolayers under normoxia or hypoxia (5% O2), and chondrocyte differentiation was evaluated using reverse transcription-quantitative PCR and fluorescence-activated cell sorting. Subsequently, cartilage differentiation of hiPSCs was conducted in 3D culture under normoxia or hypoxia (5% O2), and the formed cartilage-like tissues were evaluated on days 28 and 56 using histological analyses. Hypoxia suppressed the expression levels of the immature mesodermal markers brachyury (T) and forkhead box protein F1; however, it promoted the expression of the chondrogenic markers Acan and CD44. The number of sex-determining region Y-box 9-positive cells and the percentages of safranin O-positive and type 2 collagen-positive tissues increased under hypoxic conditions. Moreover, upon hypoxia-inducible factor (HIF)-1α staining, nuclei of tissues cultured under hypoxia stained more deeply compared with those of tissues cultured under normoxia. Overall, these findings indicated that hypoxia not only enhanced cartilage matrix production, but also improved tissue purity by promoting the expression of HIF-1α gene. Potentially, pure cartilage-like tissues could be produced rapidly and conveniently using this method.
Collapse
Affiliation(s)
- Seiji Shimomura
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Hiroaki Inoue
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Yuji Arai
- Department of Sports and Para‑Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Shuji Nakagawa
- Department of Sports and Para‑Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Yuta Fujii
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Tsunao Kishida
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Masaharu Shin-Ya
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Shohei Ichimaru
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Shinji Tsuchida
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| | - Toshikazu Kubo
- Department of Orthopedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602‑8566, Japan
| |
Collapse
|
29
|
Li K, Fan L, Lin J, Heng BC, Deng Z, Zheng Q, Zhang J, Jiang Y, Ge Z. Nanosecond pulsed electric fields prime mesenchymal stem cells to peptide ghrelin and enhance chondrogenesis and osteochondral defect repair in vivo. SCIENCE CHINA. LIFE SCIENCES 2022; 65:927-939. [PMID: 34586575 DOI: 10.1007/s11427-021-1983-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/22/2021] [Indexed: 01/07/2023]
Abstract
Mesenchymal stem cells (MSCs) are important cell sources in cartilage tissue development and homeostasis, and multiple strategies have been developed to improve MSCs chondrogenic differentiation with an aim of promoting cartilage regeneration. Here we report the effects of combining nanosecond pulsed electric fields (nsPEFs) followed by treatment with ghrelin (a hormone that stimulates release of growth hormone) to regulate chondrogenesis of MSCs. nsPEFs and ghrelin were observed to separately enhance the chondrogenesis of MSCs, and the effects were significantly enhanced when the bioelectric stimulation and hormone were combined, which in turn improved osteochondral tissue repair of these cells within Sprague Dawley rats. We further found that nsPEFs can prime MSCs to be more receptive to subsequent stimuli of differentiation by upregulated Oct4/Nanog and activated JNK signaling pathway. Ghrelin initiated chondrogenic differentiation by activation of ERK1/2 signaling pathway, and RNA-seq results indicated 243 genes were regulated, and JAK-STAT signaling pathway was involved. Interestingly, the sequential order of applying these two stimuli is critical, with nsPEFs pretreatment followed by ghrelin enhanced chondrogenesis of MSCs in vitro and subsequent cartilage regeneration in vivo, but not vice versa. This synergistic prochondrogenic effects provide us new insights and strategies for future cell-based therapies.
Collapse
Affiliation(s)
- Kejia Li
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Litong Fan
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Jianjing Lin
- Arthritis Clinical and Research Center, Peking University People's Hospital, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100871, China
| | - Boon Chin Heng
- Peking University School of Stomatology, Beijing, 100081, China
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Jue Zhang
- Institute of Biomechanics and Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Yangzi Jiang
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, China.
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, China.
| | - Zigang Ge
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, 100871, China.
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China.
| |
Collapse
|
30
|
Lan X, Liang Y, Vyhlidal M, Erkut EJN, Kunze M, Mulet-Sierra A, Osswald M, Ansari K, Seikaly H, Boluk Y, Adesida AB. In vitro maturation and in vivo stability of bioprinted human nasal cartilage. J Tissue Eng 2022; 13:20417314221086368. [PMID: 35599742 PMCID: PMC9122109 DOI: 10.1177/20417314221086368] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/23/2022] [Indexed: 11/25/2022] Open
Abstract
The removal of skin cancer lesions on the nose often results in the loss of nasal
cartilage. The cartilage loss is either surgically replaced with autologous
cartilage or synthetic grafts. However, these replacement options come with
donor-site morbidity and resorption issues. 3-dimensional (3D) bioprinting
technology offers the opportunity to engineer anatomical-shaped autologous nasal
cartilage grafts. The 3D bioprinted cartilage grafts need to embody a
mechanically competent extracellular matrix (ECM) to allow for surgical suturing
and resistance to contraction during scar tissue formation. We investigated the
effect of culture period on ECM formation and mechanical properties of 3D
bioprinted constructs of human nasal chondrocytes (hNC)-laden type I collagen
hydrogel in vitro and in vivo. Tissue-engineered nasal cartilage constructs
developed from hNC culture in clinically approved collagen type I and type III
semi-permeable membrane scaffold served as control. The resulting 3D bioprinted
engineered nasal cartilage constructs were comparable or better than the
controls both in vitro and in vivo. This study demonstrates that 3D bioprinted
constructs of engineered nasal cartilage are feasible options in nasal cartilage
reconstructive surgeries.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Yan Liang
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Margaret Vyhlidal
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Esra JN Erkut
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Martin Osswald
- Institute for Reconstructive Sciences in Medicine, Misericordia Community Hospital, Edmonton, AB, Canada
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Khalid Ansari
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Hadi Seikaly
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Yaman Boluk
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Adetola B Adesida
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
31
|
Brachtl G, Poupardin R, Hochmann S, Raninger A, Jürchott K, Streitz M, Schlickeiser S, Oeller M, Wolf M, Schallmoser K, Volk HD, Geissler S, Strunk D. Batch Effects during Human Bone Marrow Stromal Cell Propagation Prevail Donor Variation and Culture Duration: Impact on Genotype, Phenotype and Function. Cells 2022; 11:946. [PMID: 35326396 PMCID: PMC8946746 DOI: 10.3390/cells11060946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
Donor variation is a prominent critical issue limiting the applicability of cell-based therapies. We hypothesized that batch effects during propagation of bone marrow stromal cells (BMSCs) in human platelet lysate (hPL), replacing fetal bovine serum (FBS), can affect phenotypic and functional variability. We therefore investigated the impact of donor variation, hPL- vs. FBS-driven propagation and exhaustive proliferation, on BMSC epigenome, transcriptome, phenotype, coagulation risk and osteochondral regenerative function. Notably, propagation in hPL significantly increased BMSC proliferation, created significantly different gene expression trajectories and distinct surface marker signatures, already after just one passage. We confirmed significantly declining proliferative potential in FBS-expanded BMSC after proliferative challenge. Flow cytometry verified the canonical fibroblastic phenotype in culture-expanded BMSCs. We observed limited effects on DNA methylation, preferentially in FBS-driven cultures, irrespective of culture duration. The clotting risk increased over culture time. Moreover, expansion in xenogenic serum resulted in significant loss of function during 3D cartilage disk formation and significantly increased clotting risk. Superior chondrogenic function under hPL-conditions was maintained over culture. The platelet blood group and isoagglutinins had minor impact on BMSC function. These data demonstrate pronounced batch effects on BMSC transcriptome, phenotype and function due to serum factors, partly outcompeting donor variation after just one culture passage.
Collapse
Affiliation(s)
- Gabriele Brachtl
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| | - Rodolphe Poupardin
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| | - Sarah Hochmann
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| | - Anna Raninger
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| | - Karsten Jürchott
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.J.); (M.S.); (S.S.); (H.-D.V.); (S.G.)
| | - Mathias Streitz
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.J.); (M.S.); (S.S.); (H.-D.V.); (S.G.)
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Insel Riems, 17493 Greifswald, Germany
| | - Stephan Schlickeiser
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.J.); (M.S.); (S.S.); (H.-D.V.); (S.G.)
| | - Michaela Oeller
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (M.O.); (K.S.)
| | - Martin Wolf
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| | - Katharina Schallmoser
- Department of Transfusion Medicine and SCI-TReCS, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (M.O.); (K.S.)
| | - Hans-Dieter Volk
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.J.); (M.S.); (S.S.); (H.-D.V.); (S.G.)
- Berlin Center for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Sven Geissler
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.J.); (M.S.); (S.S.); (H.-D.V.); (S.G.)
- Berlin Center for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Dirk Strunk
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Cell Therapy Institute, Paracelsus Medical University (PMU), 5020 Salzburg, Austria; (G.B.); (R.P.); (S.H.); (A.R.); (M.W.)
| |
Collapse
|
32
|
Phenotypic and functional properties of dedifferentiated fat cells derived from infrapatellar fat pad. Regen Ther 2022; 19:35-46. [PMID: 35059478 PMCID: PMC8739472 DOI: 10.1016/j.reth.2021.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/12/2021] [Accepted: 12/20/2021] [Indexed: 01/03/2023] Open
Abstract
Introduction Mature adipocyte-derived dedifferentiated fat cells (DFATs) are mesenchymal stem cell (MSC)-like cells with high proliferative ability and multilineage differentiation potential. In this study, we first examined whether DFATs can be prepared from infrapatellar fat pad (IFP) and then compared phenotypic and functional properties of IFP-derived DFATs (IFP-DFATs) with those of subcutaneous adipose tissue (SC)-derived DFATs (SC-DFATs). Methods Mature adipocytes isolated from IFP and SC in osteoarthritis patients (n = 7) were cultured by ceiling culture method to generate DFATs. Obtained IFP-DFATs and SC-DFATs were subjected to flow cytometric and microarray analysis to compare their immunophenotypes and gene expression profiles. Cell proliferation assay and adipogenic, osteogenic, and chondrogenic differentiation assays were performed to evaluate their functional properties. Results DFATs could be prepared from IFP and SC with similar efficiency. IFP-DFATs and SC-DFATs exhibited similar immunophenotypes (CD73+, CD90+, CD105+, CD31-, CD45-, HLA-DR-) and tri-lineage (adipogenic, osteogenic, and chondrogenic) differentiation potential, consistent with the minimal criteria for defining MSCs. Microarray analysis revealed that the gene expression profiles in IFP-DFATs were very similar to those in SC-DFATs, although there were certain number of genes that showed different levels of expression. The proliferative activity in IFP-DFATs was significantly (p < 0.05) higher than that in the SC-DFATs. IFP-DFATs showed higher chondrogenic differentiation potential than SC-DFATs in regard to production of soluble galactosaminogalactan and gene expression of type II collagen. Conclusions IFP-DFATs showed higher cellular proliferative potential and higher chondrogenic differentiation capacity than SC-DFATs. IFP-DFAT cells may be an attractive cell source for chondrogenic regeneration.
Collapse
|
33
|
Baranovskii D, Demner J, Nürnberger S, Lyundup A, Redl H, Hilpert M, Pigeot S, Krasheninnikov M, Krasilnikova O, Klabukov I, Parshin V, Martin I, Lardinois D, Barbero A. Engineering of Tracheal Grafts Based on Recellularization of Laser-Engraved Human Airway Cartilage Substrates. Cartilage 2022; 13:19476035221075951. [PMID: 35189712 PMCID: PMC9137320 DOI: 10.1177/19476035221075951] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/20/2021] [Accepted: 12/25/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Implantation of tissue-engineered tracheal grafts represents a visionary strategy for the reconstruction of tracheal wall defects after resections and may develop into a last chance for a number of patients with severe cicatricial stenosis. The use of a decellularized tracheal substrate would offer an ideally stiff graft, but the matrix density would challenge efficient remodeling into a living cartilage. In this study, we hypothesized that the pores of decellularized laser-perforated tracheal cartilage (LPTC) tissues can be colonized by adult nasal chondrocytes (NCs) to produce new cartilage tissue suitable for the repair of tracheal defects. DESIGN Human, native tracheal specimens, isolated from cadaveric donors, were exposed to decellularized and laser engraving-controlled superficial perforation (300 μm depth). Human or rabbit NCs were cultured on the LPTCs for 1 week. The resulting revitalized tissues were implanted ectopically in nude mice or orthotopically in tracheal wall defects in rabbits. Tissues were assayed histologically and by microtomography analyses before and after implantation. RESULTS NCs were able to efficiently colonize the pores of the LPTCs. The extent of colonization (i.e., percentage of viable cells spanning >300 μm of tissue depth), cell morphology, and cartilage matrix deposition improved once the revitalized constructs were implanted ectopically in nude mice. LPTCs could be successfully grafted onto the tracheal wall of rabbits without any evidence of dislocation or tracheal stenosis, 8 weeks after implantation. Rabbit NCs, within the LPTCs, actively produced new cartilage matrix. CONCLUSION Implantation of NC-revitalized LPTCs represents a feasible strategy for the repair of tracheal wall defects.
Collapse
Affiliation(s)
- Denis Baranovskii
- Thoracic Surgery, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Regenerative Technologies and Biofabrication, National Medical Research Radiological Center, Obninsk, Russia
- Research and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia, Moscow, Russia
| | - Jan Demner
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sylvia Nürnberger
- Division of Trauma Surgery, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria
| | - Alexey Lyundup
- Research and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia, Moscow, Russia
- Department of Advanced Cell Technologies, Sechenov University, Moscow, Russia
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Vienna, Austria
| | - Morgane Hilpert
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sebastien Pigeot
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Michael Krasheninnikov
- Research and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia, Moscow, Russia
| | - Olga Krasilnikova
- Department of Regenerative Technologies and Biofabrication, National Medical Research Radiological Center, Obninsk, Russia
- Department of Advanced Cell Technologies, Sechenov University, Moscow, Russia
| | - Ilya Klabukov
- Department of Regenerative Technologies and Biofabrication, National Medical Research Radiological Center, Obninsk, Russia
- Department of Advanced Cell Technologies, Sechenov University, Moscow, Russia
| | - Vladimir Parshin
- Institute of Clinical Medicine, Sechenov University, Moscow, Russia
| | - 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
|
34
|
Cai Z, Li Y, Song W, He Y, Li H, Liu X. Anti-Inflammatory and Prochondrogenic In Situ-Formed Injectable Hydrogel Crosslinked by Strontium-Doped Bioglass for Cartilage Regeneration. ACS APPLIED MATERIALS & INTERFACES 2021; 13:59772-59786. [PMID: 34898167 DOI: 10.1021/acsami.1c20565] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Directed differentiation of bone marrow mesenchymal stem cells (BMSCs) toward chondrogenesis plays a predominant role in cartilage repair. However, the uncontrolled inflammatory response to implants is found to impair the stability of scaffolds and the cartilage regeneration outcome. Herein, we fabricated an injectable hydrogel crosslinked by strontium-doped bioglass (SrBG) to modulate both human BMSC (hBMSC) differentiation and the inflammatory response. The results revealed that the introduction of Sr ions could simultaneously enhance the proliferation of hBMSCs, upregulate cartilage-specific gene expression, and improve the secretion of glycosaminoglycan. Moreover, after cultured with SA/SrBG extracts in vitro, a majority of macrophages were polarized toward the M2 phenotype and subsequently facilitated the chondrogenic differentiation of hBMSCs. Furthermore, after the composite hydrogel was injected into a cartilage defect model, neonatal cartilage-like tissues with a smooth surface and tight integration with original tissues could be found. This study suggests that the synergistic strategy based on an enhanced differentiation ability and a regulated inflammatory response is promising and may lead the way to new anti-inflammatory biomaterials.
Collapse
Affiliation(s)
- Zhuochang Cai
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Ying Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wei Song
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yaohua He
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
- Department of Orthopedics, Jinshan Branch of Shanghai Sixth People's Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201503, China
| | - Haiyan Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Chemical and Environmental Engineering, School of Engineering, RMIT University, Melbourne, Victoria 3000, Australia
| | - Xudong Liu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
35
|
Acevedo L, Iselin L, Berkelaar MHM, Salzmann GM, Wolf F, Feliciano S, Vogel N, Pagenstert G, Martin I, Pelttari K, Barbero A, Arnold MP. Comparison of Human Articular Cartilage Tissue and Chondrocytes Isolated from Peripheral versus Central Regions of Traumatic Lesions. Cartilage 2021; 13:68S-81S. [PMID: 32959685 PMCID: PMC8804865 DOI: 10.1177/1947603520958154] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE Cellular and molecular events occurring in cartilage regions close to injury are poorly investigated, but can possibly compromise the outcome of cell-based cartilage repair. In this study, key functional properties were assessed for cartilage biopsies collected from the central part of traumatic joint lesions (central) and from regions surrounding the defect (peripheral). These properties were then correlated with the quality of the initial cartilage biopsy and the inflammatory state of the joint. DESIGN Cartilage samples were collected from knee joints of 42 patients with traumatic knee injuries and analyzed for cell phenotype (by reverse transcriptas-polymerase chain reaction), histological quality, cellularity, cell viability, proliferation capacity, and post-expansion chondrogenic capacity of chondrocytes (in pellet culture). Synovium was also harvested and analyzed for the expression of inflammatory cytokines. RESULTS Cartilage quality and post-expansion chondrogenic capacity were higher in peripheral versus central samples. Differences between these 2 parameters were more pronounced in joints with high inflammatory features characterized by >100-fold difference in the mRNA levels of IL6 and IL8 in the corresponding synovium. Peripheral chondrocytes isolated from good- versus bad-quality biopsies expressed higher levels of collagen II/I and aggrecan/versican and lower levels of MMP13 and ADAMTS5. They also exhibited reduced proliferation and enhanced cartilage-forming capacity. CONCLUSIONS Chondrocytes at the periphery of traumatic lesions better maintain properties of healthy cartilage compared to those isolated from the center, even when derived from bad-quality tissues harvested from highly inflamed joints. Future studies are necessary to investigate the change of functional properties of peripheral chondrocytes over time.
Collapse
Affiliation(s)
- Lina Acevedo
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | - Lukas Iselin
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland,Department of Orthopedics and
Traumatology, Lucerne Cantonal Hospital, Lucerne, Switzerland
| | - Majoska H. M. Berkelaar
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Francine Wolf
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | - Sandra Feliciano
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | - Nicole Vogel
- Practice LEONARDO, Hirslanden Clinic
Birshof, Münchenstein, Switzerland
| | - Geert Pagenstert
- Department of Surgery, University
Hospital Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | - Karoliina Pelttari
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland,Andrea Barbero, Department of Biomedicine,
University Hospital Basel, University of Basel, Basel 4031, Switzerland.
| | - Markus P. Arnold
- Practice LEONARDO, Hirslanden Clinic
Birshof, Münchenstein, Switzerland
| |
Collapse
|
36
|
Tamaddon M, Blunn G, Xu W, Alemán Domínguez ME, Monzón M, Donaldson J, Skinner J, Arnett TR, Wang L, Liu C. Sheep condyle model evaluation of bone marrow cell concentrate combined with a scaffold for repair of large osteochondral defects. Bone Joint Res 2021; 10:677-689. [PMID: 34665001 PMCID: PMC8559972 DOI: 10.1302/2046-3758.1010.bjr-2020-0504.r1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Aims Minimally manipulated cells, such as autologous bone marrow concentrates (BMC), have been investigated in orthopaedics as both a primary therapeutic and augmentation to existing restoration procedures. However, the efficacy of BMC in combination with tissue engineering is still unclear. In this study, we aimed to determine whether the addition of BMC to an osteochondral scaffold is safe and can improve the repair of large osteochondral defects when compared to the scaffold alone. Methods The ovine femoral condyle model was used. Bone marrow was aspirated, concentrated, and used intraoperatively with a collagen/hydroxyapatite scaffold to fill the osteochondral defects (n = 6). Tissue regeneration was then assessed versus the scaffold-only group (n = 6). Histological staining of cartilage with alcian blue and safranin-O, changes in chondrogenic gene expression, microCT, peripheral quantitative CT (pQCT), and force-plate gait analyses were performed. Lymph nodes and blood were analyzed for safety. Results The results six months postoperatively showed that there were no significant differences in bone regrowth and mineral density between BMC-treated animals and controls. A significant upregulation of messenger RNA (mRNA) for types I and II collagens in the BMC group was observed, but there were no differences in the formation of hyaline-like cartilage between the groups. A trend towards reduced sulphated glycosaminoglycans (sGAG) breakdown was detected in the BMC group but this was not statistically significant. Functional weightbearing was not affected by the inclusion of BMC. Conclusion Our results indicated that the addition of BMC to scaffold is safe and has some potentially beneficial effects on osteochondral-tissue regeneration, but not on the functional endpoint of orthopaedic interest. Cite this article: Bone Joint Res 2021;10(10):677–689.
Collapse
Affiliation(s)
- Maryam Tamaddon
- Institute of Orthopaedic & Musculoskeletal Science, Division of Surgery & Interventional Science, University College London, Royal National Orthopaedic Hospital, London, UK
| | - Gordon Blunn
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Wei Xu
- Beijing Advanced Innovation Center for Materials Genome Engineering, Institute for Advanced Materials and Technology, State Key Laboratory for Advanced Metals and Materials, University of Science and Technology Beijing, Beijing, China
| | | | - Mario Monzón
- Departamento de Ingeniería Mecánica, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - James Donaldson
- Knee and Hip Unit, Royal National Orthopaedic Hospital, London, UK
| | - John Skinner
- Institute of Orthopaedic & Musculoskeletal Science, Division of Surgery & Interventional Science, University College London, Royal National Orthopaedic Hospital, London, UK.,Knee and Hip Unit, Royal National Orthopaedic Hospital, London, UK
| | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Ling Wang
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Chaozong Liu
- Institute of Orthopaedic & Musculoskeletal Science, Division of Surgery & Interventional Science, University College London, Royal National Orthopaedic Hospital, London, UK
| |
Collapse
|
37
|
Gryadunova A, Kasamkattil J, Gay MHP, Dasen B, Pelttari K, Mironov V, Martin I, Schären S, Barbero A, Krupkova O, Mehrkens A. Nose to Spine: spheroids generated by human nasal chondrocytes for scaffold-free nucleus pulposus augmentation. Acta Biomater 2021; 134:240-251. [PMID: 34339870 DOI: 10.1016/j.actbio.2021.07.064] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 12/19/2022]
Abstract
Cell-based strategies for nucleus pulposus (NP) regeneration that adequately support the engraftment and functionality of therapeutic cells are still lacking. This study explores a scaffold-free approach for NP repair, which is based on spheroids derived from human nasal chondrocytes (NC), a resilient cell type with robust cartilage-regenerative capacity. We generated NC spheroids (NCS) in two types of medium (growth or chondrogenic) and analyzed their applicability for NP repair with regard to injectability, biomechanical and biochemical attributes, and integration potential in conditions simulating degenerative disc disease (DDD). NCS engineered in both media were compatible with a typical spinal needle in terms of size (lower than 600µm), shape (roundness greater than 0.8), and injectability (no changes in morphology and catabolic gene expression after passing through the needle). While growth medium ensured stable elastic modulus (E) at 5 kPa, chondrogenic medium time-dependently increased E of NCS, in correlation with gene/protein expression of collagen. Notably, DDD-mimicking conditions did not impair NCS viability nor NCS fusion with NP spheroids simulating degenerated NP in vitro. To assess the feasibility of this approach, NCS were injected into an ex vivo-cultured bovine intervertebral disc (IVD) without damage using a spinal needle. In conclusion, our data indicated that NC cultured as spheroids can be compatible with strategies for minimally invasive NP repair in terms of injectability, tuneability, biomechanical features, and resilience. Future studies will address the capacity of NCS to integrate within degenerated NP under long-term loading conditions. STATEMENT OF SIGNIFICANCE: Current regenerative strategies still do not sufficiently support the engraftment of therapeutic cells in the nucleus pulposus (NP). We present an injectable approach based on spheroids derived from nasal chondrocytes (NC), a resilient cell type with robust cartilage-regenerative capacity. NC spheroids (NCS) generated with their own matrix and demonstrated injectability, tuneability of biomechanical/biochemical attributes, and integration potential in conditions simulating degenerative disc disease. To our knowledge, this is the first study that explored an injectable spheroid-based scaffold-free approach, which showed potential to support the adhesion and viability of therapeutic cells in degenerated NP. The provided information can be of substantial interest to a wide audience, including biomaterial scientists, biomedical engineers, biologists and medical researchers.
Collapse
Affiliation(s)
- Anna Gryadunova
- Spine Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel & University Hospital Basel, Tissue Engineering, ZLF 402, Hebelstrasse 20, 4031 Basel, Switzerland; Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, 119991, Russian Federation
| | - Jesil Kasamkattil
- Spine Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel & University Hospital Basel, Tissue Engineering, ZLF 402, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Max Hans Peter Gay
- Spine Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel & University Hospital Basel, Tissue Engineering, ZLF 402, Hebelstrasse 20, 4031 Basel, Switzerland; Institute of Anatomy, Department of Biomedicine, University of Basel & University Hospital Basel, Pestalozzistrasse 20, 4031, Bael Switzerland
| | - Boris Dasen
- Department of Biomedicine, University of Basel & University Hospital Basel, Tissue Engineering, ZLF 402, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Karoliina Pelttari
- Department of Biomedicine, University of Basel & University Hospital Basel, Tissue Engineering, ZLF 402, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Vladimir Mironov
- Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, 119991, Russian Federation; Laboratory for Biotechnological Research 3D Bioprinting Solutions, Kashirskoe Highway, 68-2, Moscow, 115409 Russian Federation
| | - Ivan Martin
- Department of Biomedicine, University of Basel & University Hospital Basel, Tissue Engineering, ZLF 402, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Stefan Schären
- Spine Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University of Basel & University Hospital Basel, Tissue Engineering, ZLF 402, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Olga Krupkova
- Spine Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel & University Hospital Basel, Tissue Engineering, ZLF 402, Hebelstrasse 20, 4031 Basel, Switzerland; Lepage Research Institute, University of Prešov, 17. Novembra 1, 081 16 Prešov, Slovakia.
| | - Arne Mehrkens
- Spine Surgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel & University Hospital Basel, Tissue Engineering, ZLF 402, Hebelstrasse 20, 4031 Basel, Switzerland
| |
Collapse
|
38
|
Pigeot S, Klein T, Gullotta F, Dupard SJ, Garcia Garcia A, García‐García A, Prithiviraj S, Lorenzo P, Filippi M, Jaquiery C, Kouba L, Asnaghi MA, Raina DB, Dasen B, Isaksson H, Önnerfjord P, Tägil M, Bondanza A, Martin I, Bourgine PE. Manufacturing of Human Tissues as off-the-Shelf Grafts Programmed to Induce Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2103737. [PMID: 34486186 PMCID: PMC11468960 DOI: 10.1002/adma.202103737] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/18/2021] [Indexed: 06/13/2023]
Abstract
Design criteria for tissue-engineered materials in regenerative medicine include robust biological effectiveness, off-the-shelf availability, and scalable manufacturing under standardized conditions. For bone repair, existing strategies rely on primary autologous cells, associated with unpredictable performance, limited availability and complex logistic. Here, a conceptual shift based on the manufacturing of devitalized human hypertrophic cartilage (HyC), as cell-free material inducing bone formation by recapitulating the developmental process of endochondral ossification, is reported. The strategy relies on a customized human mesenchymal line expressing bone morphogenetic protein-2 (BMP-2), critically required for robust chondrogenesis and concomitant extracellular matrix (ECM) enrichment. Following apoptosis-driven devitalization, lyophilization, and storage, the resulting off-the-shelf cartilage tissue exhibits unprecedented osteoinductive properties, unmatched by synthetic delivery of BMP-2 or by living engineered grafts. Scalability and pre-clinical efficacy are demonstrated by bioreactor-based production and subsequent orthotopic assessment. The findings exemplify the broader paradigm of programming human cell lines as biological factory units to engineer customized ECMs, designed to activate specific regenerative processes.
Collapse
Affiliation(s)
- Sébastien Pigeot
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
- Department of Biomedical EngineeringUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Thibaut Klein
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Fabiana Gullotta
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
- Innovative Immunotherapies UnitDivision of Immunology, Transplantation and Infectious DiseasesSan Raffaele Hospital Scientific InstituteVita‐Salute San Raffaele UniversityMilan20132Italy
| | - Steven J. Dupard
- Laboratory for Cell, Tissue, and Organ EngineeringDepartment of Clinical SciencesLund UniversityLund221 84Sweden
- Wallenberg Center for Molecular MedicineLund UniversityLund221 84Sweden
- Stem Cell CenterLund UniversityLund221 84Sweden
| | - Alejandro Garcia Garcia
- Laboratory for Cell, Tissue, and Organ EngineeringDepartment of Clinical SciencesLund UniversityLund221 84Sweden
- Wallenberg Center for Molecular MedicineLund UniversityLund221 84Sweden
- Stem Cell CenterLund UniversityLund221 84Sweden
| | - Andres García‐García
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Sujeethkumar Prithiviraj
- Laboratory for Cell, Tissue, and Organ EngineeringDepartment of Clinical SciencesLund UniversityLund221 84Sweden
- Wallenberg Center for Molecular MedicineLund UniversityLund221 84Sweden
- Stem Cell CenterLund UniversityLund221 84Sweden
| | - Pilar Lorenzo
- Wallenberg Center for Molecular MedicineLund UniversityLund221 84Sweden
| | - Miriam Filippi
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Claude Jaquiery
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Loraine Kouba
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - M. Adelaide Asnaghi
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | | | - Boris Dasen
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Hanna Isaksson
- Department of Clinical SciencesOrthopedicsLund UniversityLund221 84Sweden
- Department of Biomedical EngineeringLund UniversityLund221 84Sweden
| | - Patrik Önnerfjord
- Rheumatology and Molecular Skeletal BiologyDepartment of Clinical SciencesLund UniversityLund221 84Sweden
| | - Magnus Tägil
- Department of Clinical SciencesOrthopedicsLund UniversityLund221 84Sweden
| | - Attilio Bondanza
- Innovative Immunotherapies UnitDivision of Immunology, Transplantation and Infectious DiseasesSan Raffaele Hospital Scientific InstituteVita‐Salute San Raffaele UniversityMilan20132Italy
| | - Ivan Martin
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
- Department of Biomedical EngineeringUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
| | - Paul E. Bourgine
- Department of BiomedicineUniversity Hospital BaselUniversity of BaselBasel4031Switzerland
- Laboratory for Cell, Tissue, and Organ EngineeringDepartment of Clinical SciencesLund UniversityLund221 84Sweden
- Wallenberg Center for Molecular MedicineLund UniversityLund221 84Sweden
- Stem Cell CenterLund UniversityLund221 84Sweden
| |
Collapse
|
39
|
Subedar OD, Chiu LLY, Waldman SD. Cell Cycle Synchronization of Primary Articular Chondrocytes Enhances Chondrogenesis. Cartilage 2021; 12:526-535. [PMID: 30971093 PMCID: PMC8461165 DOI: 10.1177/1947603519841677] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Although tissue engineering is a promising option for articular cartilage repair, it has been challenging to generate functional cartilaginous tissue. While the synthetic response of chondrocytes can be influenced by various means, most approaches treat chondrocytes as a homogeneous population that would respond similarly. However, isolated cells heterogeneously progress through the cell cycle, which can affect macromolecular biosynthesis. As it is possible to synchronize cells within discrete cell cycle phases, the purpose of this study was to investigate the effects of cell cycle synchronization on the chondrogenic potential of primary articular chondrocytes. DESIGN Different methods of cell synchronization (serum starvation, thymidine, nocodazole, aphidicolin, and RO-3306) were tested for their ability to synchronize primary articular chondrocytes during the process of cell isolation. Cells (unsynchronized and synchronized) were then encapsulated in alginate gels, cultured for 4 weeks, and analyzed for their structural and biochemical properties. RESULTS The double-thymidine method yielded the highest level of cell purity, with cells synchronized in S phase. While the cells started to lose synchronization after 24 hours, tissue constructs developed from initially S phase synchronized cells had significantly higher glycosaminoglycan and collagen II amounts than those developed using unsynchronized cells. CONCLUSIONS Initial synchronization led to long-term changes in cartilaginous tissue formation. This effect was postulated to be due to the rapid auto-induction of TGF-βs by actively dividing S phase cells, thereby stimulating chondrogenesis. Cell synchronization methods may also be applied in conjunction with redifferentiation methods to improve the chondrogenic potential of dedifferentiated or diseased chondrocytes.
Collapse
Affiliation(s)
- Omar D. Subedar
- Department of Chemical Engineering, Ryerson University, Toronto, Ontario, Canada,Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Loraine L. Y. Chiu
- Department of Chemical Engineering, Ryerson University, Toronto, Ontario, Canada,Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Stephen D. Waldman
- Department of Chemical Engineering, Ryerson University, Toronto, Ontario, Canada,Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada,Stephen D. Waldman, Department of Chemical Engineering, Faculty of Engineering & Architectural Science, Ryerson University, Kerr Hall South, KHS 241N, Toronto, Ontario, Canada M5B 2K3.
| |
Collapse
|
40
|
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
|
41
|
Desando G, Bartolotti I, Cattini L, Tschon M, Martini L, Fini M, Schiavinato A, Soranzo C, Grigolo B. Prospects on the Potential In Vitro Regenerative Features of Mechanically Treated-Adipose Tissue for Osteoarthritis Care. Stem Cell Rev Rep 2021; 17:1362-1373. [PMID: 33469783 PMCID: PMC8316247 DOI: 10.1007/s12015-020-10099-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 11/06/2022]
Abstract
Gathering a better grasp on the adipose stromal vascular fraction (SVF) is demanding among clinicians for osteoarthritis (OA) care because of its promising but multifaceted clinical outcomes. The aim of this preclinical in vitro study was to test whether the mechanical approach with Hy-Tissue SVF system, a class IIa CE marked device of adipose tissue micro-fragmentation, influences the biological features and functions of SVF. We compared mechanical generated-SVF (mSVF) with the enzymatic generated-SVF (eSVF) by testing cell survival, phenotype, differentiation, and paracrine properties using ELISA assays. Both adipose SVF showed 80% viable cells and enrichment for CD-44 marker. The mSVF product preserved the functions of cell populations within the adipose tissue; however, it displayed lowered nucleated cell recovery and CFU-F than eSVF. As for multipotency, mSVF and eSVF showed similar differentiation commitment for osteochondral lineages. Both adipose SVF exhibited an increased release of VEGF, HGF, IGF-1 and PDGF-bb, involved in pathways mediating osteochondral repair and cell migration. Both mSVF and eSVF also displayed high release for the anti-inflammatory cytokine IL-10. After in vitro culture, supernatants from both mSVF and eSVF groups showed a low release of cytokines except for IL-10, thereby giving evidence of functional changes after culture expansion. In this study, mSVF showed active cell populations in the adipose tissue comparable to eSVF with excellent survival, differentiation and paracrine properties under a new mechanical adipose tissue micro-fragmentation system; thereby suggesting its potential use as a minimally invasive technique for OA treatment.
Collapse
Affiliation(s)
- G Desando
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Bologna, 40136, Italy.
| | - I Bartolotti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Bologna, 40136, Italy
| | - L Cattini
- Laboratorio di ImmunoReumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Bologna, 40136, Italia
| | - M Tschon
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Bologna, 40136, Italia
| | - L Martini
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Bologna, 40136, Italia
| | - M Fini
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Bologna, 40136, Italia
| | - A Schiavinato
- Fidia Farmaceutici S.p.A, Abano Terme, Padova, 35031, Italy
| | - C Soranzo
- Fidia Farmaceutici S.p.A, Abano Terme, Padova, 35031, Italy
| | - B Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Bologna, 40136, Italy
| |
Collapse
|
42
|
Lan X, Liang Y, Erkut EJN, Kunze M, Mulet-Sierra A, Gong T, Osswald M, Ansari K, Seikaly H, Boluk Y, Adesida AB. Bioprinting of human nasoseptal chondrocytes-laden collagen hydrogel for cartilage tissue engineering. FASEB J 2021; 35:e21191. [PMID: 33595884 DOI: 10.1096/fj.202002081r] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/18/2020] [Accepted: 10/29/2020] [Indexed: 12/23/2022]
Abstract
Skin cancer patients often have tumorigenic lesions on their noses. Surgical resection of the lesions often results in nasal cartilage removal. Cartilage grafts taken from other anatomical sites are used for the surgical reconstruction of the nasal cartilage, but donor-site morbidity is a common problem. Autologous tissue-engineered nasal cartilage grafts can mitigate the problem, but commercially available scaffolds define the shape and sizes of the engineered grafts during tissue fabrication. Moreover, the engineered grafts suffer from the inhomogeneous distribution of the functional matrix of cartilage. Advances in 3D bioprinting technology offer the opportunity to engineer cartilages with customizable dimensions and anatomically shaped configurations without the inhomogeneous distribution of cartilage matrix. Here, we report the fidelity of Freeform Reversible Embedding of Suspended Hydrogel (FRESH) bioprinting as a strategy to generate customizable and homogenously distributed functional cartilage matrix engineered nasal cartilage. Using FRESH and in vitro chondrogenesis, we have fabricated tissue-engineered nasal cartilage from combining bovine type I collagen hydrogel and human nasoseptal chondrocytes. The engineered nasal cartilage constructs displayed molecular, biochemical and histological characteristics akin to native human nasal cartilage.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Yan Liang
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada
| | - Esra J N Erkut
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada
| | - Tianxing Gong
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada
| | - Martin Osswald
- Institute for Reconstructive Sciences in Medicine (iRSM), Misericordia Community Hospital, Edmonton, AB, Canada.,Department of Surgery, Division of Otolaryngology, University of Alberta, Edmonton, AB, Canada
| | - Khalid Ansari
- Department of Surgery, Division of Otolaryngology, University of Alberta, Edmonton, AB, Canada
| | - Hadi Seikaly
- Department of Surgery, Division of Otolaryngology, University of Alberta, Edmonton, AB, Canada
| | - Yaman Boluk
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Adetola B Adesida
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, University of Alberta, Edmonton, AB, Canada.,Department of Surgery, Division of Otolaryngology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
43
|
Basoli V, Della Bella E, Kubosch EJ, Alini M, Stoddart MJ. Effect of expansion media and fibronectin coating on growth and chondrogenic differentiation of human bone marrow-derived mesenchymal stromal cells. Sci Rep 2021; 11:13089. [PMID: 34158528 PMCID: PMC8219706 DOI: 10.1038/s41598-021-92270-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/08/2021] [Indexed: 01/01/2023] Open
Abstract
In the field of regenerative medicine, considerable advances have been made from the technological and biological point of view. However, there are still large gaps to be filled regarding translation and application of mesenchymal stromal cell (MSC)-based therapies into clinical practice. Indeed, variables such as cell type, unpredictable donor variation, and expansion/differentiation methods lead to inconsistencies. Most protocols use bovine serum (FBS) derivatives during MSC expansion. However, the xenogeneic risks associated with FBS limits the use of MSC-based products in clinical practice. Herein we compare a chemically defined, xenogeneic-free commercial growth medium with a conventional medium containing 10% FBS and 5 ng/ml FGF2. Furthermore, the effect of a fibronectin-coated growth surface was investigated. The effect of the different culture conditions on chondrogenic commitment was assessed by analyzing matrix deposition and gene expression of common chondrogenic markers. Chondrogenic differentiation potential was similar between the FBS-containing αMEM and the chemically defined medium with fibronectin coating. On the contrary, the use of fibronectin coating with FBS-containing medium appeared to reduce the differentiation potential of MSCs. Moreover, cells that were poorly responsive to in vitro chondrogenic stimuli were shown to improve their differentiation potential after expansion in a TGF-β1 containing medium. In conclusion, the use of a xenogeneic-free medium provides a suitable alternative for human bone marrow MSC expansion, due the capability to maintain cell characteristic and potency. To further improve chondrogenic potential of BMSCs, priming the cells with TGF-β1 during expansion is a promising strategy.
Collapse
Affiliation(s)
- Valentina Basoli
- Regenerative Orthopaedics, AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, Switzerland
| | - Elena Della Bella
- Regenerative Orthopaedics, AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, Switzerland
| | - Eva Johanna Kubosch
- Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Albert-Ludwigs-University of Freiburg, 79106, Freiburg, Germany
| | - Mauro Alini
- Regenerative Orthopaedics, AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, Switzerland
| | - Martin J Stoddart
- Regenerative Orthopaedics, AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, Switzerland. .,Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Albert-Ludwigs-University of Freiburg, 79106, Freiburg, Germany.
| |
Collapse
|
44
|
Ex vivo osteochondral test system with control over cartilage defect depth – A pilot study to investigate the effect of oxygen tension and chondrocyte based treatments in chondral and full thickness defects in an organ model. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100173. [DOI: 10.1016/j.ocarto.2021.100173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 11/18/2022] Open
|
45
|
Wang K, Li J, Wang Y, Wang Y, Qin Y, Yang F, Zhang M, Zhu H, Li Z. Orchestrated cellular, biochemical, and biomechanical optimizations endow platelet-rich plasma-based engineered cartilage with structural and biomechanical recovery. Bioact Mater 2021; 6:3824-3838. [PMID: 33937588 PMCID: PMC8065202 DOI: 10.1016/j.bioactmat.2021.03.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/17/2022] Open
Abstract
Recently, biomaterials for cartilage regeneration has been intensively investigated. However, the development of scaffolds that capture regenerated cartilage with biomechanical and structural recovery has rarely been reported. To address this challenge, platelet-rich plasma (PRP)-based cartilage constructs with a well-orchestrated symphony of cellular, biochemical and biomechanical elements were prepared by simultaneously employing chondrogenic progenitor cells (CPCs) as a cell source, optimizing platelet concentration, and adding an enzyme-ion activator. It was shown that this triple-optimized PRP + CPC construct possessed increased biomechanical properties and suitable biochemical signals. The following in vitro study demonstrated that the triple-optimized PRP + CPC constructs generated cartilage-like tissue with higher expression levels of chondrogenic-specific markers, more deposition of cartilage-specific extracellular matrix (ECM), and greater biomechanical values than those of the other constructs. Twelve weeks after the construct was implanted in a cartilage defect in vivo, histological analysis, qPCR, and biomechanical tests collectively showed that the triple-optimized constructs yielded a more chondrocyte-like cell phenotype with a higher synthesis of Col-II and aggrecan. More importantly, the triple-optimized constructs facilitated cartilage regeneration with better biomechanical recovery than that of the other constructs. These results demonstrate the efficacy of the triple-optimization strategy and highlight the simplicity and potency of this PRP + CPC construct for cartilage regeneration. Cartilage tissue engineering has been intensively investigated. We designed a PRP-based construct with favorable cell source, reinforced scaffold and appropriate biofactors. This designed construct can facilitate cartilage regeneration with biomechanical and structural recovery simultaneously. The favorable performance of the proposed scaffolds highlights the triple-optimization strategy to improve cartilage engineering.
Collapse
Affiliation(s)
- Ketao Wang
- Department of Orthopedics, Chinese PLA General Hospital, Haidian, Beijing, 100853, China.,Department of Foot and Ankle, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.,Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ji Li
- Department of Orthopedics, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - Yuxing Wang
- Department of Orthopedics, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - Yaqiang Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yuanyuan Qin
- Department of Blood Transfusion, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - Fei Yang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Mingzhu Zhang
- Department of Foot and Ankle, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Heng Zhu
- Beijing Institute of Radiation Medicine/Beijing Institute of Basic Medical Sciences, Haidian, Beijing, 100850, China
| | - Zhongli Li
- Department of Orthopedics, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| |
Collapse
|
46
|
Power L, Acevedo L, Yamashita R, Rubin D, Martin I, Barbero A. Deep learning enables the automation of grading histological tissue engineered cartilage images for quality control standardization. Osteoarthritis Cartilage 2021; 29:433-443. [PMID: 33422705 DOI: 10.1016/j.joca.2020.12.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To automate the grading of histological images of engineered cartilage tissues using deep learning. METHODS Cartilaginous tissues were engineered from various cell sources. Safranin O and fast green stained histological images of the tissues were graded for chondrogenic quality according to the Modified Bern Score, which ranks images on a scale from zero to six according to the intensity of staining and cell morphology. The whole images were tiled, and the tiles were graded by two experts and grouped into four categories with the following grades: 0, 1-2, 3-4, and 5-6. Deep learning was used to train models to classify images into these histological score groups. Finally, the tile grades per donor were averaged. The root mean square errors (RMSEs) were calculated between each user and the model. RESULTS Transfer learning using a pretrained DenseNet model was selected. The RMSEs of the model predictions and 95% confidence intervals were 0.49 (0.37, 0.61) and 0.78 (0.57, 0.99) for each user, which was in the same range as the inter-user RMSE of 0.71 (0.51, 0.93). CONCLUSION Using supervised deep learning, we could automate the scoring of histological images of engineered cartilage and achieve results with errors comparable to inter-user error. Thus, the model could enable the automation and standardization of assessments currently used for experimental studies as well as release criteria that ensure the quality of manufactured clinical grafts and compliance with regulatory requirements.
Collapse
Affiliation(s)
- L Power
- Department of Biomedical Engineering, University of Basel, Switzerland; Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland.
| | - L Acevedo
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland.
| | - R Yamashita
- Department of Biomedical Data Science, Stanford University School of Medicine, USA.
| | - D Rubin
- Department of Biomedical Data Science, Stanford University School of Medicine, USA.
| | - I Martin
- Department of Biomedical Engineering, University of Basel, Switzerland; Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland.
| | - A Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland.
| |
Collapse
|
47
|
Bianchi VJ, Parsons M, Backstein D, Kandel RA. Endoglin Level Is Critical for Cartilage Tissue Formation In Vitro by Passaged Human Chondrocytes. Tissue Eng Part A 2021; 27:1140-1150. [PMID: 33323019 DOI: 10.1089/ten.tea.2020.0120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Transforming growth factor beta (TGFβ) signaling is required for in vitro chondrogenesis. In animal models of osteoarthritis (OA), TGFβ receptor alterations are detected in chondrocytes in severe OA cartilage. It is not known whether such changes are dependent on the grade of human OA and if they affect chondrogenesis. Thus, the purpose of this study was to determine if human OA chondrocytes obtained from low-grade or high-grade disease could form cartilage tissue and to assess the role of the co-receptors, endoglin (ENG) and TGFβ receptor 3 (TGFBRIII), in the regulation of this tissue generation in vitro. We hypothesized that the grade of OA disease would not affect the ability of cells to form cartilage tissue and that the TGFβ co-receptor, ENG, would be critical to regulating tissue formation. Chondrocytes isolated from low-grade OA or high-grade OA human articular cartilage (AC) were analyzed directly (P0) or passaged in monolayer to P2. Expression of the primary TGFβ receptor ALK5, and the co-receptors ENG and TGFβRIII, was assessed by image flow cytometry. To assess the ability to form cartilaginous tissue, cells were placed in three-dimensional culture at high density and cultured in chondrogenic media containing TGFβ3. ENG knockdown was used to determine its role in regulating tissue formation. Overall, grade-specific differences in expression of ALK5, ENG, and TGFβRIII in primary or passaged chondrocytes were not detected; however, ENG expression increased significantly after passaging. Despite the presence of ALK5, P0 cells did not form cartilaginous tissue. In contrast, P2 cells derived from low-grade and high-grade OA AC formed hyaline-like cartilaginous tissues of similar quality. Knockdown of ENG in P2 cells inhibited cartilaginous tissue formation compared to controls indicating that the level of ENG protein expression is critical for in vitro chondrogenesis by passaged articular chondrocytes. This study demonstrates that it is not the grade of OA, but the levels of ENG in the presence of ALK5 that influences the ability of human passaged articular chondrocytes to form cartilaginous tissue in vitro in 3D culture. This has implications for cartilage repair therapies. Impact statement These findings are important clinically, given the limited availability of osteoarthritis (OA) cartilage tissue. Being able to use cells from all grades of OA will increase our ability to obtain sufficient cells for cartilage repair. In addition, it is possible that endoglin (ENG) levels, in the presence of ALK5 expression, may be suitable to use as biomarkers to identify cells able to produce cartilage.
Collapse
Affiliation(s)
- Vanessa J Bianchi
- Lunenfeld-Tanenbaum Research Institute, Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | | | - David Backstein
- Division of Orthopaedic Surgery, Mount Sinai Hospital, Toronto, Canada
| | - Rita A Kandel
- Lunenfeld-Tanenbaum Research Institute, Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
48
|
Hagen A, Lehmann H, Aurich S, Bauer N, Melzer M, Moellerberndt J, Patané V, Schnabel CL, Burk J. Scalable Production of Equine Platelet Lysate for Multipotent Mesenchymal Stromal Cell Culture. Front Bioeng Biotechnol 2021; 8:613621. [PMID: 33553119 PMCID: PMC7859354 DOI: 10.3389/fbioe.2020.613621] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/16/2020] [Indexed: 12/22/2022] Open
Abstract
Translation of multipotent mesenchymal stromal cell (MSC)-based therapies is advancing in human and veterinary medicine. One critical issue is the in vitro culture of MSC before clinical use. Using fetal bovine serum (FBS) as supplement to the basal medium is still the gold standard for cultivation of many cell types including equine MSC. Alternatives are being explored, with substantial success using platelet lysate-supplemented media for human MSC. However, progress lags behind in the veterinary field. The aim of this study was to establish a scalable protocol for equine platelet lysate (ePL) production and to test the ePL in equine MSC culture. Whole blood was harvested into blood collection bags from 20 healthy horses. After checking sample materials for pathogen contamination, samples from 19 animals were included. Platelet concentrates were prepared using a buffy coat method. Platelets, platelet-derived growth factor BB, and transforming growth factor β1 concentrations were increased in the concentrates compared with whole blood or serum (p < 0.05), while white blood cells were reduced (p < 0.05). The concentrates were lysed using freeze/thaw cycles, which eliminated the cells while growth factor concentrations were maintained. Donor age negatively correlated with platelet and growth factor concentrations after processing (p < 0.05). Finally, all lysates were pooled and the ePL was evaluated as culture medium supplement in comparison with FBS, using adipose-derived MSC from four unrelated donor horses. MSC proliferated well in 10% FBS as well as in 10% ePL. However, using 5 or 2.5% ePL entailed highly inconsistent proliferation or loss of proliferation, with significant differences in generation times and confluencies (p < 0.05). MSC expressed the surface antigens CD90, CD44, and CD29, but CD73 and CD105 detection was low in all culture media. Adipogenic and osteogenic differentiation led to similar results in MSC from different culture media. The buffy coat method is useful to produce equine platelet concentrate with increased platelet and reduced white blood cell content in large scales. The ePL obtained supports MSC expansion similar as FBS when used at the same concentration (10%). Further investigations into equine MSC functionality in culture with ePL should follow.
Collapse
Affiliation(s)
- A Hagen
- Equine Clinic (Surgery, Orthopedics), Justus-Liebig-University Giessen, Giessen, Germany
| | - H Lehmann
- Department of Veterinary Clinical Sciences, Small Animal Clinic, Justus-Liebig-University Giessen, Giessen, Germany
| | - S Aurich
- Institute of Hygiene and Infectious Diseases of Animals, Justus-Liebig-University Giessen, Giessen, Germany
| | - N Bauer
- Department of Veterinary Clinical Sciences, Clinical Pathology and Clinical Pathophysiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - M Melzer
- Equine Clinic (Surgery, Orthopedics), Justus-Liebig-University Giessen, Giessen, Germany
| | - J Moellerberndt
- Equine Clinic (Surgery, Orthopedics), Justus-Liebig-University Giessen, Giessen, Germany
| | - V Patané
- Department of Veterinary Clinical Sciences, Clinical Pathology and Clinical Pathophysiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - C L Schnabel
- Faculty of Veterinary Medicine, Institute of Immunology, Leipzig University, Leipzig, Germany
| | - J Burk
- Equine Clinic (Surgery, Orthopedics), Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
49
|
Tsuyuguchi Y, Nakasa T, Ishikawa M, Miyaki S, Matsushita R, Kanemitsu M, Adachi N. The Benefit of Minced Cartilage Over Isolated Chondrocytes in Atelocollagen Gel on Chondrocyte Proliferation and Migration. Cartilage 2021; 12:93-101. [PMID: 30311776 PMCID: PMC7755964 DOI: 10.1177/1947603518805205] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Autologous chondrocyte implantation is a necessary procedure for the repair of articular cartilage defects; however, isolated chondrocyte implantation requires a 2-step procedure (for harvesting and implantation) and is limited by cytotoxicity due to enzymatic digestion. Therefore, in this in vitro study, we evaluated the possible benefit of using minced cartilage embedded in a 3-dimensional culture scaffold and fixed with fibrin glue, in comparison with isolated chondrocytes in atelocollagen, to induce cell migration, proliferation, and matrix production, using cartilage from patients with knee joint osteoarthritis. DESIGN Cartilage fragments were obtained from 7 female patients with knee osteoarthritis (OA) and embedded in atelocollagen gels. As a control, chondrocytes were isolated and embedded in gels in the same manner. These composites were cultured for 3 weeks, and cell proliferation and matrix production were evaluated using histology and immunochemistry. RESULTS Histologically, minced cartilage showed cell migration from the cartilage fragments into the gel, with the Bern score and cell count in the minced cartilage group being significantly higher than those in the control group. Immunohistochemistry revealed that the number of Ki67-positive cells, the expression of LECT-1 and TGF-β, and the glycosaminoglycan content were significantly higher in the minced cartilage than in the control group. Minced cartilage exhibited superior cell migration, proliferation, and glycosaminoglycan content than isolated chondrocytes. CONCLUSION Our findings support that minced cartilage has a favorable potential for cell proliferation and matrix production compared with the isolated chondrocytes after enzymatic treatment.
Collapse
Affiliation(s)
- Yusuke Tsuyuguchi
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan,Yusuke Tsuyuguchi, Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Tomoyuki Nakasa
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masakazu Ishikawa
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shigeru Miyaki
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan,Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Ryosuke Matsushita
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Munekazu Kanemitsu
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
50
|
Kim JK, Bae HC, Ro DH, Lee S, Lee MC, Han HS. Enhancement of Cartilage Regeneration of Synovial Stem Cells/Hydrogel by Using Transglutaminase-4. Tissue Eng Part A 2020; 27:761-770. [PMID: 33107390 DOI: 10.1089/ten.tea.2020.0271] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Although mesenchymal stem cells (MSCs) transplantation is reportedly a promising strategy for repairing damaged articular cartilage, MSCs-based cartilage tissue engineering has numerous limitations, including poor implanted cell adhesion, phenotypic alteration of cells, regulation of mechanical properties, and engraftment rates after implantation. This study aimed to investigate the efficacy of transplantation of synovium-derived mesenchymal stem cells (SDSCs) encapsulated in a hyaluronic acid/collagen/fibrinogen (HA/COL/FG) composite gel by supplementing recombinant human transglutaminase 4 (rhTG-4) in treating osteochondral defects. RhTG-4 treatment induced the expression of integrin β1 and dynamic actin fiber, enhancing SDSCs adhesion to fibronectin. Supplementation of rhTG-4 significantly induced the proliferation of SDSCs encapsulated in the HA/COL/FG composite gel and increased the hardness of the extracellular matrix. Furthermore, supplementation of rhTG-4 significantly upregulated aggrecan and type II collagen mRNA. Pretreatment with integrin β1 siRNA markedly suppressed TG4-induced actin remodeling, activation mitogen-activated protein kinase (MAPK), and eventually the chondrogenesis-related genes. Moreover, transplantation of SDSCs encapsulated in HA/COL/FG/rhTG-4 composite gel in vivo yielded reconstructed tissue resembling native hyaline cartilage. These data suggest that rhTG-4 enhances cartilage regeneration of the SDSCs encapsulated in hydrogel in rabbits. Impact statement In this study, we investigated the effects of recombinant human transglutaminase 4 on the ability of synovium-derived mesenchymal stem cells encapsulated in a hyaluronic acid/collagen/fibrinogen composite gel to repair osteochondral defects. We believe that our study makes a significant contribution to the literature because it explores a method of improving an existing modality to mediate tissue repair.
Collapse
Affiliation(s)
- Jong-Keun Kim
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Cheol Bae
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Du Hyun Ro
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sahnghoon Lee
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Myung Chul Lee
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyuk-Soo Han
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|