1
|
Li S, Sun H, Lu Q, Qiao J, Luo Y, Chu Z, Liu D, Zhou L, Liu P. Bionic ECM scaffolds for directional articular hyaline cartilage regeneration and long-term homeostasis maintenance. BIOMATERIALS ADVANCES 2025; 173:214292. [PMID: 40157112 DOI: 10.1016/j.bioadv.2025.214292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/02/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Cartilage defects play a key role in osteoarthritis, causing functional impairment as the disease progresses. Microfracture surgery is commonly used to treat articular cartilage defects, providing early pain relief and functional improvement. However, the blood clot formed during the procedure differs from the natural cartilage microenvironment, hindering hyaline cartilage formation and promoting fibrocartilage, which limits long-term outcomes. This study proposes combining a bionic flexible extracellular matrix (ECM) scaffold with microfracture surgery as a treatment for cartilage defects. By filling the microfracture site with the scaffold and thermosensitive agarose gel, we can anchor BMSCs leaking from the bone marrow while creating a 3D microenvironment that regulates stem cell differentiation. Our results show that the scaffold's mechanical strength is comparable to that of hyaline cartilage, offering excellent biomimetic properties and biocompatibility. In vitro, BMSCs migrating into the scaffold exhibited a survival rate of nearly 90 % by day 2, significantly higher than the 25 % survival rate in the control agarose gel group, with cells observed anchoring around the scaffold. In vivo, stem cells anchored to the scaffold successfully differentiated into articular hyaline cartilage, driven by the combined effects of the scaffold's physical structure and its contained cytokines. The generated hyaline cartilage maintains homeostasis over time, reducing the risk of fibrocartilage formation. This strategy addresses a key limitation of microfracture surgery, where regenerated cartilage is often fibrocartilage, offering a promising new approach for cartilage repair.
Collapse
Affiliation(s)
- Shihao Li
- Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Houyi Sun
- Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Qunshan Lu
- Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Junran Qiao
- Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Yange Luo
- Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Ziyue Chu
- Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Dehua Liu
- Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Libo Zhou
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China.
| | - Peilai Liu
- Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
2
|
Bae BS, Jung JW, Jo GO, Kim SA, Go EJ, Cho ML, Shetty AA, Kim SJ. Treatment of osteoarthritic knee with high tibial osteotomy and allogeneic human umbilical cord blood-derived mesenchymal stem cells combined with hyaluronate hydrogel composite. Stem Cell Res Ther 2025; 16:211. [PMID: 40296133 PMCID: PMC12038988 DOI: 10.1186/s13287-025-04356-9] [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: 12/19/2024] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Delaying total knee arthroplasty is crucial for middle-aged patients with severe osteoarthritis. The long-term outcomes of high tibial osteotomy (HTO) remain uncertain. Recently, mesenchymal stem cells (MSCs) have shown promising potential in enhancing cartilage regeneration. Therefore, this study aimed to assess cartilage regeneration following the implantation of allogeneic human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) with HTO. METHODS In this case series, ten patients underwent hUCB-MSC implantation with HTO. The median age was 58.50 (range: 57.00-60.00) years, and the mean body mass index was 27.81 (range: 24.42-32.24) kg/m2. Clinical outcomes, including the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), visual analog scale (VAS), Physical Component Score (PCS) and Mental Component Score (MCS) from the 36-Item Short-Form Health Survey (SF-36), were evaluated 6 months, 1 year, and 2 years postoperatively. Cartilage status of the medial femoral condyle (MFC) was assessed during hardware removal surgery, at least 2 years after the initial procedure, and compared with preoperative MFC cartilage status regarding lesion size and International Cartilage Repair Society (ICRS) grade. Radiological assessments included the Kellgren-Lawrence (KL) grading system for medial compartment osteoarthritis and hip-knee-ankle (HKA) angle. RESULTS Significant improvements were observed in WOMAC scores (preoperative: 57.00 (range: 44.75-63.00), postoperative: 27.50 (range: 22.25-28.75)), VAS scores (preoperative: 66.25 (range: 48.00-74.25), postoperative: 26.25 (range: 14.50-31.13)), SF-36 PCS (preoperative: 27.97 (range: 26.64-31.25), postoperative: 55.31 (range: 51.64-62.50)), and SF-36 MCS (preoperative: 41.04 (range: 29.95-50.96), postoperative: 63.18 (range: 53.83-65.16)) 2 years postoperatively (p = 0.002, 0.002, 0.002, and 0.020, respectively). The MFC chondral lesion demonstrated significant improvement in both lesion size (preoperative: 7.00 cm² (range: 4.38-10.50 cm²), postoperative: 0.16 cm² (range: 0.00-1.75 cm²), p = 0.002) and ICRS grade (preoperative: 4 (range: 4-4), postoperative: 1 (range: 1-2.25), p = 0.002). Additionally, the KL grade significantly decreased from 3 (range: 3-3) preoperatively to 2 (range: 2-2) postoperatively, while the HKA angle was corrected from 7.50° (range: 7.00-10.25°) preoperatively to -1.00° (range: -3.5-0.00°) postoperatively. CONCLUSIONS hUCB-MSC implantation with HTO is an effective treatment for medial compartment osteoarthritis and varus deformities, resulting in significant improvements in cartilage regeneration and overall clinical outcomes. TRIAL REGISTRATION NCT04234412.
Collapse
Affiliation(s)
- Bo Seung Bae
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jae Woong Jung
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Gyeong Ok Jo
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seon Ae Kim
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Jeong Go
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Asode Ananthram Shetty
- Institute of Medical Sciences, Faculty of Health and Social Care, Canterbury Christ Church University, Canterbury, UK
| | - Seok Jung Kim
- Department of Orthopaedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Department of Orthopedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 271, Cheonbo-ro, Uijeongbu-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
3
|
Mukherjee A, Rajput J, Poundarik A, Das B. Development of a bovine gelatin-kappa carrageenan-based dual network biomimetic hydrogel for chondrogenic differentiation of mesenchymal stem cells. Int J Biol Macromol 2025; 309:142553. [PMID: 40203945 DOI: 10.1016/j.ijbiomac.2025.142553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/25/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Abstract
Direct stem cell delivery for cartilage tissue engineering faces significant drawbacks, including loss of cells via circulation and poor viability in a hostile microenvironment. Hence, scaffold-based approaches for stem cell delivery are gaining significant momentum. In this study, composite hydrogel films composed of gelatin and κ-carrageenan dually crosslinked with glutaraldehyde and potassium chloride have been developed through the solvent casting method. The protein-polysaccharide composite mimics the natural extracellular matrix of native cartilage and the synergistic effects of covalent and ionic crosslinking provide mechanical strength, stability, and satisfactory biological performance. The physicochemical properties of the composite were analyzed using SEM-EDS, AFM, FTIR, XPS, and XRD. Rheological analysis revealed self-healing properties of the film and mechanical analysis demonstrated the ultimate tensile strength to be 13.49 ± 2.89 MPa, which mechanically mimics the native cartilage. The composite film remained stable for approximately 4 weeks in PBS, validating its stability. Biological assessments of the film after 7 days of culture demonstrated its long-term cytocompatibility, showing cell viability of 97.56 ± 0.21 %, cell adhesion was observed using FESEM micrographs, and cell proliferation through Live/Dead assay. The dually crosslinked biomimetic composite films aided in chondrogenic differentiation, as confirmed using collagen II staining and TGF-β expression studies, and hence demonstrate promising potential for cartilage tissue regeneration.
Collapse
Affiliation(s)
- Anwesha Mukherjee
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Jayhind Rajput
- Department of Metallurgical and Materials Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Atharva Poundarik
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India; Department of Metallurgical and Materials Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Bodhisatwa Das
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India.
| |
Collapse
|
4
|
Unguryte A, Uzieliene I, Bagdonas E, Zentelyte A, Porvaneckas N, Bernotiene E. Potential of MSCA1 for isolating osteogenic cells in a chondrocyte population. Sci Rep 2025; 15:7813. [PMID: 40050298 PMCID: PMC11885582 DOI: 10.1038/s41598-025-91303-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
The aim of the present study was to elucidate the utility of Mesenchymal Stem Cell Antigen-1 (MSCA1) (tissue nonspecific alkaline phosphatase (TNAP)) as a potential marker for purification of human chondrocyte fraction with less heterogenous phenotype from those with osteogenic properties. Chondrocytes were isolated from human osteoarthritis cartilage and sorted according to MSCA1 expression by MACS (Magnetic-activated cell sorting) and FACS (Fluorescence-activated cell sorting) techniques (MSCA1high and MSCA1low), analyzed for gene expression, osteogenic and adipogenic differentiation capacities, and were compared between the sorted populations. Gene expression analyses revealed upregulation in osteogenic genes (ALPL and RUNX2) and significantly lower expression of chondrocyte-specific genes (COL2A1, SOX9 and MIA) in sorted MSCA1high, as compared to MSCA1low. Expression of Aldehyde dehydrogenase isoform gene ALDH1A2 was enriched in MSCA1low chondrocytes. Stronger osteogenic differentiation was observed in MSCA1high, as compared to the unsorted cells, and particularly, MSCA1low chondrocytes. Expression of MSCA1 in human chondrocytes is biased in their commitment to the osteogenic lineage. We demonstrate that MSCA1 can be used as a marker for separation of cells, prone to differentiate towards osteogenic lineage from those, retaining chondrogenic phenotype. This may allow enrichment of a chondrogenic population and separation from undesirable osteoprogenitors.
Collapse
Affiliation(s)
- Ausra Unguryte
- Department of Regenerative Medicine, Innovative medicine centre, Santariskiu g. 5, Vilnius, LT-08406, Lithuania
| | - Ilona Uzieliene
- Department of Regenerative Medicine, Innovative medicine centre, Santariskiu g. 5, Vilnius, LT-08406, Lithuania
| | - Edvardas Bagdonas
- Department of Regenerative Medicine, Innovative medicine centre, Santariskiu g. 5, Vilnius, LT-08406, Lithuania
| | - Aiste Zentelyte
- Department of Regenerative Medicine, Innovative medicine centre, Santariskiu g. 5, Vilnius, LT-08406, Lithuania
| | - Narūnas Porvaneckas
- Clinic of Rheumatology, Orthopaedics Traumatology and Reconstructive Surgery, Faculty of Medicine, Vilnius University, Vilnius, LT-03101, Lithuania
| | - Eiva Bernotiene
- Department of Regenerative Medicine, Innovative medicine centre, Santariskiu g. 5, Vilnius, LT-08406, Lithuania.
- Faculty of Fundamental Sciences, VilniusTech, Sauletekio al. 11, LT-10223, Vilnius, Lithuania.
| |
Collapse
|
5
|
McMillan A, Hoffman MR, Xu Y, Wu Z, Thayer E, Peel A, Guymon A, Kanotra S, Salem AK. 3D bioprinted ferret mesenchymal stem cell-laden cartilage grafts for laryngotracheal reconstruction in a ferret surgical model. Biomater Sci 2025; 13:1304-1322. [PMID: 39886992 PMCID: PMC11784027 DOI: 10.1039/d4bm01251h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Chondrogenic differentiation of mesenchymal stem cells (MSCs) within a three-dimensional (3D) environment can be guided to form cartilage-like tissue in vitro to generate cartilage grafts for implantation. 3D bioprinted, MSC-populated cartilage grafts have the potential to replace autologous cartilage in reconstructive airway surgery. Here, bone marrow-derived ferret MSCs (fMSCs) capable of directed musculoskeletal differentiation were generated for the first time. A multi-material, 3D bioprinted fMSC-laden scaffold was then engineered that was capable of in vitro cartilage regeneration, as evidenced by glycosaminoglycan (GAG) production and collagen II immunohistochemical staining. In vivo implantation of these 3D bioprinted scaffolds in a ferret model of laryngotracheal reconstruction (LTR) demonstrated healing of the defect site, epithelial mucosalization of the inner lumen, and expansion of the airway volume. While the implanted scaffold allowed for reconstruction of the created airway defect, minimal chondrocytes were identified at the implant site. Nevertheless, we have established the ferret as a biomedical research model for airway reconstruction and, although further evaluation is warranted, the generation of fMSCs provides an opportunity for realizing the potential for 3D bioprinted regenerative stem cell platforms in the ferret.
Collapse
Affiliation(s)
- Alexandra McMillan
- Department of Otolaryngology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA.
| | - Matthew R Hoffman
- Department of Otolaryngology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Yan Xu
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA.
| | - Zongliang Wu
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA.
| | - Emma Thayer
- Department of Otolaryngology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Adreann Peel
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, USA
| | - Allan Guymon
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, USA
| | - Sohit Kanotra
- Department of Head and Neck Surgery, UCLA, Los Angeles, California, USA
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
6
|
Wang T, Maldonado CC, Huang BL, Budbazar E, Martin A, Layne MD, Murphy-Ullrich JE, Grinstaff MW, Albro MB. A Bio-inspired Latent TGF-β Conjugated Scaffold Improves Neocartilage Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636279. [PMID: 39975171 PMCID: PMC11838498 DOI: 10.1101/2025.02.03.636279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
In cartilage tissue engineering, active TGF-β is conventionally supplemented in culture medium at highly supraphysiologic doses to accelerate neocartilage development. While this approach enhances cartilage extracellular matrix (ECM) biosynthesis, it further promotes tissue features detrimental to hyaline cartilage function, including the induction of tissue swelling, hyperplasia, hypertrophy, and ECM heterogeneities. In contrast, during native cartilage development, chondrocytes are surrounded by TGF-β configured in a latent complex (LTGF-β), which undergoes cell-mediated activation, giving rise to moderated, physiologic dosing regimens that enhance ECM biosynthesis while avoiding detrimental features associated with TGF-β excesses. Here, we explore a bio-inspired strategy, consisting of LTGF-β-conjugated scaffolds, providing TGF-β exposure regimens that are moderated and uniformly administered throughout the construct. Specifically, we evaluate the performance of LTGF-β scaffolds to improve neocartilage development with bovine chondrocyte-seeded agarose constructs compared to outcomes from active TGF-β media supplementation (MS) at a physiologic 0.3 ng/mL dose (MS-0.3), supraphysiologic 10 ng/mL dose (MS-10), or TGF-β free. For small-size constructs (∅3×2 mm), LTGF-β scaffolds yield neocartilage that achieves native-matched mechanical properties (800-925 kPa) and sGAG content (6.6%-7.1%), while providing a cell morphology and collagen distribution more reminiscent of hyaline cartilage. LTGF-β scaffolds further afford an optimal chondrogenic phenotype, marked by a 12-to 28-fold reduction of COL-I expression relative to TGF-β-free and a 7-to 17-fold reduction of COL-X expression relative to MS-10. Further, for large-size constructs, which approach the dimensions needed for clinical cartilage repair, LTGF-β scaffolds significantly reduce mechanical and biochemical heterogeneities relative to MS-0.3 and MS-10. Overall, the use of LTGF-β scaffolds improves the composition, structure, material properties, and cell phenotype of neocartilage.
Collapse
|
7
|
Pico OA, Espinoza F, Cádiz MI, Sossa CL, Becerra-Bayona SM, Salgado MCC, Rodríguez JER, Cárdenas OFV, Cure JMQ, Khoury M, Arango-Rodríguez ML. Efficacy of a single dose of cryopreserved human umbilical cord mesenchymal stromal cells for the treatment of knee osteoarthritis:a randomized, controlled, double-blind pilot study. Cytotherapy 2025; 27:188-200. [PMID: 39503681 DOI: 10.1016/j.jcyt.2024.09.005] [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: 06/26/2024] [Revised: 09/14/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Knee osteoarthritis (OA) is the most prevalent degenerative musculoskeletal disorder, which is particularly common in older population. While conventional treatments have limited effectiveness, the development of more effective therapeutic strategies is necessary to address this primary source of pain and disability. Umbilical cord mesenchymal stromal cells (UC-MSCs) offer a promising therapeutic approach for treating knee OA. AIM This randomized, prospective, double-blind and controlled pilot study was carried out to evaluate and compare the safety and therapeutic efficacy of a single intra-articular injection of a standardized product CellistemOA (5 × 106 ± 5 × 105 UC-MSCs), vs. triamcinolone (a synthetic corticosteroid) (10 mg/mL) in thirty patients with symptomatic knee OA (Kellgren-Lawrence grade II or III). METHODS The outcomes included changes in Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores based on a Likert scale, numerical rating score (NRS) for pain, Magnetic Resonance Imaging (MRI), and quality of life (SF-36 questionnaire), from baseline and throughout 12-months of follow-up. RESULTS Patients treated with CellistemOA showed significant improvement in WOMAC score (including the three subscale scores (pain, stiffness and function), NRS in pain, and SF-36 profile from baseline to 12 months (p < 0.05) compared to the triamcinolone group, and no severe adverse events were reported. There were no significant differences in MRI WORMS scores between the two groups. However, patients who received the cellular treatment experienced a significant improvement in their SF-36 profile (p < 0.05). CONCLUSIONS This pilot study revealed that a single dose of CellistemOA is safe and superior to the active comparator in knee OA at 1-year of follow-up, making it a compelling therapeutic alternative to treat symptomatic OA patients.
Collapse
Affiliation(s)
- Omar Amado Pico
- Fundación Oftalmológica de Santander - FOSCAL, Floridablanca, Colombia; Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia
| | - Francisco Espinoza
- Cells for Cells & Consorcio Regenero, Santiago, Chile; Program for Translational Research in Cell Therapy, Universidad de los Andes, Santiago, Chile; Department of Rheumatology, Universidad de los Andes, Santiago, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - María Ignacia Cádiz
- Cells for Cells & Consorcio Regenero, Santiago, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Laboratory of Nano-Regenerative Medicine, Universidad de los Andes, Santiago, Chile
| | - Claudia L Sossa
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia; Programa para el Tratamiento y Estudio de Enfermedades Hematológicas y Oncológicas de Santander (PROTEHOS), Floridablanca, Colombia
| | - Silvia M Becerra-Bayona
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia
| | - María C Canencio Salgado
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia
| | | | | | | | - Maroun Khoury
- Cells for Cells & Consorcio Regenero, Santiago, Chile; Program for Translational Research in Cell Therapy, Universidad de los Andes, Santiago, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Laboratory of Nano-Regenerative Medicine, Universidad de los Andes, Santiago, Chile
| | - Martha L Arango-Rodríguez
- Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga - UNAB, Bucaramanga, Colombia; Banco Multitejidos y Centro de Terapias Avanzadas, Clínica FOSCAL Internacional, Floridablanca, Colombia.
| |
Collapse
|
8
|
Trivedi J, Desai S, Molino J, Owens BD, Jayasuriya CT. Intra-articular Injections of CXCR4-Overexpressing Human Cartilage-Derived Progenitor Cells Improve Meniscus Healing and Protect Against Posttraumatic Osteoarthritis in Immunocompetent Rabbits. Am J Sports Med 2025; 53:396-405. [PMID: 39763468 PMCID: PMC11794014 DOI: 10.1177/03635465241309305] [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] [Indexed: 01/25/2025]
Abstract
BACKGROUND Meniscal injuries that fail to heal instigate catabolic changes in the knee's microenvironment, posing a high risk for developing posttraumatic osteoarthritis (PTOA). Previous research has suggested that human cartilage-derived progenitor cells (hCPCs) can stimulate meniscal repair in a manner that depends on stromal cell-derived factor 1 (SDF-1) pathway activity. HYPOTHESIS Overexpressing the SDF-1 receptor CXCR4 in hCPCs will increase cell trafficking and further improve the repair efficacy of meniscal injuries. STUDY DESIGN Controlled laboratory study. METHODS hCPCs were genetically modified to overexpress CXCR4 (CXCR4-overexpressing [OE] hCPCs) using lentivirus. In vitro characterization was performed using cell viability assay, cell migration assay, and immunoblotting. These cells were then used to treat a meniscal injury in rabbits. A medial meniscal tear was surgically created in the right knees of New Zealand White rabbits, followed by 2 intra-articular injections (5.0 × 106 cells each) of either CXCR4-OE hCPCs, wild-type hCPCs, or saline alone. A histological assessment of menisci and cartilage was performed using safranin O/fast green staining. Joints were assessed for PTOA changes using the modified Osteoarthritis Research Society International scoring system. Fluorescence imaging and DNA analysis were performed to examine tissue for human cells. RESULTS SDF-1 inhibited NF-κB and ERK pathways in both wild-type and CXCR4-OE hCPCs. CXCR4 overexpression increased hCPC trafficking toward sources of SDF-1, including injured meniscal fibrocartilage and an SDF-1-presoaked collagen scaffold. Intra-articular injections of CXCR4-OE hCPCs significantly improved meniscus healing, as evidenced by the complete absence of tears in 5 of 6 (83%) animals that received CXCR4-OE hCPCs compared with only 3 of 6 (50%) wild-type hCPC-treated animals and 2 of 6 (33%) animals in the saline control group. CXCR4-OE hCPC-treated animals also showed significantly less erosion in their knee cartilage compared with control animals. CONCLUSION Overall, CXCR4 overexpression inhibited catabolic pathway signaling in hCPCs and increased cell migration. Evidence suggests that intra-articular injections of these cells into the injured knee allow them to home in on sites of fibrocartilage injuries and ultimately result in meniscal tear healing and PTOA inhibition in immunocompetent animals. CLINICAL RELEVANCE This study demonstrated that cartilage progenitors with elevated CXCR4 expression have the potential to be a potent therapeutic tool for stimulating meniscal tear healing.
Collapse
Affiliation(s)
| | | | | | | | - Chathuraka T. Jayasuriya
- Address correspondence to Chathuraka T. Jayasuriya, PhD, Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, 1 Hoppin Street, Suite 4.313, Providence, RI 02903, USA () (X: @JayasuriyaLab)
| |
Collapse
|
9
|
Meenakshi Sundaram RS, Rupert S, Srinivasan P, Sathyanesan J, Govarthanan K, Jeyaraman N, Ramasubramanian S, Jeyaraman M, Chung HY, Gangadaran P, Ahn BC. Decoding Cytokine Dynamics: Wharton's Jelly Stromal Cells and Chondro-Differentiates in PHA-Stimulated Co-Culture. Cells 2025; 14:174. [PMID: 39936966 PMCID: PMC11817647 DOI: 10.3390/cells14030174] [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: 12/03/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
INTRODUCTION Articular cartilage damage presents a significant clinical challenge, with limited options for effective regeneration. Mesenchymal stromal cells (MSCs) derived from Wharton's jelly (WJ) are a promising cell source for cartilage repair due to their regenerative and immunomodulatory properties. While undifferentiated MSCs have demonstrated potent immunoregulatory effects, the immunomodulatory potential of chondrocytes derived from WJ-MSCs remains underexplored, particularly under inflammatory conditions. This study investigates the differential cytokine expression profiles of WJ-MSC-derived chondrocytes and undifferentiated MSCs under inflammatory stimulation with phytohemagglutinin (PHA) to understand their immunomodulatory capacities. MATERIALS AND METHODS WJ-MSCs were differentiated into chondrocytes using a micromass culture system. Differentiated chondrocytes were then co-cultured with immune cells under PHA-induced inflammatory conditions. Control groups included co-cultured cells without PHA activation and chondrocytes activated with PHA in the absence of immune cell interaction. Cytokine expression profiles were analyzed using the RT2 Customized Gene Array to evaluate pro- and anti-inflammatory markers. Morphological changes were assessed microscopically. The immunomodulatory responses of chondrocytes were compared to those of undifferentiated MSCs under the same experimental conditions. RESULTS Chondrocytes co-cultured with immune cells under PHA activation exhibited downregulation of IDO, HLA-G, PDGF, IL-10, TNF-α, IL-6, and IFN-γ compared to undifferentiated MSCs in similar conditions. In non-PHA co-cultured conditions, chondrocytes showed increased expression of IL-6, IFN-γ, IL-4, VEGF, iNOS, PDGF, PTGS-2 and TGF-β, while TNF-α, IL-10, IDO and HLA-G were decreased. In contrast, chondrocytes activated with PHA without immune cell interaction displayed reduced expression of HLA-G and TNF-α, with no significant changes in IL-6, IFN-γ, IL-4, IL-10, VEGF, PDGF, PTGS-2, TGF-β, IDO, and iNOS compared to PHA-stimulated undifferentiated MSCs. CONCLUSION This study demonstrates that chondrocytes derived from WJ-MSCs exhibit limited immunomodulatory potential compared to undifferentiated MSCs, particularly under PHA-induced inflammatory conditions. Undifferentiated MSCs showed superior regulation of key cytokines associated with immune modulation. These findings suggest that maintaining MSCs in an undifferentiated state may be advantageous for therapeutic applications targeting inflammatory conditions, such as osteoarthritis. Future research should explore strategies to enhance the immunomodulatory efficacy of chondrocytes, potentially through genetic modification or adjunctive therapies.
Collapse
Affiliation(s)
- Raja Sundari Meenakshi Sundaram
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Secunda Rupert
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Prasanna Srinivasan
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Jeswanth Sathyanesan
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Kavitha Govarthanan
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India;
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr. MGR Educational and Research Institute, Chennai 600017, Tamil Nadu, India; (N.J.); (M.J.)
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India;
| | - Swaminathan Ramasubramanian
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India;
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr. MGR Educational and Research Institute, Chennai 600017, Tamil Nadu, India; (N.J.); (M.J.)
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India;
| | - Ho Yun Chung
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea;
| | - Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
10
|
Ghamrawi A, Basso R, Shakik N, Haddad L, Nasr Z, Harmouch C. Wharton's Jelly Mesenchymal Stem Cells: Shaping the Future of Osteoarthritis Therapy with Advancements in Chitosan-Hyaluronic Acid Scaffolds. Stem Cells Dev 2025; 34:1-16. [PMID: 39605205 DOI: 10.1089/scd.2024.0033] [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: 11/29/2024] Open
Abstract
This review explores the potential of Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) in cartilage regeneration and osteoarthritis treatment. It covers key factors influencing chondrogenesis, including growth factors, cytokines, and hypoxia, focusing on precise timing. The effectiveness of three-dimensional cultures and scaffold-based strategies in chondrogenic differentiation is discussed. Specific biomaterials such as chitosan and hyaluronic acid are highlighted for tissue engineering. The document reviews clinical applications, incorporating evidence from animal research and early trials and molecular and histological assessments of chondrogenic differentiation processes. It addresses challenges and strategies for optimizing MSC-derived chondrocyte therapy, emphasizing the immunomodulatory properties of these cells. The review concludes as a comprehensive road map for future research and clinical applications in regenerative medicine.
Collapse
Affiliation(s)
- Ahed Ghamrawi
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, Tripoli, Lebanon
| | - Rasha Basso
- Department of Medical Laboratory Sciences, Faculty of Health Sciences University of Balamand, Beirut, Lebanon
| | - Nour Shakik
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, Tripoli, Lebanon
| | - Lara Haddad
- Department of Medical Laboratory Sciences, Faculty of Health Sciences University of Balamand, Beirut, Lebanon
| | - Zeina Nasr
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, Tripoli, Lebanon
| | - Chaza Harmouch
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, Tripoli, Lebanon
| |
Collapse
|
11
|
Zhong Y, Zhang B, Somoza R, Caplan AI, Welter JF, Baskaran H. Amino Acid Uptake Limitations during Human Mesenchymal Stem Cell-Based Chondrogenesis. Tissue Eng Part A 2025; 31:1-12. [PMID: 38517098 PMCID: PMC11807877 DOI: 10.1089/ten.tea.2024.0032] [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: 01/17/2024] [Accepted: 03/12/2024] [Indexed: 03/23/2024] Open
Abstract
A mino acids are the essential building blocks for collagen and proteoglycan, which are the main constituents for cartilage extracellular matrix (ECM). Synthesis of ECM proteins requires the uptake of various essential/nonessential amino acids. Analyzing amino acid metabolism during chondrogenesis can help to relate tissue quality to amino acid metabolism under different conditions. In our study, we studied amino acid uptake/secretion using human mesenchymal stem cell (hMSC)-based aggregate chondrogenesis in a serum-free induction medium with a defined chemical formulation. The initial glucose level and medium-change frequency were varied. Our results showed that essential amino acid uptake increased with time during hMSCs chondrogenesis for all initial glucose levels and medium-change frequencies. Essential amino acid uptake rates were initial glucose-level independent. The DNA-normalized glycosaminoglycans and hydroxyproline content of chondrogenic aggregates correlated with cumulative uptake of leucine, valine, and tryptophan regardless of initial glucose levels and medium-change frequencies. Collectively, our results show that amino acid uptake rates during in vitro chondrogenesis were insufficient to produce a tissue with an ECM content similar to that of human neonatal cartilage or adult cartilage. Furthermore, this deficiency was likely related to the downregulation of some key amino acid transporters in the cells. Such deficiency could be partially improved by increasing the amino acid availability in the chondrogenic medium by changing culture conditions.
Collapse
Affiliation(s)
- Yi Zhong
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| | - Bo Zhang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| | - Rodrigo Somoza
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| | - Arnold I. Caplan
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| | - Jean F. Welter
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| | - Harihara Baskaran
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
12
|
Levis H, Lewis C, Fainor M, Lawal A, Stockham E, Weston J, Farhang N, Gullbrand SE, Bowles RD. Targeted CRISPR regulation of ZNF865 enhances stem cell cartilage deposition, tissue maturation rates, and mechanical properties in engineered intervertebral discs. Acta Biomater 2025; 191:276-291. [PMID: 39521313 DOI: 10.1016/j.actbio.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/08/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Cell and tissue engineering based approaches have garnered significant interest for treating intervertebral disc degeneration and associated low back pain due to the substantial limitations of currently available clinical treatments. Herein we present a clustered regularly interspaced short palindromic repeats (CRISPR)-guided gene modulation strategy to improve the therapeutic potential of cell and tissue engineering therapies for treating intervertebral disc disease. Recently, we discovered a zinc finger (ZNF) protein, ZNF865 (BLST), which is associated with no in-depth publications and has not been functionally characterized. Utilizing CRISPR-guided gene modulation, we show that ZNF865 regulates cell cycle progression and protein processing. As a result, regulating this gene acts as a primary titratable regulator of cell activity. We also demonstrate that targeted ZNF865 regulation can enhance protein production and fibrocartilage-like matrix deposition in human adipose-derived stem cells (hASCs). Furthermore, we demonstrate CRISPR-engineered hASCs ability to increase GAG and collagen II matrix deposition in human-size tissue-engineered discs by 8.5-fold and 88.6-fold, respectively, while not increasing collagen X expression compared to naive hASCs dosed with growth factors. With this increased tissue deposition, we observe significant improvements in compressive mechanical properties, generating a stiffer and more robust tissue. Overall, we present novel biology on ZNF865 and display the power of CRISPR-cell engineering to enhance strategies treating musculoskeletal disease. STATEMENT OF SIGNIFICANCE: This work reports on a novel gene, ZNF865 (also known as BLST), that when regulated with CRISPRa, improves cartilagenous tissue deposition in human sized tissue engineering constructs. Producing tissue engineering constructs at human scale has proven difficult, and this strategy presents a broadly applicable tool to enhance a cells ability to produce tissue at these scales, as we saw an ∼8-88 fold increase in tissue deposition and significantly improved biomechanics in large tissue engineered intervertebral disc compared to traditional growth factor differentiation methods. Additionally, this work begins to elucidate the biology of this novel zinc finger protein, which appears to be critical in regulating cell function and activity.
Collapse
Affiliation(s)
- Hunter Levis
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Christian Lewis
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Matthew Fainor
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States; Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
| | - Ameerah Lawal
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States; Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
| | - Elise Stockham
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Jacob Weston
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Niloofar Farhang
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Sarah E Gullbrand
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States; Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
| | - Robby D Bowles
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States; Department of Orthopaedic Surgery, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
13
|
Tao H, Feng M, Feng H, Ren H. Research advance of 3D printing for articular cartilage regeneration. Regen Med 2025; 20:45-55. [PMID: 39957623 PMCID: PMC11881833 DOI: 10.1080/17460751.2025.2466346] [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: 12/08/2024] [Accepted: 02/10/2025] [Indexed: 02/18/2025] Open
Abstract
Articular cartilage lesion frequently leads to dysfunction and the development of degenerative diseases, posing a significant public health challenge due to the limited self-healing capacity of cartilage tissue. Current surgical treatments, including marrow stimulation techniques and osteochondral autografts/allografts, have limited efficacy or have significant drawbacks, highlighting the urgent need for alternative strategies. Advances in 3D printing for cartilage regeneration have shown promising potential in creating cartilage-mimicking constructs, thereby opening new possibilities for cartilage repair. In this review, we summarize current surgical treatment methods and their limitations for addressing articular cartilage lesion, various 3D printing strategies and their features in cartilage tissue engineering, seed cells from different sources, and different types of biomaterials. We also explore the benefits, current challenges, and future research directions for 3D printing in the treatment of articular cartilage lesion within the field of cartilage tissue engineering.
Collapse
Affiliation(s)
- Haicheng Tao
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Mingli Feng
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hui Feng
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hongchen Ren
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Sawyer M, Semodji A, Nielson O, Rektor A, Burgoyne H, Eppel M, Eixenberger J, Montenegro-Brown R, Nelson ML, Lujan T, Estrada D. Direct Scaffold-Coupled Electrical Stimulation of Chondrogenic Progenitor Cells through Graphene Foam Bioscaffolds to Control Mechanical Properties of Graphene Foam - Cell Composites. RESEARCH SQUARE 2024:rs.3.rs-5589589. [PMID: 39764126 PMCID: PMC11703340 DOI: 10.21203/rs.3.rs-5589589/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Osteoarthritis, a major global cause of pain and disability, is driven by the irreversible degradation of hyaline cartilage in joints. Cartilage tissue engineering presents a promising therapeutic avenue, but success hinges on replicating the native physiological environment to guide cellular behavior and generate tissue constructs that mimic natural cartilage. Although electrical stimulation has been shown to enhance chondrogenesis and extracellular matrix production in 2D cultures, the mechanisms underlying these effects remain poorly understood, particularly in 3D models. Here, we report that direct scaffold-coupled electrical stimulation applied to 3D graphene foam bioscaffolds significantly enhances the mechanical properties of the resulting graphene foam - cell constructs. Using custom 3D-printed electrical stimulus chambers, we applied biphasic square impulses (20, 40, 60 mVpp at 1 kHz) for 5 minutes daily over 7 days. Stimulation at 60 mVpp increased the steady-state energy dissipation and equilibrium modulus by approximately 65% and 25%, respectively, compared to unstimulated controls, while also yielding the highest cell density among stimulated samples. In addition, our custom chambers facilitated full submersion of the hydrophobic graphene foam in media, leading to enhanced cell attachment and integration across the scaffold surface and within its hollow branches. To assess this cellular integration, we employed co-localized confocal fluorescence microscopy and X-ray microCT imaging enabled by colloidal gold nanoparticle and fluorophore staining, which allowed visualization of cell distribution within the opaque scaffold's internal structure. These findings highlight the potential of direct scaffold-coupled electrical stimulus to modulate the mechanical properties of engineered tissues and offer new insights into the emergent behavior of cells within conductive 3D bioscaffolds.
Collapse
|
15
|
Adhikari B, Stager MA, Collins EG, Fischenich KM, Olusoji J, Ruble AF, Payne KA, Krebs MD. Sustained release of MAPK14-targeting siRNA from polyelectrolyte complex hydrogels mitigates MSC osteogenesis in vitro with potential application in growth plate injury. J Biomed Mater Res A 2024; 112:2346-2357. [PMID: 39145460 DOI: 10.1002/jbm.a.37784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 07/13/2024] [Accepted: 07/30/2024] [Indexed: 08/16/2024]
Abstract
The growth plate is a cartilage structure at the end of long bones which mediates growth in children. When fractured, the formation of bony repair tissue known as a "bony bar" can occur and cause limb deformities. There are currently no effective clinical solutions for the prevention of the bony bar formation or regeneration of healthy growth plate cartilage after a fracture. This study employs previously developed alginate/chitosan polyelectrolyte complex (PEC) hydrogels as a sustained release vehicle for the delivery of short-interfering RNA (siRNA). Specifically, the siRNA targets the p38-MAPK pathway in mesenchymal stem cells (MSCs) to prevent their osteogenic differentiation. In vitro experimental findings show sustained release of siRNA from the hydrogels for 6 months. Flow cytometry and confocal imaging indicate that the hydrogels release siRNA to effectively knockdown GFP expression over a sustained period. MAPK-14 targeting siRNA was used to knockdown the expression of MAPK-14 and correspondingly decrease the expression of other osteogenic genes in MSCs in vitro over the span of 21 days. These hydrogels were used in a rat model of growth plate injury to determine whether siMAPK-14 released from the gels could inhibit bony bar formation. No significant reduction of bony bar formation was seen in vivo at the one concentration of siRNA examined. This PEC hydrogel represents a significant advancement for siRNA sustained delivery, and presents an interesting potential therapeutic delivery system for growth plate injuries and other regenerative medicine applications.
Collapse
Affiliation(s)
- Bikram Adhikari
- Quantitative Biosciences and Bioengineering, Colorado School of Mines, Golden, Colorado, USA
| | - Michael A Stager
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Elise G Collins
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Kristine M Fischenich
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jesutomisin Olusoji
- Quantitative Biosciences and Bioengineering, Colorado School of Mines, Golden, Colorado, USA
| | - Ana Ferreira Ruble
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Karin A Payne
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Melissa D Krebs
- Quantitative Biosciences and Bioengineering, Colorado School of Mines, Golden, Colorado, USA
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| |
Collapse
|
16
|
Wu KC, Chang YH, Ding DC, Lin SZ. Mesenchymal Stromal Cells for Aging Cartilage Regeneration: A Review. Int J Mol Sci 2024; 25:12911. [PMID: 39684619 PMCID: PMC11641625 DOI: 10.3390/ijms252312911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Cartilage degeneration is a key feature of aging and osteoarthritis, characterized by the progressive deterioration of joint function, pain, and limited mobility. Current treatments focus on symptom relief, not cartilage regeneration. Mesenchymal stromal cells (MSCs) offer a promising therapeutic option due to their capability to differentiate into chondrocytes, modulate inflammation, and promote tissue regeneration. This review explores the potential of MSCs for cartilage regeneration, examining their biological properties, action mechanisms, and applications in preclinical and clinical settings. MSCs derived from bone marrow, adipose tissue, and other sources can self-renew and differentiate into multiple cell types. In aging cartilage, they aid in tissue regeneration by secreting growth factors and cytokines that enhance repair and modulate immune responses. Recent preclinical studies show that MSCs can restore cartilage integrity, reduce inflammation, and improve joint function, although clinical translation remains challenging due to limitations such as cell viability, scalability, and regulatory concerns. Advancements in MSC delivery, including scaffold-based approaches and engineered exosomes, may improve therapeutic effectiveness. Potential risks, such as tumorigenicity and immune rejection, are also discussed, emphasizing the need for optimized treatment protocols and large-scale clinical trials to develop effective, minimally invasive therapies for cartilage regeneration.
Collapse
Affiliation(s)
- Kun-Chi Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
- Institute of Medical Sciences, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
17
|
Wang X, Zheng Z, Zhang Y, Sun J, Liu J, Liu Y, Ding G. Application of hydrogel-loaded dental stem cells in the field of tissue regeneration. Hum Cell 2024; 38:2. [PMID: 39436502 DOI: 10.1007/s13577-024-01134-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
Mesenchymal stem cells (MSCs) are highly favored in clinical trials due to their unique characteristics, which have isolated from various human tissues. Derived from dental tissues, dental stem cells (DSCs) are particularly notable for their applications in tissue repair and regenerative medicine, attributed to their readily available sources, absence of ethical controversies, and minimal immunogenicity. Hydrogel-loaded stem cell therapy is widespread across a variety of injuries and diseases, and has good repair capabilities for both soft and hard tissues. This review comprehensively summarizes the regenerative and differentiation potential of various DSCs encapsulated in hydrogels across different tissues. In addition, the existing problems and future direction are also addressed. The application of hydrogel-DSCs composite has gained substantial progress in the field of tissue regeneration and need in-depth study in the future.
Collapse
Affiliation(s)
- Xiaolan Wang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China
| | - Zejun Zheng
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China
| | - Ying Zhang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China
| | - Jinmeng Sun
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China
| | - Jian Liu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China
| | - Yunxia Liu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China.
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Baotong West Street No.7166, Weifang, Shandong Province, China.
| |
Collapse
|
18
|
Alwohoush E, Ismail MA, Al-Kurdi B, Barham R, Al Hadidi S, Awidi A, Ababneh NA. Effect of hypoxia on proliferation and differentiation of induced pluripotent stem cell-derived mesenchymal stem cells. Heliyon 2024; 10:e38857. [PMID: 39421364 PMCID: PMC11483329 DOI: 10.1016/j.heliyon.2024.e38857] [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: 04/15/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
Although mesenchymal stem cells (MSCs) are extensively applied in the regenerative field, the majority of MSCs die after a few weeks of transplantation. Therefore, hypoxia pre-conditioning is a crucial step in increasing the MSCs' tolerance to physiological conditions. Meanwhile, induced pluripotent stem cell-derived MSCs (iMSCs) were proposed as a possible alternative to MSCs, and recently, the interest is growing in applying iMSCs in the regenerative field. This study examined the effect of hypoxia pre-conditioning on the proliferation, viability, and differentiation of iMSCs. Both iMSCs and MSCs were subjected to two rounds of severe short-term hypoxia (1 % O2 for 24h). After that, iMSCs and MSCs were characterized by testing their surface markers' expression, proliferation, viability, oxidative stress, and differentiation potential. Our findings revealed that hypoxia did not have a consistent effect among all the analyzed lines: the severe short-term hypoxia (1 % O2) reduced iMSCs proliferation, cell viability, and MMP while showing a benign effect on surface markers expression, colony formation, ROS accumulation, and osteogenic and adipogenic differentiation. Though hypoxia adversely affected iMSCs' proliferation, this does not necessarily mean that hypoxia is harmful to iMSCs; on the contrary, our results suggest that short-term hypoxia might have a beneficial long-term effect on the proliferation of iMSCs. Thus, the effect of hypoxia on proliferation, viability, and differentiation should also be tested after a long recovery period from iMSCs. Our next step will be to test the effect of hypoxia for a longer period besides uncovering the changes in the expression profile of hypoxic iMSCs.
Collapse
Affiliation(s)
- Enas Alwohoush
- Cell Therapy Center, the University of Jordan, Amman, Jordan
| | | | - Ban Al-Kurdi
- Cell Therapy Center, the University of Jordan, Amman, Jordan
| | - Raghda Barham
- Cell Therapy Center, the University of Jordan, Amman, Jordan
| | - Sabal Al Hadidi
- Cell Therapy Center, the University of Jordan, Amman, Jordan
| | - Abdalla Awidi
- Cell Therapy Center, the University of Jordan, Amman, Jordan
- Hemostasis and Thrombosis Laboratory, School of Medicine, the University of Jordan, Amman, Jordan
- Department of Hematology and Oncology, Jordan University Hospital, Amman, Jordan
| | | |
Collapse
|
19
|
Yau A, Sands I, Zhang W, Chen Y. Injectable Janus Base Nanomatrix (JBNm) in Maintaining Long-Term Homeostasis of Regenerated Cartilage for Tissue Chip Applications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.05.616785. [PMID: 39416084 PMCID: PMC11482866 DOI: 10.1101/2024.10.05.616785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Engineered cartilage tissues have wide applications in in vivo cartilage repair as well as in vitro models, such as cartilage-on-a-chip or cartilage tissue chips. Currently, most cartilage tissue engineering approaches focus on promoting chondrogenesis of stem cells to produce regenerated cartilage. However, this regenerated cartilage can dedifferentiate into fibrotic tissue or further differentiate into hypertrophic or calcified cartilage. One of the most challenging objectives in cartilage tissue engineering is to maintain long-term cartilage homeostasis. Since the microenvironment of engineered cartilage tissue is crucial for stem cell adhesion, proliferation, differentiation, and function, we aim to develop a novel scaffold that can maintain the long-term homeostasis of regenerated cartilage. Therefore, we developed a library of Janus base nanomatrices (JBNms), composed of DNA-inspired Janus nanotubes (JBNts) as well as cartilage extracellular matrix (ECM) proteins. The JBNms were developed to selectively promote chondro-lineage cell functions while inhibiting bone and endothelial cell growth. More importantly, the JBNm can effectively promote chondrogenesis while inhibiting hypertrophy, osteogenesis, angiogenesis, and dedifferentiation. Additionally, the JBNm is injectable, forming a solid scaffold suitable for producing and maintaining regenerated cartilage tissue in microfluidic chips, making it ideal for tissue chip applications. In this study, we successfully created cartilage tissue chips using JBNms. These chips can model cartilage tissue even after long-term culture and can also mimic arthritis progression, making them useful for drug screening. Thus, we have developed a novel nanomaterial approach for improved cartilage tissue engineering and cartilage tissue chip applications.
Collapse
Affiliation(s)
| | | | - Wuxia Zhang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Yupeng Chen
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
20
|
Zhang X, Cui C, Lin F. Efficacy and safety of mesenchymal stem cell injections for knee osteoarthritis: A systematic review and meta-analysis. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2024; 29:55. [PMID: 39629035 PMCID: PMC11613985 DOI: 10.4103/jrms.jrms_515_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/08/2024] [Accepted: 03/18/2024] [Indexed: 12/06/2024]
Abstract
Background There have not been any clear studies on the use of mesenchymal stem cells (MSCs) to treat osteoarthritis (OA) in the knee. Materials and Methods This study investigates the effects of different MSC dosages on pain alleviation in individuals with OA in the knee by conducting a meta-analysis of existing randomized controlled trials. Electronic resources such as Google Scholar, PubMed, Cochrane Library, and Web of Science were searched up until June 2023. Treatment effect sizes were computed using the knee osteoarthritis outcome score (KOOS), the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), and the Knee Society Score (KSS). Random or fixed effect models were applied to aggregate the data. We performed a subgroup analysis according to dosage level. The heterogeneity of the research was investigated using the Chi-square test and the I2 index. Results The meta-analysis included 26 studies with a total sample size of 739 patients. A significant reduction in pain was observed 1 year and 2 years following the injection of MSCs into the injured joint, as indicated by the Visual Analogue Scale, WOMAC, KOOS, and KSS indexes (P < 0.05). Patients on MSCs reported much reduced pain after 1 and 2 years compared to the control group (P < 0.05). Subgroup and meta-regression analyses revealed no statistically significant variations in the effectiveness of MSC dosage (P < 0.05). The studies did not report any adverse effects. Conclusion Different dosages of MSCs had the same pain-relieving effects on patients with OA in the knee. MSC injections were safe and beneficial in such cases.
Collapse
Affiliation(s)
- Xinguang Zhang
- Department of Joint Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Cunbao Cui
- Department of Joint Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Feng Lin
- Department of Joint Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
21
|
Skoracka J, Bajewska K, Kulawik M, Suchorska W, Kulcenty K. Advances in cartilage tissue regeneration: a review of stem cell therapies, tissue engineering, biomaterials, and clinical trials. EXCLI JOURNAL 2024; 23:1170-1182. [PMID: 39391058 PMCID: PMC11464958 DOI: 10.17179/excli2024-7088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/22/2024] [Indexed: 10/12/2024]
Abstract
Cartilage tissue, characterized by its limited regenerative capacity, presents significant challenges in clinical therapy. Recent advancements in cartilage regeneration have focused on integrating stem cell therapies, tissue engineering strategies, and advanced modeling techniques to overcome existing limitations. Stem cells, particularly Mesenchymal Stem Cells (MSCs) and induced pluripotent stem cells (iPSCs), hold promise for cartilage repair due to their ability to differentiate into chondrocytes, the key cells responsible for cartilage formation. Tissue engineering approaches, including 3D models, organ-on-a-chip systems, and organoids, offer innovative methods to mimic natural tissue microenvironments and evaluate potential treatments. MSC-based techniques, such as cell sheet tissue engineering, address challenges associated with traditional therapies, including cell availability and culture difficulties. Furthermore, advancements in 3D bioprinting enable the fabrication of complex tissue structures, while organ-on-a-chip systems provide microfluidic platforms for disease modeling and physiological mimicry. Organoids serve as simplified models of organs, capturing some complexity and enabling the monitoring of pathophysiological aspects of cartilage diseases. This comprehensive review underscores the transformative potential of integrating stem cell therapies, tissue engineering strategies, and advanced modeling techniques to improve cartilage regeneration and pave the way for more effective clinical treatments.
Collapse
Affiliation(s)
- Julia Skoracka
- Poznan University of Medical Sciences, Poznan, Poland, Fredry 10 Street, 61-701 Poznan, Poland
| | - Kaja Bajewska
- Poznan University of Medical Sciences, Poznan, Poland, Fredry 10 Street, 61-701 Poznan, Poland
| | - Maciej Kulawik
- Poznan University of Medical Sciences, Poznan, Poland, Fredry 10 Street, 61-701 Poznan, Poland
| | - Wiktoria Suchorska
- Department of Electroradiology, Poznan University of Medical Sciences,Garbary 15 Street, 61-866 Poznan, Poland
- Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866 Poznan, Poland
| | - Katarzyna Kulcenty
- Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866 Poznan, Poland
| |
Collapse
|
22
|
Chu YY, Hikita A, Asawa Y, Hoshi K. Advancements in chondrocyte 3-dimensional embedded culture: Implications for tissue engineering and regenerative medicine. Biomed J 2024; 48:100786. [PMID: 39236979 PMCID: PMC12018037 DOI: 10.1016/j.bj.2024.100786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/09/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
Cartilage repair necessitates regenerative medicine because of the unreliable healing mechanism of cartilage. To yield a sufficient number of cells for transplantation, chondrocytes must be expanded in culture. However, in 2D culture, chondrocytes tend to lose their distinctive phenotypes and functionalities after serial passage, thereby limiting their efficacy for tissue engineering purposes. The mechanism of dedifferentiation in 2D culture can be attributed to various factors, including abnormal nuclear strength, stress-induced mitochondrial impairment, chromatin remodeling, ERK-1/2 and the p38/mitogen-activated protein kinase (MAPK) signaling pathway. These mechanisms collectively contribute to the loss of chondrocyte phenotype and reduced production of cartilage-specific extracellular matrix (ECM) components. Chondrocyte 3D culture methods have emerged as promising solutions to prevent dedifferentiation. Techniques, such as scaffold-based culture and scaffold-free approaches, provide chondrocytes with a more physiologically relevant environment, promoting their differentiation and matrix synthesis. These methods have been used in cartilage tissue engineering to create engineered cartilage constructs for transplantation and joint repair. However, chondrocyte 3D culture still has limitations, such as low viability and proliferation rate, and also difficulties in passage under 3D condition. These indicate challenges of obtaining a sufficient number of chondrocytes for large-scale tissue production. To address these issues, ongoing studies of many research groups have been focusing on refining culture conditions, optimizing scaffold materials, and exploring novel cell sources such as stem cells to enhance the quality and quantity of engineered cartilage tissues. Although obstacles remain, continuous endeavors to enhance culture techniques and overcome limitations offer a promising outlook for the advancement of more efficient strategies for cartilage regeneration.
Collapse
Affiliation(s)
- Yu-Ying Chu
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Plastic and Reconstructive Surgery, Craniofacial Research Centre, Chang Gung Memorial Hospital at Linko, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Atsuhiko Hikita
- Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan
| | - Yukiyo Asawa
- Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan
| | - Kazuto Hoshi
- Department of Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Tissue Engineering, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
23
|
Zhang B, Berilla J, Cho S, Somoza RA, Welter JF, Alexander PE, Baskaran H. Synergistic effects of biological stimuli and flexion induce microcavities promote hypertrophy and inhibit chondrogenesis during in vitro culture of human mesenchymal stem cell aggregates. Biotechnol J 2024; 19:e2400060. [PMID: 39295570 PMCID: PMC11870314 DOI: 10.1002/biot.202400060] [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: 01/29/2024] [Revised: 06/26/2024] [Accepted: 07/30/2024] [Indexed: 09/21/2024]
Abstract
Interzone/cavitation are key steps in early stage joint formation that have not been successfully developed in vitro. Further, current models of endochondral ossification, an important step in early bone formation, lack key morphology morphological structures such as microcavities found during development in vivo. This is possibly due to the lack of appropriate strategies for incorporating chemical and mechanical stimuli that are thought to be involved in joint development. We designed a bioreactor system and investigated the synergic effect of chemical stimuli (chondrogenesis-inducing [CIM] and hypertrophy-inducing medium [HIM]) and mechanical stimuli (flexion) on the growth of human mesenchymal stem cells (hMSCs) based linear aggregates under different conditions over 4 weeks of perfusion culture. Computational studies were used to evaluate tissue stress qualitatively. After harvesting, both Safranin-O and hematoxylin & eosin (H&E) staining histology demonstrated microcavity structures and void structures in the region of higher stresses for tissue aggregates cultured only in HIM under flexion. In comparison to either HIM treatment or flexion only, increased glycosaminoglycan (GAG) content in the extracellular matrix (ECM) at this region indicates the morphological change resembles the early stage of joint cavitation; while decreased type II collagen (Col II), and increased type X collagen (Col X) and vascular endothelial growth factor (VEGF) with a clear boundary in the staining section indicates it resembles the early stage of ossification. Further, cell alignment analysis indicated that cells were mostly oriented toward the direction of flexion in high-stress region only in HIM under flexion, resembling cell morphology in both joint cavitation and hypertrophic cartilage in growth plate. Collectively, our results suggest that flexion and HIM inhibit chondrogenesis and promote hypertrophy and development of microcavities that resemble the early stage of joint cavitation and endochondral ossification. We believe the tissue model described in this work can be used to develop in vitro models of joint tissue for applications such as pathophysiology and drug discovery.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jim Berilla
- Case School of Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sungwoo Cho
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - Rodrigo A Somoza
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jean F Welter
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Peter E Alexander
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Harihara Baskaran
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
24
|
He S, Wang S, Liu R, Chen H, Wang Q, Jia D, Chen L, Dai J, Li X. Conditioned Medium of Infrapatellar Fat Stem Cells Alleviates Degradation of Chondrocyte Extracellular Matrix and Delays Development of Osteoarthritis. Gerontology 2024; 70:1171-1187. [PMID: 39159625 DOI: 10.1159/000540505] [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: 10/11/2023] [Accepted: 06/20/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Osteoarthritis (OA) is a prevalent clinical chronic degenerative condition characterized by the degeneration of articular cartilage. Currently, drug treatments for OA come with varying degrees of side effects, making the development of new therapeutic approaches for OA imperative. Mesenchymal stem cells (MSCs) are known to mitigate the progression of OA primarily through paracrine effects. The conditioned medium (CM) derived from MSCs encapsulates a variety of paracrine factors secreted by these cells. METHODS In this study, we investigated the effect of the CM of infrapatellar fat pad-derived MSCs (IPFSCs) on OA in vitro and in vivo, as well as and the potential underlying mechanisms. We established three experimental groups: the normal group, the OA group, and the CM intervention group. In vitro experiments, we used methods such as qPCR, Western blot, immunofluorescence, and flow cytometry to detect the impact of CM on OA chondrocytes. In vivo experiments, we evaluated the changes in the knee joints of OA rats after intra-articular injection of CM treatment. RESULTS The results showed that injection of CM into the knee joint inhibited OA development in a rat model induced by destabilization of the medial meniscus and anterior cruciate ligament transection. The CM increased the deposition of extracellular matrix-related components (type II collagen and Proteoglycan). The activation of PI3K/AKT/NF-κB signaling pathway was induced by IL-1β in chondrocytes, which was finally inhibited by CM-IPFSCs treatment. CONCLUSION In summary, IPFSCs-CM may have therapeutic potential for OA.
Collapse
Affiliation(s)
- Shiping He
- Panzhihua Central Hospital, Panzhihua, China
| | - Shihan Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Ruizhou Liu
- Medical College of Zhejiang University, Hangzhou, China,
| | - Hui Chen
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Qiang Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Dazhou Jia
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Longchi Chen
- Yangzhou Clinical School of Xuzhou Medical University, Yangzhou, China
| | - Jihang Dai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Xiaolei Li
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| |
Collapse
|
25
|
Gadomski SJ, Mui BW, Gorodetsky R, Paravastu SS, Featherall J, Li L, Haffey A, Kim JC, Kuznetsov SA, Futrega K, Lazmi-Hailu A, Merling RK, Martin D, McCaskie AW, Robey PG. Time- and cell-specific activation of BMP signaling restrains chondrocyte hypertrophy. iScience 2024; 27:110537. [PMID: 39193188 PMCID: PMC11347861 DOI: 10.1016/j.isci.2024.110537] [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: 04/14/2023] [Revised: 02/29/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Stem cell therapies for degenerative cartilage disease are limited by an incomplete understanding of hyaline cartilage formation and maintenance. Human bone marrow stromal cells/skeletal stem cells (hBMSCs/SSCs) produce stable hyaline cartilage when attached to hyaluronic acid-coated fibrin microbeads (HyA-FMBs), yet the mechanism remains unclear. In vitro, hBMSC/SSC/HyA-FMB organoids exhibited reduced BMP signaling early in chondrogenic differentiation, followed by restoration of BMP signaling in chondrogenic IGFBP5 + /MGP + cells. Subsequently, human-induced pluripotent stem cell (hiPSC)-derived sclerotome cells were established (BMP inhibition) and then treated with transforming growth factor β (TGF-β) -/+ BMP2 and growth differentiation factor 5 (GDF5) (BMP signaling activation). TGF-β alone elicited a weak chondrogenic response, but TGF-β/BMP2/GDF5 led to delamination of SOX9 + aggregates (chondrospheroids) with high expression of COL2A1, ACAN, and PRG4 and minimal expression of COL10A1 and ALP in vitro. While transplanted hBMSCs/SSCs/HyA-FMBs did not heal articular cartilage defects in immunocompromised rodents, chondrospheroid-derived cells/HyA-FMBs formed non-hypertrophic cartilage that persisted until at least 5 months in vivo.
Collapse
Affiliation(s)
- Stephen J. Gadomski
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- NIH Oxford-Cambridge Scholars Program in Partnership with Medical University of South Carolina, Charleston, SC 29425, USA
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | - Byron W.H. Mui
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
- NIH Oxford-Cambridge Scholars Program in Partnership with Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- NIH Medical Research Scholars Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Raphael Gorodetsky
- Lab of Biotechnology and Radiobiology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Sriram S. Paravastu
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- NIH Medical Research Scholars Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joseph Featherall
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- NIH Medical Research Scholars Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Li Li
- National Institute of Dental and Craniofacial Research Imaging Core, National Institutes of Health, Bethesda, MD 20892, USA
| | - Abigail Haffey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- National Institute of Dental and Craniofacial Research Summer Internship Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jae-Chun Kim
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
- National Institute of Dental and Craniofacial Research Summer Dental Student Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sergei A. Kuznetsov
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Kathryn Futrega
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Astar Lazmi-Hailu
- Lab of Biotechnology and Radiobiology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Randall K. Merling
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - NIDCD/NIDCR Genomics and Computational Biology Core,
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, 35A Convent Drive, Room 1F-103, Bethesda, MD 20892, USA
- Genomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Martin
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, 35A Convent Drive, Room 1F-103, Bethesda, MD 20892, USA
- Genomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew W. McCaskie
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
- Department of Surgery, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Pamela G. Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| |
Collapse
|
26
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [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: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
27
|
Mishra A, Kumar R, Harilal S, Nigam M, Datta D, Singh S. Emerging Landscape of In Vitro Models for Assessing Rheumatoid Arthritis Management. ACS Pharmacol Transl Sci 2024; 7:2280-2305. [PMID: 39144547 PMCID: PMC11320735 DOI: 10.1021/acsptsci.4c00260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 08/16/2024]
Abstract
Rheumatoid arthritis (RA) is a complex condition that is influenced by various causes, including immunological, genetic, and environmental factors. Several studies using animal models have documented immune system dysfunction and described the clinical characteristics of the disease. These studies have provided valuable insights into the pathogenesis of inflammatory arthritis and the identification of new targets for treatment. Nevertheless, none of these animal models successfully replicated all the characteristics of RA. Additionally, numerous experimental medications, which were developed based on our enhanced comprehension of the immune system's function in RA, have shown potential in animal research but ultimately proved ineffective during different stages of clinical trials. There have been several novel therapy alternatives, which do not achieve a consistently outstanding therapeutic outcome in all patients. This underscores the importance of employing the progress in in vitro models, particularly 3D models like tissue explants, and diverse multicomponent approaches such as coculture strategies, synovial membrane, articular cartilage, and subchondral bone models that accurately replicate the structural characteristics of RA pathophysiology. These methods are crucial for the advancement of potential therapeutic strategies. This review discusses the latest advancements in in vitro models and their potential to greatly impact research on managing RA.
Collapse
Affiliation(s)
- Abhay
Prakash Mishra
- Department
of Pharmacology, University of Free State, Bloemfontein 9301, South Africa
- Department
of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Rajesh Kumar
- Faculty
of Pharmaceutical Sciences, Kerala University
of Health Sciences, Kerala 680596, India
| | - Seetha Harilal
- Faculty
of Pharmaceutical Sciences, Kerala University
of Health Sciences, Kerala 680596, India
| | - Manisha Nigam
- Department
of Biochemistry, Hemvati Nandan Bahuguna
Garhwal University, Srinagar
Garhwal, Uttarakhand 246174, India
| | - Deepanjan Datta
- Department
of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sudarshan Singh
- Office of
Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
- Faculty of
Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
28
|
Park DY, Kim SH, Park SH, Jang JS, Yoo JJ, Lee SJ. 3D Bioprinting Strategies for Articular Cartilage Tissue Engineering. Ann Biomed Eng 2024; 52:1883-1893. [PMID: 37204546 DOI: 10.1007/s10439-023-03236-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023]
Abstract
Articular cartilage is the avascular and aneural tissue which is the primary connective tissue covering the surface of articulating bone. Traumatic damage or degenerative diseases can cause articular cartilage injuries that are common in the population. As a result, the demand for new therapeutic options is continually increasing for older people and traumatic young patients. Many attempts have been made to address these clinical needs to treat articular cartilage injuries, including osteoarthritis (OA); however, regenerating highly qualified cartilage tissue remains a significant obstacle. 3D bioprinting technology combined with tissue engineering principles has been developed to create biological tissue constructs that recapitulate the anatomical, structural, and functional properties of native tissues. In addition, this cutting-edge technology can precisely place multiple cell types in a 3D tissue architecture. Thus, 3D bioprinting has rapidly become the most innovative tool for manufacturing clinically applicable bioengineered tissue constructs. This has led to increased interest in 3D bioprinting in articular cartilage tissue engineering applications. Here, we reviewed current advances in bioprinting for articular cartilage tissue engineering.
Collapse
Affiliation(s)
- Do Young Park
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Orthopedic Surgery, Ajou University Hospital, Suwon, Republic of Korea
| | - Seon-Hwa Kim
- Department of Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea
| | - Sang-Hyug Park
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea
| | - Ji Su Jang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Anesthesiology and Pain Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
29
|
Imere A, Foster NC, Hajiali H, Okur KE, Wright AL, Barroso IA, Haj AJE. Enhanced chondrogenic potential in GelMA-based 3D cartilage model via Wnt3a surface immobilization. Sci Rep 2024; 14:15022. [PMID: 38951570 PMCID: PMC11217376 DOI: 10.1038/s41598-024-65970-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
Cartilage tissue engineering aims to develop functional substitutes for treating cartilage defects and osteoarthritis. Traditional two-dimensional (2D) cell culture systems lack the complexity of native cartilage, leading to the development of 3D regenerative cartilage models. In this study, we developed a 3D model using Gelatin Methacryloyl (GelMA)-based hydrogels seeded with Y201 cells, a bone marrow mesenchymal stem cell line. The model investigated chondrogenic differentiation potential in response to Wnt3a stimulation within the GelMA scaffold and validated using known chondrogenic agonists. Y201 cells demonstrated suitability for the model, with increased proteoglycan content and upregulated chondrogenic marker expression under chondrogenic conditions. Wnt3a enhanced cell proliferation, indicating activation of the Wnt/β-catenin pathway, which plays a role in cartilage development. GelMA hydrogels provided an optimal scaffold, supporting cell viability and proliferation. The 3D model exhibited consistent responses to chondrogenic agonists, with TGF-β3 enhancing cartilage-specific extracellular matrix (ECM) production and chondrogenic differentiation. The combination of Wnt3a and TGF-β3 showed synergistic effects, promoting chondrogenic differentiation and ECM production. This study presents a 3D regenerative cartilage model with potential for investigating cartilage biology, disease mechanisms, and drug screening. The model provides insights into complex cartilage regeneration mechanisms and offers a platform for developing therapeutic approaches for cartilage repair and osteoarthritis treatment.
Collapse
Affiliation(s)
- Angela Imere
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Nicola C Foster
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Hadi Hajiali
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Kerime Ebrar Okur
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Abigail L Wright
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ines A Barroso
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Alicia J El Haj
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
30
|
Ali EAM, Smaida R, Meyer M, Ou W, Li Z, Han Z, Benkirane-Jessel N, Gottenberg JE, Hua G. iPSCs chondrogenic differentiation for personalized regenerative medicine: a literature review. Stem Cell Res Ther 2024; 15:185. [PMID: 38926793 PMCID: PMC11210138 DOI: 10.1186/s13287-024-03794-1] [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: 03/28/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Cartilage, an important connective tissue, provides structural support to other body tissues, and serves as a cushion against impacts throughout the body. Found at the end of the bones, cartilage decreases friction and averts bone-on-bone contact during joint movement. Therefore, defects of cartilage can result from natural wear and tear, or from traumatic events, such as injuries or sudden changes in direction during sports activities. Overtime, these cartilage defects which do not always produce immediate symptoms, could lead to severe clinical pathologies. The emergence of induced pluripotent stem cells (iPSCs) has revolutionized the field of regenerative medicine, providing a promising platform for generating various cell types for therapeutic applications. Thus, chondrocytes differentiated from iPSCs become a promising avenue for non-invasive clinical interventions for cartilage injuries and diseases. In this review, we aim to highlight the current strategies used for in vitro chondrogenic differentiation of iPSCs and to explore their multifaceted applications in disease modeling, drug screening, and personalized regenerative medicine. Achieving abundant functional iPSC-derived chondrocytes requires optimization of culture conditions, incorporating specific growth factors, and precise temporal control. Continual improvements in differentiation methods and integration of emerging genome editing, organoids, and 3D bioprinting technologies will enhance the translational applications of iPSC-derived chondrocytes. Finally, to unlock the benefits for patients suffering from cartilage diseases through iPSCs-derived technologies in chondrogenesis, automatic cell therapy manufacturing systems will not only reduce human intervention and ensure sterile processes within isolator-like platforms to minimize contamination risks, but also provide customized production processes with enhanced scalability and efficiency.
Collapse
Affiliation(s)
- Eltahir Abdelrazig Mohamed Ali
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1260, Regenerative NanoMedicine (RNM), 1 Rue Eugène Boeckel, 67000, Strasbourg, France
- Université de Strasbourg, 67000, Strasbourg, France
| | - Rana Smaida
- Lamina Therapeutics, 1 Rue Eugène Boeckel, 67000, Strasbourg, France
| | - Morgane Meyer
- Université de Strasbourg, 67000, Strasbourg, France
- Lamina Therapeutics, 1 Rue Eugène Boeckel, 67000, Strasbourg, France
| | - Wenxin Ou
- Université de Strasbourg, 67000, Strasbourg, France
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), Service de Rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 67000, Strasbourg, France
- Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Zongjin Li
- Nankai University School of Medicine, Tianjin, 300071, China
| | - Zhongchao Han
- Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co, Beijing, 100176, China
| | - Nadia Benkirane-Jessel
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1260, Regenerative NanoMedicine (RNM), 1 Rue Eugène Boeckel, 67000, Strasbourg, France.
- Université de Strasbourg, 67000, Strasbourg, France.
- Lamina Therapeutics, 1 Rue Eugène Boeckel, 67000, Strasbourg, France.
| | - Jacques Eric Gottenberg
- Université de Strasbourg, 67000, Strasbourg, France.
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), Service de Rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 67000, Strasbourg, France.
| | - Guoqiang Hua
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1260, Regenerative NanoMedicine (RNM), 1 Rue Eugène Boeckel, 67000, Strasbourg, France.
- Université de Strasbourg, 67000, Strasbourg, France.
| |
Collapse
|
31
|
Zhra M, Magableh AM, Samhan LM, Fatani LM, Qasem RJ, Aljada A. The Expression of a Subset of Aging and Antiaging Markers Following the Chondrogenic and Osteogenic Differentiation of Mesenchymal Stem Cells of Placental Origin. Cells 2024; 13:1022. [PMID: 38920652 PMCID: PMC11201886 DOI: 10.3390/cells13121022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Mesenchymal stem cells (MSCs) of placental origin hold great promise in tissue engineering and regenerative medicine for diseases affecting cartilage and bone. However, their utility has been limited by their tendency to undergo premature senescence and phenotypic drift into adipocytes. This study aimed to explore the potential involvement of a specific subset of aging and antiaging genes by measuring their expression prior to and following in vitro-induced differentiation of placental MSCs into chondrocytes and osteoblasts as opposed to adipocytes. The targeted genes of interest included the various LMNA/C transcript variants (lamin A, lamin C, and lamin A∆10), sirtuin 7 (SIRT7), and SM22α, along with the classic aging markers plasminogen activator inhibitor 1 (PAI-1), p53, and p16INK4a. MSCs were isolated from the decidua basalis of human term placentas, expanded, and then analyzed for phenotypic properties by flow cytometry and evaluated for colony-forming efficiency. The cells were then induced to differentiate in vitro into chondrocytes, osteocytes, and adipocytes following established protocols. The mRNA expression of the targeted genes was measured by RT-qPCR in the undifferentiated cells and those fully differentiated into the three cellular lineages. Compared to undifferentiated cells, the differentiated chondrocytes demonstrated decreased expression of SIRT7, along with decreased PAI-1, lamin A, and SM22α expression, but the expression of p16INK4a and p53 increased, suggesting their tendency to undergo premature senescence. Interestingly, the cells maintained the expression of lamin C, which indicates that it is the primary lamin variant influencing the mechanoelastic properties of the differentiated cells. Notably, the expression of all targeted genes did not differ from the undifferentiated cells following osteogenic differentiation. On the other hand, the differentiation of the cells into adipocytes was associated with decreased expression of lamin A and PAI-1. The distinct patterns of expression of aging and antiaging genes following in vitro-induced differentiation of MSCs into chondrocytes, osteocytes, and adipocytes potentially reflect specific roles for these genes during and following differentiation in the fully functional cells. Understanding these roles and the network of signaling molecules involved can open opportunities to improve the handling and utility of MSCs as cellular precursors for the treatment of cartilage and bone diseases.
Collapse
Affiliation(s)
- Mahmoud Zhra
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Ahmad M. Magableh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Lara M. Samhan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Lein M. Fatani
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Rani J. Qasem
- Department of Pharmacology and Pharmacy Practice, College of Pharmacy, Middle East University, Amman 11831, Jordan
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
32
|
Upadhyay U, Kolla S, Maredupaka S, Priya S, Srinivasulu K, Chelluri LK. Development of an alginate-chitosan biopolymer composite with dECM bioink additive for organ-on-a-chip articular cartilage. Sci Rep 2024; 14:11765. [PMID: 38782958 PMCID: PMC11116456 DOI: 10.1038/s41598-024-62656-1] [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: 12/23/2023] [Accepted: 05/20/2024] [Indexed: 05/25/2024] Open
Abstract
In vitro use of articular cartilage on an organ-on-a-chip (OOAC) via microfluidics is challenging owing to the dense extracellular matrix (ECM) composed of numerous protein moieties and few chondrocytes, which has limited proliferation potential and microscale translation. Hence, this study proposes a novel approach for using a combination of biopolymers and decellularised ECM (dECM) as a bioink additive in the development of scalable OOAC using a microfluidic platform. The bioink was tested with native chondrocytes and mesenchymal stem cell-induced chondrocytes using biopolymers of alginate and chitosan composite hydrogels. Two-dimensional (2D) and three-dimensional (3D) biomimetic tissue construction approaches have been used to characterise the morphology and cellular marker expression (by histology and confocal laser scanning microscopy), viability (cell viability dye using flow cytometry), and genotypic expression of ECM-specific markers (by quantitative PCR). The results demonstrated that the bioink had a significant impact on the increase in phenotypic and genotypic expression, with a statistical significance level of p < 0.05 according to Student's t-test. The use of a cell-laden biopolymer as a bioink optimised the niche conditions for obtaining hyaline-type cartilage under culture conditions, paving the way for testing mechano-responsive properties and translating these findings to a cartilage-on-a-chip microfluidics system.
Collapse
Affiliation(s)
- Upasna Upadhyay
- Stem Cell Unit, Global Medical Education and Research Foundation (GMERF), Lakdi-ka-pul, Hyderabad, Telangana, 500004, India
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF) Deemed to be University, Vaddeswaram, Vijayawada, Andhra Pradesh, 522302, India
| | - Saketh Kolla
- Department of Orthopaedics, Gleneagles Global Hospitals, Lakdi-ka-pul, Hyderabad, Telangana, 500004, India
| | - Siddhartha Maredupaka
- Department of Orthopaedics, Gleneagles Global Hospitals, Lakdi-ka-pul, Hyderabad, Telangana, 500004, India
| | - Swapna Priya
- Stem Cell Unit, Global Medical Education and Research Foundation (GMERF), Lakdi-ka-pul, Hyderabad, Telangana, 500004, India
| | - Kamma Srinivasulu
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF) Deemed to be University, Vaddeswaram, Vijayawada, Andhra Pradesh, 522302, India
| | - Lakshmi Kiran Chelluri
- Advanced Diagnostics and Therapeutics, Gleneagles Global Hospitals, Lakdi-ka-pul, Hyderabad, Telangana, 500004, India.
- Academics and Research, Global Medical Education and Research Foundation (GMERF), Gleneagles Global Hospitals, Lakdi-ka-pul, Hyderabad, Telangana, 500004, India.
| |
Collapse
|
33
|
Foltz L, Avabhrath N, Lanchy JM, Levy T, Possemato A, Ariss M, Peterson B, Grimes M. Craniofacial chondrogenesis in organoids from human stem cell-derived neural crest cells. iScience 2024; 27:109585. [PMID: 38623327 PMCID: PMC11016914 DOI: 10.1016/j.isci.2024.109585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 04/17/2024] Open
Abstract
Knowledge of cell signaling pathways that drive human neural crest differentiation into craniofacial chondrocytes is incomplete, yet essential for using stem cells to regenerate craniomaxillofacial structures. To accelerate translational progress, we developed a differentiation protocol that generated self-organizing craniofacial cartilage organoids from human embryonic stem cell-derived neural crest stem cells. Histological staining of cartilage organoids revealed tissue architecture and staining typical of elastic cartilage. Protein and post-translational modification (PTM) mass spectrometry and snRNA-seq data showed that chondrocyte organoids expressed robust levels of cartilage extracellular matrix (ECM) components: many collagens, aggrecan, perlecan, proteoglycans, and elastic fibers. We identified two populations of chondroprogenitor cells, mesenchyme cells and nascent chondrocytes, and the growth factors involved in paracrine signaling between them. We show that ECM components secreted by chondrocytes not only create a structurally resilient matrix that defines cartilage, but also play a pivotal autocrine cell signaling role in determining chondrocyte fate.
Collapse
Affiliation(s)
- Lauren Foltz
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| | - Nagashree Avabhrath
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| | - Jean-Marc Lanchy
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| | - Tyler Levy
- Cell Signaling Technology, Danvers, MA 01923, USA
| | | | - Majd Ariss
- Cell Signaling Technology, Danvers, MA 01923, USA
| | | | - Mark Grimes
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
34
|
Casado-Losada I, Acosta M, Schädl B, Priglinger E, Wolbank S, Nürnberger S. Unlocking Potential: Low Bovine Serum Albumin Enhances the Chondrogenicity of Human Adipose-Derived Stromal Cells in Pellet Cultures. Biomolecules 2024; 14:413. [PMID: 38672430 PMCID: PMC11048491 DOI: 10.3390/biom14040413] [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: 02/21/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Bovine serum albumin (BSA) plays a crucial role in cell culture media, influencing cellular processes such as proliferation and differentiation. Although it is commonly included in chondrogenic differentiation media, its specific function remains unclear. This study explores the effect of different BSA concentrations on the chondrogenic differentiation of human adipose-derived stromal/stem cells (hASCs). hASC pellets from six donors were cultured under chondrogenic conditions with three BSA concentrations. Surprisingly, a lower BSA concentration led to enhanced chondrogenesis. The degree of this effect was donor-dependent, classifying them into two groups: (1) high responders, forming at least 35% larger, differentiated pellets with low BSA in comparison to high BSA; (2) low responders, which benefitted only slightly from low BSA doses with a decrease in pellet size and marginal differentiation, indicative of low intrinsic differentiation potential. In all cases, increased chondrogenesis was accompanied by hypertrophy under low BSA concentrations. To the best of our knowledge, this is the first study showing improved chondrogenicity and the tendency for hypertrophy with low BSA concentration compared to standard levels. Once the tendency for hypertrophy is understood, the determination of BSA concentration might be used to tune hASC chondrogenic or osteogenic differentiation.
Collapse
Affiliation(s)
- Isabel Casado-Losada
- Department of Orthopedics and Trauma-Surgery, Division of Trauma-Surgery, Medical University of Vienna, 1090 Vienna, Austria; (I.C.-L.); (M.A.)
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria (E.P.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Melanie Acosta
- Department of Orthopedics and Trauma-Surgery, Division of Trauma-Surgery, Medical University of Vienna, 1090 Vienna, Austria; (I.C.-L.); (M.A.)
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria (E.P.); (S.W.)
| | - Barbara Schädl
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria (E.P.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Eleni Priglinger
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria (E.P.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Department for Orthopedics and Traumatology, Kepler University Hospital GmbH, Johannes Kepler University Linz, 4020 Linz, Austria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria (E.P.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Sylvia Nürnberger
- Department of Orthopedics and Trauma-Surgery, Division of Trauma-Surgery, Medical University of Vienna, 1090 Vienna, Austria; (I.C.-L.); (M.A.)
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria (E.P.); (S.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
35
|
Gaissmaier C, Angele P, Spiro RC, Köhler A, Kirner A, Niemeyer P. Hydrogel-Based Matrix-Associated Autologous Chondrocyte Implantation Shows Greater Substantial Clinical Benefit at 24 Months Follow-Up than Microfracture: A Propensity Score Matched-Pair Analysis. Cartilage 2024:19476035241235928. [PMID: 38501741 PMCID: PMC11569661 DOI: 10.1177/19476035241235928] [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] [Received: 12/12/2023] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 03/20/2024] Open
Abstract
OBJECTIVE To compare substantial clinical benefit (SCB) of a hydrogel-based, matrix-associated autologous chondrocyte implantation (M-ACI) method versus microfracture (MFx) in the treatment of knee cartilage defects. DESIGN Propensity score matched-pair analysis, using the MFx control group of a phase III study as comparator for M-ACI treatment in a single-arm phase III study, resulting in 144 patients in the matched-pair set. RESULTS Groups were comparable regarding baseline Knee Injury and Osteoarthritis Outcome Score (KOOS), sex, age, body mass index, symptom duration, smoking status, and previous knee surgeries. Defect sizes in the M-ACI group were significantly larger than in the MFx group (6.4 cm2 vs. 3.7 cm2). Other differences concerned location, number, and etiology of defects that were not considered to influence the interpretation of results. At 24 months, significantly more patients in the M-ACI group achieved SCB in KOOS pain (72.2% vs. 48.6%; P = 0.0108), symptoms (84.7% vs. 61.1%, P = 0.0039), sports/recreation (84.7% vs. 56.9%, P = 0.0008), and quality of life (QoL; 72.2% vs. 44.4%, P = 0.0014). The SCBs for KOOS activities in daily living and International Knee Documentation Committee score were higher for M-ACI but not significantly different from MFx. The SCB rates consistently favored M-ACI from 3 months onward. The highest improvements from baseline at 24 months in patients with SCB were observed for KOOS sports/rec. (M-ACI: 60.8 points, MFx: 55.9 points) and QoL (M-ACI: 58.1, MFx: 57.4). CONCLUSION Hydrogel-based M-ACI demonstrated superior SCB in KOOS pain, symptoms, sports/rec., and QoL compared with MFx in patients with knee cartilage defects through 2 years follow-up.
Collapse
Affiliation(s)
| | - Peter Angele
- Sporthopaedicum Regensburg, Regensburg, Germany
- Department of Trauma Surgery, University Medical Center Regensburg, Regensburg, Germany
| | | | - Annette Köhler
- TETEC—Tissue Engineering Technologies AG, Reutlingen, Germany
| | | | - Philipp Niemeyer
- OCM Orthopädische Chirurgie München, Munich, Germany
- Department of Orthopedics and Trauma Surgery, University Medical Center Freiburg, Albert Ludwig University of Freiburg, Freiburg, Germany
| |
Collapse
|
36
|
Walton BL, Shattuck-Brandt R, Hamann CA, Tung VW, Colazo JM, Brand DD, Hasty KA, Duvall CL, Brunger JM. A programmable arthritis-specific receptor for guided articular cartilage regenerative medicine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578281. [PMID: 38352576 PMCID: PMC10862827 DOI: 10.1101/2024.01.31.578281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Objective Investigational cell therapies have been developed as disease-modifying agents for the treatment of osteoarthritis (OA), including those that inducibly respond to inflammatory factors driving OA progression. However, dysregulated inflammatory cascades do not specifically signify the presence of OA. Here, we deploy a synthetic receptor platform that regulates cell behaviors in an arthritis-specific fashion to confine transgene expression to sites characterized by cartilage degeneration. Methods An scFv specific for type II collagen (CII) was used to produce a synthetic Notch (synNotch) receptor that enables "CII-synNotch" mesenchymal stromal cells (MSCs) to recognize CII fibers exposed in damaged cartilage. Engineered cell activation by both CII-treated culture surfaces and on primary tissue samples was measured via inducible reporter transgene expression. TGFβ3-expressing cells were assessed for cartilage anabolic gene expression via qRT-PCR. In a co-culture with CII-synNotch MSCs engineered to express IL-1Ra, ATDC5 chondrocytes were stimulated with IL-1α, and inflammatory responses of ATDC5s were profiled via qRT-PCR and an NF-κB reporter assay. Results CII-synNotch MSCs are highly responsive to CII, displaying activation ranges over 40-fold in response to physiologic CII inputs. CII-synNotch cells exhibit the capacity to distinguish between healthy and damaged cartilage tissue and constrain transgene expression to regions of exposed CII fibers. Receptor-regulated TGFβ3 expression resulted in upregulation of Acan and Col2a1 in MSCs, and inducible IL-1Ra expression by engineered CII-synNotch MSCs reduced pro-inflammatory gene expression in chondrocytes. Conclusion This work demonstrates proof-of-concept that the synNotch platform guides MSCs for spatially regulated, disease-dependent delivery of OA-relevant biologic drugs.
Collapse
Affiliation(s)
- Bonnie L. Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | | | - Catherine A. Hamann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Victoria W. Tung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Juan M. Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - David D. Brand
- Research Service, Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, TN 38105, USA
| | - Karen A. Hasty
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis VA Medical Center, Memphis, TN, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
- Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - Jonathan M. Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
- Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, 37212, USA
| |
Collapse
|
37
|
Jeyaraman M, Nallakumarasamy A, Jeyaraman N, Ramasubramanian S. Tissue engineering in chondral defect. COMPUTATIONAL BIOLOGY FOR STEM CELL RESEARCH 2024:361-378. [DOI: 10.1016/b978-0-443-13222-3.00033-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
38
|
Gao J, Pei H, Lv F, Niu X, You Y, He L, Hu S, Shah KM, Liu M, Chen Y, Du B, Xiong H, Luo J. JD-312 - A novel small molecule that facilitates cartilage repair and alleviates osteoarthritis progression. J Orthop Translat 2024; 44:60-71. [PMID: 38269355 PMCID: PMC10805627 DOI: 10.1016/j.jot.2023.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/12/2023] [Accepted: 11/21/2023] [Indexed: 01/26/2024] Open
Abstract
Background The chondrogenic differentiation of mesenchymal stem cells (MSCs) to enhance cartilage repair and regeneration is a promising strategy to alleviate osteoarthritis (OA) progression. Method The potency of JD-312 in inducing chondrogenic differentiation of MSCs was assessed and verified. The efficacy of JD-312-treated MSCs was evaluated using a Sprague-Dawley rat DMM model. Additionally, the capacity of JD-312 to successfully recruit bone marrow-derived mesenchymal stem cells (BMSCs) for the treatment of OA in vitro was confirmed via intra-articular injection. The repair status of the articular cartilage was analyzed in vivo through histological examination. Result In this study, we identify JD-312 as a novel non-toxic small molecule that can promote chondrogenic differentiation in human umbilical cord-derived MSCs (hUCMSCs) and human bone marrow MSCS (hBMSCs) in vitro. We also show that transient differentiation of MSCs with JD-312 prior to in vivo administration remarkably improves the regeneration of cartilage and promotes Col2a1 and Acan expression in rat models of DMM, in comparison to kartogenin (KGN) pre-treatment or MSCs alone. Furthermore, direct intra-articular injection of JD-312 in murine model of OA showed reduced loss of articular cartilage and improved pain parameters. Lastly, we identified that the effects of JD-312 are at least in part mediated via upregulation of genes associated with the focal adhesion, PI3K-Akt signaling and the ECM-receptor interaction pathways, and specifically cartilage oligomeric matrix protein (COMP) may play a vital role. Conclusion Our study demonstrated that JD-312 showed encouraging repair effects for OA in vivo. The translational potential of this article Together, our findings demonstrate that JD-312 is a promising new therapeutic molecule for cartilage regeneration with clinical potential.
Collapse
Affiliation(s)
- Jingduo Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Haixiang Pei
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
- Institute for Advanced Study, Shenzhen University and Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, PR China
| | - Fang Lv
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Xin Niu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Yu You
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Liang He
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Shijia Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Karan M. Shah
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield, United Kingdom
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Yihua Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Bing Du
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Hai Xiong
- Institute for Advanced Study, Shenzhen University and Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, PR China
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| |
Collapse
|
39
|
Al Maruf DSA, Xin H, Cheng K, Garcia AG, Mohseni-Dargah M, Ben-Sefer E, Tomaskovic-Crook E, Crook JM, Clark JR. Bioengineered cartilaginous grafts for repairing segmental mandibular defects. J Tissue Eng 2024; 15. [DOI: 10.1177/20417314241267017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Reconstructing critical-sized craniofacial bone defects is a global healthcare challenge. Current methods, like autologous bone transplantation, face limitations. Bone tissue engineering offers an alternative to autologous bone, with traditional approaches focusing on stimulating osteogenesis via the intramembranous ossification (IMO) pathway. However, IMO falls short in addressing larger defects, particularly in clinical scenarios where there is insufficient vascularisation. This review explores redirecting bone regeneration through endochondral ossification (ECO), a process observed in long bone healing stimulated by hypoxic conditions. Despite its promise, gaps exist in applying ECO to bone tissue engineering experiments, requiring the elucidation of key aspects such as cell sources, biomaterials and priming protocols. This review discusses various scaffold biomaterials and cellular sources for chondrogenesis and hypertrophic chondrocyte priming, mirroring the ECO pathway. The review highlights challenges in current endochondral priming and proposes alternative approaches. Emphasis is on segmental mandibular defect repair, offering insights for future research and clinical application. This concise review aims to advance bone tissue engineering by addressing critical gaps in ECO strategies.
Collapse
Affiliation(s)
- D S Abdullah Al Maruf
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Hai Xin
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Kai Cheng
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Camperdown, NSW, Australia
| | - Alejandro Garcia Garcia
- Cell, Tissue and Organ Engineering Laboratory, Biomedical Centre (BMC), Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, Lund, Sweden
| | - Masoud Mohseni-Dargah
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
| | - Eitan Ben-Sefer
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Eva Tomaskovic-Crook
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Sarcoma and Surgical Research Centre, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- ARC Centre of Excellence for Electromaterials Science, The University of Wollongong, Wollongong, NSW, Australia
- Intelligent Polymer Research Institute, AIIM Facility, The University of Wollongong, Wollongong, NSW, Australia
| | - Jeremy Micah Crook
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Sarcoma and Surgical Research Centre, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- ARC Centre of Excellence for Electromaterials Science, The University of Wollongong, Wollongong, NSW, Australia
- Intelligent Polymer Research Institute, AIIM Facility, The University of Wollongong, Wollongong, NSW, Australia
| | - Jonathan Robert Clark
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Camperdown, NSW, Australia
| |
Collapse
|
40
|
Ghosh S, Pati F. Decellularized extracellular matrix and silk fibroin-based hybrid biomaterials: A comprehensive review on fabrication techniques and tissue-specific applications. Int J Biol Macromol 2023; 253:127410. [PMID: 37844823 DOI: 10.1016/j.ijbiomac.2023.127410] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
Biomaterials play a fundamental role in tissue engineering by providing biochemical and physical cues that influence cellular fate and matrix development. Decellularized extracellular matrix (dECM) as a biomaterial is distinguished by its abundant composition of matrix proteins, such as collagen, elastin, fibronectin, and laminin, as well as glycosaminoglycans and proteoglycans. However, the mechanical properties of only dECM-based constructs may not always meet tissue-specific requirements. Recent advancements address this challenge by utilizing hybrid biomaterials that harness the strengths of silk fibroin (SF), which contributes the necessary mechanical properties, while dECM provides essential cellular cues for in vitro studies and tissue regeneration. This review discusses emerging trends in developing such biopolymer blends, aiming to synergistically combine the advantages of SF and dECM through optimal concentrations and desired cross-linking density. We focus on different fabrication techniques and cross-linking methods that have been utilized to fabricate various tissue-engineered hybrid constructs. Furthermore, we survey recent applications of such biomaterials for the regeneration of various tissues, including bone, cartilage, trachea, bladder, vascular graft, heart, skin, liver, and other soft tissues. Finally, the trajectory and prospects of the constructs derived from this blend in the tissue engineering field have been summarized, highlighting their potential for clinical translation.
Collapse
Affiliation(s)
- Soham Ghosh
- BioFab Lab, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502285, Telangana, India
| | - Falguni Pati
- BioFab Lab, Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502285, Telangana, India.
| |
Collapse
|
41
|
Chen Y, Cheng RJ, Wu Y, Huang D, Li Y, Liu Y. Advances in Stem Cell-Based Therapies in the Treatment of Osteoarthritis. Int J Mol Sci 2023; 25:394. [PMID: 38203565 PMCID: PMC10779279 DOI: 10.3390/ijms25010394] [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: 11/19/2023] [Revised: 12/16/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Osteoarthritis (OA) is a chronic, degenerative joint disease presenting a significant global health threat. While current therapeutic approaches primarily target symptom relief, their efficacy in repairing joint damage remains limited. Recent research has highlighted mesenchymal stem cells (MSCs) as potential contributors to cartilage repair, anti-inflammatory modulation, and immune regulation in OA patients. Notably, MSCs from different sources and their derivatives exhibit variations in their effectiveness in treating OA. Moreover, pretreatment and gene editing techniques of MSCs can enhance their therapeutic outcomes in OA. Additionally, the combination of novel biomaterials with MSCs has shown promise in facilitating the repair of damaged cartilage. This review summarizes recent studies on the role of MSCs in the treatment of OA, delving into their advantages and exploring potential directions for development, with the aim of providing fresh insights for future research in this critical field.
Collapse
Affiliation(s)
- Ye Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| | - Rui-Juan Cheng
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| | - Yinlan Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| | - Deying Huang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| | - Yanhong Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (R.-J.C.); (Y.W.); (D.H.)
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Chengdu 610041, China
| |
Collapse
|
42
|
Franco RAG, McKenna E, Shajib MS, Guillesser B, Robey PG, Crawford RW, Doran MR, Futrega K. Microtissue Culture Provides Clarity on the Relative Chondrogenic and Hypertrophic Response of Bone-Marrow-Derived Stromal Cells to TGF-β1, BMP-2, and GDF-5. Cells 2023; 13:37. [PMID: 38201241 PMCID: PMC10778331 DOI: 10.3390/cells13010037] [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: 09/20/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 01/12/2024] Open
Abstract
Chondrogenic induction of bone-marrow-derived stromal cells (BMSCs) is typically accomplished with medium supplemented with growth factors (GF) from the transforming growth factor-beta (TGF-β)/bone morphogenetic factor (BMP) superfamily. In a previous study, we demonstrated that brief (1-3 days) stimulation with TGF-β1 was sufficient to drive chondrogenesis and hypertrophy using small-diameter microtissues generated from 5000 BMSC each. This biology is obfuscated in typical large-diameter pellet cultures, which suffer radial heterogeneity. Here, we investigated if brief stimulation (2 days) of BMSC microtissues with BMP-2 (100 ng/mL) or growth/differentiation factor (GDF-5, 100 ng/mL) was also sufficient to induce chondrogenic differentiation, in a manner comparable to TGF-β1 (10 ng/mL). Like TGF-β1, BMP-2 and GDF-5 are reported to stimulate chondrogenic differentiation of BMSCs, but the effects of transient or brief use in culture have not been explored. Hypertrophy is an unwanted outcome in BMSC chondrogenic differentiation that renders engineered tissues unsuitable for use in clinical cartilage repair. Using three BMSC donors, we observed that all GFs facilitated chondrogenesis, although the efficiency and the necessary duration of stimulation differed. Microtissues treated with 2 days or 14 days of TGF-β1 were both superior at producing extracellular matrix and expression of chondrogenic gene markers compared to BMP-2 and GDF-5 with the same exposure times. Hypertrophic markers increased proportionally with chondrogenic differentiation, suggesting that these processes are intertwined for all three GFs. The rapid action, or "temporal potency", of these GFs to induce BMSC chondrogenesis was found to be as follows: TGF-β1 > BMP-2 > GDF-5. Whether briefly or continuously supplied in culture, TGF-β1 was the most potent GF for inducing chondrogenesis in BMSCs.
Collapse
Affiliation(s)
- Rose Ann G. Franco
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
| | - Eamonn McKenna
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
| | - Md. Shafiullah Shajib
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Bianca Guillesser
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Pamela G. Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Ross W. Crawford
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Michael R. Doran
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD 20892, USA
- Mater Research Institute—University of Queensland (UQ), Translational Research Institute (TRI), Brisbane, QLD 4102, Australia
| | - Kathryn Futrega
- Centre for Biomedical Technologies (CBT), School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Department of Health and Human Services, Bethesda, MD 20892, USA
| |
Collapse
|
43
|
Yin H, Mao K, Huang Y, Guo A, Shi L. Tendon stem/progenitor cells are promising reparative cell sources for multiple musculoskeletal injuries of concomitant articular cartilage lesions associated with ligament injuries. J Orthop Surg Res 2023; 18:869. [PMID: 37968672 PMCID: PMC10647040 DOI: 10.1186/s13018-023-04313-3] [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: 07/16/2023] [Accepted: 10/23/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Trauma-related articular cartilage lesions usually occur in conjunction with ligament injuries. Torn ligaments are frequently reconstructed with tendon autograft and has been proven to achieve satisfactory clinical outcomes. However, treatments for the concomitant articular cartilage lesions are still very insufficient. The current study was aimed to evaluate whether stem cells derived from tendon tissue can be considered as an alternative reparative cell source for cartilage repair. METHODS Primary human tendon stem/progenitor cells (hTSPCs) were isolated from 4 male patients (32 ± 8 years) who underwent ACL reconstruction surgery with autologous semitendinosus and gracilis tendons. The excessive tendon tissue after graft preparation was processed for primary cell isolation with an enzyme digestion protocol. Decellularization cartilage matrix (DCM) was used to provide a chondrogenic microenvironment for hTSPCs. Cell viability, cell morphology on the DCM, as well as their chondrogenic differentiation were evaluated. RESULTS DAPI staining and DNA quantitative analysis (61.47 μg per mg dry weight before and 2.64 μg/mg after decellularization) showed that most of the cells in the cartilage lacuna were removed after decellularization process. Whilst, the basic structure of the cartilage tissue was preserved and the main ECM components, collagen type II and sGAG were retained after decellularization, which were revealed by DMMB assay and histology. Live/dead staining and proliferative assay demonstrated that DCM supported attachment, survival and proliferation of hTSPCs with an excellent biocompatibility. Furthermore, gene expression analysis indicated that chondrogenic differentiation of hTSPC was induced by the DCM microenvironment, with upregulation of chondrogenesis-related marker genes, COL 2 and SOX9, without the use of exogenous growth factors. CONCLUSION DCM supported hTSPCs attachment and proliferation with high biocompatibility. Moreover, TSPCs underwent a distinct chondrogenesis after the induction of a chondrogenic microenvironment provided by DCM. These results indicated that TSPCs are promising reparative cell sources for promoting cartilage repair. Particularly, in the cohort that articular cartilage lesions occur in conjunction with ligament injuries, autologous TSPCs can be isolated from a portion of the tendon autograph harvested for ligaments reconstruction. In future clinical practice, combined ligament reconstruction with TSPCs- based therapy for articular cartilage repair can to be considered to achieve superior repair of these associated injuries, in which autologous TSPCs can be isolated from a portion of the tendon autograph harvested for ligaments reconstruction.
Collapse
Affiliation(s)
- Heyong Yin
- Department of Orthopaedics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China
| | - Kelei Mao
- Department of Orthopaedics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China
| | - Yufu Huang
- Department of Orthopaedics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China
| | - Ai Guo
- Department of Orthopaedics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China.
| | - Lin Shi
- Department of Orthopaedics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
44
|
Vafa E, Tayebi L, Abbasi M, Azizli MJ, Bazargan-Lari R, Talaiekhozani A, Zareshahrabadi Z, Vaez A, Amani AM, Kamyab H, Chelliapan S. A better roadmap for designing novel bioactive glasses: effective approaches for the development of innovative revolutionary bioglasses for future biomedical applications. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:116960-116983. [PMID: 36456674 DOI: 10.1007/s11356-022-24176-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 11/08/2022] [Indexed: 06/17/2023]
Abstract
The introduction of bioactive glasses (BGs) precipitated a paradigm shift in the medical industry and opened the path for the development of contemporary regenerative medicine driven by biomaterials. This composition can bond to live bone and can induce osteogenesis by the release of physiologically active ions. 45S5 BG products have been transplanted effectively into millions of patients around the world, primarily to repair bone and dental defects. Over the years, many other BG compositions have been introduced as innovative biomaterials for repairing soft tissue and delivering drugs. When research first started, many of the accomplishments that have been made today were unimaginable. It appears that the true capacity of BGs has not yet been realized. Because of this, research involving BGs is extremely fascinating. However, to be successful, it requires interdisciplinary cooperation between physicians, glass chemists, and bioengineers. The present paper gives a picture of the existing clinical uses of BGs and illustrates key difficulties deserving to be faced in the future. The challenges range from the potential for BGs to be used in a wide variety of applications. We have high hopes that this paper will be of use to both novice researchers, who are just beginning their journey into the world of BGs, as well as seasoned scientists, in that it will promote conversation regarding potential additional investigation and lead to the discovery of innovative medical applications for BGs.
Collapse
Affiliation(s)
- Ehsan Vafa
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, USA
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Azizli
- Department of Chemistry and Chemical Engineering, Islamic Azad University, Rasht, Rasht Branch, Iran
| | - Reza Bazargan-Lari
- Department of Materials Science and Engineering, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Amirreza Talaiekhozani
- Department of Civil Engineering, Jami Institute of Technology, Isfahan, Iran
- Alavi Educational and Cultural Complex, Shiraz, Iran
| | - Zahra Zareshahrabadi
- Basic Sciences in Infectious Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Mohamad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hesam Kamyab
- Malaysia-Japan International Institute of Technology, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia
- Department of Biomaterials, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai 600077, India, Chennai, India
| | - Shreeshivadasan Chelliapan
- Engineering Department, Razak Faculty of Technology & Informatics, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia
| |
Collapse
|
45
|
Kováč J, Priščáková P, Gbelcová H, Heydari A, Žiaran S. Bioadhesive and Injectable Hydrogels and Their Correlation with Mesenchymal Stem Cells Differentiation for Cartilage Repair: A Mini-Review. Polymers (Basel) 2023; 15:4228. [PMID: 37959908 PMCID: PMC10648146 DOI: 10.3390/polym15214228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/06/2023] [Accepted: 10/17/2023] [Indexed: 11/15/2023] Open
Abstract
Injectable bioadhesive hydrogels, known for their capacity to carry substances and adaptability in processing, offer great potential across various biomedical applications. They are especially promising in minimally invasive stem cell-based therapies for treating cartilage damage. This approach harnesses readily available mesenchymal stem cells (MSCs) to differentiate into chondrocytes for cartilage regeneration. In this review, we investigate the relationship between bioadhesion and MSC differentiation. We summarize the fundamental principles of bioadhesion and discuss recent trends in bioadhesive hydrogels. Furthermore, we highlight their specific applications in conjunction with stem cells, particularly in the context of cartilage repair. The review also encompasses a discussion on testing methods for bioadhesive hydrogels and direct techniques for differentiating MSCs into hyaline cartilage chondrocytes. These approaches are explored within both clinical and laboratory settings, including the use of genetic tools. While this review offers valuable insights into the interconnected aspects of these topics, it underscores the need for further research to fully grasp the complexities of their relationship.
Collapse
Affiliation(s)
- Ján Kováč
- Medical Vision, Záhradnícka 55, 821 08 Bratislava, Slovakia; (J.K.); (P.P.); (H.G.); (A.H.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Petra Priščáková
- Medical Vision, Záhradnícka 55, 821 08 Bratislava, Slovakia; (J.K.); (P.P.); (H.G.); (A.H.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Helena Gbelcová
- Medical Vision, Záhradnícka 55, 821 08 Bratislava, Slovakia; (J.K.); (P.P.); (H.G.); (A.H.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Abolfazl Heydari
- Medical Vision, Záhradnícka 55, 821 08 Bratislava, Slovakia; (J.K.); (P.P.); (H.G.); (A.H.)
- Polymer Institute of the Slovak Academy of Sciences, Dúbravská Cesta 9, 845 41 Bratislava, Slovakia
| | - Stanislav Žiaran
- Medical Vision, Záhradnícka 55, 821 08 Bratislava, Slovakia; (J.K.); (P.P.); (H.G.); (A.H.)
- Department of Urology, Faculty of Medicine, Comenius University, Limbová 5, 833 05 Bratislava, Slovakia
| |
Collapse
|
46
|
Gherghel R, Macovei LA, Burlui MA, Cardoneanu A, Rezus II, Mihai IR, Rezus E. Osteoarthritis—The Role of Mesenchymal Stem Cells in Cartilage Regeneration. APPLIED SCIENCES 2023; 13:10617. [DOI: 10.3390/app131910617] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Osteoarthritis (OA) is a condition that can cause substantial pain, loss of joint function, and a decline in quality of life in patients. Numerous risk factors, including aging, genetics, and injury, have a role in the onset of OA, characterized by structural changes within the joints. Most therapeutic approaches focus on the symptoms and try to change or improve the structure of the joint tissues. Even so, no treatments have been able to stop or slow the progression of OA or give effective and long-lasting relief of symptoms. In the absence of disease-modifying drugs, regenerative medicine is being investigated as a possible treatment that can change the course of OA by changing the structure of damaged articular cartilage. In regenerative therapy for OA, mesenchymal stem cells (MSCs) have been the mainstay of translational investigations and clinical applications. In recent years, MSCs have been discovered to be an appropriate cell source for treating OA due to their ability to expand rapidly in culture, their nontumorigenic nature, and their ease of collection. MSCs’ anti-inflammatory and immunomodulatory capabilities may provide a more favorable local environment for the regeneration of injured articular cartilage, which was thought to be one of the reasons why they were seen as more suited for OA. In addition to bone marrow, MSCs have also been isolated from adipose tissue, synovium, umbilical cord, cord blood, dental pulp, placenta, periosteum, and skeletal muscle. Adipose tissue and bone marrow are two of the most essential tissues for therapeutic MSCs. Positive preclinical and clinical trial results have shown that, despite current limitations and risks, MSC-based therapy is becoming a promising approach to regenerative medicine in treating OA.
Collapse
Affiliation(s)
- Robert Gherghel
- Department of Orthopedics and Trauma Surgery, Piatra Neamt Emergency Hospital, 700115 Piatra Neamt, Romania
| | - Luana Andreea Macovei
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Maria-Alexandra Burlui
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Anca Cardoneanu
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Ioana-Irina Rezus
- Department of Dermatology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Ioana Ruxandra Mihai
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| |
Collapse
|
47
|
Xu X, Xu L, Xia J, Wen C, Liang Y, Zhang Y. Harnessing knee joint resident mesenchymal stem cells in cartilage tissue engineering. Acta Biomater 2023; 168:372-387. [PMID: 37481194 DOI: 10.1016/j.actbio.2023.07.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/26/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
Osteoarthritis (OA) is a widespread clinical disease characterized by cartilage degeneration in middle-aged and elderly people. Currently, there is no effective treatment for OA apart from total joint replacement in advanced stages. Mesenchymal stem cells (MSCs) are a type of adult stem cell with diverse differentiation capabilities and immunomodulatory potentials. MSCs are known to effectively regulate the cartilage microenvironment, promote cartilage regeneration, and alleviate OA symptoms. As a result, they are promising sources of cells for OA therapy. Recent studies have revealed the presence of resident MSCs in synovial fluid, synovial membrane, and articular cartilage, which can be collected as knee joint-derived MSCs (KJD-MSC). Several preclinical and clinical studies have demonstrated that KJD-MSCs have great potential for OA treatment, whether applied alone, in combination with biomaterials, or as exocrine MSCs. In this article, we will review the characteristics of MSCs in the joints, including their cytological characteristics, such as proliferation, cartilage differentiation, and immunomodulatory abilities, as well as the biological function of MSC exosomes. We will also discuss the use of tissue engineering in OA treatment and introduce the concept of a new generation of stem cell-based tissue engineering therapy, including the use of engineering, gene therapy, and gene editing techniques to create KJD-MSCs or KJD-MSC derivative exosomes with improved functionality and targeted delivery. These advances aim to maximize the efficiency of cartilage tissue engineering and provide new strategies to overcome the bottleneck of OA therapy. STATEMENT OF SIGNIFICANCE: This research will provide new insights into the medicinal benefit of Joint resident Mesenchymal Stem Cells (MSCs), specifically on its cartilage tissue engineering ability. Through this review, the community will further realize promoting joint resident mesenchymal stem cells, especially cartilage progenitor/MSC-like progenitor cells (CPSC), as a preventive measure against osteoarthritis and cartilage injury. People and medical institutions may also consider cartilage derived MSC as an alternative approach against cartilage degeneration. Moreover, the discussion presented in this study will convey valuable information for future research that will explore the medicinal benefits of cartilage derived MSC.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Orthopedics, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Limei Xu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Jiang Xia
- Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Caining Wen
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Yujie Liang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| | - Yuanmin Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| |
Collapse
|
48
|
Wang Z. Assessing Tumorigenicity in Stem Cell-Derived Therapeutic Products: A Critical Step in Safeguarding Regenerative Medicine. Bioengineering (Basel) 2023; 10:857. [PMID: 37508884 PMCID: PMC10376867 DOI: 10.3390/bioengineering10070857] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/08/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Stem cells hold promise in regenerative medicine due to their ability to proliferate and differentiate into various cell types. However, their self-renewal and multipotency also raise concerns about their tumorigenicity during and post-therapy. Indeed, multiple studies have reported the presence of stem cell-derived tumors in animal models and clinical administrations. Therefore, the assessment of tumorigenicity is crucial in evaluating the safety of stem cell-derived therapeutic products. Ideally, the assessment needs to be performed rapidly, sensitively, cost-effectively, and scalable. This article reviews various approaches for assessing tumorigenicity, including animal models, soft agar culture, PCR, flow cytometry, and microfluidics. Each method has its advantages and limitations. The selection of the assay depends on the specific needs of the study and the stage of development of the stem cell-derived therapeutic product. Combining multiple assays may provide a more comprehensive evaluation of tumorigenicity. Future developments should focus on the optimization and standardization of microfluidics-based methods, as well as the integration of multiple assays into a single platform for efficient and comprehensive evaluation of tumorigenicity.
Collapse
Affiliation(s)
- Zongjie Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Chan Zuckerberg Biohub Chicago, Chicago, IL 60607, USA
| |
Collapse
|
49
|
Woodbury SM, Swanson WB, Mishina Y. Mechanobiology-informed biomaterial and tissue engineering strategies for influencing skeletal stem and progenitor cell fate. Front Physiol 2023; 14:1220555. [PMID: 37520820 PMCID: PMC10373313 DOI: 10.3389/fphys.2023.1220555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/05/2023] [Indexed: 08/01/2023] Open
Abstract
Skeletal stem and progenitor cells (SSPCs) are the multi-potent, self-renewing cell lineages that form the hematopoietic environment and adventitial structures of the skeletal tissues. Skeletal tissues are responsible for a diverse range of physiological functions because of the extensive differentiation potential of SSPCs. The differentiation fates of SSPCs are shaped by the physical properties of their surrounding microenvironment and the mechanical loading forces exerted on them within the skeletal system. In this context, the present review first highlights important biomolecules involved with the mechanobiology of how SSPCs sense and transduce these physical signals. The review then shifts focus towards how the static and dynamic physical properties of microenvironments direct the biological fates of SSPCs, specifically within biomaterial and tissue engineering systems. Biomaterial constructs possess designable, quantifiable physical properties that enable the growth of cells in controlled physical environments both in-vitro and in-vivo. The utilization of biomaterials in tissue engineering systems provides a valuable platform for controllably directing the fates of SSPCs with physical signals as a tool for mechanobiology investigations and as a template for guiding skeletal tissue regeneration. It is paramount to study this mechanobiology and account for these mechanics-mediated behaviors to develop next-generation tissue engineering therapies that synergistically combine physical and chemical signals to direct cell fate. Ultimately, taking advantage of the evolved mechanobiology of SSPCs with customizable biomaterial constructs presents a powerful method to predictably guide bone and skeletal organ regeneration.
Collapse
Affiliation(s)
- Seth M. Woodbury
- Yuji Mishina Laboratory, University of Michigan School of Dentistry, Department of Biologic and Materials Science & Prosthodontics, Ann Arbor, MI, United States
- University of Michigan College of Literature, Science, and Arts, Department of Chemistry, Ann Arbor, MI, United States
- University of Michigan College of Literature, Science, and Arts, Department of Physics, Ann Arbor, MI, United States
| | - W. Benton Swanson
- Yuji Mishina Laboratory, University of Michigan School of Dentistry, Department of Biologic and Materials Science & Prosthodontics, Ann Arbor, MI, United States
| | - Yuji Mishina
- Yuji Mishina Laboratory, University of Michigan School of Dentistry, Department of Biologic and Materials Science & Prosthodontics, Ann Arbor, MI, United States
| |
Collapse
|
50
|
Kouroupis D, Kaplan LD, Huard J, Best TM. CD10-Bound Human Mesenchymal Stem/Stromal Cell-Derived Small Extracellular Vesicles Possess Immunomodulatory Cargo and Maintain Cartilage Homeostasis under Inflammatory Conditions. Cells 2023; 12:1824. [PMID: 37508489 PMCID: PMC10377825 DOI: 10.3390/cells12141824] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/23/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
The onset and progression of human inflammatory joint diseases are strongly associated with the activation of resident synovium/infrapatellar fat pad (IFP) pro-inflammatory and pain-transmitting signaling. We recently reported that intra-articularly injected IFP-derived mesenchymal stem/stromal cells (IFP-MSC) acquire a potent immunomodulatory phenotype and actively degrade substance P (SP) via neutral endopeptidase CD10 (neprilysin). Our hypothesis is that IFP-MSC robust immunomodulatory therapeutic effects are largely exerted via their CD10-bound small extracellular vesicles (IFP-MSC sEVs) by attenuating synoviocyte pro-inflammatory activation and articular cartilage degradation. Herein, IFP-MSC sEVs were isolated from CD10High- and CD10Low-expressing IFP-MSC cultures and their sEV miRNA cargo was assessed using multiplex methods. Functionally, we interrogated the effect of CD10High and CD10Low sEVs on stimulated by inflammatory/fibrotic cues synoviocyte monocultures and cocultures with IFP-MSC-derived chondropellets. Finally, CD10High sEVs were tested in vivo for their therapeutic capacity in an animal model of acute synovitis/fat pad fibrosis. Our results showed that CD10High and CD10Low sEVs possess distinct miRNA profiles. Reactome analysis of miRNAs highly present in sEVs showed their involvement in the regulation of six gene groups, particularly those involving the immune system. Stimulated synoviocytes exposed to IFP-MSC sEVs demonstrated significantly reduced proliferation and altered inflammation-related molecular profiles compared to control stimulated synoviocytes. Importantly, CD10High sEV treatment of stimulated chondropellets/synoviocyte cocultures indicated significant chondroprotective effects. Therapeutically, CD10High sEV treatment resulted in robust chondroprotective effects by retaining articular cartilage structure/composition and PRG4 (lubricin)-expressing cartilage cells in the animal model of acute synovitis/IFP fibrosis. Our study suggests that CD10High sEVs possess immunomodulatory miRNA attributes with strong chondroprotective/anabolic effects for articular cartilage in vivo. The results could serve as a foundation for sEV-based therapeutics for the resolution of detrimental aspects of immune-mediated inflammatory joint changes associated with conditions such as osteoarthritis (OA).
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA (T.M.B.)
- Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lee D. Kaplan
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA (T.M.B.)
| | - Johnny Huard
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA;
| | - Thomas M. Best
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA (T.M.B.)
| |
Collapse
|