1
|
Tueni N, Amirouche F. Branding a New Technological Outlook for Future Orthopaedics. Bioengineering (Basel) 2025; 12:494. [PMID: 40428113 PMCID: PMC12109234 DOI: 10.3390/bioengineering12050494] [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: 01/15/2025] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 05/29/2025] Open
Abstract
Orthopedics is undergoing a transformative shift driven by personalized medical technologies that enhance precision, efficiency, and patient outcomes. Virtual surgical planning, robotic assistance, and real-time 3D navigation have revolutionized procedures like total knee arthroplasty and hip replacement, offering unparalleled accuracy and reducing recovery times. Integrating artificial intelligence, advanced imaging, and 3D-printed patient-specific implants further elevates surgical precision, minimizes intraoperative complications, and supports individualized care. In sports orthopedics, wearable sensors and motion analysis technologies are revolutionizing diagnostics, injury prevention, and rehabilitation, enabling real-time decision-making and improved patient safety. Health-tracking devices are advancing recovery and supporting preventative care, transforming athletic performance management. Concurrently, breakthroughs in biologics, biomaterials, and bioprinting are reshaping treatments for cartilage defects, ligament injuries, osteoporosis, and meniscal damage. These innovations are poised to establish new benchmarks for regenerative medicine in orthopedics. By combining cutting-edge technologies with interdisciplinary collaboration, the field is redefining surgical standards, optimizing patient care, and paving the way for a highly personalized and efficient future.
Collapse
Affiliation(s)
- Nicole Tueni
- Institute of Continuum Mechanics and Biomechanics, Friedrich-Alexander-Universität Erlangen-Nuremberg, 91054 Erlangen, Germany;
| | - Farid Amirouche
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Orthopaedic Surgery, Northshore University HealthSystem, Skokie, IL 60076, USA
| |
Collapse
|
2
|
Widodo W, Aprilya D, Satria O. Regenerative Medicine: A New Horizon in Peripheral Nerve Injury and Repair. Orthop Rev (Pavia) 2025; 17:133572. [PMID: 40176924 PMCID: PMC11964392 DOI: 10.52965/001c.133572] [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: 12/08/2024] [Accepted: 12/21/2024] [Indexed: 04/05/2025] Open
Abstract
A peripheral nerve injury is a great burden for the patient and a challenge for the clinician. In a complete injury (axonotmesis or neurotmesis), the slow nature of nerve regeneration after repair or reconstruction hardly catches up to the target organ's degeneration rate, leading to a poor prognosis. The current advance in regenerative medicine has shown the potency of stem cells and their products for healing many human body structures, including the nerve. A comprehensive literature search was conducted using an internet-based search engine for current advances in regenerative medicine to augment peripheral nerve repair or reconstruction. Stem cells can differentiate into nerve cells and have paracrine and immunomodulatory effects. Its products, such as the secretome and exosome, have also been studied, and they have many benefits for the regeneration process. This novel treatment possesses significant potential to accelerate nerve healing after nerve reconstruction and potentially postpone the degenerative process in the target organ, allowing it to respond to the new signal once nerve regeneration is complete. The aim of this article is to summarized the application of stem cells and its products for nerve healing.
Collapse
Affiliation(s)
- Wahyu Widodo
- Hand and Microsurgery Division, Orthopedic and Traumatology Department, Fatmawati General Hospital, Jakarta, Indonesia
| | - Dina Aprilya
- Hand and Microsurgery Division, Orthopedic and Traumatology Department, Fatmawati General Hospital, Jakarta, Indonesia
| | - Oryza Satria
- Hand and Microsurgery Division, Orthopedic and Traumatology Department, Fatmawati General Hospital, Jakarta, Indonesia
| |
Collapse
|
3
|
Anaya-Sampayo LM, Roa NS, Martínez-Cardozo C, García-Robayo DA, Rodríguez-Lorenzo LM. Influence of Hydroxyapatite and Gelatin Content on Crosslinking Dynamics and HDFn Cell Viability in Alginate Bioinks for 3D Bioprinting. Polymers (Basel) 2024; 16:3224. [PMID: 39599315 PMCID: PMC11598013 DOI: 10.3390/polym16223224] [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: 10/18/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
This study investigates how varying concentrations of hydroxyapatite (OHAp) and the addition of gelatin influence the ionic crosslinking time of alginate-based bioinks, as well as the shear stress experienced by neonatal human dermal fibroblasts (HDFn) during extrusion. These factors are crucial for validating bioinks and developing viable 3D bioprinted models. Four bioink formulations were created with a 50/50 ratio of alginate to gelatin, incorporating different calcium phosphate concentrations (0%, 1%, 5%, and 10%). The bioink compositions were confirmed via Fourier Transform Infrared (FT-IR) spectroscopy, and rheological analyses evaluated their pseudoplastic behavior, printability limits, and crosslinking times. The results indicated a notable increase in the consistency index (k) from 0.32 for the 0% OHAp formulation to 0.48 for the 10% OHAp formulation, suggesting improved viscoelastic properties. The elastic modulus recovery after crosslinking rose significantly from 245 Pa to 455 Pa. HDFn experienced a shear stress of up to 1.5436 Pa at the tip during extrusion with the HDFn-ALG5-GEL5-OHAp10 bioinks, calculated at a shear rate as low as 2 s-1. Viability assays confirmed over 70% cell viability 24 h post-extrusion and 92% viability after 7 days for the 10% OHAp formulation, highlighting the potential of hydroxyapatite-enhanced bioinks in tissue engineering applications.
Collapse
Affiliation(s)
- Lina Maria Anaya-Sampayo
- Centro de Investigaciones Odontológicas, Facultad de Odontología, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (L.M.A.-S.); (N.S.R.)
| | - Nelly S. Roa
- Centro de Investigaciones Odontológicas, Facultad de Odontología, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (L.M.A.-S.); (N.S.R.)
| | | | - Dabeiba Adriana García-Robayo
- Centro de Investigaciones Odontológicas, Facultad de Odontología, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (L.M.A.-S.); (N.S.R.)
| | - Luis M. Rodríguez-Lorenzo
- Department of Polymeric Nanomaterials and Biomaterials, Institute Science and Technology of Polymers (ICTP-CSIC), 28006 Madrid, Spain
| |
Collapse
|
4
|
Pal D, Das P, Roy S, Mukherjee P, Halder S, Ghosh D, Nandi SK. Recent trends of stem cell therapies in the management of orthopedic surgical challenges. Int J Surg 2024; 110:6330-6344. [PMID: 38716973 PMCID: PMC11487011 DOI: 10.1097/js9.0000000000001524] [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: 12/29/2023] [Accepted: 04/14/2024] [Indexed: 10/20/2024]
Abstract
Emerged health-related problems especially with increasing population and with the wider occurrence of these issues have always put the utmost concern and led medicine to outgrow its usual mode of treatment, to achieve better outcomes. Orthopedic interventions are one of the most concerning hitches, requiring advancement in several issues, that show complications with conventional approaches. Advanced studies have been undertaken to address the issue, among which stem cell therapy emerged as a better area of growth. The capacity of the stem cells to renovate themselves and adapt into different cell types made it possible to implement its use as a regenerative slant. Harvesting the stem cells, particularly mesenchymal stem cells (MSCs) is easier and can be further grown in vitro . In this review, we have discussed orthopedic-related issues including bone defects and fractures, nonunions, ligament and tendon injuries, degenerative changes, and associated conditions, which require further approaches to execute better outcomes, and the advanced strategies that can be tagged along with various ways of application of MSCs. It aims to objectify the idea of stem cells, with a major focus on the application of MSCs from different sources in various orthopedic interventions. It also discusses the limitations, and future scopes for further approaches in the field of regenerative medicine. The involvement of MSCs may transition the procedures in orthopedic interventions from predominantly surgical substitution and reconstruction to bio-regeneration and prevention. Nevertheless, additional improvements and evaluations are required to explore the effectiveness and safety of mesenchymal stem cell treatment in orthopedic regenerative medicine.
Collapse
Affiliation(s)
| | - Pratik Das
- Department of Veterinary Surgery and Radiology
| | - Subhasis Roy
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal
| | - Prasenjit Mukherjee
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal
| | | | | | | |
Collapse
|
5
|
Subramanyam K, Poornima S, Kumar S, Hasan Q. Short-Term Clinical Results of Single-Injection Autologous Bone Marrow Aspirate Concentrate (BMAC) as a Therapeutic Option/Tool in Knee Osteoarthritis. BIOLOGICS 2024; 4:218-231. [DOI: 10.3390/biologics4020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Purpose: Knee osteoarthritis (KOA) is a very common cartilage disorder affecting millions of people globally and is characterized by pain, stiffness, swelling, loss of articular cartilage, and osteophyte formation, resulting in disability. The presently available treatments for KOA are palliative. Hence, there is a need to explore a non-surgical treatment portfolio. Bone marrow aspirate concentrate (BMAC) is one of the predominant attention-drawing managements/treatments for KOA in recent times due to its potential advantages of disease-modifying and regeneration capacities. Principle: This study aimed to evaluate the role of single-injection autologous BMAC as a therapeutic option in the treatment of KOA and evaluate the functional and clinical outcomes of KOA patients. In this study, 132 patients with KOA (Kellgren and Lawrence (KL) grade II and III) were included as per the inclusion criteria. Autologous bone marrow was aspirated and separated, and concentrated bone marrow aspirate was administered into the knee joint of the affected individual. Results: At the end of the 12th month (end of the follow-up period), 95% of patients showed complete pain relief and improvement in joint function, which shows that the results were promising and encouraging. Unpaired t-test results also indicated that the two-tailed p-value is less than 0.0001, and the difference is extremely statistically significant. No adverse effects were observed in the study patients. Conclusions: BMAC therapy has potential, with satisfactory, efficient, and durable results in KL grades II and III in KOA patients. This can be a safe alternative therapy in the treatment of KOA, especially in the early grades of OA. In summary, to the best of our knowledge, this is the first study from India that evaluated BMAC efficacy both subjectively and objectively in KOA (KL-II and KL-III) patients.
Collapse
Affiliation(s)
- Krishna Subramanyam
- Department of Orthopaedics, Kamineni Hospitals, LB Nagar, Hyderabad 500074, India
- Department of Orthopaedics, Yashoda Hospitals, Malakpet, Hyderabad 500036, India
| | - Subhadra Poornima
- Department of Genetics and Molecular Medicine, Kamineni Academy of Medical Sciences and Research Centre, LB. Nagar, Hyderabad 500074, India
- Department of Genetics and Molecular Medicine, Kamineni Life Sciences, Moula Ali, Hyderabad 500047, India
| | - Satish Kumar
- Department of Orthopaedics, Yashoda Hospitals, Malakpet, Hyderabad 500036, India
| | - Qurratulain Hasan
- Department of Genetics and Molecular Medicine, Kamineni Academy of Medical Sciences and Research Centre, LB. Nagar, Hyderabad 500074, India
| |
Collapse
|
6
|
Taguchi T, Lopez M, Takawira C. Viable tendon neotissue from adult adipose-derived multipotent stromal cells. Front Bioeng Biotechnol 2024; 11:1290693. [PMID: 38260742 PMCID: PMC10800559 DOI: 10.3389/fbioe.2023.1290693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Background: Tendon healing is frequently prolonged, unpredictable, and results in poor tissue quality. Neotissue formed by adult multipotent stromal cells has the potential to guide healthy tendon tissue formation. Objectives: The objective of this study was to characterize tendon neotissue generated by equine adult adipose-derived multipotent stromal cells (ASCs) on collagen type I (COLI) templates under 10% strain in a novel bioreactor. The tested hypothesis was that ASCs assume a tendon progenitor cell-like morphology, express tendon-related genes, and produce more organized extracellular matrix (ECM) in tenogenic versus stromal medium with perfusion and centrifugal fluid motion. Methods: Equine ASCs on COLI sponge cylinders were cultured in stromal or tenogenic medium within bioreactors during combined perfusion and centrifugal fluid motion for 7, 14, or 21 days under 10% strain. Viable cell distribution and number, tendon-related gene expression, and micro- and ultra-structure were evaluated with calcein-AM/EthD-1 staining, resazurin reduction, RT-PCR, and light, transmission, and scanning electron microscopy. Fibromodulin was localized with immunohistochemistry. Cell number and gene expression were compared between culture media and among culture periods (p < 0.05). Results: Viable cells were distributed throughout constructs for up to 21 days of culture, and cell numbers were higher in tenogenic medium. Individual cells had a round or rhomboid shape with scant ECM in stromal medium in contrast to clusters of parallel, elongated cells surrounded by highly organized ECM in tenogenic medium after 21 days of culture. Transcription factor, extracellular matrix, and mature tendon gene expression profiles confirmed ASC differentiation to a tendon progenitor-like cell in tenogenic medium. Construct micro- and ultra-structure were consistent with tendon neotissue and fibromodulin was present in the ECM after culture in tenogenic medium. Conclusion: Long-term culture in custom bioreactors with combined perfusion and centrifugal tenogenic medium circulation supports differentiation of equine adult ASCs into tendon progenitor-like cells capable of neotissue formation.
Collapse
|
7
|
Cabrera-Pérez R, Ràfols-Mitjans A, Roig-Molina Á, Beltramone S, Vives J, Batlle-Morera L. Human Wharton's jelly-derived mesenchymal stromal cells promote bone formation in immunodeficient mice when administered into a bone microenvironment. J Transl Med 2023; 21:802. [PMID: 37950242 PMCID: PMC10638709 DOI: 10.1186/s12967-023-04672-9] [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: 08/02/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Wharton's Jelly (WJ) Mesenchymal Stromal Cells (MSC) have emerged as an attractive allogeneic therapy for a number of indications, except for bone-related conditions requiring new tissue formation. This may be explained by the apparent recalcitrance of MSC,WJ to differentiate into the osteogenic lineage in vitro, as opposed to permissive bone marrow (BM)-derived MSCs (MSC,BM) that readily commit to bone cells. Consequently, the actual osteogenic in vivo capacity of MSC,WJ is under discussion. METHODS We investigated how physiological bone environments affect the osteogenic commitment of recalcitrant MSCs in vitro and in vivo. To this end, MSC of BM and WJ origin were co-cultured and induced for synchronous osteogenic differentiation in vitro using transwells. For in vivo experiments, immunodeficient mice were injected intratibially with a single dose of human MSC and bone formation was evaluated after six weeks. RESULTS Co-culture of MSC,BM and MSC,WJ resulted in efficient osteogenesis in both cell types after three weeks. However, MSC,WJ failed to commit to bone cells in the absence of MSC,BM's osteogenic stimuli. In vivo studies showed successful bone formation within the medullar cavity of tibias in 62.5% of mice treated with MSC, WJ. By contrast, new formed trabeculae were only observed in 25% of MSC,BM-treated mice. Immunohistochemical staining of human COXIV revealed the persistence of the infused cells at the site of injection. Additionally, cells of human origin were also identified in the brain, heart, spleen, kidney and gonads in some animals treated with engineered MSC,WJ (eMSC,WJ). Importantly, no macroscopic histopathological alterations, ectopic bone formation or any other adverse events were detected in MSC-treated mice. CONCLUSIONS Our findings demonstrate that in physiological bone microenvironment, osteogenic commitment of MSC,WJ is comparable to that of MSC,BM, and support the use of off-the-shelf allogeneic MSC,WJ products in bone repair and bone regeneration applications.
Collapse
Affiliation(s)
- Raquel Cabrera-Pérez
- Servei de Teràpia Cel·lular i Avançada, Blood and Tissue Bank (BST), 08005, Barcelona, Catalonia, Spain
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR) and Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Catalonia, Spain
| | - Alexis Ràfols-Mitjans
- Centre for Genomic Regulation (CRG), Genomic Regulation, Stem Cells and Cancer Program, The Barcelona Institute of Science and Technology, 08003, Barcelona, Catalonia, Spain
| | - Ángela Roig-Molina
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR) and Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Catalonia, Spain
| | - Silvia Beltramone
- Centre for Genomic Regulation (CRG), Genomic Regulation, Stem Cells and Cancer Program, The Barcelona Institute of Science and Technology, 08003, Barcelona, Catalonia, Spain
| | - Joaquim Vives
- Servei de Teràpia Cel·lular i Avançada, Blood and Tissue Bank (BST), 08005, Barcelona, Catalonia, Spain.
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR) and Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Catalonia, Spain.
- Medicine Department, Universitat Autònoma de Barcelona (UAB), 08193, Barcelona, Catalonia, Spain.
| | - Laura Batlle-Morera
- Centre for Genomic Regulation (CRG), Genomic Regulation, Stem Cells and Cancer Program, The Barcelona Institute of Science and Technology, 08003, Barcelona, Catalonia, Spain.
| |
Collapse
|
8
|
Maeng SW, Ko JY, Park TY, Yun J, Park SH, Han SJ, Joo KI, Ha S, Jee M, Im GI, Cha HJ. Adipose stem cell transplantation using adhesive protein-based viscous immiscible liquid for cartilage reconstruction. CHEMICAL ENGINEERING JOURNAL 2023; 463:142379. [DOI: 10.1016/j.cej.2023.142379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
9
|
Melo-Fonseca F, Carvalho O, Gasik M, Miranda G, Silva FS. Mechanical stimulation devices for mechanobiology studies: a market, literature, and patents review. Biodes Manuf 2023. [DOI: 10.1007/s42242-023-00232-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
AbstractSignificant advancements in various research and technological fields have contributed to remarkable findings on the physiological dynamics of the human body. To more closely mimic the complex physiological environment, research has moved from two-dimensional (2D) culture systems to more sophisticated three-dimensional (3D) dynamic cultures. Unlike bioreactors or microfluidic-based culture models, cells are typically seeded on polymeric substrates or incorporated into 3D constructs which are mechanically stimulated to investigate cell response to mechanical stresses, such as tensile or compressive. This review focuses on the working principles of mechanical stimulation devices currently available on the market or custom-built by research groups or protected by patents and highlights the main features still open to improvement. These are the features which could be focused on to perform, in the future, more reliable and accurate mechanobiology studies.
Graphic abstract
Collapse
|
10
|
Im GI. Regenerative medicine for osteonecrosis of the femoral head : present and future. Bone Joint Res 2023; 12:5-8. [PMID: 36587245 PMCID: PMC9872044 DOI: 10.1302/2046-3758.121.bjr-2022-0057.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Cite this article: Bone Joint Res 2023;12(1):5-8.
Collapse
Affiliation(s)
- Gun-Il Im
- Research Institute for Convergence Life Science, Dongguk University, Goyang, South Korea, Gun-Il Im. E-mail:
| |
Collapse
|
11
|
Eliasberg CD, Nemirov DA, Green SJE, Melancon SG, Rodeo SA. Evaluation of Patient Preference and Perception Regarding the Clinical Use of Autologous Versus Allogeneic Cell Therapy in Orthopedic Surgery. HSS J 2022; 18:240-247. [PMID: 35645640 PMCID: PMC9096988 DOI: 10.1177/15563316211014885] [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: 12/30/2020] [Accepted: 03/04/2021] [Indexed: 02/07/2023]
Abstract
Background: Cell therapy has become a hot topic in orthopedics, with significant research dedicated to improving physicians' understanding of its efficacy. However, little is known about patients' cell therapy knowledge. Questions/Purposes: The aims of this study were to (1) evaluate patients' perceptions of cell therapy in orthopedics, (2) determine whether patients have a preference for autologous or allogeneic cell therapy, and (3) assess patient concerns about cell therapy. Methods: Consecutive outpatients of an orthopedic clinic were surveyed from June 2019 to January 2020. All patients were 18 years old or older and being seen for an orthopedic intervention, including rotator cuff repair, anterior cruciate ligament (ACL) reconstruction, arthroscopic meniscectomy, or a cartilage repair procedure such as an osteochondral allograft transplantation or matrix-associated autologous chondrocyte implantation. Results: A total of 50 patients were surveyed (mean age: 53 years). The patients' average rating for likelihood to use autologous cells was 8.86 ± 2.2 out of 10 and the average rating for likelihood to use allogeneic cells was 6.24 ± 3.3; 46% of patients had no specific concerns about autologous cell therapy, while 28% expressed concerns about efficacy, and 12% had concerns about donor age. The top 2 "main concerns" about allogeneic cell therapy were disease transmission (30%) and immune reaction (24%). Conclusions: This survey found that patients asserted a preference for autologous cell therapy in orthopedics. Further research is necessary to further elucidate the factors related to cell therapy that are most important to patients.
Collapse
|
12
|
Kir MC, Onal MO, Uluer ET, Ulman C, Inan S. Continuous and intermittent parathyroid hormone administration promotes osteogenic differentiation and activity of programmable cells of monocytic origin. Biotech Histochem 2022; 97:593-603. [PMID: 35473476 DOI: 10.1080/10520295.2022.2049876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Bone healing deficiencies are challenging for orthopedic practice. The use of stem cells with scaffolds to treat bone tissue losses currently is popular for promoting regeneration of tissue. Programmable cells of monocytic origin (PCMO) may differentiate into three germ layers and may be a promising alternative treatment due to their stem cell-like properties. Parathyroid hormone (PTH) participates in bone metabolism. Intermittent administration of PTH promotes osteogenic activity of mesenchymal stem cdells (MSC). We investigated the osteogenic effects of continuous and intermittent administration of PTH on PCMO. Mononuclear cells were harvested from the peripheral blood of healthy donors. Isolated cells were cultured for six days in a de-differentiation medium. Indirect immunocytochemistry using anti-CD14, anti-CD45 and anti-CD90 primary antibodies, as well as electron microscopy were used to detect PCMO. PCMO then were cultured in an osteogenic differentiation medium supplemented with continuous or intermittent 50 ng/ml PTH. The PTH-free control group (CG), intermittent PTH treated group (IPG) and continuous PTH treated group (CPG) were cultured and assessed for their differentiation into osteogenic lineage cells by indirect immunocytochemistry using anti-collagen I, anti-osteonectin and anti-osteocalcin primary antibodies. Osteoblast-like cells obtained by continuous or intermittent PTH administration exhibited increased levels of collagen I, osteonectin and osteocalcin immunoreactivity. We found that continuous and intermittent PTH administration to PCMO enhanced their differentiation to osteogenic lineage cells and increased osteoblastic activity.
Collapse
Affiliation(s)
- M C Kir
- Department of Orthopedics and Traumatology, Okmeydani Training and Research Hospital, Istanbul, Turkey
| | - M O Onal
- Department of Histology & Embryology, Faculty of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| | - E T Uluer
- Department of Histology & Embryology, Faculty of Medicine, Manisa Celal Bayar University, Manisa, Turkey
| | - C Ulman
- Department of Biochemistry, Faculty of Medicine, Manisa Celal Bayar University, Manisa, Turkey
| | - S Inan
- Department of Histology & Embryology, Faculty of Medicine, Izmir University of Economics, Izmir, Turkey
| |
Collapse
|
13
|
Quality control methods in musculoskeletal tissue engineering: from imaging to biosensors. Bone Res 2021; 9:46. [PMID: 34707086 PMCID: PMC8551153 DOI: 10.1038/s41413-021-00167-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 04/23/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue engineering is rapidly progressing toward clinical application. In the musculoskeletal field, there has been an increasing necessity for bone and cartilage replacement. Despite the promising translational potential of tissue engineering approaches, careful attention should be given to the quality of developed constructs to increase the real applicability to patients. After a general introduction to musculoskeletal tissue engineering, this narrative review aims to offer an overview of methods, starting from classical techniques, such as gene expression analysis and histology, to less common methods, such as Raman spectroscopy, microcomputed tomography, and biosensors, that can be employed to assess the quality of constructs in terms of viability, morphology, or matrix deposition. A particular emphasis is given to standards and good practices (GXP), which can be applicable in different sectors. Moreover, a classification of the methods into destructive, noninvasive, or conservative based on the possible further development of a preimplant quality monitoring system is proposed. Biosensors in musculoskeletal tissue engineering have not yet been used but have been proposed as a novel technology that can be exploited with numerous advantages, including minimal invasiveness, making them suitable for the development of preimplant quality control systems.
Collapse
|
14
|
Peng Y, Qu R, Feng Y, Huang X, Yang Y, Fan T, Sun B, Khan AU, Wu S, Dai J, Ouyang J. Regulation of the integrin αVβ3- actin filaments axis in early osteogenesis of human fibroblasts under cyclic tensile stress. Stem Cell Res Ther 2021; 12:523. [PMID: 34620239 PMCID: PMC8496073 DOI: 10.1186/s13287-021-02597-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 07/11/2021] [Indexed: 11/23/2022] Open
Abstract
Background Integrins play a prominent role in osteogenic differentiation by transmitting both mechanical and chemical signals. Integrin expression is closely associated with tensile stress, which has a positive effect on osteogenic differentiation. We investigated the relationship between integrin αVβ3 and tensile stress. Methods Human fibroblasts were treated with c (RGDyk) and lentivirus transduction to inhibit function of integrin αVβ3. Y-15, cytochalasin D and verteporfin were used to inhibit phosphorylation of FAK, polymerization of microfilament and function of nuclear YAP, respectively. Fibroblasts were exposed to a cyclic tensile stress of 10% at 0.5 Hz, once a day for 2 h each application. Fibroblasts were harvested on day 4 and 7 post-treatment. The expression of ALP, RUNX2, integrin αVβ3, β-actin, talin-1, FAK, vinculin, and nuclear YAP was detected by Western blot or qRT-PCR. The expression and distribution of integrin αVβ3, vinculin, microfilament and nuclear YAP. Results Cyclic tensile stress was found to promote expression of ALP and RUNX2. Inhibition of integrin αVβ3 activation downregulated the rearrangement of microfilament and the expression of ALP, RUNX2 and nuclear YAP. When the polymerization of microfilament was inhibited the expression of ALP, RUNX2 and nuclear YAP were decreased. The phosphorylation of FAK induced by cyclic tensile stress reduced by the inhibition of integrin αVβ3. The expression of ALP and RUNX2 was decreased by inhibition of phosphorylation of FAK and inhibition of nuclear YAP. Conclusions Cyclic tensile stress promotes osteogenesis of human fibroblasts via integrin αVβ3-microfilament axis. Phosphorylation of FAK and nuclear YAP participates in this process. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02597-y.
Collapse
Affiliation(s)
- Yan Peng
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510000, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510000, China
| | - Yanting Feng
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, Guangdong, China
| | - Xiaolan Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510000, China
| | - Yuchao Yang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510000, China
| | - Tingyu Fan
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510000, China
| | - Bing Sun
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510000, China
| | - Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510000, China
| | - Shutong Wu
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510000, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510000, China.
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
15
|
Chondrogenic Potential of Human Dental Pulp Stem Cells Cultured as Microtissues. Stem Cells Int 2021; 2021:7843798. [PMID: 34539791 PMCID: PMC8443354 DOI: 10.1155/2021/7843798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/22/2021] [Accepted: 08/16/2021] [Indexed: 11/18/2022] Open
Abstract
Several tissue engineering stem cell-based procedures improve hyaline cartilage repair. In this work, the chondrogenic potential of dental pulp stem cell (DPSC) organoids or microtissues was studied. After several weeks of culture in proliferation or chondrogenic differentiation media, synthesis of aggrecan and type II and I collagen was immunodetected, and SOX9, ACAN, COL2A1, and COL1A1 gene expression was analysed by real-time RT-PCR. Whereas microtissues cultured in proliferation medium showed the synthesis of aggrecan and type II and I collagen at the 6th week of culture, samples cultured in chondrogenic differentiation medium showed an earlier and important increase in the synthesis of these macromolecules after 4 weeks. Gene expression analysis showed a significant increase of COL2A1 after 3 days of culture in chondrogenic differentiation medium, while COL1A1 was highly expressed after 14 days. Cell-cell proximity promotes the chondrogenic differentiation of DPSCs and important synthesis of hyaline chondral macromolecules.
Collapse
|
16
|
Vahedi P, Moghaddamshahabi R, Webster TJ, Calikoglu Koyuncu AC, Ahmadian E, Khan WS, Jimale Mohamed A, Eftekhari A. The Use of Infrapatellar Fat Pad-Derived Mesenchymal Stem Cells in Articular Cartilage Regeneration: A Review. Int J Mol Sci 2021; 22:9215. [PMID: 34502123 PMCID: PMC8431575 DOI: 10.3390/ijms22179215] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cartilage is frequently damaged with a limited capacity for repair. Current treatment strategies are insufficient as they form fibrocartilage as opposed to hyaline cartilage, and do not prevent the progression of degenerative changes. There is increasing interest in the use of autologous mesenchymal stem cells (MSC) for tissue regeneration. MSCs that are used to treat articular cartilage defects must not only present a robust cartilaginous production capacity, but they also must not cause morbidity at the harvest site. In addition, they should be easy to isolate from the tissue and expand in culture without terminal differentiation. The source of MSCs is one of the most important factors that may affect treatment. The infrapatellar fat pad (IPFP) acts as an important reservoir for MSC and is located in the anterior compartment of the knee joint in the extra-synovial area. The IPFP is a rich source of MSCs, and in this review, we discuss studies that demonstrate that these cells have shown many advantages over other tissues in terms of ease of isolation, expansion, and chondrogenic differentiation. Future studies in articular cartilage repair strategies and suitable extraction as well as cell culture methods will extend the therapeutical application of IPFP-derived MSCs into additional orthopedic fields, such as osteoarthritis. This review provides the latest research concerning the use of IPFP-derived MSCs in the treatment of articular cartilage damage, providing critical information for the field to grow.
Collapse
Affiliation(s)
- Parviz Vahedi
- Department of Anatomical Sciences, Maragheh University of Medical Sciences, Maragheh 78151-55158, Iran;
| | - Rana Moghaddamshahabi
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta 99628, North Cyprus, Turkey;
| | - Thomas J. Webster
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA;
| | - Ayse Ceren Calikoglu Koyuncu
- Materials and Metallurgical Engineering Department, Faculty of Technology, Marmara University, Istanbul 34722, Turkey;
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz 51666-15731, Iran;
| | - Wasim S. Khan
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Ali Jimale Mohamed
- Department of Pharmacology, Faculty of Medicine, Somali National University, Mogadishu 801, Somalia;
| | - Aziz Eftekhari
- Department of Toxicology and Pharmacology, Maragheh University of Medical Sciences, Maragheh 78151-55158, Iran
- Department of Synthesis and Characterization of Polymers, Polymer Institute, Slovak Academy of Sciences (SAS), Dúbravská cesta, 9, 845 41 Bratislava, Slovakia
| |
Collapse
|
17
|
Eliasberg CD, Nemirov DA, Mandelbaum BR, Pearle AD, Tokish JM, Baria MR, Millett PJ, Shapiro SA, Rodeo SA. Complications Following Biologic Therapeutic Injections: A Multicenter Case Series. Arthroscopy 2021; 37:2600-2605. [PMID: 33872744 DOI: 10.1016/j.arthro.2021.03.065] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE To describe the complications that occur following biologic therapeutic injections. METHODS We queried physician members of the Biologic Association, a multidisciplinary organization dedicated to providing a unified voice for all matters related to musculoskeletal biologics and regenerative medicine. Patients included in this study must have (1) received a biologic injection, (2) sustained an adverse reaction, and (3) had a minimum of 1-year follow-up after the injection. Patient demographic information, medical comorbidities, diagnoses, and previous treatments were recorded. The type of injection, injection setting, injection manufacturers, and specific details about the complication and outcome were collected. RESULTS In total, 14 patients were identified across 6 institutions in the United States (mean age 63 years, range: 36-83 years). The most common injections in this series were intra-articular knee injections (50%), followed intra-articular shoulder injections (21.4%). The most common underlying diagnosis was osteoarthritis (78.5%). Types of injections included umbilical cord blood, platelet-rich plasma, bone marrow aspirate concentrate, placental tissue, and unspecified "stem cell" injections. Complications included infection (50%), suspected sterile inflammatory response (42.9%), and a combination of both (7.1%). The most common pathogen identified from infection cases was Escherichia coli (n = 4). All patients who had isolated infections underwent treatment with at least one subsequent surgical intervention (mean: 3.6, range: 1-12) and intravenous antibiotic therapy. CONCLUSIONS This study demonstrates that serious complications can occur following treatment with biologic injections, including infections requiring multiple surgical procedures and inflammatory reactions. LEVEL OF EVIDENCE Level IV, case series.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Scott A Rodeo
- Hospital for Special Surgery, New York, New York, U.S.A
| |
Collapse
|
18
|
Tucker JD, Goetz LL, Duncan MB, Gilman JB, Elmore LW, Sell SA, McClure MJ, Quagliano PV, Martin CC. Randomized, Placebo-Controlled Analysis of the Knee Synovial Environment Following Platelet-Rich Plasma Treatment for Knee Osteoarthritis. PM R 2021; 13:707-719. [PMID: 33492733 DOI: 10.1002/pmrj.12561] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/24/2020] [Accepted: 01/05/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND Platelet-rich-plasma (PRP) is used to treat knee osteoarthritis; however, mechanistic evidence of PRP effectiveness for pain relief is limited. OBJECTIVE To assess molecular biomarkers and mesenchymal stem cells (MSCs) in synovial fluid during PRP treatment of the osteoarthritic knee joint. DESIGN Single blinded, randomized, placebo controlled pilot study. SETTING Veterans Affairs Medical Center. PARTICIPANTS Seventeen participants with mild to moderate knee osteoarthritis were randomized in a 2:1 placebo-controlled ratio, receiving PRP or saline (placebo) intra-articular injection into the knee joint. METHODS Knee synovial fluid was analyzed before the respective injections and again 10 days following injection. Participants were followed up to 12 months completing visual analog scale (VAS) and Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) questionnaires at intervals over that period. MAIN OUTCOME MEASURES The effects of PRP on synovial protein and MSC gene expression levels were measured by multiplex enzyme-linked immunosorbent assay and quantitative polymerase chain reaction. RESULTS Novel biomarkers including levels of interleukin (IL)-5, IL-6, IL-10, and tumor necrosis factor-α were measured in synovial fluid 10 days after PRP treatment. Altered gene expression profiles in MSCs from patients treated with PRP were observed for matrix metalloproteinases and inflammatory markers (IL-6, IL-8, CCL2, TNF-α). A2M protease was significantly increased following PRP treatment (P = .005). WOMAC scores declined for up to 3 months from baseline levels and remained low at 6 and 12 months in the PRP group. In contrast, WOMAC scores for patients receiving the saline injection were relatively unchanged for up to 12 months. CONCLUSIONS We report significant changes for the biomarker A2M (P = .005) as well as differences in expression of cellular markers and postulate that PRP modulates the local knee synovial environment by altering the inflammatory milieu, matrix degradation, and angiogenic growth factors. The PRP treatment group had less pain and stiffness and improved function scores.
Collapse
Affiliation(s)
- Jason D Tucker
- iOrthoBiologix, Charlotte, NC.,Department of Physical Medicine and Rehabilitation, Virginia Commonwealth University Health System, Richmond, VA
| | - Lance L Goetz
- Spinal Cord Injury and Disorders Service, Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA
| | | | - Jared B Gilman
- Department of Physical Medicine and Rehabilitation, Virginia Commonwealth University Health System, Richmond, VA
| | | | - Scott A Sell
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, St. Louis, MO
| | - Michael J McClure
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA
| | - Peter V Quagliano
- Department of Radiology, Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA
| | - Caroline C Martin
- Department or Pathology, Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA
| |
Collapse
|
19
|
Zhang Y, Zhuang Z, Wei Q, Li P, Li J, Fan Y, Zhang L, Hong Z, He W, Wang H, Liu Y, Li W. Inhibition of miR-93-5p promotes osteogenic differentiation in a rabbit model of trauma-induced osteonecrosis of the femoral head. FEBS Open Bio 2021. [PMID: 34092046 PMCID: PMC8329948 DOI: 10.1002/2211-5463.13218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 05/12/2021] [Accepted: 06/04/2021] [Indexed: 01/08/2023] Open
Abstract
Trauma‐induced osteonecrosis of the femoral head (TIONFH) is characterized by femoral head collapse accompanied by degenerative changes of the hip. We previously reported that miR‐93‐5p expression is abnormally high in patients with TIONFH, but the role of miR‐93‐5p in the TIONFH process remains unclear. Herein, we investigated the role of miR‐93‐5p in TIONFH in a rabbit model. Bone marrow mesenchymal stem cells (BMSCs) were used for both in vivo and in vitro experiments. A rabbit model of TIONFH was injected with BMSCs transfected with miR‐93‐5p inhibitor. In addition, both an miR‐93‐5p mimic and negative control were transfected into BMSCs. Expression of miR‐93‐5p was significantly increased in the model group compared with control samples. An miR‐93‐5p inhibitor induced the expression of bone morphogenetic protein 2 (BMP‐2) and alkaline phosphatase. Furthermore, expression of osteogenesis‐related markers (BMP‐2, secreted phosphoprotein 1, RUNX family transcription factor 2 and Osterix) was higher in the miR‐93‐5p inhibitor group, as revealed by quantitative PCR and western blotting. In addition, in vitro experimentation revealed that an miR‐93‐5p mimic decreased BMP‐2 and TNF receptor superfamily member 11b expression, but increased receptor activator of nuclear factor‐kappaB ligand expression. In summary, the miR‐93‐5p inhibitor could promote osteogenic differentiation by increasing BMP‐2 expression during the development of TIONFH. Thus, miR‐93‐5p may have potential as a therapeutic target for TIONF treatment.
Collapse
Affiliation(s)
- Ying Zhang
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), China.,Guangzhou University of Chinese Medicine, China
| | | | - Qiushi Wei
- Institute of Orthopaedics of Guangzhou University of Chinese Medicine, China.,The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, China
| | - Peifeng Li
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), China
| | - Jitian Li
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), China
| | - Yanan Fan
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), China
| | - Leilei Zhang
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), China
| | - Zhinan Hong
- Institute of Orthopaedics of Guangzhou University of Chinese Medicine, China.,The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, China
| | - Wei He
- Institute of Orthopaedics of Guangzhou University of Chinese Medicine, China.,The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, China
| | - Haibin Wang
- Guangzhou University of Chinese Medicine, China
| | - Youwen Liu
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), China
| | - Wuyin Li
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), China
| |
Collapse
|
20
|
Dakkak A, Krill M, Fogarty A, Krill M. Stem cell therapy for the management of lateral elbow tendinopathy: A systematic literature review. Sci Sports 2021. [DOI: 10.1016/j.scispo.2020.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
21
|
Brahmbhatt AN, Misra S. Stem Cell Delivery for the Treatment of Arteriovenous Fistula Failure. STEM CELL THERAPY FOR VASCULAR DISEASES 2021:281-297. [DOI: 10.1007/978-3-030-56954-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
22
|
Wu JQ, Jiang N, Yu B. Mechanisms of action of neuropeptide Y on stem cells and its potential applications in orthopaedic disorders. World J Stem Cells 2020; 12:986-1000. [PMID: 33033559 PMCID: PMC7524693 DOI: 10.4252/wjsc.v12.i9.986] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/25/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023] Open
Abstract
Musculoskeletal disorders are the leading causes of disability and result in reduced quality of life. The neuro-osteogenic network is one of the most promising fields in orthopaedic research. Neuropeptide Y (NPY) system has been reported to be involved in the regulations of bone metabolism and homeostasis, which also provide feedback to the central NPY system via NPY receptors. Currently, potential roles of peripheral NPY in bone metabolism remain unclear. Growing evidence suggests that NPY can regulate biological actions of bone marrow mesenchymal stem cells, hematopoietic stem cells, endothelial cells, and chondrocytes via a local autocrine or paracrine manner by different NPY receptors. The regulative activities of NPY may be achieved through the plasticity of NPY receptors, and interactions among the targeted cells as well. In general, NPY can influence proliferation, apoptosis, differentiation, migration, mobilization, and cytokine secretion of different types of cells, and play crucial roles in the development of bone delayed/non-union, osteoporosis, and osteoarthritis. Further basic research should clarify detailed mechanisms of action of NPY on stem cells, and clinical investigations are also necessary to comprehensively evaluate potential applications of NPY and its receptor-targeted drugs in management of musculoskeletal disorders.
Collapse
Affiliation(s)
- Jian-Qun Wu
- Department of Orthopedics and Traumatology, Huadu District People’s Hospital, Guangzhou 510800, Guangdong Province, China
| | - Nan Jiang
- Division of Orthopaedics and Traumatology, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
23
|
Citeroni MR, Ciardulli MC, Russo V, Della Porta G, Mauro A, El Khatib M, Di Mattia M, Galesso D, Barbera C, Forsyth NR, Maffulli N, Barboni B. In Vitro Innovation of Tendon Tissue Engineering Strategies. Int J Mol Sci 2020; 21:E6726. [PMID: 32937830 PMCID: PMC7555358 DOI: 10.3390/ijms21186726] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Tendinopathy is the term used to refer to tendon disorders. Spontaneous adult tendon healing results in scar tissue formation and fibrosis with suboptimal biomechanical properties, often resulting in poor and painful mobility. The biomechanical properties of the tissue are negatively affected. Adult tendons have a limited natural healing capacity, and often respond poorly to current treatments that frequently are focused on exercise, drug delivery, and surgical procedures. Therefore, it is of great importance to identify key molecular and cellular processes involved in the progression of tendinopathies to develop effective therapeutic strategies and drive the tissue toward regeneration. To treat tendon diseases and support tendon regeneration, cell-based therapy as well as tissue engineering approaches are considered options, though none can yet be considered conclusive in their reproduction of a safe and successful long-term solution for full microarchitecture and biomechanical tissue recovery. In vitro differentiation techniques are not yet fully validated. This review aims to compare different available tendon in vitro differentiation strategies to clarify the state of art regarding the differentiation process.
Collapse
Affiliation(s)
- Maria Rita Citeroni
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Maria Camilla Ciardulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (G.D.P.); (N.M.)
| | - Valentina Russo
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (G.D.P.); (N.M.)
- Interdepartment Centre BIONAM, Università di Salerno, via Giovanni Paolo I, 84084 Fisciano (SA), Italy
| | - Annunziata Mauro
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Mohammad El Khatib
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Miriam Di Mattia
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Devis Galesso
- Fidia Farmaceutici S.p.A., via Ponte della Fabbrica 3/A, 35031 Abano Terme (PD), Italy; (D.G.); (C.B.)
| | - Carlo Barbera
- Fidia Farmaceutici S.p.A., via Ponte della Fabbrica 3/A, 35031 Abano Terme (PD), Italy; (D.G.); (C.B.)
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Thornburrow Drive, Stoke on Trent ST4 7QB, UK;
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (G.D.P.); (N.M.)
- Department of Musculoskeletal Disorders, Faculty of Medicine and Surgery, University of Salerno, Via San Leonardo 1, 84131 Salerno, Italy
- Centre for Sports and Exercise Medicine, Barts and The London School of Medicine and Dentistry, Mile End Hospital, Queen Mary University of London, 275 Bancroft Road, London E1 4DG, UK
- School of Pharmacy and Bioengineering, Keele University School of Medicine, Thornburrow Drive, Stoke on Trent ST5 5BG, UK
| | - Barbara Barboni
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| |
Collapse
|
24
|
Janvier AJ, Canty-Laird E, Henstock JR. A universal multi-platform 3D printed bioreactor chamber for tendon tissue engineering. J Tissue Eng 2020; 11:2041731420942462. [PMID: 32944210 PMCID: PMC7469720 DOI: 10.1177/2041731420942462] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
A range of bioreactors use linear actuators to apply tensile forces in vitro, but differences in their culture environments can limit a direct comparison between studies. The widespread availability of 3D printing now provides an opportunity to develop a 'universal' bioreactor chamber that, with minimal exterior editing can be coupled to a wide range of commonly used linear actuator platforms, for example, the EBERS-TC3 and CellScale MCT6, resulting in a greater comparability between results and consistent testing of potential therapeutics. We designed a bioreactor chamber with six independent wells that was 3D printed in polylactic acid using an Ultimaker 2+ and waterproofed using a commercially available coating (XTC-3D), an oxirane resin. The cell culture wells were further coated with Sylgard-184 polydimethylsiloxane (PDMS) to produce a low-adhesion well surface. With appropriate coating and washing steps, all materials were shown to be non-cytotoxic by lactate dehydrogenase assay, and the bioreactor was waterproof, sterilisable and reusable. Tissue-engineered tendons were generated from human mesenchymal stem cells in a fibrin hydrogel and responded to 5% cyclic strain (0.5 Hz, 5 h/day, 21 days) in the bioreactor by increased production of collagen-Iα1 and decreased production of collagen-IIIα1. Calcification of the extracellular matrix was observed in unstretched tendon controls indicating abnormal differentiation, while tendons cultured under cyclic strain did not calcify and exhibited a tenogenic phenotype. The ease of manufacturing this bioreactor chamber enables researchers to quickly and cheaply reproduce this culture environment for use with many existing bioreactor actuator platforms by downloading the editable CAD files from a public database and following the manufacturing steps we describe.
Collapse
Affiliation(s)
- Adam J Janvier
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | | - James R Henstock
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| |
Collapse
|
25
|
Shelat R, Bhatt LK, Paunipagar B, Kurian T, Khanna A, Chandra S. Regeneration of hyaline cartilage in osteochondral lesion model using L-lysine magnetic nanoparticles labeled mesenchymal stem cells and their in vivo imaging. J Tissue Eng Regen Med 2020; 14:1604-1617. [PMID: 32840054 DOI: 10.1002/term.3120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/24/2020] [Accepted: 08/04/2020] [Indexed: 12/16/2022]
Abstract
Treatment of osteochondral defects continues to pose a major challenge for patients and orthopedic surgeons due to the limited healing potential of articular cartilage. Mesenchymal stem cells (MSCs) possess therapeutic potential for the treatment of osteochondral pain and pathology. However, it is necessary to use proper labeling and imaging agent of stem cells that can decipher its role posttransplantation. A major limitation of routinely used contrast agents is signal dilution over a period of time which limits its use for further studies. At the same time, regeneration of fibrocartilage over native hyaline cartilage also limits the use of conventional therapies. The present study evaluates the efficacy of bone marrow-derived mesenchymal stem cells (BMSCs) for the treatment of osteochondral defect in rats with the regeneration of hyaline cartilage in situ and in vivo monitoring of the stem cells using L-lysine functionalized magnetic iron oxide nanoparticles (lys-IONPs). L-lysine stabilizes the iron oxide nanoparticles, enhances the biocompatibility, and provides functionalities for efficient stem cell labeling. in vitro toxic effects of lys-IONPs on mitochondrial impairment, morphological alterations, and actin cytoskeleton reveal minimum damage to BM-MSCs. Histological data (H and E, Masson's trichrome and immunohistochemistry) describe the early initiation of healing and regeneration of hyaline-like cartilage over fibrocartilage in stem cell treated groups. MR scans demonstrate generation of hypointense signals in lys-IONPs-BMSCs with improved signal intensity and minimum loss over 28 days revealing its use as a long-term stem cell labeling and imaging agent.
Collapse
Affiliation(s)
- Ruchita Shelat
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS University, Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | | | | | - Aparna Khanna
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS University, Mumbai, India.,Computational Biology and Translational Research, Amity University, Mumbai, India
| | - Sudeshna Chandra
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS University, Mumbai, India
| |
Collapse
|
26
|
Ethical and Practical Considerations for Integrating Cellular ("Stem Cell") Therapy into Clinical Practice. Curr Rev Musculoskelet Med 2020; 13:525-529. [PMID: 32468421 DOI: 10.1007/s12178-020-09647-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Cellular therapies, also known as "stem cell" interventions (SCI), have undergone a rapid popularization in the USA and worldwide. The current review aimed at outlining (1) the ethical challenges facing the implementation of SCI; (2) the applicability of the currently available SCI; and (3) recommendations to achieve ethical, well-regulated incorporation of SCI in the clinical setting. RECENT FINDINGS Concerns regarding the inadequate characterization, poor adverse effects disclosure, and unorthodox, often inappropriate, market practices have engendered a genuine concern regarding the SCI compliance with ethical standards. Six instances of litigation on the basis of misrepresentation or inappropriate informed consent were recorded between 2012 and 2018. Such concerns have been furthered by the loopholes in the regulatory aspect governing the use of SCI coupled with the unclear literature-reported efficacy and diverse spectrum of profess indications. Similarly, the application of SCI in the clinical field is yet to prove its value. The uncertain efficacy, coupled with obscure true-costs of utilization, impedes a value-based assessment. A multidisciplinary approach involving legislative and medical professional societies should continue to advance regulations that govern SCI. A well-regulated system that allows for the ethical integration of SCI with appositely evidenced-based described benefits and risks should be sought.
Collapse
|
27
|
Ciardulli MC, Marino L, Lovecchio J, Giordano E, Forsyth NR, Selleri C, Maffulli N, Porta GD. Tendon and Cytokine Marker Expression by Human Bone Marrow Mesenchymal Stem Cells in a Hyaluronate/Poly-Lactic-Co-Glycolic Acid (PLGA)/Fibrin Three-Dimensional (3D) Scaffold. Cells 2020; 9:E1268. [PMID: 32443833 PMCID: PMC7291129 DOI: 10.3390/cells9051268] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 01/19/2023] Open
Abstract
We developed a (three-dimensional) 3D scaffold, we named HY-FIB, incorporating a force-transmission band of braided hyaluronate embedded in a cell localizing fibrin hydrogel and poly-lactic-co-glycolic acid (PLGA) nanocarriers as transient components for growth factor controlled delivery. The tenogenic supporting capacity of HY-FIB on human-Bone Marrow Mesenchymal Stem Cells (hBM-MSCs) was explored under static conditions and under bioreactor-induced cyclic strain conditions. HY-FIB elasticity enabled to deliver a mean shear stress of 0.09 Pa for 4 h/day. Tendon and cytokine marker expression by hBM-MSCs were studied. Results: hBM-MSCs embedded in HY-FIB and subjected to mechanical stimulation, resulted in a typical tenogenic phenotype, as indicated by type 1 Collagen fiber immunofluorescence. RT-qPCR showed an increase of type 1 Collagen, scleraxis, and decorin gene expression (3-fold, 1600-fold, and 3-fold, respectively, at day 11) in dynamic conditions. Cells also showed pro-inflammatory (IL-6, TNF, IL-12A, IL-1β) and anti-inflammatory (IL-10, TGF-β1) cytokine gene expressions, with a significant increase of anti-inflammatory cytokines in dynamic conditions (IL-10 and TGF-β1 300-fold and 4-fold, respectively, at day 11). Mechanical signaling, conveyed by HY-FIB to hBM-MSCs, promoted tenogenic gene markers expression and a pro-repair cytokine balance. The results provide strong evidence in support of the HY-FIB system and its interaction with cells and its potential for use as a predictive in vitro model.
Collapse
Affiliation(s)
- Maria C. Ciardulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (L.M.); (C.S.); (N.M.)
| | - Luigi Marino
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (L.M.); (C.S.); (N.M.)
| | - Joseph Lovecchio
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna, Via dell’Università 50, 47522 Cesena (FC), Italy; (J.L.); (E.G.)
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna, Via dell’Università 50, 47522 Cesena (FC), Italy; (J.L.); (E.G.)
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire ST4 7QB, UK;
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (L.M.); (C.S.); (N.M.)
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (L.M.); (C.S.); (N.M.)
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, Staffordshire ST4 7QB, UK;
- Centre for Sport and Exercise Medicine, Queen Mary University of London, Barts and The London School of Medicine, London E1 4NL, UK
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (L.M.); (C.S.); (N.M.)
- Department of Industrial Engineering, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano (SA), Italy
| |
Collapse
|
28
|
Dewey MJ, Nosatov AV, Subedi K, Harley B. Anisotropic mineralized collagen scaffolds accelerate osteogenic response in a glycosaminoglycan-dependent fashion. RSC Adv 2020; 10:15629-15641. [PMID: 32655857 PMCID: PMC7351350 DOI: 10.1039/d0ra01336f] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regeneration of critically-sized craniofacial bone defects requires a template to promote cell activity and bone remodeling. However, induced regeneration becomes more challenging with increasing defect size. Methods of repair using allografts and autografts have inconsistent results, attributed to age-related regenerative capabilities of bone. We are developing a mineralized collagen scaffold to promote craniomaxillofacial bone regeneration as an alternative to repair. Here, we hypothesize modifying the pore anisotropy and glycosaminoglycan content of the scaffold will improve cell migration, viability, and subsequent bone formation. Using anisotropic and isotropic scaffold variants, we test the role of pore orientation on human mesenchymal stem cell (MSC) activity. We subsequently explore the role of glycosaminoglycan content, notably chondroitin-6-sulfate, chondroitin-4-sulfate, and heparin sulfate on mineralization. We find that while short term MSC migration and activity was not affected by pore orientation, increased bone mineral synthesis was observed in anisotropic scaffolds. Further, while scaffold glycosaminoglycan content did not impact cell viability, heparin sulfate and chondroitin-6-sulfate containing variants increased mineral formation at the late stage of in vitro culture, respectively. Overall, these findings show scaffold microstructural and proteoglycan modifications represent a powerful tool to improve MSC osteogenic activity. Mineralized collagen scaffolds were modified to include anisotropic pore architecture and one of three glycosaminoglycans in order to improve bone mineral formation in vitro.![]()
Collapse
Affiliation(s)
| | | | | | - Brendan Harley
- Dept. of Materials Science and Engineering, USA.,School of Chemical Sciences, USA.,Dept. Chemical and Biomolecular Engineering, USA.,Dept. of Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory 600 S. Mathews Ave., Urbana, IL 61801, USA
| |
Collapse
|
29
|
Dewey MJ, Johnson EM, Slater ST, Milner DJ, Wheeler MB, Harley BAC. Mineralized collagen scaffolds fabricated with amniotic membrane matrix increase osteogenesis under inflammatory conditions. Regen Biomater 2020; 7:247-258. [PMID: 32523727 PMCID: PMC7266662 DOI: 10.1093/rb/rbaa005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/17/2020] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
Defects in craniofacial bones occur congenitally, after high-energy impacts, and during the course of treatment for stroke and cancer. These injuries are difficult to heal due to the overwhelming size of the injury area and the inflammatory environment surrounding the injury. Significant inflammatory response after injury may greatly inhibit regenerative healing. We have developed mineralized collagen scaffolds that can induce osteogenic differentiation and matrix biosynthesis in the absence of osteogenic media or supplemental proteins. The amniotic membrane is derived from placentas and has been recently investigated as an extracellular matrix to prevent chronic inflammation. Herein, we hypothesized that a mineralized collagen–amnion composite scaffold could increase osteogenic activity in the presence of inflammatory cytokines. We report mechanical properties of a mineralized collagen–amnion scaffold and investigated osteogenic differentiation and mineral deposition of porcine adipose-derived stem cells within these scaffolds as a function of inflammatory challenge. Incorporation of amniotic membrane matrix promotes osteogenesis similarly to un-modified mineralized collagen scaffolds, and increases in mineralized collagen–amnion scaffolds under inflammatory challenge. Together, these findings suggest that a mineralized collagen–amnion scaffold may provide a beneficial environment to aid craniomaxillofacial bone repair, especially in the course of defects presenting significant inflammatory complications.
Collapse
Affiliation(s)
- Marley J Dewey
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA
| | - Eileen M Johnson
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA
| | - Simona T Slater
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA
| | - Derek J Milner
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA
| | - Matthew B Wheeler
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA.,Department of Animal Sciences, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA
| | - Brendan A C Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave., Urbana, IL 61801, USA
| |
Collapse
|
30
|
López-Fernández A, Barro V, Ortiz-Hernández M, Manzanares MC, Vivas D, Vives J, Vélez R, Ginebra MP, Aguirre M. Effect of Allogeneic Cell-Based Tissue-Engineered Treatments in a Sheep Osteonecrosis Model. Tissue Eng Part A 2020; 26:993-1004. [PMID: 32122263 DOI: 10.1089/ten.tea.2019.0339] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is defined as a tissue disorder and successive subchondral bone collapse resulting from an ischemic process, which may progress to hip osteoarthritis. Cell therapy with multipotent bone marrow mesenchymal stromal cells (BM-MSC) of autologous origin appears to be safe and has shown regenerative potential in previous preclinical and clinical studies. The use of allogeneic cells is far more challenging, but may be a promising alternative to use of autologous cells. Moreover, an optimized dosage of cells from an allogeneic source is needed to obtain off-the-shelf tissue engineering products (TEPs). The purpose of this study was to evaluate the efficacy of a TEP composed of undifferentiated ex vivo expanded BM-MSC of allogeneic origin, combined with bone matrix particles in variable doses. A comparative analysis of TEP's bone regenerative properties against its autologous counterpart was performed in an early-stage ONFH preclinical model in mature sheep. Allogeneic BM-MSC groups demonstrated bone regeneration capacity in osteonecrotic lesions equivalent to autologous BM-MSC groups 6 weeks after treatment. Likewise, stimulation of bone regeneration by a low cell dose of 0.5 × 106 BM-MSC/cm3 was equivalent to that of a high cell dose, 5 × 106 BM-MSC/cm3. Neither local nor systemic immunological reactions nor tumorigenesis were reported, strengthening the safety profile of allogeneic BM-MSC therapy in this model. Our results suggest that low-dose allogeneic BM-MSC is sufficient to promote bone regeneration in femoral head osteonecrotic lesions, and should be considered in translation of new allogeneic cell-based TEPs to human clinics. Impact statement Cell therapy and tissue engineering hold promise as novel regenerative therapies for musculoskeletal diseases, and particularly in bone regeneration strategies. In this article, we report the evaluation of the efficacy of an allogeneic cell-based tissue engineering product (TEP) in an early-stage osteonecrosis of the femoral head preclinical model in skeletally mature sheep. Moreover, we demonstrate its bone regeneration capacity and safety in vivo and its equivalence to autologous counterparts. These findings have important implications for the translation of new allogeneic cell-based TEPs to human clinics.
Collapse
Affiliation(s)
- Alba López-Fernández
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Víctor Barro
- Orthopedic Surgery Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Mònica Ortiz-Hernández
- Biomaterials, Biomechanics and Tissue Engineering Group (BBT), Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
| | - Maria Cristina Manzanares
- Human Anatomy and Embryology Unit, Department of Pathology and Experimental Therapeutics, Universitat de Barcelona, Barcelona, Spain
| | - Daniel Vivas
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Servei de Teràpia Cel·lular, Banc de Sang i Teixits, Barcelona, Spain
| | - Joaquim Vives
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Servei de Teràpia Cel·lular, Banc de Sang i Teixits, Barcelona, Spain.,Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Roberto Vélez
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Orthopedic Surgery Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Maria Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group (BBT), Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain.,Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Màrius Aguirre
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Orthopedic Surgery Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
31
|
Johnstone B, Stoddart MJ, Im GI. Multi-Disciplinary Approaches for Cell-Based Cartilage Regeneration. J Orthop Res 2020; 38:463-472. [PMID: 31478253 DOI: 10.1002/jor.24458] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 08/23/2019] [Indexed: 02/04/2023]
Abstract
Articular cartilage does not regenerate in adults. A lot of time and resources have been dedicated to cartilage regeneration research. The current understanding suggests that multi-disciplinary approach including biologic, genetic, and mechanical stimulations may be needed for cell-based cartilage regeneration. This review summarizes contents of a workshop sponsored by International Combined Orthopaedic Societies during the 2019 annual meeting of the Orthopaedic Research Society held in Austin, Texas. Three approaches for cell-based cartilage regeneration were introduced, including cellular basis of chondrogenesis, gene-enhanced cartilage regeneration, and physical modulation to divert stem cells to chondrogenic cell fate. While the complicated nature of cartilage regeneration has not allowed us to achieve successful regeneration of hyaline articular cartilage so far, the utilization of multi-disciplinary approaches in various fields of biomedical engineering will provide means to achieve this goal faster. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:463-472, 2020.
Collapse
Affiliation(s)
- Brian Johnstone
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, Oregon
| | | | - Gun-Il Im
- Integrative Research Institute for Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
| |
Collapse
|
32
|
Su X, Wang J, Kang H, Bao G, Liu L. Effects of dynamic radial tensile stress on fibrocartilage differentiation of bone marrow mesenchymal stem cells. Biomed Eng Online 2020; 19:8. [PMID: 32024525 PMCID: PMC7003351 DOI: 10.1186/s12938-020-0751-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/24/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Uniaxial/biaxial tensile stress has been employed to induce chondrocyte differentiation of mesenchymal stem cells. However, the effects of radial tensile stimuli on differentiation of MSCs into fibrocartilage remain unclear. RESULTS It was found that induced bone marrow mesenchymal stem cells (BMSCs) were not only similar to TMJ disc cells in morphology, but also could synthesize type I collagen (Col I), a small amount of type II collagen (Col II) and glycosaminoglycans (GAGs). The synthesis of Col I significantly increased while that of Col II gradually decreased with increasing tensile strength. The ratio of Col I to Col II was 1.8 to 1 and 2 to 1 in the 10% and 15% stretching groups, respectively. The gene expression of Col I and GAGs was significantly upregulated, whereas that of Col II was downregulated. However, the higher tensile stimulation (15%) promoted the synthesis of α-smooth muscle actin (α-SMA). Too much α-SMA is not conducive to constructing engineered tissue. CONCLUSION Therefore, the 10% radial tensile stimulus was the optimal strength for inducing the BMSCs to differentiate into fibrochondrocytes of the temporomandibular joint (TMJ) disc. This work provided a novel approach for inducing BMSCs to differentiate into fibrochondrocytes.
Collapse
Affiliation(s)
- Xuelian Su
- College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.,Key Lab of Oral Diseases of Gansu Province, Northwest Minzu University, Lanzhou, People's Republic of China.,Key Lab of Stomatology of State Ethnic Affairs Commission, Northwest Minzu University, Lanzhou, People's Republic of China.,Key Laboratory of Mechanics on Disaster and Environment in Western China, The Ministry of Education of China, Lanzhou University, Lanzhou, People's Republic of China.,Department of Prosthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu, China
| | - Jizeng Wang
- College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China. .,Key Laboratory of Mechanics on Disaster and Environment in Western China, The Ministry of Education of China, Lanzhou University, Lanzhou, People's Republic of China.
| | - Hong Kang
- College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.,Key Laboratory of Mechanics on Disaster and Environment in Western China, The Ministry of Education of China, Lanzhou University, Lanzhou, People's Republic of China
| | - Guangjie Bao
- Key Lab of Oral Diseases of Gansu Province, Northwest Minzu University, Lanzhou, People's Republic of China.,Key Lab of Stomatology of State Ethnic Affairs Commission, Northwest Minzu University, Lanzhou, People's Republic of China.,Department of Prosthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu, China
| | - Lin Liu
- Key Lab of Oral Diseases of Gansu Province, Northwest Minzu University, Lanzhou, People's Republic of China.,Key Lab of Stomatology of State Ethnic Affairs Commission, Northwest Minzu University, Lanzhou, People's Republic of China.,Department of Prosthodontics, School of Stomatology, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
33
|
Lokeshwar SD, Patel P, Shah SM, Ramasamy R. A Systematic Review of Human Trials Using Stem Cell Therapy for Erectile Dysfunction. Sex Med Rev 2020; 8:122-130. [DOI: 10.1016/j.sxmr.2019.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/19/2019] [Accepted: 08/24/2019] [Indexed: 01/08/2023]
|
34
|
Kolber MJ, Purita J, Sterling B, Stermer J, Salamh P, Masaracchio M, Hanney WJ. Stem Cell Injections for Musculoskeletal Pathology: An Overview for the Sports Medicine Professional. Strength Cond J 2019. [DOI: 10.1519/ssc.0000000000000500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Lim WL, Liau LL, Ng MH, Chowdhury SR, Law JX. Current Progress in Tendon and Ligament Tissue Engineering. Tissue Eng Regen Med 2019; 16:549-571. [PMID: 31824819 PMCID: PMC6879704 DOI: 10.1007/s13770-019-00196-w] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/07/2019] [Accepted: 05/22/2019] [Indexed: 02/08/2023] Open
Abstract
Background Tendon and ligament injuries accounted for 30% of all musculoskeletal consultations with 4 million new incidences worldwide each year and thus imposed a significant burden to the society and the economy. Damaged tendon and ligament can severely affect the normal body movement and might lead to many complications if not treated promptly and adequately. Current conventional treatment through surgical repair and tissue graft are ineffective with a high rate of recurrence. Methods In this review, we first discussed the anatomy, physiology and pathophysiology of tendon and ligament injuries and its current treatment. Secondly, we explored the current role of tendon and ligament tissue engineering, describing its recent advances. After that, we also described stem cell and cell secreted product approaches in tendon and ligament injuries. Lastly, we examined the role of the bioreactor and mechanical loading in in vitro maturation of engineered tendon and ligament. Results Tissue engineering offers various alternative ways of treatment from biological tissue constructs to stem cell therapy and cell secreted products. Bioreactor with mechanical stimulation is instrumental in preparing mature engineered tendon and ligament substitutes in vitro. Conclusions Tissue engineering showed great promise in replacing the damaged tendon and ligament. However, more study is needed to develop ideal engineered tendon and ligament.
Collapse
Affiliation(s)
- Wei Lee Lim
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, 56000 Kuala Lumpur, Malaysia
| | - Ling Ling Liau
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, JalanYaacob Latif, 56000 Kuala Lumpur, Malaysia
| | - Min Hwei Ng
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, 56000 Kuala Lumpur, Malaysia
| | - Shiplu Roy Chowdhury
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, 56000 Kuala Lumpur, Malaysia
| | - Jia Xian Law
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, 56000 Kuala Lumpur, Malaysia
| |
Collapse
|
36
|
Cabrera-Pérez R, Monguió-Tortajada M, Gámez-Valero A, Rojas-Márquez R, Borràs FE, Roura S, Vives J. Osteogenic commitment of Wharton's jelly mesenchymal stromal cells: mechanisms and implications for bioprocess development and clinical application. Stem Cell Res Ther 2019; 10:356. [PMID: 31779673 PMCID: PMC6883559 DOI: 10.1186/s13287-019-1450-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/03/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Background Orthopaedic diseases are one of the major targets for regenerative medicine. In this context, Wharton’s jelly (WJ) is an alternative source to bone marrow (BM) for allogeneic transplantation since its isolation does not require an invasive procedure for cell collection and does not raise major ethical concerns. However, the osteogenic capacity of human WJ-derived multipotent mesenchymal stromal cells (MSC) remains unclear. Methods Here, we compared the baseline osteogenic potential of MSC from WJ and BM cell sources by cytological staining, quantitative real-time PCR and proteomic analysis, and assessed chemical and biological strategies for priming undifferentiated WJ-MSC. Concretely, different inhibitors/activators of the TGFβ1-BMP2 signalling pathway as well as the secretome of differentiating BM-MSC were tested. Results Cytochemical staining as well as gene expression and proteomic analysis revealed that osteogenic commitment was poor in WJ-MSC. However, stimulation of the BMP2 pathway with BMP2 plus tanshinone IIA and the addition of extracellular vesicles or protein-enriched preparations from differentiating BM-MSC enhanced WJ-MSC osteogenesis. Furthermore, greater outcome was obtained with the use of conditioned media from differentiating BM-MSC. Conclusions Altogether, our results point to the use of master banks of WJ-MSC as a valuable alternative to BM-MSC for orthopaedic conditions.
Collapse
Affiliation(s)
- Raquel Cabrera-Pérez
- Cell Therapy Service, Blood and Tissue Bank (BST), Barcelona, Catalonia, Spain. .,Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR) and Universitat Autònoma de Barcelona (UAB), Barcelona, Catalonia, Spain.
| | - Marta Monguió-Tortajada
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Catalonia, Spain
| | - Ana Gámez-Valero
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Catalonia, Spain
| | - Raquel Rojas-Márquez
- Cell Therapy Service, Blood and Tissue Bank (BST), Barcelona, Catalonia, Spain.,Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona, Catalonia, Spain
| | - Francesc Enric Borràs
- REMAR-IVECAT Group, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Catalonia, Spain.,Nephrology Service, Germans Trias i Pujol University Hospital, Badalona, Catalonia, Spain
| | - Santiago Roura
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Badalona, Catalonia, Spain
| | - Joaquim Vives
- Cell Therapy Service, Blood and Tissue Bank (BST), Barcelona, Catalonia, Spain. .,Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR) and Universitat Autònoma de Barcelona (UAB), Barcelona, Catalonia, Spain. .,Medicine Department, Universitat Autònoma de Barcelona (UAB), Badalona, Catalonia, Spain.
| |
Collapse
|
37
|
Liu Y, Li M, Yin Z, Zhou S, Qiu Y. SUMO-modified bone marrow mesenchymal stem cells promoted the repair of articular cartilage in rats. Cell Biol Int 2019; 44:560-568. [PMID: 31642552 DOI: 10.1002/cbin.11256] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/19/2019] [Indexed: 12/25/2022]
Abstract
Articular cartilage damage can lead to joint deformity, pain, and severe dysfunction. However, due to the lack of blood vessels and nerves in articular cartilage, the self-healing capacity of damaged cartilage is limited. In this study, we overexpressed small ubiquitin-like modifier (SUMO)1, SUMO2/3, and SUMO1/2/3 in bone marrow mesenchymal stem cells (BMSCs). Then, these cells were inoculated on surfaces of different hardness, and their differentiation into chondrocytes, hypoxic tolerance ability, and inflammatory response was detected. Finally, BMSCs were transplanted into the injured knee joint cavity of the rats, and the repair was evaluated. We found that BMSCs overexpressing SUMO1 were more likely to differentiate into articular cartilage along with the hardness of the surface, while BMSCs overexpressing SUMO2/3 could reduce inflammation response and improve the damaged cartilage microenvironment. In the rat model, BMSCs overexpressing SUMO1/2/3 transplanted on injured articular cartilage surface showed better survival, less inflammatory response, and improved tissue repair capability. In conclusion, BMSCs overexpressing SUMO are more tolerant to hypoxia conditions, and have stronger repair ability for damaged chondrocytes in vitro and for articular cartilage injury model in rats, and are excellent seed cells for repairing articular cartilage.
Collapse
Affiliation(s)
- Ying Liu
- The Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.,The Department of Orthopedics, Affiliated Hospital, Binzhou Medical University, Binzhou, 256603, China
| | - Meng Li
- The Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhanhai Yin
- The Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shuangli Zhou
- The Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yusheng Qiu
- The Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
38
|
Grier WK, Sun Han Chang RA, Ramsey MD, Harley BA. The influence of cyclic tensile strain on multi-compartment collagen-GAG scaffolds for tendon-bone junction repair. Connect Tissue Res 2019; 60:530-543. [PMID: 31007094 PMCID: PMC6706312 DOI: 10.1080/03008207.2019.1601183] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Background: Orthopedic injuries often occur at the interface between soft tissues and bone. The tendon-bone junction (TBJ) is a classic example of such an interface. Current clinical strategies for TBJ injuries prioritize mechanical reattachment over regeneration of the native interface, resulting in poor outcomes. The need to promote regenerative healing of spatially-graded tissues inspires our effort to develop new tissue engineering technologies that replicate features of the spatially-graded extracellular matrix and strain profiles across the native TBJ. Materials and Methods: We recently described a biphasic collagen-glycosaminoglycan (CG) scaffold containing distinct compartment with divergent mineral content and structural alignment (isotropic vs. anisotropic) linked by a continuous interface zone to mimic structural and compositional features of the native TBJ. Results: Here, we report application of cyclic tensile strain (CTS) to the scaffold via a bioreactor leads to non-uniform strain profiles across the spatially-graded scaffold. Further, combinations of CTS and matrix structural features promote rapid, spatially-distinct differentiation profiles of human bone marrow-derived mesenchymal stem cells (MSCs) down multiple osteotendinous lineages. CTS preferentially upregulates MSC activity and tenogenic differentiation in the anisotropic region of the scaffold. This work demonstrates a tissue engineering approach that couples instructive biomaterials with cyclic tensile stimuli to promote regenerative healing of orthopedic interfaces.
Collapse
Affiliation(s)
- William K. Grier
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Raul A. Sun Han Chang
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Matthew D. Ramsey
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Brendan A.C. Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
39
|
Vakharia RM, Roche MW, Alcerro JC, Lavernia CJ. The Current Status of Cell-Based Therapies for Primary Knee Osteoarthritis. Orthop Clin North Am 2019; 50:415-423. [PMID: 31466658 DOI: 10.1016/j.ocl.2019.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
There is a growing interest in cell therapy for knee osteoarthritis. This study systematically reviews the current status of cell-based therapies. The authors review treatment modalities, clinical outcomes, and the economics of cell therapy. Inclusion criteria were articles containing cellular therapy, platelet-rich plasma, and knee osteoarthritis in the title. Letters, editorial material, abstracts not published, and manuscripts with incomplete data were excluded. Forty-two articles met these inclusion criteria and were critically reviewed. Cell-based therapy holds promise as a means of restoring deficient local cartilage cell populations. There is no evidence-based information for the use of cell-based therapies in knee osteoarthritis.
Collapse
Affiliation(s)
- Rushabh M Vakharia
- Holy Cross Hospital, Orthopedic Research Institute, 5597 North Dixie Highway, Fort Lauderdale, FL 33308, USA
| | - Martin W Roche
- Holy Cross Hospital, Orthopedic Research Institute, 5597 North Dixie Highway, Fort Lauderdale, FL 33308, USA
| | - Jose Carlos Alcerro
- Orthopedic Surgery, Adult Joint Reconstruction, Instituto Hondureño de Seguridad Social, Tegucigalpa, Honduras
| | - Carlos Jesus Lavernia
- Orthopedic Surgery, Adult Joint Reconstruction, Arthritis Surgery Research Foundation, Coral Gables, FL, USA.
| |
Collapse
|
40
|
Reparative and Regenerative Effects of Mesenchymal Stromal Cells-Promising Potential for Kidney Transplantation? Int J Mol Sci 2019; 20:ijms20184614. [PMID: 31540361 PMCID: PMC6770554 DOI: 10.3390/ijms20184614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/15/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) possess reparative, regenerative and immunomodulatory properties. The current literature suggests that MSCs could improve kidney transplant outcome via immunomodulation. In many clinical domains, research has also focussed on the regenerative and reparative effects of therapies with MSCs. However, in the field of transplantation, data on this subject remain scarce. This review provides an overview of what is known about the regenerative and reparative effects of MSCs in various fields ranging from wound care to fracture healing and also examines the potential of these promising MSC properties to improve the outcome of kidney transplantations.
Collapse
|
41
|
Affiliation(s)
- Gun-Il Im
- 1Integrative Research Institute for Regenerative Medical Engineering, Dongguk University, 27 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326 Republic of Korea
| | - Chong-Su Cho
- 2Research Institute of Agriculture and Life Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 Republic of Korea
| |
Collapse
|
42
|
Guilak F, Pferdehirt L, Ross AK, Choi YR, Collins KH, Nims RJ, Katz DB, Klimak M, Tabbaa S, Pham CT. Designer Stem Cells: Genome Engineering and the Next Generation of Cell-Based Therapies. J Orthop Res 2019; 37:1287-1293. [PMID: 30977548 PMCID: PMC6546536 DOI: 10.1002/jor.24304] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 02/04/2023]
Abstract
Stem cells provide tremendous promise for the development of new therapeutic approaches for musculoskeletal conditions. In addition to their multipotency, certain types of stem cells exhibit immunomodulatory effects that can mitigate inflammation and enhance tissue repair. However, the translation of stem cell therapies to clinical practice has proven difficult due to challenges in intradonor and interdonor variability, engraftment, variability in recipient microenvironment and patient indications, and limited therapeutic biological activity. In this regard, the success of stem cell-based therapies may benefit from cellular engineering approaches to enhance factors such as purification, homing and cell survival, trophic effects, or immunomodulatory signaling. By combining recent advances in gene editing, synthetic biology, and tissue engineering, the potential exists to create new classes of "designer" cells that have prescribed cell-surface molecules and receptors as well as synthetic gene circuits that provide for autoregulated drug delivery or enhanced tissue repair. Published by Wiley Periodicals, Inc. J Orthop Res 37:1287-1293, 2019.
Collapse
Affiliation(s)
- Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110,Correspondence: Farshid Guilak, Ph.D. Center of Regenerative Medicine, Washington University, St. Louis, Campus Box 8233, McKinley Research Bldg, Room 3121, St. Louis, MO 63110-1624.
| | - Lara Pferdehirt
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Alison K. Ross
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Yun-Rak Choi
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110
| | - Robert J. Nims
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110
| | - Dakota B. Katz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Molly Klimak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | | | - Christine T.N. Pham
- Division of Rheumatology, Department of Medicine, Washington University in St. Louis, MO, 63110
| |
Collapse
|
43
|
CORR® International - Asia-Pacific: Stem Cell-based Treatments in Orthopaedic Clinical Practice-Is it Ready For Primetime in the Asia-Pacific Region? Clin Orthop Relat Res 2019; 477:695-697. [PMID: 30829690 PMCID: PMC6437390 DOI: 10.1097/corr.0000000000000697] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
44
|
Dewey MJ, Johnson EM, Weisgerber DW, Wheeler MB, Harley BAC. Shape-fitting collagen-PLA composite promotes osteogenic differentiation of porcine adipose stem cells. J Mech Behav Biomed Mater 2019; 95:21-33. [PMID: 30953806 DOI: 10.1016/j.jmbbm.2019.03.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/17/2018] [Accepted: 03/17/2019] [Indexed: 10/27/2022]
Abstract
Craniomaxillofacial bone defects can occur as a result of congenital, post-oncologic, and high-energy impact conditions. The scale and irregularity of such defects motivate new biomaterials to promote regeneration of the damaged bone. We have recently described a mineralized collagen scaffold capable of instructing stem cell osteogenic differentiation and new bone infill in the absence of traditional osteogenic supplements. Herein, we report the integration of a millimeter-scale reinforcing poly (lactic acid) frame fabricated via 3D-printing into the mineralized collagen scaffold with micron-scale porosity to form a multi-scale mineralized collagen-PLA composite. We describe modifications to the PLA frame design to increase the compressive strength (Young's Modulus, ultimate stress and strain) of the composite. A critical challenge beyond increasing the compressive strength of the collagen scaffold is addressing challenges inherent with the irregularity of clinical defects. As a result, we examined the potential for modifying the frame architecture to render the composite with increased compressive strength in one axis or radial compressibility and shape-fitting capacity in an orthogonal axis. A library of mineralized collagen-PLA composites was mechanically characterized via compression testing and push-out test to describe mechanical performance and shape-fitting capacity. We also report in vitro comparison of the bioactivity of porcine adipose derived stem cells in the mineralized collagen-PLA composite versus the mineralized collagen scaffold via metabolic activity, gene expression, and functional matrix synthesis. The results suggest that incorporation of the PLA reinforcing frame does not negatively influence the osteoinductive nature of the mineralized collagen scaffold. Together, these findings suggest a strategy to address often competing bioactivity, mechanical strength, and shape-fitting design requirements for biomaterials for craniomaxillofacial bone regeneration.
Collapse
Affiliation(s)
- Marley J Dewey
- Dept. of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Eileen M Johnson
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Daniel W Weisgerber
- Dept. of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Matthew B Wheeler
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Brendan A C Harley
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Dept. of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
45
|
Wang-Yang L, You-Liang Z, Tiao L, Peng Z, Wu-Ji X, Xiao-Long L, Xin-Yu Q, Hui X. Pretreatment with Lithospermic Acid Attenuates Oxidative Stress- induced Apoptosis in Bone Marrow-derived Mesenchymal Stem Cells via Anti-oxidation and Activation of PI3K/Akt Pathway. DIGITAL CHINESE MEDICINE 2019. [DOI: 10.1016/j.dcmed.2019.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
46
|
Qi H, Liu DP, Xiao DW, Tian DC, Su YW, Jin SF. Exosomes derived from mesenchymal stem cells inhibit mitochondrial dysfunction-induced apoptosis of chondrocytes via p38, ERK, and Akt pathways. In Vitro Cell Dev Biol Anim 2019; 55:203-210. [PMID: 30783864 DOI: 10.1007/s11626-019-00330-x] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/28/2019] [Indexed: 12/18/2022]
Abstract
Osteoarthritis (OA) is the most common chronic joint disease worldwide. Chondrocyte, as the only resident cell type in cartilage, its apoptosis is of pathogenetic significance in OA. Mesenchymal stem cell (MSC)-based-therapy has been proved effective in OA in animals and clinical studies. Nowadays, the regenerative potential of MSC-based therapy is mostly attributed to its paracrine secretion, in which exosomes may play an important role. In the present study, we aimed to find out the significance of MSC-derived exosomes (MSC-Exos) on the viability of chondrocytes under normal and inflammatory conditions. Bone marrow MSCs (BMSCs) and chondrocytes from rabbits were cultured in vitro. BMSC-Exos were isolated by an ultracentrifugation method. Transmission electron microscopy and Western blot were used to identify exosomes. The internalization of BMSC-Exos into chondrocytes was observed by fluorescent microscope. The viability and apoptosis of chondrocytes induced by IL-1β were tested through MTT method, Hoechst33324 dying, and mitochondrial damage measurement. Phosphorylation of p38, ERK, and Akt were evaluated by Western blot. The results showed that BMSC-Exos were round-shaped. Co-culturing BMSC-Exos with chondrocytes could observe the uptake of BMSC-Exos by chondrocytes. The viability decreased, apoptosis occurred, and the mitochondrial membrane potential of chondrocytes changed a lot when IL-1β were given, but all the changes were almost abolished when BMSC-Exos was added. Furthermore, the phosphorylation of p38 and ERK were inhibited, and phosphorylation of Akt was promoted by BMSC-Exos compared with IL-1β group. The present study demonstrated that BMSC-Exos inhibited mitochondrial-induced apoptosis in response to IL-1β, and p38, ERK, and Akt pathways were involved. BMSC-Exo might represent a novel cell-free therapeutic approach for the treatment of OA.
Collapse
Affiliation(s)
- Hui Qi
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, China.
| | - Dan-Ping Liu
- Department of Orthopaedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Da-Wei Xiao
- Department of Orthopaedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Da-Chuan Tian
- Department of Orthopaedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Yong-Wei Su
- Department of Orthopaedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Shao-Feng Jin
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, China
| |
Collapse
|
47
|
Perspective on Intra-articular Injection Cell Therapy for Osteoarthritis Treatment. Tissue Eng Regen Med 2019; 16:357-363. [PMID: 31413940 DOI: 10.1007/s13770-018-00176-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/13/2018] [Accepted: 12/24/2018] [Indexed: 01/05/2023] Open
Abstract
Background Osteoarthritis (OA), the most common arthritis, is one of the most frequently encountered orthopaedic conditions. As a small number of large joints such as knee and hip are affected in OA, OA is an ideal target for local therapy. Although corticosteroid and hyaluronic acid have been traditionally used for joints through intra-articular (IA) injection, IA injection also provides a minimally invasive route to apply cell therapy to treat OA. IA cell therapy has drawn attention because it may provide regeneration of articular cartilage in addition to palliative anti-inflammatory effects. Methods Current progress of IA injection therapy and the author's perspective on this issue are described narratively. Results It is too premature to have any conclusion on the eventual efficacy of IA cell therapy concerning regeneration of articular cartilage based on current data. Prospective radiological and histological data from larger numbers of patients are needed to prove cost effectiveness of IA cell therapy. Conclusions Expanding research in this field will produce further evidences to provide guidance on the eventual effectiveness of IA cell therapy in the future.
Collapse
|
48
|
Zhang H, Wang ZJ, Wang LJ, Li TT, He S, Li LP, Li XY, Liu SJ, Li JD, Li SJ, Zhang RP. A dual-mode nanoparticle based on natural biomaterials for photoacoustic and magnetic resonance imaging of bone mesenchymal stem cells in vivo. RSC Adv 2019; 9:35003-35010. [PMID: 35530687 PMCID: PMC9074133 DOI: 10.1039/c9ra05937g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022] Open
Abstract
Stem cell imaging in vivo is critical to elucidate the homing, distribution, survival, and repair mechanisms and to evaluate the therapeutic effects of engrafted stem cells. Unfortunately, unimodal imaging of stem cells does not simultaneously satisfy all current requirements owing to their intrinsic limitations. Obviously, bimodal or multimodal imaging of stem cells is a promising strategy for circumventing this issue. This study aimed to design and synthesize a novel dual-modal polyethylene glycol-modified magnetic nanoparticle (Fe3+-PEG-MNP) based on natural biomaterials including melanin and Fe ions for photoacoustic (PA) and magnetic resonance (MR) imaging of stem cells in vivo. The Fe3+-PEG-MNPs were characterized and their PA/MR imaging capability and cytotoxicity were evaluated. Bone marrow mesenchymal stem cells (BM-MSCs) labeled with Fe3+-PEG-MNPs were subjected to PA and MR imaging in vitro and in vivo. Consequently, Fe3+-PEG-MNPs displayed many superior properties, including ultra-small particle size, higher stability, water solubility, easy labeling of cells, lower cytotoxicity, high biosafety, excellent capability of PA/MR imaging, high sensitivity and long-term monitoring in vitro and in vivo. In particular, PA and MR signals of labeled BM-MSCs were maintained for at least 35 and 28 d, respectively, in vivo. Therefore, Fe3+-PEG-MNPs are ideal dual-modal PA/MR nanoparticles for non-invasive and effective monitoring of engrafted stem cells in vivo. Stem cell imaging in vivo is critical to elucidate the homing, distribution, survival, and repair mechanisms and to evaluate the therapeutic effects of engrafted stem cells.![]()
Collapse
|
49
|
Lee E, Ko JY, Kim J, Park JW, Lee S, Im GI. Osteogenesis and angiogenesis are simultaneously enhanced in BMP2-/VEGF-transfected adipose stem cells through activation of the YAP/TAZ signaling pathway. Biomater Sci 2019; 7:4588-4602. [DOI: 10.1039/c9bm01037h] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
While bone has the capability to heal itself, there is a great difficulty in reconstituting large bone defects created by heavy trauma or the resection of malignant tumors.
Collapse
Affiliation(s)
- Eugene Lee
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
- Department of Orthopaedics
| | - Ji-Yun Ko
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Juyoung Kim
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Jeong-Won Park
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Songhee Lee
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
| | - Gun-Il Im
- Research Institute for Integrative Regenerative Biomedical Engineering
- Dongguk University
- Goyang 10326
- Republic of Korea
- Department of Orthopaedics
| |
Collapse
|
50
|
Osteogenesis of Multipotent Progenitor Cells using the Epigallocatechin Gallate-Modified Gelatin Sponge Scaffold in the Rat Congenital Cleft-Jaw Model. Int J Mol Sci 2018; 19:ijms19123803. [PMID: 30501071 PMCID: PMC6320852 DOI: 10.3390/ijms19123803] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/25/2018] [Accepted: 11/26/2018] [Indexed: 12/21/2022] Open
Abstract
Cost-effective and functionalized scaffolds are in high demand for stem-cell-based regenerative medicine to treat refractory bone defects in craniofacial abnormalities and injuries. One potential strategy is to utilize pharmacological and cost-effective plant polyphenols and biocompatible proteins, such as gelatin. Nevertheless, the use of chemically modified proteins with plant polyphenols in this strategy has not been standardized. Here, we demonstrated that gelatin chemically modified with epigallocatechin gallate (EGCG), the major catechin isolated from green tea, can be a useful material to induce bone regeneration in a rat congenial cleft-jaw model in vivo when used with/without adipose-derived stem cells or dedifferentiated fat cells. Vacuum-heated gelatin sponges modified with EGCG (vhEGCG-GS) induced superior osteogenesis from these two cell types compared with vacuum-heated gelatin sponges (vhGS). The EGCG-modification converted the water wettability of vhGS to a hydrophilic property (contact angle: 110° to 3.8°) and the zeta potential to a negative surface charge; the modification enhanced the cell adhesion property and promoted calcium phosphate precipitation. These results suggest that the EGCG-modification with chemical synthesis can be a useful platform to modify the physicochemical property of gelatin. This alteration is likely to provide a preferable microenvironment for multipotent progenitor cells, inducing superior bone formation in vivo.
Collapse
|