1
|
Crossman J, Lai H, Kulka M, Jomha N, Flood P, El-Bialy T. The Effect of Low-Intensity Pulsed Ultrasound on Temporomandibular Joint Arthritis in Juvenile Rats. Tissue Eng Part A 2024; 30:740-751. [PMID: 38517092 DOI: 10.1089/ten.tea.2024.0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024] Open
Abstract
Juvenile idiopathic arthritis is an inflammatory disease that can affect the temporomandibular joint (TMJ) and lower jaw growth. Better treatment options are needed, so this study investigated the effect of low-intensity pulsed ultrasound (LIPUS) on TMJ arthritis. Seventy-two 3-week-old male Wistar rats were in vivo microcomputed tomography (micro-CT) scanned and divided into eight groups (n = 9). These groups were Group 1-TMJ arthritis and immediate LIPUS treatment (20 min/day, 4 weeks); Group 2-immediate LIPUS treatment and no TMJ arthritis; Group 3-TMJ arthritis and no LIPUS; Group 4-no TMJ arthritis and no LIPUS; Group 5-TMJ arthritis and LIPUS treatment with a delayed start by 4 weeks; Group 6-Delayed LIPUS and no TMJ arthritis; Group 7-TMJ arthritis and no (delayed) LIPUS; and Group 8-no TMJ arthritis and no (delayed) LIPUS. Ex vivo micro-CT scanning was completed, and samples were prepared for tissue analysis. Synovitis was observed in the TMJ arthritis (collagen-induced arthritis [CIA]) groups, but the severity appeared greater in the groups without LIPUS treatment. Fibrocartilage and hypertrophic cell layer thicknesses in the CIA group without LIPUS treatment were significantly greater (p < 0.05). Proteoglycan staining appeared greater in the LIPUS groups. Immediate LIPUS treatment increased the expression of type II collagen, type X collagen, and transforming growth factor-beta 1 (TGF-β1) immunostaining, and CIA (no LIPUS) increased MMP-13, vascular endothelial growth factor, and interleukin-1 beta (IL-1β) immunostaining. LIPUS treatment prevented growth disturbances observed in the CIA groups (no LIPUS) (p < 0.005). Our results have contributed to the understanding of the uses and limitations of the CIA juvenile rat model and have demonstrated the effects of LIPUS on the TMJ and mandibular growth. This information will help in designing future studies for investigating LIPUS and TMJ arthritis, leading to the development of new treatment options for children with juvenile arthritis in their TMJs.
Collapse
Affiliation(s)
- Jacqueline Crossman
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Hollis Lai
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Marianna Kulka
- Department of Medical Microbiology & Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Quantum and Nanotechnologies Research Center, The National Research Council Canada, Ontario, Canada
| | - Nadr Jomha
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Patrick Flood
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Tarek El-Bialy
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
2
|
Dogru S, Alba GM, Pierce KC, Wang T, Kia DS, Albro MB. Cell mediated reactions create TGF-β delivery limitations in engineered cartilage. Acta Biomater 2024; 190:178-190. [PMID: 39447669 PMCID: PMC11614674 DOI: 10.1016/j.actbio.2024.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
During native cartilage development, endogenous TGF-β activity is tightly regulated by cell-mediated chemical reactions in the extracellular milieu (e.g., matrix and receptor binding), providing spatiotemporal control in a manner that is localized and short acting. These regulatory paradigms appear to be at odds with TGF-β delivery needs in tissue engineering (TE) where administered TGF-β is required to transport long distances or reside in tissues for extended durations. In this study, we perform a novel examination of the influence of cell-mediated reactions on the spatiotemporal distribution of administered TGF-β in cartilage TE applications. Reaction rates of TGF-β binding to cell-deposited ECM and TGF-β internalization by cell receptors are experimentally characterized in bovine chondrocyte-seeded tissue constructs. TGF-β binding to the construct ECM exhibits non-linear Brunauer-Emmett-Teller (BET) adsorption behavior, indicating that as many as seven TGF-β molecules can aggregate at a binding site. Cell-mediated TGF-β internalization rates exhibit a biphasic trend, following a Michaelis-Menten relation (Vmax = 2.4 molecules cell-1 s-1, Km = 1.7 ng mL-1) at low ligand doses (≤130 ng/mL), but exhibit an unanticipated non-saturating power trend at higher doses (≥130 ng/mL). Computational models are developed to illustrate the influence of these reactions on TGF-β spatiotemporal delivery profiles for conventional TGF-β administration platforms. For TGF-β delivery via supplementation in culture medium, these reactions give rise to pronounced steady state TGF-β spatial gradients; TGF-β concentration decays by ∼90 % at a depth of only 500 μm from the media-exposed surface. For TGF-β delivery via heparin-conjugated affinity scaffolds, cell mediated internalization reactions significantly reduce the TGF-β scaffold retention time (160-360-fold reduction) relative to acellular heparin scaffolds. This work establishes the significant limitations that cell-mediated chemical reactions engender for TGF-β delivery and highlights the need for novel delivery platforms that account for these reactions to achieve optimal TGF-β exposure profiles. STATEMENT OF SIGNIFICANCE: During native cartilage development, endogenous TGF-β activity is tightly regulated by cell-mediated chemical reactions in the extracellular milieu (e.g., matrix and receptor binding), providing spatiotemporal control in a manner that is localized and short acting. However, the effect of these reactions on the delivery of exogenous TGF-β to engineered cartilage tissues remains not well understood. In this study, we demonstrate that cell-mediated reactions significantly restrict the delivery of TGF-β to cells in engineered cartilage tissue constructs. For delivery via media supplementation, reactions significantly limit TGF-β penetration into constructs. For delivery via scaffold loading, reactions significantly limit TGF-β residence time in constructs. Overall, these results illustrate the impact of cell-mediated chemical reactions on TGF-β delivery profiles and support the importance of accounting for these reactions when designing TGF-β delivery platforms for promoting cartilage regeneration.
Collapse
Affiliation(s)
- Sedat Dogru
- College of Engineering, Boston University, Boston, MA, United States
| | - Gabriela M Alba
- College of Engineering, Boston University, Boston, MA, United States
| | - Kirk C Pierce
- College of Engineering, Boston University, Boston, MA, United States
| | - Tianbai Wang
- College of Engineering, Boston University, Boston, MA, United States
| | | | - Michael B Albro
- College of Engineering, Boston University, Boston, MA, United States.
| |
Collapse
|
3
|
Semitela A, Marques PAAP, Completo A. Strategies to engineer articular cartilage with biomimetic zonal features: a review. Biomater Sci 2024; 12:5961-6005. [PMID: 39463257 DOI: 10.1039/d4bm00579a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Articular cartilage (AC) is a highly specialized tissue with restricted ability for self-regeneration, given its avascular and acellular nature. Although a considerable number of surgical treatments is available for the repair, reconstruction, and regeneration of AC defects, most of them do not prioritize the development of engineered cartilage with zonal stratification derived from biomimetic biochemical, biomechanical and topographic cues. In the absence of these zonal elements, engineered cartilage will exhibit increased susceptibility to failure and will neither be able to withstand the mechanical loading to which AC is subjected nor will it integrate well with the surrounding tissue. In this regard, new breakthroughs in the development of hierarchical stratified engineered cartilage are highly sought after. Initially, this review provides a comprehensive analysis of the composition and zonal organization of AC, aiming to enhance our understanding of the significance of the structure of AC for its function. Next, we direct our attention towards the existing in vitro and in vivo studies that introduce zonal elements in engineered cartilage to elicit appropriate AC regeneration by employing tissue engineering strategies. Finally, the advantages, challenges, and future perspectives of these approaches are presented.
Collapse
Affiliation(s)
- Angela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Paula A A P Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
4
|
Dinesh NEH, Baratang N, Rosseau J, Mohapatra R, Li L, Mahalingam R, Tiedemann K, Campeau PM, Reinhardt DP. Fibronectin isoforms promote postnatal skeletal development. Matrix Biol 2024; 133:86-102. [PMID: 39159790 DOI: 10.1016/j.matbio.2024.08.002] [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: 04/11/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024]
Abstract
Fibronectin (FN) is a ubiquitous extracellular matrix glycoprotein essential for the development of various tissues. Mutations in FN cause a unique form of spondylometaphyseal dysplasia, emphasizing its importance in cartilage and bone development. However, the relevance and functional role of FN during skeletal development has remained elusive. To address these aspects, we have generated conditional knockout mouse models targeting the cellular FN isoform in cartilage (cFNKO), the plasma FN isoform in hepatocytes (pFNKO), and both isoforms together in a double knockout (FNdKO). We used these mice to determine the relevance of the two principal FN isoforms in skeletal development from postnatal day one to the adult stage at two months. We identified a distinct topological FN deposition pattern in the mouse limb during different gestational and postnatal skeletal development phases, with prominent levels at the resting and hypertrophic chondrocyte zones and in the trabecular bone. Cartilage-specific cFN emerged as the predominant isoform in the growth plate, whereas circulating pFN remained excluded from the growth plate and confined to the primary and secondary ossification centers. Deleting either isoform independently (cFNKO or pFNKO) yielded only relatively subtle changes in the analyzed skeletal parameters. However, the double knockout of cFN in the growth plate and pFN in the circulation of the FNdKO mice significantly reduced postnatal body weight, body length, and bone length. Micro-CT analysis of the adult bone microarchitecture in FNdKO mice exposed substantial reductions in trabecular bone parameters and bone mineral density. The mice also showed elevated bone marrow adiposity. Analysis of chondrogenesis in FNdKO mice demonstrated changes in the resting, proliferating and hypertrophic growth plate zones, consistent alterations in chondrogenic markers such as collagen type II and X, decreased apoptosis of hypertrophic chondrocytes, and downregulation of bone formation markers. Transforming growth factor-β1 and downstream phospho-AKT levels were significantly lower in the FNdKO than in the control mice, revealing a crucial FN-mediated regulatory pathway in chondrogenesis and bone formation. In conclusion, the data demonstrate that FN is essential for chondrogenesis and bone development. Even though cFN and pFN act in different regions of the bone, both FN isoforms are required for the regulation of chondrogenesis, cartilage maturation, trabecular bone formation, and overall skeletal growth.
Collapse
Affiliation(s)
- Neha E H Dinesh
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | | | | | - Ronit Mohapatra
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Ling Li
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Ramshaa Mahalingam
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | | | | | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
5
|
Ao R, Liang W, Wang Z, Li Q, Pan X, Zhen Y, An Y. Delivery Strategies of Growth Factors in Cartilage Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39345121 DOI: 10.1089/ten.teb.2024.0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Cartilage plays an important role in supporting soft tissues, reducing joint friction, and distributing pressure. However, its self-repair capacity is limited due to the lack of blood vessels, nerves, and lymphatic systems. Tissue engineering offers a potential solution to promote cartilage regeneration by combining scaffolds, seed cells, and growth factors. Among these, growth factors play a critical role in regulating cell proliferation, differentiation, and extracellular matrix remodeling. However, their instability, susceptibility to degradation and potential side effects limit their effectiveness. This article reviews the main growth factors used in cartilage tissue engineering and their delivery strategies, including affinity-based delivery, carrier-assisted delivery, stimuli-responsive delivery, spatial structure-based delivery, and cell system-based delivery. Each method shows unique advantages in enhancing the delivery efficiency and specificity of growth factors but also faces challenges such as cost, biocompatibility, and safety. Future research needs to further optimize these strategies to achieve more efficient, safe, and economical delivery of growth factors, thereby advancing the clinical application of cartilage tissue engineering.
Collapse
Affiliation(s)
- Rigele Ao
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Wei Liang
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Zimo Wang
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Qiaoyu Li
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Xingyi Pan
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Yonghuan Zhen
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| |
Collapse
|
6
|
Dinesh NEH, Rousseau J, Mosher DF, Strauss M, Mui J, Campeau PM, Reinhardt DP. Mutations in fibronectin dysregulate chondrogenesis in skeletal dysplasia. Cell Mol Life Sci 2024; 81:419. [PMID: 39367925 PMCID: PMC11456097 DOI: 10.1007/s00018-024-05444-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/22/2024] [Accepted: 09/06/2024] [Indexed: 10/07/2024]
Abstract
Fibronectin (FN) is an extracellular matrix glycoprotein essential for the development and function of major vertebrate organ systems. Mutations in FN result in an autosomal dominant skeletal dysplasia termed corner fracture-type spondylometaphyseal dysplasia (SMDCF). The precise pathomechanisms through which mutant FN induces impaired skeletal development remain elusive. Here, we have generated patient-derived induced pluripotent stem cells as a cell culture model for SMDCF to investigate the consequences of FN mutations on mesenchymal stem cells (MSCs) and their differentiation into cartilage-producing chondrocytes. In line with our previous data, FN mutations disrupted protein secretion from MSCs, causing a notable increase in intracellular FN and a significant decrease in extracellular FN levels. Analyses of plasma samples from SMDCF patients also showed reduced FN in circulation. FN and endoplasmic reticulum (ER) protein folding chaperones (BIP, HSP47) accumulated in MSCs within ribosome-covered cytosolic vesicles that emerged from the ER. Massive amounts of these vesicles were not cleared from the cytosol, and a smaller subset showed the presence of lysosomal markers. The accumulation of intracellular FN and ER proteins elevated cellular stress markers and altered mitochondrial structure. Bulk RNA sequencing revealed a specific transcriptomic dysregulation of the patient-derived cells relative to controls. Analysis of MSC differentiation into chondrocytes showed impaired mesenchymal condensation, reduced chondrogenic markers, and compromised cell proliferation in mutant cells. Moreover, FN mutant cells exhibited significantly lower transforming growth factor beta-1 (TGFβ1) expression, crucial for mesenchymal condensation. Exogenous FN or TGFβ1 supplementation effectively improved the MSC condensation and promoted chondrogenesis in FN mutant cells. These findings demonstrate the cellular consequences of FN mutations in SMDCF and explain the molecular pathways involved in the associated altered chondrogenesis.
Collapse
Affiliation(s)
- Neha E H Dinesh
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montreal, QC, Canada
| | - Justine Rousseau
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Deane F Mosher
- Departments of Biomolecular Chemistry and Medicine, University of Wisconsin, Madison, WI, USA
| | - Mike Strauss
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montreal, QC, Canada
| | - Jeannie Mui
- Facility for Electron Microscopy Research of McGill University, Montreal, QC, Canada
| | - Philippe M Campeau
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, QC, Canada
| | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montreal, QC, Canada.
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
7
|
Pande S, Pati F, Chakraborty P. Harnessing Peptide-Based Hydrogels for Enhanced Cartilage Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:5885-5905. [PMID: 39159490 DOI: 10.1021/acsabm.4c00879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Cartilage tissue engineering remains a formidable challenge due to its complex, avascular structure and limited regenerative capacity. Traditional approaches, such as microfracture, autografts, and stem cell delivery, often fail to restore functional tissue adequately. Recently, there has been a surge in the exploration of new materials that mimic the extracellular microenvironment necessary to guide tissue regeneration. This review investigates the potential of peptide-based hydrogels as an innovative solution for cartilage regeneration. These hydrogels, formed via supramolecular self-assembly, exhibit excellent properties, including biocompatibility, ECM mimicry, and controlled biodegradation, making them highly suitable for cartilage tissue engineering. This review explains the structure of cartilage and the principles of supramolecular and peptide hydrogels. It also delves into their specific properties relevant to cartilage regeneration. Additionally, this review presents recent examples and a comparative analysis of various peptide-based hydrogels used for cartilage regeneration. The review also addresses the translational challenges of these materials, highlighting regulatory hurdles and the complexities of clinical application. This comprehensive investigation provides valuable insights for biomedical researchers, tissue engineers, and clinical professionals aiming to enhance cartilage repair methodologies.
Collapse
Affiliation(s)
- Shreya Pande
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| | - Priyadarshi Chakraborty
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy 502284, Telangana, India
| |
Collapse
|
8
|
Yuan SC, Álvarez Z, Lee SR, Pavlović RZ, Yuan C, Singer E, Weigand SJ, Palmer LC, Stupp SI. Supramolecular Motion Enables Chondrogenic Bioactivity of a Cyclic Peptide Mimetic of Transforming Growth Factor-β1. J Am Chem Soc 2024; 146:21555-21567. [PMID: 39054767 DOI: 10.1021/jacs.4c05170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Transforming growth factor (TGF)-β1 is a multifunctional protein that is essential in many cellular processes that include fibrosis, inflammation, chondrogenesis, and cartilage repair. In particular, cartilage repair is important to avoid physical disability since this tissue does not have the inherent capacity to regenerate beyond full development. We report here on supramolecular coassemblies of two peptide amphiphile molecules, one containing a TGF-β1 mimetic peptide, and another which is one of two constitutional isomers lacking bioactivity. Using human articular chondrocytes, we investigated the bioactivity of the supramolecular copolymers of each isomer displaying either the previously reported linear form of the mimetic peptide or a novel cyclic analogue. Based on fluorescence depolarization and 1H NMR spin-lattice relaxation times, we found that coassemblies containing the cyclic compound and the most dynamic isomer exhibited the highest intracellular TGF-β1 signaling and gene expression of cartilage extracellular matrix components. We conclude that control of supramolecular motion is emerging as an important factor in the binding of synthetic molecules to receptors that can be tuned through chemical structure.
Collapse
Affiliation(s)
- Shelby C Yuan
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois 60611, United States
| | - Zaida Álvarez
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois 60611, United States
- Department of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), Barcelona 08028, Spain
| | - Sieun Ruth Lee
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Radoslav Z Pavlović
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois 60611, United States
| | - Chunhua Yuan
- Campus Chemical Instrument Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ethan Singer
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Steven J Weigand
- DuPont-Northwestern-Dow Collaborative Access Team Synchrotron Research Center, Northwestern University, Advanced Photon Source/Argonne National Laboratory 432-A004, Argonne, Illinois 60439, United States
| | - Liam C Palmer
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois 60611, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Samuel I Stupp
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois 60611, United States
- Department of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
9
|
Soni N, Niranjane P, Purohit A. Role of Growth Factors in Nasal Cartilage Development and Molding: A Comprehensive Review. Cureus 2024; 16:e67202. [PMID: 39295663 PMCID: PMC11409944 DOI: 10.7759/cureus.67202] [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: 07/28/2024] [Accepted: 08/19/2024] [Indexed: 09/21/2024] Open
Abstract
This review aims to investigate the properties of growth factors concerning the morphogenesis and development of nasal cartilage, which is fundamentally important for facial form and appearance. Since cartilage lacks a blood supply, it is more difficult to regenerate, as cartilage tissue obtains sustenance by diffusion. Cytokines are signalling molecules that control chondrocyte metabolism and extracellular matrix formation, which is required for cartilage development, homeostasis, and healing. Some craniofacial illnesses alter the composition of the cartilage and the structural organization of growth factors, allowing for moulding. TGF-β (transforming growth factor-β) encourages chondrocyte differentiation, whereas IGF-1 (insulin-like growth factor-1) stimulates cartilage-forming collagen synthesis and chondrocyte multiplication. We used the scoping review approach to present current research on the role of growth factors in the creation and architecture of nasal cartilage. We generally observed this structure before conducting specific experiments to determine the impact of growth agents on the development of chondrocytes and cartilage. Prominent findings increase our understanding of how growth factors influence the extracellular matrix, cell activities and features, and cartilage growth rate; all are critical for cartilage tissue development and repair. Research into growth factors and their physiological interactions with cartilage may help improve treatment's functional and aesthetic outcomes and our understanding of the origins and consequences of nasal congenital anomalies. This study emphasizes the importance of expanding knowledge and experience, as well as the use of growth factors in clinical practice, to stimulate cartilage development.
Collapse
Affiliation(s)
- Nikita Soni
- Department of Orthodontics and Dentofacial Orthopaedics, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Priyanka Niranjane
- Department of Orthodontics and Dentofacial Orthopaedics, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Akanksha Purohit
- Neglected Tropical Diseases, Global Health Strategies, Delhi, IND
| |
Collapse
|
10
|
Liu K, Zhang B, Zhang X. Promoting Articular Cartilage Regeneration through Microenvironmental Regulation. J Immunol Res 2024; 2024:4751168. [PMID: 39104594 PMCID: PMC11300091 DOI: 10.1155/2024/4751168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/21/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
In recent years, as the aging population continues to grow, osteoarthritis (OA) has emerged as a leading cause of disability, with its incidence rising annually. Current treatments of OA include exercise and medications in the early stages and total joint replacement in the late stages. These approaches only relieve pain and reduce inflammation; however, they have significant side effects and high costs. Therefore, there is an urgent need to identify effective treatment methods that can delay the pathological progression of this condition. The changes in the articular cartilage microenvironment, which are complex and diverse, can aggravate the pathological progression into a vicious cycle, inhibiting the repair and regeneration of articular cartilage. Understanding these intricate changes in the microenvironment is crucial for devising effective treatment modalities. By searching relevant research articles and clinical trials in PubMed according to the keywords of articular cartilage, microenvironment, OA, mechanical force, hypoxia, cytokine, and cell senescence. This study first summarizes the factors affecting articular cartilage regeneration, then proposes corresponding treatment strategies, and finally points out the future research direction. We find that regulating the opening of mechanosensitive ion channels, regulating the expression of HIF-1, delivering growth factors, and clearing senescent cells can promote the formation of articular cartilage regeneration microenvironment. This study provides a new idea for the treatment of OA in the future, which can promote the regeneration of articular cartilage through the regulation of the microenvironment so as to achieve the purpose of treating OA.
Collapse
Affiliation(s)
- Kai Liu
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and MinistryGuangxi Medical University, Nanning, Guangxi 530021, China
| | - Bingjun Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiaoling Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and MinistryGuangxi Medical University, Nanning, Guangxi 530021, China
| |
Collapse
|
11
|
Roncada T, Blunn G, Roldo M. Collagen and Alginate Hydrogels Support Chondrocytes Redifferentiation In Vitro without Supplementation of Exogenous Growth Factors. ACS OMEGA 2024; 9:21388-21400. [PMID: 38764657 PMCID: PMC11097186 DOI: 10.1021/acsomega.4c01675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/21/2024]
Abstract
Focal cartilage defects are a prevalent knee problem affecting people of all ages. Articular cartilage (AC) possesses limited healing potential, and osteochondral defects can lead to pain and long-term complications such as osteoarthritis. Autologous chondrocyte implantation (ACI) has been a successful surgical approach for repairing osteochondral defects over the past two decades. However, a major drawback of ACI is the dedifferentiation of chondrocytes during their in vitro expansion. In this study, we isolated ovine chondrocytes and cultured them in a two-dimensional environment for ACI procedures. We hypothesized that 3D scaffolds would support the cells' redifferentiation without the need for growth factors so we encapsulated them into soft collagen and alginate (col/alg) hydrogels. Chondrocytes embedded into the hydrogels were viable and proliferated. After 7 days, they regained their original rounded morphology (aspect ratio 1.08) and started to aggregate. Gene expression studies showed an upregulation of COL2A1, FOXO3A, FOXO1, ACAN, and COL6A1 (37, 1.13, 22, 1123, and 1.08-fold change expression, respectively) as early as day one. At 21 days, chondrocytes had extensively colonized the hydrogel, forming large cell clusters. They started to replace the degrading scaffold by depositing collagen II and aggrecan, but with limited collagen type I deposition. This approach allows us to overcome the limitations of current approaches such as the dedifferentiation occurring in 2D in vitro expansion and the necrotic formation in spheroids. Further studies are warranted to assess long-term ECM deposition and integration with native cartilage. Though limitations exist, this study suggests a promising avenue for cartilage repair with col/alg hydrogel scaffolds.
Collapse
Affiliation(s)
- Tosca Roncada
- School
of Pharmacy and Biomedical Sciences, University
of Portsmouth, St Michael’s
Building, White Swan Road, Portsmouth PO1 2DT, U.K.
| | - Gordon Blunn
- School
of Pharmacy and Biomedical Sciences, University
of Portsmouth, St Michael’s
Building, White Swan Road, Portsmouth PO1 2DT, U.K.
| | - Marta Roldo
- School
of Pharmacy and Biomedical Sciences, University
of Portsmouth, St Michael’s
Building, White Swan Road, Portsmouth PO1 2DT, U.K.
| |
Collapse
|
12
|
Hussain MT, Austin-Williams S, Wright TD, Dhawan UK, Pinto AL, Cooper D, Norling LV. β1-Integrin-Mediated Uptake of Chondrocyte Extracellular Vesicles Regulates Chondrocyte Homeostasis. Int J Mol Sci 2024; 25:4756. [PMID: 38731975 PMCID: PMC11083596 DOI: 10.3390/ijms25094756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Osteoarthritis (OA) is the most prevalent age-related degenerative disorder, which severely reduces the quality of life of those affected. Whilst management strategies exist, no cures are currently available. Virtually all joint resident cells generate extracellular vesicles (EVs), and alterations in chondrocyte EVs during OA have previously been reported. Herein, we investigated factors influencing chondrocyte EV release and the functional role that these EVs exhibit. Both 2D and 3D models of culturing C28I/2 chondrocytes were used for generating chondrocyte EVs. We assessed the effect of these EVs on chondrogenic gene expression as well as their uptake by chondrocytes. Collectively, the data demonstrated that chondrocyte EVs are sequestered within the cartilage ECM and that a bi-directional relationship exists between chondrocyte EV release and changes in chondrogenic differentiation. Finally, we demonstrated that the uptake of chondrocyte EVs is at least partially dependent on β1-integrin. These results indicate that chondrocyte EVs have an autocrine homeostatic role that maintains chondrocyte phenotype. How this role is perturbed under OA conditions remains the subject of future work.
Collapse
Affiliation(s)
- Mohammed Tayab Hussain
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Shani Austin-Williams
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Thomas Dudley Wright
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Umesh Kumar Dhawan
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
| | - Andreia L. Pinto
- Royal Brompton & Harefield NHS Foundation Trust, London SW3 6PY, UK;
| | - Dianne Cooper
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London EC1M 6BQ, UK
| | - Lucy V. Norling
- The William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (M.T.H.); (T.D.W.); (U.K.D.); (D.C.)
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
13
|
Sulcanese L, Prencipe G, Canciello A, Cerveró-Varona A, Perugini M, Mauro A, Russo V, Barboni B. Stem-Cell-Driven Chondrogenesis: Perspectives on Amnion-Derived Cells. Cells 2024; 13:744. [PMID: 38727280 PMCID: PMC11083072 DOI: 10.3390/cells13090744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Regenerative medicine harnesses stem cells' capacity to restore damaged tissues and organs. In vitro methods employing specific bioactive molecules, such as growth factors, bio-inductive scaffolds, 3D cultures, co-cultures, and mechanical stimuli, steer stem cells toward the desired differentiation pathways, mimicking their natural development. Chondrogenesis presents a challenge for regenerative medicine. This intricate process involves precise modulation of chondro-related transcription factors and pathways, critical for generating cartilage. Cartilage damage disrupts this process, impeding proper tissue healing due to its unique mechanical and anatomical characteristics. Consequently, the resultant tissue often forms fibrocartilage, which lacks adequate mechanical properties, posing a significant hurdle for effective regeneration. This review comprehensively explores studies showcasing the potential of amniotic mesenchymal stem cells (AMSCs) and amniotic epithelial cells (AECs) in chondrogenic differentiation. These cells exhibit innate characteristics that position them as promising candidates for regenerative medicine. Their capacity to differentiate toward chondrocytes offers a pathway for developing effective regenerative protocols. Understanding and leveraging the innate properties of AMSCs and AECs hold promise in addressing the challenges associated with cartilage repair, potentially offering superior outcomes in tissue regeneration.
Collapse
Affiliation(s)
- Ludovica Sulcanese
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Angelo Canciello
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Monia Perugini
- Department of Bioscience and Technology for Food, Agriculture, and Environment, University of Teramo, 64100 Teramo, Italy;
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Valentina Russo
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| |
Collapse
|
14
|
Foltz L, Avabhrath N, Lanchy JM, Levy T, Possemato A, Ariss M, Peterson B, Grimes M. Craniofacial chondrogenesis in organoids from human stem cell-derived neural crest cells. iScience 2024; 27:109585. [PMID: 38623327 PMCID: PMC11016914 DOI: 10.1016/j.isci.2024.109585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 04/17/2024] Open
Abstract
Knowledge of cell signaling pathways that drive human neural crest differentiation into craniofacial chondrocytes is incomplete, yet essential for using stem cells to regenerate craniomaxillofacial structures. To accelerate translational progress, we developed a differentiation protocol that generated self-organizing craniofacial cartilage organoids from human embryonic stem cell-derived neural crest stem cells. Histological staining of cartilage organoids revealed tissue architecture and staining typical of elastic cartilage. Protein and post-translational modification (PTM) mass spectrometry and snRNA-seq data showed that chondrocyte organoids expressed robust levels of cartilage extracellular matrix (ECM) components: many collagens, aggrecan, perlecan, proteoglycans, and elastic fibers. We identified two populations of chondroprogenitor cells, mesenchyme cells and nascent chondrocytes, and the growth factors involved in paracrine signaling between them. We show that ECM components secreted by chondrocytes not only create a structurally resilient matrix that defines cartilage, but also play a pivotal autocrine cell signaling role in determining chondrocyte fate.
Collapse
Affiliation(s)
- Lauren Foltz
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| | - Nagashree Avabhrath
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| | - Jean-Marc Lanchy
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| | - Tyler Levy
- Cell Signaling Technology, Danvers, MA 01923, USA
| | | | - Majd Ariss
- Cell Signaling Technology, Danvers, MA 01923, USA
| | | | - Mark Grimes
- Division of Biological Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, The University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
15
|
Diederichs S, Dreher SI, Nüesch SA, Schmidt S, Merle C, Richter W. Mesenchymal stromal cell chondrogenesis under ALK1/2/3-specific BMP inhibition: a revision of the prohypertrophic signalling network concept. Stem Cell Res Ther 2024; 15:98. [PMID: 38581019 PMCID: PMC10998299 DOI: 10.1186/s13287-024-03710-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/27/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND In vitro chondrogenesis of mesenchymal stromal cells (MSCs) driven by the essential chondro-inducer transforming growth factor (TGF)-β is instable and yields undesired hypertrophic cartilage predisposed to bone formation in vivo. TGF-β can non-canonically activate bone morphogenetic protein-associated ALK1/2/3 receptors. These have been accused of driving hypertrophic MSC misdifferentiation, but data remained conflicting. We here tested the antihypertrophic capacity of two highly specific ALK1/2/3 inhibitors - compound A (CompA) and LDN-212854 (LDN21) - in order to reveal potential prohypertrophic contributions of these BMP/non-canonical TGF-β receptors during MSC in vitro chondrogenesis. METHODS Standard chondrogenic pellet cultures of human bone marrow-derived MSCs were treated with TGF-β and CompA (500 nM) or LDN21 (500 nM). Daily 6-hour pulses of parathyroid hormone-related peptide (PTHrP[1-34], 2.5 nM, from day 7) served as potent antihypertrophic control treatment. Day 28 samples were subcutaneously implanted into immunodeficient mice. RESULTS All groups underwent strong chondrogenesis, but GAG/DNA deposition and ACAN expression were slightly but significantly reduced by ALK inhibition compared to solvent controls along with a mild decrease of the hypertrophy markers IHH-, SPP1-mRNA, and Alkaline phosphatase (ALP) activity. When corrected for the degree of chondrogenesis (COL2A1 expression), only pulsed PTHrP but not ALK1/2/3 inhibition qualified as antihypertrophic treatment. In vivo, all subcutaneous cartilaginous implants mineralized within 8 weeks, but PTHrP pretreated samples formed less bone and attracted significantly less haematopoietic marrow than ALK1/2/3 inhibitor groups. CONCLUSIONS Overall, our data show that BMP-ALK1/2/3 inhibition cannot program mesenchymal stromal cells toward stable chondrogenesis. BMP-ALK1/2/3 signalling is no driver of hypertrophic MSC misdifferentiation and BMP receptor induction is not an adverse prohypertrophic side effect of TGF-β that leads to endochondral MSC misdifferentiation. Instead, the prohypertrophic network comprises misregulated PTHrP/hedgehog signalling and WNT activity, and a potential contribution of TGF-β-ALK4/5-mediated SMAD1/5/9 signalling should be further investigated to decide about its postulated prohypertrophic activity. This will help to successfully engineer cartilage replacement tissues from MSCs in vitro and translate these into clinical cartilage regenerative therapies.
Collapse
Affiliation(s)
- Solvig Diederichs
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, Heidelberg, 69118, Germany.
| | - Simon I Dreher
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, Heidelberg, 69118, Germany
| | - Sarah Anna Nüesch
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, Heidelberg, 69118, Germany
| | - Sven Schmidt
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, Heidelberg, 69118, Germany
| | - Christian Merle
- Orthopaedic University Hospital, Heidelberg University Hospital, Heidelberg, Germany
- Orthopädische Klinik Paulinenhilfe, Diakonieklinikum Stuttgart, Stuttgart, Germany
| | - Wiltrud Richter
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, Heidelberg, 69118, Germany
| |
Collapse
|
16
|
Majumder N, Seit S, Bhabesh NS, Ghosh S. An Advanced Bioconjugation Strategy for Covalent Tethering of TGFβ3 with Silk Fibroin Matrices and its Implications in the Chondrogenesis Profile of Human BMSCs and Human Chondrocytes: A Paradigm Shift in Cartilage Tissue Engineering. Adv Healthc Mater 2024; 13:e2303513. [PMID: 38291832 DOI: 10.1002/adhm.202303513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/25/2024] [Indexed: 02/01/2024]
Abstract
The transforming growth factor-β class of cytokines plays a significant role in articular cartilage formation from mesenchymal condensation to chondrogenic differentiation. However, their exogenous addition to the chondrogenic media makes the protocol expensive. It reduces the bioavailability of the cytokine to the cells owing to their burst release. The present study demonstrates an advanced bioconjugation strategy to conjugate transforming growth factor-β3 (TGFβ3) with silk fibroin matrix covalently via a cyanuric chloride coupling reaction. The tethering and change in secondary conformation are confirmed using various spectroscopic analyses. To assess the functionality of the chemically modified silk matrix, human bone marrow-derived mesenchymal stem cells (hBMSCs) and chondrocytes are cultured for 28 days in a chondrogenic differentiation medium. Gene expression and histological analysis reveal enhanced expression of chondrogenic markers with intense Safranin-O and Alcian Blue staining in TGFβ3 conjugated silk matrices than where TGFβ3 is exogenously added to the media for both hBMSCs and chondrocytes. Therefore, this study successfully recapitulates the native niche of TGFβ3 and the role of the silk as a growth factor stabilizer. When cultured over TGFβ3 conjugated silk matrices, hBMSCs display increased proteoglycan secretion and maximum chondrogenic trait with attenuation of chondrocyte hypertrophy over human chondrocytes.
Collapse
Affiliation(s)
- Nilotpal Majumder
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Sinchan Seit
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Neel Sarovar Bhabesh
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Transcription Regulation group, New Delhi, 110067, India
| | - Sourabh Ghosh
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| |
Collapse
|
17
|
Xu J, Zhang Q, Jiang T, Liu L, Gu H, Tan Y, Wang H. Dose- and stage-dependent toxic effects of prenatal prednisone exposure on fetal articular cartilage development. Toxicol Lett 2024; 393:14-23. [PMID: 38211732 DOI: 10.1016/j.toxlet.2024.01.004] [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: 09/10/2023] [Revised: 12/16/2023] [Accepted: 01/06/2024] [Indexed: 01/13/2024]
Abstract
Prednisone is frequently used to treat rheumatoid diseases in pregnant women because of its high degree of safety. Whether prenatal prednisone exposure (PPE) negatively impacts fetal articular cartilage development is unclear. In this study, we simulated a clinical prednisone treatment regimen to examine the effects of different timings and doses of PPE on cartilage development in female and male fetal mice. Prednisone doses (0.25, 0.5, and 1 mg/kg/d) was administered to Kunming mice at different gestational stages (0-9 gestational days, GD0-9), mid-late gestation (GD10-18), or during the entire gestation (GD0-18) by oral gavage. The amount of matrix aggrecan (ACAN) and collagen type II a1(COL2a1), and expression of transforming growth factor β1 (TGFβ1) signaling pathway also demonstrated that the chondrocyte count and ACAN and COL2a1 expression reduced in fetal mice with early and mid-late PPE, with the reduction being more significant in the mice with early PPE than that in those with PPE at other stages. Prenatal exposure to different prednisone doses prevented the reduction of TGFβ signaling pathway-related genes [TGFβR1, SMAD family member 3 (Smad3), SRY-box9 (SOX9)] as well as ACAN and COL2a1 mRNA expression levels in fetal mouse cartilage, with the most significant decrease after 1 mg/kg·d PPE. In conclusion, PPE can inhibit/restrain fetal cartilage development, with the greatest effect at higher clinical dose (1 mg/kg·d) and early stage of pregnancy (GD0-9), and the mechanism may be related to TGFβ signaling pathway inhibition. The result of this study provide a theoretical and experimental foundation for the rational clinical use of prednisone.
Collapse
Affiliation(s)
- Junmiao Xu
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qi Zhang
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Tao Jiang
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Liang Liu
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hanwen Gu
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yang Tan
- Division of Joint surgery and sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
18
|
Vágó J, Somogyi C, Takács R, Barna KB, Jin EJ, Zákány R, Matta C. Isolation and Culturing of Primary Murine Chondroprogenitor Cells: A Mammalian Model of Chondrogenesis. Curr Protoc 2024; 4:e1005. [PMID: 38465381 DOI: 10.1002/cpz1.1005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Embryonic limb bud-derived micromass cultures are valuable tools for investigating cartilage development, tissue engineering, and therapeutic strategies for cartilage-related disorders. This collection of fine-tuned protocols used in our laboratories outlines step-by-step procedures for the isolation, expansion, and differentiation of primary mouse limb bud cells into chondrogenic micromass cultures. Key aspects covered in these protocols include synchronized fertilization of mice (Basic Protocol 1), tissue dissection, cell isolation, micromass formation, and culture optimization parameters, such as cell density and medium composition (Basic Protocol 2). We describe techniques for characterizing the chondrogenic differentiation process by histological analysis (Basic Protocol 3). The protocols also address common challenges encountered during the process and provide troubleshooting strategies. This fine-tuned comprehensive protocol serves as a valuable resource for scientists working in the fields of developmental biology, cartilage tissue engineering, and regenerative medicine, offering an updated methodology for the study of efficient chondrogenic differentiation and cartilage tissue regeneration. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Synchronized fertilization of mice Basic Protocol 2: Micromass culture of murine embryonic limb bud-derived cells Basic Protocol 3: Qualitative assessment of cartilage matrix production using Alcian blue staining.
Collapse
Affiliation(s)
- Judit Vágó
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Hungary
- These authors contributed equally to the work
| | - Csilla Somogyi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Hungary
- These authors contributed equally to the work
| | - Roland Takács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Hungary
| | - Krisztina Bíróné Barna
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Hungary
| | - Eun-Jung Jin
- Department of Biological Sciences, College of Natural Sciences, Wonkwang University, South Korea
| | - Róza Zákány
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Hungary
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Hungary
| |
Collapse
|
19
|
Lee J, Lee E, Huh SJ, Kang JI, Park KM, Byun H, Lee S, Kim E, Shin H. Composite Spheroid-Laden Bilayer Hydrogel for Engineering Three-Dimensional Osteochondral Tissue. Tissue Eng Part A 2024; 30:225-243. [PMID: 38062771 DOI: 10.1089/ten.tea.2023.0299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2024] Open
Abstract
A combination of hydrogels and stem cell spheroids has been used to engineer three-dimensional (3D) osteochondral tissue, but precise zonal control directing cell fate within the hydrogel remains a challenge. In this study, we developed a composite spheroid-laden bilayer hydrogel to imitate osteochondral tissue by spatially controlled differentiation of human adipose-derived stem cells. Meticulous optimization of the spheroid-size and mechanical strength of gelatin methacryloyl (GelMA) hydrogel enables the cells to homogeneously sprout within the hydrogel. Moreover, fibers immobilizing transforming growth factor beta-1 (TGF-β1) or bone morphogenetic protein-2 (BMP-2) were incorporated within the spheroids, which induced chondrogenic or osteogenic differentiation of cells in general media, respectively. The spheroids-filled GelMA solution was crosslinked to create the bilayer hydrogel, which demonstrated a strong interfacial adhesion between the two layers. The cell sprouting enhanced the adhesion of each hydrogel, demonstrated by increase in tensile strength from 4.8 ± 0.4 to 6.9 ± 1.2 MPa after 14 days of culture. Importantly, the spatially confined delivery of BMP-2 within the spheroids increased mineral deposition and more than threefold enhanced osteogenic genes of cells in the bone layer while the cells induced by TGF-β1 signals were apparently differentiated into chondrocytes within the cartilage layer. The results suggest that our composite spheroid-laden hydrogel could be used for the biofabrication of osteochondral tissue, which can be applied to engineer other complex tissues by delivery of appropriate biomolecules.
Collapse
Affiliation(s)
- Jinkyu Lee
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, Republic of Korea
| | - Eunjin Lee
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, Republic of Korea
| | - Seung Jae Huh
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, Republic of Korea
| | - Jeon Il Kang
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Kyung Min Park
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Hayeon Byun
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Sangmin Lee
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Eunhyung Kim
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, Republic of Korea
- Department of Bioengineering, Institute of Nano Science and Technology, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
20
|
Tomaszewska E, Hułas-Stasiak M, Dobrowolski P, Świątkiewicz M, Muszyński S, Tomczyk-Warunek A, Blicharski T, Donaldson J, Arciszewski MB, Świetlicki M, Puzio I, Bonior J. Does Chronic Pancreatitis in Growing Pigs Lead to Articular Cartilage Degradation and Alterations in Subchondral Bone? Int J Mol Sci 2024; 25:1989. [PMID: 38396667 PMCID: PMC10888541 DOI: 10.3390/ijms25041989] [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: 12/18/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Chronic pancreatitis (CP), a progressive inflammatory disease, poses diagnostic challenges due to its initially asymptomatic nature. While CP's impact on exocrine and endocrine functions is well-recognized, its potential influence on other body systems, particularly in young individuals, remains underexplored. This study investigates the hypothesis that CP in growing pigs leads to alterations in articular cartilage and subchondral bone, potentially contributing to osteoarthritis (OA) development. Utilizing a pig model of cerulein-induced CP, we examined the structural and compositional changes in subchondral bone, articular cartilage, and synovial fluid. Histological analyses, including Picrosirius Red and Safranin-O staining, were employed alongside immuno-histochemistry and Western blotting techniques. Our findings reveal significant changes in the subchondral bone, including reduced bone volume and alterations in collagen fiber composition. Articular cartilage in CP pigs exhibited decreased proteoglycan content and alterations in key proteins such as MMP-13 and TGF-β1, indicative of early cartilage degradation. These changes suggest a link between CP and musculoskeletal alterations, underscoring the need for further research into CP's systemic effects. Our study provides foundational insights into the relationship between CP and skeletal health, potentially guiding future pediatric healthcare strategies for early CP diagnosis and management.
Collapse
Affiliation(s)
- Ewa Tomaszewska
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| | - Monika Hułas-Stasiak
- Department of Functional Anatomy and Cytobiology, Faculty of Biology and Biotechnology, Maria Curie-Sklodowska University, 20-033 Lublin, Poland; (M.H.-S.); (P.D.)
| | - Piotr Dobrowolski
- Department of Functional Anatomy and Cytobiology, Faculty of Biology and Biotechnology, Maria Curie-Sklodowska University, 20-033 Lublin, Poland; (M.H.-S.); (P.D.)
| | - Małgorzata Świątkiewicz
- Department of Animal Nutrition and Feed Science, National Research Institute of Animal Production, 32-083 Balice, Poland;
| | - Siemowit Muszyński
- Department of Biophysics, Faculty of Environmental Biology, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| | - Agnieszka Tomczyk-Warunek
- Laboratory of Locomotor System Research, Department of Rehabilitation and Physiotherapy, Medical University in Lublin, 20-090 Lublin, Poland;
| | - Tomasz Blicharski
- Department of Orthopaedics and Rehabilitation, Medical University in Lublin, 20-090 Lublin, Poland;
| | - Janine Donaldson
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Parktown, Johannesburg 2193, South Africa;
| | - Marcin B. Arciszewski
- Department of Animal Anatomy and Histology, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| | - Michał Świetlicki
- Department of Applied Physics, Faculty of Mechanical Engineering, Lublin University of Technology, 20-618 Lublin, Poland;
| | - Iwona Puzio
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| | - Joanna Bonior
- Department of Medical Physiology, Chair of Biomedical Sciences, Institute of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, 31-501 Cracow, Poland;
| |
Collapse
|
21
|
Kapat K, Kumbhakarn S, Sable R, Gondane P, Takle S, Maity P. Peptide-Based Biomaterials for Bone and Cartilage Regeneration. Biomedicines 2024; 12:313. [PMID: 38397915 PMCID: PMC10887361 DOI: 10.3390/biomedicines12020313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
The healing of osteochondral defects (OCDs) that result from injury, osteochondritis, or osteoarthritis and bear lesions in the cartilage and bone, pain, and loss of joint function in middle- and old-age individuals presents challenges to clinical practitioners because of non-regenerative cartilage and the limitations of current therapies. Bioactive peptide-based osteochondral (OC) tissue regeneration is becoming more popular because it does not have the immunogenicity, misfolding, or denaturation problems associated with original proteins. Periodically, reviews are published on the regeneration of bone and cartilage separately; however, none of them addressed the simultaneous healing of these tissues in the complicated heterogeneous environment of the osteochondral (OC) interface. As regulators of cell adhesion, proliferation, differentiation, angiogenesis, immunomodulation, and antibacterial activity, potential therapeutic strategies for OCDs utilizing bone and cartilage-specific peptides should be examined and investigated. The main goal of this review was to study how they contribute to the healing of OCDs, either alone or in conjunction with other peptides and biomaterials.
Collapse
Affiliation(s)
- Kausik Kapat
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Sakshi Kumbhakarn
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Rahul Sable
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Prashil Gondane
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Shruti Takle
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Pritiprasanna Maity
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
22
|
Grzelak A, Hnydka A, Higuchi J, Michalak A, Tarczynska M, Gaweda K, Klimek K. Recent Achievements in the Development of Biomaterials Improved with Platelet Concentrates for Soft and Hard Tissue Engineering Applications. Int J Mol Sci 2024; 25:1525. [PMID: 38338805 PMCID: PMC10855389 DOI: 10.3390/ijms25031525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Platelet concentrates such as platelet-rich plasma, platelet-rich fibrin or concentrated growth factors are cost-effective autologous preparations containing various growth factors, including platelet-derived growth factor, transforming growth factor β, insulin-like growth factor 1 and vascular endothelial growth factor. For this reason, they are often used in regenerative medicine to treat wounds, nerve damage as well as cartilage and bone defects. Unfortunately, after administration, these preparations release growth factors very quickly, which lose their activity rapidly. As a consequence, this results in the need to repeat the therapy, which is associated with additional pain and discomfort for the patient. Recent research shows that combining platelet concentrates with biomaterials overcomes this problem because growth factors are released in a more sustainable manner. Moreover, this concept fits into the latest trends in tissue engineering, which include biomaterials, bioactive factors and cells. Therefore, this review presents the latest literature reports on the properties of biomaterials enriched with platelet concentrates for applications in skin, nerve, cartilage and bone tissue engineering.
Collapse
Affiliation(s)
- Agnieszka Grzelak
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland; (A.G.); (A.H.)
| | - Aleksandra Hnydka
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland; (A.G.); (A.H.)
| | - Julia Higuchi
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Prymasa Tysiaclecia Avenue 98, 01-142 Warsaw, Poland;
| | - Agnieszka Michalak
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, Chodzki 4 a Street, 20-093 Lublin, Poland;
| | - Marta Tarczynska
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-090 Lublin, Poland; (M.T.); (K.G.)
- Arthros Medical Centre, Chodzki 31 Street, 20-093 Lublin, Poland
| | - Krzysztof Gaweda
- Department and Clinic of Orthopaedics and Traumatology, Medical University of Lublin, Jaczewskiego 8 Street, 20-090 Lublin, Poland; (M.T.); (K.G.)
- Arthros Medical Centre, Chodzki 31 Street, 20-093 Lublin, Poland
| | - Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland; (A.G.); (A.H.)
| |
Collapse
|
23
|
Uysal O, Erybeh H, Canbek M, Ekenel EQ, Gunes S, Büyükköroğlu G, Semerci Sevimli T, Cemrek F, Sariboyaci AE. Stem Cell-Based or Cell-Free Gene Therapy in Chondrocyte Regeneration: Synovial Fluid-Derived Mesenchymal Stem Cell Exosomes. Curr Mol Med 2024; 24:906-919. [PMID: 37859306 PMCID: PMC11327740 DOI: 10.2174/0115665240266016231014081916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Cartilage injuries are currently the most prevalent joint disease. Previous studies have emphasized the use of stem cells as the effective treatment for regenerating cartilage damage. OBJECTIVE In this study, considering the difficulties of the cellular therapy method, it was hypothesized that human synovial fluid-derived mesenchymal stem cell (hSFMSC) exosomes as a SC source could be used to treat these injuries as a safer and cell-free therapeutic alternative procedure due to its direct relevance to cartilage regeneration. Moreover, this study aimed to determine the miRNA and target genes required for the formation of SC treatment combined with gene therapy in order to reveal the mechanism of cartilage regeneration and increase its effectiveness. METHODS MSCs were characterized by flow cytometry, and immunocytochemical and differentiation analyses were done. To characterize functionally isolated exosomes, in vitro uptake analysis was performed. RT-qPCR was used to examine in terms of the advantages of cellular and cell-free therapy, mature human chondroblasts derived by differentiation from hSF-MSCs and human chondrocyte profiles were compared in order to demonstrate the above profile of hSF-MSCs and exosomes isolated from them, and the effectiveness of SC therapy in repairing cartilage damage. RESULTS According to our findings, the expression level of hsa-miR-155-5p was found to be considerably higher in chondrocytes differentiated from human synovial fluid MSCs than in mature human chondrocytes. These findings were also supported by the TGF-signalling pathway and chondrogenesis marker genes. CONCLUSION It was concluded that hSF-MSCs and exosomes can be used in the treatment of cartilage damage, and hsa-miR-155-5p can be used as a target miRNA in a new gene therapy approach because it increases the therapeutic effect on cartilage damage.
Collapse
Affiliation(s)
- Onur Uysal
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Eskisehir, Turkiye
| | - Haya Erybeh
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Molecular Biology, Science Faculty, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Mediha Canbek
- Department of Molecular Biology, Science Faculty, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Emilia Qomi Ekenel
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Sibel Gunes
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Eskisehir, Turkiye
| | - Gülay Büyükköroğlu
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkiye
| | - Tugba Semerci Sevimli
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Fatih Cemrek
- Department of Statistics, Faculty of Science and Letters, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Ayla Eker Sariboyaci
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Eskisehir, Turkiye
| |
Collapse
|
24
|
Iqbal Z, Xia J, Murtaza G, Shabbir M, Rehman K, Yujie L, Duan L. Targeting WNT signalling pathways as new therapeutic strategies for osteoarthritis. J Drug Target 2023; 31:1027-1049. [PMID: 37969105 DOI: 10.1080/1061186x.2023.2281861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/21/2023] [Indexed: 11/17/2023]
Abstract
Osteoarthritis (OA) is a highly prevalent chronic joint disease and the leading cause of disability. Currently, no drugs are available to control joint damage or ease the associated pain. The wingless-type (WNT) signalling pathway is vital in OA progression. Excessive activation of the WNT signalling pathway is pertinent to OA progression and severity. Therefore, agonists and antagonists of the WNT pathway are considered potential drug candidates for OA treatment. For example, SM04690, a novel small molecule inhibitor of WNT signalling, has demonstrated its potential in a recent phase III clinical trial as a disease-modifying osteoarthritis drug (DMOAD). Therefore, targeting the WNT signalling pathway may be a distinctive approach to developing particular agents helpful in treating OA. This review aims to update the most recent progress in OA drug development by targeting the WNT pathway. In this, we introduce WNT pathways and their crosstalk with other signalling pathways in OA development and highlight the role of the WNT signalling pathway as a key regulator in OA development. Several articles have reviewed the Wnt pathway from different aspects. This candid review provides an introduction to WNT pathways and their crosstalk with other signalling pathways in OA development, highlighting the role of the WNT signalling pathway as a key regulator in OA development with the latest research. Particularly, we emphasise the state-of-the-art in targeting the WNT pathway as a promising therapeutic approach for OA and challenges in their development and the nanocarrier-based delivery of WNT modulators for treating OA.
Collapse
Affiliation(s)
- Zoya Iqbal
- Department of Orthopedics, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Pakistan
| | - Maryam Shabbir
- Faculty of Pharmacy, The University of Lahore, Lahore Campus, Pakistan
| | - Khurrum Rehman
- Department of Allied health sciences, The University of Agriculture, D.I.Khan, Pakistan
| | - Liang Yujie
- Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Li Duan
- Department of Orthopedics, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
25
|
St Amant J, Michaud J, Hinds D, Coyle M, Pozzi A, Clark AL. Depleting transforming growth factor beta receptor 2 signalling in the cartilage of itga1-null mice attenuates spontaneous knee osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100399. [PMID: 37649532 PMCID: PMC10462827 DOI: 10.1016/j.ocarto.2023.100399] [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: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Objectives Integrin α1β1 protects against osteoarthritis (OA) when it is upregulated in the superficial zone of cartilage in the early stages of disease. However, the mechanism behind this protection is unknown. Integrin α1β1 moderates transforming growth factor β receptor II (TGFBR2) signalling, a critical regulator of chondrocyte anabolic activity. To this end, mice lacking integrin α1β1 have increased baseline activation of TGFBR2 signalling and overall fibrosis. The purpose of this study was to evaluate the interplay between integrin α1β1 and TGFBR2 in the development of spontaneous OA. We hypothesized that dampening TGFBR2 signalling in the cartilage of itga1-null mice would attenuate OA. Methods Behavioural and histological manifestations of spontaneous knee OA were measured at 4, 8, 12 and 16 months in mice with and without a ubiquitous itga1 deletion and with and without a tamoxifen-induced cartilage specific TGFBR2 depletion. Results Knee cartilage degeneration, collateral ligament ossification and pain responses increased with age. Itga1-null mice with intact TGFBR2 signalling developed earlier and more severe OA compared to controls. In agreement with our hypothesis, depleting TGFBR2 signalling in the cartilage of itga1-null mice attenuated OA progression. Conclusion Intact TGFBR2 signalling drives early and worse knee OA in itga1-null mice. This result supports the hypothesis that the increased expression of integrin α1β1 by superficial zone chondrocytes early in OA development dampens TGFBR2 signalling and thus protects against degeneration.
Collapse
Affiliation(s)
- Jennifer St Amant
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Jana Michaud
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Daniel Hinds
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Madison Coyle
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Veterans Affairs, Nashville, TN, USA
| | - Andrea L. Clark
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
26
|
Pitou M, Papachristou E, Bratsios D, Kefala GM, Tsagkarakou AS, Leonidas DD, Aggeli A, Papadopoulos GE, Papi RM, Choli-Papadopoulou T. In Vitro Chondrogenesis Induction by Short Peptides of the Carboxy-Terminal Domain of Transforming Growth Factor β1. Biomedicines 2023; 11:3182. [PMID: 38137403 PMCID: PMC10740954 DOI: 10.3390/biomedicines11123182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 12/24/2023] Open
Abstract
Τransforming growth factor β1 (TGF-β1) comprises a key regulator protein in many cellular processes, including in vivo chondrogenesis. The treatment of human dental pulp stem cells, separately, with Leu83-Ser112 (C-terminal domain of TGF-β1), as well as two very short peptides, namely, 90-YYVGRKPK-97 (peptide 8) and 91-YVGRKP-96 (peptide 6) remarkably enhanced the chondrogenic differentiation capacity in comparison to their full-length mature TGF-β1 counterpart either in monolayer cultures or 3D scaffolds. In 3D scaffolds, the reduction of the elastic modulus and viscous modulus verified the production of different amounts and types of ECM components. Molecular dynamics simulations suggested a mode of the peptides' binding to the receptor complex TβRII-ALK5 and provided a possible structural explanation for their role in inducing chondrogenesis, along with endogenous TGF-β1. Further experiments clearly verified the aforementioned hypothesis, indicating the signal transduction pathway and the involvement of TβRII-ALK5 receptor complex. Real-time PCR experiments and Western blot analysis showed that peptides favor the ERK1/2 and Smad2 pathways, leading to an articular, extracellular matrix formation, while TGF-β1 also favors the Smad1/5/8 pathway which leads to the expression of the metalloproteinases ADAMTS-5 and MMP13 and, therefore, to a hypertrophic chondrocyte phenotype. Taken together, the two short peptides, and, mainly, peptide 8, could be delivered with a scaffold to induce in vivo chondrogenesis in damaged articular cartilage, constituting, thus, an alternative therapeutic approach for osteoarthritis.
Collapse
Affiliation(s)
- Maria Pitou
- Laboratory of Biochemistry, School of Chemistry, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| | - Eleni Papachristou
- Laboratory of Biochemistry, School of Chemistry, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| | - Dimitrios Bratsios
- Laboratory of Biomedical Engineering, School of Chemical Engineering, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| | - Georgia-Maria Kefala
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Anastasia S. Tsagkarakou
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Demetrios D. Leonidas
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Amalia Aggeli
- Laboratory of Biomedical Engineering, School of Chemical Engineering, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| | - Georgios E. Papadopoulos
- Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
| | - Rigini M. Papi
- Laboratory of Biochemistry, School of Chemistry, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| | - Theodora Choli-Papadopoulou
- Laboratory of Biochemistry, School of Chemistry, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece
| |
Collapse
|
27
|
Gu F, Zhang K, Zhu WA, Sui Z, Li J, Xie X, Yu T. Silicone rubber sealed channel induced self-healing of large bone defects: Where is the limit of self-healing of bone? J Orthop Translat 2023; 43:21-35. [PMID: 37965195 PMCID: PMC10641457 DOI: 10.1016/j.jot.2023.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/02/2023] [Accepted: 09/12/2023] [Indexed: 11/16/2023] Open
Abstract
Background Large defects of long tubular bones due to severe trauma, bone tumor resection, or osteomyelitis debridement are challenging in orthopedics. Bone non-union and other complications often lead to serious consequences. At present, autologous bone graft is still the gold standard for the treatment of large bone defects. However, autologous bone graft sources are limited. Silicon rubber (SR) materials are widely used in biomedical fields, due to their safety and biocompatibility, and even shown to induce nerve regeneration. Materials and methods We extracted rat bone marrow mesenchymal stem cells (BMMSCs) in vitro and verified the biocompatibility of silicone rubber through cell experiments. Then we designed a rabbit radius critical sized bone defect model to verify the effect of silicone rubber sealed channel inducing bone repair in vivo. Results SR sealed channel could prevent the fibrous tissue from entering the fracture end and forming bone nonunion, thereby inducing self-healing of long tubular bone through endochondral osteogenesis. The hematoma tissue formed in the early stage was rich in osteogenesis and angiogenesis related proteins, and gradually turned into vascularization and endochondral osteogenesis, and finally realized bone regeneration. Conclusions In summary, our study proved that SR sealed channel could prevent the fibrous tissue from entering the fracture end and induce self-healing of long tubular bone through endochondral osteogenesis. In this process, the sealed environment provided by the SR channel was key, and this might indicate that the limit of self-healing of bone exceeded the previously thought. The translational potential of this article This study investigated a new concept to induce the self-healing of large bone defects. It could avoid trauma caused by autologous bone extraction and possible rejection reactions caused by bone graft materials. Further research based on this study, including the innovation of induction materials, might invent a new type of bone inducing production, which could bring convenience to patients. We believed that this study had significant meaning for the treatment of large bone defects in clinical practice.
Collapse
Affiliation(s)
- Feng Gu
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| | - Ke Zhang
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| | - Wan-an Zhu
- Department of Radiology, First Hospital of Jilin University, Changchun, 130021, China
| | - Zhenjiang Sui
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| | - Jiangbi Li
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaoping Xie
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| | - Tiecheng Yu
- Department of Orthopedics, First Hospital of Jilin University, Changchun, 130021, China
| |
Collapse
|
28
|
Thielen NGM, van Caam APM, V Beuningen HM, Vitters EL, van den Bosch MHJ, Koenders MI, van de Loo FAJ, Blaney Davidson EN, van der Kraan PM. Separating friend from foe: Inhibition of TGF-β-induced detrimental SMAD1/5/9 phosphorylation while maintaining protective SMAD2/3 signaling in OA chondrocytes. Osteoarthritis Cartilage 2023; 31:1481-1490. [PMID: 37652257 DOI: 10.1016/j.joca.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/26/2023] [Accepted: 08/14/2023] [Indexed: 09/02/2023]
Abstract
OBJECTIVE Transforming growth factor-β (TGF-β) signaling via SMAD2/3 is crucial to control cartilage homeostasis. However, TGF-β can also have detrimental effects by signaling via SMAD1/5/9 and thereby contribute to diseases like osteoarthritis (OA). In this study, we aimed to block TGF-β-induced SMAD1/5/9 signaling in primary human OA chondrocytes, while maintaining functional SMAD2/3 signaling. DESIGN Human OA chondrocytes were pre-incubated with different concentrations of ALK4/5/7 kinase inhibitor SB-505124 before stimulation with TGF-β. Changes in SMAD C-terminal phosphorylation were analyzed using Western blot and response genes were measured with quantitative Polymerase Chain Reaction. To further explore the consequences of our ability to separate pathways, we investigated TGF-β-induced chondrocyte hypertrophy. RESULTS Pre-incubation with 0.5 µM SB-505124, maintained ±50% of C-terminal SMAD2/3 phosphorylation and induction of JUNB and SERPINE1, but blocked SMAD1/5/9-C phosphorylation and expression of ID1 and ID3. Furthermore, TGF-β, in levels comparable to those in the synovial fluid of OA patients, resulted in regulation of hypertrophic and dedifferentiation markers in OA chondrocytes; i.e. an increase in COL10, RUNX2, COL1A1, and VEGF and a decrease in ACAN expression. Interestingly, in a subgroup of OA chondrocyte donors, blocking only SMAD1/5/9 caused stronger inhibition on TGF-β-induced RUNX2 than blocking both SMAD pathways. CONCLUSION Our findings indicate that using low dose of SB-505124 we maintained functional SMAD2/3 signaling that blocks RUNX2 expression in a subgroup of OA patients. We are the first to show that SMAD2/3 and SMAD1/5/9 pathways can be separately modulated using low and high doses of SB-505124 and thereby split TGF-β's detrimental from protective function in chondrocytes.
Collapse
Affiliation(s)
- Nathalie G M Thielen
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Arjan P M van Caam
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Henk M V Beuningen
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Elly L Vitters
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Martijn H J van den Bosch
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marije I Koenders
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Fons A J van de Loo
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Esmeralda N Blaney Davidson
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Peter M van der Kraan
- Department of Experimental Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
29
|
Mazzella M, Walker K, Cormier C, Kapanowski M, Ishmakej A, Saifee A, Govind Y, Chaudhry GR. Regulation of self-renewal and senescence in primitive mesenchymal stem cells by Wnt and TGFβ signaling. Stem Cell Res Ther 2023; 14:305. [PMID: 37880755 PMCID: PMC10601332 DOI: 10.1186/s13287-023-03533-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND The therapeutic application of multipotent mesenchymal stem cells (MSCs) encounters significant challenges, primarily stemming from their inadequate growth and limited self-renewal capabilities. Additionally, as MSCs are propagated, their ability to self-renew declines, and the exact cellular and molecular changes responsible for this are poorly understood. This study aims to uncover the complex molecular mechanisms that govern the self-renewal of primitive (p) MSCs. METHODS We grew pMSCs using two types of medium, fetal bovine serum (FM) and xeno-free (XM), at both low passage (LP, P3) and high passage (HP, P20). To evaluate LP and HP pMSCs, we examined their physical characteristics, cell surface markers, growth rate, colony-forming ability, BrdU assays for proliferation, telomerase activity, and potential to differentiate into three lineages. Moreover, we conducted RNA-seq to analyze their transcriptome and MNase-seq analysis to investigate nucleosome occupancies. RESULTS When grown in FM, pMSCs underwent changes in their cellular morphology, becoming larger and elongated. This was accompanied by a decrease in the expression of CD90 and CD49f, as well as a reduction in CFE, proliferation rate, and telomerase activity. In addition, these cells showed an increased tendency to differentiate into the adipogenic lineage. However, when grown in XM, pMSCs maintained their self-renewal capacity and ability to differentiate into multiple lineages while preserving their fibroblastoid morphology. Transcriptomic analysis showed an upregulation of genes associated with self-renewal, cell cycle regulation, and DNA replication in XM-cultured pMSCs, while senescence-related genes were upregulated in FM-cultured cells. Further analysis demonstrated differential nucleosomal occupancies in self-renewal and senescence-related genes for pMSCs grown in XM and FM, respectively. These findings were confirmed by qRT-PCR analysis, which revealed alterations in the expression of genes related to self-renewal, cell cycle regulation, DNA replication, differentiation, and senescence. To understand the underlying mechanisms, we investigated the involvement of Wnt and TGFβ signaling pathways by modulating them with agonists and antagonists. This experimental manipulation led to the upregulation and downregulation of self-renewal genes in pMSCs, providing further insights into the signaling pathways governing the self-renewal and senescence of pMSCs. CONCLUSION Our study shows that the self-renewal potential of pMSCs is associated with the Wnt pathway, while senescence is linked to TGFβ.
Collapse
Affiliation(s)
- Matteo Mazzella
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Keegan Walker
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Christina Cormier
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Michael Kapanowski
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Albi Ishmakej
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Azeem Saifee
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - Yashvardhan Govind
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA
| | - G Rasul Chaudhry
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA.
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI, 48309, USA.
| |
Collapse
|
30
|
Zavala G, Viafara-García SM, Novoa J, Hidalgo C, Contardo I, Díaz-Calderón P, Alejandro González-Arriagada W, Khoury M, Acevedo JP. An advanced biphasic porous and injectable scaffold displays a fine balance between mechanical strength and remodeling capabilities essential for cartilage regeneration. Biomater Sci 2023; 11:6801-6822. [PMID: 37622217 DOI: 10.1039/d3bm00703k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
An important challenge in tissue engineering is the regeneration of functional articular cartilage (AC). In the field, biomimetic hydrogels are being extensively studied as scaffolds that recapitulate microenvironmental features or as mechanical supports for transplanted cells. New advanced hydrogel formulations based on salmon methacrylate gelatin (sGelMA), a cold-adapted biomaterial, are presented in this work. The psychrophilic nature of this biomaterial provides rheological advantages allowing the fabrication of scaffolds with high concentrations of the biopolymer and high mechanical strength, suitable for formulating injectable hydrogels with high mechanical strength for cartilage regeneration. However, highly intricate cell-laden scaffolds derived from highly concentrated sGelMA solutions could be deleterious for cells and scaffold remodeling. On this account, the current study proposes the use of sGelMA supplemented with a mesophilic sacrificial porogenic component. The cytocompatibility of different sGelMA-based formulations is tested through the encapsulation of osteoarthritic chondrocytes (OACs) and stimulated to synthesize extracellular matrix (ECM) components in vitro and in vivo. The sGelMA-derived scaffolds reach high levels of stiffness, and the inclusion of porogens impacts positively the scaffold degradability and molecular diffusion, improved fitness of OACs, increased the expression of cartilage-related genes, increased glycosaminoglycan (GAG) synthesis, and improved remodeling toward cartilage-like tissues. Altogether, these data support the use of sGelMA solutions in combination with mammalian solid gelatin beads for highly injectable formulations for cartilage regeneration, strengthening the importance of the balance between mechanical properties and remodeling capabilities.
Collapse
Affiliation(s)
- Gabriela Zavala
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Sergio M Viafara-García
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Javier Novoa
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Carmen Hidalgo
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Ingrid Contardo
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Facultad de Medicina, Escuela de Nutrición y Dietética, Biopolymer Research & Engineering Laboratory (BiopREL), Universidad de los Andes, Chile
| | - Paulo Díaz-Calderón
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Facultad de Medicina, Escuela de Nutrición y Dietética, Biopolymer Research & Engineering Laboratory (BiopREL), Universidad de los Andes, Chile
| | | | - Maroun Khoury
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Juan Pablo Acevedo
- Centro de Investigación e Innovación Biomédica (CIIB), Universidad de los Andes, Chile.
- Cells for Cells and REGENERO, The Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| |
Collapse
|
31
|
Kwok B, Chandrasekaran P, Wang C, He L, Mauck RL, Dyment NA, Koyama E, Han L. Rapid specialization and stiffening of the primitive matrix in developing articular cartilage and meniscus. Acta Biomater 2023; 168:235-251. [PMID: 37414114 PMCID: PMC10529006 DOI: 10.1016/j.actbio.2023.06.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/02/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023]
Abstract
Understanding early patterning events in the extracellular matrix (ECM) formation can provide a blueprint for regenerative strategies to better recapitulate the function of native tissues. Currently, there is little knowledge on the initial, incipient ECM of articular cartilage and meniscus, two load-bearing counterparts of the knee joint. This study elucidated distinctive traits of their developing ECMs by studying the composition and biomechanics of these two tissues in mice from mid-gestation (embryonic day 15.5) to neo-natal (post-natal day 7) stages. We show that articular cartilage initiates with the formation of a pericellular matrix (PCM)-like primitive matrix, followed by the separation into distinct PCM and territorial/interterritorial (T/IT)-ECM domains, and then, further expansion of the T/IT-ECM through maturity. In this process, the primitive matrix undergoes a rapid, exponential stiffening, with a daily modulus increase rate of 35.7% [31.9 39.6]% (mean [95% CI]). Meanwhile, the matrix becomes more heterogeneous in the spatial distribution of properties, with concurrent exponential increases in the standard deviation of micromodulus and the slope correlating local micromodulus with the distance from cell surface. In comparison to articular cartilage, the primitive matrix of meniscus also exhibits exponential stiffening and an increase in heterogeneity, albeit with a much slower daily stiffening rate of 19.8% [14.9 24.9]% and a delayed separation of PCM and T/IT-ECM. These contrasts underscore distinct development paths of hyaline versus fibrocartilage. Collectively, these findings provide new insights into how knee joint tissues form to better guide cell- and biomaterial-based repair of articular cartilage, meniscus and potentially other load-bearing cartilaginous tissues. STATEMENT OF SIGNIFICANCE: Successful regeneration of articular cartilage and meniscus is challenged by incomplete knowledge of early events that drive the initial formation of the tissues' extracellular matrix in vivo. This study shows that articular cartilage initiates with a pericellular matrix (PCM)-like primitive matrix during embryonic development. This primitive matrix then separates into distinct PCM and territorial/interterritorial domains, undergoes an exponential daily stiffening of ≈36% and an increase in micromechanical heterogeneity. At this early stage, the meniscus primitive matrix shows differential molecular traits and exhibits a slower daily stiffening of ≈20%, underscoring distinct matrix development between these two tissues. Our findings thus establish a new blueprint to guide the design of regenerative strategies to recapitulate the key developmental steps in vivo.
Collapse
Affiliation(s)
- Bryan Kwok
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Lan He
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Nathaniel A Dyment
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
32
|
Robinson ME, Rauch D, Glorieux FH, Rauch F. Standardized growth charts for children with osteogenesis imperfecta. Pediatr Res 2023; 94:1075-1082. [PMID: 36922619 DOI: 10.1038/s41390-023-02550-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/06/2023] [Accepted: 02/17/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Osteogenesis imperfecta (OI) is associated with short stature, which is mild, severe and moderate in OI types I, III and IV, respectively. Standardized OI type- and sex-specific growth charts across all pediatric ages do not exist. METHODS We assessed 573 individuals with OI (type I, III or IV), each with at least one height measurement between ages 3 months and 20 years (total 6523 observations). Analogous to the Centers for Disease Control pediatric growth charts, we generated OI type- and sex-specific growth charts for infants (ages 3-36 months) as well as children and adolescents (ages 2-20 years). Growth curves were fitted to the data using the LMS method and percentiles were smoothed. RESULTS Age was associated with a decline in height z-scores (p < 0.001 for all OI types), which was more pronounced in females. Height multiplier curves were produced to predict adult height in children with OI. Among individuals with OI type I, those with COL1A1 pathogenic variants leading to haploinsufficiency were taller than those with COL1A1 or COL1A2 pathogenic variants not leading to haploinsufficiency. CONCLUSION Our standardized OI type- and sex-specific growth charts can be used to assess the growth of individuals with OI from infancy to adulthood. IMPACT Standardized osteogenesis imperfecta (OI) type- and sex-specific growth charts across all pediatric ages do not exist. Our study is the first to generate OI type- and sex-specific growth charts across all pediatric ages. Our height multiplier curves can be utilized to predict adult height in children with OI.
Collapse
Affiliation(s)
- Marie-Eve Robinson
- Shriners Hospital for Children - Canada, McGill University, Montreal, QC, Canada.
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada.
| | - Damian Rauch
- Shriners Hospital for Children - Canada, McGill University, Montreal, QC, Canada
| | - Francis H Glorieux
- Shriners Hospital for Children - Canada, McGill University, Montreal, QC, Canada
| | - Frank Rauch
- Shriners Hospital for Children - Canada, McGill University, Montreal, QC, Canada
| |
Collapse
|
33
|
Wen Y, Chen Y, Wu W, Zhang H, Peng Z, Yao X, Zhang X, Jiang W, Liao Y, Xie Y, Shen X, Sun H, Hu J, Liu H, Chen X, Chen J, Ouyang H. Hyperplastic Human Macromass Cartilage for Joint Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301833. [PMID: 37395375 PMCID: PMC10502860 DOI: 10.1002/advs.202301833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/07/2023] [Indexed: 07/04/2023]
Abstract
Cartilage damage affects millions of people worldwide. Tissue engineering strategies hold the promise to provide off-the-shelf cartilage analogs for tissue transplantation in cartilage repair. However, current strategies hardly generate sufficient grafts, as tissues cannot maintain size growth and cartilaginous phenotypes simultaneously. Herein, a step-wise strategy is developed for fabricating expandable human macromass cartilage (macro-cartilage) in a 3D condition by employing human polydactyly chondrocytes and a screen-defined serum-free customized culture (CC). CC-induced chondrocytes demonstrate improved cell plasticity, expressing chondrogenic biomarkers after a 14.59-times expansion. Crucially, CC-chondrocytes form large-size cartilage tissues with average diameters of 3.25 ± 0.05 mm, exhibiting abundant homogenous matrix and intact structure without a necrotic core. Compared with typical culture, the cell yield in CC increases 2.57 times, and the expression of cartilage marker collagen type II increases 4.70 times. Transcriptomics reveal that this step-wise culture drives a proliferation-to-differentiation process through an intermediate plastic stage, and CC-chondrocytes undergo a chondral lineage-specific differentiation with an activated metabolism. Animal studies show that CC macro-cartilage maintains a hyaline-like cartilage phenotype in vivo and significantly promotes the healing of large cartilage defects. Overall, an efficient expansion of human macro-cartilage with superior regenerative plasticity is achieved, providing a promising strategy for joint regeneration.
Collapse
|
34
|
Loder S, Patel N, Morgani S, Sambon M, Leucht P, Levi B. Genetic models for lineage tracing in musculoskeletal development, injury, and healing. Bone 2023; 173:116777. [PMID: 37156345 PMCID: PMC10860167 DOI: 10.1016/j.bone.2023.116777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/07/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
Musculoskeletal development and later post-natal homeostasis are highly dynamic processes, marked by rapid structural and functional changes across very short periods of time. Adult anatomy and physiology are derived from pre-existing cellular and biochemical states. Consequently, these early developmental states guide and predict the future of the system as a whole. Tools have been developed to mark, trace, and follow specific cells and their progeny either from one developmental state to the next or between circumstances of health and disease. There are now many such technologies alongside a library of molecular markers which may be utilized in conjunction to allow for precise development of unique cell 'lineages'. In this review, we first describe the development of the musculoskeletal system beginning as an embryonic germ layer and at each of the key developmental stages that follow. We then discuss these structures in the context of adult tissues during homeostasis, injury, and repair. Special focus is given in each of these sections to the key genes involved which may serve as markers of lineage or later in post-natal tissues. We then finish with a technical assessment of lineage tracing and the techniques and technologies currently used to mark cells, tissues, and structures within the musculoskeletal system.
Collapse
Affiliation(s)
- Shawn Loder
- Department of Plastic Surgery, University of Pittsburgh, Scaife Hall, Suite 6B, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | - Nicole Patel
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | - Benjamin Levi
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
35
|
Kouroupis D, Kaplan LD, Huard J, Best TM. CD10-Bound Human Mesenchymal Stem/Stromal Cell-Derived Small Extracellular Vesicles Possess Immunomodulatory Cargo and Maintain Cartilage Homeostasis under Inflammatory Conditions. Cells 2023; 12:1824. [PMID: 37508489 PMCID: PMC10377825 DOI: 10.3390/cells12141824] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/23/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
The onset and progression of human inflammatory joint diseases are strongly associated with the activation of resident synovium/infrapatellar fat pad (IFP) pro-inflammatory and pain-transmitting signaling. We recently reported that intra-articularly injected IFP-derived mesenchymal stem/stromal cells (IFP-MSC) acquire a potent immunomodulatory phenotype and actively degrade substance P (SP) via neutral endopeptidase CD10 (neprilysin). Our hypothesis is that IFP-MSC robust immunomodulatory therapeutic effects are largely exerted via their CD10-bound small extracellular vesicles (IFP-MSC sEVs) by attenuating synoviocyte pro-inflammatory activation and articular cartilage degradation. Herein, IFP-MSC sEVs were isolated from CD10High- and CD10Low-expressing IFP-MSC cultures and their sEV miRNA cargo was assessed using multiplex methods. Functionally, we interrogated the effect of CD10High and CD10Low sEVs on stimulated by inflammatory/fibrotic cues synoviocyte monocultures and cocultures with IFP-MSC-derived chondropellets. Finally, CD10High sEVs were tested in vivo for their therapeutic capacity in an animal model of acute synovitis/fat pad fibrosis. Our results showed that CD10High and CD10Low sEVs possess distinct miRNA profiles. Reactome analysis of miRNAs highly present in sEVs showed their involvement in the regulation of six gene groups, particularly those involving the immune system. Stimulated synoviocytes exposed to IFP-MSC sEVs demonstrated significantly reduced proliferation and altered inflammation-related molecular profiles compared to control stimulated synoviocytes. Importantly, CD10High sEV treatment of stimulated chondropellets/synoviocyte cocultures indicated significant chondroprotective effects. Therapeutically, CD10High sEV treatment resulted in robust chondroprotective effects by retaining articular cartilage structure/composition and PRG4 (lubricin)-expressing cartilage cells in the animal model of acute synovitis/IFP fibrosis. Our study suggests that CD10High sEVs possess immunomodulatory miRNA attributes with strong chondroprotective/anabolic effects for articular cartilage in vivo. The results could serve as a foundation for sEV-based therapeutics for the resolution of detrimental aspects of immune-mediated inflammatory joint changes associated with conditions such as osteoarthritis (OA).
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA (T.M.B.)
- Diabetes Research Institute & Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lee D. Kaplan
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA (T.M.B.)
| | - Johnny Huard
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA;
| | - Thomas M. Best
- Department of Orthopaedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA (T.M.B.)
| |
Collapse
|
36
|
Mantsou A, Papachristou E, Keramidas P, Lamprou P, Pitou M, Papi RM, Dimitriou K, Aggeli A, Choli-Papadopoulou T. Fabrication of a Smart Fibrous Biomaterial That Harbors an Active TGF-β1 Peptide: A Promising Approach for Cartilage Regeneration. Biomedicines 2023; 11:1890. [PMID: 37509529 PMCID: PMC10377373 DOI: 10.3390/biomedicines11071890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
The regeneration of articular cartilage remains a serious problem in various pathological conditions such as osteoarthritis, due to the tissue's low self-healing capacity. The latest therapeutic approaches focus on the construction of biomaterials that induce cartilage repair. This research describes the design, synthesis, and investigation of a safe, "smart", fibrous scaffold containing a genetically incorporated active peptide for chondrogenic induction. While possessing specific sequences and the respective mechanical properties from natural fibrous proteins, the fibers also incorporate a Transforming Growth Factor-β1 (TGF-β1)-derived peptide (YYVGRKPK) that can promote chondrogenesis. The scaffold formed stable porous networks with shear-thinning properties at 37 °C, as shown by SEM imaging and rheological characterization, and were proven to be non-toxic to human dental pulp stem cells (hDPSCs). Its chondrogenic capacity was evidenced by a strong increase in the expression of specific chondrogenesis gene markers SOX9, COL2, ACAN, TGFBR1A, and TGFBR2 in cells cultured on "scaffold-TGFβ1" for 21 days and by increased phosphorylation of intracellular signaling proteins Smad-2 and Erk-1/2. Additionally, intense staining of glycosaminoglycans was observed in these cells. According to our results, "scaffold-TGFβ1" is proposed for clinical studies as a safe, injectable treatment for cartilage degeneration.
Collapse
Affiliation(s)
- Aglaia Mantsou
- Laboratory of Biochemistry, School of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Eleni Papachristou
- Laboratory of Biochemistry, School of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Panagiotis Keramidas
- Laboratory of Biochemistry, School of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Paraskevas Lamprou
- Laboratory of Biochemistry, School of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Maria Pitou
- Laboratory of Biochemistry, School of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Rigini M Papi
- Laboratory of Biochemistry, School of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Katerina Dimitriou
- Laboratory of Chemical Engineering A', School of Chemical Engineering, Faculty of Engineering, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Amalia Aggeli
- Laboratory of Chemical Engineering A', School of Chemical Engineering, Faculty of Engineering, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Theodora Choli-Papadopoulou
- Laboratory of Biochemistry, School of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| |
Collapse
|
37
|
Zujur D, Al-Akashi Z, Nakamura A, Zhao C, Takahashi K, Aritomi S, Theoputra W, Kamiya D, Nakayama K, Ikeya M. Enhanced chondrogenic differentiation of iPS cell-derived mesenchymal stem/stromal cells via neural crest cell induction for hyaline cartilage repair. Front Cell Dev Biol 2023; 11:1140717. [PMID: 37234772 PMCID: PMC10206169 DOI: 10.3389/fcell.2023.1140717] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Background: To date, there is no effective long-lasting treatment for cartilage tissue repair. Primary chondrocytes and mesenchymal stem/stromal cells are the most commonly used cell sources in regenerative medicine. However, both cell types have limitations, such as dedifferentiation, donor morbidity, and limited expansion. Here, we report a stepwise differentiation method to generate matrix-rich cartilage spheroids from induced pluripotent stem cell-derived mesenchymal stem/stromal cells (iMSCs) via the induction of neural crest cells under xeno-free conditions. Methods: The genes and signaling pathways regulating the chondrogenic susceptibility of iMSCs generated under different conditions were studied. Enhanced chondrogenic differentiation was achieved using a combination of growth factors and small-molecule inducers. Results: We demonstrated that the use of a thienoindazole derivative, TD-198946, synergistically improves chondrogenesis in iMSCs. The proposed strategy produced controlled-size spheroids and increased cartilage extracellular matrix production with no signs of dedifferentiation, fibrotic cartilage formation, or hypertrophy in vivo. Conclusion: These findings provide a novel cell source for stem cell-based cartilage repair. Furthermore, since chondrogenic spheroids have the potential to fuse within a few days, they can be used as building blocks for biofabrication of larger cartilage tissues using technologies such as the Kenzan Bioprinting method.
Collapse
Affiliation(s)
- Denise Zujur
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Ziadoon Al-Akashi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Anna Nakamura
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan
| | - Chengzhu Zhao
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Kazuma Takahashi
- Research Institute for Bioscience Product and Fine Chemicals, Ajinomoto Co., Inc, Kawasaki, Japan
| | - Shizuka Aritomi
- Research Institute for Bioscience Product and Fine Chemicals, Ajinomoto Co., Inc, Kawasaki, Japan
| | - William Theoputra
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Daisuke Kamiya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Takeda-CiRA Joint Program (T-CiRA), Kanagawa, Japan
| | - Koichi Nakayama
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan
| | - Makoto Ikeya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Takeda-CiRA Joint Program (T-CiRA), Kanagawa, Japan
| |
Collapse
|
38
|
Mazzella M, Walker K, Cormier C, Kapanowski M, Ishmakej A, Saifee A, Govind Y, Chaudhry GR. WNT and VEGF/PDGF signaling regulate self-renewal in primitive mesenchymal stem cells. RESEARCH SQUARE 2023:rs.3.rs-2512048. [PMID: 37090660 PMCID: PMC10120760 DOI: 10.21203/rs.3.rs-2512048/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Background Therapeutic use of multipotent mesenchymal stem cells (MSCs) is hampered due to poor growth and limited self-renewal potential. The self-renewal potential of MSCs is also affected during propagation and changes are poorly understood. This study investigated the molecular mechanism involved in the self-renewal of primitive (p) MSCs. Methods pMSCs were cultured to low passage (LP), P3, and high passage (HP), P20, in fetal bovine serum medium (FM) and xeno-free medium (XM). The characteristics of LP and HP pMSCs were evaluated for morphology, expression of cell surface markers, doubling time (DT), colony forming efficiency (CFE), proliferation by BrdU assay, telomerase activity and trilineage differentiation. We then examined transcriptome and nucleosome occupancies using RNA-seq and MNase-seq, respectively analyses. Results pMSCs grown in FM gradually changed morphology to large elongated cells and showed a significant reduction in the expression of CD90 and CD49f, CFE, proliferation, and telomerase activity. In addition, cells had a greater propensity to differentiate into the adipogenic lineage. In contrast, pMSCs grown in XM maintained small fibroblastoid morphology, self-renewal, and differentiation potential. Transcriptomic analysis showed upregulation of genes involved in self-renewal, cell cycle, and DNA replication in XM-grown pMSCs. Whereas senescence genes were upregulated in cells in FM. MNase-seq analysis revealed less nucleosomal occupancies in self-renewal genes and senescence genes in pMSCs grown in XM and FM, respectively. The expression of selected genes associated with self-renewal, cell cycle, DNA replication, differentiation, and senescence was confirmed by qRT-PCR. These results led us to propose signaling pathways involved in the self-renewal and senescence of pMSCs. Conclusion We conclude that the self-renewal potential of pMSCs is controlled by WNT and VEGF/PDGF, but TGFβ and PI3K signaling induce senescence.
Collapse
|
39
|
Loh HY, Norman BP, Lai KS, Cheng WH, Nik Abd Rahman NMA, Mohamed Alitheen NB, Osman MA. Post-Transcriptional Regulatory Crosstalk between MicroRNAs and Canonical TGF-β/BMP Signalling Cascades on Osteoblast Lineage: A Comprehensive Review. Int J Mol Sci 2023; 24:ijms24076423. [PMID: 37047394 PMCID: PMC10094338 DOI: 10.3390/ijms24076423] [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: 11/07/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 04/14/2023] Open
Abstract
MicroRNAs (miRNAs) are a family of small, single-stranded, and non-protein coding RNAs about 19 to 22 nucleotides in length, that have been reported to have important roles in the control of bone development. MiRNAs have a strong influence on osteoblast differentiation through stages of lineage commitment and maturation, as well as via controlling the activities of osteogenic signal transduction pathways. Generally, miRNAs may modulate cell stemness, proliferation, differentiation, and apoptosis by binding the 3'-untranslated regions (3'-UTRs) of the target genes, which then can subsequently undergo messenger RNA (mRNA) degradation or protein translational repression. MiRNAs manage the gene expression in osteogenic differentiation by regulating multiple signalling cascades and essential transcription factors, including the transforming growth factor-beta (TGF-β)/bone morphogenic protein (BMP), Wingless/Int-1(Wnt)/β-catenin, Notch, and Hedgehog signalling pathways; the Runt-related transcription factor 2 (RUNX2); and osterix (Osx). This shows that miRNAs are essential in regulating diverse osteoblast cell functions. TGF-βs and BMPs transduce signals and exert diverse functions in osteoblastogenesis, skeletal development and bone formation, bone homeostasis, and diseases. Herein, we highlighted the current state of in vitro and in vivo research describing miRNA regulation on the canonical TGF-β/BMP signalling, their effects on osteoblast linage, and understand their mechanism of action for the development of possible therapeutics. In this review, particular attention and comprehensive database searches are focused on related works published between the years 2000 to 2022, using the resources from PubMed, Google Scholar, Scopus, and Web of Science.
Collapse
Affiliation(s)
- Hui-Yi Loh
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Brendan P Norman
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Kok-Song Lai
- Health Sciences Division, Abu Dhabi Women's College, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Wan-Hee Cheng
- Faculty of Health and Life Sciences, INTI International University, Persiaran Perdana BBN, Putra Nilai, Nilai 71800, Negeri Sembilan, Malaysia
| | - Nik Mohd Afizan Nik Abd Rahman
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Noorjahan Banu Mohamed Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Mohd Azuraidi Osman
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
40
|
Richard D, Pregizer S, Venkatasubramanian D, Raftery RM, Muthuirulan P, Liu Z, Capellini TD, Craft AM. Lineage-specific differences and regulatory networks governing human chondrocyte development. eLife 2023; 12:e79925. [PMID: 36920035 PMCID: PMC10069868 DOI: 10.7554/elife.79925] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 03/14/2023] [Indexed: 03/16/2023] Open
Abstract
To address large gaps in our understanding of the molecular regulation of articular and growth plate cartilage development in humans, we used our directed differentiation approach to generate these distinct cartilage tissues from human embryonic stem cells. The resulting transcriptomic profiles of hESC-derived articular and growth plate chondrocytes were similar to fetal epiphyseal and growth plate chondrocytes, with respect to genes both known and previously unknown to cartilage biology. With the goal to characterize the regulatory landscapes accompanying these respective transcriptomes, we mapped chromatin accessibility in hESC-derived chondrocyte lineages, and mouse embryonic chondrocytes, using ATAC-sequencing. Integration of the expression dataset with the differentially accessible genomic regions revealed lineage-specific gene regulatory networks. We validated functional interactions of two transcription factors (TFs) (RUNX2 in growth plate chondrocytes and RELA in articular chondrocytes) with their predicted genomic targets. The maps we provide thus represent a framework for probing regulatory interactions governing chondrocyte differentiation. This work constitutes a substantial step towards comprehensive and comparative molecular characterizations of distinct chondrogenic lineages and sheds new light on human cartilage development and biology.
Collapse
Affiliation(s)
- Daniel Richard
- Human Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Steven Pregizer
- Department of Orthopedic Research, Boston Children’s HospitalBostonUnited States
- Department of Orthopedic Surgery, Harvard Medical SchoolBostonUnited States
| | - Divya Venkatasubramanian
- Department of Orthopedic Research, Boston Children’s HospitalBostonUnited States
- Department of Orthopedic Surgery, Harvard Medical SchoolBostonUnited States
- Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| | - Rosanne M Raftery
- Department of Orthopedic Research, Boston Children’s HospitalBostonUnited States
- Department of Orthopedic Surgery, Harvard Medical SchoolBostonUnited States
| | | | - Zun Liu
- Human Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Terence D Capellini
- Human Evolutionary Biology, Harvard UniversityCambridgeUnited States
- Broad Institute of MIT and HarvardCambridgeUnited States
| | - April M Craft
- Department of Orthopedic Research, Boston Children’s HospitalBostonUnited States
- Department of Orthopedic Surgery, Harvard Medical SchoolBostonUnited States
- Harvard Stem Cell InstituteCambridgeUnited States
| |
Collapse
|
41
|
Conditioned Medium - Is it an Undervalued Lab Waste with the Potential for Osteoarthritis Management? Stem Cell Rev Rep 2023:10.1007/s12015-023-10517-1. [PMID: 36790694 PMCID: PMC10366316 DOI: 10.1007/s12015-023-10517-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND The approaches currently used in osteoarthritis (OA) are mainly short-term solutions with unsatisfactory outcomes. Cell-based therapies are still controversial (in terms of the sources of cells and the results) and require strict culture protocol, quality control, and may have side-effects. A distinct population of stromal cells has an interesting secretome composition that is underrated and commonly ends up as biological waste. Their unique properties could be used to improve the existing techniques due to protective and anti-ageing properties. SCOPE OF REVIEW In this review, we seek to outline the advantages of the use of conditioned media (CM) and exosomes, which render them superior to other cell-based methods, and to summarise current information on the composition of CM and their effect on chondrocytes. MAJOR CONCLUSIONS CM are obtainable from a variety of mesenchymal stromal cell (MSC) sources, such as adipose tissue, bone marrow and umbilical cord, which is significant to their composition. The components present in CMs include proteins, cytokines, growth factors, chemokines, lipids and ncRNA with a variety of functions. In most in vitro and in vivo studies CM from MSCs had a beneficial effect in enhance processes associated with chondrocyte OA pathomechanism. GENERAL SIGNIFICANCE This review summarises the information available in the literature on the function of components most commonly detected in MSC-conditioned media, as well as the effect of CM on OA chondrocytes in in vitro culture. It also highlights the need to standardise protocols for obtaining CM, and to conduct clinical trials to transfer the effects obtained in vitro to human subjects.
Collapse
|
42
|
Tu B, Li J, Sun Z, Zhang T, Liu H, Yuan F, Fan C. Macrophage-Derived TGF-β and VEGF Promote the Progression of Trauma-Induced Heterotopic Ossification. Inflammation 2023; 46:202-216. [PMID: 35986177 DOI: 10.1007/s10753-022-01723-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/17/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022]
Abstract
Heterotopic ossification (HO) is a pathological bone formation process caused by musculoskeletal trauma. HO is characterized by aberrant endochondral ossification and angiogenesis. Our previous studies have indicated that macrophage inflammation is involved in traumatic HO formation. In this study, we found that macrophage infiltration and TGF-β signaling activation are presented in human HO. Depletion of macrophages effectively suppressed traumatic HO formation in a HO mice model, and macrophage depletion significantly inhibited the activation of TGF-β/Smad2/3 signaling. In addition, the TGF-β blockade created by a neutralizing antibody impeded ectopic bone formation in vivo. Notably, endochondral ossification and angiogenesis are attenuated following macrophage depletion or TGF-β inhibition. Furthermore, our observations on macrophage polarization revealed that M2 macrophages, rather than M1 macrophages, play a critical role in supporting HO development by enhancing the osteogenic and chondrogenic differentiation of mesenchymal stem cells. Our findings on ectopic bone formation in HO patients and the mice model indicate that M2 macrophages are an important contributor for HO development, and that inhibition of M2 polarization or TGF-β activity may be a potential method of therapy for traumatic HO.
Collapse
Affiliation(s)
- Bing Tu
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Juehong Li
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Ziyang Sun
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Tongtong Zhang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Hang Liu
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Feng Yuan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
43
|
Hasan MR, Koskenranta A, Alakurtti K, Takatalo M, Rice DP. RAB23 regulates musculoskeletal development and patterning. Front Cell Dev Biol 2023; 11:1049131. [PMID: 36910145 PMCID: PMC9995984 DOI: 10.3389/fcell.2023.1049131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
RAB23 is a small GTPase which functions at the plasma membrane to regulate growth factor signaling. Mutations in RAB23 cause Carpenter syndrome, a condition that affects normal organogenesis and patterning. In this study, we investigate the role of RAB23 in musculoskeletal development and show that it is required for patella bone formation and for the maintenance of tendon progenitors. The patella is the largest sesamoid bone in mammals and plays a critical role during movement by providing structural and mechanical support to the knee. Rab23 -/- mice fail to form a patella and normal knee joint. The patella is formed from Sox9 and scleraxis (Scx) double-positive chondroprogenitor cells. We show that RAB23 is required for the specification of SOX9 and scleraxis double-positive patella chondroprogenitors during the formation of patella anlagen and the subsequent establishment of patellofemoral joint. We find that scleraxis and SOX9 expression are disrupted in Rab23 -/- mice, and as a result, development of the quadriceps tendons, cruciate ligaments, patella tendons, and entheses is either abnormal or lost. TGFβ-BMP signaling is known to regulate patella initiation and patella progenitor differentiation and growth. We find that the expression of TGFβR2, BMPR1, BMP4, and pSmad are barely detectable in the future patella site and in the rudimentary tendons and ligaments around the patellofemoral joint in Rab23 -/- mice. Also, we show that GLI1, SOX9, and scleraxis, which regulate entheses establishment and maturation, are weakly expressed in Rab23 -/- mice. Further analysis of the skeletal phenotype of Rab23 -/- mice showed a close resemblance to that of Tgfβ2 -/- mice, highlighting a possible role for RAB23 in regulating TGFβ superfamily signaling.
Collapse
Affiliation(s)
- Md Rakibul Hasan
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Anna Koskenranta
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Kirsi Alakurtti
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Maarit Takatalo
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - David P Rice
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland.,Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
44
|
Li DX, Ma Z, Szojka ARA, Lan X, Kunze M, Mulet-Sierra A, Westover L, Adesida AB. Non-hypertrophic chondrogenesis of mesenchymal stem cells through mechano-hypoxia programing. J Tissue Eng 2023; 14:20417314231172574. [PMID: 37216035 PMCID: PMC10192798 DOI: 10.1177/20417314231172574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/09/2023] [Indexed: 05/24/2023] Open
Abstract
Cartilage tissue engineering aims to generate functional replacements to treat cartilage defects from damage and osteoarthritis. Human bone marrow-derived mesenchymal stem cells (hBM-MSC) are a promising cell source for making cartilage, but current differentiation protocols require the supplementation of growth factors like TGF-β1 or -β3. This can lead to undesirable hypertrophic differentiation of hBM-MSC that progress to bone. We have found previously that exposing engineered human meniscus tissues to physiologically relevant conditions of the knee (mechanical loading and hypoxia; hence, mechano-hypoxia conditioning) increased the gene expression of hyaline cartilage markers, SOX9 and COL2A1, inhibited hypertrophic marker COL10A1, and promoted bulk mechanical property development. Adding further to this protocol, we hypothesize that combined mechano-hypoxia conditioning with TGF-β3 growth factor withdrawal will promote stable, non-hypertrophic chondrogenesis of hBM-MSC embedded in an HA-hydrogel. We found that the combined treatment upregulated many cartilage matrix- and development-related markers while suppressing many hypertrophic- and bone development-related markers. Tissue level assessments with biochemical assays, immunofluorescence, and histochemical staining confirmed the gene expression data. Further, mechanical property development in the dynamic compression treatment shows promise toward generating functional engineered cartilage through more optimized and longer culture conditions. In summary, this study introduced a novel protocol to differentiate hBM-MSC into stable, cartilage-forming cells.
Collapse
Affiliation(s)
- David Xinzheyang Li
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Civil and Environmental
Engineering, Faculty of Engineering, AB, University of Alberta, Edmonton, AB,
Canada
| | - Zhiyao Ma
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Alexander RA Szojka
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Xiaoyi Lan
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Civil and Environmental
Engineering, Faculty of Engineering, AB, University of Alberta, Edmonton, AB,
Canada
| | - Melanie Kunze
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Lindsey Westover
- Department of Mechanical Engineering,
Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Adetola B Adesida
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
45
|
Ragni E, Perucca Orfei C, De Luca P, Libonati F, de Girolamo L. Tissue-Protective and Anti-Inflammatory Landmark of PRP-Treated Mesenchymal Stromal Cells Secretome for Osteoarthritis. Int J Mol Sci 2022; 23:ijms232415908. [PMID: 36555578 PMCID: PMC9788137 DOI: 10.3390/ijms232415908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Bone-marrow-mesenchymal-stromal-cells (BMSCs)- and platelet-rich-plasma (PRP)-based therapies have shown potential for treating osteoarthritis (OA). Recently, the combination of these two approaches was proposed, with results that overcame those observed with the separate treatments, indicating a possible role of PRP in ameliorating BMSCs' regenerative properties. Since a molecular fingerprint of BMSCs cultivated in the presence of PRP is missing, the aim of this study was to characterize the secretome in terms of soluble factors and extracellular-vesicle (EV)-embedded miRNAs from the perspective of tissues, pathways, and molecules which frame OA pathology. One hundred and five soluble factors and one hundred eighty-four EV-miRNAs were identified in the PRP-treated BMSCs' secretome, respectively. Several soluble factors were related to the migration of OA-related immune cells, suggesting the capacity of BMSCs to attract lympho-, mono-, and granulocytes and modulate their inflammatory status. Accordingly, several EV-miRNAs had an immunomodulating role at both the single-factor and cell level, together with the ability to target OA-characterizing extracellular-matrix-degrading enzymes and cartilage destruction pathways. Overall, anti-inflammatory and protective signals far exceeded inflammation and destruction cues for cartilage, macrophages, and T cells. This study demonstrates that BMSCs cultivated in the presence of PRP release therapeutic molecules and give molecular ground for the use of this combined and innovative therapy for OA treatment.
Collapse
|
46
|
Hsieh CC, Yen BL, Chang CC, Hsu PJ, Lee YW, Yen ML, Yet SF, Chen L. Wnt antagonism without TGFβ induces rapid MSC chondrogenesis via increasing AJ interactions and restricting lineage commitment. iScience 2022; 26:105713. [PMID: 36582823 PMCID: PMC9792887 DOI: 10.1016/j.isci.2022.105713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 07/26/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Human mesenchymal stem cells (MSCs) remain one of the best cell sources for cartilage, a tissue without regenerative capacity. However, MSC chondrogenesis is commonly induced through TGFβ, a pleomorphic growth factor without specificity for this lineage. Using tissue- and induced pluripotent stem cell-derived MSCs, we demonstrate an efficient and precise approach to induce chondrogenesis through Wnt/β-catenin antagonism alone without TGFβ. Compared to TGFβ, Wnt/β-catenin antagonism more rapidly induced MSC chondrogenesis without eliciting off-target lineage specification toward smooth muscle or hypertrophy; this was mediated through increasing N-cadherin levels and β-catenin interactions-key components of the adherens junctions (AJ)-and increasing cytoskeleton-mediated condensation. Validation with transcriptomic analysis of human chondrocytes compared to MSCs and osteoblasts showed significant downregulation of Wnt/β-catenin and TGFβ signaling along with upregulation of α-catenin as an upstream regulator. Our findings underscore the importance of understanding developmental pathways and structural modifications in achieving efficient MSC chondrogenesis for translational application.
Collapse
Affiliation(s)
- Chen-Chan Hsieh
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, Miaoli County35053, Taiwan
| | - B. Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, Miaoli County35053, Taiwan
- Corresponding author
| | - Chia-Chi Chang
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, Miaoli County35053, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center (NDMC), Taipei, Taiwan
| | - Pei-Ju Hsu
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, Miaoli County35053, Taiwan
| | - Yu-Wei Lee
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, Miaoli County35053, Taiwan
| | - Men-Luh Yen
- Department of Obstetrics/Gynecology, National Taiwan University (NTU) Hospital and College of Medicine, NTU, Taipei, Taiwan
| | - Shaw-Fang Yet
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, Miaoli County35053, Taiwan
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
47
|
Xiong Y, Mi BB, Lin Z, Hu YQ, Yu L, Zha KK, Panayi AC, Yu T, Chen L, Liu ZP, Patel A, Feng Q, Zhou SH, Liu GH. The role of the immune microenvironment in bone, cartilage, and soft tissue regeneration: from mechanism to therapeutic opportunity. Mil Med Res 2022; 9:65. [PMID: 36401295 PMCID: PMC9675067 DOI: 10.1186/s40779-022-00426-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/30/2022] [Indexed: 11/21/2022] Open
Abstract
Bone, cartilage, and soft tissue regeneration is a complex spatiotemporal process recruiting a variety of cell types, whose activity and interplay must be precisely mediated for effective healing post-injury. Although extensive strides have been made in the understanding of the immune microenvironment processes governing bone, cartilage, and soft tissue regeneration, effective clinical translation of these mechanisms remains a challenge. Regulation of the immune microenvironment is increasingly becoming a favorable target for bone, cartilage, and soft tissue regeneration; therefore, an in-depth understanding of the communication between immune cells and functional tissue cells would be valuable. Herein, we review the regulatory role of the immune microenvironment in the promotion and maintenance of stem cell states in the context of bone, cartilage, and soft tissue repair and regeneration. We discuss the roles of various immune cell subsets in bone, cartilage, and soft tissue repair and regeneration processes and introduce novel strategies, for example, biomaterial-targeting of immune cell activity, aimed at regulating healing. Understanding the mechanisms of the crosstalk between the immune microenvironment and regeneration pathways may shed light on new therapeutic opportunities for enhancing bone, cartilage, and soft tissue regeneration through regulation of the immune microenvironment.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Bo-Bin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ze Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yi-Qiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Kang-Kang Zha
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.,Key Laboratory of Biorheological Science and Technology,Ministry of Education College of Bioengineering, Chongqing University, Shapingba, Chongqing, 400044, China
| | - Adriana C Panayi
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
| | - Tao Yu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.,Department of Physics, Center for Hybrid Nanostructure (CHyN), University of Hamburg, Hamburg, 22761, Germany
| | - Zhen-Ping Liu
- Department of Physics, Center for Hybrid Nanostructure (CHyN), University of Hamburg, Hamburg, 22761, Germany.,Joint Laboratory of Optofluidic Technology and System,National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, 510006, China
| | - Anish Patel
- Skeletal Biology Laboratory, Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02120, USA
| | - Qian Feng
- Key Laboratory of Biorheological Science and Technology,Ministry of Education College of Bioengineering, Chongqing University, Shapingba, Chongqing, 400044, China.
| | - Shuan-Hu Zhou
- Skeletal Biology Laboratory, Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02120, USA. .,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.
| | - Guo-Hui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
48
|
Ragni E, Perucca Orfei C, de Girolamo L. Secreted Factors and Extracellular Vesicles Account for the Immunomodulatory and Tissue Regenerative Properties of Bone-Marrow-Derived Mesenchymal Stromal Cells for Osteoarthritis. Cells 2022; 11:3501. [PMID: 36359897 PMCID: PMC9658264 DOI: 10.3390/cells11213501] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 07/28/2023] Open
Abstract
Bone-marrow-derived mesenchymal stromal cells (BMSCs) showed therapeutic potential in the treatment of musculoskeletal diseases, including osteoarthritis (OA). Their soluble mediators and extracellular vesicles (EVs), which make up the secretome, suppress immune response, attenuate inflammation and promote cartilage repair. EVs, as well as the whole secretome, have been investigated as cell free approaches for OA although, to date, a disease-tailored molecular fingerprint is missing. In this study, soluble mediators and miRNAs were sifted in the BMSCs' secretome and EVs, respectively, and analyzed in the frame of cell types and factors involved in OA. The majority of identified molecules repress the activation of immune cells and the production of OA-related inflammatory mediators, as well as promote cartilage protection by acting on both chondrocytes homeostasis and extracellular matrix-degrading enzymes. These data provide the molecular ground for the therapeutic potential of BMSCs for regenerative applications for OA and support the use of secretome or EVs as cell-free applications in joint diseases.
Collapse
|
49
|
Cho S, Choi H, Jeong H, Kwon SY, Roh EJ, Jeong KH, Baek I, Kim BJ, Lee SH, Han I, Cha JM. Preclinical Study of Human Bone Marrow-Derived Mesenchymal Stem Cells Using a 3-Dimensional Manufacturing Setting for Enhancing Spinal Fusion. Stem Cells Transl Med 2022; 11:1072-1088. [PMID: 36180050 PMCID: PMC9585955 DOI: 10.1093/stcltm/szac052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/12/2022] [Indexed: 11/29/2022] Open
Abstract
Spinal fusion surgery is a surgical technique that connects one or more vertebrae at the same time to prevent movement between the vertebrae. Although synthetic bone substitutes or osteogenesis-inducing recombinant proteins were introduced to promote bone union, the rate of revision surgery is still high due to pseudarthrosis. To promote successful fusion after surgery, stem cells with or without biomaterials were introduced; however, conventional 2D-culture environments have resulted in a considerable loss of the innate therapeutic properties of stem cells. Therefore, we conducted a preclinical study applying 3D-spheroids of human bone marrow-dewrived mesenchymal stem cells (MSCs) to a mouse spinal fusion model. First, we built a large-scale manufacturing platform for MSC spheroids, which is applicable to good manufacturing practice (GMP). Comprehensive biomolecular examinations, which include liquid chromatography-mass spectrometry and bioinformatics could suggest a framework of quality control (QC) standards for the MSC spheroid product regarding the identity, purity, viability, and potency. In our animal study, the mass-produced and quality-controlled MSC spheroids, either undifferentiated or osteogenically differentiated were well-integrated into decorticated bone of the lumbar spine, and efficiently improved angiogenesis, bone regeneration, and mechanical stability with statistical significance compared to 2D-cultured MSCs. This study proposes a GMP-applicable bioprocessing platform and QC directions of MSC spheroids aiming for their clinical application in spinal fusion surgery as a new bone graft substitute.
Collapse
Affiliation(s)
- Sumin Cho
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea.,3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology, Incheon National University, Incheon, Republic of Korea
| | - Hyemin Choi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hyundoo Jeong
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea
| | - Su Yeon Kwon
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Eun Ji Roh
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kwang-Hun Jeong
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea.,3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology, Incheon National University, Incheon, Republic of Korea
| | - Inho Baek
- Department of Biomedical Technology, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Byoung Ju Kim
- Department of Biomedical Technology, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Technology, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jae Min Cha
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea.,3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology, Incheon National University, Incheon, Republic of Korea
| |
Collapse
|
50
|
Cui J, Shibata Y, Itaka K, Zhou J, Zhang J. Unbiased comparison and modularization identify time-related transcriptomic reprogramming in exercised rat cartilage: Integrated data mining and experimental validation. Front Physiol 2022; 13:974266. [PMID: 36187764 PMCID: PMC9520919 DOI: 10.3389/fphys.2022.974266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Exercise is indispensable for maintaining cartilage integrity in healthy joints and remains a recommendation for knee osteoarthritis. Although the effects of exercise on cartilage have been implied, the detailed mechanisms, such as the effect of exercise time which is important for exercise prescription, remain elusive. In this study, bioinformatic analyses, including unbiased comparisons and modularization, were performed on the transcriptomic data of rat cartilage to identify the time-related genes and signaling pathways. We found that exercise had a notable effect on cartilage transcriptome. Exercise prominently suppressed the genes related to cell division, hypertrophy, catabolism, inflammation, and immune response. The downregulated genes were more prominent and stable over time than the upregulated genes. Although exercise time did not prominently contribute to the effects of exercise, it was a factor related to a batch of cellular functions and signaling pathways, such as extracellular matrix (ECM) homeostasis and cellular response to growth factors and stress. Two clusters of genes, including early and late response genes, were identified according to the expression pattern over time. ECM organization, BMP signaling, and PI3K-Akt signaling were early responsive in the exercise duration. Moreover, time-related signaling pathways, such as inositol phosphate metabolism, nicotinate/nicotinamide metabolism, cell cycle, and Fc epsilon RI signaling pathway, were identified by unbiased mapping and polarization of the highly time-correlated genes. Immunohistochemistry staining showed that Egfr was a late response gene that increased on day 15 of exercise. This study elucidated time-related transcriptomic reprogramming induced by exercise in cartilage, advancing the understanding of cartilage homeostasis.
Collapse
Affiliation(s)
- Jiarui Cui
- School of Rehabilitation and Health Preservation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yo Shibata
- Department of Conservative Dentistry, Division of Biomaterials and Engineering, Showa University School of Dentistry, Tokyo, Japan
| | - Keiji Itaka
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Jun Zhou
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- *Correspondence: Jun Zhou, ; Jiaming Zhang,
| | - Jiaming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jun Zhou, ; Jiaming Zhang,
| |
Collapse
|