1
|
Le QB, Ezhilarasu H, Chan WW, Patra AT, Murugan P, Venkatesh SA, Tay YK, Lim SR, Abdul Rahim AA, Lee JSZ, Bi X, Choudhury D. A platform for Bioengineering Tissue Membranes from cell spheroids. Mater Today Bio 2025; 31:101526. [PMID: 40026618 PMCID: PMC11869014 DOI: 10.1016/j.mtbio.2025.101526] [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: 10/08/2024] [Revised: 01/16/2025] [Accepted: 01/26/2025] [Indexed: 03/05/2025] Open
Abstract
Cell spheroids are essential building blocks for engineering tissues like cartilage, bone, liver, cardiac, pancreatic, and neural tissues, but controlling their fusion and organisation is challenging. Spheroids tend to fuse into a larger mass, impeding nutrient and waste diffusion. To overcome this, we developed a method to assemble spheroids into a thin layer by using two mesh scaffolds to spread them evenly, and a solid frame with grid to secure the assembly. This allows the spheroids to fuse into a thin membrane-like tissue, allowing better medium diffusion during cell culture. We demonstrated this method by producing cartilage tissue membranes from human mesenchymal stem cell spheroids undergoing chondrogenic differentiation, evaluating spheroid sizes, assembly timing, fusion process and membrane thickness. Our method is a versatile platform for producing tissue membranes from cell spheroids, with significant potential in tissue engineering for creating functional tissue constructs from various cell types.
Collapse
Affiliation(s)
- Quang Bach Le
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
| | - Hariharan Ezhilarasu
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
| | - Weng Wan Chan
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
| | - Alok Tanala Patra
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
| | - Priya Murugan
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
| | - Shashaank Abhinav Venkatesh
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
| | - Yean Kai Tay
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
| | - Shin Ru Lim
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
| | - Ahmad Amirul Abdul Rahim
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
| | - Jia Sheng Zach Lee
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
| | - Xuezhi Bi
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
| | - Deepak Choudhury
- Biomanufacturing Technology (BMT), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A∗STAR), 20 Biopolis Way, 138668, Singapore
- Department of Food Science and Technology, National University of Singapore, Singapore
| |
Collapse
|
2
|
Choudhery MS, Arif T, Mahmood R, Mushtaq A, Niaz A, Hassan Z, Zahid H, Nayab P, Arshad I, Arif M, Majid M, Harris DT. Induced Mesenchymal Stem Cells: An Emerging Source for Regenerative Medicine Applications. J Clin Med 2025; 14:2053. [PMID: 40142860 PMCID: PMC11943107 DOI: 10.3390/jcm14062053] [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: 02/17/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Regenerative medicine is gaining interest in the medical field due to the limitations of conventional treatments, which often fail to address the underlying cause of disease. In recent years, stem cell-based therapies have evolved as a promising alternative approach to treat those diseases that cannot be cured using conventional medicine. Adult stem cells, particularly the mesenchymal stem cells (MSCs), have attracted a lot of attention due to their ability to regenerate and repair human tissues and organs. MSCs isolated from adult tissues are well characterized and are currently the most common type of cells for use in regenerative medicine. However, their low number in adult donor tissues, donor-age and cell-source related heterogeneity, limited proliferative and differentiation potential, and early senescence in in vitro cultures, negatively affect MSC regenerative potential. These factors restrict MSC use for research as well as for clinical applications. To overcome these problems, MSCs with superior regenerative potential are required. Induced MSCs (iMSCs) are obtained from induced pluripotent stem cells (iPSCs). These cells are patient-specific, readily available, and have relatively superior regenerative potential and, therefore, can overcome the problems associated with the use of primary MSCs. In this review, the authors aim to discuss the characteristics, regenerative potential, and limitations of MSCs for regenerative medicine applications. The main methods to generate iMSCs from iPSCs have been discussed in detail. In addition, the proposed criteria for their molecular characterization, applications of iMSCs for disease modeling and drug discovery, as well as potential use in regenerative medicine have been explored in detail.
Collapse
Affiliation(s)
- Mahmood S. Choudhery
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Taqdees Arif
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Ruhma Mahmood
- Department of Pediatric Surgery, Allama Iqbal Medical College, Jinnah Hospital, Lahore 54700, Pakistan;
| | - Asad Mushtaq
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Ahmad Niaz
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Zaeema Hassan
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Hamda Zahid
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Pakeeza Nayab
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Iqra Arshad
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Mehak Arif
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Mashaim Majid
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - David T. Harris
- Department of Immunobiology, University of Arizona Health Sciences Biorepository, College of Medicine, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
3
|
Kanda T, Iwasaki K, Taguchi Y, Umeda M. Role of sodium-dependent vitamin C transporter 2 in human periodontal ligament fibroblasts. J Periodontal Res 2025; 60:265-277. [PMID: 39225294 DOI: 10.1111/jre.13322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 09/04/2024]
Abstract
AIM Ascorbic acid (AA) is a water-soluble vitamin that has antioxidant properties and regulates homeostasis of connective tissue through controlling various enzymatic activities. Two cell surface glycoproteins, sodium-dependent vitamin C transporter (SVCT) 1 and SVCT2, are known as ascorbate transporters. The purpose of this study was to investigate the expression pattern and functions of SVCTs in periodontal ligament (PDL) and PDL fibroblast (PDLF). METHODS Gene expression was examined using real-time polymerase chain reaction (PCR) and reverse transcription PCR. SVCT2 expression was determined by immunofluorescence staining, western blot and flow cytometry. ALP activity and collagen production were examined using ALP staining and collagen staining. Short interfering RNA was used to knock down the gene level of SVCT2. Change of comprehensive gene expression under SVCT2 knockdown condition was examined by RNA-sequencing analysis. RESULTS Real-time PCR, fluorescent immunostaining, western blot and flowy cytometry showed that SVCT2 was expressed in PDLF and PDL. ALP activity, collagen production, and SVCT2 expression were enhanced upon AA stimulation in PDLF. The enhancement of ALP activity, collagen production, and SVCT2 expression by AA was abolished under SVCT2 knockdown condition. RNA-sequencing revealed that gene expression of CLDN4, Cyclin E2, CAMK4, MSH5, DMC1, and Nidgen2 were changed by SVCT2 knockdown. Among them, the expression of MSH5 and DMC1, which are related to DNA damage sensor activity, was enhanced by AA, suggesting the new molecular target of AA in PDLF. CONCLUSION Our study reveals the SVCT2 expression in PDL and the pivotal role of SVCT2 in mediating AA-induced enhancements of ALP activity and collagen production in PDLF. Additionally, we identify alterations in gene expression profiles, highlighting potential molecular targets influenced by AA through SVCT2. These findings deepen our understanding of periodontal tissue homeostasis mechanisms and suggest promising intervention targeting AA metabolism.
Collapse
Affiliation(s)
- Tomoko Kanda
- Graduate School of Dentistry (Department of Periodontology), Osaka Dental University, Osaka, Japan
| | - Kengo Iwasaki
- Division of Creative and Integrated Medicine, Advanced Medicine Research Center, Translational Research Institute for Medical Innovation (TRIMI), Osaka Dental University, Osaka, Japan
| | - Yoichiro Taguchi
- Department of Periodontology, Osaka Dental University, Osaka, Japan
| | - Makoto Umeda
- Department of Periodontology, Osaka Dental University, Osaka, Japan
| |
Collapse
|
4
|
Tam HY, Liu J, Yiu TC, Leung AOW, Li C, Gu S, Rennert O, Huang B, Cheung HH. Amelioration of premature aging in Werner syndrome stem cells by targeting SHIP/AKT pathway. Cell Biosci 2025; 15:10. [PMID: 39863890 PMCID: PMC11765919 DOI: 10.1186/s13578-025-01355-4] [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: 08/20/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Pathogenic or null mutations in WRN helicase is a cause of premature aging disease Werner syndrome (WS). WRN is known to protect somatic cells including adult stem cells from premature senescence. Loss of WRN in mesenchymal stem cells (MSCs) not only drives the cells to premature senescence but also significantly impairs the function of the stem cells in tissue repair or regeneration. RESULTS In this study, we profiled the signaling pathways altered in WRN-deficient MSC and applied pharmacological method to activate the AKT signaling in these cells and examined their cellular phenotype related to aging. We found that the AKT signaling in WRN-deficient MSCs was significantly suppressed while the AKT upstream phosphatases (SHIP1/2) were upregulated. Knockdown or inhibition of SHIP1/2 could ameliorate premature senescence in WRN-deficient MSCs. Moreover, SHIP inhibition stimulated MSC proliferation and suppressed expression of pro-inflammatory cytokines IL-6 and IL-8. The stemness of WRN-deficient MSC was also improved upon pharmacological treatments with the inhibitors. CONCLUSIONS These results suggested that targeting the SHIP/AKT signaling pathway is beneficial to WRN-deficient stem cells and fibroblasts, which might be applied for improving the trophic function of MSC in, for instance, promoting angiogenesis.
Collapse
Affiliation(s)
- Hei-Yin Tam
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong S.A.R., China
| | - Jiaxing Liu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong S.A.R., China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong, China
| | - Tsz-Ching Yiu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong S.A.R., China
| | - Adrian On-Wah Leung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong S.A.R., China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong, China
| | - Chang Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong S.A.R., China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong, China
| | - Shen Gu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong S.A.R., China
| | - Owen Rennert
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, USA
| | - Boxian Huang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Hoi-Hung Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong S.A.R., China.
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong S.A.R., China.
| |
Collapse
|
5
|
Migliorini F, Pilone M, Ascani J, Schäfer L, Jeyaraman M, Maffulli N. Management of knee osteoarthritis using bone marrow aspirate concentrate: a systematic review. Br Med Bull 2025; 153:ldae016. [PMID: 39506910 DOI: 10.1093/bmb/ldae016] [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: 01/11/2024] [Revised: 08/20/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024]
Abstract
INTRODUCTION Knee osteoarthritis (OA) is a common degenerative joint condition and a major cause of disability in the general population. SOURCE OF DATA Recent published literature identified from PubMed, EMBASE, Google Scholar, and Scopus. AREAS OF AGREEMENT Orthobiological therapies try to regenerate articular cartilage and stop the progression of the degenerative lesion. Intra-articular injections of biological derivates have been increasingly used in the last decade. AREAS OF CONTROVERSY The indications for the use of bone marrow aspirate concentrate (BMAC) are still unclear. GROWING POINTS We systematically reviewed the current literature on BMAC in the management of knee OA, giving an update on the current indications for the selection of the ideal patient and the preparations and efficacy of BMAC compared to other biological alternatives. AREAS TIMELY FOR DEVELOPING RESEARCH BMAC is a valuable source of mesenchymal stem cells, offering potential benefits in attenuating the inflammatory pathway associated with knee OA. Intra-articular injection of BMAC has shown effectiveness in clinical trials improving functional outcomes of knee OA patients. The superiority of BMAC over other orthobiological treatments cannot be assessed because of conflicting results.
Collapse
Affiliation(s)
- Filippo Migliorini
- Department of Orthopaedic and Trauma Surgery, Academic Hospital of Bolzano (SABES-ASDAA), Via Lorenz Böhler 5, 39100, Bolzano, Italy
- Department of Life Sciences, Health, and Health Professions, Link Campus University, Via del Casale di San Pio V, 00165 Rome, Italy
| | - Marco Pilone
- Residency Program in Orthopedics and Traumatology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Jacopo Ascani
- Department of Trauma and Orthopaedic Surgery, Faculty of Medicine and Psychology, University "La Sapienza" of Rome, Via di Grottarossa 1035, 00189 Roma, Italy
| | - Luise Schäfer
- Department of Orthopaedic and Trauma Surgery, Academic Hospital of Bolzano (SABES-ASDAA), Via Lorenz Böhler 5, 39100, Bolzano, Italy
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Poonamallee High Rd, Velappanchavadi, Chennai 600077, Tamil Nadu, India
| | - Nicola Maffulli
- Department of Trauma and Orthopaedic Surgery, Faculty of Medicine and Psychology, University "La Sapienza" of Rome, Via di Grottarossa 1035, 00189 Roma, Italy
- School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, Stoke on Trent ST4 7QB, United Kingdom
- Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Mile End Hospital, 275 Bancroft Road, E1 4DG London, UK
| |
Collapse
|
6
|
Chathoth BM, Helmholz H, Angrisani N, Wiese B, Reifenrath J, Willumeit-Römer R. Investigating the Potential of Magnesium Microparticles on Cartilage and Bone Regeneration Utilizing an In Vitro Osteoarthritis Model. J Biomed Mater Res A 2025; 113:e37862. [PMID: 39719870 DOI: 10.1002/jbm.a.37862] [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: 06/28/2024] [Revised: 12/06/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
Osteoarthritis (OA) is a significant condition that profoundly impacts synovial joints, including cartilage and subchondral bone plate. Biomaterials that can impede OA progression are a promising alternative or supplement to anti-inflammatory and surgical interventions. Magnesium (Mg) alloys known for bone regeneration potential were assessed in the form of Mg microparticles regarding their impact on tissue regeneration and prevention of OA progression. In vitro assays based on mesenchymal stem cells (SCP-1) were applied to evaluate the Mg microparticle's compatibility and function. Biocompatibility documented through live-dead staining and lactate dehydrogenase assay revealed a 90% cell viability at a concentration below 10 mM after 3 days of exposure. An in vitro OA model based on the supplementation of the cytokines IL-1β, and TNF-α was established and disclosed the effect of Mg degradation products in differentiating SCP-1 cells. Sustained differentiation was confirmed through extracellular matrix staining and increased gene marker expression. The Mg supplementation reduced the release of inflammatory cytokines (IL-6 and IL-8) while promoting the expression of proteins such as collagen X, collagen I, and osteopontin in a time-dependent manner. The in vitro study suggests that Mg microparticles hold a therapeutic potential for OA treatment with their ability to support bone and cartilage repair mechanisms even under inflammatory conditions.
Collapse
Affiliation(s)
| | - Heike Helmholz
- Helmholtz Zentrum Hereon, Institute of Metallic Biomaterials, Geesthacht, Germany
| | - Nina Angrisani
- Hannover Medical School, Department of Orthopedic Surgery, DIAKOVERE Annastift, Hannover, Germany
| | - Björn Wiese
- Helmholtz Zentrum Hereon, Institute of Metallic Biomaterials, Geesthacht, Germany
| | - Janin Reifenrath
- Hannover Medical School, Department of Orthopedic Surgery, DIAKOVERE Annastift, Hannover, Germany
| | | |
Collapse
|
7
|
Zhong Y, Zhang B, Somoza R, Caplan AI, Welter JF, Baskaran H. Amino Acid Uptake Limitations during Human Mesenchymal Stem Cell-Based Chondrogenesis. Tissue Eng Part A 2025; 31:1-12. [PMID: 38517098 PMCID: PMC11807877 DOI: 10.1089/ten.tea.2024.0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/12/2024] [Indexed: 03/23/2024] Open
Abstract
A mino acids are the essential building blocks for collagen and proteoglycan, which are the main constituents for cartilage extracellular matrix (ECM). Synthesis of ECM proteins requires the uptake of various essential/nonessential amino acids. Analyzing amino acid metabolism during chondrogenesis can help to relate tissue quality to amino acid metabolism under different conditions. In our study, we studied amino acid uptake/secretion using human mesenchymal stem cell (hMSC)-based aggregate chondrogenesis in a serum-free induction medium with a defined chemical formulation. The initial glucose level and medium-change frequency were varied. Our results showed that essential amino acid uptake increased with time during hMSCs chondrogenesis for all initial glucose levels and medium-change frequencies. Essential amino acid uptake rates were initial glucose-level independent. The DNA-normalized glycosaminoglycans and hydroxyproline content of chondrogenic aggregates correlated with cumulative uptake of leucine, valine, and tryptophan regardless of initial glucose levels and medium-change frequencies. Collectively, our results show that amino acid uptake rates during in vitro chondrogenesis were insufficient to produce a tissue with an ECM content similar to that of human neonatal cartilage or adult cartilage. Furthermore, this deficiency was likely related to the downregulation of some key amino acid transporters in the cells. Such deficiency could be partially improved by increasing the amino acid availability in the chondrogenic medium by changing culture conditions.
Collapse
Affiliation(s)
- Yi Zhong
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| | - Bo Zhang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| | - Rodrigo Somoza
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| | - Arnold I. Caplan
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| | - Jean F. Welter
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| | - Harihara Baskaran
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Modular Manufacturing of Structural Tissues (CMOST), Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
8
|
Sawyer M, Semodji A, Nielson O, Rektor A, Burgoyne H, Eppel M, Eixenberger J, Montenegro-Brown R, Nelson ML, Lujan T, Estrada D. Direct Scaffold-Coupled Electrical Stimulation of Chondrogenic Progenitor Cells through Graphene Foam Bioscaffolds to Control Mechanical Properties of Graphene Foam - Cell Composites. RESEARCH SQUARE 2024:rs.3.rs-5589589. [PMID: 39764126 PMCID: PMC11703340 DOI: 10.21203/rs.3.rs-5589589/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Osteoarthritis, a major global cause of pain and disability, is driven by the irreversible degradation of hyaline cartilage in joints. Cartilage tissue engineering presents a promising therapeutic avenue, but success hinges on replicating the native physiological environment to guide cellular behavior and generate tissue constructs that mimic natural cartilage. Although electrical stimulation has been shown to enhance chondrogenesis and extracellular matrix production in 2D cultures, the mechanisms underlying these effects remain poorly understood, particularly in 3D models. Here, we report that direct scaffold-coupled electrical stimulation applied to 3D graphene foam bioscaffolds significantly enhances the mechanical properties of the resulting graphene foam - cell constructs. Using custom 3D-printed electrical stimulus chambers, we applied biphasic square impulses (20, 40, 60 mVpp at 1 kHz) for 5 minutes daily over 7 days. Stimulation at 60 mVpp increased the steady-state energy dissipation and equilibrium modulus by approximately 65% and 25%, respectively, compared to unstimulated controls, while also yielding the highest cell density among stimulated samples. In addition, our custom chambers facilitated full submersion of the hydrophobic graphene foam in media, leading to enhanced cell attachment and integration across the scaffold surface and within its hollow branches. To assess this cellular integration, we employed co-localized confocal fluorescence microscopy and X-ray microCT imaging enabled by colloidal gold nanoparticle and fluorophore staining, which allowed visualization of cell distribution within the opaque scaffold's internal structure. These findings highlight the potential of direct scaffold-coupled electrical stimulus to modulate the mechanical properties of engineered tissues and offer new insights into the emergent behavior of cells within conductive 3D bioscaffolds.
Collapse
|
9
|
Wu KC, Chang YH, Ding DC, Lin SZ. Mesenchymal Stromal Cells for Aging Cartilage Regeneration: A Review. Int J Mol Sci 2024; 25:12911. [PMID: 39684619 PMCID: PMC11641625 DOI: 10.3390/ijms252312911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Cartilage degeneration is a key feature of aging and osteoarthritis, characterized by the progressive deterioration of joint function, pain, and limited mobility. Current treatments focus on symptom relief, not cartilage regeneration. Mesenchymal stromal cells (MSCs) offer a promising therapeutic option due to their capability to differentiate into chondrocytes, modulate inflammation, and promote tissue regeneration. This review explores the potential of MSCs for cartilage regeneration, examining their biological properties, action mechanisms, and applications in preclinical and clinical settings. MSCs derived from bone marrow, adipose tissue, and other sources can self-renew and differentiate into multiple cell types. In aging cartilage, they aid in tissue regeneration by secreting growth factors and cytokines that enhance repair and modulate immune responses. Recent preclinical studies show that MSCs can restore cartilage integrity, reduce inflammation, and improve joint function, although clinical translation remains challenging due to limitations such as cell viability, scalability, and regulatory concerns. Advancements in MSC delivery, including scaffold-based approaches and engineered exosomes, may improve therapeutic effectiveness. Potential risks, such as tumorigenicity and immune rejection, are also discussed, emphasizing the need for optimized treatment protocols and large-scale clinical trials to develop effective, minimally invasive therapies for cartilage regeneration.
Collapse
Affiliation(s)
- Kun-Chi Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
- Institute of Medical Sciences, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
10
|
Tee CA, Roxby DN, Othman R, Denslin V, Bhat KS, Yang Z, Han J, Tucker-Kellogg L, Boyer LA. Metabolic modulation to improve MSC expansion and therapeutic potential for articular cartilage repair. Stem Cell Res Ther 2024; 15:308. [PMID: 39285485 PMCID: PMC11406821 DOI: 10.1186/s13287-024-03923-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Articular cartilage degeneration can result from injury, age, or arthritis, causing significant joint pain and disability without surgical intervention. Currently, the only FDA cell-based therapy for articular cartilage injury is Autologous Chondrocyte Implantation (ACI); however, this procedure is costly, time-intensive, and requires multiple treatments. Mesenchymal stromal cells (MSCs) are an attractive alternative autologous therapy due to their availability and ability to robustly differentiate into chondrocytes for transplantation with good safety profiles. However, treatment outcomes are variable due to donor-to-donor variability as well as intrapopulation heterogeneity and unstandardized MSC manufacturing protocols. Process improvements that reduce cell heterogeneity while increasing donor cell numbers with improved chondrogenic potential during expansion culture are needed to realize the full potential of MSC therapy. METHODS In this study, we investigated the potential of MSC metabolic modulation during expansion to enhance their chondrogenic commitment by varying the nutrient composition, including glucose, pyruvate, glutamine, and ascorbic acid in culture media. We tested the effect of metabolic modulation in short-term (one passage) and long-term (up to seven passages). We measured metabolic state, cell size, population doubling time, and senescence and employed novel tools including micro-magnetic resonance relaxometry (µMRR) relaxation time (T2) to characterize the effects of AA on improved MSC expansion and chondrogenic potential. RESULTS Our data show that the addition of 1 mM L-ascorbic acid-2-phosphate (AA) to cultures for one passage during MSC expansion prior to initiation of differentiation improves chondrogenic differentiation. We further demonstrate that AA treatment reduced the proportion of senescent cells and cell heterogeneity also allowing for long-term expansion that led to a > 300-fold increase in yield of MSCs with enhanced chondrogenic potential compared to untreated cells. AA-treated MSCs with improved chondrogenic potential showed a robust shift in metabolic profile to OXPHOS and higher µMRR T2 values, identifying critical quality attributes that could be implemented in MSC manufacturing for articular cartilage repair. CONCLUSIONS Our results suggest an improved MSC manufacturing process that can enhance chondrogenic potential by targeting MSC metabolism and integrating process analytic tools during expansion.
Collapse
Affiliation(s)
- Ching Ann Tee
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
| | - Daniel Ninio Roxby
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
| | - Rashidah Othman
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
| | - Vinitha Denslin
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore, 117510, Republic of Singapore
| | - Kiesar Sideeq Bhat
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
- Department of Bioresources, University of Kashmir, Hazratbal, Srinagar, 190006, India
| | - Zheng Yang
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore, 117510, Republic of Singapore
- Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block 11, Singapore, 119288, Republic of Singapore
| | - Jongyoon Han
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 50 Vassar St, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Lisa Tucker-Kellogg
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore.
- Cancer and Stem Cell Biology and Centre for Computational Biology, Duke-NUS Medical School, 8 College Rd, Singapore, 169857, Republic of Singapore.
| | - Laurie A Boyer
- Critical Analytics for Manufacturing Personalised-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1 Create Way, Enterprise Wing, #04-13/14, Singapore, 138602, Republic of Singapore.
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
- Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| |
Collapse
|
11
|
Yao L, Flynn N, Kaphle P. Effects of cell culture time and cytokines on migration of dental pulp stem cell-derived chondrogenic cells in collagen hydrogels. Physiol Rep 2024; 12:e70063. [PMID: 39327065 PMCID: PMC11427086 DOI: 10.14814/phy2.70063] [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: 02/26/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
The transplantation of collagen hydrogels encapsulating human dental pulp stem cell (DPSC)-derived chondrogenic cells is potentially a novel approach for the regeneration of degenerated nucleus pulposus (NP) and cartilage. Grafted cell migration allows cells to disperse in the hydrogels and the treated tissue from the grafted location. We previously reported the cell migration in type I and type II hydrogels. It is important to explore further how cell culture time affect the cell motility. In this study, we observed the decreased motility of DPSC-derived chondrogenic cells after culturing for 2 weeks compared with cells cultured for 2 days in these gels. The Alamarblue assay showed the cell proliferation during the two-week cell culture period. The findings suggest that the transitions of cell motility and proliferation during the longer culture time. The result indicates that the early culture stage is an optimal time for cell transplantation. In a degenerated disc, the expression of IL-1β and TNFα increased significantly compared with healthy tissue and therefore may affect grafted cell migration. The incorporation of IL-1β and TNFα into the collagen hydrogels decreased cell motility. The study indicates that the control of IL-1β and TNFα production may help to maintain cell motility after transplantation.
Collapse
Affiliation(s)
- Li Yao
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| | - Nikol Flynn
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| | - Pranita Kaphle
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| |
Collapse
|
12
|
Zhang B, Berilla J, Cho S, Somoza RA, Welter JF, Alexander PE, Baskaran H. Synergistic effects of biological stimuli and flexion induce microcavities promote hypertrophy and inhibit chondrogenesis during in vitro culture of human mesenchymal stem cell aggregates. Biotechnol J 2024; 19:e2400060. [PMID: 39295570 PMCID: PMC11870314 DOI: 10.1002/biot.202400060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/26/2024] [Accepted: 07/30/2024] [Indexed: 09/21/2024]
Abstract
Interzone/cavitation are key steps in early stage joint formation that have not been successfully developed in vitro. Further, current models of endochondral ossification, an important step in early bone formation, lack key morphology morphological structures such as microcavities found during development in vivo. This is possibly due to the lack of appropriate strategies for incorporating chemical and mechanical stimuli that are thought to be involved in joint development. We designed a bioreactor system and investigated the synergic effect of chemical stimuli (chondrogenesis-inducing [CIM] and hypertrophy-inducing medium [HIM]) and mechanical stimuli (flexion) on the growth of human mesenchymal stem cells (hMSCs) based linear aggregates under different conditions over 4 weeks of perfusion culture. Computational studies were used to evaluate tissue stress qualitatively. After harvesting, both Safranin-O and hematoxylin & eosin (H&E) staining histology demonstrated microcavity structures and void structures in the region of higher stresses for tissue aggregates cultured only in HIM under flexion. In comparison to either HIM treatment or flexion only, increased glycosaminoglycan (GAG) content in the extracellular matrix (ECM) at this region indicates the morphological change resembles the early stage of joint cavitation; while decreased type II collagen (Col II), and increased type X collagen (Col X) and vascular endothelial growth factor (VEGF) with a clear boundary in the staining section indicates it resembles the early stage of ossification. Further, cell alignment analysis indicated that cells were mostly oriented toward the direction of flexion in high-stress region only in HIM under flexion, resembling cell morphology in both joint cavitation and hypertrophic cartilage in growth plate. Collectively, our results suggest that flexion and HIM inhibit chondrogenesis and promote hypertrophy and development of microcavities that resemble the early stage of joint cavitation and endochondral ossification. We believe the tissue model described in this work can be used to develop in vitro models of joint tissue for applications such as pathophysiology and drug discovery.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jim Berilla
- Case School of Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sungwoo Cho
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - Rodrigo A Somoza
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jean F Welter
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Peter E Alexander
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Harihara Baskaran
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
13
|
Yaqoob F, Hayat MK, Chaughtai MS, Khan S, Bashir MB. Mesenchymal stem cells derived from human adipose tissue exhibit significantly higher chondrogenic differentiation potential compared to those from rats. Biomed Mater Eng 2024:BME240062. [PMID: 39240621 DOI: 10.3233/bme-240062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
BACKGROUND Osteoarthritis is a prevalent joint disease affecting both humans and animals. It is characterized by articular cartilage degeneration and joint surface eburnation. Currently, no effective pharmacological treatment is available to restore the original function and structure of defective cartilage. OBJECTIVE This study explores the potential of stem cell-based therapy in treating joint diseases involving cartilage degeneration, offering a promising avenue for future research and treatment. The primary aim was to compare the characteristics and, more importantly, the chondrogenic differentiation potential of human and rat adipose-derived mesenchymal stem cells (AD-MSCs). METHODS Rat adipose tissue was collected from Sprague Dawley rats, while human adipose tissue was obtained in the form of lipoaspirate. The mesenchymal stem cells (MSCs) were then harvested using collagenase enzyme and subcultured. We meticulously evaluated and compared the cell morphology, percentage of cell viability, population doubling time, metabolic proliferation, and chondrogenic differentiation potential of MSCs harvested from both sources. Chondrogenic differentiation was induced at passage 3 using the 3D pellet culture method and assessed through histological and molecular analysis. RESULTS The findings revealed that human and rat AD-MSCs were phenotypically identical, and an insignificant difference was found in cell morphology, percentage of cell viability, metabolic proliferation, and population doubling time. However, the chondrogenic differentiation potential of human AD-MSCs was evaluated as significantly higher than that of rat AD-MSCs. CONCLUSION The current study suggests that research regarding chondrogenic differentiation of rat AD-MSCs can be effectively translated to humans. This discovery is a significant contribution to the field of regenerative medicine and has the potential to advance our understanding of stem cell-based therapy for joint diseases.
Collapse
Affiliation(s)
- Faisal Yaqoob
- Institute of Biochemistry and Biotechnology, University of Veterinary & Animal Sciences, Lahore, Pakistan
- Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Muhammad Khizer Hayat
- Center for Animal Diagnostics, Chughtai Lab, Lahore, Pakistan
- Department of Pathology, University of Veterinary & Animal Sciences, Lahore, Pakistan
| | - Muhammad Sharjeel Chaughtai
- Department of Pathology, University of Veterinary & Animal Sciences, Lahore, Pakistan
- Department of Farm Animals & Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Sehrish Khan
- Department of Clinical Medicine & Surgery, University of Veterinary & Animal Sciences, Lahore, Pakistan
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Musa Bin Bashir
- Department of Internal Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
14
|
Hoffman J, Zheng S, Zhang H, Murphy RF, Dahl KN. Image-based discrimination of the early stages of mesenchymal stem cell differentiation. Mol Biol Cell 2024; 35:ar103. [PMID: 38837346 PMCID: PMC11321037 DOI: 10.1091/mbc.e24-02-0095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are self-renewing, multipotent cells, which can be used in cellular and tissue therapeutics. MSCs cell number can be expanded in vitro, but premature differentiation results in reduced cell number and compromised therapeutic efficacies. Current techniques fail to discriminate the "stem-like" population from early stages (12 h) of differentiated MSC population. Here, we imaged nuclear structure and actin architecture using immunofluorescence and used deep learning-based computer vision technology to discriminate the early stages (6-12 h) of MSC differentiation. Convolutional neural network models trained by nucleus and actin images have high accuracy in reporting MSC differentiation; nuclear images alone can identify early stages of differentiation. Concurrently, we show that chromatin fluidity and heterochromatin levels or localization change during early MSC differentiation. This study quantifies changes in cell architecture during early MSC differentiation and describes a novel image-based diagnostic tool that could be widely used in MSC culture, expansion and utilization.
Collapse
Affiliation(s)
- Justin Hoffman
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Shiyuan Zheng
- Department of Biomedical Engineering, College of Engineering, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Huaiying Zhang
- Department of Biological Sciences, Mellon College of Science, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Robert F. Murphy
- Department of Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Kris Noel Dahl
- Department of Biomedical Engineering, College of Engineering, Carnegie Mellon University, Pittsburgh, PA 15213
| |
Collapse
|
15
|
Ravera F, Efeoglu E, Byrne HJ. A comparative analysis of stem cell differentiation on 2D and 3D substrates using Raman microspectroscopy. Analyst 2024; 149:4041-4053. [PMID: 38973486 DOI: 10.1039/d4an00315b] [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: 07/09/2024]
Abstract
Chondrogenesis is a complex cellular process that involves the transformation of mesenchymal stem cells (MSCs) into chondrocytes, the specialised cells that form cartilage. In recent years, three-dimensional (3D) culture systems have emerged as a promising approach to studying cell behaviour and development in a more physiologically relevant environment compared to traditional two-dimensional (2D) cell culture. The use of these systems provided insights into the molecular mechanisms that regulate chondrogenesis and has the potential to revolutionise the development of new therapies for cartilage repair and regeneration. This study demonstrates the successful application of Raman microspectroscopy (RMS) as a label-free, non-destructive, and sensitive method to monitor the chondrogenic differentiation of bone marrow-derived rat mesenchymal stem cells (rMSCs) in a collagen type I hydrogel, and explores the potential benefits of 3D hydrogels compared to conventional 2D cell culture environments. rMSCs were cultured on 3D substrates for 3 weeks and their differentiation was monitored by measuring the spectral signatures of their subcellular compartments. Additionally, the evolution of high-density micromass cultures was investigated to provide a comprehensive understanding of the process and complex interactions between cells and their surrounding extracellular matrix. For comparison, rMSCs were induced into chondrogenesis in identical medium conditions for 21 days in monolayer culture. Raman spectra showed that rMSCs cultured in a collagen type I hydrogel are able to undergo a distinct chondrogenic differentiation pathway at a significantly higher rate than the 2D culture cells. 3D cultures expressed stronger and more homogeneous chondrogenesis-associated peaks such as collagens, glycosaminoglycans (GAGs), and aggrecan while manifesting changes in proteins and lipidic content. These results suggest that 3D type I collagen hydrogel substrates are promising for in vitro chondrogenesis studies, and that RMS is a valuable tool for monitoring chondrogenesis in 3D environments.
Collapse
Affiliation(s)
- F Ravera
- FOCAS Research Institute, Technological University Dublin, City Campus, Dublin 8, Ireland.
| | - E Efeoglu
- NICB (National Institute for Cellular Biotechnology) at Dublin City University, Dublin 9, Ireland
| | - H J Byrne
- FOCAS Research Institute, Technological University Dublin, City Campus, Dublin 8, Ireland.
| |
Collapse
|
16
|
Hasan SMK, Islam SR, Zerin I, Ahmed T, Rahman S. Gelatin/EGDE Ultrafine Composite Fibers Reinforced with 3D Spacer Fabric as Bicomponent Scaffolds for Tissue Engineering. ACS APPLIED BIO MATERIALS 2024; 7:4593-4601. [PMID: 38914048 DOI: 10.1021/acsabm.4c00469] [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: 06/26/2024]
Abstract
Protein-based ultrafine fibrous scaffolds can mimic the native extracellular matrices (ECMs) with regard to the morphology and chemical composition but suffer from poor mechanical and wet stability. As a result, cells cannot get a true three-dimensional (3D) environment as they find in native ECMs. In this study, an epoxide, ethylene glycol diglycidylether (EGDE), with high reactivity to active hydrogen is introduced to gelatin solution, serving as an effective cross-linker. The gelatin/EGDE 3D-ultrafine (∼500 nm in diameter) fibrous composite scaffolds are made by an ultralow-concentration phase separation technique (ULCPS). The effects of the polymer content and modification conditions on the morphology and wet stability of the constructs are investigated. It is revealed that ultrafine fibers with 3D random orientation could be formed at low concentrations (0.01, 0.05, and 0.1 wt %, respectively). The wet stability of the constructs could be effectively improved by introducing EGDE into the gelatin system. The shrinkage is reduced to merely 2.14% after the modification at 120 °C for 2 h and could be maintained for up to 3 days. In order to improve the compression properties, the same technique is utilized with the presence of a poly(lactic acid) (PLA) spacer fabric to produce a bicomponent scaffold. The mechanical property and cell viability of the bicomponent scaffolds are investigated, and it is found that cells could enter deep inside and orient themselves randomly at the central area of the bicomponent scaffold. The modification and design approach presented in this study has the potential to provide various protein-based ultrafine fibrous biomaterials for a variety of biomedical applications.
Collapse
Affiliation(s)
- S M Kamrul Hasan
- Department of Textile Engineering, National Institute of Textile Engineering and Research (NITER), University of Dhaka, Dhaka 1350, Bangladesh
- Shanghai Frontier Science Research Center for Modern Textiles, College of Textiles, Donghua University, Shanghai 201620, China
- Department of Fashion and Textiles, School of Design and Social Context, RMIT University, 25 Dawson Street, Brunswick, Victoria 3054, Australia
| | - Syed Rashedul Islam
- Department of Textile Engineering, National Institute of Textile Engineering and Research (NITER), University of Dhaka, Dhaka 1350, Bangladesh
- Shanghai Frontier Science Research Center for Modern Textiles, College of Textiles, Donghua University, Shanghai 201620, China
- Department of Textile Engineering, Apparel Manufacture and Technology, BGMEA University of Fashion and Technology, Dhaka 1230, Bangladesh
| | - Ismat Zerin
- Department of Textile Engineering, National Institute of Textile Engineering and Research (NITER), University of Dhaka, Dhaka 1350, Bangladesh
| | - Toufique Ahmed
- Department of Textile Engineering, National Institute of Textile Engineering and Research (NITER), University of Dhaka, Dhaka 1350, Bangladesh
| | - Sadikur Rahman
- Department of Textile Engineering, National Institute of Textile Engineering and Research (NITER), University of Dhaka, Dhaka 1350, Bangladesh
| |
Collapse
|
17
|
Saleh AS, Abdel-Gabbar M, Gabr H, Shams A, Tamur S, Mahdi EA, Ahmed OM. Ameliorative effects of undifferentiated and differentiated BM-MSCs in MIA-induced osteoarthritic Wistar rats: roles of NF-κB and MMPs signaling pathways. Am J Transl Res 2024; 16:2793-2813. [PMID: 39114694 PMCID: PMC11301505 DOI: 10.62347/fghv2647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/15/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVES Osteoarthritis (OA) is a degenerative joint condition that is persistent. OA affects millions of people throughout the world. Both people and society are heavily economically burdened by osteoarthritis. There is currently no medication that can structurally alter the OA processes or stop the disease from progressing. Stem cells have the potential to revolutionize medicine due to their capacity to differentiate into chondrocytes, capacity to heal tissues and organs including osteoarthritic joints, and immunomodulatory capabilities. Therefore, the goal of the current investigation was to determine how bone marrow-derived mesenchymal stem cells (BM-MSCs) and chondrogenic differentiated mesenchymal stem cells (CD-MSCs) affected the treatment of OA in rats with monosodium iodoacetate (MIA)-induced osteoarthritis. METHODS Male Wistar rats were injected three times with MIA (1 mg)/100 µL isotonic saline to induce osteoarthritis in the ankle joint of the right hind leg. Following the MIA injection, the osteoarthritic rats were given weekly treatments of 1 × 106 BM-MSCs and CD-MSCs into the tail vein for three weeks. RESULTS The obtained results showed that in osteoarthritic rats, BM-MSCs and CD-MSCs dramatically decreased ankle diameter measurements, decreased oxidized glutathione (GSSG) level, and boosted glutathione peroxidase (GPx) and glutathione reductase (GR) activities. Additionally, in rats with MIA-induced OA, BM-MSCs and CD-MSCs dramatically boosted interleukin-10 (IL-10) serum levels while considerably decreasing serum anticitrullinated protein antibodies (ACPA), tumour necrosis factor-α (TNF-α), and interleukin-17 (IL-17) levels as well as ankle transforming growth factor-β1 (TGF-β1) expression. Analysis of histology, immunohistochemistry, and western blots in osteoarthritic joints showed that cartilage breakdown and joint inflammation gradually decreased over time. CONCLUSIONS It is possible to conclude from these results that BM-MSCs and CD-MSCs have anti-arthritic potential in MIA-induced OA, which may be mediated via inhibitory effects on oxidative stress, MMPs and inflammation through suppressing the NF-κB pathway. In osteoarthritis, using CD-MSCs as a treatment is more beneficial therapeutically than using BM-MSCs.
Collapse
Affiliation(s)
- Ablaa S Saleh
- Department of Biochemistry, Faculty of Science, Beni-Suef UniversityBeni-Suef 62521, Egypt
| | - Mohammed Abdel-Gabbar
- Department of Biochemistry, Faculty of Science, Beni-Suef UniversityBeni-Suef 62521, Egypt
| | - Hala Gabr
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo UniversityCairo 11435, Egypt
| | - Anwar Shams
- Department of Pharmacology, College of Medicine, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif UniversityTaif 26432, Saudi Arabia
- High Altitude Research Center, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
| | - Shadi Tamur
- Department of Pediatric, College of Medicine, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
| | - Emad A Mahdi
- Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef UniversityBeni-Suef 62521, Egypt
| | - Osama M Ahmed
- Division of Physiology, Department of Zoology, Faculty of Science, Beni-Suef UniversityBeni-Suef 62521, Egypt
| |
Collapse
|
18
|
Velikova T, Dekova T, Miteva DG. Controversies regarding transplantation of mesenchymal stem cells. World J Transplant 2024; 14:90554. [PMID: 38947963 PMCID: PMC11212595 DOI: 10.5500/wjt.v14.i2.90554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/07/2024] [Accepted: 04/03/2024] [Indexed: 06/13/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have tantalized regenerative medicine with their therapeutic potential, yet a cloud of controversies looms over their clinical transplantation. This comprehensive review navigates the intricate landscape of MSC controversies, drawing upon 15 years of clinical experience and research. We delve into the fundamental properties of MSCs, exploring their unique immunomodulatory capabilities and surface markers. The heart of our inquiry lies in the controversial applications of MSC transplantation, including the perennial debate between autologous and allogeneic sources, concerns about efficacy, and lingering safety apprehensions. Moreover, we unravel the enigmatic mechanisms surrounding MSC transplantation, such as homing, integration, and the delicate balance between differentiation and paracrine effects. We also assess the current status of clinical trials and the ever-evolving regulatory landscape. As we peer into the future, we examine emerging trends, envisioning personalized medicine and innovative delivery methods. Our review provides a balanced and informed perspective on the controversies, offering readers a clear understanding of the complexities, challenges, and potential solutions in MSC transplantation.
Collapse
Affiliation(s)
- Tsvetelina Velikova
- Department of Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Tereza Dekova
- Department of Genetics, Faculty of Biology, Sofia University St. Kliment Ohridski, Sofia 1164, Bulgaria
| | | |
Collapse
|
19
|
Voga M. Modulation of Canine Adipose-Derived Mesenchymal Stem/Medicinal Signalling Cells with Ascorbic Acid: Effect on Proliferation and Chondrogenic Differentiation on Standard Plastic and Silk Fibroin Surfaces. Bioengineering (Basel) 2024; 11:513. [PMID: 38790380 PMCID: PMC11118827 DOI: 10.3390/bioengineering11050513] [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: 04/29/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Ascorbic acid (AA) plays a crucial role in both the proliferation and chondrogenic differentiation potential of mesenchymal stem/medicinal signalling cells (MSCs); these are both key aspects of their general therapeutic use and their increasing use in veterinary medicine. Current immunomodulatory therapies require efficient expansion of MSCs in the laboratory, while emerging tissue regeneration strategies, such as cartilage or bone repair, aim to use differentiated MSCs and modulate the expression of chondrogenic and hypertrophic markers. Our aim was to investigate whether the addition of AA to the growth medium enhances the proliferation of canine adipose-derived MSCs (cAMSCs) grown on standard plastic surfaces and whether it affects chondrogenic differentiation potential on silk fibroin (SF) films. We assessed cell viability with trypan blue and proliferation potential by calculating population doubling. Chondrogenic induction on SF films was assessed by Alcian blue staining and gene expression analysis of chondrogenic and hypertrophic genes. The results showed that growth medium with AA significantly enhanced the proliferation of cAMSCs without affecting cell viability and modulated the expression of chondrogenic and hypertrophic genes of cAMSCs grown on SF films. Our results suggest that AA may be used in growth medium for expansion of cAMSCs and, at the same time, provide the basis for future studies to investigate the role of AA and SF in chondrogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Metka Voga
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia
| |
Collapse
|
20
|
Ramírez DG, Inostroza C, Rouabhia M, Rodriguez CA, Gómez LA, Losada M, Muñoz AL. Osteogenic potential of apical papilla stem cells mediated by platelet-rich fibrin and low-level laser. Odontology 2024; 112:399-407. [PMID: 37874511 PMCID: PMC10925562 DOI: 10.1007/s10266-023-00851-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/31/2023] [Indexed: 10/25/2023]
Abstract
To evaluate the osteogenic potential of platelet-rich fibrin (PRF) and low-level laser therapy (LLLT) on human stem cells from the apical papilla (SCAP) we isolated, characterized, and then cultured in an osteogenic medium cells with PRF and/or LLLT (660 nm, 6 J/m2-irradiation). Osteogenic differentiation was assessed by bone nodule formation and expression of bone morphogenetic proteins (BMP-2 and BMP-4), whereas the molecular mechanisms were achieved by qRT-PCR and RNA-seq analysis. Statistical analysis was performed by ANOVA and Tukey's post hoc tests (p < 0.05* and p < 0.01**). Although PRF and LLLT increased bone nodule formation after 7 days and peaked at 21 days, the combination of PRF + LLLT led to the uppermost nodule formation. This was supported by increased levels of BMP-2 and -4 osteogenic proteins (p < 0.005). Furthermore, the PRF + LLLT relative expression of specific genes involved in osteogenesis, such as osteocalcin, was 2.4- (p = 0.03) and 28.3- (p = 0.001) fold higher compared to the PRF and LLLT groups, and osteopontin was 22.9- and 1.23-fold higher, respectively (p < 0.05), after 7 days of interaction. The transcriptomic profile revealed that the combination of PRF + LLLT induces MSX1, TGFB1, and SMAD1 expression, after 21 days of osteogenic differentiation conditions exposition. More studies are required to understand the complete cellular and molecular mechanisms of PRF plus LLLT on stem cells. Overall, we demonstrated for the first time that the combination of PRF and LLLT would be an excellent therapeutic tool that can be employed for dental, oral, and craniofacial repair and other tissue engineering applications.
Collapse
Affiliation(s)
- David Gutiérrez Ramírez
- Buccal Innovation Research Group, Faculty of Dentistry, Universidad Antonio Nariño, Popayán, Colombia
| | | | | | - Camilo Alfonso Rodriguez
- Faculty of Dentistry. Research Group of Oral Health, Universidad Antonio Nariño, Bogotá, Colombia
| | - Lina Andrea Gómez
- School of Medicine, Biomedical Research Center (CIBUS), Universidad de La Sabana, Chía, Colombia
| | - Mónica Losada
- Cellular and Functional Biology and Biomolecule Engineering Research Group, Faculty of Science, Universidad Antonio Nariño, Bogotá, Colombia
| | - Ana Luisa Muñoz
- Cellular and Functional Biology and Biomolecule Engineering Research Group, Faculty of Science, Universidad Antonio Nariño, Bogotá, Colombia.
- Fundación Banco Nacional de Sangre Hemolife, Calle 23 No. 116-31, Bodega 26. Parque Industrial Puerto Central, Bogotá, Colombia.
| |
Collapse
|
21
|
Nguyen TT, Kil YS, Sung JH, Youn YS, Jeong JH, Lee JH, Jiang HL, Yook S, Nam JW, Jeong JH. Fabrication of stem cell heterospheroids with sustained-release chitosan and poly(lactic-co-glycolic acid) microspheres to guide cell fate toward chondrogenic differentiation. Int J Biol Macromol 2024; 263:130356. [PMID: 38395283 DOI: 10.1016/j.ijbiomac.2024.130356] [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: 10/30/2023] [Revised: 02/19/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024]
Abstract
Mesenchymal stem cell (MSC)-based therapies show great potential in treating various diseases. However, control of the fate of injected cells needs to be improved. In this work, we developed an efficient methodology for modulating chondrogenic differentiation of MSCs. We fabricated heterospheroids with two sustained-release depots, a quaternized chitosan microsphere (QCS-MP) and a poly (lactic-co-glycolic acid) microsphere (PLGA-MP). The results show that heterospheroids composed of 1 × 104 to 5 × 104 MSCs formed rapidly during incubation in methylcellulose medium and maintained high cell viability in long-term culture. The MPs were uniformly distributed in the heterospheroids, as shown by confocal laser scanning microscopy. Incorporation of transforming growth factor beta 3 into QCS-MPs and of dexamethasone into PLGA-MPs significantly promoted the expression of chondrogenic genes and high accumulation of glycosaminoglycan in heterospheroids. Changes in crucial metabolites in the dual drug depot-engineered heterospheroids were also evaluated using 1H NMR-based metabolomics analysis to verify their successful chondrogenic differentiation. Our heterospheroid fabrication platform could be used in tissue engineering to study the effects of various therapeutic agents on stem cell fate.
Collapse
Affiliation(s)
- Tiep Tien Nguyen
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi 16419, Republic of Korea; College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea; Epibiotech Co. Ltd., Incheon 21983, Republic of Korea
| | - Yun-Seo Kil
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea; College of Pharmacy, Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam 50834, Republic of Korea
| | - Jong-Hyuk Sung
- Epibiotech Co. Ltd., Incheon 21983, Republic of Korea; College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Republic of Korea
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi 16419, Republic of Korea
| | - Ji Hoon Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi 16419, Republic of Korea
| | - Jung Heon Lee
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, Gyeonggi 16419, Republic of Korea
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi 16419, Republic of Korea; Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Joo-Won Nam
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi 16419, Republic of Korea.
| |
Collapse
|
22
|
Makhija E, Zheng Y, Wang J, Leong HR, Othman RB, Ng EX, Lee EH, Kellogg LT, Lee YH, Yu H, Poon Z, Van Vliet KJ. Topological defects in self-assembled patterns of mesenchymal stromal cells in vitro are predictive attributes of condensation and chondrogenesis. PLoS One 2024; 19:e0297769. [PMID: 38547243 PMCID: PMC10977694 DOI: 10.1371/journal.pone.0297769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/11/2024] [Indexed: 04/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising therapeutic agents for cartilage regeneration, including the potential of cells to promote chondrogenesis in vivo. However, process development and regulatory approval of MSCs as cell therapy products benefit from facile in vitro approaches that can predict potency for a given production run. Current standard in vitro approaches include a 21 day 3D differentiation assay followed by quantification of cartilage matrix proteins. We propose a novel biophysical marker that is cell population-based and can be measured from in vitro monolayer culture of MSCs. We hypothesized that the self-assembly pattern that emerges from collective-cell behavior would predict chondrogenesis motivated by our observation that certain features in this pattern, namely, topological defects, corresponded to mesenchymal condensations. Indeed, we observed a strong predictive correlation between the degree-of-order of the pattern at day 9 of the monolayer culture and chondrogenic potential later estimated from in vitro 3D chondrogenic differentiation at day 21. These findings provide the rationale and the proof-of-concept for using self-assembly patterns to monitor chondrogenic commitment of cell populations. Such correlations across multiple MSC donors and production batches suggest that self-assembly patterns can be used as a candidate biophysical attribute to predict quality and efficacy for MSCs employed therapeutically for cartilage regeneration.
Collapse
Affiliation(s)
- Ekta Makhija
- Critical Analytics for Manufacturing Personalized-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
| | - Yang Zheng
- Critical Analytics for Manufacturing Personalized-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
- NUS Tissue Engineering Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Orthopaedic Surgery, National University of Singapore, Singapore, Singapore
| | - Jiahao Wang
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Han Ren Leong
- Critical Analytics for Manufacturing Personalized-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
- Engineering Science Programme, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Rashidah Binte Othman
- Critical Analytics for Manufacturing Personalized-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
| | - Ee Xien Ng
- Critical Analytics for Manufacturing Personalized-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
| | - Eng Hin Lee
- Critical Analytics for Manufacturing Personalized-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
- NUS Tissue Engineering Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Orthopaedic Surgery, National University of Singapore, Singapore, Singapore
| | - Lisa Tucker Kellogg
- Critical Analytics for Manufacturing Personalized-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
- Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Yie Hou Lee
- Critical Analytics for Manufacturing Personalized-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
- Obstetrics and Gynaecology Academic Clinical Programme, SingHealth Duke-NUS, Singapore, Singapore
- SingHealth Duke-NUS Cell Therapy Centre, Singapore, Singapore
| | - Hanry Yu
- Critical Analytics for Manufacturing Personalized-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Physiology, National University of Singapore, Singapore, Singapore
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research, Singapore, Singapore
| | - Zhiyong Poon
- Critical Analytics for Manufacturing Personalized-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
- SingHealth Duke-NUS Cell Therapy Centre, Singapore, Singapore
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Krystyn J. Van Vliet
- Critical Analytics for Manufacturing Personalized-medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore, Singapore
- Department of Materials Science and Engineering, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| |
Collapse
|
23
|
Loukelis K, Koutsomarkos N, Mikos AG, Chatzinikolaidou M. Advances in 3D bioprinting for regenerative medicine applications. Regen Biomater 2024; 11:rbae033. [PMID: 38845855 PMCID: PMC11153344 DOI: 10.1093/rb/rbae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 06/09/2024] Open
Abstract
Biofabrication techniques allow for the construction of biocompatible and biofunctional structures composed from biomaterials, cells and biomolecules. Bioprinting is an emerging 3D printing method which utilizes biomaterial-based mixtures with cells and other biological constituents into printable suspensions known as bioinks. Coupled with automated design protocols and based on different modes for droplet deposition, 3D bioprinters are able to fabricate hydrogel-based objects with specific architecture and geometrical properties, providing the necessary environment that promotes cell growth and directs cell differentiation towards application-related lineages. For the preparation of such bioinks, various water-soluble biomaterials have been employed, including natural and synthetic biopolymers, and inorganic materials. Bioprinted constructs are considered to be one of the most promising avenues in regenerative medicine due to their native organ biomimicry. For a successful application, the bioprinted constructs should meet particular criteria such as optimal biological response, mechanical properties similar to the target tissue, high levels of reproducibility and printing fidelity, but also increased upscaling capability. In this review, we highlight the most recent advances in bioprinting, focusing on the regeneration of various tissues including bone, cartilage, cardiovascular, neural, skin and other organs such as liver, kidney, pancreas and lungs. We discuss the rapidly developing co-culture bioprinting systems used to resemble the complexity of tissues and organs and the crosstalk between various cell populations towards regeneration. Moreover, we report on the basic physical principles governing 3D bioprinting, and the ideal bioink properties based on the biomaterials' regenerative potential. We examine and critically discuss the present status of 3D bioprinting regarding its applicability and current limitations that need to be overcome to establish it at the forefront of artificial organ production and transplantation.
Collapse
Affiliation(s)
- Konstantinos Loukelis
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Nikos Koutsomarkos
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, Heraklion 70013, Greece
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion 70013, Greece
| |
Collapse
|
24
|
Knill C, Henderson EJ, Johnson C, Wah VY, Cheng K, Forster AJ, Itasaki N. Defects of the spliceosomal gene SNRPB affect osteo- and chondro-differentiation. FEBS J 2024; 291:272-291. [PMID: 37584444 DOI: 10.1111/febs.16934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/25/2023] [Accepted: 08/14/2023] [Indexed: 08/17/2023]
Abstract
Although gene splicing occurs throughout the body, the phenotype of spliceosomal defects is largely limited to specific tissues. Cerebro-costo-mandibular syndrome (CCMS) is one such spliceosomal disease, which presents as congenital skeletal dysmorphism and is caused by mutations of SNRPB gene encoding Small Nuclear Ribonucleoprotein Polypeptides B/B' (SmB/B'). This study employed in vitro cell cultures to monitor osteo- and chondro-differentiation and examined the role of SmB/B' in the differentiation process. We found that low levels of SmB/B' by knockdown or mutations of SNRPB led to suppressed osteodifferentiation in Saos-2 osteoprogenitor-like cells, which was accompanied by affected splicing of Dlx5. On the other hand, low SmB/B' led to promoted chondrogenesis in HEPM mesenchymal stem cells. Consistent with other reports, osteogenesis was promoted by the Wnt/β-catenin pathway activator and suppressed by Wnt and BMP blockers, whereas chondrogenesis was promoted by Wnt inhibitors. Suppressed osteogenic markers by SNRPB knockdown were partly rescued by Wnt/β-catenin pathway activation. Reporter analysis revealed that suppression of SNRPB results in attenuated Wnt pathway and/or enhanced BMP pathway activities. SNRPB knockdown altered splicing of TCF7L2 which impacts Wnt/β-catenin pathway activities. This work helps unravel the mechanism underlying CCMS whereby reduced expression of spliceosomal proteins causes skeletal phenotypes.
Collapse
Affiliation(s)
- Chris Knill
- Faculty of Life Sciences, University of Bristol, UK
| | | | - Craig Johnson
- Faculty of Health Sciences, University of Bristol, UK
| | - Vun Yee Wah
- Faculty of Life Sciences, University of Bristol, UK
| | - Kevin Cheng
- Faculty of Life Sciences, University of Bristol, UK
| | | | - Nobue Itasaki
- Faculty of Health Sciences, University of Bristol, UK
| |
Collapse
|
25
|
Vermeulen S, Knoops K, Duimel H, Parvizifard M, van Beurden D, López-Iglesias C, Giselbrecht S, Truckenmüller R, Habibović P, Tahmasebi Birgani Z. An in vitro model system based on calcium- and phosphate ion-induced hMSC spheroid mineralization. Mater Today Bio 2023; 23:100844. [PMID: 38033367 PMCID: PMC10682137 DOI: 10.1016/j.mtbio.2023.100844] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
A challenge in regenerative medicine is creating the three-dimensional organic and inorganic in vitro microenvironment of bone, which would allow the study of musculoskeletal disorders and the generation of building blocks for bone regeneration. This study presents a microwell-based platform for creating spheroids of human mesenchymal stromal cells, which are then mineralized using ionic calcium and phosphate supplementation. The resulting mineralized spheroids promote an osteogenic gene expression profile through the influence of the spheroids' biophysical environment and inorganic signaling and require less calcium or phosphate to achieve mineralization compared to a monolayer culture. We found that mineralized spheroids represent an in vitro model for studying small molecule perturbations and extracellular mediated calcification. Furthermore, we demonstrate that understanding pathway signaling elicited by the spheroid environment allows mimicking these pathways in traditional monolayer culture, enabling similar rapid mineralization events. In sum, this study demonstrates the rapid generation and employment of a mineralized cell model system for regenerative medicine applications.
Collapse
Affiliation(s)
- Steven Vermeulen
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Kèvin Knoops
- Microscopy CORE Lab, M4I Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Hans Duimel
- Microscopy CORE Lab, M4I Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Maryam Parvizifard
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Denis van Beurden
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Carmen López-Iglesias
- Microscopy CORE Lab, M4I Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Zeinab Tahmasebi Birgani
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
26
|
Kim M, Jang HJ, Baek SY, Choi KJ, Han DH, Sung JS. Regulation of base excision repair during adipogenesis and osteogenesis of bone marrow-derived mesenchymal stem cells. Sci Rep 2023; 13:16384. [PMID: 37773206 PMCID: PMC10542337 DOI: 10.1038/s41598-023-43737-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 09/27/2023] [Indexed: 10/01/2023] Open
Abstract
Bone marrow-derived human mesenchymal stem cells (hMSCs) can differentiate into various lineages, such as chondrocytes, adipocytes, osteoblasts, and neuronal lineages. It has been shown that the high-efficiency DNA-repair capacity of hMSCs is decreased during their differentiation. However, the underlying its mechanism during adipogenesis and osteogenesis is unknown. Herein, we investigated how alkyl-damage repair is modulated during adipogenic and osteogenic differentiation, especially focusing on the base excision repair (BER) pathway. Response to an alkylation agent was assessed via quantification of the double-strand break (DSB) foci and activities of BER-related enzymes during differentiation in hMSCs. Adipocytes showed high resistance against methyl methanesulfonate (MMS)-induced alkyl damage, whereas osteoblasts were more sensitive than hMSCs. During the differentiation, activities, and protein levels of uracil-DNA glycosylase were found to be regulated. In addition, ligation-related proteins, such as X-ray repair cross-complementing protein 1 (XRCC1) and DNA polymerase β, were upregulated in adipocytes, whereas their levels and recruitment declined during osteogenesis. These modulations of BER enzyme activity during differentiation influenced DNA repair efficiency and the accumulation of DSBs as repair intermediates in the nucleus. Taken together, we suggest that BER enzymatic activity is regulated in adipogenic and osteogenic differentiation and these alterations in the BER pathway led to different responses to alkyl damage from those in hMSCs.
Collapse
Affiliation(s)
- Min Kim
- Department of Life Science, Dongguk University-Seoul, Biomedi Campus, 32 Dongguk-ro, Ilsandong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Hyun-Jin Jang
- Department of Life Science, Dongguk University-Seoul, Biomedi Campus, 32 Dongguk-ro, Ilsandong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Song-Yi Baek
- Department of Life Science, Dongguk University-Seoul, Biomedi Campus, 32 Dongguk-ro, Ilsandong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Kyung-Jin Choi
- Department of Life Science, Dongguk University-Seoul, Biomedi Campus, 32 Dongguk-ro, Ilsandong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Dong-Hee Han
- Department of Life Science, Dongguk University-Seoul, Biomedi Campus, 32 Dongguk-ro, Ilsandong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Jung-Suk Sung
- Department of Life Science, Dongguk University-Seoul, Biomedi Campus, 32 Dongguk-ro, Ilsandong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
27
|
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
|
28
|
Decarli MC, Seijas‐Gamardo A, Morgan FLC, Wieringa P, Baker MB, Silva JVL, Moraes ÂM, Moroni L, Mota C. Bioprinting of Stem Cell Spheroids Followed by Post-Printing Chondrogenic Differentiation for Cartilage Tissue Engineering. Adv Healthc Mater 2023; 12:e2203021. [PMID: 37057819 PMCID: PMC11468754 DOI: 10.1002/adhm.202203021] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/13/2023] [Indexed: 04/15/2023]
Abstract
Cartilage tissue presents low self-repair capability and lesions often undergo irreversible progression. Structures obtained by tissue engineering, such as those based in extrusion bioprinting of constructs loaded with stem cell spheroids may offer valuable alternatives for research and therapeutic purposes. Human mesenchymal stromal cell (hMSC) spheroids can be chondrogenically differentiated faster and more efficiently than single cells. This approach allows obtaining larger tissues in a rapid, controlled and reproducible way. However, it is challenging to control tissue architecture, construct stability, and cell viability during maturation. Herein, this work reports a reproducible bioprinting process followed by a successful post-bioprinting chondrogenic differentiation procedure using large quantities of hMSC spheroids encapsulated in a xanthan gum-alginate hydrogel. Multi-layered constructs are bioprinted, ionically crosslinked, and post chondrogenically differentiated for 28 days. The expression of glycosaminoglycan, collagen II and IV are observed. After 56 days in culture, the bioprinted constructs are still stable and show satisfactory cell metabolic activity with profuse extracellular matrix production. These results show a promising procedure to obtain 3D models for cartilage research and ultimately, an in vitro proof-of-concept of their potential use as stable chondral tissue implants.
Collapse
Affiliation(s)
- Monize Caiado Decarli
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
- Department of Engineering of Biomaterials and of BioprocessesSchool of Chemical EngineeringUniversity of Campinas ‐ UNICAMPAv. Albert Einstein, 500, Cidade Universitária “Zeferino Vaz”CampinasSP13083‐852Brazil
| | - Adrián Seijas‐Gamardo
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| | - Francis L. C. Morgan
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| | - Paul Wieringa
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| | - Matthew B. Baker
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| | - Jorge Vicente L. Silva
- Three‐Dimensional Technologies Research GroupCTI Renato ArcherRodovia Dom Pedro I SP‐65, Km 143,6 ‐ AmaraisCampinasSP13069‐901Brazil
| | - Ângela Maria Moraes
- Department of Engineering of Biomaterials and of BioprocessesSchool of Chemical EngineeringUniversity of Campinas ‐ UNICAMPAv. Albert Einstein, 500, Cidade Universitária “Zeferino Vaz”CampinasSP13083‐852Brazil
| | - Lorenzo Moroni
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| | - Carlos Mota
- MERLN Institute for Technology‐Inspired Regenerative MedicineDepartment of Complex Tissue RegenerationMaastricht UniversityUniversiteitssingel, 40MaastrichtLimburg6229 ERthe Netherlands
| |
Collapse
|
29
|
Peng Y, Jiang H, Zuo HD. Factors affecting osteogenesis and chondrogenic differentiation of mesenchymal stem cells in osteoarthritis. World J Stem Cells 2023; 15:548-560. [PMID: 37424946 PMCID: PMC10324504 DOI: 10.4252/wjsc.v15.i6.548] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 06/26/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that often involves progressive cartilage degeneration and bone destruction of subchondral bone. At present, clinical treatment is mainly for pain relief, and there are no effective methods to delay the progression of the disease. When this disease progresses to the advanced stage, the only treatment option for most patients is total knee replacement surgery, which causes patients great pain and anxiety. As a type of stem cell, mesenchymal stem cells (MSCs) have multidirectional differentiation potential. The osteogenic differentiation and chondrogenic differentiation of MSCs can play vital roles in the treatment of OA, as they can relieve pain in patients and improve joint function. The differentiation direction of MSCs is accurately controlled by a variety of signaling pathways, so there are many factors that can affect the differentiation direction of MSCs by acting on these signaling pathways. When MSCs are applied to OA treatment, the microenvironment of the joints, injected drugs, scaffold materials, source of MSCs and other factors exert specific impacts on the differentiation direction of MSCs. This review aims to summarize the mechanisms by which these factors influence MSC differentiation to produce better curative effects when MSCs are applied clinically in the future.
Collapse
Affiliation(s)
- Yi Peng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Hai Jiang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Hou-Dong Zuo
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
- Department of Radiology, Chengdu Xinhua Hospital, Chengdu 610067, Sichuan Province, China
| |
Collapse
|
30
|
Uzieliene I, Bironaite D, Miksiunas R, Bagdonas E, Vaiciuleviciute R, Mobasheri A, Bernotiene E. The Effect of CaV1.2 Inhibitor Nifedipine on Chondrogenic Differentiation of Human Bone Marrow or Menstrual Blood-Derived Mesenchymal Stem Cells and Chondrocytes. Int J Mol Sci 2023; 24:ijms24076730. [PMID: 37047701 PMCID: PMC10095444 DOI: 10.3390/ijms24076730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/27/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
Cartilage is an avascular tissue and sensitive to mechanical trauma and/or age-related degenerative processes leading to the development of osteoarthritis (OA). Therefore, it is important to investigate the mesenchymal cell-based chondrogenic regenerating mechanisms and possible their regulation. The aim of this study was to investigate the role of intracellular calcium (iCa2+) and its regulation through voltage-operated calcium channels (VOCC) on chondrogenic differentiation of mesenchymal stem/stromal cells derived from human bone marrow (BMMSCs) and menstrual blood (MenSCs) in comparison to OA chondrocytes. The level of iCa2+ was highest in chondrocytes, whereas iCa2+ store capacity was biggest in MenSCs and they proliferated better as compared to other cells. The level of CaV1.2 channels was also highest in OA chondrocytes than in other cells. CaV1.2 antagonist nifedipine slightly suppressed iCa2+, Cav1.2 and the proliferation of all cells and affected iCa2+ stores, particularly in BMMSCs. The expression of the CaV1.2 gene during 21 days of chondrogenic differentiation was highest in MenSCs, showing the weakest chondrogenic differentiation, which was stimulated by the nifedipine. The best chondrogenic differentiation potential showed BMMSCs (SOX9 and COL2A1 expression); however, purposeful iCa2+ and VOCC regulation by blockers can stimulate a chondrogenic response at least in MenSCs.
Collapse
Affiliation(s)
- Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| | - Rokas Miksiunas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| | - Edvardas Bagdonas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000 Liège, Belgium
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
| |
Collapse
|
31
|
Kilian D, Poddar A, Desrochers V, Heinemann C, Halfter N, Liu S, Rother S, Gelinsky M, Hintze V, Lode A. Cellular adhesion and chondrogenic differentiation inside an alginate-based bioink in response to tailorable artificial matrices and tannic acid treatment. BIOMATERIALS ADVANCES 2023; 147:213319. [PMID: 36758282 DOI: 10.1016/j.bioadv.2023.213319] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/30/2022] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
Many established bioinks fulfill important requirements regarding fabrication standards and cytocompatibility. Current research focuses on development of functionalized bioinks with an improved support of tissue-specific cell differentiation. Many approaches primarily depend on decellularized extracellular matrices or blood components. In this study, we investigated the combination of a highly viscous alginate-methylcellulose (algMC) bioink with collagen-based artificial extracellular matrix (aECM) as a finely controllable and tailorable system composed of collagen type I (col) with and without chondroitin sulfate (CS) or sulfated hyaluronan (sHA). As an additional stabilizer, the polyphenol tannic acid (TA) was integrated into the inks. The assessment of rheological properties and printability as well as hydrogel microstructure revealed no adverse effect of the integrated components on the inks. Viability, adhesion, and proliferation of bioprinted immortalized human mesenchymal stem cells (hTERT-MSC) was improved indicating enhanced interaction with the designed microenvironment. Furthermore, chondrogenic matrix production (collagen type II and sulfated glycosaminoglycans) by primary human chondrocytes (hChon) was enhanced by aECM. Supplementing the inks with TA was required for these positive effects but caused cytotoxicity as soon as TA concentrations exceeded a certain amount. Thus, combining tailorable aECM with algMC and balanced TA addition proved to be a promising approach for promoting adhesion of immortalized stem cells and differentiation of chondrocytes in bioprinted scaffolds.
Collapse
Affiliation(s)
- David Kilian
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Aayush Poddar
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Vanessa Desrochers
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Christiane Heinemann
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany
| | - Norbert Halfter
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany
| | - Suihong Liu
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany; Rapid Manufacturing Engineering Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Sandra Rother
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany; Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Saar, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Strasse 27, 01069 Dresden, Germany
| | - Anja Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany.
| |
Collapse
|
32
|
Musa M, Zeppieri M, Enaholo ES, Salati C, Parodi PC. Adipose Stem Cells in Modern-Day Ophthalmology. Clin Pract 2023; 13:230-245. [PMID: 36826163 PMCID: PMC9955457 DOI: 10.3390/clinpract13010021] [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: 12/28/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Stem cells (SCs) have evolved as an interesting and viable factor in ophthalmologic patient care in the past decades. SCs have been classified as either embryonic, mesenchymal, tissue-specific, or induced pluripotent cells. Multiple novel management techniques and clinical trials have been established to date. While available publications are predominantly animal-model-based, significant material is derived from human studies and case-selected scenarios. This possibility of explanting cells from viable tissue to regenerate/repair damaged tissue points to an exciting future of therapeutic options in all fields of medicine, and ophthalmology is surely not left out. Adipose tissue obtained from lipo-aspirates has been shown to produce mesenchymal SCs that are potentially useful in different body parts, including the oculo-visual system. An overview of the anatomy, physiology, and extraction process for adipose-tissue-derived stem cells (ADSC) is important for better understanding the potential therapeutic benefits. This review examines published data on ADSCs in immune-modulatory, therapeutic, and regenerative treatments. We also look at the future of ADSC applications for ophthalmic patient care. The adverse effects of this relatively novel therapy are also discussed.
Collapse
Affiliation(s)
- Mutali Musa
- Department of Optometry, University of Benin, Benin City 300238, Nigeria
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
- Correspondence:
| | | | - Carlo Salati
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| | - Pier Camillo Parodi
- Department of Plastic Surgery, University Hospital of Udine, 33100 Udine, Italy
| |
Collapse
|
33
|
Textor M, Hoburg A, Lehnigk R, Perka C, Duda GN, Reinke S, Blankenstein A, Hochmann S, Stockinger A, Resch H, Wolf M, Strunk D, Geissler S. Chondrocyte Isolation from Loose Bodies-An Option for Reducing Donor Site Morbidity for Autologous Chondrocyte Implantation. Int J Mol Sci 2023; 24:ijms24021484. [PMID: 36675010 PMCID: PMC9867247 DOI: 10.3390/ijms24021484] [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: 11/30/2022] [Revised: 12/21/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023] Open
Abstract
Loose bodies (LBs) from patients with osteochondritis dissecans (OCD) are usually removed and discarded during surgical treatment of the defect. In this study, we address the question of whether these LBs contain sufficient viable and functional chondrocytes that could serve as a source for autologous chondrocyte implantation (ACI) and how the required prolonged in vitro expansion affects their phenotype. Chondrocytes were isolated from LBs of 18 patients and compared with control chondrocyte from non-weight-bearing joint regions (n = 7) and bone marrow mesenchymal stromal cells (BMSCs, n = 6) obtained during primary arthroplasty. No significant differences in the initial cell yield per isolation and the expression of the chondrocyte progenitor cell markers CD44 + /CD146+ were found between chondrocyte populations from LBs (LB-CH) and control patients (Ctrl-CH). During long-term expansion, LB-CH exhibited comparable viability and proliferation rates to control cells and no ultimate cell cycle arrest was observed within 12 passages respectively 15.3 ± 1.1 mean cumulative populations doublings (CPD). The chondrogenic differentiation potential was comparable between LB-CH and Ctrl-CH, but both groups showed a significantly higher ability to form a hyaline cartilage matrix in vitro than BMSC. Our data suggest that LBs are a promising cell source for obtaining qualitatively and quantitatively suitable chondrocytes for therapeutic applications, thereby circumventing donor site morbidity as a consequence of the biopsies required for the current ACI procedure.
Collapse
Affiliation(s)
- Martin Textor
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Arnd Hoburg
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Centrum für Muskuloskelettale Chirugie (CBMSC), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Med Center 360 Degree Berlin, Kieler Straße 1, 12163 Berlin, Germany
| | - Rex Lehnigk
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Centrum für Muskuloskelettale Chirugie (CBMSC), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Carsten Perka
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Centrum für Muskuloskelettale Chirugie (CBMSC), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Georg N. Duda
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Centrum für Muskuloskelettale Chirugie (CBMSC), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02138, USA
| | - Simon Reinke
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Antje Blankenstein
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sarah Hochmann
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | | | - Herbert Resch
- Department of Traumatology, Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Martin Wolf
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Dirk Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Sven Geissler
- Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Center for Advanced Therapies (BECAT), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany
- Correspondence:
| |
Collapse
|
34
|
Abstract
Cartilage resides under a low oxygen tension within articulating joints. The oxygen tension within cartilage of the knee joint has been measured to be between 2% and 5% oxygen. Although the literature has historically termed this level of oxygen as hypoxia, particularly when doing experiments in vitro in this range, this is actually the physiological oxygen tension experienced in vivo and is more accurately termed physioxia. In general, culture of chondrogenic cells under physioxia has demonstrated a donor-dependent beneficial effect on chondrogenesis, with an upregulation in cartilage genes (SOX9, COL2A1, ACAN) and matrix deposition (sulfated glycosaminoglycans (sGAGs), collagen II). Physioxia also reduces the expression of hypertrophic markers (COL10A1, MMP13). This chapter will outline the methods for the expansion and differentiation of chondrogenic cells under physioxia using oxygen-controlled incubators and glove box environments, with the typical assays used for qualitative and quantitative assessment of chondrogenesis.
Collapse
Affiliation(s)
- Girish Pattappa
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center of Regensburg, Regensburg, Germany
| | - Brandon D Markway
- Department of Orthopaedics & Rehabilitation, Oregon Health & Science University, Portland, OR, USA
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Medical Center of Regensburg, Regensburg, Germany
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Wurzburg, Wurzburg, Germany
| | - Brian Johnstone
- Department of Orthopaedics & Rehabilitation, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
35
|
de Souza TV, Pastena Giorno L, Malmonge SM, Santos AR. Bioprinting: From Technique to Application in Tissue Engineering and Regenerative Medicine. Curr Mol Med 2023; 23:934-951. [PMID: 36017861 DOI: 10.2174/1566524023666220822152448] [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: 04/11/2022] [Revised: 05/22/2022] [Accepted: 05/30/2022] [Indexed: 11/22/2022]
Abstract
Among the different approaches present in regenerative medicine and tissue engineering, the one that has attracted the most interest in recent years is the possibility of printing functional biological tissues. Bioprinting is a technique that has been applied to create cellularized three-dimensional structures that mimic biological tissues and thus allow their replacement. Hydrogels are interesting materials for this type of technique. Hydrogels based on natural polymers are known due to their biocompatible properties, in addition to being attractive biomaterials for cell encapsulation. They provide a threedimensional aqueous environment with biologically relevant chemical and physical signals, mimicking the natural environment of the extracellular matrix (ECM). Bioinks are ink formulations that allow the printing of living cells. The controlled deposition of biomaterials by bioinks needs to maintain cell viability and offer specific biochemical and physical stimuli capable of guiding cell migration, proliferation, and differentiation. In this work, we analyze the theoretical and practical issues of bioprinting, citing currently used methods, their advantages, and limitations. We present some important molecules that have been used to compose bioinks, as well as the cellular responses that have been observed in different tissues. Finally, we indicate future perspectives of the method.
Collapse
Affiliation(s)
- Thaís Vieira de Souza
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Luciana Pastena Giorno
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Sonia Maria Malmonge
- Centro de Engenharia, Modelagem e Ciências Sociais Aplicadas, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Arnaldo R Santos
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| |
Collapse
|
36
|
Leung AOW, Yiu TC, Liu L, Tam HY, Gu S, Tu J, Pei D, Cheung HH. Targeting G-quadruplex for rescuing impaired chondrogenesis in WRN-deficient stem cells. Cell Biosci 2022; 12:212. [PMID: 36587229 PMCID: PMC9805690 DOI: 10.1186/s13578-022-00939-8] [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: 08/13/2022] [Accepted: 12/08/2022] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Pathogenic mutations in WRN are a cause of premature aging disease Werner syndrome (WS). Besides accelerated aging phenotypes and cancer predisposition, patients with WS also display underdevelopment in the skeletal system, characterized by short stature, light body weight and unusually thin extremities. The reasons for these developmental defects are not completely understood and the underlying molecular mechanism remains to be elucidated. RESULTS In this study, WRN was found to modulate transcription of short stature homeobox gene SHOX. Loss of WRN resulted in insufficient expression of SHOX, the gene dose of which is critical for driving chondrocyte differentiation. WRN could bind the G-quadruplex (G4) structures in the SHOX promoter and stimulate transcription. Aberrant formation of G4 structures in WRN-deficient cells impeded normal transcription of SHOX, thus resulting in impaired chondrogenesis. Chondrogenesis could be rescued by overexpression of WRN helicase or SHOX, suggesting that SHOX is a downstream target of WRN. Gene editing of the G4 structures in the SHOX promoter could increase SHOX expression, therefore rescuing the impaired chondrogenesis in WRN-deficient cells. CONCLUSIONS Our data suggest that dysgenesis of the developing bone in WS might be caused by SHOX insufficiency. Aberrant formation of G4 structures in SHOX promoter suppresses SHOX expression and impairs chondrogenesis. Targeted mutagenesis in the G4 structures enhances SHOX expression and thus providing an opportunity to rescue the chondrogenic defect.
Collapse
Affiliation(s)
- Adrian On-Wah Leung
- grid.10784.3a0000 0004 1937 0482Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China ,grid.9227.e0000000119573309Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Tsz-Ching Yiu
- grid.10784.3a0000 0004 1937 0482Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lingxiao Liu
- grid.10784.3a0000 0004 1937 0482Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China ,grid.9227.e0000000119573309Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Hei-Yin Tam
- grid.10784.3a0000 0004 1937 0482Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shen Gu
- grid.10784.3a0000 0004 1937 0482Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jiajie Tu
- grid.186775.a0000 0000 9490 772XInstitute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Duanqing Pei
- grid.9227.e0000000119573309Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong SAR, China ,grid.494629.40000 0004 8008 9315Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, 310024 Hangzhou, China
| | - Hoi-Hung Cheung
- grid.10784.3a0000 0004 1937 0482Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
37
|
Mesenchymal Stromal Cells Laden in Hydrogels for Osteoarthritis Cartilage Regeneration: A Systematic Review from In Vitro Studies to Clinical Applications. Cells 2022; 11:cells11243969. [PMID: 36552733 PMCID: PMC9777087 DOI: 10.3390/cells11243969] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
This systematic review is focused on the main characteristics of the hydrogels used for embedding the mesenchymal stromal cells (MSCs) in in vitro/ex vivo studies, in vivo OA models and clinical trials for favoring cartilage regeneration in osteoarthritis (OA). PubMED and Embase databases were used to select the papers that were submitted to a public reference manager Rayyan Systematic Review Screening Software. A total of 42 studies were considered eligible: 25 articles concerned in vitro studies, 2 in vitro and ex vivo ones, 5 in vitro and in vivo ones, 8 in vivo ones and 2 clinical trials. Some in vitro studies evidenced a rheological characterization of the hydrogels and description of the crosslinking methods. Only 37.5% of the studies considered at the same time chondrogenic, fibrotic and hypertrophic markers. Ex vivo studies focused on hydrogel adhesion properties and the modification of MSC-laden hydrogels subjected to compression tests. In vivo studies evidenced the effect of cell-laden hydrogels in OA animal models or defined the chondrogenic potentiality of the cells in subcutaneous implantation models. Clinical studies confirmed the positive impact of these treatments on patients with OA. To speed the translation to the clinical use of cell-laden hydrogels, further studies on hydrogel characteristics, injection modalities, chemo-attractant properties and adhesion strength are needed.
Collapse
|
38
|
Ranmuthu CKI, Ranmuthu CDS, Wijewardena CK, Seah MKT, Khan WS. Evaluating the Effect of Hypoxia on Human Adult Mesenchymal Stromal Cell Chondrogenesis In Vitro : A Systematic Review. Int J Mol Sci 2022; 23:ijms232315210. [PMID: 36499531 PMCID: PMC9741425 DOI: 10.3390/ijms232315210] [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: 09/26/2022] [Revised: 11/18/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Human adult mesenchymal stromal cells (MSCs) from a variety of sources may be used to repair defects in articular cartilage by inducing them into chondrogenic differentiation. The conditions in which optimal chondrogenic differentiation takes place are an area of interest in the field of tissue engineering. Chondrocytes exist in vivo in a normally hypoxic environment and thus it has been suggested that exposing MSCs to hypoxia may also contribute to a beneficial effect on their differentiation. There are two main stages in which MSCs can be exposed to hypoxia, the expansion phase when cells are cultured, and the differentiation phase when cells are induced with a chondrogenic medium. This systematic review sought to explore the effect of hypoxia at these two stages on human adult MSC chondrogenesis in vitro. A literature search was performed on PubMed, EMBASE, Medline via Ovid, and Cochrane, and 24 studies were ultimately included. The majority of these studies showed that hypoxia during the expansion phase or the differentiation phase enhances at least some markers of chondrogenic differentiation in adult MSCs. These results were not always demonstrated at the protein level and there were also conflicting reports. Studies evaluating continuous exposure to hypoxia during the expansion and differentiation phases also had mixed results. These inconsistent results can be explained by the heterogeneity of studies, including factors such as different sources of MSCs used, donor variability, level of hypoxia used in each study, time exposed to hypoxia, and differences in culture methodology.
Collapse
|
39
|
Zhu S, Donovan EL, Makosa D, Mehta-D'souza P, Jopkiewicz A, Batushansky A, Cortassa D, Simmons AD, Lopes EBP, Kinter M, Griffin TM. Sirt3 Promotes Chondrogenesis, Chondrocyte Mitochondrial Respiration and the Development of High-Fat Diet-Induced Osteoarthritis in Mice. J Bone Miner Res 2022; 37:2531-2547. [PMID: 36214465 PMCID: PMC10091721 DOI: 10.1002/jbmr.4721] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 09/09/2022] [Accepted: 10/06/2022] [Indexed: 12/31/2022]
Abstract
Understanding how obesity-induced metabolic stress contributes to synovial joint tissue damage is difficult because of the complex role of metabolism in joint development, maintenance, and repair. Chondrocyte mitochondrial dysfunction is implicated in osteoarthritis (OA) pathology, which motivated us to study the mitochondrial deacetylase enzyme sirtuin 3 (Sirt3). We hypothesized that combining high-fat-diet (HFD)-induced obesity and cartilage Sirt3 loss at a young age would impair chondrocyte mitochondrial function, leading to cellular stress and accelerated OA. Instead, we unexpectedly found that depleting cartilage Sirt3 at 5 weeks of age using Sirt3-flox and Acan-CreERT2 mice protected against the development of cartilage degeneration and synovial hyperplasia following 20 weeks of HFD. This protection was associated with increased cartilage glycolysis proteins and reduced mitochondrial fatty acid metabolism proteins. Seahorse-based assays supported a mitochondrial-to-glycolytic shift in chondrocyte metabolism with Sirt3 deletion. Additional studies with primary murine juvenile chondrocytes under hypoxic and inflammatory conditions showed an increased expression of hypoxia-inducible factor (HIF-1) target genes with Sirt3 deletion. However, Sirt3 deletion impaired chondrogenesis using a murine bone marrow stem/stromal cell pellet model, suggesting a context-dependent role of Sirt3 in cartilage homeostasis. Overall, our data indicate that Sirt3 coordinates HFD-induced changes in mature chondrocyte metabolism that promote OA. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Shouan Zhu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Elise L Donovan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Dawid Makosa
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Padmaja Mehta-D'souza
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Anita Jopkiewicz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Dominic Cortassa
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Aaron D Simmons
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Timothy M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.,Department of Biochemistry and Molecular Biology, Department of Physiology, Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, USA.,Biomedical Laboratory Research and Development, Veterans Affairs Medical Center, Oklahoma City, USA
| |
Collapse
|
40
|
Nadine S, Fernandes IJ, Correia CR, Mano JF. Close-to-native bone repair via tissue-engineered endochondral ossification approaches. iScience 2022; 25:105370. [PMID: 36339269 PMCID: PMC9626746 DOI: 10.1016/j.isci.2022.105370] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In order to solve the clinical challenges related to bone grafting, several tissue engineering (TE) strategies have been proposed to repair critical-sized defects. Generally, the classical TE approaches are designed to promote bone repair via intramembranous ossification. Although promising, strategies that direct the osteogenic differentiation of mesenchymal stem/stromal cells are usually characterized by a lack of functional vascular supply, often resulting in necrotic cores. A less explored alternative is engineering bone constructs through a cartilage-mediated approach, resembling the embryological process of endochondral ossification. The remodeling of an intermediary hypertrophic cartilaginous template triggers vascular invasion and bone tissue deposition. Thus, employing this knowledge can be a promising direction for the next generation of bone TE constructs. This review highlights the most recent biomimetic strategies for applying endochondral ossification in bone TE while discussing the plethora of cell types, culture conditions, and biomaterials essential to promote a successful bone regeneration process.
Collapse
|
41
|
Heyman E, Meeremans M, Devriendt B, Olenic M, Chiers K, De Schauwer C. Validation of a color deconvolution method to quantify MSC tri-lineage differentiation across species. Front Vet Sci 2022; 9:987045. [PMID: 36311666 PMCID: PMC9608146 DOI: 10.3389/fvets.2022.987045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/20/2022] [Indexed: 11/04/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a promising candidate for both human and veterinary regenerative medicine applications because of their abundance and ability to differentiate into several lineages. Mesenchymal stem cells are however a heterogeneous cell population and as such, it is imperative that they are unequivocally characterized to acquire reproducible results in clinical trials. Although the tri-lineage differentiation potential of MSCs is reported in most veterinary studies, a qualitative evaluation of representative histological images does not always unambiguously confirm tri-lineage differentiation. Moreover, potential differences in differentiation capacity are not identified. Therefore, quantification of tri-lineage differentiation would greatly enhance proper characterization of MSCs. In this study, a method to quantify the tri-lineage differentiation potential of MSCs is described using digital image analysis, based on the color deconvolution plug-in (ImageJ). Mesenchymal stem cells from three species, i.e., bovine, equine, and porcine, were differentiated toward adipocytes, chondrocytes, and osteocytes. Subsequently, differentiated MSCs were stained with Oil Red O, Alcian Blue, and Alizarin Red S, respectively. Next, a differentiation ratio (DR) was obtained by dividing the area % of the differentiation signal by the area % of the nuclear signal. Although MSCs isolated from all donors in all species were capable of tri-lineage differentiation, differences were demonstrated between donors using this quantitative DR. Our straightforward, simple but robust method represents an elegant approach to determine the degree of MSC tri-lineage differentiation across species. As such, differences in differentiation potential within the heterogeneous MSC population and between different MSC sources can easily be identified, which will support further optimization of regenerative therapies.
Collapse
Affiliation(s)
- Emma Heyman
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium,*Correspondence: Emma Heyman
| | - Marguerite Meeremans
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Maria Olenic
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium,Tissue Engineering Lab, Muscles and Movement Group, Faculty of Medicine, Catholic University of Leuven, Kortrijk, Belgium
| | - Koen Chiers
- Laboratory of Veterinary Pathology, Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Catharina De Schauwer
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
42
|
Decellularised Cartilage ECM Culture Coatings Drive Rapid and Robust Chondrogenic Differentiation of Human Periosteal Cells. Bioengineering (Basel) 2022; 9:bioengineering9050203. [PMID: 35621481 PMCID: PMC9137502 DOI: 10.3390/bioengineering9050203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
The control of cell behaviour in an effort to create highly homogeneous cultures is becoming an area of intense research, both to elucidate fundamental biology and for regenerative applications. The extracellular matrix (ECM) controls many cellular processes in vivo, and as such is a rich source of cues that may be translated in vitro. Herein, we describe the creation of cell culture coatings from porcine decellularised hyaline cartilage through enzymatic digestion. Surprisingly, heat-mediated sterilisation created a coating with the capacity to rapidly and robustly induce chondrogenic differentiation of human periosteal cells. This differentiation was validated through the alteration of cell phenotype from a fibroblastic to a cuboidal/cobblestone chondrocyte-like appearance. Moreover, chondrogenic gene expression further supported this observation, where cells cultured on heat sterilised ECM-coated plastic displayed higher expression of COL2A1, ACAN and PRG4 (p < 0.05) compared to non-coated plastic cultures. Interestingly, COL2A1 and ACAN expression in this context were sensitive to initial cell density; however, SOX9 expression appeared to be mainly driven by the coating independent of seeding density. The creation of a highly chondrogenic coating may provide a cost-effective solution for the differentiation and/or expansion of human chondrocytes aimed towards cartilage repair strategies.
Collapse
|
43
|
Nothdurfter D, Ploner C, Coraça-Huber DC, Wilflingseder D, Müller T, Hermann M, Hagenbuchner J, Ausserlechner MJ. 3D bioprinted, vascularized neuroblastoma tumor environment in fluidic chip devices for precision medicine drug testing. Biofabrication 2022; 14. [PMID: 35333193 DOI: 10.1088/1758-5090/ac5fb7] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 03/22/2022] [Indexed: 11/12/2022]
Abstract
Neuroblastoma is an extracranial solid tumor which develops in early childhood and still has a poor prognosis. One strategy to increase cure rates is the identification of patient-specific drug responses in tissue models that mimic the interaction between patient cancer cells and tumor environment. We therefore developed a perfused and micro-vascularized tumor-environment model that is directly bioprinted into custom-manufactured fluidic chips. A gelatin-methacrylate/fibrin-based matrix containing multiple cell types mimics the tumor-microenvironment that promotes spontaneous micro-vessel formation by embedded endothelial cells. We demonstrate that both, adipocyte- and iPSC-derived mesenchymal stem cells can guide this process. Bioprinted channels are coated with endothelial cells post printing to form a dense vessel - tissue barrier. The tissue model thereby mimics structure and function of human soft tissue with endothelial cell-coated larger vessels for perfusion and micro-vessel networks within the hydrogel-matrix. Patient-derived neuroblastoma spheroids are added to the matrix during the printing process and grown for more than two weeks. We demonstrate that micro-vessels are attracted by and grow into tumor spheroids and that neuroblastoma cells invade the tumor-environment as soon as the spheroids disrupt. In summary, we describe the first bioprinted, micro-vascularized neuroblastoma - tumor-environment model directly printed into fluidic chips and a novel medium-throughput biofabrication platform suitable for studying tumor angiogenesis and metastasis in precision medicine approaches in future.
Collapse
Affiliation(s)
- Daniel Nothdurfter
- Department of Pediatrics I and 3D Bioprinting Lab, Medical University Innsbruck, Austria
| | - Christian Ploner
- Department of Plastic and Reconstructive Surgery, Medical University Innsbruck, Austria
| | - Débora C Coraça-Huber
- Research Laboratory for Biofilms and Implant Associated Infections (BIOFILM LAB), Experimental Orthopedics, Department of Orthopedic Surgery, Medical University Innsbruck, Austria
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University Innsbruck, Austria
| | - Thomas Müller
- Department of Pediatrics I and 3D Bioprinting Lab, Medical University Innsbruck, Austria
| | - Martin Hermann
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Judith Hagenbuchner
- Department of Pediatrics II and 3D Bioprinting Lab, Medical University Innsbruck, Austria
| | | |
Collapse
|
44
|
Prajwal GS, Jeyaraman N, Kanth V K, Jeyaraman M, Muthu S, Rajendran SNS, Rajendran RL, Khanna M, Oh EJ, Choi KY, Chung HY, Ahn BC, Gangadaran P. Lineage Differentiation Potential of Different Sources of Mesenchymal Stem Cells for Osteoarthritis Knee. Pharmaceuticals (Basel) 2022; 15:386. [PMID: 35455383 PMCID: PMC9028477 DOI: 10.3390/ph15040386] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 02/05/2023] Open
Abstract
Tissue engineering and regenerative medicine (TERM) have paved a way for treating musculoskeletal diseases in a minimally invasive manner. The regenerative medicine cocktail involves the usage of mesenchymal stem/stromal cells (MSCs), either uncultured or culture-expanded cells along with growth factors, cytokines, exosomes, and secretomes to provide a better regenerative milieu in degenerative diseases. The successful regeneration of cartilage depends on the selection of the appropriate source of MSCs, the quality, quantity, and frequency of MSCs to be injected, and the selection of the patient at an appropriate stage of the disease. However, confirmation on the most favorable source of MSCs remains uncertain to clinicians. The lack of knowledge in the current cellular treatment is uncertain in terms of how beneficial MSCs are in the long-term or short-term (resolution of pain) and improved quality of life. Whether MSCs treatments have any superiority, exists due to sources of MSCs utilized in their potential to objectively regenerate the cartilage at the target area. Many questions on source and condition remain unanswered. Hence, in this review, we discuss the lineage differentiation potentials of various sources of MSCs used in the management of knee osteoarthritis and emphasize the role of tissue engineering in cartilage regeneration.
Collapse
Affiliation(s)
- Gollahalli Shivashankar Prajwal
- Research Fellow, Fellowship in Orthopaedic Rheumatology (FEIORA), Dr. Ram Manohar Lohiya National Law University, Lucknow 226010, Uttar Pradesh, India; (G.S.P.); (N.J.)
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 110048, Uttar Pradesh, India; (S.M.); (M.K.)
- Department of Orthopaedics, Mallika Spine Centre, Guntur 522001, Andhra Pradesh, India
| | - Naveen Jeyaraman
- Research Fellow, Fellowship in Orthopaedic Rheumatology (FEIORA), Dr. Ram Manohar Lohiya National Law University, Lucknow 226010, Uttar Pradesh, India; (G.S.P.); (N.J.)
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 110048, Uttar Pradesh, India; (S.M.); (M.K.)
- Department of Orthopaedics, Atlas Hospitals, Tiruchirappalli 620002, Tamil Nadu, India
| | - Krishna Kanth V
- Department of Orthopaedics, Government Medical College, Mahabubabad 506104, Telangana, India;
| | - Madhan Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 110048, Uttar Pradesh, India; (S.M.); (M.K.)
- Department of Orthopaedics, Faculty of Medicine—Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, Tamil Nadu, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201306, Uttar Pradesh, India
- Orthopaedic Research Group, Coimbatore 641001, Tamil Nadu, India
| | - Sathish Muthu
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 110048, Uttar Pradesh, India; (S.M.); (M.K.)
- Department of Orthopaedics, Government Medical College, Mahabubabad 506104, Telangana, India;
- Department of Orthopaedics, Faculty of Medicine—Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, Tamil Nadu, India
- Orthopaedic Research Group, Coimbatore 641001, Tamil Nadu, India
| | - Sree Naga Sowndary Rajendran
- Department of Medicine, Sri Venkateshwaraa Medical College Hospital and Research Centre, Puducherry 605102, Puducherry, India;
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
| | - Manish Khanna
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 110048, Uttar Pradesh, India; (S.M.); (M.K.)
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624001, Tamil Nadu, India
- Department of Orthopaedics, Prasad Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| | - Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea; (E.J.O.); (K.Y.C.); (H.Y.C.)
| | - Kang Young Choi
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea; (E.J.O.); (K.Y.C.); (H.Y.C.)
| | - Ho Yun Chung
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea; (E.J.O.); (K.Y.C.); (H.Y.C.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
45
|
Ricci A, Cataldi A, Zara S, Gallorini M. Graphene-Oxide-Enriched Biomaterials: A Focus on Osteo and Chondroinductive Properties and Immunomodulation. MATERIALS (BASEL, SWITZERLAND) 2022; 15:2229. [PMID: 35329679 PMCID: PMC8955105 DOI: 10.3390/ma15062229] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/28/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022]
Abstract
Due to its exceptional physical properties, such as high electronic conductivity, good thermal stability, excellent mechanical strength, and chemical versatility, graphene has sparked a lot of interest in the scientific community for various applications. It has therefore been employed as an antibacterial agent, in photothermal therapy (PTT) and biosensors, in gene delivery systems, and in tissue engineering for regenerative purposes. Since it was first discovered in 1947, different graphene derivatives have been synthetized from pristine graphene. The most adaptable derivate is graphene oxide (GO). Owing to different functional groups, the amphiphilic structure of GO can interact with cells and exogenous or endogenous growth/differentiation factors, allowing cell adhesion, growth, and differentiation. When GO is used as a coating for scaffolds and nanomaterials, it has been found to enhance bone, chondrogenic, cardiac, neuronal, and skin regeneration. This review focuses on the applications of graphene-based materials, in particular GO, as a coating for scaffolds in bone and chondrogenic tissue engineering and summarizes the most recent findings. Moreover, novel developments on the immunomodulatory properties of GO are reported.
Collapse
Affiliation(s)
| | | | | | - Marialucia Gallorini
- Department of Pharmacy, “G. d'Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (A.R.); (A.C.); (S.Z.)
| |
Collapse
|
46
|
Ravanbakhsh H, Luo Z, Zhang X, Maharjan S, Mirkarimi HS, Tang G, Chávez-Madero C, Mongeau L, Zhang YS. Freeform Cell-Laden Cryobioprinting for Shelf-Ready Tissue Fabrication and Storage. MATTER 2022; 5:573-593. [PMID: 35695821 PMCID: PMC9173715 DOI: 10.1016/j.matt.2021.11.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
One significant drawback of existing bioprinted tissues is their lack of shelf-availability caused by complications in both fabrication and storage. Here, we report a cryobioprinting strategy for simultaneously fabricating and storing cell-laden volumetric tissue constructs through seamlessly combining extrusion bioprinting and cryopreservation. The cryobioprinting performance was investigated by designing, fabricating, and storing cell-laden constructs made of our optimized cryoprotective gelatin-based bioinks using a freezing plate with precisely controllable temperature. The in situ freezing process further promoted the printability of cell-laden hydrogel bioinks to achieve freeform structures otherwise inconvenient with direct extrusion bioprinting. The effects of bioink composition on printability and cell viability were evaluated. The functionality of the method was finally investigated using cell differentiation and chick ex ovo assays. The results confirmed the feasibility and efficacy of cryobioprinting as a single-step method for concurrent tissue biofabrication and storage.
Collapse
Affiliation(s)
- Hossein Ravanbakhsh
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, McGill University, Montreal, QC, H3A0C3, Canada
| | - Zeyu Luo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Orthopedics, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, 610041, P.R. China
| | - Xiang Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, P. R. China
- National Center for International Joint Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Sushila Maharjan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Hengameh S. Mirkarimi
- Department of Mechanical Engineering, École de technologie supérieure, Montreal, QC, H3C1K3, Canada
| | - Guosheng Tang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Carolina Chávez-Madero
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Departamento de Ingeniería Mecatrónica y Electrónica, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey 64849, NL, México
| | - Luc Mongeau
- Department of Mechanical Engineering, McGill University, Montreal, QC, H3A0C3, Canada
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
47
|
Harryvan TJ, de Lange S, Hawinkels LJ, Verdegaal EM. The ABCs of Antigen Presentation by Stromal Non-Professional Antigen-Presenting Cells. Int J Mol Sci 2021; 23:ijms23010137. [PMID: 35008560 PMCID: PMC8745042 DOI: 10.3390/ijms23010137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 12/29/2022] Open
Abstract
Professional antigen-presenting cells (APCs), such as dendritic cells and macrophages, are known for their ability to present exogenous antigens to T cells. However, many other cell types, including endothelial cells, fibroblasts, and lymph node stromal cells, are also capable of presenting exogenous antigens to either CD8+ or CD4+ T cells via cross-presentation or major histocompatibility complex (MHC) class II-mediated presentation, respectively. Antigen presentation by these stromal nonprofessional APCs differentially affect T cell function, depending on the type of cells that present the antigen, as well as the local (inflammatory) micro-environment. It has been recently appreciated that nonprofessional APCs can, as such, orchestrate immunity against pathogens, tumor survival, or rejection, and aid in the progression of various auto-immune pathologies. Therefore, the interest for these nonprofessional APCs is growing as they might be an important target for enhancing various immunotherapies. In this review, the different nonprofessional APCs are discussed, as well as their functional consequences on the T cell response, with a focus on immuno-oncology.
Collapse
Affiliation(s)
- Tom J. Harryvan
- Department of Gastroenterology & Hepatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
- Correspondence: (T.J.H.); (L.J.A.C.H.); (E.M.E.V.); Tel.: +0031-715266736 (L.J.A.C.H.)
| | - Sabine de Lange
- Department of Gastroenterology & Hepatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Lukas J.A.C. Hawinkels
- Department of Gastroenterology & Hepatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
- Correspondence: (T.J.H.); (L.J.A.C.H.); (E.M.E.V.); Tel.: +0031-715266736 (L.J.A.C.H.)
| | - Els M.E. Verdegaal
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Correspondence: (T.J.H.); (L.J.A.C.H.); (E.M.E.V.); Tel.: +0031-715266736 (L.J.A.C.H.)
| |
Collapse
|
48
|
James JL, Umapathy A, Srinivasan S, Barker CN, Brooks A, Hearn J, Chhana A, Williams E, Sheppard H, McGlashan SR. The Chondrogenic Potential of First-Trimester and Term Placental Mesenchymal Stem/Stromal Cells. Cartilage 2021; 13:544S-558S. [PMID: 34521248 PMCID: PMC8804733 DOI: 10.1177/19476035211044822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES Mesenchymal stem/stromal cells (MSCs) are a well-established cell source for cartilage engineering, but challenges remain as differentiation often results in chondrocyte hypertrophy. Chondrogenic potential also varies with MSC source and donor age. We assessed the chondrogenic potential of first-trimester and term placental MSCs and compared their response to commonly used bone marrow MSCs (BM-MSCs). DESIGN MSCs were isolated from first-trimester and term placentae. BM-MSCs were commercially obtained. Chondrogenesis was induced by micromass culture in commercial chondrogenic media for 7, 14, or 21 days. Pellets were assessed for glycosaminoglycan (GAG) content, and types I, II, and X collagen. Gene expression was profiled using Qiagen RT2 human MSC arrays. RESULTS At day 0, first-trimester and term MSCs expression levels of many chondrogenic genes to BM-MSC after 21 days of culture. Only first trimester MSCs showed significant changes in chondrogenic gene expression during induction compared to day 0 undifferentiated MSCs (greater BMP4, KAT2B, and reduced GDF6 expression). Additionally, first-trimester MSCs showed significantly greater expression of ABCB1 (at days 14 and 21) and BMP4 (at days 7, 14, 21) compared with term MSCs. Both first-trimester and term pellets showed increased GAG content over time and term MSCs had significantly GAG greater compared with BM-MSCs at days 7 and 14. Type II collagen was present in all pellets but unlike BM-MSCs, type I collagen was not observed in first-trimester or term MSC pellets. CONCLUSIONS These data highlight differences in BM-MSC and placental MSC chondrogenesis and demonstrate that placental MSCs may be an alternative cell source.
Collapse
Affiliation(s)
- Joanna L. James
- Obstetrics and Gynaecology, The
University of Auckland, Auckland, New Zealand
| | - Anandita Umapathy
- Obstetrics and Gynaecology, The
University of Auckland, Auckland, New Zealand
| | - Sonia Srinivasan
- Obstetrics and Gynaecology, The
University of Auckland, Auckland, New Zealand
| | - Claire N. Barker
- Anatomy and Medical Imaging, The
University of Auckland, Auckland, New Zealand
| | - Anna Brooks
- School of Biological Sciences, The
University of Auckland, Auckland, New Zealand
| | - James Hearn
- Molecular Medicine and Pathology, The
University of Auckland, Auckland, New Zealand
| | - Ashika Chhana
- Anatomy and Medical Imaging, The
University of Auckland, Auckland, New Zealand
| | - Eloise Williams
- Obstetrics and Gynaecology, The
University of Auckland, Auckland, New Zealand
| | - Hilary Sheppard
- School of Biological Sciences, The
University of Auckland, Auckland, New Zealand
| | - Sue R. McGlashan
- Anatomy and Medical Imaging, The
University of Auckland, Auckland, New Zealand,Sue R. McGlashan, Department of Anatomy and
Medical Imaging, School of Medical Sciences, The University of Auckland, Faculty
of Medical and Health Sciences, 85 Park Road, Grafton, Auckland 1142, New
Zealand.
| |
Collapse
|
49
|
Autologous Bone Marrow-Derived Chondrocytes for Patients with Knee Osteoarthritis: A Randomized Controlled Trial. Adv Orthop 2021; 2021:2146722. [PMID: 34845428 PMCID: PMC8627347 DOI: 10.1155/2021/2146722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/23/2021] [Accepted: 11/01/2021] [Indexed: 11/18/2022] Open
Abstract
Results There were a total of 60 patients who were followed up. Three patients in Group II were removed from the analysis as they underwent total knee arthroplasty (TKA). A notably significant improvement was noticed in the ABMDC group on all scores of VAS and MKSSSF with P < 0.0001. The control group continued to be dissatisfied with the treatment they were taking. Conclusions This study reveals that a single injection of 5 million of ABMDC was efficient in reducing the symptoms, improving the functional score and betterment of QOL.
Collapse
|
50
|
Pant T, Juric M, Bosnjak ZJ, Dhanasekaran A. Recent Insight on the Non-coding RNAs in Mesenchymal Stem Cell-Derived Exosomes: Regulatory and Therapeutic Role in Regenerative Medicine and Tissue Engineering. Front Cardiovasc Med 2021; 8:737512. [PMID: 34660740 PMCID: PMC8517144 DOI: 10.3389/fcvm.2021.737512] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022] Open
Abstract
Advances in the field of regenerative medicine and tissue engineering over the past few decades have paved the path for cell-free therapy. Numerous stem cell types, including mesenchymal stem cells (MSCs), have been reported to impart therapeutic effects via paracrine secretion of exosomes. The underlying factors and the associated mechanisms contributing to these MSC-derived exosomes' protective effects are, however, poorly understood, limiting their application in the clinic. The exosomes exhibit a diversified repertoire of functional non-coding RNAs (ncRNAs) and have the potential to transfer these biologically active transcripts to the recipient cells, where they are found to modulate a diverse array of functions. Altered expression of the ncRNAs in the exosomes has been linked with the regenerative potential and development of various diseases, including cardiac, neurological, skeletal, and cancer. Also, modulating the expression of ncRNAs in these exosomes has been found to improve their therapeutic impact. Moreover, many of these ncRNAs are expressed explicitly in the MSC-derived exosomes, making them ideal candidates for regenerative medicine, including tissue engineering research. In this review, we detail the recent advances in regenerative medicine and summarize the evidence supporting the altered expression of the ncRNA repertoire specific to MSCs under different degenerative diseases. We also discuss the therapeutic role of these ncRNA for the prevention of these various degenerative diseases and their future in translational medicine.
Collapse
Affiliation(s)
- Tarun Pant
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Matea Juric
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Zeljko J Bosnjak
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | | |
Collapse
|