1
|
Jeyaraman N, Jeyaraman M, Muthu S, Balaji S, Ramasubramanian S, Patro BP. Chondrogenic Potential of Umbilical Cord-Derived Mesenchymal Stromal Cells: Insights and Innovations. Indian J Orthop 2024; 58:1349-1361. [PMID: 39324097 PMCID: PMC11420429 DOI: 10.1007/s43465-024-01239-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024]
Abstract
Background The advent of tissue engineering and regenerative medicine has introduced innovative approaches to treating degenerative and traumatic injuries, particularly in cartilage, a tissue with limited self-repair capabilities. Among the various stem cell sources, umbilical cord-derived mesenchymal stromal cells (UC-MSCs) have garnered significant interest due to their non-invasive collection, minimal ethical concerns, and robust regenerative potential, particularly in cartilage regeneration. Methods A comprehensive literature review was conducted using multiple databases, including PubMed, Scopus, Web of Science, and Google Scholar. Search terms focused on "umbilical cordderived mesenchymal stromal cells," "chondrogenesis," "cartilage regeneration," and related topics. Studies published in the past two decades were included, with selection criteria emphasizing methodological rigor and relevance to UC-MSC chondrogenesis. The review synthesizes findings from various sources to provide a thorough analysis of the potential of UC-MSCs in cartilage tissue engineering. Results UC-MSCs exhibit significant chondrogenic potential, supported by their ability to differentiate into chondrocytes under specific conditions. Recent advancements include the development of biomaterial scaffolds and the application of genetic engineering techniques, such as CRISPR/Cas9, to enhance chondrogenic differentiation. Despite these advancements, challenges remain in standardizing cell isolation techniques, scaling up production for clinical use, and ensuring the long-term functionality of regenerated cartilage. Conclusion UC-MSCs offer a promising solution for cartilage regeneration in the field of regenerative medicine. Ongoing research is focused on overcoming current challenges through the use of advanced technologies, including bioreactors and gene editing. Collaborative efforts among researchers, clinicians, and bioengineers are essential to translating the potential of UC-MSCs into effective clinical therapies, which could significantly advance tissue regeneration and therapeutic innovation. Graphical Abstract
Collapse
Affiliation(s)
- Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Tamil Nadu, Chennai, 600077 India
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Tamil Nadu, Chennai, 600077 India
- VirginiaTech India, Dr MGR Educational and Research Institute, Tamil Nadu, Chennai, 600095 India
- Department of Orthopaedics, Orthopaedic Research Group, Tamil Nadu, Coimbatore, 641045 India
| | - Sathish Muthu
- Department of Orthopaedics, Orthopaedic Research Group, Tamil Nadu, Coimbatore, 641045 India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Tamil Nadu, Coimbatore, 641021 India
- Department of Orthopaedics, Government Karur Medical College, Tamil Nadu, Karur, 639004 India
| | - Sangeetha Balaji
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Tamil Nadu, Chennai, 600002 India
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Tamil Nadu, Chennai, 600002 India
| | - Bishnu Prasad Patro
- Department of Orthopaedics, All India Institute of Medical Sciences, Bhubaneswar, Odisha 751019 India
| |
Collapse
|
2
|
Velot É, Balmayor ER, Bertoni L, Chubinskaya S, Cicuttini F, de Girolamo L, Demoor M, Grigolo B, Jones E, Kon E, Lisignoli G, Murphy M, Noël D, Vinatier C, van Osch GJVM, Cucchiarini M. Women's contribution to stem cell research for osteoarthritis: an opinion paper. Front Cell Dev Biol 2023; 11:1209047. [PMID: 38174070 PMCID: PMC10762903 DOI: 10.3389/fcell.2023.1209047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/18/2023] [Indexed: 01/05/2024] Open
Affiliation(s)
- Émilie Velot
- Laboratory of Molecular Engineering and Articular Physiopathology (IMoPA), French National Centre for Scientific Research, University of Lorraine, Nancy, France
| | - Elizabeth R. Balmayor
- Experimental Orthopaedics and Trauma Surgery, Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen, Germany
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN, United States
| | - Lélia Bertoni
- CIRALE, USC 957, BPLC, École Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | | | - Flavia Cicuttini
- Musculoskeletal Unit, Monash University and Rheumatology, Alfred Hospital, Melbourne, VIC, Australia
| | - Laura de Girolamo
- IRCCS Ospedale Galeazzi - Sant'Ambrogio, Orthopaedic Biotechnology Laboratory, Milan, Italy
| | - Magali Demoor
- Normandie University, UNICAEN, BIOTARGEN, Caen, France
| | - Brunella Grigolo
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio RAMSES, Bologna, Italy
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom
| | - Elizaveta Kon
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department ofBiomedical Sciences, Humanitas University, Milan, Italy
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Bologna, Italy
| | - Mary Murphy
- Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - Danièle Noël
- IRMB, University of Montpellier, Inserm, CHU Montpellier, Montpellier, France
| | - Claire Vinatier
- Nantes Université, Oniris, INSERM, Regenerative Medicine and Skeleton, Nantes, France
| | - Gerjo J. V. M. van Osch
- Department of Orthopaedics and Sports Medicine and Department of Otorhinolaryngology, Department of Biomechanical Engineering, University Medical Center Rotterdam, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, Delft, Netherlands
| | - Magali Cucchiarini
- Center of Experimental Orthopedics, Saarland University and Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
3
|
Zhang Z, Zhao S, Sun Z, Zhai C, Xia J, Wen C, Zhang Y, Zhang Y. Enhancement of the therapeutic efficacy of mesenchymal stem cell-derived exosomes in osteoarthritis. Cell Mol Biol Lett 2023; 28:75. [PMID: 37770821 PMCID: PMC10540339 DOI: 10.1186/s11658-023-00485-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/29/2023] [Indexed: 09/30/2023] Open
Abstract
Osteoarthritis (OA), a common joint disorder with articular cartilage degradation as the main pathological change, is the major source of pain and disability worldwide. Despite current treatments, the overall treatment outcome is unsatisfactory. Thus, patients with severe OA often require joint replacement surgery. In recent years, mesenchymal stem cells (MSCs) have emerged as a promising therapeutic option for preclinical and clinical palliation of OA. MSC-derived exosomes (MSC-Exos) carrying bioactive molecules of the parental cells, including non-coding RNAs (ncRNAs) and proteins, have demonstrated a significant impact on the modulation of various physiological behaviors of cells in the joint cavity, making them promising candidates for cell-free therapy for OA. This review provides a comprehensive overview of the biosynthesis and composition of MSC-Exos and their mechanisms of action in OA. We also discussed the potential of MSC-Exos as a therapeutic tool for modulating intercellular communication in OA. Additionally, we explored bioengineering approaches to enhance MSC-Exos' therapeutic potential, which may help to overcome challenges and achieve clinically meaningful OA therapies.
Collapse
Affiliation(s)
- Zehao Zhang
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Sheng Zhao
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Zhaofeng Sun
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Chuanxing Zhai
- School of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, China
| | - Caining Wen
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
| | - Yuge Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
| | - Yuanmin Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
| |
Collapse
|
4
|
Yousefifard M, Sarveazad A, Janzadeh A, Behroozi Z, Nasirinezhad F. Pain Alleviating Effect of Adipose-Derived Stem Cells Transplantation on the Injured Spinal Cord: A Behavioral and Electrophysiological Evaluation. J Stem Cells Regen Med 2022; 18:53-63. [PMID: 36713791 PMCID: PMC9837693 DOI: 10.46582/jsrm.1802010] [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: 05/18/2022] [Accepted: 09/15/2022] [Indexed: 01/01/2023]
Abstract
Few studies are conducted on the efficacy of human adipose-derived stem cells (ADSCs) in spinal cord injury (SCI) management and electrophysiological changes in the spinal cord. Therefore, the present study aimed to determine the effect of ADSCs on neuropathic pain, motor function recovery, and electrophysiology assessment. For the purpose of this study, adult male Wistar rats (weight: 140-160 gr, n = 42) were randomly allocated into five groups namely intact animals, sham-operated, SCI non-treated animals, vehicle-treated (culture media), and ADSCs treated groups. One week after clips compression SCI induction, about 1×106 cells were transplanted into the spinal cord. As well, both neuropathic pain (allodynia and hyperalgesia) and motor function were measured weekly. Cavity size, ADSCs survival, and electrophysiology assessments were measured at the end of the eighth week. The transplantation of ADSCs resulted in a significant improvement in the locomotion of SCI animals (p<0.0001), mechanical allodynia (p<0.0001), cold allodynia (p<0.0001), mechanical hyperalgesia (p<0.0001), and thermal hyperalgesia (p<0.0001). The cavity size was significantly smaller among the ADSCs-treated animals (p <0.0001). The single-unit recording showed that the transplantation of ADSCs decreased wide dynamic range (WDR) in neurons and it evoked potential in response to receiving signals from Aβ (p<0.0001) and Aδ (p=0.003) C-fiber (p<0.0001) neurons. Post-discharge recorded from WDR neurons decreased after the transplantation of ADSCs (p<0.0001) and wind up in the ADSCs-treated group was lower than that of the SCI group (p=0.003). Our results showed that the transplantation of ADSCs could significantly alleviate neuropathic pain, enhance motor function recovery, and improve electrophysiology findings after SCI.
Collapse
Affiliation(s)
- Mahmoud Yousefifard
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Arash Sarveazad
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran,Nursing care Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Atousa Janzadeh
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Behroozi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Farinaz Nasirinezhad
- Cellular and molecular research center, Iran University of Medical Sciences, Tehran, Iran,Centre for Experimental and Comparative Study, Iran University of Medical Sciences, Tehran, Iran,Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran,Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran,Farinaz Nasirinezhad, Cellular, and molecular research center, Iran University of Medical Sciences, Tehran, Iran., Tel/Fax: +982188622709.
| |
Collapse
|
5
|
The Effect of Human Bone Marrow Mesenchymal Stem Cell-Derived Exosomes on Cartilage Repair in Rabbits. Stem Cells Int 2022; 2022:5760107. [PMID: 36117721 PMCID: PMC9477595 DOI: 10.1155/2022/5760107] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/22/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have shown chondroprotective effects in cartilage repair. However, side effects caused by MSC treatment limit their application in clinic. As a cell-free therapy, MSC-derived exosomes (EXOs) have attracted much more attention in recent years. In the present study, we prepared EXOs from human bone marrow mesenchymal stem cells (hBMSCs) and examined their therapeutic potentials in cartilage repair. Our results showed that the prepared extracellular vesicles exhibit classical features of EXOs, such as cup-like shape, around 100 nm diameter, positive protein markers (CD81, TSG101, and Flotillin 1), and ability of internalization. In primary chondrocytes, the treatment of hBMSC-EXOs markedly increases cell viability and proliferation in a dose-dependent manner. Moreover, wound healing assay showed that hBMSC-EXOs accelerate cell migration in primary chondrocytes. JC-1 staining revealed that the mitochondrial membrane potential was enhanced by hBMSC-EXOs, indicating cell apoptosis was decreased in the presence of hBMSC-EXOs. In rabbits with articular cartilage defects, local administration with hBMSC-EXOs facilitates cartilage regeneration as evidenced by gross view and hematoxylin-eosin (H&E) and Saf-O/Fast Green staining. In addition, the International Cartilage Repair Society (ICRS) score was increased by the application of hBMSC-EXOs. Overall, our data indicate that the treatment with hBMSC-EXOs is a suitable cell-free therapy for treating cartilage defects, and these benefits are likely due to improved cell proliferation and migration in chondrocytes.
Collapse
|
6
|
Chen YC, Liao HJ, Hsu YM, Shen YS, Chang CH. Delivery of Mesenchymal Stem Cell in Dialdehyde Methylcellulose-Succinyl-Chitosan Hydrogel Promotes Chondrogenesis in a Porcine Model. Polymers (Basel) 2022; 14:polym14071474. [PMID: 35406348 PMCID: PMC9002496 DOI: 10.3390/polym14071474] [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: 02/22/2022] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 02/04/2023] Open
Abstract
Due to the limitation in the current treatment modalities, such as secondary surgery in ACI and fibrocartilage formation in microfracture surgery, various scaffolds or hydrogels have been developed for cartilage regeneration. In the present study, we used sodium periodate to oxidize methylcellulose and formed dialdehyde methylcellulose (DAC) after dialysis and freeze-drying process, DAC was further mixed with succinyl-chitosan (SUC) to form an DAC-SUC in situ forming hydrogel. The hydrogel is a stiffness, elastic-like and porous hydrogel according to the observation of SEM and rheological analysis. DAC-SUC13 hydrogel possess well cell-compatibility as well as biodegradability. Most bone marrow mesenchymal stem cells (BM-pMSCs) were alive in the hydrogel and possess chondrogenesis potential. According to the results of animal study, we found DAC-SUC13 hydrogel can function as a stem cell carrier to promote glycosaminoglycans and type II collagen synthesis in the osteochondral defects of porcine knee. These findings suggested that DAC-SUC13 hydrogel combined with stem cell is a potential treatment for cartilage defects repair in the future.
Collapse
Affiliation(s)
- Yu-Chun Chen
- Department of Chemical Engineering, National United University, Miaoli 360302, Taiwan;
| | - Hsiu-Jung Liao
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City 220216, Taiwan; (H.-J.L.); (Y.-M.H.); (Y.-S.S.)
| | - Yuan-Ming Hsu
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City 220216, Taiwan; (H.-J.L.); (Y.-M.H.); (Y.-S.S.)
| | - Yi-Shan Shen
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City 220216, Taiwan; (H.-J.L.); (Y.-M.H.); (Y.-S.S.)
- Department of Biomedical Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Chih-Hung Chang
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City 220216, Taiwan; (H.-J.L.); (Y.-M.H.); (Y.-S.S.)
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan City 320315, Taiwan
- Correspondence:
| |
Collapse
|
7
|
Urlić I, Ivković A. Cell Sources for Cartilage Repair-Biological and Clinical Perspective. Cells 2021; 10:cells10092496. [PMID: 34572145 PMCID: PMC8468484 DOI: 10.3390/cells10092496] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 01/04/2023] Open
Abstract
Cell-based therapy represents a promising treatment strategy for cartilage defects. Alone or in combination with scaffolds/biological signals, these strategies open many new avenues for cartilage tissue engineering. However, the choice of the optimal cell source is not that straightforward. Currently, various types of differentiated cells (articular and nasal chondrocytes) and stem cells (mesenchymal stem cells, induced pluripotent stem cells) are being researched to objectively assess their merits and disadvantages with respect to the ability to repair damaged articular cartilage. In this paper, we focus on the different cell types used in cartilage treatment, first from a biological scientist’s perspective and then from a clinician’s standpoint. We compare and analyze the advantages and disadvantages of these cell types and offer a potential outlook for future research and clinical application.
Collapse
Affiliation(s)
- Inga Urlić
- Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Correspondence: (I.U.); (A.I.)
| | - Alan Ivković
- Department of Orthopaedic Surgery, University Hospital Sveti Duh, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Clinical Medicine, University of Applied Health Sciences, 10000 Zagreb, Croatia
- Correspondence: (I.U.); (A.I.)
| |
Collapse
|
8
|
Muthu S, Jeyaraman M, Jain R, Gulati A, Jeyaraman N, Prajwal GS, Mishra PC. Accentuating the sources of mesenchymal stem cells as cellular therapy for osteoarthritis knees-a panoramic review. Stem Cell Investig 2021; 8:13. [PMID: 34386542 PMCID: PMC8327191 DOI: 10.21037/sci-2020-055] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 04/25/2021] [Indexed: 02/05/2023]
Abstract
The large economic burden on the global health care systems is due to the increasing number of symptomatic osteoarthritis (OA) knee patients whereby accounting for greater morbidity and impaired functional quality of life. The recent developments and impulses in molecular and regenerative medicine have paved the way for inducing the biological active cells such as stem cells, bioactive materials, and growth factors towards the healing and tissue regenerative process. Mesenchymal stem cells (MSCs) act as a minimally invasive procedure that bridges the gap between pharmacological treatment and surgical treatment for OA. MSCs are the ideal cell-based therapy for treating disorders under a minimally invasive environment in conjunction with cartilage regeneration. Due to the worldwide recognized animal model for such cell-based therapies, global researchers have started using the various sources of MSCs towards cartilage regeneration. However, there is a lacuna in literature on the comparative efficacy and safety of various sources of MSCs in OA of the knee. Hence, the identification of a potential source for therapeutic use in this clinical scenario remains unclear. In this article, we compared the therapeutic effects of various sources of MSCs in terms of efficacy, safety, differentiation potential, durability, accessibility, allogenic preparation and culture expandability to decide the optimal source of MSCs for OA knee.
Collapse
Affiliation(s)
- Sathish Muthu
- Assistant Orthopaedic Surgeon, Government Hospital, Velayuthampalayam, Karur, Tamil Nadu, India
- International Association of Stemcell and Regenerative Medicine (IASRM), New Delhi, India
| | - Madhan Jeyaraman
- International Association of Stemcell and Regenerative Medicine (IASRM), New Delhi, India
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Rashmi Jain
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Arun Gulati
- Department of Orthopaedics, Kalpana Chawla Government Medical College & Hospital, Karnal, Haryana, India
| | - Naveen Jeyaraman
- International Association of Stemcell and Regenerative Medicine (IASRM), New Delhi, India
- Department of Orthopaedics, Kasturba Medical College, MAHE University, Manipal, Karnataka, India
| | | | - Prabhu Chandra Mishra
- International Association of Stemcell and Regenerative Medicine (IASRM), New Delhi, India
| |
Collapse
|
9
|
Um S, Ha J, Choi SJ, Oh W, Jin HJ. Prospects for the therapeutic development of umbilical cord blood-derived mesenchymal stem cells. World J Stem Cells 2020; 12:1511-1528. [PMID: 33505598 PMCID: PMC7789129 DOI: 10.4252/wjsc.v12.i12.1511] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/23/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Umbilical cord blood (UCB) is a primitive and abundant source of mesenchymal stem cells (MSCs). UCB-derived MSCs have a broad and efficient therapeutic capacity to treat various diseases and disorders. Despite the high latent self-renewal and differentiation capacity of these cells, the safety, efficacy, and yield of MSCs expanded for ex vivo clinical applications remains a concern. However, immunomodulatory effects have emerged in various disease models, exhibiting specific mechanisms of action, such as cell migration and homing, angiogenesis, anti-apoptosis, proliferation, anti-cancer, anti-fibrosis, anti-inflammation and tissue regeneration. Herein, we review the current literature pertaining to the UCB-derived MSC application as potential treatment strategies, and discuss the concerns regarding the safety and mass production issues in future applications.
Collapse
Affiliation(s)
- Soyoun Um
- Research Team for Immune Cell Therapy, Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Jueun Ha
- Research Team for Osteoarthritis, Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Hye Jin Jin
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| |
Collapse
|
10
|
Ma L, Zhang Y, Wang C. Coaction of TGF-β1 and CDMP1 in BMSCs-induced laryngeal cartilage repair in rabbits. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:130. [PMID: 33252704 DOI: 10.1007/s10856-020-06454-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 10/13/2020] [Accepted: 10/27/2020] [Indexed: 06/12/2023]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are well-known for tissue regeneration and bone repair. This study intended to evaluate the potential efficiency BMSCs in poly(lactide-co-glycolide) (PLGA) scaffolds for the treatment of laryngeal cartilage defects. BMSCs were isolated and identified, and added with 10 ng/mL transforming growth factor-beta1 (TGF-β1) or/and 300 ng/mL CDMP1 to coculture with PLGA scaffolds. The chondrogenic differentiation, migration, and apoptosis of BMSCs were detected under the action of TGF-β1 or/and CDMP1. After successful modeling of laryngeal cartilage defects, PLGA scaffolds were transplanted into the rabbits correspondingly. After 8 weeks, laryngeal cartilage defects were assessed. Levels of collagen II, aggrecan, Sox9, Smad2, Smad3, ERK, and JNK were detected. The TGF-β1 or/and CDMP1-induced BMSCs expressed collagen II, aggrecan, and Sox9, with enhanced cell migration and inhibited apoptosis. In addition, laryngeal cartilage defect in rabbits with TGF-β1 or/and CDMP1 was alleviated, and levels of specific cartilage matrix markers were decreased. The combined effects of TGF-β1 and CDMP1 were more significant. The TGF-β1/Smad and ERK/JNK pathways were activated after TGF-β1 or/and CDMP1 were added to BMSCs or rabbits. In summary, BMSCs and PLGA scaffolds repair laryngeal cartilage defects in rabbits by activating the TGF-β1/Smad and ERK/JNK pathways under the coaction of TGF-β1 and CDMP1.
Collapse
Affiliation(s)
- Linxiang Ma
- Department of Otolaryngology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, PR China
| | - Yonghong Zhang
- Department of Otolaryngology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, PR China
| | - Caihua Wang
- Department of Otolaryngology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, PR China.
| |
Collapse
|
11
|
Zucchelli E, Birchall M, Bulstrode NW, Ferretti P. Modeling Normal and Pathological Ear Cartilage in vitro Using Somatic Stem Cells in Three-Dimensional Culture. Front Cell Dev Biol 2020; 8:666. [PMID: 32850801 PMCID: PMC7402373 DOI: 10.3389/fcell.2020.00666] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/01/2020] [Indexed: 01/07/2023] Open
Abstract
Microtia (underdeveloped ear) is a rare congenital dysmorphology affecting the development of the outer ear. Although human microtic cartilage has not been fully characterized, chondrogenic cells derived from this tissue have been proposed as a suitable source for autologous auricular reconstruction. The aim of this study was to further characterize native microtic cartilage and investigate the properties of cartilage stem/progenitor cells (CSPCs) derived from it. Two-dimensional (2D) systems are most commonly used to assess the chondrogenic potential of somatic stem cells in vitro, but limit cell interactions and differentiation. Hence here we investigated the behavior of microtic CSPCs in three-dimensional spheroid cultures. Remarkable similarities between human microtic cartilages from five patients, as compared to normal cartilage, were observed notwithstanding possibly different etiologies of the disease. Native microtic cartilage displayed poorly defined perichondrium and hyper-cellularity, an immature phenotype that resembled that of the normal developing human auricular cartilage we studied in parallel. Crucially, our analysis of microtic ears revealed for the first time that, unlike normal cartilage, microtic cartilages are vascularized. Importantly, CSPCs isolated from human microtic and normal ear cartilages were found to recapitulate many characteristics of pathological and healthy tissues, respectively, when allowed to differentiate as spheroids, but not in monolayer cultures. Noteworthily, starting from initially homogeneous cell pellets, CSPC spheroids spontaneously underwent a maturation process in culture, and formed two regions (inner and outer region) separated by a boundary, with distinct cell types that differed in chondrogenic commitment as indicated by expression of chondrogenic markers. Compared to normal ear-derived spheroids, microtic spheroids were asymmetric, hyper-cellularized and the inner and outer regions did not develop properly. Hence, their organization resembled that of native microtic cartilage. Together, our results identify novel features of microtic ears and highlight the importance of 3D self-organizing in vitro systems for better understanding somatic stem cell behavior and disease modeling. Our observations of ear-derived chondrogenic stem cell behavior have implications for choice of cells for tissue engineered reconstructive purposes and for modeling the etiopathogenesis of microtia.
Collapse
Affiliation(s)
- Eleonora Zucchelli
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Martin Birchall
- UCL Ear Institute, University College London, London, United Kingdom
| | - Neil W. Bulstrode
- Department of Plastic Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Patrizia Ferretti
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
12
|
Neybecker P, Henrionnet C, Pape E, Grossin L, Mainard D, Galois L, Loeuille D, Gillet P, Pinzano A. Respective stemness and chondrogenic potential of mesenchymal stem cells isolated from human bone marrow, synovial membrane, and synovial fluid. Stem Cell Res Ther 2020; 11:316. [PMID: 32711576 PMCID: PMC7382063 DOI: 10.1186/s13287-020-01786-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/31/2020] [Accepted: 06/23/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND MSCs isolated from bone marrow (BM-MSCs) have well-established chondrogenic potential, but MSCs derived from the synovial membrane (SM-MSCs) and synovial fluid (SF-MSCs) are thought to possess superior chondrogenicity. This study aimed to compare the in vitro immunophenotype and trilineage and chondrogenic potential of BM-MSCs to SM-MSCs and SF-MSCs. METHODS MSCs were isolated from bone marrow (BM-MSCs), synovial membrane (SM-MSCs), and synovial fluid (SF-MSCs) extracted from the hips (BM) and knees (SM and SF) of advanced OA patients undergoing arthroplasty. Flow cytometric analysis was used at P2 to evaluate cell stemness. The trilinear differentiation test was performed at P2. At P3, MSC-seeded collagen sponges were cultured in chondrogenic medium for 28 days. Chondrogenic gene expression was quantified by qRT-PCR. Finally, the implants were stained to assess the deposition of proteoglycans and type II collagen. RESULTS Despite variability, the immunophenotyping of BM-MSCs, SM-MSCs, and SF-MSCs was quite similar. All cell types were positive for the expression of stem cell markers and negative for exclusion markers. Additionally, chondrogenic differentiation and hypertrophy were more pronounced in BM-MSCs (ACAN, SOX9, COL2B, and COL10A) than in SF-MSCs, with SM-MSCs having intermediate characteristics. Concerning matrix synthesis, the three cell types were equipotent in terms of GAG content, while BM-MSC ECM synthesis of type II collagen was superior. CONCLUSIONS Chondrogenic MSCs are easily collected from SM and SF in advanced human OA, but in vitro chondrogenesis that is superior to age-matched BM-MSCs should not be expected. However, due to intra-articular priming, SF-MSCs did not overexpress hypertrophic gene.
Collapse
Affiliation(s)
- Paul Neybecker
- UMR 7365 CNRS-Université de Lorraine, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, F54505, Vandœuvre-Lès-Nancy, France
| | - Christel Henrionnet
- UMR 7365 CNRS-Université de Lorraine, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, F54505, Vandœuvre-Lès-Nancy, France
| | - Elise Pape
- UMR 7365 CNRS-Université de Lorraine, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, F54505, Vandœuvre-Lès-Nancy, France.,Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511, Vandœuvre-lès-Nancy, France
| | - Laurent Grossin
- UMR 7365 CNRS-Université de Lorraine, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, F54505, Vandœuvre-Lès-Nancy, France
| | - Didier Mainard
- UMR 7365 CNRS-Université de Lorraine, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, F54505, Vandœuvre-Lès-Nancy, France.,Service de Chirurgie Orthopédique, Traumatologique et Arthroscopique, CHRU Nancy, 29 avenue du Maréchal de Lattre de Tassigny CO 60034, F54035, Nancy, France
| | - Laurent Galois
- UMR 7365 CNRS-Université de Lorraine, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, F54505, Vandœuvre-Lès-Nancy, France.,Service de Chirurgie Orthopédique, Traumatologique et Arthroscopique, CHRU Nancy, 29 avenue du Maréchal de Lattre de Tassigny CO 60034, F54035, Nancy, France
| | - Damien Loeuille
- UMR 7365 CNRS-Université de Lorraine, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, F54505, Vandœuvre-Lès-Nancy, France.,Service de Rhumatologie, CHRU de Nancy, Hôpitaux de Brabois, Bâtiment des Spécialités Médicales, 5 rue du Morvan, F54511, Vandœuvre-lès-Nancy, France
| | - Pierre Gillet
- UMR 7365 CNRS-Université de Lorraine, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, F54505, Vandœuvre-Lès-Nancy, France.,Laboratoire de Pharmacologie, Toxicologie et Pharmacovigilance, Bâtiment de Biologie Médicale et de Biopathologie, CHRU de Nancy-Brabois, 5 Rue du Morvan, F54511, Vandœuvre-lès-Nancy, France
| | - Astrid Pinzano
- UMR 7365 CNRS-Université de Lorraine, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, F54505, Vandœuvre-Lès-Nancy, France. .,Service de Rhumatologie, CHRU de Nancy, Hôpitaux de Brabois, Bâtiment des Spécialités Médicales, 5 rue du Morvan, F54511, Vandœuvre-lès-Nancy, France. .,Contrat d'Interface, Service de Rhumatologie, Hôpital de Brabois, Bâtiment Spécialités Médicales, F54511, Vandœuvre lès Nancy, France.
| |
Collapse
|
13
|
Chang YH, Wu KC, Wang CC, Ding DC. Enhanced chondrogenesis of human umbilical cord mesenchymal stem cells in a gelatin honeycomb scaffold. J Biomed Mater Res A 2020; 108:2069-2079. [PMID: 32323440 DOI: 10.1002/jbm.a.36966] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 03/19/2020] [Accepted: 03/28/2020] [Indexed: 12/16/2022]
Abstract
Transplantation of chondrogenic stem cells is a promising strategy for cartilage repair, but requires improvements in cell sourcing, maintenance, and chondrogenic differentiation efficiency. We examined whether gelatin honeycomb scaffolds can enhance the proliferation, viability, and chondrogenic capability of human umbilical cord mesenchymal stem cells (HUCMSCs) compared to standard plate cultures. In addition, the survival and phenotypic stability of HUCMSC-derived chondrocytes in a scaffold were evaluated in mice over 6 weeks post-transplantation. Survival and proliferation rates of HUCMSCs were comparable between scaffold and plate culture. Scaffold culture in a chondrogenic differentiation medium induced more stable expression of the key hyaline cartilage genes COL2A1 and ACAN and the production of the respective proteins Type II collagen and aggrecan as well as glycosaminoglycan. These HUCMSC-differentiated chondrocytes also stably expressed cartilage genes and proteins in the scaffold 6 weeks after transplantation into nonobese diabetic/severe combined immunodeficient mice. These findings indicate that honeycomb-like gelatin scaffolds can promote the survival and chondrogenic differentiation of HUCMSCs to form hyaline-like cartilage.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
| | - Kun-Chi Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
| | - Chen-Chie Wang
- Department of Orthopedics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, New Taipei City, Taiwan
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,Stem Cell Laboratory, Department of Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| |
Collapse
|
14
|
3D Culture of MSCs on a Gelatin Microsphere in a Dynamic Culture System Enhances Chondrogenesis. Int J Mol Sci 2020; 21:ijms21082688. [PMID: 32294921 PMCID: PMC7215541 DOI: 10.3390/ijms21082688] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 12/17/2022] Open
Abstract
Recent advancement in cartilage tissue engineering has explored the potential of 3D culture to mimic the in vivo environment of human cartilaginous tissue. Three-dimensional culture using microspheres was described to play a role in driving the differentiation of mesenchymal stem cells to chondrocyte lineage. However, factors such as mechanical agitation on cell chondrogenesis during culture on the microspheres has yet to be elucidated. In this study, we compared the 2D and 3D culture of bone-marrow-derived mesenchymal stem cells (BMSCs) on gelatin microspheres (GMs) in terms of MSC stemness properties, immune-phenotype, multilineage differentiation properties, and proliferation rate. Then, to study the effect of mechanical agitation on chondrogenic differentiation in 3D culture, we cultured BMSCs on GM (BMSCs-GM) in either static or dynamic bioreactor system with two different mediums, i.e., F12: DMEM (1:1) + 10% FBS (FD) and chondrogenic induction medium (CIM). Our results show that BMSCs attached to the GM surface and remained viable in 3D culture. BMSCs-GM proliferated faster and displayed higher stemness properties than BMSCs on a tissue culture plate (BMSCs-TCP). GMs also enhanced the efficiency of in-vitro chondrogenesis of BMSCs, especially in a dynamic culture with higher cell proliferation, RNA expression, and protein expression compared to that in a static culture. To conclude, our results indicate that the 3D culture of BMSCs on gelatin microsphere was superior to 2D culture on a standard tissue culture plate. Furthermore, culturing BMSCs on GM in dynamic culture conditions enhanced their chondrogenic differentiation.
Collapse
|
15
|
Up-Regulation of Superoxide Dismutase 2 in 3D Spheroid Formation Promotes Therapeutic Potency of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells. Antioxidants (Basel) 2020; 9:antiox9010066. [PMID: 31940867 PMCID: PMC7023074 DOI: 10.3390/antiox9010066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 02/07/2023] Open
Abstract
Umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) are accessible, available in abundance, and have been shown to be a promising source that can regenerate cartilage in patients with osteoarthritis or other orthopedic diseases. Recently, a three-dimensional (3D) cell culture system was developed to mimic the naive tissue microenvironment. However, the efficacy of cells generated from the 3D spheroid culture system has not yet been elucidated. In the present study, we demonstrate the changes in superoxide dismutase 2 (SOD2) gene expression, an indicator of oxidative stress, on 3D spheroid MSCs. Moreover, siRNA transfection and neutralizing antibody investigations were performed to confirm the function of SOD2 and E-cadherin. Overall, we found that SOD2 siRNA transfection in the spheroid form of MSCs increases the expression of apoptotic genes and decreases the clearance of mitochondrial reactive oxygen species (ROS). As a result, we confirm that 3D spheroid formation increases E-cadherin and SOD2 expression, ultimately regulating the phosphoinositide 3-kinase (PI3K/pAkt/pNrf2 and pERK/pNrf2 signaling pathway. Additionally, we show that SOD2 expression on 3D spheroid MSCs affects the regeneration rates of destructive cartilage in an osteoarthritic model. We postulate that the impact of SOD2 expression on 3D spheroid MSCs reduces oxidative stress and apoptosis, and also promotes cartilage regeneration.
Collapse
|
16
|
Gao S, Guo X, Zhao S, Jin Y, Zhou F, Yuan P, Cao L, Wang J, Qiu Y, Sun C, Kang Z, Gao F, Xu W, Hu X, Yang D, Qin Y, Ning K, Shaw PJ, Zhong G, Cheng L, Zhu H, Gao Z, Chen X, Xu J. Differentiation of human adipose-derived stem cells into neuron/motoneuron-like cells for cell replacement therapy of spinal cord injury. Cell Death Dis 2019; 10:597. [PMID: 31395857 PMCID: PMC6687731 DOI: 10.1038/s41419-019-1772-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/24/2019] [Accepted: 05/31/2019] [Indexed: 01/06/2023]
Abstract
Human adipose-derived stem cells (hADSCs) are increasingly presumed to be a prospective stem cell source for cell replacement therapy in various degenerative and/or traumatic diseases. The potential of trans-differentiating hADSCs into motor neuron cells indisputably provides an alternative way for spinal cord injury (SCI) treatment. In the present study, a stepwise and efficient hADSC trans-differentiation protocol with retinoic acid (RA), sonic hedgehog (SHH), and neurotrophic factors were developed. With this protocol hADSCs could be converted into electrophysiologically active motoneuron-like cells (hADSC-MNs), which expressed both a cohort of pan neuronal markers and motor neuron specific markers. Moreover, after being primed for neuronal differentiation with RA/SHH, hADSCs were transplanted into SCI mouse model and they survived, migrated, and integrated into injured site and led to partial functional recovery of SCI mice. When ablating the transplanted hADSC-MNs harboring HSV-TK-mCherry overexpression system with antivirial Ganciclovir (GCV), functional relapse was detected by motor-evoked potential (MEP) and BMS assays, implying that transplanted hADSC-MNs participated in rebuilding the neural circuits, which was further confirmed by retrograde neuronal tracing system (WGA). GFP-labeled hADSC-MNs were subjected to whole-cell patch-clamp recording in acute spinal cord slice preparation and both action potentials and synaptic activities were recorded, which further confirmed that those pre-conditioned hADSCs indeed became functionally active neurons in vivo. As well, transplanted hADSC-MNs largely prevented the formation of injury-induced cavities and exerted obvious immune-suppression effect as revealed by preventing astrocyte reactivation and favoring the secretion of a spectrum of anti-inflammatory cytokines and chemokines. Our work suggests that hADSCs can be readily transformed into MNs in vitro, and stay viable in spinal cord of the SCI mouse and exert multi-therapeutic effects by rebuilding the broken circuitry and optimizing the microenvironment through immunosuppression.
Collapse
Affiliation(s)
- Shane Gao
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xuanxuan Guo
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Simeng Zhao
- iHuman Institute, Shanghai Science and Technology University, Shanghai, 201210, China
| | - Yinpeng Jin
- Shanghai Public Health Clinical Center, Fudan University, JinShan, Shanghai, 201508, China
| | - Fei Zhou
- Department of Neurology, Third Affiliated Hospital of Navy Military Medical University, Shanghai, 200438, China
| | - Ping Yuan
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China
| | - Limei Cao
- Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China
| | - Jian Wang
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yue Qiu
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Chenxi Sun
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhanrong Kang
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 200137, China
| | - Fengjuan Gao
- Zhoupu hospital, Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Wei Xu
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xiao Hu
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China
| | - Danjing Yang
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Ying Qin
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Ke Ning
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ, UK
| | - Pamela J Shaw
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ, UK
| | - Guisheng Zhong
- iHuman Institute, Shanghai Science and Technology University, Shanghai, 201210, China.
| | - Liming Cheng
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Hongwen Zhu
- Tianjin Hospital, Tianjin, 300211, China. .,BOE Technology Group Co., Ltd., Beijing, 100176, China.
| | - Zhengliang Gao
- Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Xu Chen
- Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China.
| | - Jun Xu
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
17
|
Intra-articular delivery of synovium-resident mesenchymal stem cells via BMP-7-loaded fibrous PLGA scaffolds for cartilage repair. J Control Release 2019; 302:169-180. [DOI: 10.1016/j.jconrel.2019.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 03/31/2019] [Accepted: 04/03/2019] [Indexed: 12/16/2022]
|
18
|
Safari F, Fani N, Eglin D, Alini M, Stoddart MJ, Baghaban Eslaminejad M. Human umbilical cord-derived scaffolds for cartilage tissue engineering. J Biomed Mater Res A 2019; 107:1793-1802. [PMID: 30983084 DOI: 10.1002/jbm.a.36698] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 04/06/2019] [Accepted: 04/11/2019] [Indexed: 01/19/2023]
Abstract
Since articular cartilage is an avascular tissue, it has limited self-regeneration capacity after damage. Current methods for human bone marrow-derived mesenchymal stem cell (hBM-MSC) differentiation into cartilage result in tissues with a lower quality as compared to native articular cartilage. Decellularized biological scaffolds have the potential to provide appropriate signals, in order to support cellular retention, migration, proliferation, and differentiation. Given the high amount of collagen, hyaluronic acid (HA), and glycosaminoglycan (GAG) in umbilical cord, this tissue can be considered as an abundant natural biomaterial for tissue engineering applications. Human umbilical cord-derived scaffolds were prepared, and the chondrogenic induction of hBM-MSCs loaded onto the scaffolds was investigated. Gelatin-based scaffolds as a commercial material was used as a control. The results show that hBM-MSCs in tissue-derived scaffolds have an increased expression of chondrogenic markers compared with gelatin, whereas there are no significant differences between the expression of hypertrophic and osteogenic markers between tissue and gelatin scaffolds. In conclusion, it is confirmed that umbilical cord-derived scaffolds are able to provide a native environment for the cells and can promote cartilage differentiation. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1793-1802, 2019.
Collapse
Affiliation(s)
- Fatemeh Safari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Nesa Fani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - David Eglin
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
19
|
Neybecker P, Henrionnet C, Pape E, Mainard D, Galois L, Loeuille D, Gillet P, Pinzano A. In vitro and in vivo potentialities for cartilage repair from human advanced knee osteoarthritis synovial fluid-derived mesenchymal stem cells. Stem Cell Res Ther 2018; 9:329. [PMID: 30486903 PMCID: PMC6263063 DOI: 10.1186/s13287-018-1071-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/29/2018] [Accepted: 11/09/2018] [Indexed: 12/18/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) are found in synovial fluid (SF) and can easily be harvested during arthrocentesis or arthroscopy. However, SF-MSC characterization and chondrogenicity in collagen sponges have been poorly documented as well as their hypothetical in vivo chondroprotective properties with intra-articular injections during experimental osteoarthritis (OA). Methods SF-MSCs were isolated from human SF aspirates in patients suffering from advanced OA undergoing total knee joint replacements. SF-MSCs at passage 2 (P2) were characterized by flow cytometry for epitope profiling. SF-MSCs at P2 were subsequently cultured in vitro to assess their multilineage potentials. To assess their chondrogenicity, SF-MSCs at P4 were seeded in collagen sponges for 4 weeks under various oxygen tensions and growth factors combinations to estimate their gene profile and matrix production. Also, SF-MSCs were injected into the joints in a nude rat anterior cruciate ligament transection (ACLT) to macroscopically and histologically assess their possible chondroprotective properties,. Results We characterized the stemness (CD73+, CD90+, CD105+, CD34−, CD45−) and demonstrated the multilineage potency of SF-MSCs in vitro. Furthermore, the chondrogenic induction (TGF-ß1 ± BMP-2) of these SF-MSCs in collagen sponges demonstrated a good capacity of chondrogenic gene induction and extracellular matrix synthesis. Surprisingly, hypoxia did not enhance matrix synthesis, although it boosted chondrogenic gene expression (ACAN, SOX9, COL2A1). Besides, intra-articular injections of xenogenic SF-MSCs did exert neither chondroprotection nor inflammation in ACLT-induced OA in the rat knee. Conclusions Advanced OA SF-MSCs seem better candidates for cell-based constructs conceived for cartilage defects rather than intra-articular injections for diffuse OA.
Collapse
Affiliation(s)
- Paul Neybecker
- UMR 7365 CNRS-UL, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, 54505, Vandœuvre-Lès-Nancy, France
| | - Christel Henrionnet
- UMR 7365 CNRS-UL, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, 54505, Vandœuvre-Lès-Nancy, France
| | - Elise Pape
- UMR 7365 CNRS-UL, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, 54505, Vandœuvre-Lès-Nancy, France
| | - Didier Mainard
- UMR 7365 CNRS-UL, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, 54505, Vandœuvre-Lès-Nancy, France.,Service de Chirurgie Orthopédique, Traumatologique et Arthroscopique, CHRU Nancy, 29 Avenue du Maréchal de Lattre de Tassigny CO 60034, F54035, Nancy, France
| | - Laurent Galois
- UMR 7365 CNRS-UL, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, 54505, Vandœuvre-Lès-Nancy, France.,Service de Chirurgie Orthopédique, Traumatologique et Arthroscopique, CHRU Nancy, 29 Avenue du Maréchal de Lattre de Tassigny CO 60034, F54035, Nancy, France
| | - Damien Loeuille
- UMR 7365 CNRS-UL, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, 54505, Vandœuvre-Lès-Nancy, France.,Service de Rhumatologie, CHRU de Nancy, Hôpitaux de Brabois, Bâtiment des Spécialités Médicales, 5 rue du Morvan, F54511, Vandœuvre-lès-Nancy, France
| | - Pierre Gillet
- UMR 7365 CNRS-UL, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, 54505, Vandœuvre-Lès-Nancy, France
| | - Astrid Pinzano
- UMR 7365 CNRS-UL, IMoPA (Ingénierie Moléculaire et Physiopathologie Articulaire), Biopôle de l'Université de Lorraine, Campus Brabois-Santé, 9 Avenue de la Forêt de Haye, BP 20199, 54505, Vandœuvre-Lès-Nancy, France. .,Service de Rhumatologie, CHRU de Nancy, Hôpitaux de Brabois, Bâtiment des Spécialités Médicales, 5 rue du Morvan, F54511, Vandœuvre-lès-Nancy, France.
| |
Collapse
|
20
|
Rim YA, Nam Y, Ju JH. Application of Cord Blood and Cord Blood-Derived Induced Pluripotent Stem Cells for Cartilage Regeneration. Cell Transplant 2018; 28:529-537. [PMID: 30251563 PMCID: PMC7103603 DOI: 10.1177/0963689718794864] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Regeneration of articular cartilage is of great interest in cartilage tissue engineering
since articular cartilage has a low regenerative capacity. Due to the difficulty in
obtaining healthy cartilage for transplantation, there is a need to develop an alternative
and effective regeneration therapy to treat degenerative or damaged joint diseases. Stem
cells including various adult stem cells and pluripotent stem cells are now actively used
in tissue engineering. Here, we provide an overview of the current status of cord blood
cells and induced pluripotent stem cells derived from these cells in cartilage
regeneration. The abilities of these cells to undergo chondrogenic differentiation are
also described. Finally, the technical challenges of articular cartilage regeneration and
future directions are discussed.
Collapse
Affiliation(s)
- Yeri Alice Rim
- 1 CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoojun Nam
- 1 CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Hyeon Ju
- 1 CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,2 Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
21
|
Differences in the intrinsic chondrogenic potential of equine umbilical cord matrix and cord blood mesenchymal stromal/stem cells for cartilage regeneration. Sci Rep 2018; 8:13799. [PMID: 30217993 PMCID: PMC6138671 DOI: 10.1038/s41598-018-28164-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 06/13/2018] [Indexed: 12/15/2022] Open
Abstract
Umbilical cord blood mesenchymal stromal/stem cells (UCB-MSCs) and umbilical cord matrix MSCs (UCM-MSCs) have chondrogenic potential and are alternative sources to standard surgically derived bone marrow or adipose tissue collection for cartilage engineering. However, the majority of comparative studies explore neonatal MSCs potential only on ISCT benchmark assays accounting for some bias in the reproducibility between in vitro and in clinical studies. Therefore, we characterized equine UCB-MSCs and UCM-MSCs and investigated with particular attention their chondrogenesis potential in 3D culture with BMP-2 + TGF-ß1 in normoxia or hypoxia. We carried out an exhaustive characterization of the extracellular matrix generated by both these two types of MSCs after the induction of chondrogenesis through evaluation of hyaline cartilage, hypertrophic and osteogenic markers (mRNA, protein and histology levels). Some differences in hypoxia sensitivity and chondrogenesis were observed. UCB-MSCs differentiated into chondrocytes express an abundant, dense and a hyaline-like cartilage matrix. By contrast, despite their expression of cartilage markers, UCM-MSCs failed to express a relevant cartilage matrix after chondrogenic induction. Both MSCs types also displayed intrinsic differences at their undifferentiated basal status, UCB-MSCs expressing higher levels of chondrogenic markers whereas UCM-MSCs synthesizing higher amounts of osteogenic markers. Our results suggest that UCB-MSCs should be preferred for ex-vivo horse cartilage engineering. How those results should be translated to in vivo direct cartilage regeneration remains to be determined through dedicated study.
Collapse
|
22
|
Characterization and use of Equine Bone Marrow Mesenchymal Stem Cells in Equine Cartilage Engineering. Study of their Hyaline Cartilage Forming Potential when Cultured under Hypoxia within a Biomaterial in the Presence of BMP-2 and TGF-ß1. Stem Cell Rev Rep 2018; 13:611-630. [PMID: 28597211 DOI: 10.1007/s12015-017-9748-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Articular cartilage presents a poor capacity for self-repair. Its structure-function are frequently disrupted or damaged upon physical trauma or osteoarthritis in humans. Similar musculoskeletal disorders also affect horses and are the leading cause of poor performance or early retirement of sport- and racehorses. To develop a therapeutic solution for horses, we tested the autologous chondrocyte implantation technique developed on human bone marrow (BM) mesenchymal stem cells (MSCs) on horse BM-MSCs. This technique involves BM-MSC chondrogenesis using a combinatory approach based on the association of 3D-culture in collagen sponges, under hypoxia in the presence of chondrogenic factors (BMP-2 + TGF-β1) and siRNA to knockdown collagen I and HtrA1. Horse BM-MSCs were characterized before being cultured in chondrogenic conditions to find the best combination to enhance, stabilize, the chondrocyte phenotype. Our results show a very high proliferation of MSCs and these cells satisfy the criteria defining stem cells (pluripotency-surface markers expression). The combination of BMP-2 + TGF-β1 strongly induces the chondrogenic differentiation of MSCs and prevents HtrA1 expression. siRNAs targeting Col1a1 and Htra1 were functionally validated. Ultimately, the combined use of specific culture conditions defined here with specific growth factors and a Col1a1 siRNAs (50 nM) association leads to the in vitro synthesis of a hyaline-type neocartilage whose chondrocytes present an optimal phenotypic index similar to that of healthy, differentiated chondrocytes. Our results lead the way to setting up pre-clinical trials in horses to better understand the reaction of neocartilage substitute and to carry out a proof-of-concept of this therapeutic strategy on a large animal model.
Collapse
|
23
|
Someya T, Sano K, Hara K, Sagane Y, Watanabe T, Wijesekara RGS. Fibroblast and keratinocyte gene expression following exposure to the extracts of holy basil plant ( Ocimum tenuiflorum), malabar nut plant ( Justicia adhatoda), and emblic myrobalan plant ( Phyllanthus emblica). Data Brief 2018; 17:24-46. [PMID: 29876372 PMCID: PMC5988028 DOI: 10.1016/j.dib.2017.12.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/20/2017] [Accepted: 12/28/2017] [Indexed: 01/08/2023] Open
Abstract
This data article provides gene expression profiles, determined by using real-time PCR, of fibroblasts and keratinocytes treated with 0.01% and 0.001% extracts of holy basil plant (Ocimum tenuiflorum), sri lankan local name “maduruthala”, 0.1% and 0.01% extracts of malabar nut plant (Justicia adhatoda), sri lankan local name “adayhoda” and 0.003% and 0.001% extracts of emblic myrobalan plant (Phyllanthus emblica), sri lankan local name “nelli”, harvested in Sri Lanka. For fibroblasts, the dataset includes expression profiles for genes encoding hyaluronan synthase 1 (HAS1), hyaluronan synthase 2 (HAS2), hyaluronidase-1 (HYAL1), hyaluronidase-2 (HYAL2), versican, aggrecan, CD44, collagen, type I, alpha 1 (COL1A1), collagen, type III, alpha 1 (COL3A1), collagen, type VII, alpha 1 (COL7A1), matrix metalloproteinase 1 (MMP1), acid ceramidase, basic fibroblast growth factor (bFGF), fibroblast growth factor-7 (FGF7), vascular endothelial growth factor (VEGF), interleukin-1 alpha (IL-1α), cyclooxygenase-2 (cox2), transforming growth factor beta (TGF-β), and aquaporin 3 (AQP3). For keratinocytes, the expression profiles are for genes encoding HAS1, HAS2, HYAL1, HYAL2, versican, CD44, IL-1α, cox2, TGF-β, AQP3, Laminin5, collagen, type XVII, alpha 1 (COL17A1), integrin alpha-6 (ITGA6), ceramide synthase 3 (CERS3), elongation of very long chain fatty acids protein 1 (ELOVL1), elongation of very long chain fatty acids protein 4 (ELOVL4), filaggrin (FLG), transglutaminase 1 (TGM1), and keratin 1 (KRT1). The expression profiles are provided as bar graphs.
Collapse
Affiliation(s)
- Takao Someya
- ALBION Co. Ltd., 1-7-10 Ginza, Chuo-ku, Tokyo 104-0061, Japan
| | - Katsura Sano
- ALBION Co. Ltd., 1-7-10 Ginza, Chuo-ku, Tokyo 104-0061, Japan
| | - Kotaro Hara
- ALBION Co. Ltd., 1-7-10 Ginza, Chuo-ku, Tokyo 104-0061, Japan
| | - Yoshimasa Sagane
- Department of food and Cosmetic Science, Faculty of Bioindustry, Tokyo University of Agriculture, 196 Yasaka, Abashiri, Hokkaido 099-2493, Japan
| | - Toshihiro Watanabe
- Department of food and Cosmetic Science, Faculty of Bioindustry, Tokyo University of Agriculture, 196 Yasaka, Abashiri, Hokkaido 099-2493, Japan
| | - R G S Wijesekara
- Department of Aquaculture and Fisheries, Faculty of Livestock, Fisheries and Nutrition, Wayamba University of Sri Lanka, Makandura, Gonawila 60170, Sri Lanka
| |
Collapse
|
24
|
Desancé M, Contentin R, Bertoni L, Gomez-Leduc T, Branly T, Jacquet S, Betsch JM, Batho A, Legendre F, Audigié F, Galéra P, Demoor M. Chondrogenic Differentiation of Defined Equine Mesenchymal Stem Cells Derived from Umbilical Cord Blood for Use in Cartilage Repair Therapy. Int J Mol Sci 2018; 19:ijms19020537. [PMID: 29439436 PMCID: PMC5855759 DOI: 10.3390/ijms19020537] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/03/2018] [Accepted: 02/07/2018] [Indexed: 12/13/2022] Open
Abstract
Cartilage engineering is a new strategy for the treatment of cartilage damage due to osteoarthritis or trauma in humans. Racehorses are exposed to the same type of cartilage damage and the anatomical, cellular, and biochemical properties of their cartilage are comparable to those of human cartilage, making the horse an excellent model for the development of cartilage engineering. Human mesenchymal stem cells (MSCs) differentiated into chondrocytes with chondrogenic factors in a biomaterial appears to be a promising therapeutic approach for direct implantation and cartilage repair. Here, we characterized equine umbilical cord blood-derived MSCs (eUCB-MSCs) and evaluated their potential for chondrocyte differentiation for use in cartilage repair therapy. Our results show that isolated eUCB-MSCs had high proliferative capacity and differentiated easily into osteoblasts and chondrocytes, but not into adipocytes. A three-dimensional (3D) culture approach with the chondrogenic factors BMP-2 and TGF-β1 potentiated chondrogenic differentiation with a significant increase in cartilage-specific markers at the mRNA level (Col2a1, Acan, Snorc) and the protein level (type II and IIB collagen) without an increase in hypertrophic chondrocyte markers (Col10a1 and Mmp13) in normoxia and in hypoxia. However, these chondrogenic factors caused an increase in type I collagen, which can be reduced using small interfering RNA targeting Col1a2. This study provides robust data on MSCs characterization and demonstrates that eUCB-MSCs have a great potential for cartilage tissue engineering.
Collapse
Affiliation(s)
- Mélanie Desancé
- Normandie University, UNICAEN, BIOTARGEN, 14000 Caen, France.
| | | | - Lélia Bertoni
- Center of Imaging and Research on Locomotor Affections in Equines, Ecole Vétérinaire d'Alfort, Université Paris-Est, 14430 Goustranville, France.
| | | | - Thomas Branly
- Normandie University, UNICAEN, BIOTARGEN, 14000 Caen, France.
| | - Sandrine Jacquet
- Center of Imaging and Research on Locomotor Affections in Equines, Ecole Vétérinaire d'Alfort, Université Paris-Est, 14430 Goustranville, France.
| | - Jean-Marc Betsch
- Clinique Vétérinaire Equine de Méheudin, Méheudin, 61150 Ecouché, France.
| | - Agnès Batho
- Normandie University, UNICAEN, BIOTARGEN, 14000 Caen, France.
- EFS Caen, 14000 Caen, France.
| | | | - Fabrice Audigié
- Center of Imaging and Research on Locomotor Affections in Equines, Ecole Vétérinaire d'Alfort, Université Paris-Est, 14430 Goustranville, France.
| | - Philippe Galéra
- Normandie University, UNICAEN, BIOTARGEN, 14000 Caen, France.
| | - Magali Demoor
- Normandie University, UNICAEN, BIOTARGEN, 14000 Caen, France.
| |
Collapse
|
25
|
Stem Cells for Cartilage Repair: Preclinical Studies and Insights in Translational Animal Models and Outcome Measures. Stem Cells Int 2018. [PMID: 29535784 PMCID: PMC5832141 DOI: 10.1155/2018/9079538] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Due to the restricted intrinsic capacity of resident chondrocytes to regenerate the lost cartilage postinjury, stem cell-based therapies have been proposed as a novel therapeutic approach for cartilage repair. Moreover, stem cell-based therapies using mesenchymal stem cells (MSCs) or induced pluripotent stem cells (iPSCs) have been used successfully in preclinical and clinical settings. Despite these promising reports, the exact mechanisms underlying stem cell-mediated cartilage repair remain uncertain. Stem cells can contribute to cartilage repair via chondrogenic differentiation, via immunomodulation, or by the production of paracrine factors and extracellular vesicles. But before novel cell-based therapies for cartilage repair can be introduced into the clinic, rigorous testing in preclinical animal models is required. Preclinical models used in regenerative cartilage studies include murine, lapine, caprine, ovine, porcine, canine, and equine models, each associated with its specific advantages and limitations. This review presents a summary of recent in vitro data and from in vivo preclinical studies justifying the use of MSCs and iPSCs in cartilage tissue engineering. Moreover, the advantages and disadvantages of utilizing small and large animals will be discussed, while also describing suitable outcome measures for evaluating cartilage repair.
Collapse
|
26
|
Improvement of the Chondrocyte-Specific Phenotype upon Equine Bone Marrow Mesenchymal Stem Cell Differentiation: Influence of Culture Time, Transforming Growth Factors and Type I Collagen siRNAs on the Differentiation Index. Int J Mol Sci 2018; 19:ijms19020435. [PMID: 29389887 PMCID: PMC5855657 DOI: 10.3390/ijms19020435] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/26/2018] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
Articular cartilage is a tissue characterized by its poor intrinsic capacity for self-repair. This tissue is frequently altered upon trauma or in osteoarthritis (OA), a degenerative disease that is currently incurable. Similar musculoskeletal disorders also affect horses and OA incurs considerable economic loss for the equine sector. In the view to develop new therapies for humans and horses, significant progress in tissue engineering has led to the emergence of new generations of cartilage therapy. Matrix-associated autologous chondrocyte implantation is an advanced 3D cell-based therapy that holds promise for cartilage repair. This study aims to improve the autologous chondrocyte implantation technique by using equine mesenchymal stem cells (MSCs) from bone marrow differentiated into chondrocytes that can be implanted in the chondral lesion. The optimized protocol relies on culture under hypoxia within type I/III collagen sponges. Here, we explored three parameters that influence MSC differentiation: culture times, growth factors and RNA interference strategies. Our results suggest first that an increase in culture time from 14 to 28 or 42 days lead to a sharp increase in the expression of chondrocyte markers, notably type II collagen (especially the IIB isoform), along with a concomitant decrease in HtrA1 expression. Nevertheless, the expression of type I collagen also increased with longer culture times. Second, regarding the growth factor cocktail, TGF-β3 alone showed promising result but the previously tested association of BMP-2 and TGF-β1 better limits the expression of type I collagen. Third, RNA interference targeting Col1a2 as well as Col1a1 mRNA led to a more significant knockdown, compared with a conventional strategy targeting Col1a1 alone. This chondrogenic differentiation strategy showed a strong increase in the Col2a1:Col1a1 mRNA ratio in the chondrocytes derived from equine bone marrow MSCs, this ratio being considered as an index of the functionality of cartilage. These data provide evidence of a more stable chondrocyte phenotype when combining Col1a1 and Col1a2 siRNAs associated to a longer culture time in the presence of BMP-2 and TGF-β1, opening new opportunities for preclinical trials in the horse. In addition, because the horse is an excellent model for human articular cartilage disorders, the equine therapeutic approach developed here can also serve as a preclinical step for human medicine.
Collapse
|
27
|
Someya T, Sano K, Hara K, Sagane Y, Watanabe T, Wijesekara R. Fibroblast and keratinocyte gene expression following exposure to extracts of neem plant ( Azadirachta indica). Data Brief 2017; 16:982-992. [PMID: 29322079 PMCID: PMC5752095 DOI: 10.1016/j.dib.2017.12.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 12/05/2017] [Accepted: 12/14/2017] [Indexed: 01/08/2023] Open
Abstract
This data article provides gene expression profiles, determined by using real-time PCR, of fibroblasts and keratinocytes treated with 0.01% and 0.001% extracts of neem plant (Azadirachta indica), local name “Kohomba” in Sri Lanka, harvested in Sri Lanka. For fibroblasts, the dataset includes expression profiles for genes encoding hyaluronan synthase 1 (HAS1), hyaluronan synthase 2 (HAS2), hyaluronidase-1 (HYAL1), hyaluronidase-2 (HYAL2), versican, aggrecan, CD44, collagen, type I, alpha 1 (COL1A1), collagen, type III, alpha 1 (COL3A1), collagen, type VII, alpha 1 (COL7A1), matrix metalloproteinase 1 (MMP1), acid ceramidase, basic fibroblast growth factor (bFGF), fibroblast growth factor-7 (FGF7), vascular endothelial growth factor (VEGF), interleukin-1 alpha (IL-1α), cyclooxygenase-2 (cox2), transforming growth factor beta (TGF-β), and aquaporin 3 (AQP3). For keratinocytes, the expression profiles are for genes encoding HAS1, HAS2, HYAL1, HYAL2, versican, CD44, IL-1α, cox2, TGF-β, AQP3, Laminin5, collagen, type XVII, alpha 1 (COL17A1), integrin alpha-6 (ITGA6), ceramide synthase 3 (CERS3), elongation of very long chain fatty acids protein 1 (ELOVL1), elongation of very long chain fatty acids protein 4 (ELOVL4), filaggrin (FLG), transglutaminase 1 (TGM1), and keratin 1 (KRT1). The expression profiles are provided as bar graphs.
Collapse
Affiliation(s)
- Takao Someya
- ALBION Co. Ltd., 1–7-10 Ginza, Chuo-ku, Tokyo 104-0061, Japan
- Corresponding author.
| | - Katsura Sano
- ALBION Co. Ltd., 1–7-10 Ginza, Chuo-ku, Tokyo 104-0061, Japan
| | - Kotaro Hara
- ALBION Co. Ltd., 1–7-10 Ginza, Chuo-ku, Tokyo 104-0061, Japan
| | - Yoshimasa Sagane
- Department of food and Cosmetic Science, Faculty of Bioindustry, Tokyo University of Agriculture, 196 Yasaka, Abashiri, Hokkaido 099–2493, Japan
| | - Toshihiro Watanabe
- Department of food and Cosmetic Science, Faculty of Bioindustry, Tokyo University of Agriculture, 196 Yasaka, Abashiri, Hokkaido 099–2493, Japan
| | - R.G.S. Wijesekara
- Department of Aquaculture and Fisheries, Faculty of Livestock, Fisheries and Nutrition, Wayamba University of Sri Lanka, Makandura, Gonawila 60170, Sri Lanka
| |
Collapse
|
28
|
Hypoxia Is a Critical Parameter for Chondrogenic Differentiation of Human Umbilical Cord Blood Mesenchymal Stem Cells in Type I/III Collagen Sponges. Int J Mol Sci 2017; 18:ijms18091933. [PMID: 28885597 PMCID: PMC5618582 DOI: 10.3390/ijms18091933] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/29/2017] [Accepted: 09/01/2017] [Indexed: 12/18/2022] Open
Abstract
Umbilical cord blood (UCB) is an attractive alternative to bone marrow for isolation of mesenchymal stem cells (MSCs) to treat articular cartilage defects. Here, we set out to determine the growth factors (bone morphogenetic protein 2 (BMP-2) and transforming growth factor-β (TGF-β1)) and oxygen tension effects during chondrogenesis of human UCB-MSCs for cartilage engineering. Chondrogenic differentiation was induced using 3D cultures in type I/III collagen sponges with chondrogenic factors in normoxia (21% O₂) or hypoxia (<5% O₂) for 7, 14 and 21 days. Our results show that UCB-MSCs can be committed to chondrogenesis in the presence of BMP-2+TGF-β1. Normoxia induced the highest levels of chondrocyte-specific markers. However, hypoxia exerted more benefit by decreasing collagen X and matrix metalloproteinase-13 (MMP13) expression, two chondrocyte hypertrophy markers. However, a better chondrogenesis was obtained by switching oxygen conditions, with seven days in normoxia followed by 14 days in hypoxia, since these conditions avoid hypertrophy of hUCB-MSC-derived chondrocytes while maintaining the expression of chondrocyte-specific markers observed in normoxia. Our study demonstrates that oxygen tension is a key factor for chondrogenesis and suggests that UBC-MSCs 3D-culture should begin in normoxia to obtain a more efficient chondrocyte differentiation before placing them in hypoxia for chondrocyte phenotype stabilization. UCB-MSCs are therefore a reliable source for cartilage engineering.
Collapse
|
29
|
RNA Interference and BMP-2 Stimulation Allows Equine Chondrocytes Redifferentiation in 3D-Hypoxia Cell Culture Model: Application for Matrix-Induced Autologous Chondrocyte Implantation. Int J Mol Sci 2017; 18:ijms18091842. [PMID: 28837082 PMCID: PMC5618491 DOI: 10.3390/ijms18091842] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 12/29/2022] Open
Abstract
As in humans, osteoarthritis (OA) causes considerable economic loss to the equine industry. New hopes for cartilage repair have emerged with the matrix-associated autologous chondrocyte implantation (MACI). Nevertheless, its limitation is due to the dedifferentiation occurring during the chondrocyte amplification phase, leading to the loss of its capacity to produce a hyaline extracellular matrix (ECM). To enhance the MACI therapy efficiency, we have developed a strategy for chondrocyte redifferentiation, and demonstrated its feasibility in the equine model. Thus, to mimic the cartilage microenvironment, the equine dedifferentiated chondrocytes were cultured in type I/III collagen sponges for 7 days under hypoxia in the presence of BMP-2. In addition, chondrocytes were transfected by siRNA targeting Col1a1 and Htra1 mRNAs, which are overexpressed during dedifferentiation and OA. To investigate the quality of the neo-synthesized ECM, specific and atypical cartilage markers were evaluated by RT-qPCR and Western blot. Our results show that the combination of 3D hypoxia cell culture, BMP-2 (Bone morphogenetic protein-2), and RNA interference, increases the chondrocytes functional indexes (Col2a1/Col1a1, Acan/Col1a1), leading to an effective chondrocyte redifferentiation. These data represent a proof of concept for this process of application, in vitro, in the equine model, and will lead to the improvement of the MACI efficiency for cartilage tissue engineering therapy in preclinical/clinical trials, both in equine and human medicine.
Collapse
|
30
|
Kwon H, Haudenschild AK, Brown WE, Vapniarsky N, Paschos NK, Arzi B, Hu JC, Athanasiou KA. Tissue engineering potential of human dermis-isolated adult stem cells from multiple anatomical locations. PLoS One 2017; 12:e0182531. [PMID: 28767737 PMCID: PMC5540597 DOI: 10.1371/journal.pone.0182531] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/19/2017] [Indexed: 02/06/2023] Open
Abstract
Abundance and accessibility render skin-derived stem cells an attractive cell source for tissue engineering applications. Toward assessing their utility, the variability of constructs engineered from human dermis-isolated adult stem (hDIAS) cells was examined with respect to different anatomical locations (foreskin, breast, and abdominal skin), both in vitro and in a subcutaneous, athymic mouse model. All anatomical locations yielded hDIAS cells with multi-lineage differentiation potentials, though adipogenesis was not seen for foreskin-derived hDIAS cells. Using engineered cartilage as a model, tissue engineered constructs from hDIAS cells were compared. Construct morphology differed by location. The mechanical properties of human foreskin- and abdominal skin-derived constructs were similar at implantation, remaining comparable after 4 additional weeks of culture in vivo. Breast skin-derived constructs were not mechanically testable. For all groups, no signs of abnormality were observed in the host. Addition of aggregate redifferentiation culture prior to construct formation improved chondrogenic differentiation of foreskin-derived hDIAS cells, as evident by increases in glycosaminoglycan and collagen contents. More robust Alcian blue staining and homogeneous cell populations were also observed compared to controls. Human DIAS cells elicited no adverse host responses, reacted positively to chondrogenic regimens, and possessed multi-lineage differentiation potential with the caveat that efficacy may differ by anatomical origin of the skin. Taken together, these results suggest that hDIAS cells hold promise as a potential cell source for a number of tissue engineering applications.
Collapse
Affiliation(s)
- Heenam Kwon
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Anne K. Haudenschild
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Wendy E. Brown
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Natalia Vapniarsky
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Nikolaos K. Paschos
- Department of Orthopaedic Surgery, Penn Sports Medicine, University of Pennsylvania Health System, Philadelphia, PA, United States of America
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Jerry C. Hu
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering, University of California, Davis, CA, United States of America
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA, United States of America
- * E-mail:
| |
Collapse
|
31
|
Rai V, Dilisio MF, Dietz NE, Agrawal DK. Recent strategies in cartilage repair: A systemic review of the scaffold development and tissue engineering. J Biomed Mater Res A 2017; 105:2343-2354. [PMID: 28387995 DOI: 10.1002/jbm.a.36087] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/29/2017] [Indexed: 12/19/2022]
Abstract
Osteoarthritis results in irreparable loss of articular cartilage. Due to its avascular nature and low mitotic activity, cartilage has little intrinsic capacity for repair. Cartilage loss leads to pain, physical disability, movement restriction, and morbidity. Various treatment strategies have been proposed for cartilage regeneration, but the optimum treatment is yet to be defined. Tissue engineering with engineered constructs aimed towards developing a suitable substrate may help in cartilage regeneration by providing the mechanical, biological and chemical support to the cells. The use of scaffold as a substrate to support the progenitor cells or autologous chondrocytes has given promising results. Leakage of cells, poor cell survival, poor cell differentiation, inadequate integration into the host tissue, incorrect distribution of cells, and dedifferentiation of the normal cartilage are the common problems in tissue engineering. Current research is focused on improving mechanical and biochemical properties of scaffold to make it more efficient. The aim of this review is to provide a critical discussion on existing challenges, scaffold type and properties, and an update on ongoing recent developments in the architecture and composition of scaffold to enhance the proliferation and viability of mesenchymal stem cells. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2343-2354, 2017.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, 68178
| | - Matthew F Dilisio
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, 68178
- Department of Orthopedics, Creighton University School of Medicine, Omaha, Nebraska, 68178
| | - Nicholas E Dietz
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, 68178
- Department of Pathology, Creighton University School of Medicine, Omaha, Nebraska, 68178
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska, 68178
| |
Collapse
|
32
|
Nam Y, Rim YA, Ju JH. Chondrogenic Pellet Formation from Cord Blood-derived Induced Pluripotent Stem Cells. J Vis Exp 2017. [PMID: 28654049 DOI: 10.3791/55988] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Human articular cartilage lacks the ability to repair itself. Cartilage degeneration is thus treated not by curative but by conservative treatments. Currently, efforts are being made to regenerate damaged cartilage with ex vivo expanded chondrocytes or bone marrow-derived mesenchymal stem cells (BMSCs). However, the restricted viability and instability of these cells limit their application in cartilage reconstruction. Human induced pluripotent stem cells (hiPSCs) have received scientific attention as a new alternative for regenerative applications. With unlimited self-renewal ability and multipotency, hiPSCs have been highlighted as a new replacement cell source for cartilage repair. However, obtaining a high quantity of high-quality chondrogenic pellets is a major challenge to their clinical application. In this study, we used embryoid body (EB)-derived outgrowth cells for chondrogenic differentiation. Successful chondrogenesis was confirmed by PCR and staining with alcian blue, toluidine blue, and antibodies against collagen types I and II (COL1A1 and COL2A1, respectively). We provide a detailed method for the differentiation of cord blood mononuclear cell-derived iPSCs (CBMC-hiPSCs) into chondrogenic pellets.
Collapse
Affiliation(s)
- Yoojun Nam
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Division of Rheumatology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea
| | - Yeri Alice Rim
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Division of Rheumatology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea
| | - Ji Hyeon Ju
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea;
| |
Collapse
|
33
|
Cao Z, Liu C, Bai Y, Dou C, Li JM, Shi DW, Dong SW, Xiang Q. Inhibitory effect of dihydroartemisinin on chondrogenic and hypertrophic differentiation of mesenchymal stem cells. Am J Transl Res 2017; 9:2748-2759. [PMID: 28670366 PMCID: PMC5489878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/19/2017] [Indexed: 06/07/2023]
Abstract
Chondrocytes located in hyaline cartilage may maintain phenotype while the chondrocytes situated in calcified cartilage differentiate into hypertrophy. Chondrogenic and hypertrophic differentiation of mesenchymal stem cells (MSCs) are two subsequent processes during endochondral ossification. However, it is necessary for chondrocytes to hold homeostasis and to inhibit hypertrophic differentiation in stem cell-based regenerated cartilage. Dihydroartemisinin (DHA) is derived from artemisia apiacea which has many biological functions such as anti-malarial and anti-tumor. Whereas the effects of DHA on chondrogenic and hypertrophic differentiation are poorly understand. In this study, the cytotoxicity of DHA was determined by CCK8 assay and the cell apoptosis was analyzed by flow cytometry. Additionally, the effects of DHA on chondrogenic and hypertrophic differentiation of MSCs are explored by RT-PCR, western blotting and immunohistochemistry. The results showed that DHA inhibited expression of chondrogenic markers including Sox9 and Col2a1 by activating Nrf2 and Notch signaling. After induced to chondrogenesis, cells were treated with hypertrophic induced medium with DHA. The results revealed that hypertrophic markers including Runx2 and Col10a1 were down-regulated following DHA treatment through Pax6/HOXA2 and Gli transcription factors. These findings indicate that DHA is negative to chondrogenesis and is protective against chondrocyte hypertrophy to improve chondrocytes stability. Therefore, DHA might be not suited for chondogenesis but be potential as a new therapeutic candidate to maintain the biological function of regenerated cartilage.
Collapse
Affiliation(s)
- Zhen Cao
- Department of Anatomy, Third Military Medical UniversityChongqing, China
- Department of Biomedical Materials Science, Third Military Medical UniversityChongqing, China
| | - Chuan Liu
- Department of Biomedical Materials Science, Third Military Medical UniversityChongqing, China
| | - Yun Bai
- Department of Biomedical Materials Science, Third Military Medical UniversityChongqing, China
| | - Ce Dou
- Department of Biomedical Materials Science, Third Military Medical UniversityChongqing, China
| | - Jian-Mei Li
- Department of Biomedical Materials Science, Third Military Medical UniversityChongqing, China
| | - Duo-Wei Shi
- Department of Orthopaedics, Jinchuan Group Company Workers HospitalJinchang 737103, Gansu, China
| | - Shi-Wu Dong
- Department of Biomedical Materials Science, Third Military Medical UniversityChongqing, China
| | - Qiang Xiang
- Department of Emergency, Southwest Hospital, Third Military Medical UniversityChongqing, China
| |
Collapse
|
34
|
Legendre F, Ollitrault D, Gomez-Leduc T, Bouyoucef M, Hervieu M, Gruchy N, Mallein-Gerin F, Leclercq S, Demoor M, Galéra P. Enhanced chondrogenesis of bone marrow-derived stem cells by using a combinatory cell therapy strategy with BMP-2/TGF-β1, hypoxia, and COL1A1/HtrA1 siRNAs. Sci Rep 2017; 7:3406. [PMID: 28611369 PMCID: PMC5469741 DOI: 10.1038/s41598-017-03579-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 05/02/2017] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) hold promise for cartilage engineering. Here, we aimed to determine the best culture conditions to induce chondrogenesis of MSCs isolated from bone marrow (BM) of aged osteoarthritis (OA) patients. We showed that these BM-MSCs proliferate slowly, are not uniformly positive for stem cell markers, and maintain their multilineage potential throughout multiple passages. The chondrogenic lineage of BM-MSCs was induced in collagen scaffolds, under normoxia or hypoxia, by BMP-2 and/or TGF-β1. The best chondrogenic induction, with the least hypertrophic induction, was obtained with the combination of BMP-2 and TGF-β1 under hypoxia. Differentiated BM-MSCs were then transfected with siRNAs targeting two markers overexpressed in OA chondrocytes, type I collagen and/or HtrA1 protease. siRNAs significantly decreased mRNA and protein levels of type I collagen and HtrA1, resulting in a more typical chondrocyte phenotype, but with frequent calcification of the subcutaneously implanted constructs in a nude mouse model. Our 3D culture model with BMP-2/TGF-β1 and COL1A1/HtrA1 siRNAs was not effective in producing a cartilage-like matrix in vivo. Further optimization is needed to stabilize the chondrocyte phenotype of differentiated BM-MSCs. Nevertheless, this study offers the opportunity to develop a combinatory cellular therapy strategy for cartilage tissue engineering.
Collapse
Affiliation(s)
- Florence Legendre
- Caen Normandy University, France; UNICAEN EA7450 BioTARGen (Biologie, Génétique et Thérapies ostéoArticulaires et Respiratoires), 3 rue Nelson Mandela, 14280, Saint-Contest, France
| | - David Ollitrault
- Caen Normandy University, France; UNICAEN EA7450 BioTARGen (Biologie, Génétique et Thérapies ostéoArticulaires et Respiratoires), 3 rue Nelson Mandela, 14280, Saint-Contest, France
| | - Tangni Gomez-Leduc
- Caen Normandy University, France; UNICAEN EA7450 BioTARGen (Biologie, Génétique et Thérapies ostéoArticulaires et Respiratoires), 3 rue Nelson Mandela, 14280, Saint-Contest, France
| | - Mouloud Bouyoucef
- Caen Normandy University, France; UNICAEN EA7450 BioTARGen (Biologie, Génétique et Thérapies ostéoArticulaires et Respiratoires), 3 rue Nelson Mandela, 14280, Saint-Contest, France
| | - Magalie Hervieu
- Caen Normandy University, France; UNICAEN EA7450 BioTARGen (Biologie, Génétique et Thérapies ostéoArticulaires et Respiratoires), 3 rue Nelson Mandela, 14280, Saint-Contest, France
| | - Nicolas Gruchy
- Caen Normandy University, France; UNICAEN EA7450 BioTARGen (Biologie, Génétique et Thérapies ostéoArticulaires et Respiratoires), 3 rue Nelson Mandela, 14280, Saint-Contest, France
- Laboratoire de Cytogénétique Prénatale, Service de Génétique, CHU Caen, France
| | - Frédéric Mallein-Gerin
- Institute for Biology and Chemistry of Proteins, CNRS, UMR 5305 Laboratory of Tissue Biology and Therapeutic Engineering, Université Claude Bernard-Lyon 1 and University of Lyon, Lyon, France
| | - Sylvain Leclercq
- Caen Normandy University, France; UNICAEN EA7450 BioTARGen (Biologie, Génétique et Thérapies ostéoArticulaires et Respiratoires), 3 rue Nelson Mandela, 14280, Saint-Contest, France
- Service de Chirurgie Orthopédique, Clinique Saint-Martin, Caen, France
| | - Magali Demoor
- Caen Normandy University, France; UNICAEN EA7450 BioTARGen (Biologie, Génétique et Thérapies ostéoArticulaires et Respiratoires), 3 rue Nelson Mandela, 14280, Saint-Contest, France
| | - Philippe Galéra
- Caen Normandy University, France; UNICAEN EA7450 BioTARGen (Biologie, Génétique et Thérapies ostéoArticulaires et Respiratoires), 3 rue Nelson Mandela, 14280, Saint-Contest, France.
| |
Collapse
|
35
|
Li X, Teng Y, Liu J, Lin H, Fan Y, Zhang X. Chondrogenic differentiation of BMSCs encapsulated in chondroinductive polysaccharide/collagen hybrid hydrogels. J Mater Chem B 2017; 5:5109-5119. [PMID: 32264096 DOI: 10.1039/c7tb01020f] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Although BMSC-based therapy is one of the most front-line technologies for cartilage repair, it is still a big challenge to attain ideal niches for BMSC chondrogenic differentiation. In this study, we developed hyaluronate and chondroitin sulfate derivatives to prepare covalently crosslinked polysaccharide hydrogels. Based on these binary hydrogels, collagen was added to prepare ternary hybrid hydrogels and its effect on encapsulated BMSCs was studied. After culturing with different cell densities in vitro without the addition of growth factors for 3 weeks, the chondrogenesis of BMSCs was evaluated by CLSM, mechanical testing, histological staining, immunohistochemical staining and gene expression. The results indicated that BMSCs in high cell density (50 million per mL) cell-laden constructs had a more obvious chondrogenic phenotype than those in low cell density ones (5 million per mL). However, the components of hydrogels had a significant influence on chondrogenic differentiation. With the addition of collagen, the BMSCs in ternary hybrid hydrogels showed more significant chondrogenesis, possessing with more amounts of secreted glycosaminoglycans (GAGs) and type II collagen deposition, higher mechanical properties and chondrogenic gene expression over 3 weeks of culture in vitro. It can be concluded that the bioactive collagen is beneficial to the chondrogenesis of BMSCs. This hybrid hydrogels deserve further studies to have a prospective application in tissue engineering for cartilage defect repair.
Collapse
Affiliation(s)
- Xiupeng Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | | | | | | | | | | |
Collapse
|
36
|
Turajane T, Chaveewanakorn U, Fongsarun W, Aojanepong J, Papadopoulos KI. Avoidance of Total Knee Arthroplasty in Early Osteoarthritis of the Knee with Intra-Articular Implantation of Autologous Activated Peripheral Blood Stem Cells versus Hyaluronic Acid: A Randomized Controlled Trial with Differential Effects of Growth Factor Addition. Stem Cells Int 2017; 2017:8925132. [PMID: 29056974 PMCID: PMC5625803 DOI: 10.1155/2017/8925132] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/25/2017] [Accepted: 08/24/2017] [Indexed: 02/08/2023] Open
Abstract
In this randomized controlled trial, in early osteoarthritis (OA) that failed conservative intervention, the need for total knee arthroplasty (TKA) and WOMAC scores were evaluated, following a combination of arthroscopic microdrilling mesenchymal cell stimulation (MCS) and repeated intra-articular (IA) autologous activated peripheral blood stem cells (AAPBSCs) with growth factor addition (GFA) and hyaluronic acid (HA) versus IA-HA alone. Leukapheresis-harvested AAPBSCs were administered as three weekly IA injections combined with HA and GFA (platelet-rich plasma [PRP] and granulocyte colony-stimulating factor [hG-CSF]) and MCS in group 1 and in group 2 but without hG-CSF while group 3 received IA-HA alone. Each group of 20 patients was evaluated at baseline and at 1, 6, and, 12 months. At 12 months, all patients in the AAPBSC groups were surgical intervention free compared to three patients needing TKA in group 3 (p < 0.033). Total WOMAC scores showed statistically significant improvements at 6 and 12 months for the AAPBSC groups versus controls. There were no notable adverse events. We have shown avoidance of TKA in the AAPBSC groups at 12 months and potent, early, and sustained symptom alleviation through GFA versus HA alone. Differential effects of hG-CSF were noted with an earlier onset of symptom alleviation throughout.
Collapse
Affiliation(s)
- Thana Turajane
- 1Department of Orthopedic Surgery, Police General Hospital, Bangkok, Thailand
| | | | | | - Jongjate Aojanepong
- 3Department of Gynecology and Obstetrics, Police General Hospital, Bangkok, Thailand
| | | |
Collapse
|