1
|
Rosochowicz MA, Lach MS, Richter M, Jagiełło I, Suchorska WM, Trzeciak T. The iPSC secretome is beneficial for in vitro propagation of primary osteoarthritic chondrocytes cell lines. Biochem Biophys Res Commun 2024; 730:150392. [PMID: 39003867 DOI: 10.1016/j.bbrc.2024.150392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND One of the obstacles to autologous chondrocyte implantation (ACI) is obtaining a large quantity of chondrocytes without depletion of their properties. The conditioned medium (CM) from different subpopulations of stem cells (mesenchymal stromal cells (MSC) or induced pluripotent stem cells (iPSC)) could be a gamechanger. MSCs' potential is related to the donor's health and age, which could be omitted when, as a source, iPSCs are used. There is a lack of data regarding their use in the chondrocyte culture expansion. Thus, we wanted to verify whether iPSC-CM could be beneficial for the cell culture of primary chondrocyte cells. METHODS We added the iPSC-CMs from GPCCi001-A and ND 41658*H cells to the culture of primary chondrocyte cell lines isolated from OA patients (n = 6) for other two passages. The composition of the CM was evaluated using Luminex technology. Then, we analysed the senescence, proliferation rate and using flow cytometry: viability, distribution of cell cycle phases, production of reactive oxygen species (ROS) and double-strand breaks. The cartilage-related markers were evaluated using Western blot and immunofluorescence. Additionally, a three-dimensional cell culture was used to determine the potential to form cartilage particles. RESULTS iPSC-CM increased proliferation and diminished cell ROS production and senescence. CM influenced the cartilage-related protein expression and promoted the growth of cartilage particles. The cell exposed to CM did not lose the ECM proteins, suggesting the chondroprotective effect for prolonged culture time. CONCLUSION Our preliminary results suggest a beneficial effect on maintaining chondrocyte biology during in vitro expansion.
Collapse
Affiliation(s)
- Monika A Rosochowicz
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland; Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland; Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland.
| | - Michał S Lach
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland; Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland
| | - Magdalena Richter
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland
| | - Inga Jagiełło
- Department of Tumour Pathology, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland
| | - Wiktoria M Suchorska
- Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland; Department of Electroradiology, Poznan University of Medical Sciences, Garbary 15 Street, 61-866, Poznan, Poland
| | - Tomasz Trzeciak
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland
| |
Collapse
|
2
|
Ali EAM, Smaida R, Meyer M, Ou W, Li Z, Han Z, Benkirane-Jessel N, Gottenberg JE, Hua G. iPSCs chondrogenic differentiation for personalized regenerative medicine: a literature review. Stem Cell Res Ther 2024; 15:185. [PMID: 38926793 PMCID: PMC11210138 DOI: 10.1186/s13287-024-03794-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Cartilage, an important connective tissue, provides structural support to other body tissues, and serves as a cushion against impacts throughout the body. Found at the end of the bones, cartilage decreases friction and averts bone-on-bone contact during joint movement. Therefore, defects of cartilage can result from natural wear and tear, or from traumatic events, such as injuries or sudden changes in direction during sports activities. Overtime, these cartilage defects which do not always produce immediate symptoms, could lead to severe clinical pathologies. The emergence of induced pluripotent stem cells (iPSCs) has revolutionized the field of regenerative medicine, providing a promising platform for generating various cell types for therapeutic applications. Thus, chondrocytes differentiated from iPSCs become a promising avenue for non-invasive clinical interventions for cartilage injuries and diseases. In this review, we aim to highlight the current strategies used for in vitro chondrogenic differentiation of iPSCs and to explore their multifaceted applications in disease modeling, drug screening, and personalized regenerative medicine. Achieving abundant functional iPSC-derived chondrocytes requires optimization of culture conditions, incorporating specific growth factors, and precise temporal control. Continual improvements in differentiation methods and integration of emerging genome editing, organoids, and 3D bioprinting technologies will enhance the translational applications of iPSC-derived chondrocytes. Finally, to unlock the benefits for patients suffering from cartilage diseases through iPSCs-derived technologies in chondrogenesis, automatic cell therapy manufacturing systems will not only reduce human intervention and ensure sterile processes within isolator-like platforms to minimize contamination risks, but also provide customized production processes with enhanced scalability and efficiency.
Collapse
Affiliation(s)
- Eltahir Abdelrazig Mohamed Ali
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1260, Regenerative NanoMedicine (RNM), 1 Rue Eugène Boeckel, 67000, Strasbourg, France
- Université de Strasbourg, 67000, Strasbourg, France
| | - Rana Smaida
- Lamina Therapeutics, 1 Rue Eugène Boeckel, 67000, Strasbourg, France
| | - Morgane Meyer
- Université de Strasbourg, 67000, Strasbourg, France
- Lamina Therapeutics, 1 Rue Eugène Boeckel, 67000, Strasbourg, France
| | - Wenxin Ou
- Université de Strasbourg, 67000, Strasbourg, France
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), Service de Rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 67000, Strasbourg, France
- Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Zongjin Li
- Nankai University School of Medicine, Tianjin, 300071, China
| | - Zhongchao Han
- Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co, Beijing, 100176, China
| | - Nadia Benkirane-Jessel
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1260, Regenerative NanoMedicine (RNM), 1 Rue Eugène Boeckel, 67000, Strasbourg, France.
- Université de Strasbourg, 67000, Strasbourg, France.
- Lamina Therapeutics, 1 Rue Eugène Boeckel, 67000, Strasbourg, France.
| | - Jacques Eric Gottenberg
- Université de Strasbourg, 67000, Strasbourg, France.
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), Service de Rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 67000, Strasbourg, France.
| | - Guoqiang Hua
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR 1260, Regenerative NanoMedicine (RNM), 1 Rue Eugène Boeckel, 67000, Strasbourg, France.
- Université de Strasbourg, 67000, Strasbourg, France.
| |
Collapse
|
3
|
Owaidah A. Induced pluripotent stem cells in cartilage tissue engineering: a literature review. Biosci Rep 2024; 44:BSR20232102. [PMID: 38563479 PMCID: PMC11088306 DOI: 10.1042/bsr20232102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024] Open
Abstract
Osteoarthritis (OA) is a long-term, persistent joint disorder characterized by bone and cartilage degradation, resulting in tightness, pain, and restricted movement. Current attempts in cartilage regeneration are cell-based therapies using stem cells. Multipotent stem cells, such as mesenchymal stem cells (MSCs), and pluripotent stem cells, such as embryonic stem cells (ESCs), have been used to regenerate cartilage. However, since the discovery of human-induced pluripotent stem cells (hiPSCs) in 2007, it was seen as a potential source for regenerative chondrogenic therapy as it overcomes the ethical issues surrounding the use of ESCs and the immunological and differentiation limitations of MSCs. This literature review focuses on chondrogenic differentiation and 3D bioprinting technologies using hiPSCS, suggesting them as a viable source for successful tissue engineering. METHODS A literature search was conducted using scientific search engines, PubMed, MEDLINE, and Google Scholar databases with the terms 'Cartilage tissue engineering' and 'stem cells' to retrieve published literature on chondrogenic differentiation and tissue engineering using MSCs, ESCs, and hiPSCs. RESULTS hiPSCs may provide an effective and autologous treatment for focal chondral lesions, though further research is needed to explore the potential of such technologies. CONCLUSIONS This review has provided a comprehensive overview of these technologies and the potential applications for hiPSCs in regenerative medicine.
Collapse
Affiliation(s)
- Amani Y. Owaidah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
4
|
Yammine KM, Abularach SM, Kim SY, Bikovtseva AA, Lilianty J, Butty VL, Schiavoni RP, Bateman JF, Lamandé SR, Shoulders MD. ER procollagen storage defect without coupled unfolded protein response drives precocious arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.19.562780. [PMID: 37905055 PMCID: PMC10614947 DOI: 10.1101/2023.10.19.562780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Collagenopathies are a group of clinically diverse disorders caused by defects in collagen folding and secretion. For example, mutations in the gene encoding collagen type-II, the primary collagen in cartilage, can lead to diverse chondrodysplasias. One example is the Gly1170Ser substitution in procollagen-II, which causes precocious osteoarthritis. Here, we biochemically and mechanistically characterize an induced pluripotent stem cell-based cartilage model of this disease, including both hetero- and homozygous genotypes. We show that Gly1170Ser procollagen-II is notably slow to fold and secrete. Instead, procollagen-II accumulates intracellularly, consistent with an endoplasmic reticulum (ER) storage disorder. Owing to unique features of the collagen triple helix, this accumulation is not recognized by the unfolded protein response. Gly1170Ser procollagen-II interacts to a greater extent than wild-type with specific proteostasis network components, consistent with its slow folding. These findings provide mechanistic elucidation into the etiology of this disease. Moreover, the cartilage model will enable rapid testing of therapeutic strategies to restore proteostasis in the collagenopathies.
Collapse
|
5
|
Zhang Z, Mu Y, Zhou H, Yao H, Wang DA. Cartilage Tissue Engineering in Practice: Preclinical Trials, Clinical Applications, and Prospects. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:473-490. [PMID: 36964757 DOI: 10.1089/ten.teb.2022.0190] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
Articular cartilage defects significantly compromise the quality of life in the global population. Although many strategies are needed to repair articular cartilage, including microfracture, autologous osteochondral transplantation, and osteochondral allograft, the therapeutic effects remain suboptimal. In recent years, with the development of cartilage tissue engineering, scientists have continuously improved the formulations of therapeutic cells, biomaterial-based scaffolds, and biological factors, which have opened new avenues for better therapeutics of cartilage lesions. This review focuses on advances in cartilage tissue engineering, particularly in preclinical trials and clinical applications, prospects, and challenges.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, P.R. China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, P.R. China
| |
Collapse
|
6
|
Rastegar Adib F, Bagheri F, Sharifi AM. Osteochondral regeneration in rabbit using xenograft decellularized ECM in combination with different biological products; platelet-rich fibrin, amniotic membrane extract, and mesenchymal stromal cells. J Biomed Mater Res B Appl Biomater 2022; 110:2089-2099. [PMID: 35383398 DOI: 10.1002/jbm.b.35063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 11/11/2022]
Abstract
This study aimed to investigate the regenerative effect of decellularized osteochondral ECM xenograft in combination with various biological products in an osteochondral (OC) defect. OC tissue from the sheep femur were obtained and decellularized. The decellularized ECM (dECM) was combined with either platelet-rich fibrin (PRF), amniotic membrane extract (AME), or rabbit bone marrow-derived mesenchymal stromal cells (rBMSCs). The hybrid dECM-biological products were then utilized for the treatment of rabbit OC critical size defects. The regenerative potential of different groups was compared using; MRI, macroscopic assessment, histopathology, and histomorphometry. All characterizations analysis verified successful decellularization. Three months post-surgery, macroscopic findings indicated that dECM was better compared to controls. Also, dECM in combination with AME, PRF, and rBMSCs showed enhanced OC regeneration compared to only dECM (AME: +100%, PRF: +61%, rBMSCs: +28%). In particular, the dECM+AME group results in the best integration of new cartilage into surrounding cartilage tissue. The histomorphometric evaluations demonstrated enhancement in new cartilage formation and bone tissue (86.5 ± 5.9% and 90 ± 7.7%, respectively) for the dECM+AME group compared to other groups. Furthermore, histological results for the dECM+AME elucidated a mature hyaline cartilage tissue that covered the new and symmetrically formed subchondral bone, exhibiting a significantly higher regenerative effect compared to all other treated groups. This finding was also confirmed with MRI images. The current study revealed that in addition to the benefits of dECM alone, its combination with AME indicated to have a superior regenerative effect on OC regeneration. Overall, dECM+AME may be considered a suitable construct for treating knee OC injuries.
Collapse
Affiliation(s)
- Fatemeh Rastegar Adib
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Bagheri
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Ali Mohammad Sharifi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Tissue Engineering Group, (NOCERAL), Department of Orthopedics Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
7
|
The Induced Pluripotent Stem Cells in Articular Cartilage Regeneration and Disease Modelling: Are We Ready for Their Clinical Use? Cells 2022; 11:cells11030529. [PMID: 35159338 PMCID: PMC8834349 DOI: 10.3390/cells11030529] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 02/01/2023] Open
Abstract
The development of induced pluripotent stem cells has brought unlimited possibilities to the field of regenerative medicine. This could be ideal for treating osteoarthritis and other skeletal diseases, because the current procedures tend to be short-term solutions. The usage of induced pluripotent stem cells in the cell-based regeneration of cartilage damages could replace or improve on the current techniques. The patient’s specific non-invasive collection of tissue for reprogramming purposes could also create a platform for drug screening and disease modelling for an overview of distinct skeletal abnormalities. In this review, we seek to summarise the latest achievements in the chondrogenic differentiation of pluripotent stem cells for regenerative purposes and disease modelling.
Collapse
|
8
|
Kamaraj A, Kyriacou H, Seah KTM, Khan WS. Use of human induced pluripotent stem cells for cartilage regeneration in vitro and within chondral defect models of knee joint cartilage in vivo: a Preferred Reporting Items for Systematic Reviews and Meta-Analyses systematic literature review. Cytotherapy 2021; 23:647-661. [PMID: 34059422 DOI: 10.1016/j.jcyt.2021.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 03/16/2021] [Accepted: 03/27/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS Articular cartilage has limited regenerative ability when damaged through trauma or disease. Failure to treat focal chondral lesions results in changes that inevitably progress to osteoarthritis. Osteoarthritis is a major contributor to disability globally, which results in significant medical costs and lost wages every year. Human induced pluripotent stem cells (hiPSCs) have long been considered a potential autologous therapeutic option for the treatment of focal chondral lesions. Although there are significant advantages to hiPSCs over other stem cell options, such as mesenchymal and embryonic stem cells, there are concerns regarding their ability to form bona fide cartilage and their tumorgenicity in vivo. METHODS The authors carried out a systematic literature review on the use of hiPSCs to produce differentiated progeny capable of producing high-quality cartilage in vitro and regenerate cartilage in osteochondral defects in vivo in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Eight studies were included in the review that used hiPSCs or their derived progeny in xenogeneic transplants in animal models to regenerate cartilage in osteochondral defects of the knee joint. The in vitro-differentiated, hiPSC-derived and in vivo defect repair ability of the hiPSC-derived progeny transplants were assessed. RESULTS Most studies reported the generation of high-quality cartilage-producing progeny that were able to successfully repair cartilage defects in vivo. No tumorigenicity was observed. CONCLUSIONS The authors conclude that hiPSCs offer a valuable source of cartilage-producing progeny that show promise as an effective cell-based therapy in treating focal chondral lesions.
Collapse
Affiliation(s)
- Achi Kamaraj
- Division of Trauma and Orthopedic Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Harry Kyriacou
- Division of Trauma and Orthopedic Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - K T Matthew Seah
- Division of Trauma and Orthopedic Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Wasim S Khan
- Division of Trauma and Orthopedic Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Lee MS, Stebbins MJ, Jiao H, Huang HC, Leiferman EM, Walczak BE, Palecek SP, Shusta EV, Li WJ. Comparative evaluation of isogenic mesodermal and ectomesodermal chondrocytes from human iPSCs for cartilage regeneration. SCIENCE ADVANCES 2021; 7:eabf0907. [PMID: 34138734 PMCID: PMC8133756 DOI: 10.1126/sciadv.abf0907] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/29/2021] [Indexed: 05/12/2023]
Abstract
Generating phenotypic chondrocytes from pluripotent stem cells is of great interest in the field of cartilage regeneration. In this study, we differentiated human induced pluripotent stem cells into the mesodermal and ectomesodermal lineages to prepare isogenic mesodermal cell-derived chondrocytes (MC-Chs) and neural crest cell-derived chondrocytes (NCC-Chs), respectively, for comparative evaluation. Our results showed that both MC-Chs and NCC-Chs expressed hyaline cartilage-associated markers and were capable of generating hyaline cartilage-like tissue ectopically and at joint defects. Moreover, NCC-Chs revealed closer morphological and transcriptional similarities to native articular chondrocytes than MC-Chs. NCC-Ch implants induced by our growth factor mixture demonstrated increased matrix production and stiffness compared to MC-Ch implants. Our findings address how chondrocytes derived from pluripotent stem cells through mesodermal and ectomesodermal differentiation are different in activities and functions, providing the crucial information that helps make appropriate cell choices for effective regeneration of articular cartilage.
Collapse
Affiliation(s)
- Ming-Song Lee
- Laboratory of Musculoskeletal Biology and Regenerative Medicine, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Matthew J Stebbins
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Hongli Jiao
- Laboratory of Musculoskeletal Biology and Regenerative Medicine, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Hui-Ching Huang
- Laboratory of Musculoskeletal Biology and Regenerative Medicine, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ellen M Leiferman
- Laboratory of Musculoskeletal Biology and Regenerative Medicine, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Brian E Walczak
- Laboratory of Musculoskeletal Biology and Regenerative Medicine, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Wan-Ju Li
- Laboratory of Musculoskeletal Biology and Regenerative Medicine, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA.
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
10
|
Rejuvenated Stem/Progenitor Cells for Cartilage Repair Using the Pluripotent Stem Cell Technology. Bioengineering (Basel) 2021; 8:bioengineering8040046. [PMID: 33920285 PMCID: PMC8070387 DOI: 10.3390/bioengineering8040046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 01/19/2023] Open
Abstract
It is widely accepted that chondral defects in articular cartilage of adult joints are never repaired spontaneously, which is considered to be one of the major causes of age-related degenerative joint disorders, such as osteoarthritis. Since mobilization of subchondral bone (marrow) cells and addition of chondrocytes or mesenchymal stromal cells into full-thickness defects show some degrees of repair, the lack of self-repair activity in adult articular cartilage can be attributed to lack of reparative cells in adult joints. In contrast, during a fetal or embryonic stage, joint articular cartilage has a scar-less repair activity, suggesting that embryonic joints may contain cells responsible for such activity, which can be chondrocytes, chondroprogenitors, or other cell types such as skeletal stem cells. In this respect, the tendency of pluripotent stem cells (PSCs) to give rise to cells of embryonic characteristics will provide opportunity, especially for humans, to obtain cells carrying similar cartilage self-repair activity. Making use of PSC-derived cells for cartilage repair is still in a basic or preclinical research phase. This review will provide brief overviews on how human PSCs have been used for cartilage repair studies.
Collapse
|
11
|
Csobonyeiova M, Polak S, Nicodemou A, Zamborsky R, Danisovic L. iPSCs in Modeling and Therapy of Osteoarthritis. Biomedicines 2021; 9:186. [PMID: 33673154 PMCID: PMC7917981 DOI: 10.3390/biomedicines9020186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA) belongs to chronic degenerative disorders and is often a leading cause of disability in elderly patients. Typically, OA is manifested by articular cartilage erosion, pain, stiffness, and crepitus. Currently, the treatment options are limited, relying mostly on pharmacological therapy, which is often related to numerous complications. The proper management of the disease is challenging because of the poor regenerative capacity of articular cartilage. During the last decade, cell-based approaches such as implantation of autologous chondrocytes or mesenchymal stem cells (MSCs) have shown promising results. However, the mentioned techniques face their hurdles (cell harvesting, low proliferation capacity). The invention of induced pluripotent stem cells (iPSCs) has created new opportunities to increase the efficacy of the cartilage healing process. iPSCs may represent an unlimited source of chondrocytes derived from a patient's somatic cells, circumventing ethical and immunological issues. Aside from the regenerative potential of iPSCs, stem cell-derived cartilage tissue models could be a useful tool for studying the pathological process of OA. In our recent article, we reviewed the progress in chondrocyte differentiation techniques, disease modeling, and the current status of iPSC-based regenerative therapy of OA.
Collapse
Affiliation(s)
- Maria Csobonyeiova
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia; (M.C.); (S.P.)
| | - Stefan Polak
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia; (M.C.); (S.P.)
| | - Andreas Nicodemou
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia;
| | - Radoslav Zamborsky
- National Institute of Children’s Diseases, Department of Orthopedics, Faculty of Medicine, Comenius University, Limbova 1, 833 40 Bratislava, Slovakia;
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia;
- Regenmed Ltd., Medena 29, 811 01 Bratislava, Slovakia
| |
Collapse
|
12
|
Yamashita A, Tsumaki N. Recent progress of animal transplantation studies for treating articular cartilage damage using pluripotent stem cells. Dev Growth Differ 2021; 63:72-81. [PMID: 33411345 DOI: 10.1111/dgd.12706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022]
Abstract
Focal articular cartilage damage can eventually lead to the onset of osteoarthritis with degradation around healthy articular cartilage. Currently, there are no drugs available that effectively repair articular cartilage damage. Several surgical techniques exist and are expected to prevent progression to osteoarthritis, but they do not offer a long-term clinical solution. Recently, regenerative medicine approaches using human pluripotent stem cells (PSCs) have gained attention as new cell sources for therapeutic products. To translate PSCs to clinical application, appropriate cultures that produce large amounts of chondrocytes and hyaline cartilage are needed. So too are assays for the safety and efficacy of the cellular materials in preclinical studies including animal transplantation models. To confirm safety and efficacy, transplantation into the subcutaneous space and articular cartilage defects have been performed in animal models. All but one study we reviewed that transplanted PSC-derived cellular products into articular cartilage defects found safe and effective recovery. However, for most of those studies, the quality of the PSCs was not verified, and the evaluations were done with small animals over short observation periods. Large animals and longer observation times are preferred. We will discuss the recent progress and future direction of the animal transplantation studies for the treatment of focal articular cartilage damages using PSCs.
Collapse
Affiliation(s)
- Akihiro Yamashita
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Noriyuki Tsumaki
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
13
|
Taghiyar L, Jahangir S, Khozaei Ravari M, Shamekhi MA, Eslaminejad MB. Cartilage Repair by Mesenchymal Stem Cell-Derived Exosomes: Preclinical and Clinical Trial Update and Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1326:73-93. [PMID: 33629260 DOI: 10.1007/5584_2021_625] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) and other degenerative joint diseases are characterized by articular cartilage destruction, synovial inflammation, sclerosis of subchondral bone, and loss of extracellular matrix (ECM). Worldwide, these diseases are major causes of disability. Cell therapies have been considered to be the best therapeutic strategies for long-term treatment of articular cartilage diseases. It has been suggested that the mechanism of stem cell-based therapy is related to paracrine secretion of extracellular vesicles (EVs), which are recognized as the main secretion factors of stem cells. EVs, and in particular the subclass exosomes (Exos), are novel therapeutic approaches for treatment of cartilage lesions and OA. The results of recent studies have shown that EVs isolated from mesenchymal stem cells (MSCs) could inhibit OA progression. EVs isolated from various stem cell sources, such as MSCs, may contribute to tissue regeneration of the limbs, skin, heart, and other tissues. Here, we summarize recent findings of preclinical and clinical studies on different MSC-derived EVs and their effectiveness as a treatment for damaged cartilage. The Exos isolation techniques in OA treatment are also highlighted.
Collapse
Affiliation(s)
- Leila Taghiyar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahrbano Jahangir
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mojtaba Khozaei Ravari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - 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
|
14
|
Zhao Y, Liu H, Zhao C, Dang P, Li H, Farzaneh M. Paracrine Interactions Involved in Human Induced Pluripotent Stem Cells Differentiation into Chondrocytes. Curr Stem Cell Res Ther 2020; 15:233-242. [PMID: 31889496 DOI: 10.2174/1574888x15666191224122058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/24/2019] [Accepted: 10/30/2019] [Indexed: 12/20/2022]
Abstract
Osteoarthritis (OA), as a degenerative joint disease, is the most common form of joint disorder that causes pain, stiffness, and other symptoms associated with OA. Various genetic, biomechanical, and environmental factors have a relevant role in the development of OA. To date, extensive efforts are currently being made to overcome the poor self-healing capacity of articular cartilage. Despite the pivotal role of chondrocytes, their proliferation and repair capacity after tissue injury are limited. Therefore, the development of new strategies to overcome these constraints is urgently needed. Recent advances in regenerative medicine suggest that pluripotent stem cells are promising stem cell sources for cartilage repair. Pluripotent stem cells are undifferentiated cells that have the capacity to differentiate into different types of cells and can self-renew indefinitely. In the past few decades, numerous attempts have been made to regenerate articular cartilage by using induced pluripotent stem cells (iPSCs). The potential applications of patient-specific iPSCs hold great promise for regenerative medicine and OA treatment. However, there are different culture conditions for the preparation and characterization of human iPSCs-derived chondrocytes (hiChondrocytes). Recent biochemical analyses reported that several paracrine factors such as TGFb, BMPs, WNT, Ihh, and Runx have been shown to be involved in cartilage cell proliferation and differentiation from human iPSCs. In this review, we summarize and discuss the paracrine interactions involved in human iPSCs differentiation into chondrocytes in different cell culture media.
Collapse
Affiliation(s)
- Yunchang Zhao
- Department of Orthopedics III, Zhoukou Central Hospital, Zhoukou, Henan 466000, China
| | - Honghao Liu
- Department of Orthopedics III, Zhoukou Central Hospital, Zhoukou, Henan 466000, China
| | - Chunjie Zhao
- Department of Orthopedics III, Zhoukou Central Hospital, Zhoukou, Henan 466000, China
| | - Peng Dang
- Department of Orthopedics III, Zhoukou Central Hospital, Zhoukou, Henan 466000, China
| | - Haijian Li
- Department of Orthopedics III, Zhoukou Central Hospital, Zhoukou, Henan 466000, China
| | - Maryam Farzaneh
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
15
|
Shah S, Otsuka T, Bhattacharjee M, Laurencin CT. Minimally Invasive Cellular Therapies for Osteoarthritis Treatment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-020-00184-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Yamashita A, Yoshitomi H, Kihara S, Toguchida J, Tsumaki N. Culture substrate-associated YAP inactivation underlies chondrogenic differentiation of human induced pluripotent stem cells. Stem Cells Transl Med 2020; 10:115-127. [PMID: 32822104 PMCID: PMC7780802 DOI: 10.1002/sctm.20-0058] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/23/2020] [Accepted: 07/12/2020] [Indexed: 12/11/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are a promising cell source for the creation of cartilage to treat articular cartilage damage. The molecular mechanisms that translate culture conditions to the chondrogenic differentiation of hiPSCs remain to be analyzed. To analyze the effects of culture substrates, we chondrogenically differentiated hiPSCs on Matrigel or laminin 511‐E8 while holding the composition of the chondrogenic medium constant. Cartilage was formed from hiPSCs on Matrigel, but not on laminin 511‐E8. On Matrigel, the hiPSCs were round and yes‐associated protein (YAP) was inactive. In contrast, on laminin 511‐E8, the hiPSCs were flat and YAP was active. Treating the laminin 511‐E8 hiPSCs in a bioreactor caused cell aggregates, in which the cells were round and YAP was inactive. Subsequent culture of the aggregates in chondrogenic medium resulted in cartilage formation. Transient knockdown of YAP in hiPSCs around the start of chondrogenic differentiation successfully formed cartilage on laminin 511‐E8, suggesting that the activation of YAP is responsible for the failure of cartilage formation from hiPSCs on laminin 511‐E8. Consistently, the addition of YAP inhibitors to laminin 511‐E8 hiPSCs caused partial cartilage formation. This study contributes to identifying the molecules that mediate the effects of culture substrates on the chondrogenic differentiation of hiPSCs as well as to developing clinically applicable chondrogenic differentiation methods.
Collapse
Affiliation(s)
- Akihiro Yamashita
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Hiroyuki Yoshitomi
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University (CiRA), Kyoto, Japan.,Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shunsuke Kihara
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Junya Toguchida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University (CiRA), Kyoto, Japan.,Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Noriyuki Tsumaki
- Cell Induction and Regulation Field, Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
17
|
Clinical Application Status of Articular Cartilage Regeneration Techniques: Tissue-Engineered Cartilage Brings New Hope. Stem Cells Int 2020; 2020:5690252. [PMID: 32676118 PMCID: PMC7345961 DOI: 10.1155/2020/5690252] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 05/22/2020] [Accepted: 05/28/2020] [Indexed: 12/16/2022] Open
Abstract
Hyaline articular cartilage lacks blood vessels, lymphatics, and nerves and is characterised by limited self-repair ability following injury. Traditional techniques of articular cartilage repair and regeneration all have certain limitations. The development of tissue engineering technology has brought hope to the regeneration of articular cartilage. The strategies of tissue-engineered articular cartilage can be divided into three types: “cell-scaffold construct,” cell-free, and scaffold-free. In “cell-scaffold construct” strategies, seed cells can be autologous chondrocytes or stem. Among them, some commercial products with autologous chondrocytes as seed cells, such as BioSeed®-C and CaReS®, have been put on the market and some products are undergoing clinical trials, such as NOVOCART® 3D. The stem cells are mainly pluripotent stem cells and mesenchymal stem cells from different sources. Cell-free strategies that indirectly utilize the repair and regeneration potential of stem cells have also been used in clinical settings, such as TruFit and MaioRegen. Finally, the scaffold-free strategy is also a new development direction, and the short-term repair results of related products, such as NOVOCART® 3D, are encouraging. In this paper, the commonly used techniques of articular cartilage regeneration in surgery are reviewed. By studying different strategies and different seed cells, the clinical application status of tissue-engineered articular cartilage is described in detail.
Collapse
|
18
|
Marconi A, Hancock-Ronemus A, Gillis JA. Adult chondrogenesis and spontaneous cartilage repair in the skate, Leucoraja erinacea. eLife 2020; 9:e53414. [PMID: 32393435 PMCID: PMC7217701 DOI: 10.7554/elife.53414] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/21/2020] [Indexed: 12/13/2022] Open
Abstract
Mammalian articular cartilage is an avascular tissue with poor capacity for spontaneous repair. Here, we show that embryonic development of cartilage in the skate (Leucoraja erinacea) mirrors that of mammals, with developing chondrocytes co-expressing genes encoding the transcription factors Sox5, Sox6 and Sox9. However, in skate, transcriptional features of developing cartilage persist into adulthood, both in peripheral chondrocytes and in cells of the fibrous perichondrium that ensheaths the skeleton. Using pulse-chase label retention experiments and multiplexed in situ hybridization, we identify a population of cycling Sox5/6/9+ perichondral progenitor cells that generate new cartilage during adult growth, and we show that persistence of chondrogenesis in adult skates correlates with ability to spontaneously repair cartilage injuries. Skates therefore offer a unique model for adult chondrogenesis and cartilage repair and may serve as inspiration for novel cell-based therapies for skeletal pathologies, such as osteoarthritis.
Collapse
Affiliation(s)
| | - Amy Hancock-Ronemus
- Charles River LaboratoriesWilmington, MassachusettsUnited States
- Marine Biological LaboratoryWoods Hole, MassachusettsUnited States
| | - J Andrew Gillis
- Department of Zoology, University of CambridgeCambridgeUnited Kingdom
- Marine Biological LaboratoryWoods Hole, MassachusettsUnited States
| |
Collapse
|
19
|
Chijimatsu R, Saito T. Mechanisms of synovial joint and articular cartilage development. Cell Mol Life Sci 2019; 76:3939-3952. [PMID: 31201464 PMCID: PMC11105481 DOI: 10.1007/s00018-019-03191-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/30/2019] [Accepted: 06/11/2019] [Indexed: 12/29/2022]
Abstract
Articular cartilage is formed at the end of epiphyses in the synovial joint cavity and permanently contributes to the smooth movement of synovial joints. Most skeletal elements develop from transient cartilage by a biological process known as endochondral ossification. Accumulating evidence indicates that articular and growth plate cartilage are derived from different cell sources and that different molecules and signaling pathways regulate these two kinds of cartilage. As the first sign of joint development, the interzone emerges at the presumptive joint site within a pre-cartilage tissue. After that, joint cavitation occurs in the center of the interzone, and the cells in the interzone and its surroundings gradually form articular cartilage and the synovial joint. During joint development, the interzone cells continuously migrate out to the epiphyseal cartilage and the surrounding cells influx into the joint region. These complicated phenomena are regulated by various molecules and signaling pathways, including GDF5, Wnt, IHH, PTHrP, BMP, TGF-β, and FGF. Here, we summarize current literature and discuss the molecular mechanisms underlying joint formation and articular development.
Collapse
Affiliation(s)
- Ryota Chijimatsu
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Taku Saito
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
20
|
Kawata M, Mori D, Kanke K, Hojo H, Ohba S, Chung UI, Yano F, Masaki H, Otsu M, Nakauchi H, Tanaka S, Saito T. Simple and Robust Differentiation of Human Pluripotent Stem Cells toward Chondrocytes by Two Small-Molecule Compounds. Stem Cell Reports 2019; 13:530-544. [PMID: 31402337 PMCID: PMC6739881 DOI: 10.1016/j.stemcr.2019.07.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 01/08/2023] Open
Abstract
A simple induction protocol to differentiate chondrocytes from pluripotent stem cells (PSCs) using small-molecule compounds is beneficial for cartilage regenerative medicine and mechanistic studies of chondrogenesis. Here, we demonstrate that chondrocytes are robustly induced from human PSCs by simple combination of two compounds, CHIR99021, a glycogen synthase kinase 3 inhibitor, and TTNPB, a retinoic acid receptor (RAR) agonist, under serum- and feeder-free conditions within 5-9 days. An excellent differentiation efficiency and potential to form hyaline cartilaginous tissues in vivo were demonstrated. Comprehensive gene expression and open chromatin analyses at each protocol stage revealed step-by-step differentiation toward chondrocytes. Genome-wide analysis of RAR and β-catenin association with DNA showed that retinoic acid and Wnt/β-catenin signaling collaboratively regulated the key marker genes at each differentiation stage. This method provides a promising cell source for regenerative medicine and, as an in vitro model, may facilitate elucidation of the molecular mechanisms underlying chondrocyte differentiation.
Collapse
Affiliation(s)
- Manabu Kawata
- Sensory & Motor System Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Bone and Cartilage Regenerative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daisuke Mori
- Sensory & Motor System Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Bone and Cartilage Regenerative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kosuke Kanke
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hironori Hojo
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinsuke Ohba
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ung-Il Chung
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Fumiko Yano
- Bone and Cartilage Regenerative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideki Masaki
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Makoto Otsu
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sakae Tanaka
- Sensory & Motor System Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taku Saito
- Sensory & Motor System Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Bone and Cartilage Regenerative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
21
|
Gorecka J, Kostiuk V, Fereydooni A, Gonzalez L, Luo J, Dash B, Isaji T, Ono S, Liu S, Lee SR, Xu J, Liu J, Taniguchi R, Yastula B, Hsia HC, Qyang Y, Dardik A. The potential and limitations of induced pluripotent stem cells to achieve wound healing. Stem Cell Res Ther 2019; 10:87. [PMID: 30867069 PMCID: PMC6416973 DOI: 10.1186/s13287-019-1185-1] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Wound healing is the physiologic response to a disruption in normal skin architecture and requires both spatial and temporal coordination of multiple cell types and cytokines. This complex process is prone to dysregulation secondary to local and systemic factors such as ischemia and diabetes that frequently lead to chronic wounds. Chronic wounds such as diabetic foot ulcers are epidemic with great cost to the healthcare system as they heal poorly and recur frequently, creating an urgent need for new and advanced therapies. Stem cell therapy is emerging as a potential treatment for chronic wounds, and adult-derived stem cells are currently employed in several commercially available products; however, stem cell therapy is limited by the need for invasive harvesting techniques, immunogenicity, and limited cell survival in vivo. Induced pluripotent stem cells (iPSC) are an exciting cell type with enhanced therapeutic and translational potential. iPSC are derived from adult cells by in vitro induction of pluripotency, obviating the ethical dilemmas surrounding the use of embryonic stem cells; they are harvested non-invasively and can be transplanted autologously, reducing immune rejection; and iPSC are the only cell type capable of being differentiated into all of the cell types in healthy skin. This review focuses on the use of iPSC in animal models of wound healing including limb ischemia, as well as their limitations and methods aimed at improving iPSC safety profile in an effort to hasten translation to human studies.
Collapse
Affiliation(s)
- Jolanta Gorecka
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Valentyna Kostiuk
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Arash Fereydooni
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Jiesi Luo
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA
| | - Biraja Dash
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, PO Box 208062, New Haven, CT, 06520-8062, USA
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Shun Ono
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Shirley Liu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Shin Rong Lee
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Jianbiao Xu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Jia Liu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Bogdan Yastula
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Henry C Hsia
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, PO Box 208062, New Haven, CT, 06520-8062, USA
| | - Yibing Qyang
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA.,Yale Stem Cell Center, Yale University, New Haven, USA.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, USA.,Department of Pathology, Yale University, New Haven, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA.
| |
Collapse
|
22
|
Xiao S, Liu W, Bi J, Liu S, Zhao H, Gong N, Xing D, Gao H, Gong M. Anti-inflammatory effect of hesperidin enhances chondrogenesis of human mesenchymal stem cells for cartilage tissue repair. JOURNAL OF INFLAMMATION-LONDON 2018; 15:14. [PMID: 30038551 PMCID: PMC6053785 DOI: 10.1186/s12950-018-0190-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 07/13/2018] [Indexed: 01/01/2023]
Abstract
Background Articular cartilage diseases are considered a major health problem, and tissue engineering using human mesenchymal stem cells (MSCs) have been shown as a promising solution for cartilage tissue repair. Hesperidin is a flavonoid extract from citrus fruits with anti-inflammatory properties. We aimed to investigate the effect of hesperidin on MSCs for cartilage tissue repair. MSCs were treated by hesperidin, and colony formation and proliferation assays were performed to evaluate self-renewal ability of MSCs. Alcian blue staining and Sox9 expression were measured to evaluate chondrogenesis of MSCs. Secretion of pro-inflammatory cytokines IFN-γ, IL-2, IL-4 and IL-10, and expression of nuclear factor kappa B (NF-κB) subunit p65 were also assessed. Results Hesperidin improved self-renewal ability and chondrogenesis of MSCs, inhibited secretion of pro-inflammatory cytokines IFN-γ, IL-2, IL-4 and IL-10, and suppressed the expression of p65. Overexpression of p65 was able to reverse the hesperidin inhibited secretions of pro-inflammatory cytokines, and abolish the enhancing effect of hesperidin on chondrogenesis of MSCs. Conclusion Hesperidin could serve as a therapeutic agent to effectively enhance chondrogenesis of human MSCs by inhibiting inflammation to facilitate cartilage tissue repair.
Collapse
Affiliation(s)
- Shipeng Xiao
- 1Department of Joint Surgery, The Second Hospital of Shandong University, Jinan, 250033 Shandong China
| | - Wenguang Liu
- 1Department of Joint Surgery, The Second Hospital of Shandong University, Jinan, 250033 Shandong China
| | - Jianqiang Bi
- 2Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials, Ministry of Education, Shandong University, Jinan, 250061 Shandong China
| | - Shenghou Liu
- 1Department of Joint Surgery, The Second Hospital of Shandong University, Jinan, 250033 Shandong China
| | - Heng Zhao
- 1Department of Joint Surgery, The Second Hospital of Shandong University, Jinan, 250033 Shandong China
| | - Ningji Gong
- 3Department of emergency, The Second Hospital of Shandong University, Jinan, 250033 Shandong China
| | - Deguo Xing
- 4Department of Trauma and Orthopedics, The Second Hospital of Shandong University, Jinan, 250033 Shandong China
| | - Hongwei Gao
- 4Department of Trauma and Orthopedics, The Second Hospital of Shandong University, Jinan, 250033 Shandong China
| | - Mingzhi Gong
- 4Department of Trauma and Orthopedics, The Second Hospital of Shandong University, Jinan, 250033 Shandong China
| |
Collapse
|
23
|
Combating Osteoarthritis through Stem Cell Therapies by Rejuvenating Cartilage: A Review. Stem Cells Int 2018; 2018:5421019. [PMID: 29765416 PMCID: PMC5885495 DOI: 10.1155/2018/5421019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/05/2018] [Indexed: 12/13/2022] Open
Abstract
Knee osteoarthritis (OA) is a chronic degenerative disorder which could be distinguished by erosion of articular cartilage, pain, stiffness, and crepitus. Not only aging-associated alterations but also the metabolic factors such as hyperglycemia, dyslipidemia, and obesity affect articular tissues and may initiate or exacerbate the OA. The poor self-healing ability of articular cartilage due to limited regeneration in chondrocytes further adversely affects the osteoarthritic microenvironment. Traditional and current surgical treatment procedures for OA are limited and incapable to reverse the damage of articular cartilage. To overcome these limitations, cell-based therapies are currently being employed to repair and regenerate the structure and function of articular tissues. These therapies not only depend upon source and type of stem cells but also on environmental conditions, growth factors, and chemical and mechanical stimuli. Recently, the pluripotent and various multipotent mesenchymal stem cells have been employed for OA therapy, due to their differentiation potential towards chondrogenic lineage. Additionally, the stem cells have also been supplemented with growth factors to achieve higher healing response in osteoarthritic cartilage. In this review, we summarized the current status of stem cell therapies in OA pathophysiology and also highlighted the potential areas of further research needed in regenerative medicine.
Collapse
|
24
|
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: 9.0] [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
|
25
|
Kalamegam G, Memic A, Budd E, Abbas M, Mobasheri A. A Comprehensive Review of Stem Cells for Cartilage Regeneration in Osteoarthritis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1089:23-36. [PMID: 29725971 DOI: 10.1007/5584_2018_205] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Osteoarthritis (OA) is an age related joint disease associated with degeneration and loss of articular cartilage. Consequently, OA patients suffer from chronic joint pain and disability. Weight bearing joints and joints that undergo repetitive stress and excessive 'wear and tear' are particularly prone to developing OA. Cartilage has a poor regenerative capacity and current pharmacological agents only provide symptomatic pain relief. OA patients that respond poorly to conventional therapies are ultimately treated with surgical procedures to promote cartilage repair by implantation of artificial joint structures (arthroplasty) or total joint replacement (TJR). In the last two decades, stem cells derived from various tissues with varying differentiation and tissue regeneration potential have been used for the treatment of OA either alone or in combination with natural or synthetic scaffolds to aid cartilage repair. Although stem cells can be differentiated into chondrocytes in vitro or aid cartilage regeneration in vivo, their potential for OA management remains limited as cartilage regenerated by stem cells fails to fully recapitulate the structural and biomechanical properties of the native tissue. Efficient tissue regeneration remains elusive despite the simple design of cartilage, which unlike most other tissues is avascular and aneural, consisting of a single cell type. In this article, we have comprehensively reviewed the types of stem cells that have been proposed or tested for the management of OA, their potential efficacy as well as their limitations. We also touch on the role of biomaterials in cartilage tissue engineering and examine the prospects for their use in cell-based therapies.
Collapse
Affiliation(s)
- Gauthaman Kalamegam
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Sheikh Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis with Stem Cells, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adnan Memic
- Center of Nanotechnology, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Emma Budd
- Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Mohammed Abbas
- Sheikh Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis with Stem Cells, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Orthopaedic Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ali Mobasheri
- Sheikh Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis with Stem Cells, King Abdulaziz University, Jeddah, Saudi Arabia. .,Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK. .,Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Queen's Medical Centre, Nottingham, UK. .,Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
| |
Collapse
|
26
|
Suchorska WM, Augustyniak E, Richter M, Trzeciak T. Comparison of Four Protocols to Generate Chondrocyte-Like Cells from Human Induced Pluripotent Stem Cells (hiPSCs). Stem Cell Rev Rep 2017; 13:299-308. [PMID: 27987073 PMCID: PMC5380716 DOI: 10.1007/s12015-016-9708-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stem cells (SCs) are a promising approach to regenerative medicine, with the potential to treat numerous orthopedic disorders, including osteo-degenerative diseases. The development of human-induced pluripotent stem cells (hiPSCs) has increased the potential of SCs for new treatments. However, current methods of differentiating hiPSCs into chondrocyte-like cells are suboptimal and better methods are needed. The aim of the present study was to assess four different chondrogenic differentiation protocols to identify the most efficient method of generating hiPSC-derived chondrocytes. For this study, hiPSCs were obtained from primary human dermal fibroblasts (PHDFs) and differentiated into chondrocyte-like cells using four different protocols: 1) monolayer culture with defined growth factors (GF); 2) embryoid bodies (EBs) in a chondrogenic medium with TGF-β3 cells; 3) EBs in chondrogenic medium conditioned with human chondrocytes (HC-402-05a cell line) and 4) EBs in chondrogenic medium conditioned with human chondrocytes and supplemented with TGF-β3. The cells obtained through these four protocols were evaluated and compared at the mRNA and protein levels. Although chondrogenic differentiation of hiPSCs was successfully achieved with all of these protocols, the two fastest and most cost-effective methods were the monolayer culture with GFs and the medium conditioned with human chondrocytes. Both of these methods are superior to other available techniques. The main advantage of the conditioned medium is that the technique is relatively simple and inexpensive while the directed method (i.e., monolayer culture with GFs) is faster than any protocol described to date because it is does not require additional steps such as EB formation.
Collapse
Affiliation(s)
- Wiktoria Maria Suchorska
- Radiobiology Lab, Greater Poland Cancer Centre, 61- 866, Poznan, Poland
- Department of Electroradiology, Poznan University of Medical Sciences, 61-866, Poznan, Poland
| | - Ewelina Augustyniak
- Radiobiology Lab, Greater Poland Cancer Centre, 61- 866, Poznan, Poland.
- The Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091, Warsaw, Poland.
| | - Magdalena Richter
- Department of Orthopedics and Traumatology, Poznan University of Medical Sciences, 61-545, Poznan, Poland
| | - Tomasz Trzeciak
- Department of Orthopedics and Traumatology, Poznan University of Medical Sciences, 61-545, Poznan, Poland
| |
Collapse
|
27
|
Choi JR, Yong KW, Choi JY. Effects of mechanical loading on human mesenchymal stem cells for cartilage tissue engineering. J Cell Physiol 2017; 233:1913-1928. [PMID: 28542924 DOI: 10.1002/jcp.26018] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/18/2017] [Indexed: 12/22/2022]
Abstract
Today, articular cartilage damage is a major health problem, affecting people of all ages. The existing conventional articular cartilage repair techniques, such as autologous chondrocyte implantation (ACI), microfracture, and mosaicplasty, have many shortcomings which negatively affect their clinical outcomes. Therefore, it is essential to develop an alternative and efficient articular repair technique that can address those shortcomings. Cartilage tissue engineering, which aims to create a tissue-engineered cartilage derived from human mesenchymal stem cells (MSCs), shows great promise for improving articular cartilage defect therapy. However, the use of tissue-engineered cartilage for the clinical therapy of articular cartilage defect still remains challenging. Despite the importance of mechanical loading to create a functional cartilage has been well demonstrated, the specific type of mechanical loading and its optimal loading regime is still under investigation. This review summarizes the most recent advances in the effects of mechanical loading on human MSCs. First, the existing conventional articular repair techniques and their shortcomings are highlighted. The important parameters for the evaluation of the tissue-engineered cartilage, including chondrogenic and hypertrophic differentiation of human MSCs are briefly discussed. The influence of mechanical loading on human MSCs is subsequently reviewed and the possible mechanotransduction signaling is highlighted. The development of non-hypertrophic chondrogenesis in response to the changing mechanical microenvironment will aid in the establishment of a tissue-engineered cartilage for efficient articular cartilage repair.
Collapse
Affiliation(s)
- Jane Ru Choi
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia.,UBC Engineering Lab, University of British Columbia, Vancouver, Canada
| | - Kar Wey Yong
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia.,Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Canada
| | - Jean Yu Choi
- Faculty of Medicine, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
28
|
Characterization of Mesenchymal Stem Cell-Like Cells Derived From Human iPSCs via Neural Crest Development and Their Application for Osteochondral Repair. Stem Cells Int 2017; 2017:1960965. [PMID: 28607560 PMCID: PMC5451770 DOI: 10.1155/2017/1960965] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/03/2017] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) derived from induced pluripotent stem cells (iPSCs) are a promising cell source for the repair of skeletal disorders. Recently, neural crest cells (NCCs) were reported to be effective for inducing mesenchymal progenitors, which have potential to differentiate into osteochondral lineages. Our aim was to investigate the feasibility of MSC-like cells originated from iPSCs via NCCs for osteochondral repair. Initially, MSC-like cells derived from iPSC-NCCs (iNCCs) were generated and characterized in vitro. These iNCC-derived MSC-like cells (iNCMSCs) exhibited a homogenous population and potential for osteochondral differentiation. No upregulation of pluripotent markers was detected during culture. Second, we implanted iNCMSC-derived tissue-engineered constructs into rat osteochondral defects without any preinduction for specific differentiation lineages. The implanted cells remained alive at the implanted site, whereas they failed to repair the defects, with only scarce development of osteochondral tissue in vivo. With regard to tumorigenesis, the implanted cells gradually disappeared and no malignant cells were detected throughout the 2-month follow-up. While this study did not show that iNCMSCs have efficacy for repair of osteochondral defects when implanted under undifferentiated conditions, iNCMSCs exhibited good chondrogenic potential in vitro under appropriate conditions. With further optimization, iNCMSCs may be a new source for tissue engineering of cartilage.
Collapse
|
29
|
Augustyniak E, Suchorska WM, Trzeciak T, Richter M. Gene expression profile in human induced pluripotent stem cells: Chondrogenic differentiation in vitro, part B. Mol Med Rep 2017; 15:2402-2414. [PMID: 28447733 PMCID: PMC5428858 DOI: 10.3892/mmr.2017.6335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 01/26/2017] [Indexed: 01/15/2023] Open
Abstract
The development of human induced pluripotent stem cells (hiPSCs) is considered a turning point in tissue engineering. However, more data are required to improve understanding of key aspects of the cell differentiation process, including how specific chondrogenic processes affect the gene expression profile of chondrocyte‑like cells and the relative value of cell differentiation markers. The main aims of the present study were as follows: To determine the gene expression profile of chondrogenic‑like cells derived from hiPSCs cultured in mediums conditioned with HC‑402‑05a cells or supplemented with transforming growth factor β3 (TGF‑β3), and to assess the relative utility of the most commonly‑used chondrogenic markers as indicators of cell differentiation. These issues are relevant with regard to the use of human fibroblasts in the reprogramming process to obtain hiPSCs. Human fibroblasts are derived from mesoderm and thus share a wide range of properties with chondrocytes, which originate from the mesenchyme. The hiPSCs were obtained from human primary dermal fibroblasts during a reprogramming process. Two methods, both involving embryoid bodies (EB), were used to obtain chondrocytes from the hiPSCs: EBs formed in the presence of a chondrogenic medium with TGF‑β3 (10 ng/ml) and EBs formed in a medium conditioned with growth factors from HC‑402‑05a cells. Based on reverse transcription-quantitative polymerase chain reaction analysis, the results demonstrated that hiPSCs are capable of effective chondrogenic differentiation, with the cells obtained in the HC‑402‑05a medium presenting with morphological features and markers characteristic of mature human chondrocytes. In contrast, cells differentiated in the presence of TGF‑β3 presented with certain undesirable hypertrophic characteristics. Several genes, most notably runt‑related transcription factor 2, transforming growth factor β2 and transforming growth factor β3, were good markers of advanced and late hiPSC chondrogenic differentiation, whereas transforming growth factor β3I, II, III receptors and bone morphogenetic protein-2, bone morphogenetic protein-4 and growth differentiation factor 5 were less valuable. These findings provide valuable data on the use of stem cells in cartilage tissue regeneration.
Collapse
Affiliation(s)
- Ewelina Augustyniak
- Radiobiology Laboratory, Greater Poland Cancer Centre, 61‑866 Poznan, Poland
| | | | - Tomasz Trzeciak
- Department of Orthopedics and Traumatology, Poznan University of Medical Sciences, 61‑545 Poznan, Poland
| | - Magdalena Richter
- Department of Orthopedics and Traumatology, Poznan University of Medical Sciences, 61‑545 Poznan, Poland
| |
Collapse
|
30
|
Gibson JD, O'Sullivan MB, Alaee F, Paglia DN, Yoshida R, Guzzo RM, Drissi H. Regeneration of Articular Cartilage by Human ESC-Derived Mesenchymal Progenitors Treated Sequentially with BMP-2 and Wnt5a. Stem Cells Transl Med 2016; 6:40-50. [PMID: 28170184 PMCID: PMC5442752 DOI: 10.5966/sctm.2016-0020] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/16/2016] [Indexed: 01/12/2023] Open
Abstract
The success of cell‐based therapies to restore joint cartilage requires an optimal source of reparative progenitor cells and tight control of their differentiation into a permanent cartilage phenotype. Bone morphogenetic protein 2 (BMP‐2) has been extensively shown to promote mesenchymal cell differentiation into chondrocytes in vitro and in vivo. Conversely, developmental studies have demonstrated decreased chondrocyte maturation by Wingless‐Type MMTV Integration Site Family, Member 5A (Wnt5a). Thus, we hypothesized that treatment of human embryonic stem cell (hESC)‐derived chondroprogenitors with BMP‐2 followed by Wnt5a may control the maturational progression of these cells into a hyaline‐like chondrocyte phenotype. We examined the effects of sustained exposure of hESC‐derived mesenchymal‐like progenitors to recombinant Wnt5a or BMP‐2 in vitro. Our data indicate that BMP‐2 promoted a strong chondrogenic response leading to terminal maturation, whereas recombinant Wnt5a induced a mild chondrogenic response without promoting hypertrophy. Moreover, Wnt5a suppressed BMP‐2‐mediated chondrocyte maturation, preventing the formation of fibrocartilaginous tissue in high‐density cultures treated sequentially with BMP‐2 and Wnt5a. Implantation of scaffoldless pellets of hESC‐derived chondroprogenitors pretreated with BMP‐2 followed by Wnt5a into rat chondral defects induced an articular‐like phenotype in vivo. Together, the data establish a novel role for Wnt5a in controlling the progression from multipotency into an articular‐like cartilage phenotype in vitro and in vivo. Stem Cells Translational Medicine2017;6:40–50
Collapse
Affiliation(s)
- Jason D. Gibson
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Stem Cell Institute, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| | - Michael B. O'Sullivan
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Stem Cell Institute, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| | - Farhang Alaee
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Stem Cell Institute, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| | - David N. Paglia
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Stem Cell Institute, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| | - Ryu Yoshida
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Stem Cell Institute, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| | - Rosa M. Guzzo
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Stem Cell Institute, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| | - Hicham Drissi
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Stem Cell Institute, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| |
Collapse
|
31
|
Human Pluripotent Stem Cells: Advances in Chondrogenic Differentiation and Articular Cartilage Regeneration. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s40610-016-0041-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
32
|
Massie I, Dietrich J, Roth M, Geerling G, Mertsch S, Schrader S. Development of Causative Treatment Strategies for Lacrimal Gland Insufficiency by Tissue Engineering and Cell Therapy. Part 2: Reconstruction of Lacrimal Gland Tissue: What Has Been Achieved So Far and What Are the Remaining Challenges? Curr Eye Res 2016; 41:1255-1265. [DOI: 10.3109/02713683.2016.1151531] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Isobel Massie
- Labor für Experimentelle Ophthalmologie, University of Düsseldorf, Düsseldorf, Germany
| | - Jana Dietrich
- Labor für Experimentelle Ophthalmologie, University of Düsseldorf, Düsseldorf, Germany
| | - Mathias Roth
- Labor für Experimentelle Ophthalmologie, University of Düsseldorf, Düsseldorf, Germany
| | - Gerd Geerling
- Augenklinik, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Sonja Mertsch
- Labor für Experimentelle Ophthalmologie, University of Düsseldorf, Düsseldorf, Germany
| | - Stefan Schrader
- Labor für Experimentelle Ophthalmologie, University of Düsseldorf, Düsseldorf, Germany
- Augenklinik, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
33
|
Bernhard JC, Vunjak-Novakovic G. Should we use cells, biomaterials, or tissue engineering for cartilage regeneration? Stem Cell Res Ther 2016; 7:56. [PMID: 27089917 PMCID: PMC4836146 DOI: 10.1186/s13287-016-0314-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
For a long time, cartilage has been a major focus of the whole field of tissue engineering, both because of the constantly growing need for more effective options for joint repair and the expectation that this apparently simple tissue will be easy to engineer. After several decades, cartilage regeneration has proven to be anything but easy. With gratifying progress in our understanding of the factors governing cartilage development and function, and cell therapy being successfully used for several decades, there is still a lot to do. We lack reliable methods to generate durable articular cartilage that would resemble the original tissue lost to injury or disease. The question posed here is whether the answer would come from the methods using cells, biomaterials, or tissue engineering. We present a concise review of some of the most meritorious efforts in each area, and propose that the solution will most likely emerge from the ongoing attempts to recapitulate certain aspects of native cartilage development. While an ideal recipe for cartilage regeneration is yet to be formulated, we believe that it will contain cell, biomaterial, and tissue engineering approaches, blended into an effective method for seamless repair of articular cartilage.
Collapse
Affiliation(s)
- Jonathan C Bernhard
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY, 10032, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY, 10032, USA. .,Department of Medicine, Columbia University, 622 West 168th Street, VC12-234, New York, NY, 10032, USA.
| |
Collapse
|
34
|
Diederichs S, Gabler J, Autenrieth J, Kynast KL, Merle C, Walles H, Utikal J, Richter W. Differential Regulation of SOX9 Protein During Chondrogenesis of Induced Pluripotent Stem Cells Versus Mesenchymal Stromal Cells: A Shortcoming for Cartilage Formation. Stem Cells Dev 2016; 25:598-609. [DOI: 10.1089/scd.2015.0312] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Solvig Diederichs
- Research Center for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica Gabler
- Research Center for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| | - Jennifer Autenrieth
- Research Center for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| | - Katharina L. Kynast
- Research Center for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| | - Christian Merle
- Clinic for Orthopedics and Trauma Surgery, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| | - Heike Walles
- Department Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, Translational Center Würzburg ‘Regenerative Therapies in Oncology and Musculoskeletal Diseases’–Würzburg Branch, Würzburg, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology, and Allergology, University Medical Center Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopedics, Orthopedic University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|