1
|
Wang B, Lu Z, Gao G, Mikaeiliagah E, Wang L, Yu Q, Wang Z, Hu G, Chen S, Zhang X, Pei M. Distinct role of perlecan in mesenchymal tissue regeneration via genetic and epigenetic modification. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2025; 508:161103. [PMID: 40124846 PMCID: PMC11928145 DOI: 10.1016/j.cej.2025.161103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Perlecan (HSPG2), a key component of basement membrane proteins, plays a crucial role in tissue development and regeneration. However, its direct impact on mesenchymal tissue differentiation, mediated through both genetic modification (gain- and loss-of-function mutations) and epigenetic changes (matrix microenvironment alterations), remains underexplored. In this study, we utilized CRISPR/Cas9 to achieve knockout (KO) and overexpression (OE) of HSPG2 in human fetal nucleus pulposus stem/progenitor cells (NPSCs) and adult infrapatellar fat pad-derived stem cells (IPFSCs) to investigate perlecan's influence on mesenchymal differentiation. We also assessed the effects of decellularized extracellular matrix (dECM) derived from fetal NPSCs with modified HSPG2 expression on the proliferation and differentiation of adult NPSCs. Our findings demonstrate that HSPG2-KO enhance chondrogenic differentiation, while HSPG2-OE suppressed adipogenic differentiation in both fetal NPSCs and adult IPFSCs. Notably, dECM from HSPG2-OE fetal NPSCs significantly promoted chondrogenic differentiation in adult NPSCs, suggesting potential applications for perlecan in developing advanced biomaterials for cartilage regeneration.
Collapse
Affiliation(s)
- Bin Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Orthopaedics, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, China
| | - Zhihua Lu
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Medical School, Yangzhou Polytechnic College, Yangzhou, Jiangsu 225009, China
| | - Gongming Gao
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Orthopaedics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Elmira Mikaeiliagah
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Lei Wang
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
| | - Qingqing Yu
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Zhuo Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Orthopaedics, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, China
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Bioinformatics Core, West Virginia University, Morgantown, WV, USA
| | - Song Chen
- Department of Orthopaedics, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xiaobing Zhang
- Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Peking Union Medical College, Tianjin, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
2
|
Liu X, Guo X, Pei YA, Pei M, Ge Z. Charting a quarter-century of commercial cartilage regeneration products. J Orthop Translat 2025; 50:354-363. [PMID: 39968336 PMCID: PMC11833628 DOI: 10.1016/j.jot.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/04/2024] [Accepted: 10/30/2024] [Indexed: 02/20/2025] Open
Abstract
Functional cartilage regeneration remains difficult to achieve despite decades of research. Dozens of commercial products have been proposed, with each targeting different facets of successful cartilage engineering, including mechanical properties, integration, lubrication and inflammation; however, there remains a lack of breakthroughs in meaningful clinical outcomes. Prior research categorized commercial products based on their components and elucidated challenges faced during the market approval process. This paper, for the first time, comprehensively reviews the properties of commercial products covering the last 25 years, including design trends in components, compatibility with minimally invasive surgery, indications for cartilage defects, long-term follow-up, as well as active sponsorship support of the International Cartilage Regeneration and Joint Preservation Society (ICRS). We aim to summarize the key factors for potentially successful commercial products and elucidate overarching trends in technology development in this field. Given that no revolutionary products have yielded significantly improved clinical results, emerging products compete with one another on user-friendliness and cost-efficiency. Other relevant characteristics include compatibility with minimally invasive surgery, extensiveness of required surgery (one-stage vs. two-stage), use of versatile artificial polymers and application of cells and biomaterials. Specific products continue to lead the market due to their cost-efficiency or indications for larger cartilage defects. However, they have been shown to result in no significant improvement upon clinical follow-up. Thus, there is a need for products that surpass current commercial products and show clinical effectiveness. Translation potential of this article: This review analyzes product components, compatibility with minimally invasive surgery, indication for cartilage defect areas, clinical performance as well as sponsorship for the World Conference of International Cartilage Regeneration & Joint Preservation Society, based on information about cartilage regeneration products from 1997 to 2023. It shines a light on future development of design and commercialization of cartilage products.
Collapse
Affiliation(s)
- Xinyi Liu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
| | - Xiaolei Guo
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, China
| | - Yixuan Amy Pei
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA, 19104
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Zigang Ge
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
| |
Collapse
|
3
|
Górska A, Trubalski M, Borowski B, Brachet A, Szymańczyk S, Markiewicz R. Navigating stem cell culture: insights, techniques, challenges, and prospects. Front Cell Dev Biol 2024; 12:1435461. [PMID: 39588275 PMCID: PMC11586186 DOI: 10.3389/fcell.2024.1435461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/09/2024] [Indexed: 11/27/2024] Open
Abstract
Stem cell research holds huge promise for regenerative medicine and disease modeling, making the understanding and optimization of stem cell culture a critical aspect of advancing these therapeutic applications. This comprehensive review provides an in-depth overview of stem cell culture, including general information, contemporary techniques, encountered problems, and future perspectives. The article begins by explaining the fundamental characteristics of various stem cell types, elucidating the importance of proper culture conditions in maintaining pluripotency or lineage commitment. A detailed exploration of established culture techniques sheds light on the evolving landscape of stem cell culture methodologies. Common challenges such as genetic stability, heterogeneity, and differentiation efficiency are thoroughly discussed, with insights into cutting-edge strategies and technologies aimed at addressing these hurdles. Moreover, the article delves into the impact of substrate materials, culture media components, and biophysical cues on stem cell behavior, emphasizing the intricate interplay between the microenvironment and cell fate decisions. As stem cell research advances, ethical considerations and regulatory frameworks become increasingly important, prompting a critical examination of these aspects in the context of culture practices. Lastly, the article explores emerging perspectives, including the integration of artificial intelligence and machine learning in optimizing culture conditions, and the potential applications of stem cell-derived products in personalized medicine. This comprehensive overview aims to serve as a valuable resource for researchers and clinicians, fostering a deeper understanding of stem cell culture and its key role in advancing regenerative medicine and biomedical research.
Collapse
Affiliation(s)
- Aleksandra Górska
- Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, Lublin, Poland
| | - Mateusz Trubalski
- Students Scientific Association, Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, Lublin, Poland
| | - Bartosz Borowski
- Students Scientific Association, Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, Lublin, Poland
| | - Adam Brachet
- Student Scientific Association, Department of Forensic Medicine, Medical University of Lublin, Lublin, Poland
| | - Sylwia Szymańczyk
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Renata Markiewicz
- Occupational Therapy Laboratory, Chair of Nursing Development, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
4
|
Laiva AL, O'Brien FJ, Keogh MB. Dual delivery gene-activated scaffold directs fibroblast activity and keratinocyte epithelization. APL Bioeng 2024; 8:016104. [PMID: 38283135 PMCID: PMC10821797 DOI: 10.1063/5.0174122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/12/2024] [Indexed: 01/30/2024] Open
Abstract
Fibroblasts are the most abundant cell type in dermal skin and keratinocytes are the most abundant cell type in the epidermis; both play a crucial role in wound remodeling and maturation. We aim to assess the functionality of a novel dual gene activated scaffold (GAS) on human adult dermal fibroblasts (hDFs) and see how the secretome produced could affect human dermal microvascular endothelial cells (HDMVECs) and human epidermal keratinocyte (hEKs) growth and epithelization. Our GAS is a collagen chondroitin sulfate scaffold loaded with pro-angiogenic stromal derived factor (SDF-1α) and/or an anti-aging β-Klotho plasmids. hDFs were grown on GAS for two weeks and compared to gene-free scaffolds. GAS produced a significantly better healing outcome in the fibroblasts than in the gene-free scaffold group. Among the GAS groups, the dual GAS induced the most potent pro-regenerative maturation in fibroblasts with a downregulation in proliferation (twofold, p < 0.05), fibrotic remodeling regulators TGF-β1 (1.43-fold, p < 0.01) and CTGF (1.4-fold, p < 0.05), fibrotic cellular protein α-SMA (twofold, p < 0.05), and fibronectin matrix deposition (twofold, p < 0.05). The dual GAS secretome also showed enhancements of paracrine keratinocyte pro-epithelializing ability (1.3-fold, p < 0.05); basement membrane regeneration through laminin (6.4-fold, p < 0.005) and collagen IV (8.7-fold, p < 0.005) deposition. Our findings demonstrate enhanced responses in dual GAS containing hDFs by proangiogenic SDF-1α and β-Klotho anti-fibrotic rejuvenating activities. This was demonstrated by activating hDFs on dual GAS to become anti-fibrotic in nature while eliciting wound repair basement membrane proteins; enhancing a proangiogenic HDMVECs paracrine signaling and greater epithelisation of hEKs.
Collapse
Affiliation(s)
| | | | - Michael B. Keogh
- Author to whom correspondence should be addressed:. Tel.: +973 17351450
| |
Collapse
|
5
|
Pei YA, Mikaeiliagah E, Wang B, Zhang X, Pei M. The matrix microenvironment influences but does not dominate tissue-specific stem cell lineage differentiation. Mater Today Bio 2023; 23:100805. [PMID: 37766896 PMCID: PMC10519827 DOI: 10.1016/j.mtbio.2023.100805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/25/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Mesenchymal stem cells (MSCs) play a pivotal role in tissue engineering and regenerative medicine, with their clinical application often hindered by cell senescence during ex vivo expansion. Recent studies suggest that MSC-deposited decellularized extracellular matrix (dECM) offers a conducive microenvironment that fosters cell proliferation and accentuates stem cell differentiation. However, the ability of this matrix environment to govern lineage differentiation of tissue-specific stem cells remains ambiguous. This research employs human adipose-derived MSCs (ADSCs) and synovium-derived MSCs (SDSCs) as models for adipogenesis and chondrogenesis differentiation pathways, respectively. Genetically modified dECM (GMdECM), produced by SV40LT-transduced immortalized cells, was studied for its influence on cell differentiation. Both types of immortalized cells displayed a reduction in chondrogenic ability but an enhancement in adipogenic potential. ADSCs grown on ADSC-deposited dECM showed stable chondrogenic potential but increased adipogenic capacity; conversely, SDSCs expanded on SDSC-generated dECM displayed elevated chondrogenic capacity and diminished adipogenic potential. This cell-dependent response was confirmed through GMdECM expansion, with SDSCs showing enhanced chondrogenesis. However, ADSCs did not exhibit improved chondrogenic potential on GMdECM, suggesting that the matrix microenvironment does not dictate the final differentiation path of tissue-specific stem cells. Potential molecular mechanisms, such as elevated basement membrane protein expression in GMdECMs and dynamic TWIST1 expression during expansion and chondrogenic induction, may underpin the strong chondrogenic differentiation of GMdECM-expanded SDSCs.
Collapse
Affiliation(s)
- Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elmira Mikaeiliagah
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Biology, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| | - Bin Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Foot and Hand Surgery, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu, China
| | - Xiaobing Zhang
- Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Peking Union Medical College, Tianjin, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
6
|
Matveeva D, Buravkov S, Andreeva E, Buravkova L. Hypoxic Extracellular Matrix Preserves Its Competence after Expansion of Human MSCs under Physiological Hypoxia In Vitro. Biomimetics (Basel) 2023; 8:476. [PMID: 37887607 PMCID: PMC10604705 DOI: 10.3390/biomimetics8060476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/14/2023] [Accepted: 09/30/2023] [Indexed: 10/28/2023] Open
Abstract
Tissue-relevant O2 levels are considered as an important tool for the preconditioning of multipotent mesenchymal stromal cells (MSCs) for regenerative medicine needs. The present study investigated the quality and functions of the extracellular matrix (ECM) of MSCs under low O2 levels. Human adipose tissue-derived MSCs were continuously expanded under normoxia (20% O2, N) or "physiological" hypoxia (5% O2, Hyp). Decellularized ECM (dcECM) was prepared. The structure of the dcECM was analyzed using confocal laser and scanning electron microscopy. Collagen, dcECM-N, and dcECM-Hyp were recellularized with MSC-N and further cultured at normoxia. The efficacy of adhesion, spreading, growth, osteogenic potential, and paracrine activity of recellularized MSC-N were evaluated. At low O2, the dcECM showed an increased alignment of fibrillar structures and provided accelerated spreading of MSC-N, indicating increased dcECM-Hyp stiffness. We described O2-dependent "ECM-education" of MSC-N when cultured on dcECM-Hyp. This was manifested as attenuated spontaneous osteo-commitment, increased susceptibility to osteo-induction, and a shift in the paracrine profile. It has been suggested that the ECM after physiological hypoxia is able to ensure the maintenance of a low-commitment state of MSCs. DcECM, which preserves the competence of the natural microenvironment of cells and is capable of "educating" others, appears to be a prospective tool for guiding cell modifications for cell therapy and tissue engineering.
Collapse
Affiliation(s)
| | | | - Elena Andreeva
- Institute of Biomedical Problems of Russian Academy of Sciences, Moscow 123007, Russia; (D.M.); (S.B.); (L.B.)
| | | |
Collapse
|
7
|
Xu X, Xu L, Xia J, Wen C, Liang Y, Zhang Y. Harnessing knee joint resident mesenchymal stem cells in cartilage tissue engineering. Acta Biomater 2023; 168:372-387. [PMID: 37481194 DOI: 10.1016/j.actbio.2023.07.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/26/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
Osteoarthritis (OA) is a widespread clinical disease characterized by cartilage degeneration in middle-aged and elderly people. Currently, there is no effective treatment for OA apart from total joint replacement in advanced stages. Mesenchymal stem cells (MSCs) are a type of adult stem cell with diverse differentiation capabilities and immunomodulatory potentials. MSCs are known to effectively regulate the cartilage microenvironment, promote cartilage regeneration, and alleviate OA symptoms. As a result, they are promising sources of cells for OA therapy. Recent studies have revealed the presence of resident MSCs in synovial fluid, synovial membrane, and articular cartilage, which can be collected as knee joint-derived MSCs (KJD-MSC). Several preclinical and clinical studies have demonstrated that KJD-MSCs have great potential for OA treatment, whether applied alone, in combination with biomaterials, or as exocrine MSCs. In this article, we will review the characteristics of MSCs in the joints, including their cytological characteristics, such as proliferation, cartilage differentiation, and immunomodulatory abilities, as well as the biological function of MSC exosomes. We will also discuss the use of tissue engineering in OA treatment and introduce the concept of a new generation of stem cell-based tissue engineering therapy, including the use of engineering, gene therapy, and gene editing techniques to create KJD-MSCs or KJD-MSC derivative exosomes with improved functionality and targeted delivery. These advances aim to maximize the efficiency of cartilage tissue engineering and provide new strategies to overcome the bottleneck of OA therapy. STATEMENT OF SIGNIFICANCE: This research will provide new insights into the medicinal benefit of Joint resident Mesenchymal Stem Cells (MSCs), specifically on its cartilage tissue engineering ability. Through this review, the community will further realize promoting joint resident mesenchymal stem cells, especially cartilage progenitor/MSC-like progenitor cells (CPSC), as a preventive measure against osteoarthritis and cartilage injury. People and medical institutions may also consider cartilage derived MSC as an alternative approach against cartilage degeneration. Moreover, the discussion presented in this study will convey valuable information for future research that will explore the medicinal benefits of cartilage derived MSC.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Orthopedics, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Limei Xu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, 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, Shandong 272029, China
| | - Yujie Liang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| | - Yuanmin Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| |
Collapse
|
8
|
Ji S, Xiong M, Chen H, Liu Y, Zhou L, Hong Y, Wang M, Wang C, Fu X, Sun X. Cellular rejuvenation: molecular mechanisms and potential therapeutic interventions for diseases. Signal Transduct Target Ther 2023; 8:116. [PMID: 36918530 PMCID: PMC10015098 DOI: 10.1038/s41392-023-01343-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/16/2022] [Accepted: 01/19/2023] [Indexed: 03/16/2023] Open
Abstract
The ageing process is a systemic decline from cellular dysfunction to organ degeneration, with more predisposition to deteriorated disorders. Rejuvenation refers to giving aged cells or organisms more youthful characteristics through various techniques, such as cellular reprogramming and epigenetic regulation. The great leaps in cellular rejuvenation prove that ageing is not a one-way street, and many rejuvenative interventions have emerged to delay and even reverse the ageing process. Defining the mechanism by which roadblocks and signaling inputs influence complex ageing programs is essential for understanding and developing rejuvenative strategies. Here, we discuss the intrinsic and extrinsic factors that counteract cell rejuvenation, and the targeted cells and core mechanisms involved in this process. Then, we critically summarize the latest advances in state-of-art strategies of cellular rejuvenation. Various rejuvenation methods also provide insights for treating specific ageing-related diseases, including cellular reprogramming, the removal of senescence cells (SCs) and suppression of senescence-associated secretory phenotype (SASP), metabolic manipulation, stem cells-associated therapy, dietary restriction, immune rejuvenation and heterochronic transplantation, etc. The potential applications of rejuvenation therapy also extend to cancer treatment. Finally, we analyze in detail the therapeutic opportunities and challenges of rejuvenation technology. Deciphering rejuvenation interventions will provide further insights into anti-ageing and ageing-related disease treatment in clinical settings.
Collapse
Affiliation(s)
- Shuaifei Ji
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Mingchen Xiong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Huating Chen
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Yiqiong Liu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Laixian Zhou
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Yiyue Hong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Mengyang Wang
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau SAR, China.
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China.
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China.
| |
Collapse
|
9
|
Wang B, Qinglai T, Yang Q, Li M, Zeng S, Yang X, Xiao Z, Tong X, Lei L, Li S. Functional acellular matrix for tissue repair. Mater Today Bio 2023; 18:100530. [PMID: 36601535 PMCID: PMC9806685 DOI: 10.1016/j.mtbio.2022.100530] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022] Open
Abstract
In view of their low immunogenicity, biomimetic internal environment, tissue- and organ-like physicochemical properties, and functionalization potential, decellularized extracellular matrix (dECM) materials attract considerable attention and are widely used in tissue engineering. This review describes the composition of extracellular matrices and their role in stem-cell differentiation, discusses the advantages and disadvantages of existing decellularization techniques, and presents methods for the functionalization and characterization of decellularized scaffolds. In addition, we discuss progress in the use of dECMs for cartilage, skin, nerve, and muscle repair and the transplantation or regeneration of different whole organs (e.g., kidneys, liver, uterus, lungs, and heart), summarize the shortcomings of using dECMs for tissue and organ repair after refunctionalization, and examine the corresponding future prospects. Thus, the present review helps to further systematize the application of functionalized dECMs in tissue/organ transplantation and keep researchers up to date on recent progress in dECM usage.
Collapse
Affiliation(s)
- Bin Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Tang Qinglai
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Mengmeng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Shiying Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Zian Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinying Tong
- Department of Hemodialysis, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
10
|
Guo X, Ma Y, Min Y, Sun J, Shi X, Gao G, Sun L, Wang J. Progress and prospect of technical and regulatory challenges on tissue-engineered cartilage as therapeutic combination product. Bioact Mater 2023; 20:501-518. [PMID: 35846847 PMCID: PMC9253051 DOI: 10.1016/j.bioactmat.2022.06.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/19/2022] [Accepted: 06/19/2022] [Indexed: 12/18/2022] Open
Abstract
Hyaline cartilage plays a critical role in maintaining joint function and pain. However, the lack of blood supply, nerves, and lymphatic vessels greatly limited the self-repair and regeneration of damaged cartilage, giving rise to various tricky issues in medicine. In the past 30 years, numerous treatment techniques and commercial products have been developed and practiced in the clinic for promoting defected cartilage repair and regeneration. Here, the current therapies and their relevant advantages and disadvantages will be summarized, particularly the tissue engineering strategies. Furthermore, the fabrication of tissue-engineered cartilage under research or in the clinic was discussed based on the traid of tissue engineering, that is the materials, seed cells, and bioactive factors. Finally, the commercialized cartilage repair products were listed and the regulatory issues and challenges of tissue-engineered cartilage repair products and clinical application would be reviewed. Tissue engineered cartilage, a promising strategy for articular cartilage repair. Nearly 20 engineered cartilage repair products in clinic based on clinical techniques. Combination product, the classification of tissue-engineered cartilage. Key regulatory compliance issues for combination products.
Collapse
Affiliation(s)
- Xiaolei Guo
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
- Corresponding author.
| | - Yuan Ma
- State Key Laboratory of Tribology, Tsinghua University, Beijing, PR China
| | - Yue Min
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Jiayi Sun
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Xinli Shi
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
- Corresponding author. Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100081, PR China
| | - Guobiao Gao
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Lei Sun
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Jiadao Wang
- State Key Laboratory of Tribology, Tsinghua University, Beijing, PR China
- Corresponding author. State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
11
|
Kim J, Lee SK, Jung M, Jeong SY, You H, Won JY, Han SD, Cho HJ, Park S, Park J, Kim TM, Kim S. Extracellular vesicles from IFN-γ-primed mesenchymal stem cells repress atopic dermatitis in mice. J Nanobiotechnology 2022; 20:526. [PMID: 36496385 PMCID: PMC9741801 DOI: 10.1186/s12951-022-01728-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin disorder characterized by immune dysregulation, pruritus, and abnormal epidermal barrier function. Compared with conventional mesenchymal stem cell (MSC), induced pluripotent stem cell (iPSC)-derived mesenchymal stem cell (iMSC) is recognized as a unique source for producing extracellular vesicles (EVs) because it can be obtained in a scalable manner with an enhanced homogeneity. Stimulation of iMSCs with inflammatory cytokines can improve the immune-regulatory, anti-inflammatory, and tissue-repairing potential of iMSC-derived EVs. RESULTS Proteome analysis showed that IFN-γ-iMSC-EVs are enriched with protein sets that are involved in regulating interferon responses and inflammatory pathways. In AD mice, expression of interleukin receptors for Th2 cytokines (IL-4Rα/13Rα1/31Rα) and activation of their corresponding intracellular signaling molecules was reduced. IFN-γ-iMSC-EVs decreased itching, which was supported by reduced inflammatory cell infiltration and mast cells in AD mouse skin; reduced IgE receptor expression and thymic stromal lymphopoietin and NF-kB activation; and recovered impaired skin barrier, as evidenced by upregulation of key genes of epidermal differentiation and lipid synthesis. CONCLUSIONS IFN-γ-iMSC-EVs inhibit Th2-induced immune responses, suppress inflammation, and facilitate skin barrier restoration, contributing to AD improvement.
Collapse
Affiliation(s)
- Jimin Kim
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul, 05855 South Korea
| | - Seul Ki Lee
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul, 05855 South Korea
| | - Minyoung Jung
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul, 05855 South Korea
| | - Seon-Yeong Jeong
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul, 05855 South Korea
| | - Haedeun You
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul, 05855 South Korea
| | - Ji-Yeon Won
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul, 05855 South Korea
| | - Sang-Deok Han
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul, 05855 South Korea
| | - Hye Jin Cho
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul, 05855 South Korea
| | - Somi Park
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul, 05855 South Korea
| | - Joonghoon Park
- grid.31501.360000 0004 0470 5905Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Gangwon-do 25354 South Korea ,grid.31501.360000 0004 0470 5905Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354 South Korea
| | - Tae Min Kim
- grid.31501.360000 0004 0470 5905Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Gangwon-do 25354 South Korea ,grid.31501.360000 0004 0470 5905Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354 South Korea
| | - Soo Kim
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul, 05855 South Korea
| |
Collapse
|
12
|
Matrix from urine stem cells boosts tissue-specific stem cell mediated functional cartilage reconstruction. Bioact Mater 2022; 23:353-367. [PMID: 36474659 PMCID: PMC9709166 DOI: 10.1016/j.bioactmat.2022.11.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022] Open
Abstract
Articular cartilage has a limited capacity to self-heal once damaged. Tissue-specific stem cells are a solution for cartilage regeneration; however, ex vivo expansion resulting in cell senescence remains a challenge as a large quantity of high-quality tissue-specific stem cells are needed for cartilage regeneration. Our previous report demonstrated that decellularized extracellular matrix (dECM) deposited by human synovium-derived stem cells (SDSCs), adipose-derived stem cells (ADSCs), urine-derived stem cells (UDSCs), or dermal fibroblasts (DFs) provided an ex vivo solution to rejuvenate human SDSCs in proliferation and chondrogenic potential, particularly for dECM deposited by UDSCs. To make the cell-derived dECM (C-dECM) approach applicable clinically, in this study, we evaluated ex vivo rejuvenation of rabbit infrapatellar fat pad-derived stem cells (IPFSCs), an easily accessible alternative for SDSCs, by the abovementioned C-dECMs, in vivo application for functional cartilage repair in a rabbit osteochondral defect model, and potential cellular and molecular mechanisms underlying this rejuvenation. We found that C-dECM rejuvenation promoted rabbit IPFSCs' cartilage engineering and functional regeneration in both ex vivo and in vivo models, particularly for the dECM deposited by UDSCs, which was further confirmed by proteomics data. RNA-Seq analysis indicated that both mesenchymal-epithelial transition (MET) and inflammation-mediated macrophage activation and polarization are potentially involved in the C-dECM-mediated promotion of IPFSCs' chondrogenic capacity, which needs further investigation.
Collapse
|
13
|
Ciavarella C, Valente S, Pasquinelli G. The Characteristics and Survival Potential Under Sub-lethal Stress of Mesenchymal Stromal/Stem Cells Isolated from the Human Vascular Wall. Stem Cells 2022; 40:1071-1077. [PMID: 36099050 PMCID: PMC9806765 DOI: 10.1093/stmcls/sxac066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 08/30/2022] [Indexed: 01/05/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) have been identified in multiple human tissues, including the vascular wall. High proliferative potential, multilineage, and immunomodulatory properties make vascular MSCs promising candidates for regenerative medicine. Indeed, their location is strategic for controlling vascular and extra-vascular tissue homeostasis. However, the clinical application of MSCs, and in particular vascular MSCs, is still challenging. Current studies are focused on developing strategies to improve MSC therapeutic applications, like priming MSCs with stress conditions (hypoxia, nutrient deprivation) to achieve a higher therapeutic potential. The goal of the present study is to review the main findings regarding the MSCs isolated from the human vascular wall. Further, the main priming strategies tested on MSCs from different sources are reported, together with the experience on vascular MSCs isolated from healthy cryopreserved and pathological arteries. Stress induction can be a priming approach able to improve MSC effectiveness through several mechanisms that are discussed in this review. Nevertheless, these issues have not been completely explored in vascular MSCs and potential side effects need to be investigated.
Collapse
Affiliation(s)
| | - Sabrina Valente
- Corresponding author: Sabrina Valente, PhD, DIMES - Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via Massarenti, 9, 40138 Bologna, Italy. Tel: +39 0512144520;
| | - Gianandrea Pasquinelli
- DIMES - Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy,Subcellular Nephro-Vascular Diagnostic Program, Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
14
|
Liu C, Pei M, Li Q, Zhang Y. Decellularized extracellular matrix mediates tissue construction and regeneration. Front Med 2022; 16:56-82. [PMID: 34962624 PMCID: PMC8976706 DOI: 10.1007/s11684-021-0900-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023]
Abstract
Contributing to organ formation and tissue regeneration, extracellular matrix (ECM) constituents provide tissue with three-dimensional (3D) structural integrity and cellular-function regulation. Containing the crucial traits of the cellular microenvironment, ECM substitutes mediate cell-matrix interactions to prompt stem-cell proliferation and differentiation for 3D organoid construction in vitro or tissue regeneration in vivo. However, these ECMs are often applied generically and have yet to be extensively developed for specific cell types in 3D cultures. Cultured cells also produce rich ECM, particularly stromal cells. Cellular ECM improves 3D culture development in vitro and tissue remodeling during wound healing after implantation into the host as well. Gaining better insight into ECM derived from either tissue or cells that regulate 3D tissue reconstruction or organ regeneration helps us to select, produce, and implant the most suitable ECM and thus promote 3D organoid culture and tissue remodeling for in vivo regeneration. Overall, the decellularization methodologies and tissue/cell-derived ECM as scaffolds or cellular-growth supplements used in cell propagation and differentiation for 3D tissue culture in vitro are discussed. Moreover, current preclinical applications by which ECM components modulate the wound-healing process are reviewed.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27109, USA.
| |
Collapse
|
15
|
The essential anti-angiogenic strategies in cartilage engineering and osteoarthritic cartilage repair. Cell Mol Life Sci 2022; 79:71. [PMID: 35029764 PMCID: PMC9805356 DOI: 10.1007/s00018-021-04105-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/01/2021] [Accepted: 12/18/2021] [Indexed: 01/16/2023]
Abstract
In the cartilage matrix, complex interactions occur between angiogenic and anti-angiogenic components, growth factors, and environmental stressors to maintain a proper cartilage phenotype that allows for effective load bearing and force distribution. However, as seen in both degenerative disease and tissue engineering, cartilage can lose its vascular resistance. This vascularization then leads to matrix breakdown, chondrocyte apoptosis, and ossification. Research has shown that articular cartilage inflammation leads to compromised joint function and decreased clinical potential for regeneration. Unfortunately, few articles comprehensively summarize what we have learned from previous investigations. In this review, we summarize our current understanding of the factors that stabilize chondrocytes to prevent terminal differentiation and applications of these factors to rescue the cartilage phenotype during cartilage engineering and osteoarthritis treatment. Inhibiting vascularization will allow for enhanced phenotypic stability so that we are able to develop more stable implants for cartilage repair and regeneration.
Collapse
|
16
|
Pei YA, Pei M. Hypoxia Modulates Regenerative Potential of Fetal Stem Cells. APPLIED SCIENCES (BASEL, SWITZERLAND) 2022; 12:363. [PMID: 36660242 PMCID: PMC9846719 DOI: 10.3390/app12010363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Adult mesenchymal stem cells (MSCs) are prone to senescence, which limits the scope of their use in tissue engineering and regeneration and increases the likelihood of post-implantation failure. As a robust alternative cell source, fetal stem cells can prevent an immune reaction and senescence. However, few studies use this cell type. In this study, we sought to characterize fetal cells' regenerative potential in hypoxic conditions. Specifically, we examined whether hypoxic exposure during the expansion and differentiation phases would affect human fetal nucleus pulposus cell (NPC) and fetal synovium-derived stem cell (SDSC) plasticity and three-lineage differentiation potential. We concluded that fetal NPCs represent the most promising cell source for chondrogenic differentiation, as they are more responsive and display stronger phenotypic stability, particularly when expanded and differentiated in hypoxic conditions. Fetal SDSCs have less potential for chondrogenic differentiation compared to their adult counterpart. This study also indicated that fetal SDSCs exhibit a discrepancy in adipogenic and osteogenic differentiation in response to hypoxia.
Collapse
Affiliation(s)
- Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA
- WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
17
|
Cai Y, Wang S, Qu J, Belmonte JCI, Liu GH. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:231-238. [PMID: 35303745 PMCID: PMC8968747 DOI: 10.1093/stcltm/szab012] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/17/2021] [Indexed: 11/14/2022] Open
Abstract
Stem cell therapies, including stem cell transplantation and rejuvenation of stem cells in situ, are promising avenues for tackling a broad range of diseases. Stem cells can both self-renew and differentiate into other cell types, and play a significant role in the regulation of tissue homeostasis and regeneration after cell degeneration or injury. However, stem cell exhaustion or dysfunction increases with age and impedes the normal function of multiple tissues and systems. Thus, stem cell therapies could provide a solution to aging and age-associated diseases. Here, we discuss recent advances in understanding the mechanisms that regulate stem cell regeneration. We also summarize potential strategies for rejuvenating stem cells that leverage intrinsic and extrinsic factors. These approaches may pave the way toward therapeutic interventions aiming at extending both health and life span.
Collapse
Affiliation(s)
- Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People’s Republic of China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, People’s Republic of China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, People’s Republic of China
- Aging Translational Medicine Center, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People’s Republic of China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, People’s Republic of China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- Corresponding author: Jing Qu, PhD, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, People’s Republic of China. Tel: +86-10-64807768;
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- Juan Carlos Izpisúa-Belmonte, PhD, Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA, USA. Tel: (858) 453-4100 x1130;
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People’s Republic of China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, People’s Republic of China
- Guanghui Liu, PhD, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 3A Datun Road, Chaoyang District, Beijing 100101, People’s Republic of China. Tel: +86-10-64807852;
| |
Collapse
|
18
|
Wang Y, Pei YA, Sun Y, Zhou S, Zhang XB, Pei M. Stem cells immortalized by hTERT perform differently from those immortalized by SV40LT in proliferation, differentiation, and reconstruction of matrix microenvironment. Acta Biomater 2021; 136:184-198. [PMID: 34551328 PMCID: PMC8627502 DOI: 10.1016/j.actbio.2021.09.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 11/22/2022]
Abstract
Although matrix microenvironment has the potential to improve expanded stem cell proliferation and differentiation capacity, decellularized extracellular matrix (dECM) deposited by senescent cells does not contribute to the rejuvenation of adult stem cells, which has become a barrier to personalized stem cell therapy. Genetic modification is an effective strategy to protect cells from senescence but it carries the increased risk of malignant transformation and genetic instability. In this study, lentivirus carrying either human telomerase reverse transcriptase (hTERT) or simian virus 40 large T antigen (SV40LT) was used to transduce human infrapatellar fat pad-derived stem cells (IPFSCs). We found that virus transduction modified the proliferative, chondrogenic, and adipogenic abilities of IPFSCs. Interestingly, dECM deposited by immortalized cells significantly influenced replicative senescent IPFSCs in proliferation and differentiation preference, the effect of which is hinged on the approach of immortalization using either SV40LT or hTERT. Our findings indicate both dECM expansion and immortalization strategies can be used for replicative senescent adult stem cells' proliferation and lineage-specific differentiation, which benefits future stem cell-based tissue regeneration. This approach may also work for adult stem cells with premature senescence in elderly/aged patients, which needs further investigation. STATEMENT OF SIGNIFICANCE: Adult stem cells are a promising solution for autologous cell-based therapy. Unfortunately, cell senescence due to donor age and/or ex vivo expansion prevents clinical application. Recent progress with decellularized extracellular matrix provides a potential for the rejuvenation of senescent stem cells by improving their proliferation and differentiation capacities. Given the fact that the young matrix can provide a healthy and energetic microenvironment, in this study, two approaches using lentivirus transduction of hTERT and SV40LT were compared. The goal was to immortalize donor cells for deposition of decellularized extracellular matrix. The matrix was demonstrated to contribute diverging effects on the chondrogenic and adipogenic differentiation of expanded stem cells and exhibited proliferation benefits as well. These findings provide an invaluable asset for stem cell-based tissue regeneration.
Collapse
Affiliation(s)
- Yiming Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; Department of Joint Surgery, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yuan Sun
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Sheng Zhou
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Medicine, Loma Linda University, Loma Linda, CA, USA.
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
19
|
Impact of perlecan, a core component of basement membrane, on regeneration of cartilaginous tissues. Acta Biomater 2021; 135:13-26. [PMID: 34454085 DOI: 10.1016/j.actbio.2021.08.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/02/2021] [Accepted: 08/20/2021] [Indexed: 02/03/2023]
Abstract
As an indispensable component of the extracellular matrix, perlecan (Pln) plays an essential role in cartilaginous tissue function. Although there exist studies suggesting that Pln expressed by cartilaginous tissues is critical for chondrogenesis, few papers have discussed the potential impact Pln may have on cartilage regeneration. In this review, we delineate Pln structure, biomechanical properties, and interactive ligands-which together contribute to the effect Pln has on cartilaginous tissue development. We also review how the signaling pathways of Pln affect cartilage development and scrutinize the potential application of Pln to divisions of cartilage regeneration, spanning vascularization, stem cell differentiation, and biomaterial improvement. The aim of this review is to deepen our understanding of the spatial and temporal interactions that occur between Pln and cartilaginous tissue and ultimately apply Pln in scaffold design to improve cell-based cartilage engineering and regeneration. STATEMENT OF SIGNIFICANCE: As a key component of the basement membrane, Pln plays a critical role in tissue development and repair. Recent findings suggest that Pln existing in the pericellular matrix surrounding mature chondrocytes is actively involved in cartilage regeneration and functionality. We propose that Pln is essential to developing an in vitro matrix niche within biological scaffolds for cartilage tissue engineering.
Collapse
|
20
|
Foo JB, Looi QH, How CW, Lee SH, Al-Masawa ME, Chong PP, Law JX. Mesenchymal Stem Cell-Derived Exosomes and MicroRNAs in Cartilage Regeneration: Biogenesis, Efficacy, miRNA Enrichment and Delivery. Pharmaceuticals (Basel) 2021; 14:1093. [PMID: 34832875 PMCID: PMC8618513 DOI: 10.3390/ph14111093] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes are the small extracellular vesicles secreted by cells for intercellular communication. Exosomes are rich in therapeutic cargos such as microRNA (miRNA), long non-coding RNA (lncRNA), small interfering RNA (siRNA), DNA, protein, and lipids. Recently, many studies have focused on miRNAs as a promising therapeutic factor to support cartilage regeneration. Exosomes are known to contain a substantial amount of a variety of miRNAs. miRNAs regulate the post-transcriptional gene expression by base-pairing with the target messenger RNA (mRNA), leading to gene silencing. Several exosomal miRNAs have been found to play a role in cartilage regeneration by promoting chondrocyte proliferation and matrix secretion, reducing scar tissue formation, and subsiding inflammation. The exosomal miRNA cargo can be modulated using techniques such as cell transfection and priming as well as post-secretion modifications to upregulate specific miRNAs to enhance the therapeutic effect. Exosomes are delivered to the joints through direct injection or via encapsulation within a scaffold for sustained release. To date, exosome therapy for cartilage injuries has yet to be optimized as the ideal cell source for exosomes, and the dose and method of delivery have yet to be identified. More importantly, a deeper understanding of the role of exosomal miRNAs in cartilage repair is paramount for the development of more effective exosome therapy.
Collapse
Affiliation(s)
- Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
- Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Qi Hao Looi
- My Cytohealth Sdn. Bhd., D353a, Menara Suezcap 1, KL Gateway, no. 2, Jalan Kerinchi, Gerbang Kerinchi Lestari, Kuala Lumpur 59200, Malaysia;
- National Orthopaedic Centre of Excellence in Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Bandar Sunway 47500, Malaysia;
| | - Sau Har Lee
- Centre for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Maimonah Eissa Al-Masawa
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia;
| | - Pei Pei Chong
- Faculty of Health and Medical Sciences, School of Biosciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
21
|
Peck SH, Bendigo JR, Tobias JW, Dodge GR, Malhotra NR, Mauck RL, Smith LJ. Hypoxic Preconditioning Enhances Bone Marrow-Derived Mesenchymal Stem Cell Survival in a Low Oxygen and Nutrient-Limited 3D Microenvironment. Cartilage 2021; 12:512-525. [PMID: 30971109 PMCID: PMC8461160 DOI: 10.1177/1947603519841675] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Skeletal tissues such as intervertebral disc and articular cartilage possess limited innate potential to regenerate, in part due to their avascularity and low cell density. Despite recent advances in mesenchymal stem cell (MSC)-based disc and cartilage regeneration, key challenges remain, including the sensitivity of these cells to in vivo microenvironmental stress such as low oxygen and limited nutrition. The objective of this study was to investigate whether preconditioning with hypoxia and/or transforming growth factor-β 3 (TGF-β3) can enhance MSC survival and extracellular matrix production in a low oxygen and nutrient-limited microenvironment. DESIGN MSCs from multiple bovine donors were preconditioned in monolayer in normoxia or hypoxia, with or without TGF-β3, and the global effects on gene expression were examined using microarrays. Subsequently, the effects of preconditioning on MSC survival and extracellular matrix production were examined using low oxygen and nutrient-limited pellet culture experiments. RESULTS Hypoxic preconditioning resulted in upregulation of genes associated with growth, cell-cell signaling, metabolism, and cell stress response pathways, and significantly enhanced MSC survival for all donors in low oxygen and nutrient-limited pellet culture. In contrast, TGF-β3 preconditioning diminished survival. The nature and magnitude of the effects of preconditioning with either hypoxia or TGF-β3 on glycosaminoglycan production were donor dependent. CONCLUSIONS These results strongly support the use of hypoxic preconditioning to improve postimplantation MSC survival in avascular tissues such as disc and cartilage.
Collapse
Affiliation(s)
- Sun H. Peck
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Justin R. Bendigo
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - John W. Tobias
- Penn Genomic Analysis Core, University of Pennsylvania, Philadelphia, PA, USA
| | - George R. Dodge
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Neil R. Malhotra
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Lachlan J. Smith
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| |
Collapse
|
22
|
Vahedi P, Moghaddamshahabi R, Webster TJ, Calikoglu Koyuncu AC, Ahmadian E, Khan WS, Jimale Mohamed A, Eftekhari A. The Use of Infrapatellar Fat Pad-Derived Mesenchymal Stem Cells in Articular Cartilage Regeneration: A Review. Int J Mol Sci 2021; 22:9215. [PMID: 34502123 PMCID: PMC8431575 DOI: 10.3390/ijms22179215] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cartilage is frequently damaged with a limited capacity for repair. Current treatment strategies are insufficient as they form fibrocartilage as opposed to hyaline cartilage, and do not prevent the progression of degenerative changes. There is increasing interest in the use of autologous mesenchymal stem cells (MSC) for tissue regeneration. MSCs that are used to treat articular cartilage defects must not only present a robust cartilaginous production capacity, but they also must not cause morbidity at the harvest site. In addition, they should be easy to isolate from the tissue and expand in culture without terminal differentiation. The source of MSCs is one of the most important factors that may affect treatment. The infrapatellar fat pad (IPFP) acts as an important reservoir for MSC and is located in the anterior compartment of the knee joint in the extra-synovial area. The IPFP is a rich source of MSCs, and in this review, we discuss studies that demonstrate that these cells have shown many advantages over other tissues in terms of ease of isolation, expansion, and chondrogenic differentiation. Future studies in articular cartilage repair strategies and suitable extraction as well as cell culture methods will extend the therapeutical application of IPFP-derived MSCs into additional orthopedic fields, such as osteoarthritis. This review provides the latest research concerning the use of IPFP-derived MSCs in the treatment of articular cartilage damage, providing critical information for the field to grow.
Collapse
Affiliation(s)
- Parviz Vahedi
- Department of Anatomical Sciences, Maragheh University of Medical Sciences, Maragheh 78151-55158, Iran;
| | - Rana Moghaddamshahabi
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta 99628, North Cyprus, Turkey;
| | - Thomas J. Webster
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA;
| | - Ayse Ceren Calikoglu Koyuncu
- Materials and Metallurgical Engineering Department, Faculty of Technology, Marmara University, Istanbul 34722, Turkey;
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz 51666-15731, Iran;
| | - Wasim S. Khan
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Ali Jimale Mohamed
- Department of Pharmacology, Faculty of Medicine, Somali National University, Mogadishu 801, Somalia;
| | - Aziz Eftekhari
- Department of Toxicology and Pharmacology, Maragheh University of Medical Sciences, Maragheh 78151-55158, Iran
- Department of Synthesis and Characterization of Polymers, Polymer Institute, Slovak Academy of Sciences (SAS), Dúbravská cesta, 9, 845 41 Bratislava, Slovakia
| |
Collapse
|
23
|
Noh YK, Kim SW, Kim IH, Park K. Human nasal septal chondrocytes (NSCs) preconditioned on NSC-derived matrix improve their chondrogenic potential. Biomater Res 2021; 25:10. [PMID: 33823936 PMCID: PMC8025325 DOI: 10.1186/s40824-021-00211-z] [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: 11/30/2020] [Accepted: 03/17/2021] [Indexed: 01/22/2023] Open
Abstract
Background Extracellular matrix (ECM) has a profound effect on cell behaviors. In this study, we prepare a decellularized human nasal septal chondrocyte (NSC)-derived ECM (CHDM), as a natural (N-CHDM) or soluble form (S-CHDM), and investigate their impact on NSCs differentiation. Methods N-CHDM, S-CHDM were obtained from NSC. To evaluate function of NSC cultured on each substrate, gene expression using chondrogenic marker, and chondrogenic protein expression were tested. Preconditioned NSCs-loaded scaffolds were transplanted in nude mice for 3 weeks and analyzed. Results When cultivated on each substrate, NSCs exhibited similar cell spread area but showed distinct morphology on N-CHDM with significantly lower cell circularity. They were highly proliferative on N-CHDM than S-CHDM and tissue culture plastic (TCP), and showed more improved cell differentiation, as assessed via chondrogenic marker (Col2, Sox9, and Aggrecan) expression and immunofluorescence of COL II. We also investigated the effect of NSCs preconditioning on three different 2D substrates while NSCs were isolated from those substrates, subsequently transferred to 3D mesh scaffold, then cultivated them in vitro or transplanted in vivo. The number of cells in the scaffolds was similar to each other at 5 days but cell differentiation was notably better with NSCs preconditioned on N-CHDM, as assessed via real-time q-PCR, Western blot, and immunofluorescence. Moreover, when those NSCs-loaded polymer scaffolds were transplanted subcutaneously in nude mice for 3 weeks and analyzed, the NSCs preconditioned on the N-CHDM showed significantly advanced cell retention in the scaffold, more cells with a chondrocyte lacunae structure, and larger production of cartilage ECM (COL II, glycosaminoglycan). Conclusions Taken together, a natural form of decellularized ECM, N-CHDM would present an advanced chondrogenic potential over a reformulated ECM (S-CHDM) or TCP substrate, suggesting that N-CHDM may hold more diverse signaling cues, not just limited to ECM component.
Collapse
Affiliation(s)
- Yong Kwan Noh
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), 02792, Seoul, Republic of Korea.,Department of Biotechnology, Korea University, 02841, Seoul, Republic of Korea
| | - Sung Won Kim
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
| | - Ik-Hwan Kim
- Department of Biotechnology, Korea University, 02841, Seoul, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), 02792, Seoul, Republic of Korea. .,Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), 02792, Seoul, Republic of Korea.
| |
Collapse
|
24
|
Nouri Barkestani M, Naserian S, Uzan G, Shamdani S. Post-decellularization techniques ameliorate cartilage decellularization process for tissue engineering applications. J Tissue Eng 2021; 12:2041731420983562. [PMID: 33738088 PMCID: PMC7934046 DOI: 10.1177/2041731420983562] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/06/2020] [Indexed: 12/17/2022] Open
Abstract
Due to the current lack of innovative and effective therapeutic approaches, tissue engineering (TE) has attracted much attention during the last decades providing new hopes for the treatment of several degenerative disorders. Tissue engineering is a complex procedure, which includes processes of decellularization and recellularization of biological tissues or functionalization of artificial scaffolds by active cells. In this review, we have first discussed those conventional steps, which have led to great advancements during the last several years. Moreover, we have paid special attention to the new methods of post-decellularization that can significantly ameliorate the efficiency of decellularized cartilage extracellular matrix (ECM) for the treatment of osteoarthritis (OA). We propose a series of post-decellularization procedures to overcome the current shortcomings such as low mechanical strength and poor bioactivity to improve decellularized ECM scaffold towards much more efficient and higher integration.
Collapse
Affiliation(s)
| | - Sina Naserian
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France.,Université Paris-Saclay, CNRS, Centre de Nanosciences et Nanotechnologies C2N, UMR9001, Palaiseau, France.,CellMedEx, Saint Maur Des Fossés, France
| | - Georges Uzan
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France.,Paris-Saclay University, Villejuif, France
| | - Sara Shamdani
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France.,CellMedEx, Saint Maur Des Fossés, France
| |
Collapse
|
25
|
Research Progress on Stem Cell Therapies for Articular Cartilage Regeneration. Stem Cells Int 2021; 2021:8882505. [PMID: 33628274 PMCID: PMC7895563 DOI: 10.1155/2021/8882505] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/11/2021] [Accepted: 01/28/2021] [Indexed: 02/07/2023] Open
Abstract
Injury of articular cartilage can cause osteoarthritis and seriously affect the physical and mental health of patients. Unfortunately, current surgical treatment techniques that are commonly used in the clinic cannot regenerate articular cartilage. Regenerative medicine involving stem cells has entered a new stage and is considered the most promising way to regenerate articular cartilage. In terms of theories on the mechanism, it was thought that stem cell-mediated articular cartilage regeneration was achieved through the directional differentiation of stem cells into chondrocytes. However, recent evidence has shown that the stem cell secretome plays an important role in biological processes such as the immune response, inflammation regulation, and drug delivery. At the same time, the stem cell secretome can effectively mediate the process of tissue regeneration. This new theory has attributed the therapeutic effect of stem cells to their paracrine effects. The application of stem cells is not limited to exogenous stem cell transplantation. Endogenous stem cell homing and in situ regeneration strategies have received extensive attention. The application of stem cell derivatives, such as conditioned media, extracellular vesicles, and extracellular matrix, is an extension of stem cell paracrine theory. On the other hand, stem cell pretreatment strategies have also shown promising therapeutic effects. This article will systematically review the latest developments in these areas, summarize challenges in articular cartilage regeneration strategies involving stem cells, and describe prospects for future development.
Collapse
|
26
|
Mao C, Li D, Zhou E, Gao E, Zhang T, Sun S, Gao L, Fan Y, Wang C. Extracellular vesicles from anoxia preconditioned mesenchymal stem cells alleviate myocardial ischemia/reperfusion injury. Aging (Albany NY) 2021; 13:6156-6170. [PMID: 33578393 PMCID: PMC7950238 DOI: 10.18632/aging.202611] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) produced by anoxia-preconditioned mesenchymal stem cells (MSCs) may afford greater cardioprotection against myocardial ischemia-reperfusion injury (MIRI) than EVs derived from normoxic MSCs. Here, we isolated EVs from mouse adipose-derived MSCs (ADSCs) subjected to anoxia preconditioning or normoxia and evaluated their ability to promote survival of mouse cardiomyocytes following MIRI in vivo and anoxia/reoxygenation (AR) in vitro. Injection of anoxia-preconditioned ADSC EVs (Int-EVs) reduced both infarct size and cardiomyocyte apoptosis to a greater extent than normoxic ADSC EVs (NC-EVs) in mice subjected to MIRI. Sequencing EV-associated miRNAs revealed differential upregulation of ten miRNAs predicted to bind thioredoxin-interacting protein (TXNIP), an inflammasome- and pyroptosis-related protein. We confirmed direct binding of miRNA224-5p, the most upregulated miRNA in Int-EVs, to TXNIP and asserted through western blotting and apoptosis assays a critical protective role for this miRNA against AR-induced cardiomyocyte death. Our results suggest that ischemia-reperfusion triggers TXNIP-induced inflammasome activation in cardiomyocytes, which leads to apoptosis rather than pyroptosis due to low basal levels of the pyroptosis executioner protein gasdermin D in these cells. The antiapoptotic effect of EV-associated miRNA224-5p would in turn result from TXNIP downregulation, which prevents caspase-1-mediated degradation of GATA4 and sustains the expression of Bcl-2.
Collapse
Affiliation(s)
- Chengyu Mao
- Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongjiu Li
- Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - En Zhou
- Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Erhe Gao
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Tiantian Zhang
- Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shufang Sun
- Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Gao
- Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuqi Fan
- Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changqian Wang
- Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Grigoryan EN. Study of Natural Longlife Juvenility and Tissue Regeneration in Caudate Amphibians and Potential Application of Resulting Data in Biomedicine. J Dev Biol 2021; 9:2. [PMID: 33477527 PMCID: PMC7838874 DOI: 10.3390/jdb9010002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 12/14/2022] Open
Abstract
The review considers the molecular, cellular, organismal, and ontogenetic properties of Urodela that exhibit the highest regenerative abilities among tetrapods. The genome specifics and the expression of genes associated with cell plasticity are analyzed. The simplification of tissue structure is shown using the examples of the sensory retina and brain in mature Urodela. Cells of these and some other tissues are ready to initiate proliferation and manifest the plasticity of their phenotype as well as the correct integration into the pre-existing or de novo forming tissue structure. Without excluding other factors that determine regeneration, the pedomorphosis and juvenile properties, identified on different levels of Urodele amphibians, are assumed to be the main explanation for their high regenerative abilities. These properties, being fundamental for tissue regeneration, have been lost by amniotes. Experiments aimed at mammalian cell rejuvenation currently use various approaches. They include, in particular, methods that use secretomes from regenerating tissues of caudate amphibians and fish for inducing regenerative responses of cells. Such an approach, along with those developed on the basis of knowledge about the molecular and genetic nature and age dependence of regeneration, may become one more step in the development of regenerative medicine.
Collapse
Affiliation(s)
- Eleonora N Grigoryan
- Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
28
|
Wang Y, Hu G, Hill RC, Dzieciatkowska M, Hansen KC, Zhang XB, Yan Z, Pei M. Matrix reverses immortalization-mediated stem cell fate determination. Biomaterials 2021; 265:120387. [PMID: 32987274 PMCID: PMC7944411 DOI: 10.1016/j.biomaterials.2020.120387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/24/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Primary cell culture in vitro suffers from cellular senescence. We hypothesized that expansion on decellularized extracellular matrix (dECM) deposited by simian virus 40 large T antigen (SV40LT) transduced autologous infrapatellar fat pad stem cells (IPFSCs) could rejuvenate high-passage IPFSCs in both proliferation and chondrogenic differentiation. In the study, we found that SV40LT transduced IPFSCs exhibited increased proliferation and adipogenic potential but decreased chondrogenic potential. Expansion on dECMs deposited by passage 5 IPFSCs yielded IPFSCs with dramatically increased proliferation and chondrogenic differentiation capacity; however, this enhanced capacity diminished if IPFSCs were grown on dECM deposited by passage 15 IPFSCs. Interestingly, expansion on dECM deposited by SV40LT transduced IPFSCs yielded IPFSCs with enhanced proliferation and chondrogenic capacity but decreased adipogenic potential, particularly for the dECM group derived from SV40LT transduced passage 15 cells. Our immunofluorescence staining and proteomics data identify matrix components such as basement membrane proteins as top candidates for matrix mediated IPFSC rejuvenation. Both cell proliferation and differentiation were endorsed by transcripts measured by RNASeq during the process. This study provides a promising model for in-depth investigation of the matrix protein influence on surrounding stem cell differentiation.
Collapse
Affiliation(s)
- Yiming Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA; Bioinformatics Core, West Virginia University, Morgantown, WV, USA
| | - Ryan C Hill
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Kirk C Hansen
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, USA
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Tianjin, China; Department of Medicine, Loma Linda University, Loma Linda, CA, USA.
| | - Zuoqin Yan
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
29
|
Zong Z, Zhang X, Yang Z, Yuan W, Huang J, Lin W, Chen T, Yu J, Chen J, Cui L, Li G, Wei B, Lin S. Rejuvenated ageing mesenchymal stem cells by stepwise preconditioning ameliorates surgery-induced osteoarthritis in rabbits. Bone Joint Res 2021; 10:10-21. [PMID: 33382341 PMCID: PMC7845463 DOI: 10.1302/2046-3758.101.bjr-2020-0249.r1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIMS Ageing-related incompetence becomes a major hurdle for the clinical translation of adult stem cells in the treatment of osteoarthritis (OA). This study aims to investigate the effect of stepwise preconditioning on cellular behaviours in human mesenchymal stem cells (hMSCs) from ageing patients, and to verify their therapeutic effect in an OA animal model. METHODS Mesenchymal stem cells (MSCs) were isolated from ageing patients and preconditioned with chondrogenic differentiation medium, followed by normal growth medium. Cellular assays including Bromodeoxyuridine / 5-bromo-2'-deoxyuridine (BrdU), quantitative polymerase chain reaction (q-PCR), β-Gal, Rosette forming, and histological staining were compared in the manipulated human mesenchymal stem cells (hM-MSCs) and their controls. The anterior cruciate ligament transection (ACLT) rabbit models were locally injected with two millions, four millions, or eight millions of hM-MSCs or phosphate-buffered saline (PBS). Osteoarthritis Research Society International (OARSI) scoring was performed to measure the pathological changes in the affected joints after staining. Micro-CT analysis was conducted to determine the microstructural changes in subchondral bone. RESULTS Stepwise preconditioning approach significantly enhanced the proliferation and chondrogenic potential of ageing hMSCs at early passage. Interestingly, remarkably lower immunogenicity and senescence was also found in hM-MSCs. Data from animal studies showed cartilage damage was retarded and subchondral bone remodelling was prevented by the treatment of preconditioned MSCs. The therapeutic effect depended on the number of cells applied to animals, with the best effect observed when treated with eight millions of hM-MSCs. CONCLUSION This study demonstrated a reliable and feasible stepwise preconditioning strategy to improve the safety and efficacy of ageing MSCs for the prevention of OA development. Cite this article: Bone Joint Res 2021;10(1):10-21.
Collapse
Affiliation(s)
- Zhixian Zong
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Xiaoting Zhang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong
| | - Zhengmeng Yang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong
| | - Weihao Yuan
- Department of Biomedical Engineering, Faculty of Engineering, Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Jianping Huang
- Department of Stomatology, Second Clinical Medical College, Guangdong Medical University, Dongguan, China
| | - Weiping Lin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong
| | - Ting Chen
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Jiahao Yu
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Jiming Chen
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Liao Cui
- Department of Pharmacology, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, Hong Kong
| | - Bo Wei
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
| | - Sien Lin
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, First Clinical Medical College, Guangdong Medical University, Zhanjiang, China
- Department of Pharmacology, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, USA
| |
Collapse
|
30
|
Lu Z, Zhou S, Vaida J, Gao G, Stewart A, Parenti J, Yan L, Pei M. Unfavorable Contribution of a Tissue-Engineering Cartilage Graft to Osteochondral Defect Repair in Young Rabbits. Front Cell Dev Biol 2020; 8:595518. [PMID: 33195273 PMCID: PMC7658375 DOI: 10.3389/fcell.2020.595518] [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: 08/16/2020] [Accepted: 10/12/2020] [Indexed: 11/17/2022] Open
Abstract
A stem cell-based tissue-engineering approach is a promising strategy for treatment of cartilage defects. However, there are conflicting data in the feasibility of using this approach in young recipients. A young rabbit model with an average age of 7.7 months old was used to evaluate the effect of a tissue-engineering approach on the treatment of osteochondral defects. Following in vitro evaluation of proliferation and chondrogenic capacity of infrapatellar fat pad-derived stem cells (IPFSCs) after expansion on either tissue culture plastic (TCP) or decellularized extracellular matrix (dECM), a premature tissue construct engineered from pretreated IPFSCs was used to repair osteochondral defects in young rabbits. We found that dECM expanded IPFSCs exhibited higher proliferation and chondrogenic differentiation compared to TCP expanded cells in both pellet and tissue construct culture systems. Six weeks after creation of bilateral osteochondral defects in the femoral trochlear groove of rabbits, the Empty group (left untreated) had the best cartilage resurfacing with the highest score in Modified O’Driscoll Scale (MODS) than the other groups; however, this score had no significant difference compared to that of 15-week samples, indicating that young rabbits stop growing cartilage once they reach 9 months old. Interestingly, implantation of premature tissue constructs from both dECM and TCP groups exhibited significantly improved cartilage repair at 15 weeks compared to those at six weeks (about 9 months old), indicating that a tissue-engineering approach is able to repair adult cartilage defects. We also found that implanted pre-labeled cells in premature tissue constructs were undetectable in resurfaced cartilage at both time points. This study suggests that young rabbits (less than 9 months old) might respond differently to the classical tissue-engineering approach that is considered as a potential treatment for cartilage defects in adult rabbits.
Collapse
Affiliation(s)
- Zhihua Lu
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States.,Department of Orthopedics, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, China
| | - Sheng Zhou
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| | - Justin Vaida
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| | - Gongming Gao
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| | - Amanda Stewart
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| | - Joshua Parenti
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
| | - Lianqi Yan
- Department of Orthopedics, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States.,WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
31
|
Zhu W, Cao L, Song C, Pang Z, Jiang H, Guo C. Cell-derived decellularized extracellular matrix scaffolds for articular cartilage repair. Int J Artif Organs 2020; 44:269-281. [PMID: 32945220 DOI: 10.1177/0391398820953866] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Articular cartilage repair remains a great clinical challenge. Tissue engineering approaches based on decellularized extracellular matrix (dECM) scaffolds show promise for facilitating articular cartilage repair. Traditional regenerative approaches currently used in clinical practice, such as microfracture, mosaicplasty, and autologous chondrocyte implantation, can improve cartilage repair and show therapeutic effect to some degree; however, the long-term curative effect is suboptimal. As dECM prepared by proper decellularization procedures is a biodegradable material, which provides space for regeneration tissue growth, possesses low immunogenicity, and retains most of its bioactive molecules that maintain tissue homeostasis and facilitate tissue repair, dECM scaffolds may provide a biomimetic microenvironment promoting cell attachment, proliferation, and chondrogenic differentiation. Currently, cell-derived dECM scaffolds have become a research hotspot in the field of cartilage tissue engineering, as ECM derived from cells cultured in vitro has many advantages compared with native cartilage ECM. This review describes cell types used to secrete ECM, methods of inducing cells to secrete cartilage-like ECM and decellularization methods to prepare cell-derived dECM. The potential mechanism of dECM scaffolds on cartilage repair, methods for improving the mechanical strength of cell-derived dECM scaffolds, and future perspectives on cell-derived dECM scaffolds are also discussed in this review.
Collapse
Affiliation(s)
- Wenrun Zhu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lu Cao
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunfeng Song
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiying Pang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haochen Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Changan Guo
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
32
|
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
|
33
|
Dai H, Chen R, Gui C, Tao T, Ge Y, Zhao X, Qin R, Yao W, Gu S, Jiang Y, Gui J. Eliminating senescent chondrogenic progenitor cells enhances chondrogenesis under intermittent hydrostatic pressure for the treatment of OA. Stem Cell Res Ther 2020; 11:199. [PMID: 32450920 PMCID: PMC7249424 DOI: 10.1186/s13287-020-01708-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 04/19/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
Background Osteoarthritis (OA) is a major cause of limb dysfunction, and distraction arthroplasty which generates intermittent hydrostatic pressure (IHP) is an effective approach for OA treatment. However, the result was not always satisfactory and the reasons remained unresolved. Because aging is recognized as an important risk factor for OA and chondrogenic progenitor cells (CPCs) could acquire senescent phenotype, we made a hypothesis that CPCs senescence could have harmful effect on chondrogenesis and the outcome of distraction arthroplasty could be improved by eliminating senescent CPCs pharmacologically. Methods The role of senescent CPCs on distraction arthroplasty was first determined by comparing the cartilage samples from the failure and non-failure patients. Next, the biological behaviors of senescent CPCs were observed in the in vitro cell culture and IHP model. Finally, the beneficial effect of senescent CPCs clearance by senolytic dasatinib and quercetin (DQ) on cartilage regeneration was observed in the in vitro and in vivo IHP model. Results Larger quantities of senescent CPCs along with increased IL-1 β secretion were demonstrated in the failure patients of distraction arthroplasty. Senescent CPCs revealed impaired proliferation and chondrogenic capability and also had increased IL-1 β synthesis, typical of senescence-associated secretory phenotype (SASP). CPCs senescence and SASP formation were mutually dependent in vitro. Greater amounts of senescent CPCs were negatively correlated with IHP-induced chondrogenesis. In contrast, chondrogenesis could be significantly improved by DQ pretreatment which selectively induced senescent CPCs into apoptosis in the in vitro and in vivo IHP model. Mechanistically, senescent CPCs elimination could decrease SASP formation and therefore promote the proliferation and chondrogenic regeneration capacity of the surrounding survived CPCs under IHP stimulation. Conclusions Eliminating senescent CPCs by senolytics could decrease SASP formation and improve the result of joint distraction arthroplasty effectively. Our study provided a novel CPCs senescence-based therapeutic target for improving the outcome of OA treatment.
Collapse
Affiliation(s)
- Hanhao Dai
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ran Chen
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Chang Gui
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Tianqi Tao
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yingbin Ge
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Xilian Zhao
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ran Qin
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Wangxiang Yao
- Department of Orthopaedics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Song Gu
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yiqiu Jiang
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jianchao Gui
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
34
|
Zhao L, Hu C, Han F, Cai F, Wang J, Chen J. Preconditioning is an effective strategy for improving the efficiency of mesenchymal stem cells in kidney transplantation. Stem Cell Res Ther 2020; 11:197. [PMID: 32448356 PMCID: PMC7245776 DOI: 10.1186/s13287-020-01721-8] [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: 03/10/2020] [Revised: 04/22/2020] [Accepted: 05/08/2020] [Indexed: 12/13/2022] Open
Abstract
The inevitable side effects caused by lifelong immunosuppressive agents in kidney transplantation patients spurred the exploration of novel immunosuppressive strategies with definite curative effects and minimal adverse effects. Mesenchymal stem cells (MSCs) have become a promising candidate due to their role in modulating the immune system. Encouraging results obtained from experimental models have promoted the translation of this strategy into clinical settings. However, the demonstration of only marginal or transient benefits by several recent clinical controlled studies has made physicians hesitant to adopt the routine utilization of this procedure in clinical settings. Impaired MSC function after infusion in vivo was thought to be the main reason for their limited effects. For this reason, some preconditioning methods were developed. In this review, we aim to outline the current understanding of the preconditioning methods being explored as a strategy to improve the therapeutic effects of MSCs in kidney transplantation and promote its clinical translation.
Collapse
Affiliation(s)
- Lingfei Zhao
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Fei Han
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Fanghao Cai
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Junni Wang
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jianghua Chen
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China. .,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang Province, People's Republic of China. .,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
35
|
Identifying the Therapeutic Significance of Mesenchymal Stem Cells. Cells 2020; 9:cells9051145. [PMID: 32384763 PMCID: PMC7291143 DOI: 10.3390/cells9051145] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
The pleiotropic behavior of mesenchymal stem cells (MSCs) has gained global attention due to their immense potential for immunosuppression and their therapeutic role in immune disorders. MSCs migrate towards inflamed microenvironments, produce anti-inflammatory cytokines and conceal themselves from the innate immune system. These signatures are the reason for the uprising in the sciences of cellular therapy in the last decades. Irrespective of their therapeutic role in immune disorders, some factors limit beneficial effects such as inconsistency of cell characteristics, erratic protocols, deviating dosages, and diverse transfusion patterns. Conclusive protocols for cell culture, differentiation, expansion, and cryopreservation of MSCs are of the utmost importance for a better understanding of MSCs in therapeutic applications. In this review, we address the immunomodulatory properties and immunosuppressive actions of MSCs. Also, we sum up the results of the enhancement, utilization, and therapeutic responses of MSCs in treating inflammatory diseases, metabolic disorders and diabetes.
Collapse
|
36
|
Lu Z, Yan L, Pei M. Commentary on 'Surface markers associated with chondrogenic potential of human mesenchymal stromal/stem cells'. F1000Res 2020; 9. [PMID: 32047607 PMCID: PMC6979467 DOI: 10.12688/f1000research.21207.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/17/2020] [Indexed: 12/27/2022] Open
Abstract
In the last decade, researchers have searched for predictive surface markers of multipotent mesenchymal stromal/stem cells (MSCs) for ensuring improved therapeutic outcomes following cartilage damage in humans. However, we have achieved only limited progress because of the challenge presented by conflicting data. This commentary provides some evidence to prove a lack of success with current efforts, including an inconsistency in accepted surface markers and chondrogenic potential of MSCs as well as the tissue source-dependent MSC surface markers that correlate with chondrogenic potential. A brief discussion on these disputed topics and perspective about functionally predictive surface markers and standardization of analytic procedures are also highlighted.
Collapse
Affiliation(s)
- Zhihua Lu
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA.,Department of Orthopaedics, Orthopaedics Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu, 225001, China
| | - Lianqi Yan
- Department of Orthopaedics, Orthopaedics Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu, 225001, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA.,WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506, USA
| |
Collapse
|
37
|
Li J, Narayanan K, Zhang Y, Hill RC, He F, Hansen KC, Pei M. Role of lineage-specific matrix in stem cell chondrogenesis. Biomaterials 2019; 231:119681. [PMID: 31864016 DOI: 10.1016/j.biomaterials.2019.119681] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
Cartilage repair in clinics is a challenge owing to the limited regenerative capacities of cartilage. Synovium-derived stem cells (SDSCs) are suggested as tissue-specific stem cells for chondrogenesis. In this study, we hypothesize that decellularized extracellular matrix (dECM) deposited by SDSCs could provide a superior tissue-specific matrix microenvironment for optimal rejuvenation of adult SDSCs for cartilage regeneration. dECMs were deposited by adult stem cells with varying chondrogenic capacities; SDSCs (strong) (SECM), adipose-derived stem cells (weak) (AECM) and dermal fibroblasts (weak) (DECM), and urine-derived stem cells (none) (UECM). Plastic flasks (Plastic) were used as a control substrate. Human SDSCs were expanded on the above substrates for one passage and examined for chondrogenic capacities. We found that each dECM consisted of unique matrix proteins and exhibited varied stiffnesses, which affected cell morphology and elasticity. Human SDSCs grown on dECMs displayed a significant increase in cell proliferation and unique surface phenotypes. Under induction media, dECM expanded cells yielded pellets with a dramatically increased number of chondrogenic markers. Interestingly, SECM expanded cells had less potential for hypertrophy compared to those grown on other dECMs, indicating that a tissue-specific matrix might provide a superior microenvironment for stem cell chondrogenic differentiation.
Collapse
Affiliation(s)
- Jingting Li
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Karthikeyan Narayanan
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Ying Zhang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, Shandong, 272067, China
| | - Ryan C Hill
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Fan He
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Kirk C Hansen
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
38
|
Guan S, Zhang K, Li J. Recent Advances in Extracellular Matrix for Engineering Stem Cell Responses. Curr Med Chem 2019; 26:6321-6338. [DOI: 10.2174/0929867326666190704121309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/02/2018] [Accepted: 01/25/2019] [Indexed: 02/06/2023]
Abstract
Stem cell transplantation is an advanced medical technology, which brings hope for the
treatment of some difficult diseases in the clinic. Attributed to its self-renewal and differential
ability, stem cell research has been pushed to the forefront of regenerative medicine and has become
a hot topic in tissue engineering. The surrounding extracellular matrix has physical functions
and important biological significance in regulating the life activities of cells, which may play crucial
roles for in situ inducing specific differentiation of stem cells. In this review, we discuss the
stem cells and their engineering application, and highlight the control of the fate of stem cells, we
offer our perspectives on the various challenges and opportunities facing the use of the components
of extracellular matrix for stem cell attachment, growth, proliferation, migration and differentiation.
Collapse
Affiliation(s)
- Shuaimeng Guan
- School of Life Science, Zhengzhou University, Zhengzhou 450000, China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou 450000, China
| | - Jingan Li
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
39
|
Barakat AH, Elwell VA, Lam KS. Stem cell therapy in discogenic back pain. JOURNAL OF SPINE SURGERY (HONG KONG) 2019; 5:561-583. [PMID: 32043007 PMCID: PMC6989932 DOI: 10.21037/jss.2019.09.22] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/09/2019] [Indexed: 04/23/2023]
Abstract
Chronic low back pain has both substantial social and economic impacts on patients and healthcare budgets. Adding to the magnitude of the problem is the difficulty in identifying the exact causes of disc degeneration with modern day diagnostic and imaging techniques. With that said, current non-operative and surgical treatment modalities for discogenic low back pain fails to meet the expectations in many patients and hence the challenge. The objective for newly emerging stem cell regenerative therapy is to treat degenerative disc disease (DDD) by restoring the disc's cellularity and modulating the inflammatory response. Appropriate patient selection is crucial for the success of stem cell therapy. Regenerative modalities for discogenic pain currently focus on the use of either primary cells harvested from the intervertebral discs or stem cells from other sources whether autogenic or allogenic. The microenvironment in which stem cells are being cultured has been recognized to play a crucial role in directing or maintaining the production of the desired phenotypes and may enhance their regenerative potential. This has led to a more specific focus on innovating more effective culturing techniques, delivery vehicles and scaffolds for stem cell application. Although stem cell therapy might offer an attractive alternative treatment option, more clinical studies are still needed to establish on the safety and feasibility of such therapy. In this literature review, we aim to present the most recent in vivo and in vitro studies related to the use of stem cell therapy in the treatment of discogenic low back pain.
Collapse
Affiliation(s)
- Ahmed H. Barakat
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | - Vivian A. Elwell
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | | |
Collapse
|
40
|
Wang Y, Fu Y, Yan Z, Zhang XB, Pei M. Impact of Fibronectin Knockout on Proliferation and Differentiation of Human Infrapatellar Fat Pad-Derived Stem Cells. Front Bioeng Biotechnol 2019; 7:321. [PMID: 31803729 PMCID: PMC6873900 DOI: 10.3389/fbioe.2019.00321] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022] Open
Abstract
Fibronectin plays an essential role in tissue development and regeneration. However, the effects of fibronectin knockout (FN1-KO) on stem cells' proliferation and differentiation remain unknown. In this study, CRISPR/Cas9 generated FN1-KO in human infrapatellar fat pad-derived stem cells (IPFSCs) was evaluated for proliferation ability including cell cycle and surface markers as well as stemness gene expression and for differentiation capacity including chondrogenic and adipogenic differentiation. High passage IPFSCs were also evaluated for proliferation and differentiation capacity after expansion on decellularized ECM (dECM) deposited by FN1-KO cells. Successful FN1-KO in IPFSCs was confirmed by Sanger sequencing and Inference of CRISPR Edits analysis (ICE) as well as immunostaining for fibronectin expression. Compared to the GFP control, FN1-KO cells showed an increase in cell growth, percentage of cells in the S and G2 phases, and CD105 and CD146 expression but a decrease in expression of stemness markers CD73, CD90, SSEA4, and mesenchymal condensation marker CDH2 gene. FN1-KO decreased both chondrogenic and adipogenic differentiation capacity. Interestingly, IPFSCs grown on dECMs deposited by FN1-KO cells exhibited a decrease in cell proliferation along with a decline in CDH2 expression. After induction, IPFSCs plated on dECMs deposited by FN1-KO cells also displayed decreased expression of both chondrogenic and adipogenic capacity. We concluded that FN1-KO increased human IPFSCs' proliferation capacity; however, this capacity was reversed after expansion on dECM deposited by FN1-KO cells. Significance of fibronectin in chondrogenic and adipogenic differentiation was demonstrated in both FN1-KO IPFSCs and FN(-) matrix microenvironment.
Collapse
Affiliation(s)
- Yiming Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yawen Fu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Tianjin, China
- Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Zuoqin Yan
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Tianjin, China
- Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
41
|
Liu F, Huang X, Luo Z, He J, Haider F, Song C, Peng L, Chen T, Wu B. Hypoxia-Activated PI3K/Akt Inhibits Oxidative Stress via the Regulation of Reactive Oxygen Species in Human Dental Pulp Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6595189. [PMID: 30728888 PMCID: PMC6343138 DOI: 10.1155/2019/6595189] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/19/2018] [Accepted: 10/30/2018] [Indexed: 01/02/2023]
Abstract
In order to use stem cells as a resource for tissue regeneration, it is necessary to induce expansion in vitro. However, during culture, stem cells often lose functional properties and become senescent. Increasing evidence indicates that hypoxic preconditioning with physiological oxygen concentration can maintain the functional properties of stem cells in vitro. The purpose of the current study was to test the hypothesis that hypoxic preconditioning with physiological oxygen concentration can maintain the functional properties of stem cells in culture by reducing oxidative stress. In vitro studies were performed in primary human dental pulp cells (hDPCs). Reduced levels of oxidative stress and increased cellular "stemness" in response to physiological hypoxia were dependent upon the expression of reactive oxygen species (ROS). Subsequently, RNA-sequencing analysis revealed the increased expression of phosphoinositide 3-kinase (PI3K)/Akt signaling in culture, a pathway which regulates oxidative stress. Furthermore, we found evidence that PI3K/Akt signaling might affect intracellular ROS production by negatively regulating expression of the downstream protein Forkhead Box Protein O1 (FOXO1) and Caspase 3. Collectively, our data show that the PI3K/Akt pathway is activated in response to hypoxia and inhibits oxidative stress in a ROS-dependent manner. This study identified redox-mediated hypoxic preconditioning regulatory mechanisms that may be significant for tissue regeneration.
Collapse
Affiliation(s)
- Fei Liu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- College of Stomatology, Southern Medical University, Guangzhou 510515, China
- International Medical Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Xin Huang
- Department of Stomatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Zhenhua Luo
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - Jingjun He
- International Medical Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Farhan Haider
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ci Song
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ling Peng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ting Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Buling Wu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
42
|
Wang Y, Chen S, Yan Z, Pei M. A prospect of cell immortalization combined with matrix microenvironmental optimization strategy for tissue engineering and regeneration. Cell Biosci 2019; 9:7. [PMID: 30627420 PMCID: PMC6321683 DOI: 10.1186/s13578-018-0264-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/21/2018] [Indexed: 12/20/2022] Open
Abstract
Cellular senescence is a major hurdle for primary cell-based tissue engineering and regenerative medicine. Telomere erosion, oxidative stress, the expression of oncogenes and the loss of tumor suppressor genes all may account for the cellular senescence process with the involvement of various signaling pathways. To establish immortalized cell lines for research and clinical use, strategies have been applied including internal genomic or external matrix microenvironment modification. Considering the potential risks of malignant transformation and tumorigenesis of genetic manipulation, environmental modification methods, especially the decellularized cell-deposited extracellular matrix (dECM)-based preconditioning strategy, appear to be promising for tissue engineering-aimed cell immortalization. Due to few review articles focusing on this topic, this review provides a summary of cell senescence and immortalization and discusses advantages and limitations of tissue engineering and regeneration with the use of immortalized cells as well as a potential rejuvenation strategy through combination with the dECM approach.
Collapse
Affiliation(s)
- Yiming Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, PO Box 9196, 64 Medical Center Drive, Morgantown, WV 26506-9196 USA
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai, 200032 China
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, 610083 Sichuan China
| | - Zuoqin Yan
- Department of Orthopaedics, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai, 200032 China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, PO Box 9196, 64 Medical Center Drive, Morgantown, WV 26506-9196 USA
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506 USA
| |
Collapse
|
43
|
Smith LJ, Silverman L, Sakai D, Le Maitre CL, Mauck RL, Malhotra NR, Lotz JC, Buckley CT. Advancing cell therapies for intervertebral disc regeneration from the lab to the clinic: Recommendations of the ORS spine section. JOR Spine 2018; 1:e1036. [PMID: 30895277 PMCID: PMC6419951 DOI: 10.1002/jsp2.1036] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/09/2018] [Accepted: 09/10/2018] [Indexed: 12/28/2022] Open
Abstract
Intervertebral disc degeneration is strongly associated with chronic low back pain, a leading cause of disability worldwide. Current back pain treatment approaches (both surgical and conservative) are limited to addressing symptoms, not necessarily the root cause. Not surprisingly therefore, long-term efficacy of most approaches is poor. Cell-based disc regeneration strategies have shown promise in preclinical studies, and represent a relatively low-risk, low-cost, and durable therapeutic approach suitable for a potentially large patient population, thus making them attractive from both clinical and commercial standpoints. Despite such promise, no such therapies have been broadly adopted clinically. In this perspective we highlight primary obstacles and provide recommendations to help accelerate successful clinical translation of cell-based disc regeneration therapies. The key areas addressed include: (a) Optimizing cell sources and delivery techniques; (b) Minimizing potential risks to patients; (c) Selecting physiologically and clinically relevant efficacy metrics; (d) Maximizing commercial potential; and (e) Recognizing the importance of multidisciplinary collaborations and engaging with clinicians from inception through to clinical trials.
Collapse
Affiliation(s)
- Lachlan J. Smith
- Department of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPennsylvania
- Department of Orthopaedic SurgeryUniversity of PennsylvaniaPhiladelphiaPennsylvania
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvania
| | - Lara Silverman
- DiscGenics Inc.Salt Lake CityUtah
- Department of NeurosurgeryUniversity of Tennessee Health Science CenterMemphisTennessee
| | - Daisuke Sakai
- Department of Orthopaedic Surgery, Surgical ScienceTokai University School of MedicineIseharaJapan
| | | | - Robert L. Mauck
- Department of Orthopaedic SurgeryUniversity of PennsylvaniaPhiladelphiaPennsylvania
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvania
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Neil R. Malhotra
- Department of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Jeffrey C. Lotz
- Department of Orthopaedic SurgeryUniversity of CaliforniaSan FranciscoCalifornia
| | - Conor T. Buckley
- Trinity Centre for BioengineeringTrinity Biomedical Sciences Institute, Trinity College Dublin, The University of DublinDublinIreland
- School of EngineeringTrinity College Dublin, The University of DublinDublinIreland
- Advanced Materials and Bioengineering Research (AMBER) CentreRoyal College of Surgeons in Ireland & Trinity College Dublin, The University of DublinDublinIreland
| |
Collapse
|
44
|
Pizzute T, He F, Zhang XB, Pei M. Impact of Wnt signals on human intervertebral disc cell regeneration. J Orthop Res 2018; 36:3196-3207. [PMID: 30035326 PMCID: PMC7261601 DOI: 10.1002/jor.24115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/16/2018] [Indexed: 02/04/2023]
Abstract
Although preconditioning strategies are growing areas of interest for therapies targeting intervertebral discs (IVDs), it is unknown whether the Wnt signals previously implicated in chondrogenesis, Wnt3A, Wnt5A, and Wnt11, play key roles in the promotion of human nucleus pulposus (NP) cell redifferentiation. In this study, NP cells isolated from herniated disc patients were transduced with lentiviral vectors to overexpress the WNT3A, WNT5A, or WNT11 genes, or CRISPR associated protein 9 (Cas9)/single-guide RNA (sgRNA) vectors to knock out these genes. Following expansion, transduced NP cells were induced for redifferentiation toward the NP phenotype. The overexpression of specific WNT factors led to increases in both glycosaminoglycan (GAG) deposition and expression of redifferentiation genes. These effects were attenuated by knockout of the same WNT genes. These results indicate that specific WNT signals can regulate the expression of redifferentiation genes, unequally impact GAG deposition, and contribute to the redifferentiation of human NP cells. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:3196-3207, 2018.
Collapse
Affiliation(s)
- Tyler Pizzute
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA;,Exercise Physiology, West Virginia University, Morgantown, WV, USA
| | - Fan He
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Tianjin, China;,Department of Medicine, Loma Linda University, Loma Linda, CA, USA;,Co-Corresponding Author: Xiao-Bing Zhang PhD, Division of Regenerative Medicine MC 1528B, Department of Medicine, Loma Linda University, 11234 Anderson Street, Loma Linda, CA 92350, USA,
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA;,Exercise Physiology, West Virginia University, Morgantown, WV, USA;,WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA,Corresponding author: Ming Pei MD, PhD, Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, PO Box 9196, One Medical Center Drive, Morgantown, WV 26506-9196, USA, Telephone: 304-293-1072; Fax: 304-293-7070;
| |
Collapse
|
45
|
Sun Y, Chen S, Pei M. Comparative advantages of infrapatellar fat pad: an emerging stem cell source for regenerative medicine. Rheumatology (Oxford) 2018; 57:2072-2086. [PMID: 29373763 PMCID: PMC6256334 DOI: 10.1093/rheumatology/kex487] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 11/13/2017] [Indexed: 12/15/2022] Open
Abstract
Growing evidence indicates that infrapatellar fat pad (IPFP)-derived stem cells (IPFSCs) exert robust proliferation capacities and multilineage differentiation potentials. However, few papers summarize the advantages that the IPFP and IPFSCs have in regenerative medicine. In this review we delineate the development and anatomy of the IPFP by comparing it with an adjacent fibrous tissue, synovium, and a more frequently harvested fat depot, subcutaneous adipose tissue. Furthermore, we explore the similarities and differences of stem cells from these three tissues in terms of IPFSCs, synovium-derived stem cells and subcutaneous adipose tissue-derived stem cells in proliferation capacity and tri-lineage differentiation potentials, including chondrogenesis, osteogenesis and adipogenesis. Finally, we highlight the advantages of IPFSCs in regenerative medicine, such as the abundant accessibility and the ability to resist inflammation and senescence, two hurdles for cell-based tissue regeneration. Considering the comparative advantages of IPFSCs, the IPFP can serve as an excellent stem cell source for regenerative medicine, particularly for cartilage regeneration.
Collapse
Affiliation(s)
- Yu Sun
- Department of Orthopaedics, Orthopaedics Institute, Subei People’s Hospital of Jiangsu Province, Yangzhou, Jiangsu, China
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Song Chen
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Exercise Physiology, West Virginia University, Morgantown, WV, USA
- Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
46
|
Farrag M, Leipzig ND. Subcutaneous Maturation of Neural Stem Cell-Loaded Hydrogels Forms Region-Specific Neuroepithelium. Cells 2018; 7:cells7100173. [PMID: 30336590 PMCID: PMC6210402 DOI: 10.3390/cells7100173] [Citation(s) in RCA: 12] [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: 09/13/2018] [Revised: 10/09/2018] [Accepted: 10/11/2018] [Indexed: 01/14/2023] Open
Abstract
A combinatorial approach integrating stem cells and capable of exploiting available cues is likely needed to regenerate lost neural tissues and ultimately restore neurologic functions. This study investigates the effects of the subcutaneous maturation of adult-derived neural stem cell (aNSCs) seeded into biomaterial constructs on aNSC differentiation and ultimate regional neuronal identity as a first step toward a future spinal cord injury treatment. To achieve this, we encapsulated rat aNSCs in chitosan-based hydrogels functionalized with immobilized azide-tagged interferon-γ inside a chitosan conduit. Then, we implanted these constructs in the subcutaneous tissues in the backs of rats in the cervical, thoracic, and lumbar regions for 4, 6, and 8 weeks. After harvesting the scaffolds, we analyzed cell differentiation qualitatively using immunohistochemical analysis and quantitatively using RT-qPCR. Results revealed that the hydrogels supported aNSC survival and differentiation up to 4 weeks in the subcutaneous environment as marked by the expression of several neurogenesis markers. Most interesting, the aNSCs expressed region-specific Hox genes corresponding to their region of implantation. This study lays the groundwork for further translational work to recapitulate the potentially undiscovered patterning cues in the subcutaneous tissue and provide support for the conceptual premise that our bioengineering approach can form caudalized region-specific neuroepithelium.
Collapse
Affiliation(s)
- Mahmoud Farrag
- Integrated Bioscience Program, The University of Akron, Akron, OH 44325, USA.
| | - Nic D Leipzig
- Integrated Bioscience Program, The University of Akron, Akron, OH 44325, USA.
- Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH 44325, USA.
| |
Collapse
|
47
|
Thorpe AA, Bach FC, Tryfonidou MA, Le Maitre CL, Mwale F, Diwan AD, Ito K. Leaping the hurdles in developing regenerative treatments for the intervertebral disc from preclinical to clinical. JOR Spine 2018; 1:e1027. [PMID: 31463447 PMCID: PMC6686834 DOI: 10.1002/jsp2.1027] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/07/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022] Open
Abstract
Chronic back and neck pain is a prevalent disability, often caused by degeneration of the intervertebral disc. Because current treatments for this condition are less than satisfactory, a great deal of effort is being applied to develop new solutions, including regenerative strategies. However, the path from initial promising idea to clinical use is fraught with many hurdles to overcome. Many of the keys to success are not necessarily linked to science or innovation. Successful translation to clinic will also rely on planning and awareness of the hurdles. It will be essential to plan your entire path to clinic from the outset and to do this with a multidisciplinary team. Take advice early on regulatory aspects and focus on generating the proof required to satisfy regulatory approval. Scientific demonstration and societal benefits are important, but translation cannot occur without involving commercial parties, which are instrumental to support expensive clinical trials. This will only be possible when intellectual property can be protected sufficiently to support a business model. In this manner, commercial, societal, medical, and scientific partners can work together to ultimately improve patient health. Based on literature surveys and experiences of the co-authors, this opinion paper presents this pathway, highlights the most prominent issues and hopefully will aid in your own translational endeavors.
Collapse
Affiliation(s)
- Abbey A. Thorpe
- Biomolecular Sciences Research CentreSheffield Hallam UniversitySheffieldUK
| | - Frances C. Bach
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Marianna A. Tryfonidou
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | | | - Fackson Mwale
- Department of SurgeryMcGill UniversityMontrealCanada
| | - Ashish D. Diwan
- Spine Service, Department of Orthopaedic SurgerySt. George & Sutherland Clinical School, University of New South WalesSydneyAustralia
| | - Keita Ito
- Orthopaedic Biomechanics Division, Department of Biomedical EngineeringEindhoven University of TechnologyEindhoventhe Netherlands
- Department of OrthopedicsUniversity Medical Centre UtrechtUtrechtthe Netherlands
| |
Collapse
|
48
|
Sun Y, Yan L, Chen S, Pei M. Functionality of decellularized matrix in cartilage regeneration: A comparison of tissue versus cell sources. Acta Biomater 2018; 74:56-73. [PMID: 29702288 PMCID: PMC7307012 DOI: 10.1016/j.actbio.2018.04.048] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 01/12/2023]
Abstract
Increasing evidence indicates that decellularized extracellular matrices (dECMs) derived from cartilage tissues (T-dECMs) or chondrocytes/stem cells (C-dECMs) can support proliferation and chondrogenic differentiation of cartilage-forming cells. However, few review papers compare the differences between these dECMs when they serve as substrates for cartilage regeneration. In this review, after an introduction of cartilage immunogenicity and decellularization methods to prepare T-dECMs and C-dECMs, a comprehensive comparison focuses on the effects of T-dECMs and C-dECMs on proliferation and chondrogenic differentiation of chondrocytes/stem cells in vitro and in vivo. Key factors within dECMs, consisting of microarchitecture characteristics and micromechanical properties as well as retained insoluble and soluble matrix components, are discussed in-depth for potential mechanisms underlying the functionality of these dECMs in regulating chondrogenesis. With this information, we hope to benefit dECM based cartilage engineering and tissue regeneration for future clinical application. STATEMENT OF SIGNIFICANCE The use of decellularized extracellular matrix (dECM) is becoming a promising approach for tissue engineering and regeneration. Compared to dECM derived from cartilage tissue, recently reported dECM from cell sources exhibits a distinct role in cell based cartilage regeneration. In this review paper, for the first time, tissue and cell based dECMs are comprehensively compared for their functionality in cartilage regeneration. This information is expected to provide an update for dECM based cartilage regeneration.
Collapse
Affiliation(s)
- Yu Sun
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Department of Orthopaedics, Orthopaedics Institute, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, China
| | - Lianqi Yan
- Department of Orthopaedics, Orthopaedics Institute, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, China
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan 610083, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
49
|
Yang Y, Lin H, Shen H, Wang B, Lei G, Tuan RS. Mesenchymal stem cell-derived extracellular matrix enhances chondrogenic phenotype of and cartilage formation by encapsulated chondrocytes in vitro and in vivo. Acta Biomater 2018; 69:71-82. [PMID: 29317369 DOI: 10.1016/j.actbio.2017.12.043] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 12/27/2017] [Accepted: 12/29/2017] [Indexed: 01/24/2023]
Abstract
Mesenchymal stem cell derived extracellular matrix (MSC-ECM) is a natural biomaterial with robust bioactivity and good biocompatibility, and has been studied as a scaffold for tissue engineering. In this investigation, we tested the applicability of using decellularized human bone marrow derived MSC-ECM (hBMSC-ECM) as a culture substrate for chondrocyte expansion in vitro, as well as a scaffold for chondrocyte-based cartilage repair. hBMSC-ECM deposited by hBMSCs cultured on tissue culture plastic (TCP) was harvested, and then subjected to a decellularization process to remove hBMSCs. Compared with chondrocytes grown on TCP, chondrocytes seeded onto hBMSC-ECM exhibited significantly increased proliferation rate, and maintained better chondrocytic phenotype than TCP group. After being expanded to the same cell number and placed in high-density micromass cultures, chondrocytes from the ECM group showed better chondrogenic differentiation profile than those from the TCP group. To test cartilage formation ability, composites of hBMSC-ECM impregnated with chondrocytes were subjected to brief trypsin treatment to allow cell-mediated contraction, and folded to form 3-dimensional chondrocyte-impregnated hBMSC-ECM (Cell/ECM constructs). Upon culture in vitro in chondrogenic medium for 21 days, robust cartilage formation was observed in the Cell/ECM constructs. Similarly prepared Cell/ECM constructs were tested in vivo by subcutaneous implantation into SCID mice. Prominent cartilage formation was observed in the implanted Cell/ECM constructs 14 days post-implantation, with higher sGAG deposition compared to controls consisting of chondrocyte cell sheets. Taken together, these findings demonstrate that hBMSC-ECM is a superior culture substrate for chondrocyte expansion and a bioactive matrix potentially applicable for cartilage regeneration in vivo. STATEMENT OF SIGNIFICANCE Current cell-based treatments for focal cartilage defects face challenges, including chondrocyte dedifferentiation, need for xenogenic scaffolds, and suboptimal cartilage formation. We present here a novel technique that utilizes adult stem cell-derived extracellular matrix, as a culture substrate and/or encapsulation scaffold for human adult chondrocytes, for the repair of cartilage defects. Chondrocytes cultured in stem cell-derived matrix showed higher proliferation, better chondrocytic phenotype, and improved redifferentiation ability upon in vitro culture expansion. Most importantly, 3-dimensional constructs formed from chondrocytes folded within stem cell matrix manifested excellent cartilage formation both in vitro and in vivo. These findings demonstrate the suitability of stem cell-derived extracellular matrix as a culture substrate for chondrocyte expansion as well as a candidate bioactive matrix for cartilage regeneration.
Collapse
Affiliation(s)
- Yuanheng Yang
- Department of Orthopaedic Surgery, Xiangya hospital, Central South University, Changsha, Hunan, China; Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; The Third Xiangya hospital, Central South University, Changsha, Hunan, China
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - He Shen
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-tech and Nano-bionics, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Bing Wang
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Guanghua Lei
- Department of Orthopaedic Surgery, Xiangya hospital, Central South University, Changsha, Hunan, China.
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
50
|
The Hypoxia-Mimetic Agent Cobalt Chloride Differently Affects Human Mesenchymal Stem Cells in Their Chondrogenic Potential. Stem Cells Int 2018; 2018:3237253. [PMID: 29731777 PMCID: PMC5872594 DOI: 10.1155/2018/3237253] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 12/10/2017] [Accepted: 01/01/2018] [Indexed: 12/19/2022] Open
Abstract
Adult stem cells are a promising cell source for cartilage regeneration. They resided in a special microenvironment known as the stem-cell niche, characterized by the presence of low oxygen concentration. Cobalt chloride (CoCl2) imitates hypoxia in vitro by stabilizing hypoxia-inducible factor-alpha (HIF-1α), which is the master regulator in the cellular adaptive response to hypoxia. In this study, the influence of CoCl2 on the chondrogenic potential of human MSCs, isolated from dental pulp, umbilical cord, and adipose tissue, was investigated. Cells were treated with concentrations of CoCl2 ranging from 50 to 400 μM. Cell viability, HIF-1α protein synthesis, and the expression of the chondrogenic markers were analyzed. The results showed that the CoCl2 supplementation had no effect on cell viability, while the upregulation of chondrogenic markers such as SOX9, COL2A1, VCAN, and ACAN was dependent on the cellular source. This study shows that hypoxia, induced by CoCl2 treatment, can differently influence the behavior of MSCs, isolated from different sources, in their chondrogenic potential. These findings should be taken into consideration in the treatment of cartilage repair and regeneration based on stem cell therapies.
Collapse
|