1
|
Lu J, Zhang W, Zhu Y, Luo P, Tong X, Xie S, Jiang L, Guo X, Huang J, Gu M, Ding X, Xian S, Huang R, Ji S, Xia Z. Revealing the Therapeutic Potential of Stem Cells in Burn Healing: A Deeper Understanding of the Therapeutic Mechanisms of Epidermal Stem Cells and Mesenchymal Stem Cells. Stem Cells Int 2024; 2024:1914585. [PMID: 39717868 PMCID: PMC11666318 DOI: 10.1155/2024/1914585] [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: 09/14/2023] [Revised: 07/20/2024] [Accepted: 08/19/2024] [Indexed: 12/25/2024] Open
Abstract
Background: Burns are a global public health issue and a major cause of disability and death around the world. Stem cells, which are the undifferentiated cells with the potential for indefinite proliferation and multilineage differentiation, have the ability to replace injured skin and facilitate the wound repair process through paracrine mechanisms. In light of this, the present study aims to conduct a bibliometric analysis in order to identify research hotspots of stem cell-related burns and assess global research tendencies. Methods: To achieve this objective, we retrieved scientific publications on burns associated with stem cells covering the period from January 1, 1978, to October 13, 2022, from the Web of Science Core Collection (WoSCC). Bibliometric analyses, including production and collaboration analyses between countries, institutions, authors, and journals, as well as keyword and topic analyses, were conducted using the bibliometrix R package, CiteSpace, and VOSviewer. Results: A total of 1648 burns associated with stem cell documents were published and listed on WOSCC. The most contributive country, institution, journal, and author were the United States, LV Prasad Eye Institute, Burns, and Scheffer C.G. Tseng, respectively. More importantly, combined with historical direct citation network, trend topic analysis, keyword co-occurrence network, and substantial literature analysis, we eventually summarized the research hotspots and frontiers on burns associated stem cell reasearch. Conclusion: The present study obtained deep insight into the developing trends and research hotspots on burns associated with stem cells, which arouses growing concerns and implies increasing clinical implications. The mechanism and therapeutics of epidermal stem cells (ESCs) for burn wounds and the mechanism of mesenchymal stem cells (MSCs) and MSC-derived exosomes for burns wounds are two research hotspots in this field.
Collapse
Affiliation(s)
- Jianyu Lu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Wei Zhang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Yushu Zhu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Pengfei Luo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Xirui Tong
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Sujie Xie
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Luofeng Jiang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Xinya Guo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Jie Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Minyi Gu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Xinran Ding
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Shuyuan Xian
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Zhaofan Xia
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| |
Collapse
|
2
|
Wu X, Wang K, Chen H, Cao B, Wang Y, Wang Z, Dai C, Yao M, Ji X, Jiang X, Zhang W, Pan Z, Xue D. Hypoxia-induced mitochondrial fission regulates the fate of bone marrow mesenchymal stem cells by maintaining HIF1α stabilization. Free Radic Biol Med 2024; 225:127-144. [PMID: 39366470 DOI: 10.1016/j.freeradbiomed.2024.10.256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/19/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
For mesenchymal stem cells derived from bone marrow, a controlled reduction in ambient oxygen concentration has been recognized as a facilitator of osteogenic differentiation and the formation of calcium nodules. However, the specific molecular mechanisms underlying this phenotype remain unclear. The aim of this study was to elucidate the impact of hypoxia on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and to explore the involvement of mitophagy and the regulation of mitochondrial dynamics mediated by the mitochondrial dynamic regulatory factor FUN14 domain-containing 1 (FUNDC1). Our findings suggest that FUNDC1 is required for promoting osteogenic differentiation in BMSCs under hypoxic conditions. However, this effect was not dependent on FUNDC1-mediated mitophagy but rather on FUNDC1-mediated regulation of mitochondrial fission. At the mechanistic level, FUNDC1 binds more DNM1L and less OPA1 under hypoxic conditions, leading to an upsurge in mitochondrial division. This heightened mitochondrial division culminates in the increased translocation of Parkin to mitochondria, diminishing its interactions with HIF1α in the cytoplasm and consequently facilitating HIF1α deubiquitination and stabilization. In summary, FUNDC1-regulated mitochondrial division in hypoxic culture emerges as a critical determinant for the translocation of Parkin to mitochondria, ultimately maintaining HIF1α stabilization and promoting osteogenic differentiation.
Collapse
Affiliation(s)
- Xiaoyong Wu
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Kanbin Wang
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Hongyu Chen
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Binhao Cao
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Yibo Wang
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China; Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhongxiang Wang
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Chengxin Dai
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Minjun Yao
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Xiaoxiao Ji
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Xiaowen Jiang
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Weijun Zhang
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
| | - Zhijun Pan
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China.
| | - Deting Xue
- Department of Orthopedic Surgery of the Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou, 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China.
| |
Collapse
|
3
|
Li Y, Jin M, Guo D, Shen S, Lu K, Pan R, Sun L, Zhang H, Shao J, Pan G. Unveiling the immunogenicity of allogeneic mesenchymal stromal cells: Challenges and strategies for enhanced therapeutic efficacy. Biomed Pharmacother 2024; 180:117537. [PMID: 39405918 DOI: 10.1016/j.biopha.2024.117537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 11/14/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) exhibit significant potential in the context of cell therapy because of their capacity to perform a range of interconnected functions in damaged tissues, including immune modulation, hematopoietic support, and tissue regeneration. MSCs are hypoimmunogenic because of their diminished expression of major histocompatibility molecules, absence of costimulatory molecules, and presence of coinhibitory molecules. While autologous MSCs reduce the risk of rejection and infection, variability in cell numbers and proliferation limits their potential applications. Conversely, allogeneic MSCs (allo-MSCs) possess broad clinical applications unconstrained by donor physiology. Nonetheless, preclinical and clinical investigations highlight that transplanted allo-MSCs are subject to immune attack from recipients. These cells exhibit anti-inflammatory and proinflammatory phenotypes contingent on the microenvironment. Notably, the proinflammatory phenotype features enhanced immunogenicity and diminished immunosuppression, potentially triggering allogeneic immune reactions that impede long-term clinical efficacy. Consequently, preserving the low immunogenicity of allo-MSCs in vivo and mitigating immune rejection in diverse microenvironments represent crucial challenges for the widespread clinical application of MSCs. In this review, we elucidate the immune regulation of allo-MSCs, specifically focusing on two distinct subgroups, MSC1 and MSC2, that exhibit varying polarization states and immunogenicity. We discuss the factors and underlying mechanisms that induce MSC immunogenicity and polarization, highlighting the crucial role of major histocompatibility complex class I/II molecules in rejection post-transplantation. Additionally, we summarize the immunogenic regulatory targets and applications of allo-MSCs and outline strategies to address challenges in this promising field, aiming to enhance allo-MSC therapeutic efficacy for patients.
Collapse
Affiliation(s)
- Yuanhui Li
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Mengting Jin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Dongyang Guo
- Hangzhou City University, School of Medicine, 50 Huzhou Street, Hangzhou, China
| | - Shuang Shen
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Kaining Lu
- Breast Disease Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ruolang Pan
- Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
| | - Li Sun
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Hongchen Zhang
- Department of Gatroenterology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, No. 261 HuanSha Road, Hangzhou, China.
| | - Jianzhong Shao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Gang Pan
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China.
| |
Collapse
|
4
|
Re F, Sartore L, Pasini C, Ferroni M, Borsani E, Pandini S, Bianchetti A, Almici C, Giugno L, Bresciani R, Mutti S, Trenta F, Bernardi S, Farina M, Russo D. In Vitro Biocompatibility Assessment of Bioengineered PLA-Hydrogel Core-Shell Scaffolds with Mesenchymal Stromal Cells for Bone Regeneration. J Funct Biomater 2024; 15:217. [PMID: 39194655 DOI: 10.3390/jfb15080217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Human mesenchymal stromal cells (hMSCs), whether used alone or together with three-dimensional scaffolds, are the best-studied postnatal stem cells in regenerative medicine. In this study, innovative composite scaffolds consisting of a core-shell architecture were seeded with bone-marrow-derived hMSCs (BM-hMSCs) and tested for their biocompatibility and remarkable capacity to promote and support bone regeneration and mineralization. The scaffolds were prepared by grafting three different amounts of gelatin-chitosan (CH) hydrogel into a 3D-printed polylactic acid (PLA) core (PLA-CH), and the mechanical and degradation properties were analyzed. The BM-hMSCs were cultured in the scaffolds with the presence of growth medium (GM) or osteogenic medium (OM) with differentiation stimuli in combination with fetal bovine serum (FBS) or human platelet lysate (hPL). The primary objective was to determine the viability, proliferation, morphology, and spreading capacity of BM-hMSCs within the scaffolds, thereby confirming their biocompatibility. Secondly, the BM-hMSCs were shown to differentiate into osteoblasts and to facilitate scaffold mineralization. This was evinced by a positive Von Kossa result, the modulation of differentiation markers (osteocalcin and osteopontin), an expression of a marker of extracellular matrix remodeling (bone morphogenetic protein-2), and collagen I. The results of the energy-dispersive X-ray analysis (EDS) clearly demonstrate the presence of calcium and phosphorus in the samples that were incubated in OM, in the presence of FBS and hPL, but not in GM. The chemical distribution maps of calcium and phosphorus indicate that these elements are co-localized in the same areas of the sections, demonstrating the formation of hydroxyapatite. In conclusion, our findings show that the combination of BM-hMSCs and PLA-CH, regardless of the amount of hydrogel content, in the presence of differentiation stimuli, can provide a construct with enhanced osteogenicity for clinically relevant bone regeneration.
Collapse
Affiliation(s)
- Federica Re
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
| | - Luciana Sartore
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Materials Science and Technology Laboratory, Department of Mechanical and Industrial Engineering, University of Brescia, 25123 Brescia, Italy
| | - Chiara Pasini
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Materials Science and Technology Laboratory, Department of Mechanical and Industrial Engineering, University of Brescia, 25123 Brescia, Italy
| | - Matteo Ferroni
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Department of Civil, Environmental, Architectural Engineering and Mathematics (DICATAM), University of Brescia, Via Valotti 9, 25123 Brescia, Italy
- National Research Council (CNR)-Institute for Microelectronics and Microsystems, Via Gobetti 101, 40129 Bologna, Italy
| | - Elisa Borsani
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
- Interdepartmental University Center of Research "Adaption and Regeneration of Tissues and Organs (ARTO)", University of Brescia, 25123 Brescia, Italy
| | - Stefano Pandini
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Materials Science and Technology Laboratory, Department of Mechanical and Industrial Engineering, University of Brescia, 25123 Brescia, Italy
| | - Andrea Bianchetti
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Laboratory for Stem Cells Manipulation and Cryopreservation, Department of Transfusion Medicine, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Camillo Almici
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- Laboratory for Stem Cells Manipulation and Cryopreservation, Department of Transfusion Medicine, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Lorena Giugno
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
| | - Roberto Bresciani
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Highly Specialized Laboratory, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Silvia Mutti
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
| | - Federica Trenta
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
| | - Simona Bernardi
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
- National Center for Gene Therapy and Drugs based on RNA Technology-CN3, 35122 Padua, Italy
| | - Mirko Farina
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
| | - Domenico Russo
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- University Center of Research "STem cells, bioENgineering and regenerative MEDicine"-STENMED, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
5
|
Hao M, Xue L, Wen X, Sun L, Zhang L, Xing K, Hu X, Xu J, Xing D. Advancing bone regeneration: Unveiling the potential of 3D cell models in the evaluation of bone regenerative materials. Acta Biomater 2024; 183:1-29. [PMID: 38815683 DOI: 10.1016/j.actbio.2024.05.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Bone, a rigid yet regenerative tissue, has garnered extensive attention for its impressive healing abilities. Despite advancements in understanding bone repair and creating treatments for bone injuries, handling nonunions and large defects remains a major challenge in orthopedics. The rise of bone regenerative materials is transforming the approach to bone repair, offering innovative solutions for nonunions and significant defects, and thus reshaping orthopedic care. Evaluating these materials effectively is key to advancing bone tissue regeneration, especially in difficult healing scenarios, making it a critical research area. Traditional evaluation methods, including two-dimensional cell models and animal models, have limitations in predicting accurately. This has led to exploring alternative methods, like 3D cell models, which provide fresh perspectives for assessing bone materials' regenerative potential. This paper discusses various techniques for constructing 3D cell models, their pros and cons, and crucial factors to consider when using these models to evaluate bone regenerative materials. We also highlight the significance of 3D cell models in the in vitro assessments of these materials, discuss their current drawbacks and limitations, and suggest future research directions. STATEMENT OF SIGNIFICANCE: This work addresses the challenge of evaluating bone regenerative materials (BRMs) crucial for bone tissue engineering. It explores the emerging role of 3D cell models as superior alternatives to traditional methods for assessing these materials. By dissecting the construction, key factors of evaluating, advantages, limitations, and practical considerations of 3D cell models, the paper elucidates their significance in overcoming current evaluation method shortcomings. It highlights how these models offer a more physiologically relevant and ethically preferable platform for the precise assessment of BRMs. This contribution is particularly significant for "Acta Biomaterialia" readership, as it not only synthesizes current knowledge but also propels the discourse forward in the search for advanced solutions in bone tissue engineering and regeneration.
Collapse
Affiliation(s)
- Minglu Hao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Linyuan Xue
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Xiaobo Wen
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Li Sun
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario N2L3G1, Canada
| | - Kunyue Xing
- Alliance Manchester Business School, The University of Manchester, Manchester M139PL, UK
| | - Xiaokun Hu
- Department of Interventional Medical Center, Affiliated Hospital of Qingdao University, Qingdao 26600, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
6
|
Yang H, Wang W, Liu H, Zhang C, Cao Y, Long L, Han X, Wang Y, Yan F, Li G, Zhu M, Jin L, Fan Z. miR615-3p inhibited FBLN1 and osteogenic differentiation of umbilical cord mesenchymal stem cells by associated with YTHDF2 in a m 6A-miRNA interaction manner. Cell Prolif 2024; 57:e13607. [PMID: 38353178 PMCID: PMC11150146 DOI: 10.1111/cpr.13607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/09/2024] [Accepted: 01/27/2024] [Indexed: 06/06/2024] Open
Abstract
To investigate the role and mechanism of FBLN1 in the osteogenic differentiation and bone regeneration by using umbilical cord mesenchymal stem cells (WJCMSCs). We found that FBLN1 promoted osteogenic differentiation of WJCMSCs and WJCMSC-mediated bone regeneration. It was showed that there was an m6A methylation site in 3'UTR of FBLN1 mRNA, and the mutation of the m6A site enhanced the stability of FBLN1 mRNA, subsequently fostering the FBLN1 enhanced osteogenic differentiation of WJCMSCs. YTHDF2 was identified as capable of recognizing and binding to the m6A site, consequently inducing FBLN1 instability and repressed the osteogenic differentiation of WJCMSCs. Meanwhile, miR-615-3p negatively regulated FBLN1 by binding FBLN1 3'UTR and inhibited the osteogenic differentiation of WJCMSCs and WJCMSC-mediated bone regeneration. Then, we discovered miR-615-3p was found to regulate the functions of FBLN1 facilitated by YTHDF2 through an m6A-miRNA regulation mechanism. We demonstrated that FBLN1 is critical for regulating the osteogenic differentiation potentials of WJCMSCs and have identified that miR615-3p mediated the decay of FBLN1 mRNA which facilitated by m6A reading protein YTHDF2. This provided a novel m6A-miRNA epigenetic regulatory pattern for MSC regulation and bone regeneration.
Collapse
Affiliation(s)
- Haoqing Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of StomatologyBeijingChina
| | - Wanqing Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of StomatologyBeijingChina
| | - Huina Liu
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of StomatologyBeijingChina
| | - Chen Zhang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of StomatologyBeijingChina
| | - Yangyang Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of StomatologyBeijingChina
| | - Lujue Long
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of StomatologyBeijingChina
| | - Xiao Han
- Jiangsu Province Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingChina
| | - Yuejun Wang
- Department of General Dentistry and Integrated Emergency Dental Care, Beijing Stomatological HospitalCapital Medical UniversityBeijingChina
| | - Fei Yan
- Xiangya Stomatological Hospital and School of StomatologyCentral South UniversityChangshaChina
| | - Guoqing Li
- Jiangsu Province Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingChina
| | - Mengyuan Zhu
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of StomatologyBeijingChina
| | - Luyuan Jin
- Department of General Dentistry and Integrated Emergency Dental Care, Beijing Stomatological HospitalCapital Medical UniversityBeijingChina
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of StomatologyBeijingChina
- Beijing Laboratory of Oral HealthCapital Medical UniversityBeijingChina
- Research Unit of Tooth Development and RegenerationChinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
7
|
Li C, Sun Y, Xu W, Chang F, Wang Y, Ding J. Mesenchymal Stem Cells-Involved Strategies for Rheumatoid Arthritis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305116. [PMID: 38477559 PMCID: PMC11200100 DOI: 10.1002/advs.202305116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/13/2023] [Indexed: 03/14/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of the joints and bone destruction. Because of systemic administration and poor targeting, traditional anti-rheumatic drugs have unsatisfactory treatment efficacy and strong side effects, including myelosuppression, liver or kidney function damage, and malignant tumors. Consequently, mesenchymal stem cells (MSCs)-involved therapy is proposed for RA therapy as a benefit of their immunosuppressive and tissue-repairing effects. This review summarizes the progress of MSCs-involved RA therapy through suppressing inflammation and promoting tissue regeneration and predicts their potential clinical application.
Collapse
Affiliation(s)
- Chaoyang Li
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Yifu Sun
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
| | - Weiguo Xu
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Fei Chang
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
| | - Yinan Wang
- Department of BiobankDivision of Clinical ResearchThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of EducationThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
8
|
Qin W, Xing T, Qin S, Tang B, Chen W. BMSCs-driven graphite oxide-grafted-carbon fibers reinforced polyetheretherketone composites as functional implants: in vivo biosafety and osteogenesis. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:1343-1358. [PMID: 38493406 DOI: 10.1080/09205063.2024.2328877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 01/08/2024] [Indexed: 03/18/2024]
Abstract
Mesenchymal stem cells (MSCs) are increasingly becoming a potential treatment approach for bone injuries due to the multi-lineage differentiation potential, ability to recognize damaged tissue sites and secrete bioactive factors that can enhance tissue repair. The aim of this work was to improve osteogenesis of carbon fibers reinforced polyetheretherketone (CF/PEEK) implants through bone marrow mesenchymal stem cells (BMSCs)-based therapy. Moreover, bioactive graphene oxide (GO) was introduced into CF/PEEK by grafting GO onto CF to boost the osteogenic efficiency of BMSCs. Subsequently, CF/PEEK was implanted into the symmetrical skull defect models of SD rats. Then in vivo biosafety and osteogenesis were evaluated. The results indicated that surface wettability of CF/PEEK was effectively improved by GO, which was beneficial for the adhesion of BMSCs. The pathological tissue sections stained with H&E showed no significant pathological change in the main organs including heart, liver, spleen, lung and kidney, which indicated no acute systemic toxicity. Furthermore, bone mineralization deposition rate of CF/PEEK containing GO was 2.2 times that of pure CF/PEEK. The X-ray test showed that the surface of CF/PEEK containing GO was obviously covered by more newly formed bone tissue than pure CF/PEEK after 8 weeks of implantation. This work demonstrated that GO effectively enhanced surface bioactivity of CF/PEEK and assisted BMSCs in accelerating differentiation into bone tissue, providing a feasible strategy for improving osteogenesis of PEEK and CF/PEEK.
Collapse
Affiliation(s)
- Wen Qin
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Tong Xing
- Engineering Research Center of Heavy Mechanical, Ministry of Education, Taiyuan University of Science and Technology, Taiyuan, China
| | - Shengnan Qin
- Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Bin Tang
- College of Material Science and Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Weiyi Chen
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, China
| |
Collapse
|
9
|
Han H, Zhou Z, Shang T, Li S, Shen X, Fang J, Cui L. Silk Fibroin-Laponite Porous Microspheres as Cell Microcarriers for Osteogenic Differentiation. Tissue Eng Part A 2024. [PMID: 38666700 DOI: 10.1089/ten.tea.2024.0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2024] Open
Abstract
Silk fibroin (SF) has garnered significant attention as a natural polymer for fabricating porous scaffolds in various engineering applications. However, the limited osteoinductive property of SF has hindered its efficacy in bone repair applications. In this study, we constructed an SF-based injectable porous microcarrier that is doped with laponite (LAP), containing magnesium ions (Mg2+). The influence of freezing temperatures and concentrations of SF and LAP on the structural parameters of SF-LAP microcarriers was investigated. The SF-LAP microcarrier exhibited a porosity of 76.7 ± 1.2% and a controlled pore size of 24.6 ± 4.0 μm. At the 6 weeks of in vitro degradation test, a mild alkaline level in culture medium containing SF-LAP microcarriers was detected. The release of Mg2+ from the SF-LAP microcarrier was maintained at a concentration within the range of 1.2-2.3 mM during the 6 weeks. The seeded human adipose-derived stem cells in the SF-LAP microcarrier demonstrated a significant enhancement in osteogenic differentiation compared with cells seeded in the pure SF microcarrier, as evidenced by quantitative alkaline phosphatase activity and the expression of osteogenic marker genes. These findings underscore the potential of the SF-LAP microcarrier as an ideal cell carrier in the treatment of bone defects.
Collapse
Affiliation(s)
- Haotian Han
- Department of Plastic and Cosmetic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Zhihua Zhou
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of the Ministry of Education, Hunan Provincial Key Laboratory of Controllable Preparation and Functional Application of Fine Polymers, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, China
| | - Ting Shang
- Department of Plastic and Cosmetic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shuaijun Li
- Department of Reconstructive and Regenerative Surgery, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Stem Cells and Regenerative Medicine, Tongji University School of Medicine, Shanghai, China
| | - Xiang Shen
- Department of Orthopedics, The Fourth Hospital of Changsha, Changsha, China
| | - Jianjun Fang
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule of the Ministry of Education, Hunan Provincial Key Laboratory of Controllable Preparation and Functional Application of Fine Polymers, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, China
| | - Lei Cui
- Department of Plastic and Cosmetic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of Reconstructive and Regenerative Surgery, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Stem Cells and Regenerative Medicine, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Cheng Y, Zhu Y, Liu Y, Liu X, Ding Y, Li D, Zhang X, Liu Y. Tailored apoptotic vesicles promote bone regeneration by releasing the osteoinductive brake. Int J Oral Sci 2024; 16:31. [PMID: 38627392 PMCID: PMC11021547 DOI: 10.1038/s41368-024-00293-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 04/19/2024] Open
Abstract
Accumulating evidence has demonstrated that apoptotic vesicles (apoVs) derived from mesenchymal stem cells (MSCs; MSC-apoVs) are vital for bone regeneration, and possess superior capabilities compared to MSCs and other extracellular vesicles derived from MSCs (such as exosomes). The osteoinductive effect of MSC-apoVs is attributed to their diverse contents, especially enriched proteins or microRNAs (miRNAs). To optimize their osteoinduction activity, it is necessary to determine the unique cargo profiles of MSC-apoVs. We previously established the protein landscape and identified proteins specific to MSC-apoVs. However, the features and functions of miRNAs enriched in MSC-apoVs are unclear. In this study, we compared MSCs, MSC-apoVs, and MSC-exosomes from two types of MSC. We generated a map of miRNAs specific to MSC-apoVs and identified seven miRNAs specifically enriched in MSC-apoVs compared to MSCs and MSC-exosomes, which we classified as apoV-specific miRNAs. Among these seven specific miRNAs, hsa-miR-4485-3p was the most abundant and stable. Next, we explored its function in apoV-mediated osteoinduction. Unexpectedly, hsa-miR-4485-3p enriched in MSC-apoVs inhibited osteogenesis and promoted adipogenesis by targeting the AKT pathway. Tailored apoVs with downregulated hsa-miR-4485-3p exhibited a greater effect on bone regeneration than control apoVs. Like releasing the brake, we acquired more powerful osteoinductive apoVs. In summary, we identified the miRNA cargos, including miRNAs specific to MSC-apoVs, and generated tailored apoVs with high osteoinduction activity, which is promising in apoV-based therapies for bone regeneration.
Collapse
Affiliation(s)
- Yawen Cheng
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
- Second Clinical Division, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yuan Zhu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yaoshan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Xuenan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yanan Ding
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Deli Li
- Second Clinical Division, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China.
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Beijing, China.
| |
Collapse
|
11
|
Linke P, Munding N, Kimmle E, Kaufmann S, Hayashi K, Nakahata M, Takashima Y, Sano M, Bastmeyer M, Holstein T, Dietrich S, Müller‐Tidow C, Harada A, Ho AD, Tanaka M. Reversible Host-Guest Crosslinks in Supramolecular Hydrogels for On-Demand Mechanical Stimulation of Human Mesenchymal Stem Cells. Adv Healthc Mater 2024; 13:e2302607. [PMID: 38118064 PMCID: PMC11481031 DOI: 10.1002/adhm.202302607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/12/2023] [Indexed: 12/22/2023]
Abstract
Stem cells are regulated not only by biochemical signals but also by biophysical properties of extracellular matrix (ECM). The ECM is constantly monitored and remodeled because the fate of stem cells can be misdirected when the mechanical interaction between cells and ECM is imbalanced. A well-defined ECM model for bone marrow-derived human mesenchymal stem cells (hMSCs) based on supramolecular hydrogels containing reversible host-guest crosslinks is fabricated. The stiffness (Young's modulus E) of the hydrogels can be switched reversibly by altering the concentration of non-cytotoxic, free guest molecules dissolved in the culture medium. Fine-adjustment of substrate stiffness enables the authors to determine the critical stiffness level E* at which hMSCs turn the mechano-sensory machinery on or off. Next, the substrate stiffness across E* is switched and the dynamic adaptation characteristics such as morphology, traction force, and YAP/TAZ signaling of hMSCs are monitored. These data demonstrate the instantaneous switching of traction force, which is followed by YAP/TAZ signaling and morphological adaptation. Periodical switching of the substrate stiffness across E* proves that frequent applications of mechanical stimuli drastically suppress hMSC proliferation. Mechanical stimulation across E* level using dynamic hydrogels is a promising strategy for the on-demand control of hMSC transcription and proliferation.
Collapse
Affiliation(s)
- Philipp Linke
- Physical Chemistry of BiosystemsInstitute of Physical ChemistryHeidelberg University69120HeidelbergGermany
| | - Natalie Munding
- Physical Chemistry of BiosystemsInstitute of Physical ChemistryHeidelberg University69120HeidelbergGermany
| | - Esther Kimmle
- Physical Chemistry of BiosystemsInstitute of Physical ChemistryHeidelberg University69120HeidelbergGermany
| | - Stefan Kaufmann
- Physical Chemistry of BiosystemsInstitute of Physical ChemistryHeidelberg University69120HeidelbergGermany
| | - Kentaro Hayashi
- Center for Integrative Medicine and PhysicsInstitute for Advanced StudyKyoto UniversityKyoto606‐8501Japan
| | - Masaki Nakahata
- Department of Macromolecular ScienceGraduate School of ScienceOsaka UniversityOsaka560‐0043Japan
| | - Yoshinori Takashima
- Department of Macromolecular ScienceGraduate School of ScienceOsaka UniversityOsaka560‐0043Japan
| | - Masaki Sano
- Institute of Natural SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Martin Bastmeyer
- Center for Integrative Medicine and PhysicsInstitute for Advanced StudyKyoto UniversityKyoto606‐8501Japan
- Cell and NeurobiologyZoological InstituteKarlsruhe Institute of Technology76131KarlsruheGermany
- Institute for Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP)Karlsruhe Institute of Technology76334Eggenstein‐LeopoldshafenGermany
| | - Thomas Holstein
- Center for Integrative Medicine and PhysicsInstitute for Advanced StudyKyoto UniversityKyoto606‐8501Japan
- Molecular Genetics and EvolutionCentre for Organismal StudiesHeidelberg University69221HeidelbergGermany
| | - Sascha Dietrich
- Department of Internal Medicine VHematology, Oncology, RheumatologyUniversity Hospital Heidelberg69120HeidelbergGermany
- Department of Haematology, Oncology, and Clinical ImmunologyUniversitätsklinikum Düsseldorf40225DüsseldorfGermany
| | - Carsten Müller‐Tidow
- Department of Internal Medicine VHematology, Oncology, RheumatologyUniversity Hospital Heidelberg69120HeidelbergGermany
| | - Akira Harada
- The Institute of Scientific and Industrial ResearchOsaka University8‐1 MihogaokaIbarakiOsaka567‐0047Japan
| | - Anthony D. Ho
- Center for Integrative Medicine and PhysicsInstitute for Advanced StudyKyoto UniversityKyoto606‐8501Japan
- Department of Internal Medicine VHematology, Oncology, RheumatologyUniversity Hospital Heidelberg69120HeidelbergGermany
- Molecular Medicine Partnership Unit HeidelbergEMBL and Heidelberg University69120HeidelbergGermany
| | - Motomu Tanaka
- Physical Chemistry of BiosystemsInstitute of Physical ChemistryHeidelberg University69120HeidelbergGermany
- Center for Integrative Medicine and PhysicsInstitute for Advanced StudyKyoto UniversityKyoto606‐8501Japan
| |
Collapse
|
12
|
He Z, Li H, Zhang Y, Gao S, Liang K, Su Y, Du Y, Wang D, Xing D, Yang Z, Lin J. Enhanced bone regeneration via endochondral ossification using Exendin-4-modified mesenchymal stem cells. Bioact Mater 2024; 34:98-111. [PMID: 38186959 PMCID: PMC10770633 DOI: 10.1016/j.bioactmat.2023.12.007] [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: 09/07/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 01/09/2024] Open
Abstract
Nonunions and delayed unions pose significant challenges in orthopedic treatment, with current therapies often proving inadequate. Bone tissue engineering (BTE), particularly through endochondral ossification (ECO), emerges as a promising strategy for addressing critical bone defects. This study introduces mesenchymal stem cells overexpressing Exendin-4 (MSC-E4), designed to modulate bone remodeling via their autocrine and paracrine functions. We established a type I collagen (Col-I) sponge-based in vitro model that effectively recapitulates the ECO pathway. MSC-E4 demonstrated superior chondrogenic and hypertrophic differentiation and enhanced the ECO cell fate in single-cell sequencing analysis. Furthermore, MSC-E4 encapsulated in microscaffold, effectively facilitated bone regeneration in a rat calvarial defect model, underscoring its potential as a therapeutic agent for bone regeneration. Our findings advocate for MSC-E4 within a BTE framework as a novel and potent approach for treating significant bone defects, leveraging the intrinsic ECO process.
Collapse
Affiliation(s)
- Zihao He
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Hui Li
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Yuanyuan Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Shuang Gao
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Yiqi Su
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Du Wang
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Zhen Yang
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Jianhao Lin
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| |
Collapse
|
13
|
Yu L, Dou G, Kuang H, Bao L, Liu H, Ye Q, Wang Z, Yang X, Ren L, Li Z, Liu H, Li B, Liu S, Ge S, Liu S. Apoptotic Extracellular Vesicles Induced Endothelial Cell-Mediated Autologous Stem Cell Recruitment Dominates Allogeneic Stem Cell Therapeutic Mechanism for Bone Repair. ACS NANO 2024; 18:8718-8732. [PMID: 38465955 DOI: 10.1021/acsnano.3c11050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Although stem cell therapy is proved to be a promising strategy for bone repair and regeneration, transplanted allogeneic stem cells generally suffer from unfavorable apoptosis instead of differentiation into osteocytes. How the apoptotic stem cells promote bone regeneration still needs to be uncovered. In this work, we found that apoptotic extracellular vesicles released by allogeneic stem cells are critical mediators for promoting bone regeneration. Based on the results of in vivo experiments, a mechanism of apoptotic stem cells determined autologous stem cell recruitment and enhance osteogenesis was proposed. The nanoscaled apoptotic extracellular vesicles released from transplanted stem cells were endocytosed by vascular endothelial cells and preferentially distribute at endoplasmic reticular region. The oxidized phosphatidylcholine enriched in the vesicles activated the endoplasmic reticulum stress and triggered the reflective elevation of adhesion molecules, which induced the recruitment of autologous stem cells located in the blood vessels, transported them into the defect region, and promoted osteogenesis and bone repair. These findings not only reveal the mechanism of stem cell therapy of bone defects but also provide a cue for investigation of the biological process of stem cell therapy for other diseases and develop stem cell therapeutic strategies.
Collapse
Affiliation(s)
- Lu Yu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong 250012, China
| | - Geng Dou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Huijuan Kuang
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Lili Bao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Huan Liu
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Qingyuan Ye
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Digital Dentistry Center, School of Stomatology, The Fourth Military Medical University, Shaanxi 710032, China
| | - Zhengyan Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong 250012, China
| | - Xiaoshan Yang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Lili Ren
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zihan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China
| | - Bei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Siying Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Shaohua Ge
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong 250012, China
| | - Shiyu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
14
|
Hoveidaei AH, Sadat-Shojai M, Mosalamiaghili S, Salarikia SR, Roghani-Shahraki H, Ghaderpanah R, Ersi MH, Conway JD. Nano-hydroxyapatite structures for bone regenerative medicine: Cell-material interaction. Bone 2024; 179:116956. [PMID: 37951520 DOI: 10.1016/j.bone.2023.116956] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/04/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
Bone tissue engineering holds great promise for the regeneration of damaged or severe bone defects. However, several challenges hinder its translation into clinical practice. To address these challenges, interdisciplinary efforts and advances in biomaterials, cell biology, and bioengineering are required. In recent years, nano-hydroxyapatite (nHA)-based scaffolds have emerged as a promising approach for the development of bone regenerative agents. The unique similarity of nHA with minerals found in natural bones promotes remineralization and stimulates bone growth, which are crucial factors for efficient bone regeneration. Moreover, nHA exhibits desirable properties, such as strong chemical interactions with bone and facilitation of tissue growth, without inducing inflammation or toxicity. It also promotes osteoblast survival, adhesion, and proliferation, as well as increasing alkaline phosphatase activity, osteogenic differentiation, and bone-specific gene expression. However, it is important to note that the effect of nHA on osteoblast behavior is dose-dependent, with cytotoxic effects observed at higher doses. Additionally, the particle size of nHA plays a crucial role, with smaller particles having a more significant impact. Therefore, in this review, we highlighted the potential of nHA for improving bone regeneration processes and summarized the available data on bone cell response to nHA-based scaffolds. In addition, an attempt is made to portray the current status of bone tissue engineering using nHA/polymer hybrids and some recent scientific research in the field.
Collapse
Affiliation(s)
- Amir Human Hoveidaei
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Mehdi Sadat-Shojai
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz, Iran
| | - Seyedarad Mosalamiaghili
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Rezvan Ghaderpanah
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hamed Ersi
- Evidence Based Medicine Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Clinical Research Development Center of Shahid Mohammadi Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Janet D Conway
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA.
| |
Collapse
|
15
|
Bury MI, Fuller NJ, Wang X, Chan YY, Sturm RM, Oh SS, Sofer LA, Arora HC, Sharma TT, Nolan BG, Feng W, Rabizadeh RR, Barac M, Edassery SS, Goedegebuure MM, Wang LW, Ganesh B, Halliday LC, Seniw ME, Edassery SL, Mahmud NB, Hofer MD, McKenna KE, Cheng EY, Ameer GA, Sharma AK. Multipotent bone marrow cell-seeded polymeric composites drive long-term, definitive urinary bladder tissue regeneration. PNAS NEXUS 2024; 3:pgae038. [PMID: 38344009 PMCID: PMC10855019 DOI: 10.1093/pnasnexus/pgae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/16/2024] [Indexed: 03/02/2024]
Abstract
To date, there are no efficacious translational solutions for end-stage urinary bladder dysfunction. Current surgical strategies, including urinary diversion and bladder augmentation enterocystoplasty (BAE), utilize autologous intestinal segments (e.g. ileum) to increase bladder capacity to protect renal function. Considered the standard of care, BAE is fraught with numerous short- and long-term clinical complications. Previous clinical trials employing tissue engineering approaches for bladder tissue regeneration have also been unable to translate bench-top findings into clinical practice. Major obstacles still persist that need to be overcome in order to advance tissue-engineered products into the clinical arena. These include scaffold/bladder incongruencies, the acquisition and utility of appropriate cells for anatomic and physiologic tissue recapitulation, and the choice of an appropriate animal model for testing. In this study, we demonstrate that the elastomeric, bladder biomechanocompatible poly(1,8-octamethylene-citrate-co-octanol) (PRS; synthetic) scaffold coseeded with autologous bone marrow-derived mesenchymal stem cells and CD34+ hematopoietic stem/progenitor cells support robust long-term, functional bladder tissue regeneration within the context of a clinically relevant baboon bladder augmentation model simulating bladder trauma. Partially cystectomized baboons were independently augmented with either autologous ileum or stem-cell-seeded small-intestinal submucosa (SIS; a commercially available biological scaffold) or PRS grafts. Stem-cell synergism promoted functional trilayer bladder tissue regeneration, including whole-graft neurovascularization, in both cell-seeded grafts. However, PRS-augmented animals demonstrated fewer clinical complications and more advantageous tissue characterization metrics compared to ileum and SIS-augmented animals. Two-year study data demonstrate that PRS/stem-cell-seeded grafts drive bladder tissue regeneration and are a suitable alternative to BAE.
Collapse
Affiliation(s)
- Matthew I Bury
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Natalie J Fuller
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Xinlong Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Yvonne Y Chan
- Department of Urologic Surgery, University of California at Davis, Davis, CA 95817, USA
| | - Renea M Sturm
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Sang Su Oh
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Laurel A Sofer
- Department of Urology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Hans C Arora
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Tiffany T Sharma
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Bonnie G Nolan
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Wei Feng
- Flow Cytometry Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rebecca R Rabizadeh
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Milica Barac
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Sonia S Edassery
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Madeleine M Goedegebuure
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Larry W Wang
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Balaji Ganesh
- Flow Cytometry Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lisa C Halliday
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mark E Seniw
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
| | - Seby L Edassery
- Center for Translational Research and Education, Loyola University Chicago, Chicago, IL 60153, USA
| | - Nadim B Mahmud
- Division of Hematology/Oncology, Department of Medicine, University of Illinois Cancer Center, Chicago, IL 60612, USA
| | | | - Kevin E McKenna
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60612, USA
| | - Earl Y Cheng
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Louis A. Simpson and Kimberly K. Querrey Biomedical Research Center, Chicago, IL 60611, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Guillermo A Ameer
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
- Vascular Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60612, USA
| | - Arun K Sharma
- Division of Pediatric Urology, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Louis A. Simpson and Kimberly K. Querrey Biomedical Research Center, Chicago, IL 60611, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
16
|
Tao X, Wang J, Liu B, Cheng P, Mu D, Du H, Niu B. Plasticity and crosstalk of mesenchymal stem cells and macrophages in immunomodulation in sepsis. Front Immunol 2024; 15:1338744. [PMID: 38352879 PMCID: PMC10861706 DOI: 10.3389/fimmu.2024.1338744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Sepsis is a multisystem disease characterized by dysregulation of the host immune response to infection. Immune response kinetics play a crucial role in the pathogenesis and progression of sepsis. Macrophages, which are known for their heterogeneity and plasticity, actively participate in the immune response during sepsis. These cells are influenced by the ever-changing immune microenvironment and exhibit two-sided immune regulation. Recently, the immunomodulatory function of mesenchymal stem cells (MSCs) in sepsis has garnered significant attention. The immune microenvironment can profoundly impact MSCs, prompting them to exhibit dual immunomodulatory functions akin to a double-edged sword. This discovery holds great importance for understanding sepsis progression and devising effective treatment strategies. Importantly, there is a close interrelationship between macrophages and MSCs, characterized by the fact that during sepsis, these two cell types interact and cooperate to regulate inflammatory processes. This review summarizes the plasticity of macrophages and MSCs within the immune microenvironment during sepsis, as well as the intricate crosstalk between them. This remains an important concern for the future use of these cells for immunomodulatory treatments in the clinic.
Collapse
Affiliation(s)
- Xingyu Tao
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Jialian Wang
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Bin Liu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Peifeng Cheng
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Dan Mu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Huimin Du
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bailin Niu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
17
|
Lee SY, Phuc HD, Um SH, Mongrain R, Yoon JK, Bhang SH. Photocuring 3D printing technology as an advanced tool for promoting angiogenesis in hypoxia-related diseases. J Tissue Eng 2024; 15:20417314241282476. [PMID: 39345255 PMCID: PMC11437565 DOI: 10.1177/20417314241282476] [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: 04/11/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
Three-dimensional (3D) bioprinting has emerged as a promising strategy for fabricating complex tissue analogs with intricate architectures, such as vascular networks. Achieving this necessitates bioink formulations that possess highly printable properties and provide a cell-friendly microenvironment mimicking the native extracellular matrix. Rapid advancements in printing techniques continue to expand the capabilities of researchers, enabling them to overcome existing biological barriers. This review offers a comprehensive examination of ultraviolet-based 3D bioprinting, renowned for its exceptional precision compared to other techniques, and explores its applications in inducing angiogenesis across diverse tissue models related to hypoxia. The high-precision and rapid photocuring capabilities of 3D bioprinting are essential for accurately replicating the intricate complexity of vascular networks and extending the diffusion limits for nutrients and gases. Addressing the lack of vascular structure is crucial in hypoxia-related diseases, as it can significantly improve oxygen delivery and overall tissue health. Consequently, high-resolution 3D bioprinting facilitates the creation of vascular structures within three-dimensional engineered tissues, offering a potential solution for addressing hypoxia-related diseases. Emphasis is placed on fundamental components essential for successful 3D bioprinting, including cell types, bioink compositions, and growth factors highlighted in recent studies. The insights provided in this review underscore the promising prospects of leveraging 3D printing technologies for addressing hypoxia-related diseases through the stimulation of angiogenesis, complementing the therapeutic efficacy of cell therapy.
Collapse
Affiliation(s)
- Sang Yoon Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Huynh Dai Phuc
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Soong Ho Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Rosaire Mongrain
- Mechanical Engineering Department, McGill University, Montréal, QC, Canada
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
18
|
Dudaryeva OY, Bernhard S, Tibbitt MW, Labouesse C. Implications of Cellular Mechanical Memory in Bioengineering. ACS Biomater Sci Eng 2023; 9:5985-5998. [PMID: 37797187 PMCID: PMC10646820 DOI: 10.1021/acsbiomaterials.3c01007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
The ability to maintain and differentiate cells in vitro is critical to many advances in the field of bioengineering. However, on traditional, stiff (E ≈ GPa) culture substrates, cells are subjected to sustained mechanical stress that can lead to phenotypic changes. Such changes may remain even after transferring the cells to another scaffold or engrafting them in vivo and bias the outcomes of the biological investigation or clinical treatment. This persistence─or mechanical memory─was initially observed for sustained myofibroblast activation of pulmonary fibroblasts after culturing them on stiff (E ≈ 100 kPa) substrates. Aspects of mechanical memory have now been described in many in vitro contexts. In this Review, we discuss the stiffness-induced effectors of mechanical memory: structural changes in the cytoskeleton and activity of transcription factors and epigenetic modifiers. We then focus on how mechanical memory impacts cell expansion and tissue regeneration outcomes in bioengineering applications relying on prolonged 2D plastic culture, such as stem cell therapies and disease models. We propose that alternatives to traditional cell culture substrates can be used to mitigate or erase mechanical memory and improve the efficiency of downstream cell-based bioengineering applications.
Collapse
Affiliation(s)
- Oksana Y Dudaryeva
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland
- Department of Orthopedics, University Medical Center Utrecht, Utrecht 3584, Netherlands
| | - Stéphane Bernhard
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland
| | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland
| | - Céline Labouesse
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland
| |
Collapse
|
19
|
Xu W, Yang Y, Li N, Hua J. Interaction between Mesenchymal Stem Cells and Immune Cells during Bone Injury Repair. Int J Mol Sci 2023; 24:14484. [PMID: 37833933 PMCID: PMC10572976 DOI: 10.3390/ijms241914484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
Fractures are the most common large organ trauma in humans. The initial inflammatory response promotes bone healing during the initial post-fracture phase, but chronic and persistent inflammation due to infection or other factors does not contribute to the healing process. The precise mechanisms by which immune cells and their cytokines are regulated in bone healing remain unclear. The use of mesenchymal stem cells (MSCs) for cellular therapy of bone injuries is a novel clinical treatment approach. Bone progenitor MSCs not only differentiate into bone, but also interact with the immune system to promote the healing process. We review in vitro and in vivo studies on the role of the immune system and bone marrow MSCs in bone healing and their interactions. A deeper understanding of this paradigm may provide clues to potential therapeutic targets in the healing process, thereby improving the reliability and safety of clinical applications of MSCs to promote bone healing.
Collapse
Affiliation(s)
| | | | - Na Li
- Shaanxi Centre of Stem Cells Engineering & Technology, College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China; (W.X.); (Y.Y.)
| | - Jinlian Hua
- Shaanxi Centre of Stem Cells Engineering & Technology, College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China; (W.X.); (Y.Y.)
| |
Collapse
|
20
|
Wen Y, Chen Y, Wu W, Zhang H, Peng Z, Yao X, Zhang X, Jiang W, Liao Y, Xie Y, Shen X, Sun H, Hu J, Liu H, Chen X, Chen J, Ouyang H. Hyperplastic Human Macromass Cartilage for Joint Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301833. [PMID: 37395375 PMCID: PMC10502860 DOI: 10.1002/advs.202301833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/07/2023] [Indexed: 07/04/2023]
Abstract
Cartilage damage affects millions of people worldwide. Tissue engineering strategies hold the promise to provide off-the-shelf cartilage analogs for tissue transplantation in cartilage repair. However, current strategies hardly generate sufficient grafts, as tissues cannot maintain size growth and cartilaginous phenotypes simultaneously. Herein, a step-wise strategy is developed for fabricating expandable human macromass cartilage (macro-cartilage) in a 3D condition by employing human polydactyly chondrocytes and a screen-defined serum-free customized culture (CC). CC-induced chondrocytes demonstrate improved cell plasticity, expressing chondrogenic biomarkers after a 14.59-times expansion. Crucially, CC-chondrocytes form large-size cartilage tissues with average diameters of 3.25 ± 0.05 mm, exhibiting abundant homogenous matrix and intact structure without a necrotic core. Compared with typical culture, the cell yield in CC increases 2.57 times, and the expression of cartilage marker collagen type II increases 4.70 times. Transcriptomics reveal that this step-wise culture drives a proliferation-to-differentiation process through an intermediate plastic stage, and CC-chondrocytes undergo a chondral lineage-specific differentiation with an activated metabolism. Animal studies show that CC macro-cartilage maintains a hyaline-like cartilage phenotype in vivo and significantly promotes the healing of large cartilage defects. Overall, an efficient expansion of human macro-cartilage with superior regenerative plasticity is achieved, providing a promising strategy for joint regeneration.
Collapse
|
21
|
Álvarez-López A, Rubio RT, Hernández-Escobar S, Daza R, Colchero L, Rezvanian P, Elices M, Guinea GV, González-Nieto D, Pérez-Rigueiro J. Application of single cell force spectroscopy (SCFS) to the assessment of cell adhesion to peptide-decorated surfaces. Int J Biol Macromol 2023; 244:125369. [PMID: 37321435 DOI: 10.1016/j.ijbiomac.2023.125369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/08/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
The adhesion forces of cells to peptide-coated functionalized materials were assessed through the Single Cell Force Spectroscopy (SCFS) technique in order to develop a methodology that allows the fast selection of peptide motifs that favor the interaction between cells and the biomaterial. Borosilicate glasses were functionalized using the activated vapor silanization process (AVS) and subsequently decorated with an RGD- containing peptide using the EDC/NHS crosslinking chemistry. It is shown that the RGD-coated glass induces larger attachment forces on mesenchymal stem cell cultures (MSCs), compared to the bare glass substrates. These higher forces correlate well with the enhanced adhesion of the MSCs observed on RGD-coated substrates through conventional adhesion cell cultures and inverse centrifugation tests. The methodology based on the SCFS technique presented in this work constitutes a fast procedure for the screening of new peptides or their combinations to select candidates that may enhance the response of the organism to the implant of the functionalized biomaterials.
Collapse
Affiliation(s)
- Aroa Álvarez-López
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón (Madrid), Spain; Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Raquel Tabraue Rubio
- Bioactive Surfaces S.L, C/ Puerto de Navacerrada 18, 28260 Galapagar (Madrid), Spain)
| | - Sandra Hernández-Escobar
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón (Madrid), Spain; Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Rafael Daza
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón (Madrid), Spain; Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Luis Colchero
- Bioactive Surfaces S.L, C/ Puerto de Navacerrada 18, 28260 Galapagar (Madrid), Spain)
| | - Parsa Rezvanian
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón (Madrid), Spain; Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain; Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran
| | - Manuel Elices
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Gustavo V Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón (Madrid), Spain; Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof, Martín Lagos s/n., 28040 Madrid, Spain
| | - Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón (Madrid), Spain; Bioactive Surfaces S.L, C/ Puerto de Navacerrada 18, 28260 Galapagar (Madrid), Spain); Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain; Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón (Madrid), Spain; Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain; Bioactive Surfaces S.L, C/ Puerto de Navacerrada 18, 28260 Galapagar (Madrid), Spain); Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof, Martín Lagos s/n., 28040 Madrid, Spain.
| |
Collapse
|
22
|
Yang X, Li Q, Liu W, Zong C, Wei L, Shi Y, Han Z. Mesenchymal stromal cells in hepatic fibrosis/cirrhosis: from pathogenesis to treatment. Cell Mol Immunol 2023; 20:583-599. [PMID: 36823236 PMCID: PMC10229624 DOI: 10.1038/s41423-023-00983-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/29/2023] [Indexed: 02/25/2023] Open
Abstract
Hepatic fibrosis/cirrhosis is a significant health burden worldwide, resulting in liver failure or hepatocellular carcinoma (HCC) and accounting for many deaths each year. The pathogenesis of hepatic fibrosis/cirrhosis is very complex, which makes treatment challenging. Endogenous mesenchymal stromal cells (MSCs) have been shown to play pivotal roles in the pathogenesis of hepatic fibrosis. Paradoxically, exogenous MSCs have also been used in clinical trials for liver cirrhosis, and their effectiveness has been observed in most completed clinical trials. There are still many issues to be resolved to promote the use of MSCs in the clinic in the future. In this review, we will examine the controversial role of MSCs in the pathogenesis and treatment of hepatic fibrosis/cirrhosis. We also investigated the clinical trials involving MSCs in liver cirrhosis, summarized the parameters that need to be standardized, and discussed how to promote the use of MSCs from a clinical perspective.
Collapse
Affiliation(s)
- Xue Yang
- Department of Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Naval Medical University, Shanghai, 200438, China
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Medical College of Soochow University, Soochow University, Suzhou, 215000, China
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Qing Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenting Liu
- Department of Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Naval Medical University, Shanghai, 200438, China
| | - Chen Zong
- Department of Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Naval Medical University, Shanghai, 200438, China
| | - Lixin Wei
- Department of Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, China
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Naval Medical University, Shanghai, 200438, China
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Medical College of Soochow University, Soochow University, Suzhou, 215000, China.
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Zhipeng Han
- Department of Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, China.
- Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Naval Medical University, Shanghai, 200438, China.
| |
Collapse
|
23
|
Re F, Borsani E, Rezzani R, Sartore L, Russo D. Bone Regeneration Using Mesenchymal Stromal Cells and Biocompatible Scaffolds: A Concise Review of the Current Clinical Trials. Gels 2023; 9:gels9050389. [PMID: 37232981 DOI: 10.3390/gels9050389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/27/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023] Open
Abstract
Bone regenerative medicine is a clinical approach combining live osteoblast progenitors, such as mesenchymal stromal cells (MSCs), with a biocompatible scaffold that can integrate into host bone tissue and restore its structural integrity. Over the last few years, many tissue engineering strategies have been developed and thoroughly investigated; however, limited approaches have been translated to clinical application. Consequently, the development and clinical validation of regenerative approaches remain a centerpiece of investigational efforts towards the clinical translation of advanced bioengineered scaffolds. The aim of this review was to identify the latest clinical trials related to the use of scaffolds with or without MSCs to regenerate bone defects. A revision of the literature was performed in PubMed, Embase, and Clinicaltrials.gov from 2018 up to 2023. Nine clinical trials were analyzed according to the inclusion criteria: six presented in the literature and three reported in Clinicaltrials.gov. Data were extracted covering background trial information. Six of the clinical trials added cells to scaffolds, while three used scaffolds alone. The majority of scaffolds were composed of calcium phosphate ceramic alone, such as β-tricalcium phosphate (TCP) (two clinical trials), biphasic calcium phosphate bioceramic granules (three clinical trials), and anorganic bovine bone (two clinical trials), while bone marrow was the primary source of the MSCs (five clinical trials). The MSC expansion was performed in GMP facilities, using human platelet lysate (PL) as a supplement without osteogenic factors. Only one trial reported minor adverse events. Overall, these findings highlight the importance and efficacy of cell-scaffold constructs in regenerative medicine under different conditions. Despite the encouraging clinical results obtained, further studies are needed to assess their clinical efficacy in treating bone diseases to optimize their application.
Collapse
Affiliation(s)
- Federica Re
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
| | - Elisa Borsani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
- Interdepartmental University Center of Research "Adaption and Regeneration of Tissues and Organs (ARTO)", University of Brescia, 25123 Brescia, Italy
| | - Rita Rezzani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
- Interdepartmental University Center of Research "Adaption and Regeneration of Tissues and Organs (ARTO)", University of Brescia, 25123 Brescia, Italy
| | - Luciana Sartore
- Department of Mechanical and Industrial Engineering, Materials Science and Technology Laboratory, University of Brescia, 25123 Brescia, Italy
| | - Domenico Russo
- Unit of Blood Diseases and Cell Therapies, Department of Clinical and Experimental Sciences, University of Brescia, "ASST-Spedali Civili" Hospital of Brescia, 25123 Brescia, Italy
| |
Collapse
|
24
|
Li Y, He M, Zhang W, Liu W, Xu H, Yang M, Zhang H, Liang H, Li W, Wu Z, Fu W, Xu S, Liu X, Fan S, Zhou L, Wang C, Zhang L, Li Y, Gu J, Yin J, Zhang Y, Xia Y, Mao X, Cheng T, Shi J, Du Y, Gao Y. Expansion of human megakaryocyte-biased hematopoietic stem cells by biomimetic Microniche. Nat Commun 2023; 14:2207. [PMID: 37072407 PMCID: PMC10113370 DOI: 10.1038/s41467-023-37954-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/04/2023] [Indexed: 04/20/2023] Open
Abstract
Limited numbers of available hematopoietic stem cells (HSCs) limit the widespread use of HSC-based therapies. Expansion systems for functional heterogenous HSCs remain to be optimized. Here, we present a convenient strategy for human HSC expansion based on a biomimetic Microniche. After demonstrating the expansion of HSC from different sources, we find that our Microniche-based system expands the therapeutically attractive megakaryocyte-biased HSC. We demonstrate scalable HSC expansion by applying this strategy in a stirred bioreactor. Moreover, we identify that the functional human megakaryocyte-biased HSCs are enriched in the CD34+CD38-CD45RA-CD90+CD49f lowCD62L-CD133+ subpopulation. Specifically, the expansion of megakaryocyte-biased HSCs is supported by a biomimetic niche-like microenvironment, which generates a suitable cytokine milieu and supplies the appropriate physical scaffolding. Thus, beyond clarifying the existence and immuno-phenotype of human megakaryocyte-biased HSC, our study demonstrates a flexible human HSC expansion strategy that could help realize the strong clinical promise of HSC-based therapies.
Collapse
Affiliation(s)
- Yinghui Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Mei He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Wenshan Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- Beijing CytoNiche Biotechnology Co. Ltd., 100195, Beijing, China
| | - Hui Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Ming Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Hexiao Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Haiwei Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Wenjing Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Zhaozhao Wu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Weichao Fu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Shiqi Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xiaolei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Sibin Fan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Liwei Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Chaoqun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Lele Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yafang Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Jiali Gu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Jingjing Yin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yiran Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yonghui Xia
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xuemei Mao
- Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western Medicine, Tianjin, 300100, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Jun Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, 100084, Beijing, China.
- Beijing CytoNiche Biotechnology Co. Ltd., 100195, Beijing, China.
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
25
|
Song X, Xu L, Zhang W. Biomimetic synthesis and optimization of extracellular vesicles for bone regeneration. J Control Release 2023; 355:18-41. [PMID: 36706840 DOI: 10.1016/j.jconrel.2023.01.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/29/2023]
Abstract
Critical-size bone defect repair is in high demand but is difficult to treat. Modern therapies, such as autograft and cell-based treatments, face limitations, including potential immunological rejection and tumorigenesis. Therefore, extracellular vesicle (EV)-based strategies have been proposed as a novel approach for tissue regeneration owing to EVs' complex composition of lipids, proteins, and nucleic acids, as well as their low immunogenicity and congenital cell-targeting features. Despite these remarkable features of EVs, biomimetic synthesis and optimization of natural EVs can lead to enhanced bioactivity, increased cellular uptake, and specific cell targeting, aiming to achieve optimal therapeutic efficacy. To maximize their function, these nanoparticles can be integrated into bone graft biomaterials for superior bone regeneration. Herein, we summarize the role of naturally occurring EVs from distinct cell types in bone regeneration, the current strategies for optimizing biomimetic synthetic EVs in bone regeneration, and discuss the recent advances in applying bone graft biomaterials for the delivery of EVs to bone defect repair. We focused on distinct strategies for optimizing EVs with different functions and the most recent research on achieving time-controlled release of nanoparticles from EV-loaded biomaterials. Furthermore, we thoroughly discuss several current challenges and proposed solutions, aiming to provide insight into current progress, inspiration for future development directions, and incentives for clinical application in this field.
Collapse
Affiliation(s)
- Xinyu Song
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China
| | - Ling Xu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.
| | - Wenjie Zhang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.
| |
Collapse
|
26
|
He Y, Li F, Jiang P, Cai F, Lin Q, Zhou M, Liu H, Yan F. Remote control of the recruitment and capture of endogenous stem cells by ultrasound for in situ repair of bone defects. Bioact Mater 2023; 21:223-238. [PMID: 36157244 PMCID: PMC9465026 DOI: 10.1016/j.bioactmat.2022.08.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Stem cell-based tissue engineering has provided a promising platform for repairing of bone defects. However, the use of exogenous bone marrow mesenchymal stem cells (BMSCs) still faces many challenges such as limited sources and potential risks. It is important to develop new approach to effectively recruit endogenous BMSCs and capture them for in situ bone regeneration. Here, we designed an acoustically responsive scaffold (ARS) and embedded it into SDF-1/BMP-2 loaded hydrogel to obtain biomimetic hydrogel scaffold complexes (BSC). The SDF-1/BMP-2 cytokines can be released on demand from the BSC implanted into the defected bone via pulsed ultrasound (p-US) irradiation at optimized acoustic parameters, recruiting the endogenous BMSCs to the bone defected or BSC site. Accompanied by the daily p-US irradiation for 14 days, the alginate hydrogel was degraded, resulting in the exposure of ARS to these recruited host stem cells. Then another set of sinusoidal continuous wave ultrasound (s-US) irradiation was applied to excite the ARS intrinsic resonance, forming highly localized acoustic field around its surface and generating enhanced acoustic trapping force, by which these recruited endogenous stem cells would be captured on the scaffold, greatly promoting them to adhesively grow for in situ bone tissue regeneration. Our study provides a novel and effective strategy for in situ bone defect repairing through acoustically manipulating endogenous BMSCs. We designed ARS and embedded it into SDF-1/BMP-2 loaded hydrogel to form BSC. The BSC can release SDF-1/BMP-2 by p-US irradiation for recruitment of endogenous BMSCs and capture them by s-US irradiation. The in situ repair of bone defects were successfully realized by US-mediated control of the recruitment and capture of BMSCs.
Collapse
Affiliation(s)
- Yanni He
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Fei Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Peng Jiang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Feiyan Cai
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Qin Lin
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
| | - Meijun Zhou
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China
| | - Hongmei Liu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
- Corresponding author. Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China.
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China
- Corresponding author. Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People's Republic of China.
| |
Collapse
|
27
|
Dai K, Zhang Q, Deng S, Yu Y, Zhu F, Zhang S, Pan Y, Long D, Wang J, Liu C. A BMP-2-triggered in vivo osteo-organoid for cell therapy. SCIENCE ADVANCES 2023; 9:eadd1541. [PMID: 36608118 PMCID: PMC9821865 DOI: 10.1126/sciadv.add1541] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Cell therapies and regenerative medicine interventions require an adequate source of therapeutic cells. Here, we demonstrate that constructing in vivo osteo-organoids by implanting bone morphogenetic protein-2-loaded scaffolds into the internal muscle pocket near the femur of mice supports the growth and subsequent harvest of therapeutically useful cells including hematopoietic stem/progenitor cells (HSPCs), mesenchymal stem cells (MSCs), lymphocytes, and myeloid cells. Profiling of the in vivo osteo-organoid maturation process delineated three stages-fibroproliferation, osteochondral differentiation, and marrow generation-each of which entailed obvious changes in the organoid structure and cell type distribution. The MSCs harvested from the osteochondral differentiation stage mitigated carbon tetrachloride (CCl4)-induced chronic liver fibrosis in mice, while HSPCs and immune cells harvested during the marrow generation stage rapidly and effectively reconstituted the impaired peripheral and solid immune organs of irradiated mice. These findings demonstrate the therapeutic potentials of in vivo osteo-organoid-derived cells in cell therapies.
Collapse
Affiliation(s)
- Kai Dai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, PR China
| | - Qinghao Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Shunshu Deng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Yuanman Yu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Fuwei Zhu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Shuang Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - YuanZhong Pan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Dandan Long
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Jing Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| | - Changsheng Liu
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, PR China
- Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR China
| |
Collapse
|
28
|
Jiang Z, Li N, Shao Q, Zhu D, Feng Y, Wang Y, Yu M, Ren L, Chen Q, Yang G. Light-controlled scaffold- and serum-free hard palatal-derived mesenchymal stem cell aggregates for bone regeneration. Bioeng Transl Med 2023; 8:e10334. [PMID: 36684075 PMCID: PMC9842060 DOI: 10.1002/btm2.10334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/10/2022] [Accepted: 04/18/2022] [Indexed: 01/25/2023] Open
Abstract
Cell aggregates that mimic in vivo cell-cell interactions are promising and powerful tools for tissue engineering. This study isolated a new, easily obtained, population of mesenchymal stem cells (MSCs) from rat hard palates named hard palatal-derived mesenchymal stem cells (PMSCs). The PMSCs were positive for CD90, CD44, and CD29 and negative for CD34, CD45, and CD146. They exhibited clonogenicity, self-renewal, migration, and multipotent differentiation capacities. Furthermore, this study fabricated scaffold-free 3D aggregates using light-controlled cell sheet technology and a serum-free method. PMSC aggregates were successfully constructed with good viability. Transplantation of the PMSC aggregates and the PMSC aggregate-implant complexes significantly enhanced bone formation and implant osseointegration in vivo, respectively. This new cell resource is easy to obtain and provides an alternative strategy for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Zhiwei Jiang
- Stomatology Hospital, School of StomatologyZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Oral DiseasesHangzhouZhejiangChina
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiangChina
| | - Na Li
- Stomatology Hospital, School of StomatologyZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Oral DiseasesHangzhouZhejiangChina
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiangChina
| | - Qin Shao
- Stomatology Hospital, School of StomatologyZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Oral DiseasesHangzhouZhejiangChina
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiangChina
| | - Danji Zhu
- Stomatology Hospital, School of StomatologyZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Oral DiseasesHangzhouZhejiangChina
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiangChina
| | - Yuting Feng
- Stomatology Hospital, School of StomatologyZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Oral DiseasesHangzhouZhejiangChina
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiangChina
| | - Yang Wang
- Stomatology Hospital, School of StomatologyZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Oral DiseasesHangzhouZhejiangChina
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiangChina
| | - Mengjia Yu
- Stomatology Hospital, School of StomatologyZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Oral DiseasesHangzhouZhejiangChina
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiangChina
| | - Lingfei Ren
- Stomatology Hospital, School of StomatologyZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Oral DiseasesHangzhouZhejiangChina
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiangChina
| | - Qianming Chen
- Stomatology Hospital, School of StomatologyZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Oral DiseasesHangzhouZhejiangChina
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiangChina
| | - Guoli Yang
- Stomatology Hospital, School of StomatologyZhejiang University School of MedicineHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Oral DiseasesHangzhouZhejiangChina
- Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiangChina
| |
Collapse
|
29
|
Wang X, Yu F, Ye L. Epigenetic control of mesenchymal stem cells orchestrates bone regeneration. Front Endocrinol (Lausanne) 2023; 14:1126787. [PMID: 36950693 PMCID: PMC10025550 DOI: 10.3389/fendo.2023.1126787] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/17/2023] [Indexed: 03/08/2023] Open
Abstract
Recent studies have revealed the vital role of MSCs in bone regeneration. In both self-healing bone regeneration processes and biomaterial-induced healing of bone defects beyond the critical size, MSCs show several functions, including osteogenic differentiation and thus providing seed cells. However, adverse factors such as drug intake and body senescence can significantly affect the functions of MSCs in bone regeneration. Currently, several modalities have been developed to regulate MSCs' phenotype and promote the bone regeneration process. Epigenetic regulation has received much attention because of its heritable nature. Indeed, epigenetic regulation of MSCs is involved in the pathogenesis of a variety of disorders of bone metabolism. Moreover, studies using epigenetic regulation to treat diseases are also being reported. At the same time, the effects of epigenetic regulation on MSCs are yet to be fully understood. This review focuses on recent advances in the effects of epigenetic regulation on osteogenic differentiation, proliferation, and cellular senescence in MSCs. We intend to illustrate how epigenetic regulation of MSCs orchestrates the process of bone regeneration.
Collapse
Affiliation(s)
- Xiaofeng Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fanyuan Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Fanyuan Yu, ; Ling Ye,
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Fanyuan Yu, ; Ling Ye,
| |
Collapse
|
30
|
Tai C, Xie Z, Li Y, Feng Y, Xie Y, Yang H, Wang L, Wang B. Human skin dermis-derived fibroblasts are a kind of functional mesenchymal stromal cells: judgements from surface markers, biological characteristics, to therapeutic efficacy. Cell Biosci 2022; 12:105. [PMID: 35831878 PMCID: PMC9277801 DOI: 10.1186/s13578-022-00842-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/03/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Human mesenchymal stromal cells (MSCs) have been widely advocated to clinical use. Human skin dermis-derived fibroblasts shared similar cellular morphology and biological characteristics to MSCs, while it still keeps elusive whether fibroblasts are functionally equivalent to MSCs for therapeutic use.
Methods
We isolated various fibroblasts derived from human foreskins (HFFs) and human double-fold eyelids (HDF) and MSCs derived from human umbilical cords (UC-MSCs), and then comprehensively investigated their similarities and differences in morphology, surface markers, immunoregulation, multilineage differentiation, transcriptome sequencing, and metabolomics, and therapeutic efficacies in treating 2,4,6-Trinitrobenzenesulfonic acid (TNBS) induced colitis and carbontetrachloride (CCL4) induced liver fibrosis.
Results
Fibroblasts and UC-MSCs shared similar surface markers, strong multilineage differentiation capacity, ability of inhibiting Th1/Th17 differentiation and promoting Treg differentiation in vitro, great similarities in mRNA expression profile and metabolites, and nearly equivalent therapeutic efficacy on TNBS-induced colitis and CCL4-induced hepatic fibrosis.
Conclusion
Human skin dermis-derived fibroblasts were a kind of functional MSCs with functionally equivalent therapeutic efficacy in treating specific complications, indicating fibroblasts potentially had the same lineage hierarchy of origin as MSCs and had a remarkable potential as an alternative to MSCs in the treatment of a variety of diseases.
Collapse
|
31
|
Xie X, Wu S, Mou S, Guo N, Wang Z, Sun J. Microtissue-Based Bioink as a Chondrocyte Microshelter for DLP Bioprinting. Adv Healthc Mater 2022; 11:e2201877. [PMID: 36085440 PMCID: PMC11468467 DOI: 10.1002/adhm.202201877] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/06/2022] [Indexed: 01/28/2023]
Abstract
Bioprinting specific tissues with robust viability is a great challenge, requiring a delicate balance between a densely cellular distribution and hydrogel network crosslinking density. Microtissues composed of tissue-specific mesenchymal stem cells and extra cellular matrix (ECM) particles provide an alternative scheme for realizing biomimetic cell density and microenvironment. Nevertheless, due to their instability during manufacturing, scarce efforts have been made to date to assemble them using rapid prototyping methods. Here, a novel microtissue bioink with good printability and cellular viability maintenance for digital light processing (DLP) bioprinting is introduced. Generally, the microtissue bioink is prepared by crosslinking acellular matrix microparticles and GelMA hydrogel with a specific proportion. The microtissue bioink exhibits the desired mechanical properties, swelling ratio, and has almost no influences on printability. For instance, a DLP bioprinted ear with a precise auricle structure using microtia chondrocytes microtissue boink is created. Additionally, the chondrocytes in the printed ears show obvious advantages in cell proliferation in vitro and auricular cartilage regeneration in vivo. The microtissue composite bioink for DLP printing not only enables accurate assembly of organ building blocks but also provides a 3D shelter to ensure printed cells' viability.
Collapse
Affiliation(s)
- Xinfang Xie
- Department of Plastic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Wuhan Clinical Research Center for Superficial Organ ReconstructionWuhan430022China
| | - Shuang Wu
- Department of Plastic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Wuhan Clinical Research Center for Superficial Organ ReconstructionWuhan430022China
| | - Shan Mou
- Department of Plastic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Wuhan Clinical Research Center for Superficial Organ ReconstructionWuhan430022China
| | - Nengqiang Guo
- Department of Plastic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Wuhan Clinical Research Center for Superficial Organ ReconstructionWuhan430022China
| | - Zhenxing Wang
- Department of Plastic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Wuhan Clinical Research Center for Superficial Organ ReconstructionWuhan430022China
| | - Jiaming Sun
- Department of Plastic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Wuhan Clinical Research Center for Superficial Organ ReconstructionWuhan430022China
| |
Collapse
|
32
|
Yigitbilek F, Ozdogan E, Abrol N, Park W, Hansen M, Dasari S, Stegall M, Taner T. Liver mesenchymal stem cells are superior inhibitors of NK cell functions through differences in their secretome compared to other mesenchymal stem cells. Front Immunol 2022; 13:952262. [PMID: 36211345 PMCID: PMC9534521 DOI: 10.3389/fimmu.2022.952262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022] Open
Abstract
Liver-resident mesenchymal stem cells (L-MSCs) are superior inhibitors of alloreactive T cell responses compared to their counterparts from bone marrow (BM-MSCs) or adipose tissue (A-MSCs), suggesting a role in liver’s overall tolerogenic microenvironment. Whether L-MSCs also impact NK cell functions differently than other MSCs is not known. We generated and characterized L-MSCs, A-MSCs and BM-MSCs from human tissues. The mass spectrometry analysis demonstrated that L-MSC secretome is uniquely different than that of A-MSC/BM-MSC, with enriched protein sets involved in IFNγ responses and signaling. When co-cultured with primary human NK cells, L-MSCs but not other MSCs, decreased surface expression of activating receptors NKp44 and NKG2D. L-MSCs also decreased IFNγ secretion by IL-2-stimulated NK cells more effectively than other MSCs. Cytolytic function of NK cells were reduced significantly when co-cultured with L-MSCs, whereas A-MSCs or BM-MSCs did not have a major impact. Mechanistic studies showed that the L-MSC-mediated reduction in NK cell cytotoxicity is not through changes in secretion of the cytotoxic proteins Perforin, Granzyme A or B, but through increased production of HLA-C1 found in L-MSC secretome that inhibits NK cells by stimulating their inhibitory receptor KIRDL2/3. L-MSCs are more potent inhibitors of NK cell functions than A-MSC or BM-MSC. Combined with their T cell inhibitory features, these results suggest L-MSCs contribute to the tolerogenic liver microenvironment and liver-induced systemic tolerance often observed after liver transplantation.
Collapse
Affiliation(s)
| | - Elif Ozdogan
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Nitin Abrol
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Walter D. Park
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | | | - Surendra Dasari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Mark D. Stegall
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Timucin Taner
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- *Correspondence: Timucin Taner,
| |
Collapse
|
33
|
Zhang X, Jiang W, Xie C, Wu X, Ren Q, Wang F, Shen X, Hong Y, Wu H, Liao Y, Zhang Y, Liang R, Sun W, Gu Y, Zhang T, Chen Y, Wei W, Zhang S, Zou W, Ouyang H. Msx1 + stem cells recruited by bioactive tissue engineering graft for bone regeneration. Nat Commun 2022; 13:5211. [PMID: 36064711 PMCID: PMC9445030 DOI: 10.1038/s41467-022-32868-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 08/17/2022] [Indexed: 12/11/2022] Open
Abstract
Critical-sized bone defects often lead to non-union and full-thickness defects of the calvarium specifically still present reconstructive challenges. In this study, we show that neurotrophic supplements induce robust in vitro expansion of mesenchymal stromal cells, and in situ transplantation of neurotrophic supplements-incorporated 3D-printed hydrogel grafts promote full-thickness regeneration of critical-sized bone defects. Single-cell RNA sequencing analysis reveals that a unique atlas of in situ stem/progenitor cells is generated during the calvarial bone healing in vivo. Notably, we find a local expansion of resident Msx1+ skeletal stem cells after transplantation of the in situ cell culture system. Moreover, the enhanced calvarial bone regeneration is accompanied by an increased endochondral ossification that closely correlates to the Msx1+ skeletal stem cells. Our findings illustrate the time-saving and regenerative efficacy of in situ cell culture systems targeting major cell subpopulations in vivo for rapid bone tissue regeneration. Critical-sized bone defects still present clinical challenges. Here the authors show that transplantation of neurotrophic supplement-incorporated hydrogel grafts promote full-thickness regeneration of the calvarium and perform scRNA-seq to reveal contributing stem/progenitor cells, notably a resident Msx1+ skeletal stem cell population.
Collapse
Affiliation(s)
- Xianzhu Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Jiang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Chang Xie
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyu Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Ren
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Fei Wang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xilin Shen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Hong
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongwei Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Youguo Liao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Renjie Liang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Sun
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuqing Gu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yishan Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Wei
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Shufang Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China. .,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China. .,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
34
|
Huang EE, Zhang N, Ganio EA, Shen H, Li X, Ueno M, Utsunomiya T, Maruyama M, Gao Q, Su N, Yao Z, Yang F, Gaudillière B, Goodman SB. Differential dynamics of bone graft transplantation and mesenchymal stem cell therapy during bone defect healing in a murine critical size defect. J Orthop Translat 2022; 36:64-74. [PMID: 35979174 PMCID: PMC9357712 DOI: 10.1016/j.jot.2022.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 10/24/2022] Open
Abstract
Background A critical size bone defect is a clinical scenario in which bone is lost or excised due to trauma, infection, tumor, or other causes, and cannot completely heal spontaneously. The most common treatment for this condition is autologous bone grafting to the defect site. However, autologous bone graft is often insufficient in quantity or quality for transplantation to these large defects. Recently, tissue engineering methods using mesenchymal stem cells (MSCs) have been proposed as an alternative treatment. However, the underlying biological principles and optimal techniques for tissue regeneration of bone using stem cell therapy have not been completely elucidated. Methods In this study, we compare the early cellular dynamics of healing between bone graft transplantation and MSC therapy in a murine chronic femoral critical-size bone defect. We employ high-dimensional mass cytometry to provide a comprehensive view of the differences in cell composition, stem cell functionality, and immunomodulatory activity between these two treatment methods one week after transplantation. Results We reveal distinct cell compositions among tissues from bone defect sites compared with original bone graft, show active recruitment of MSCs to the bone defect sites, and demonstrate the phenotypic diversity of macrophages and T cells in each group that may affect the clinical outcome. Conclusion Our results provide critical data and future directions on the use of MSCs for treating critical size defects to regenerate bone.Translational Potential of this article: This study showed systematic comparisons of the cellular and immunomodulatory profiles among different interventions to improve the healing of the critical-size bone defect. The results provided potential strategies for designing robust therapeutic interventions for the unmet clinical need of treating critical-size bone defects.
Collapse
Affiliation(s)
- Elijah Ejun Huang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Edward A. Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Xueping Li
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Masaya Ueno
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Takeshi Utsunomiya
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Masahiro Maruyama
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Ni Su
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
35
|
Xu T, Yang Y, Suo D, Bei HP, Xu X, Zhao X. Electrosprayed Regeneration-Enhancer-Element Microspheres Power Osteogenesis and Angiogenesis Coupling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200314. [PMID: 35261154 DOI: 10.1002/smll.202200314] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/07/2022] [Indexed: 06/14/2023]
Abstract
Electrosprayed microspheres for bone regeneration are conventionally restricted by the lack of osteogenic modulation for both encapsulated stem cells and surrounding cells at the defect site. Here, sodium alginate microspheres encapsulating L-arginine doped hydroxyapatite nanoparticles (Arg/HA NPs) and bone mesenchymal stem cells (BMSCs) as regeneration-enhancer-element reservoirs (Arg/HA-SA@BMSC) for bone healing are electrosprayed. The Arg/HA NPs serve as a container of L-arginine and Ca2+ and the BMSCs inside the microspheres metabolize the released L-arginine into bioactive gas nitric oxide (NO) in the presence of Ca2+ to activate the nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) signaling pathway. Meanwhile, the generated NO diffuses out of the microspheres together with the Ca2+ and L-arginine as exterior enhancers to promote the osteogenesis-angiogenesis coupling of surrounding BMSCs and endothelial cells (ECs) at the bone defect site, generating an internal/external modulation loop between the encapsulated cells and surrounding native cells. It is demonstrated that such regeneration-enhancer-element reservoirs could effectively increase the bone tissue formation and neovasculature using rat calvarial defect models. It is envisioned that the microsphere system could streamline vascularized bone regeneration therapy as a high throughput, minimally invasive yet highly effective strategy to accelerate bone healing.
Collapse
Affiliation(s)
- Tianpeng Xu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, P. R. China
| | - Yuhe Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, P. R. China
| | - Di Suo
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, P. R. China
| | - Ho Pan Bei
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, P. R. China
| | - Xiaoxiao Xu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, P. R. China
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, P. R. China
| |
Collapse
|
36
|
Sarviya N, Basu SM, Mani R, Chauhan M, Kingshott P, Giri J. Biomimicking nanofibrous gelatin microspheres recreating the stem cell niche for their ex-vivo expansion and in-vivo like differentiation for injectable stem cell transplantation. BIOMATERIALS ADVANCES 2022; 139:212981. [PMID: 35882137 DOI: 10.1016/j.bioadv.2022.212981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
Stem cells based novel treatment modality for degenerative and immune dysfunction diseases created a huge demand of suitable carriers to support ex-vivo production of quality stem cells, and effective in-vivo transplantation of stem cells and their fate. In spite of promising candidature of nanofibrous microspheres (NFM) to recreate native stem cell niches to be used for possible scaling-up for ex-vivo stem cells expansion, it remains fairly unexplored. A systematic study on the stem cell-NFM interaction comparative with commercial microspheres (CM) has been performed for the first time. Gelatin NFM with variable physicochemical properties such as size, surface properties, surface chemistry, and variable degradability were prepared using microemulsion coupled with thermally induced phase separation (TIPS) method. Effect of physicochemical properties of NFM and their cellular interaction such as binding, morphology, metabolic activity and proliferation studies were performed using human bone marrow-derived mesenchymal stem cells (hBMSCs), human dental follicle stem cells (hDFSCs) and human gingival fibroblast (HGF) cells and compared with the commercial and solid microspheres. Gelatin NFM supports excellent cell binding, proliferation, metabolic activities and chemical cues specific differentiation. All out-turns indicate that NFM stand to be an outstanding candidate for ex-vivo cells' expansion and injectable carriers for stem cell transplantation.
Collapse
Affiliation(s)
- Nandini Sarviya
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India; Department of Chemistry and Biotechnology, Swinburne Institute of Technology, Victoria, Australia
| | - Suparna Mercy Basu
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Rajesh Mani
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Meenakshi Chauhan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India
| | - Peter Kingshott
- Department of Chemistry and Biotechnology, Swinburne Institute of Technology, Victoria, Australia
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana, India.
| |
Collapse
|
37
|
Song MK, Sun HJ, Cho SW. Conditioned medium of amniotic fluid-derived stromal cells exerts a bone anabolic effect by enhancing progenitor population and angiogenesis. J Tissue Eng Regen Med 2022; 16:923-933. [PMID: 35819750 DOI: 10.1002/term.3340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 06/14/2022] [Accepted: 06/29/2022] [Indexed: 11/10/2022]
Abstract
A cell-free approach utilizing the paracrine effects of mesenchymal stromal cells is receiving attention in regenerative medicine. In the present study, we evaluated the effects of a conditioned medium of amniotic fluid-derived stromal cells (AFSC-CM) on bone metabolism. In mice, intraperitoneal injections of AFSC-CM increased bone mass and enhanced bone turnover. The precursor populations of myeloid and mesenchymal lineages, as well as endothelial cells in bone marrow, were also augmented by AFSC-CM administration. In an in vitro culture experiment, AFSC-CM increased osteoclast differentiation of bone marrow-derived macrophages, but had no significant effect on the osteogenic differentiation of preosteoblasts. However, AFSC-CM administration dramatically accelerated the migration and tube formation of endothelial cells, and a cytokine array showed that AFSC-CM contained many angiogenic factors. These results indicate that AFSC-CM exerts a bone anabolic effect by changing the bone marrow microenvironment, including angiogenesis and precursor expansion. Therefore, ameliorating marrow angiogenesis is a potential therapeutic strategy for bone regeneration, for which AFSCs can be a good cellular source.
Collapse
Affiliation(s)
- Min-Kyoung Song
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine, Seoul National University, Daehak-ro, Jongno-gu, Seoul, Korea
| | - Hyun Jin Sun
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, Korea
| | - Sun Wook Cho
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Korea
| |
Collapse
|
38
|
Epigenetic therapy targeting bone marrow mesenchymal stem cells for age-related bone diseases. Stem Cell Res Ther 2022; 13:201. [PMID: 35578312 PMCID: PMC9109405 DOI: 10.1186/s13287-022-02852-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/14/2022] [Indexed: 02/08/2023] Open
Abstract
As global aging accelerates, the prevention and treatment of age-related bone diseases are becoming a critical issue. In the process of senescence, bone marrow mesenchymal stem cells (BMSCs) gradually lose the capability of self-renewal and functional differentiation, resulting in impairment of bone tissue regeneration and disorder of bone tissue homeostasis. Alteration in epigenetic modification is an essential factor of BMSC dysfunction during aging. Its transferability and reversibility provide the possibility to combat BMSC aging by reversing age-related modifications. Emerging evidence demonstrates that epigenetic therapy based on aberrant epigenetic modifications could alleviate the senescence and dysfunction of stem cells. This review summarizes potential therapeutic targets for BMSC aging, introduces some potential approaches to alleviating BMSC aging, and analyzes its prospect in the clinical application of age-related bone diseases.
Collapse
|
39
|
Enhanced Repaired Enthesis Using Tenogenically Differentiated Adipose-Derived Stem Cells in a Murine Rotator Cuff Injury Model. Stem Cells Int 2022; 2022:1309684. [PMID: 35607399 PMCID: PMC9124132 DOI: 10.1155/2022/1309684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 01/10/2022] [Indexed: 11/18/2022] Open
Abstract
Rotator cuff tear (RCT) is among the most common shoulder injuries and is prone to rerupture after surgery. Selecting suitable subpopulations of stem cells as a new specific cell type of mesenchymal stem cells has been increasingly used as a potential therapeutic tool in regenerative medicine. In this study, murine adipose-derived SSEA-4+CD90+PDGFRA+ subpopulation cells were successfully sorted, extracted, and identified. These cells showed good proliferation and differentiation potential, especially in the direction of tendon differentiation, as evidenced by qRT-PCR and immunofluorescence. Subsequently, we established a murine rotator cuff injury model and repaired it with subpopulation cells. Our results showed that the subpopulation cells embedded in a fibrin sealant significantly improved the histological score, as well as the biomechanical strength of the repaired tendon enthesis at four weeks after surgery, compared with the other groups. Hence, these findings indicated that the subpopulation of cells could augment the repaired enthesis and lead to better outcomes, thereby reducing the retear rate after rotator cuff repair. Our study provides a potential therapeutic strategy for rotator cuff healing in the future.
Collapse
|
40
|
Ohori-Morita Y, Niibe K, Limraksasin P, Nattasit P, Miao X, Yamada M, Mabuchi Y, Matsuzaki Y, Egusa H. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:434-449. [PMID: 35267026 PMCID: PMC9052431 DOI: 10.1093/stcltm/szab030] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 12/02/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
- Yumi Ohori-Morita
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Kunimichi Niibe
- Corresponding authors: Kunimichi Niibe, DDS, PhD, Associate Professor, Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai-city, Miyagi 980-8575, Japan. Tel: +81-22-717-8363; Fax: +81-22-717-8367;
| | - Phoonsuk Limraksasin
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Praphawi Nattasit
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Xinchao Miao
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Masahiro Yamada
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Yo Mabuchi
- Department of Biochemistry and Biophysics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yumi Matsuzaki
- Department of Life Science, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane 693-8501, Japan
| | - Hiroshi Egusa
- Hiroshi Egusa, DDS, PhD, Director, Center for Advanced Stem Cell and Regenerative Research, Professor and Chair, Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai-city 980-8575, Japan. Tel: +81-22-717-8363; Fax: +81-22-717-8367;
| |
Collapse
|
41
|
Hu X, Xia Z, Cai K. Recent advances of 3D hydrogel culture systems for mesenchymal stem cell-based therapy and cell behavior regulation. J Mater Chem B 2022; 10:1486-1507. [DOI: 10.1039/d1tb02537f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mesenchymal stem cells (MSCs) have been increasingly recognized as resources for disease treatments and regenerative medicine. Meanwhile, the unique chemical and physical properties of hydrogels provide innate advantages to achieve...
Collapse
|
42
|
Zhang XF, Wang T, Wang ZX, Huang KP, Zhang YW, Wang GL, Zhang HJ, Chen ZH, Wang CY, Zhang JX, Wang H. Hypoxic ucMSC-secreted exosomal miR-125b promotes endothelial cell survival and migration during wound healing by targeting TP53INP1. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:347-359. [PMID: 34513314 PMCID: PMC8416974 DOI: 10.1016/j.omtn.2021.07.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/17/2021] [Indexed: 12/29/2022]
Abstract
A hypoxic microenvironment is a common feature of skin wounds. Our previous study demonstrated that three-dimensional coculture of umbilical cord-derived mesenchymal stem cells (ucMSCs) and endothelial cells facilitates cell communication and host integration in skin tissue engineering. Here, we aimed to identify the mechanism by which ucMSCs affect endothelial cells under hypoxic conditions after skin injury. We demonstrate that hypoxia enhances the exosome-mediated paracrine function of ucMSCs, which increases endothelial cell proliferation and migration. In a mouse full-thickness skin injury model, ucMSC-derived exosomes can be taken up by endothelial cells and accelerate wound healing. Hypoxic exosomes lead to a better outcome than normoxic exosomes by promoting proliferation and inhibiting apoptosis. Mechanistically, microRNA-125b (miR-125b) transcription is induced by hypoxia in ucMSCs. After being packaged into hypoxic exosomes and transported to endothelial cells, miR-125b targets and suppresses the expression of tumor protein p53 inducible nuclear protein 1 (TP53INP1) and alleviates hypoxia-induced cell apoptosis. Inhibition of miR-125b-TP53INP1 interaction attenuates the protective effect of hypoxic exosomes. Moreover, artificial agomiR-125b can accelerate wound healing in vivo. Our findings reveal communication between ucMSCs and endothelial cells via exosomal miR-125b/TP53INP1 signaling in the hypoxic microenvironment and present hypoxic exosomes as a promising therapeutic strategy to enhance cutaneous repair.
Collapse
Affiliation(s)
- Xiao-Fei Zhang
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ting Wang
- Department of Medical Genetics, Basic school of Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Zi-Xuan Wang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kun-Peng Huang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yun-Wei Zhang
- Department of Emergency, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guo-Liang Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hong-Ji Zhang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zi-Han Chen
- Department of Medical Genetics, Basic school of Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Chang-Yan Wang
- Department of Medical Genetics, Basic school of Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Jin-Xiang Zhang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Wang
- Department of Medical Genetics, Basic school of Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| |
Collapse
|
43
|
Guo P, Liu X, Zhang P, He Z, Li Z, Alini M, Richards RG, Grad S, Stoddart MJ, Zhou G, Zou X, Chan D, Tian W, Chen D, Gao M, Zhou Z, Liu S. A single-cell transcriptome of mesenchymal stromal cells to fabricate bioactive hydroxyapatite materials for bone regeneration. Bioact Mater 2021; 9:281-298. [PMID: 34820571 PMCID: PMC8586438 DOI: 10.1016/j.bioactmat.2021.08.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/01/2021] [Accepted: 08/05/2021] [Indexed: 01/07/2023] Open
Abstract
The osteogenic microenvironment of bone-repairing materials plays a key role in accelerating bone regeneration but remains incompletely defined, which significantly limits the application of such bioactive materials. Here, the transcriptional landscapes of different osteogenic microenvironments, including three-dimensional (3D) hydroxyapatite (HA) scaffolds and osteogenic medium (OM), for mesenchymal stromal cells (MSCs) in vitro were mapped at single-cell resolution. Our findings suggested that an osteogenic process reminiscent of endochondral ossification occurred in HA scaffolds through sequential activation of osteogenic-related signaling pathways, along with inflammation and angiogenesis, but inhibition of adipogenesis and fibrosis. Moreover, we revealed the mechanism during OM-mediated osteogenesis involves the ZBTB16 and WNT signaling pathways. Heterogeneity of MSCs was also demonstrated. In vitro ossification of LRRC75A+ MSCs was shown to have better utilization of WNT-related ossification process, and PCDH10+ MSCs with superiority in hydroxyapatite-related osteogenic process. These findings provided further understanding of the cellular activity modulated by OM conditions and HA scaffolds, providing new insights for the improvement of osteogenic biomaterials. This atlas provides a blueprint for research on MSC heterogeneity and the osteogenic microenvironment of HA scaffolds and a database reference for the application of bioactive materials for bone regeneration.
Collapse
Affiliation(s)
- Peng Guo
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xizhe Liu
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Penghui Zhang
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhongyuan He
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen Li
- AO Research Institute Davos, Davos, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | | | | | | - Guangqian Zhou
- Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Health Sciences Center, Shenzhen University, Shenzhen, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Danny Chan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Wei Tian
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopaedics and Traumatology, Beijing JiShuiTan Hospital, Beijing, China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopaedics and Traumatology, Beijing JiShuiTan Hospital, Beijing, China
- Corresponding author.
| | - Manman Gao
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Sport Medicine, Inst Translat Med, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
- Corresponding author. Department of Sport Medicine, Inst Translat Med, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China.
| | - Zhiyu Zhou
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Corresponding author. Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Shaoyu Liu
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
44
|
Cell-based therapeutics for the treatment of hematologic diseases inside the bone marrow. J Control Release 2021; 339:1-13. [PMID: 34536449 DOI: 10.1016/j.jconrel.2021.09.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022]
Abstract
Cell-based therapies could overcome the limitations of traditional drugs for the treatment of refractory diseases. Cell exchange between the bone marrow and blood is bidirectional. Several kinds of cells in the blood have the capability to enter the bone marrow by interacting with sinusoidal cells under specific physiological or pathological conditions. These cells are the potential living therapeutics or delivery vehicles to treat or prevent bone marrow-related hematologic diseases. In this review, we summarized the in vivo molecular mechanisms and kinetics of these cells in entering the bone marrow. The advances in the fabrication of living cell drugs and the strategies to design cell-based carriers into the bone marrow were discussed. The latest studies on how to use blood cells as living drugs or as drug carriers to improve therapeutic outcomes of hematologic diseases inside the bone marrow were highlighted.
Collapse
|
45
|
Lu Y, Yu CH, Yang G, Sun N, Jiang F, Zhou M, Wu X, Luo J, Huang C, Zhang W, Jiang X. A rapidly magnetically assembled stem cell microtissue with "hamburger" architecture and enhanced vascularization capacity. Bioact Mater 2021; 6:3756-3765. [PMID: 33898876 PMCID: PMC8044908 DOI: 10.1016/j.bioactmat.2021.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/23/2021] [Accepted: 03/02/2021] [Indexed: 12/19/2022] Open
Abstract
With the development of magnetic manipulation technology based on magnetic nanoparticles (MNPs), scaffold-free microtissues can be constructed utilizing the magnetic attraction of MNP-labeled cells. The rapid in vitro construction and in vivo vascularization of microtissues with complex hierarchical architectures are of great importance to the viability and function of stem cell microtissues. Endothelial cells are indispensable for the formation of blood vessels and can be used in the prevascularization of engineered tissue constructs. Herein, safe and rapid magnetic labeling of cells was achieved by incubation with MNPs for 1 h, and ultrathick scaffold-free microtissues with different sophisticated architectures were rapidly assembled, layer by layer, in 5 min intervals. The in vivo transplantation results showed that in a stem cell microtissue with trisection architecture, the two separated human umbilical vein endothelial cell (HUVEC) layers would spontaneously extend to the stem cell layers and connect with each other to form a spatial network of functional blood vessels, which anastomosed with the host vasculature. The "hamburger" architecture of stem cell microtissues with separated HUVEC layers could promote vascularization and stem cell survival. This study will contribute to the construction and application of structural and functional tissues or organs in the future.
Collapse
Affiliation(s)
- Yuezhi Lu
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chun-Hua Yu
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Guangzheng Yang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ningjia Sun
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Fei Jiang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Mingliang Zhou
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiaolin Wu
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jiaxin Luo
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Cui Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedical Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, 430079, China
| | - Wenjie Zhang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
46
|
Mollentze J, Durandt C, Pepper MS. An In Vitro and In Vivo Comparison of Osteogenic Differentiation of Human Mesenchymal Stromal/Stem Cells. Stem Cells Int 2021; 2021:9919361. [PMID: 34539793 PMCID: PMC8443361 DOI: 10.1155/2021/9919361] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/23/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
The use of stem cells in regenerative medicine, including tissue engineering and transplantation, has generated a great deal of enthusiasm. Mesenchymal stromal/stem cells (MSCs) can be isolated from various tissues, most commonly, bone marrow but more recently adipose tissue, dental pulp, and Wharton's jelly, to name a few. MSCs display varying phenotypic profiles and osteogenic differentiating capacity depending and their site of origin. MSCs have been successfully differentiated into osteoblasts both in vitro an in vivo but discrepancies exist when the two are compared: what happens in vitro does not necessarily happen in vivo, and it is therefore important to understand why these differences occur. The osteogenic process is a complex network of transcription factors, stimulators, inhibitors, proteins, etc., and in vivo experiments are helpful in evaluating the various aspects of this osteogenic process without distractions and confounding variables. With that in mind, the results of in vitro experiments need to be carefully considered and interpreted with caution as they do not perfectly replicate the conditions found within living organisms. This is where in vivo experiments help us better understand interactions that might occur in the osteogenic process that cannot be replicated in vitro. Potentially, these differences could also be exploited to develop an optimal MSC cell therapeutic product that can be used for bone disorders. There are many bone disorders, most of which cause a great deal of discomfort. Clinically acceptable protocols could be developed in which MSCs are used to aid in bone regeneration providing relief for patients with chronic pain. The aim of this review is to examine the differences between studies conducted in vitro and in vivo with regard to the osteogenic process to better define the gaps in current osteogenic research. By better understanding osteogenic differentiation, we can better define treatment strategies for various bone disorders.
Collapse
Affiliation(s)
- Jamie Mollentze
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Chrisna Durandt
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
47
|
Functional heterogeneity of IFN-γ-licensed mesenchymal stromal cell immunosuppressive capacity on biomaterials. Proc Natl Acad Sci U S A 2021; 118:2105972118. [PMID: 34446555 DOI: 10.1073/pnas.2105972118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are increasingly combined with biomaterials to enhance their therapeutic properties, including their immunosuppressive function. However, clinical trials utilizing MSCs with or without biomaterials have shown limited success, potentially due to their functional heterogeneity across different donors and among different subpopulations of cells. Here, we evaluated the immunosuppressive capacity, as measured by the ability to reduce T-cell proliferation and activation, of interferon-gamma (IFN-γ)-licensed MSCs from multiple donors on fibrin and collagen hydrogels, the two most commonly utilized biomaterials in combination with MSCs in clinical trials worldwide according to ClinicalTrials.gov Variations in the immunosuppressive capacity between IFN-γ-licensed MSC donors on the biomaterials correlated with the magnitude of indoleamine-2,3-dioxygenase activity. Immunosuppressive capacity of the IFN-γ-licensed MSCs depended on the αV/α5 integrins when cultured on fibrin and on the α2/β1 integrins when cultured on collagen. While all tested MSCs were nearly 100% positive for these integrins, sorted MSCs that expressed higher levels of αV/α5 integrins demonstrated greater immunosuppressive capacity with IFN-γ licensing than MSCs that expressed lower levels of these integrins on fibrin. These findings were equivalent for MSCs sorted based on the α2/β1 integrins on collagen. These results demonstrate the importance of integrin engagement to IFN-γ licensed MSC immunosuppressive capacity and that IFN-γ-licensed MSC subpopulations of varying immunosuppressive capacity can be identified by the magnitude of integrin expression specific to each biomaterial.
Collapse
|
48
|
Trejo-Iriarte CG, Ortega MA, Asúnsolo Á, Gómez-Clavel JF, Muñoz AG, Mon MÁ, Buján J, Acero J, García-Honduvilla N. Mesenchymal adipose stem cells maintain the capacity for differentiation and survival in culture beyond the long term. J Histotechnol 2021; 44:217-233. [PMID: 34412574 DOI: 10.1080/01478885.2021.1953248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mesenchymal cells (MSCs) are considered to be cellular populations of common embryological origin. For clinical research applications, MSCs are expanded and increased with cells obtained from a primary culture. By extracting cells from tissue and encouraging them to reproduce, the stem cell population ends up dominating the culture due to a high proliferation rate and self-renewal. The first subcultures between the third and sixth are chosen in order to obtain the maximum number of cells with optimal differentiation capacity. However, few studies have reported long-term cultivation of MSCs. The objective of this study was to advance the knowledge on the characteristics of MSCs by assessing their capacity for self-renewal and phenotypic maintenance beyond 50 cell subcultures, which is defined as the normal limit for cellular survival. Rat subcutaneous adipose tissue was the source of mesenchymal adipose stem cells (MASCs) cultured over 175 subcultures. Early 1 to 5 and late 25 to 30 subcultures were used to induce cellular differentiation to become adipogenic, chondrogenic and osteogenic connective tissue cells. MASCs characteristics were studied using flow cytometry, transmission electron microscopy (TEM), and immunohistochemical and reverse transcription polymerase chain reaction (RT-qPCR) assays. The MASCs maintained cell differentiation capacity for more than 30 subcultures but lost potentiality starting at 60 up to 175 subcultures. MASCs showed the embryonic phenotypes OCT3/4 and Nanog indefinitely, and developed compensatory mechanisms, such as autophagy, to achieve cell survival over a long time period. Therefore, long-term subcultures showed that MASCs could maintain their potential for clinical research use.
Collapse
Affiliation(s)
- Cynthia G Trejo-Iriarte
- Research Group in Stem Cells and Tissue Engineering, Almaraz Dentistry Research Laboratory, Dentist Surgeon Studies, Iztacala Higher Studies School, National Autonomous University of Mexico, Mexico DF, Mexico
| | - Miguel A Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health, University of Alcalá, Alcalá de Henares, Spain.,Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Ángel Asúnsolo
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain.,Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
| | - José F Gómez-Clavel
- Laboratory of Research in Education and Dentistry; Dentist Surgeon Studies, School of Higher Studies Iztacala, National Autonomous University of Mexico, Mexico DF, Mexico
| | - Alejandro García Muñoz
- Research Group in Stem Cells and Tissue Engineering, Almaraz Dentistry Research Laboratory, Dentist Surgeon Studies, Iztacala Higher Studies School, National Autonomous University of Mexico, Mexico DF, Mexico
| | - Melchor Álvarez- Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health, University of Alcalá, Alcalá de Henares, Spain.,Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain.,Immune System Diseases-Rheumatology and Oncology Service, CIBEREHD, University Hospital Príncipe de Asturias, Alcalá de Henares, Spain
| | - Julia Buján
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health, University of Alcalá, Alcalá de Henares, Spain.,Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| | - Julio Acero
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain.,Department of Oral and Maxillofacial Surgery, Ramon y Cajal University Hospital, Alcalá University, Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health, University of Alcalá, Alcalá de Henares, Spain.,Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
| |
Collapse
|
49
|
Bao C, He C. The role and therapeutic potential of MSC-derived exosomes in osteoarthritis. Arch Biochem Biophys 2021; 710:109002. [PMID: 34352243 DOI: 10.1016/j.abb.2021.109002] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/27/2021] [Accepted: 07/31/2021] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is the most common painful disease with chronic articular cartilage degeneration. The pathological process of OA is complex and characterized by the imbalance between the synthesis and catabolism of chondrocytes and extracellular matrix, leading to the progressive destruction of articular cartilage damage. Because of the self-renewal and differentiation of mesenchymal stem cells (MSCs), various exogenous MSC-based cell therapies have been developed to treat OA. Moreover, the efficacy of MSC- based therapy is mainly attributed to the paracrine of cytokines, growth factors, and exosomes. Exosomes derived from MSCs can deliver various DNAs, RNAs, proteins and lipids, thus promoting MSCs migration and cartilage repair. Therefore, MSC-derived exosomes are considered as a promising alternative therapy for OA. In this review, we summarized properties of MSC-derived exosomes and the new role of MSC-derived exosomes in the treatment of OA. We also proposed possible perspectives of MSC-derived exosomes as cell-free regenerative reagents in the treatment of OA.
Collapse
Affiliation(s)
- Chuncha Bao
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chengqi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
50
|
Zhang K, Feng Q, Fang Z, Gu L, Bian L. Structurally Dynamic Hydrogels for Biomedical Applications: Pursuing a Fine Balance between Macroscopic Stability and Microscopic Dynamics. Chem Rev 2021; 121:11149-11193. [PMID: 34189903 DOI: 10.1021/acs.chemrev.1c00071] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Owing to their unique chemical and physical properties, hydrogels are attracting increasing attention in both basic and translational biomedical studies. Although the classical hydrogels with static networks have been widely reported for decades, a growing number of recent studies have shown that structurally dynamic hydrogels can better mimic the dynamics and functions of natural extracellular matrix (ECM) in soft tissues. These synthetic materials with defined compositions can recapitulate key chemical and biophysical properties of living tissues, providing an important means to understanding the mechanisms by which cells sense and remodel their surrounding microenvironments. This review begins with the overall expectation and design principles of dynamic hydrogels. We then highlight recent progress in the fabrication strategies of dynamic hydrogels including both degradation-dependent and degradation-independent approaches, followed by their unique properties and use in biomedical applications such as regenerative medicine, drug delivery, and 3D culture. Finally, challenges and emerging trends in the development and application of dynamic hydrogels are discussed.
Collapse
Affiliation(s)
- Kunyu Zhang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Qian Feng
- Bioengineering College, Chongqing University, Chongqing 400044, People's Republic of China
| | - Zhiwei Fang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Luo Gu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, People's Republic of China.,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, People's Republic of China.,Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, People's Republic of China.,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, People's Republic of China.,Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, People's Republic of China
| |
Collapse
|