1
|
Fan ZQ, Xie YY, Liu C, Chen YF, Yi YF, Tang ZW, Wen J, Xiao S, Li YF. Most vexing problem in pediatric fractures: Epiphyseal fractures. World J Orthop 2025; 16:106265. [DOI: 10.5312/wjo.v16.i5.106265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/27/2025] [Accepted: 04/15/2025] [Indexed: 05/16/2025] Open
Abstract
Epiphyseal fracture is a significant etiology of limb deformity in children following fractures. However, unlike the rapid advancements in orthopedics, progress regarding the pathological changes, diagnosis, and treatment of epiphyseal fractures has been slow. This review provides an overview of the epidemiology and classification of epiphyseal fractures, as well as the post-fracture pathological changes occurring within the epiphysis and its surrounding areas. Furthermore, it reviews recent advancements in the treatment of epiphyseal fractures. By summarizing laboratory-to-clinical progress related to this type of fracture, this article aims to assist pediatric orthopedists in accurately recognizing, diagnosing, and treating such injuries.
Collapse
Affiliation(s)
- Zi-Qi Fan
- School of Medicine, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
| | - Yu-Yin Xie
- Department of Pediatric Orthopedics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Can Liu
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
| | - Yi-Fan Chen
- Department of Pediatric Orthopedics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Yang-Fei Yi
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
| | - Zhong-Wen Tang
- Department of Pediatric Orthopedics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Jie Wen
- Department of Pediatric Orthopedics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Sheng Xiao
- Department of Pediatric Orthopedics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Yu-Fei Li
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha 410005, Hunan Province, China
| |
Collapse
|
2
|
Li X, Sheng S, Li G, Hu Y, Zhou F, Geng Z, Su J. Research Progress in Hydrogels for Cartilage Organoids. Adv Healthc Mater 2024; 13:e2400431. [PMID: 38768997 DOI: 10.1002/adhm.202400431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/29/2024] [Indexed: 05/22/2024]
Abstract
The repair and regeneration of cartilage has always been a hot topic in medical research. Cartilage organoids (CORGs) are special cartilage tissue created using tissue engineering techniques outside the body. These engineered organoids tissues provide models that simulate the complex biological functions of cartilage, opening new possibilities for cartilage regenerative medicine and treatment strategies. However, it is crucial to establish suitable matrix scaffolds for the cultivation of CORGs. In recent years, utilizing hydrogel to culture stem cells and induce their differentiation into chondrocytes has emerged as a promising method for the in vitro construction of CORGs. In this review, the methods for establishing CORGs are summarized and an overview of the advantages and limitations of using matrigel in the cultivation of such organoids is provided. Furthermore, the importance of cartilage tissue ECM and alternative hydrogel substitutes for Matrigel, such as alginate, peptides, silk fibroin, and DNA derivatives is discussed, and the pros and cons of using these hydrogels for the cultivation of CORGs are outlined. Finally, the challenges and future directions in hydrogel research for CORGs are discussed. It is hoped that this article provides valuable references for the design and development of hydrogels for CORGs.
Collapse
Affiliation(s)
- Xiaolong Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics and Traumatology, Nanning Hospital of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530000, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
3
|
Yamada M, Nakajima A, Sakurai K, Tamada Y, Nakagawa K. Cell Proliferation, Chondrogenic Differentiation, and Cartilaginous Tissue Formation in Recombinant Silk Fibroin with Basic Fibroblast Growth Factor Binding Peptide. J Funct Biomater 2024; 15:230. [PMID: 39194668 DOI: 10.3390/jfb15080230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024] Open
Abstract
Regeneration of articular cartilage remains a challenge for patients who have undergone cartilage injury, osteochondritis dissecans and osteoarthritis. Here, we describe a new recombinant silk fibroin with basic fibroblast growth factor (bFGF) binding peptide, which has a genetically introduced sequence PLLQATLGGGS, named P7. In this study, we cultured a human mesenchymal cell line derived from bone marrow, UE6E7-16, in wild-type fibroin sponge (FS) and recombinant silk fibroin sponge with P7 peptide (P7 FS). We compared cell proliferation, chondrogenic differentiation and cartilaginous tissue formation between the two types of sponge. After stimulation with bFGF at 3 ng/mL, P7 FS showed significantly higher cell growth (1.2-fold) and higher cellular DNA content (5.6-fold) than did wild-type FS. To promote chondrogenic differentiation, cells were cultured in the presence of TGF-β at 10 ng/mL for 28 days. Immunostaining of P7 FS showed SOX9-positive cells comparable to wild-type FS. Alcian-Blue staining of P7 FS also showed cartilaginous tissue formation equivalent to wild-type FS. A significant increase in cell proliferation in P7 FS implies future clinical application of this transgenic fibroin for regeneration of articular cartilage. To produce cartilaginous tissue efficiently, transgenic fibroin sponges and culture conditions must be improved. Such changes should include the selection of growth factors involved in chondrogenic differentiation and cartilage formation.
Collapse
Affiliation(s)
- Manabu Yamada
- Department of Orthopaedic Surgery, Toho University Graduate School of Medicine, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan
- Department of Orthopaedic Surgery, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura, Chiba 285-0841, Japan
| | - Arata Nakajima
- Department of Orthopaedic Surgery, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura, Chiba 285-0841, Japan
- Department of Rehabilitation, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura, Chiba 285-0841, Japan
| | - Kayo Sakurai
- Department of Orthopaedic Surgery, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura, Chiba 285-0841, Japan
| | - Yasushi Tamada
- Faculty of Textile Science and Technology, Shinshu University, 3-15-1 Tokida, Ueda, Nagano 386-8567, Japan
| | - Koichi Nakagawa
- Department of Orthopaedic Surgery, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura, Chiba 285-0841, Japan
| |
Collapse
|
4
|
Mamachan M, Sharun K, Banu SA, Muthu S, Pawde AM, Abualigah L, Maiti SK. Mesenchymal stem cells for cartilage regeneration: Insights into molecular mechanism and therapeutic strategies. Tissue Cell 2024; 88:102380. [PMID: 38615643 DOI: 10.1016/j.tice.2024.102380] [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: 01/11/2024] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
The use of mesenchymal stem cells (MSCs) in cartilage regeneration has gained significant attention in regenerative medicine. This paper reviews the molecular mechanisms underlying MSC-based cartilage regeneration and explores various therapeutic strategies to enhance the efficacy of MSCs in this context. MSCs exhibit multipotent capabilities and can differentiate into various cell lineages under specific microenvironmental cues. Chondrogenic differentiation, a complex process involving signaling pathways, transcription factors, and growth factors, plays a pivotal role in the successful regeneration of cartilage tissue. The chondrogenic differentiation of MSCs is tightly regulated by growth factors and signaling pathways such as TGF-β, BMP, Wnt/β-catenin, RhoA/ROCK, NOTCH, and IHH (Indian hedgehog). Understanding the intricate balance between these pathways is crucial for directing lineage-specific differentiation and preventing undesirable chondrocyte hypertrophy. Additionally, paracrine effects of MSCs, mediated by the secretion of bioactive factors, contribute significantly to immunomodulation, recruitment of endogenous stem cells, and maintenance of chondrocyte phenotype. Pre-treatment strategies utilized to potentiate MSCs, such as hypoxic conditions, low-intensity ultrasound, kartogenin treatment, and gene editing, are also discussed for their potential to enhance MSC survival, differentiation, and paracrine effects. In conclusion, this paper provides a comprehensive overview of the molecular mechanisms involved in MSC-based cartilage regeneration and outlines promising therapeutic strategies. The insights presented contribute to the ongoing efforts in optimizing MSC-based therapies for effective cartilage repair.
Collapse
Affiliation(s)
- Merlin Mamachan
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India; Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan.
| | - S Amitha Banu
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Sathish Muthu
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India; Orthopaedic Research Group, Coimbatore, Tamil Nadu, India; Department of Orthopaedics, Government Medical College, Kaur, Tamil Nadu, India
| | - Abhijit M Pawde
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Laith Abualigah
- Artificial Intelligence and Sensing Technologies (AIST) Research Center, University of Tabuk, Tabuk 71491, Saudi Arabia; Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman 19328, Jordan; Computer Science Department, Al al-Bayt University, Mafraq 25113, Jordan; MEU Research Unit, Middle East University, Amman 11831, Jordan; Department of Electrical and Computer Engineering, Lebanese American University, Byblos 13-5053, Lebanon; Applied Science Research Center, Applied Science Private University, Amman 11931, Jordan; School of Engineering and Technology, Sunway University Malaysia, Petaling Jaya 27500, Malaysia
| | - Swapan Kumar Maiti
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| |
Collapse
|
5
|
da Rocha LR, Dias RB, Fernandes MBC, Prinz R, Eirado TP, Costa IDS, Monteiro MJ, da Silva CER, Dos Santos CT, Fogagnolo F. A new option for bone regeneration: a rapid methodology for cellularization of allograft with human bone marrow stromal cells with in vivo bone-forming potential. Injury 2023; 54 Suppl 6:110777. [PMID: 38143129 DOI: 10.1016/j.injury.2023.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/26/2023] [Accepted: 05/01/2023] [Indexed: 12/26/2023]
Abstract
The treatment of severe musculoskeletal injuries, such as loss of bone tissue and consolidation disorders, requires bone transplantation, and the success of this bone reconstruction depends on the grafts transplant's osteogenic, osteoconductive, and osteoinductive properties. Although the gold standard is autograft, it is limited by availability, morbidity, and infection risk. Despite their low capacity for osteoinduction and osteogenesis, decellularized bone allografts have been used in the search for alternative therapeutic strategies to improve bone regeneration. Considering that bone marrow stromal cells (BMSCs) are responsible for the maintenance of bone turnover throughout life, we believe that associating BMSCs with allograft could produce a material that is biologically similar to autologous bone graft. For this reason, this study evaluated the osteogenic potential of bone allograft cellularized with BMSCs. First, BMSC was characterized and allograft decellularization was confirmed by histology, scanning electron microscopy, and DNA quantification. Subsequently, the BMSCs and allografts were associated and evaluated for adhesion, proliferation, and in vitro and in vivo osteogenic potential. We demonstrated that, after 2 hours, BMSCs had already adhered to the surface of allografts and remained viable for 14 days. In vitro osteogenic assays indicated increased osteogenic potential of allografts compared with beta-tricalcium phosphate (β-TCP). In vivo transplantation assays in immunodeficient mice confirmed the allograft's potential to induce bone formation, with significantly better results than β-TCP. Finally, our results indicate that allograft can provide structural support for BMSC adhesion, offering a favorable microenvironment for cell survival and differentiation and inducing new bone formation. Taken together, our data indicate that this rapid methodology for cellularization of allograft with BMSCs might be a new therapeutic alternative in regenerative medicine and bone bioengineering.
Collapse
Affiliation(s)
- Leonardo Rosa da Rocha
- Teaching and Research Division, Instituto Nacional de Traumatologia e Ortopedia Jamil Haddad (INTO), Av. Brasil, 500, Rio de Janeiro, RJ 20940-070, Brazil.
| | - Rhayra Braga Dias
- Teaching and Research Division, INTO, Av. Brasil, 500, Rio de Janeiro, RJ 20940-070, Brazil
| | | | - Rafael Prinz
- Teaching and Research Division, INTO, Av. Brasil, 500, Rio de Janeiro, RJ 20940-070, Brazil
| | - Thiago Penna Eirado
- Teaching and Research Division, INTO, Av. Brasil, 500, Rio de Janeiro, RJ 20940-070, Brazil
| | - Isabela de Souza Costa
- Teaching and Research Division, INTO, Av. Brasil, 500, Rio de Janeiro, RJ 20940-070, Brazil
| | - Mauricio J Monteiro
- Materials Division, Instituto Nacional de Tecnologia (INT), Av. Venezuela 82, Rio de Janeiro, RJ 20081-312, Brazil.
| | | | | | - Fabricio Fogagnolo
- Department of Orthopaedics and Anaesthesiology, Ribeirão Preto Medical School, Universidade de São Paulo (USP), Av. Bandeirantes, 3900, São Paulo, SP 14049900, Brazil
| |
Collapse
|
6
|
Benavente-Perez A. Evidence of vascular involvement in myopia: a review. Front Med (Lausanne) 2023; 10:1112996. [PMID: 37275358 PMCID: PMC10232763 DOI: 10.3389/fmed.2023.1112996] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
The benign public perception of myopia (nearsightedness) as a visual inconvenience masks the severity of its sight-threatening consequences. Myopia is a significant risk factor for posterior pole conditions such as maculopathy, choroidal neovascularization and glaucoma, all of which have a vascular component. These associations strongly suggest that myopic eyes might experience vascular alterations prior to the development of complications. Myopic eyes are out of focus because they are larger in size, which in turn affects their overall structure and function, including those of the vascular beds. By reviewing the vascular changes that characterize myopia, this review aims to provide an understanding of the gross, cellular and molecular alterations identified at the structural and functional levels with the goal to provide an understanding of the latest evidence in the field of experimental and clinical myopia vascular research. From the evidence presented, we hypothesize that the interaction between excessive myopic eye growth and vascular alterations are tipping-points for the development of sight-threatening changes.
Collapse
|
7
|
Guo R, Zhuang H, Chen X, Ben Y, Fan M, Wang Y, Zheng P. Tissue engineering in growth plate cartilage regeneration: Mechanisms to therapeutic strategies. J Tissue Eng 2023; 14:20417314231187956. [PMID: 37483459 PMCID: PMC10359656 DOI: 10.1177/20417314231187956] [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: 03/02/2023] [Accepted: 06/29/2023] [Indexed: 07/25/2023] Open
Abstract
The repair of growth plate injuries is a highly complex process that involves precise spatiotemporal regulation of multiple cell types. While significant progress has been made in understanding the pathological mechanisms underlying growth plate injuries, effectively regulating this process to regenerate the injured growth plate cartilage remains a challenge. Tissue engineering technology has emerged as a promising therapeutic approach for achieving tissue regeneration through the use of functional biological materials, seed cells and biological factors, and it is now widely applied to the regeneration of bone and cartilage. However, due to the unique structure and function of growth plate cartilage, distinct strategies are required for effective regeneration. Thus, this review provides an overview of current research on the application of tissue engineering to promote growth plate regeneration. It aims to elucidates the underlying mechanisms by which tissue engineering promotes growth plate regeneration and to provide novel insights and therapeutic strategies for future research on the regeneration of growth plate.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Pengfei Zheng
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Dicks AR, Steward N, Guilak F, Wu CL. Chondrogenic Differentiation of Human-Induced Pluripotent Stem Cells. Methods Mol Biol 2023; 2598:87-114. [PMID: 36355287 PMCID: PMC9830630 DOI: 10.1007/978-1-0716-2839-3_8] [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] [Indexed: 11/12/2022]
Abstract
The generation of large quantities of genetically defined human chondrocytes remains a critical step for the development of tissue engineering strategies for cartilage regeneration and high-throughput drug screening. This protocol describes chondrogenic differentiation of human-induced pluripotent stem cells (hiPSCs), which can undergo genetic modification and the capacity for extensive cell expansion. The hiPSCs are differentiated in a stepwise manner in monolayer through the mesodermal lineage for 12 days using defined growth factors and small molecules. This is followed by 28 days of chondrogenic differentiation in a 3D pellet culture system using transforming growth factor beta 3 and specific compounds to inhibit off-target differentiation. The 6-week protocol results in hiPSC-derived cartilaginous tissue that can be characterized by histology, immunohistochemistry, and gene expression or enzymatically digested to isolate chondrocyte-like cells. Investigators can use this protocol for experiments including genetic engineering, in vitro disease modeling, or tissue engineering.
Collapse
Affiliation(s)
- Amanda R Dicks
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Nancy Steward
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA.
- Shriners Hospitals for Children - St. Louis, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA.
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA.
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery and Rehabilitation, Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
9
|
Nishino K, Hashimoto Y, Nishida Y, Orita K, Takigami J, Nakamura H. Transplantation of Parathyroid Hormone-Treated Achilles Tendon Promotes Meniscal Regeneration in a Rat Meniscal Defect Model. Am J Sports Med 2022; 50:3102-3111. [PMID: 35914290 DOI: 10.1177/03635465221112954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Autologous tendon grafts are used for meniscal reconstruction of surgically removed knee joint meniscus. However, as meniscal reconstruction cannot prevent the progression of cartilage degeneration, additional procedures that confer meniscus-like histological properties to the transplanted tendon are required for improved outcomes. HYPOTHESES Parathyroid hormone (PTH)(1-34) induces cartilage formation in the rat tendon, and transplantation of PTH-treated tendon promotes meniscal regeneration. STUDY DESIGN Controlled laboratory study. METHODS Rat Achilles tendon-derived cells were cultured with or without PTH for 28 days and stained with Alcian blue to determine chondrogenic differentiation. After 14 and 28 days of incubation, gene expression was assessed using quantitative real-time polymerase chain reaction. In an in vivo study, rat Achilles tendon was injected with PTH and then transplanted onto a medial meniscal defect. Macroscopic and histological assessments of the regenerated meniscus and of cartilage degeneration in the tibial plateau were performed at 4 and 8 weeks after surgery. RESULTS In vitro, PTH-treated cells showed better staining with Alcian blue than the control (normal medium) group. PTH1R, Col2a1, Sox9, and RUNX2 were significantly upregulated in PTH-treated cells (P < .05). Macroscopically, the in vivo results revealed more prominent meniscal coverage and lesser progression of articular cartilage degeneration in the PTH group than in the phosphate-buffered saline-injected group. Histologically, toluidine blue staining revealed metachromasia in the PTH-injected tissue at 4 and 8 weeks. The PTH-treated regenerated meniscus showed positive immunostaining for type II collagen in the area exhibiting metachromasia. Moreover, PTH-treated tendon had an enhanced histological score compared with the untreated group at 4 and 8 weeks (P < .05). CONCLUSION PTH(1-34) induced cartilage formation in the rat tendon. Transplantation of PTH(1-34)-treated Achilles tendon in a rat meniscal defect model induced meniscal regeneration and preserved knee articular cartilage. Macroscopically, PTH groups showed a greater coverage of the regenerated meniscus. Histologically, the regenerated meniscus had higher cartilaginous matrix content in rats transplanted with PTH-treated tendons. PTH(1-34) stimulated tendon-derived cells to promote chondrogenic differentiation. CLINICAL RELEVANCE Meniscal transplantation using PTH-injected autologous tendon grafts might promote meniscal regeneration and prevent progression of cartilage degeneration by stimulating chondrogenic differentiation of tendon-derived cells.
Collapse
Affiliation(s)
- Kazuya Nishino
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Hashimoto
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yohei Nishida
- Department of Orthopaedic Surgery, Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Kumi Orita
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Junsei Takigami
- Department of Orthopaedic Surgery, Shimada Hospital, Osaka, Japan
| | - Hiroaki Nakamura
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
10
|
Ke C, Li H, Yang D, Ying H, Zhu H, Wang J, Xu J, Wang L. Melatonin Attenuates the Progression of Osteoarthritis in Rats by Inhibiting Inflammation and Related Oxidative Stress on the Surface of Knee Cartilage. Orthop Surg 2022; 14:2230-2237. [PMID: 35894841 PMCID: PMC9483081 DOI: 10.1111/os.13408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/02/2022] [Accepted: 06/26/2022] [Indexed: 11/16/2022] Open
Abstract
Objective To investigate the correlation between melatonin and osteoarthritis (OA) in rats. To explore the relevant mechanisms in the occurrence and development of osteoarthritis in rats, and to further understand the disease of osteoarthritis. Methods Forty healthy 6‐month‐old male SD rats were randomly divided into two groups: sham and drug intervention groups. Pre‐OA modeling, enzyme‐linked immunosorbent assay was employed to detect the levels of IL‐1β, IL‐6, COX‐2, and melatonin in the serum of the rats in each group. For OA modeling, we administered an injection of papain into the knee cavity of all rats. The levels of IL‐1β, IL‐6, and COX‐2 in the serum of rats in each group were detected 2 weeks after the modeling. Additionally, 2 weeks after the modeling, the rats in the drug intervention group were intraperitoneally injected with melatonin antagonists. The rats in the sham group were intraperitoneally injected with normal saline for 2 weeks. The levels of IL‐1β, IL‐6, and COX‐2 in the serum of each group were measured at the second, third, and fourth weeks after the drug intervention, and the levels of melatonin in the serum were measured at the second week after the drug intervention. Finally, the rats were euthanized by cervical dislocation, and pathological sections were collected from the knee joint to observe the pathological tissue changes under a microscope, and Mankin score was determined. The independent samples t‐test method was used for analysis. Results The imaging examination after the drug intervention showed that the modeling of knee osteoarthritis in rats was successful. In the pathological findings, HE staining showed a legible cartilage structure of each layer, with cartilage proliferation and partial cartilage tearing to the radial layer. The tide line was intact; toluidine blue staining revealed more obvious changes. The differences among the mean values of IL‐6, IL‐1β, and COX‐2 measured in each period were statistically significant (t = 5.50, p < 0.05). The measured mean values of IL‐6, IL‐1β, and COX‐2 revealed statistically significant differences among the groups (t = 2.01, p < 0.05). The intergroup comparison of the Mankin scores in each period showed statistically significant differences. Conclusion Melatonin may inhibit inflammation and associated oxidative stress on the surface of knee cartilage. It may be related to the repair and regeneration of articular surface cartilage during the development of OA in the rat knee joint.
Collapse
Affiliation(s)
- Chenghui Ke
- Department of Orthopaedics, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongyun Li
- Department of Anesthesiology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Yang
- Department of Orthopaedics, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Ying
- Department of Orthopaedics, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongwen Zhu
- Tianjin Hospital, Tianjin Academy of Integrative Medicine, Tianjin, China
| | - Jian Wang
- Tongji University School of Medicine, Shanghai, China
| | - Jun Xu
- Tongji University School of Medicine, Shanghai, China
| | - Lin Wang
- Department of Orthopaedics, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
11
|
Optimization of loading protocols for tissue engineering experiments. Sci Rep 2022; 12:5094. [PMID: 35332169 PMCID: PMC8948220 DOI: 10.1038/s41598-022-08849-y] [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: 12/06/2021] [Accepted: 03/14/2022] [Indexed: 11/08/2022] Open
Abstract
Tissue engineering (TE) combines cells and biomaterials to treat orthopedic pathologies. Maturation of de novo tissue is highly dependent on local mechanical environments. Mechanical stimulation influences stem cell differentiation, however, the role of various mechanical loads remains unclear. While bioreactors simplify the complexity of the human body, the potential combination of mechanical loads that can be applied make it difficult to assess how different factors interact. Human bone marrow-derived mesenchymal stromal cells were seeded within a fibrin-polyurethane scaffold and exposed to joint-mimicking motion. We applied a full factorial design of experiment to investigate the effect that the interaction between different mechanical loading parameters has on biological markers. Additionally, we employed planned contrasts to analyze differences between loading protocols and a linear mixed model with donor as random effect. Our approach enables screening of multiple mechanical loading combinations and identification of significant interactions that could not have been studied using classical mechanobiology studies. This is useful to screen the effect of various loading protocols and could also be used for TE experiments with small sample sizes and further combinatorial medication studies.
Collapse
|
12
|
De la Fuente-Hernandez MA, Sarabia-Sanchez MA, Melendez-Zajgla J, Maldonado-Lagunas V. Role of lncRNAs into Mesenchymal Stromal Cell Differentiation. Am J Physiol Cell Physiol 2022; 322:C421-C460. [PMID: 35080923 DOI: 10.1152/ajpcell.00364.2021] [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: 11/22/2022]
Abstract
Currently, findings support that 75% of the human genome is actively transcribed, but only 2% is translated into a protein, according to databases such as ENCODE (Encyclopedia of DNA Elements) [1]. The development of high-throughput sequencing technologies, computational methods for genome assembly and biological models have led to the realization of the importance of the previously unconsidered non-coding fraction of the genome. Along with this, noncoding RNAs have been shown to be epigenetic, transcriptional and post-transcriptional regulators in a large number of cellular processes [2]. Within the group of non-coding RNAs, lncRNAs represent a fascinating field of study, given the functional versatility in their mode of action on their molecular targets. In recent years, there has been an interest in learning about lncRNAs in MSC differentiation. The aim of this review is to address the signaling mechanisms where lncRNAs are involved, emphasizing their role in either stimulating or inhibiting the transition to differentiated cell. Specifically, the main types of MSC differentiation are discussed: myogenesis, osteogenesis, adipogenesis and chondrogenesis. The description of increasingly new lncRNAs reinforces their role as players in the well-studied field of MSC differentiation, allowing a step towards a better understanding of their biology and their potential application in the clinic.
Collapse
Affiliation(s)
- Marcela Angelica De la Fuente-Hernandez
- Facultad de Medicina, Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Laboratorio de Epigenética, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Miguel Angel Sarabia-Sanchez
- Facultad de Medicina, Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jorge Melendez-Zajgla
- Laboratorio de Genómica Funcional del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | | |
Collapse
|
13
|
Wang X, Li Z, Wang C, Bai H, Wang Z, Liu Y, Bao Y, Ren M, Liu H, Wang J. Enlightenment of Growth Plate Regeneration Based on Cartilage Repair Theory: A Review. Front Bioeng Biotechnol 2021; 9:654087. [PMID: 34150725 PMCID: PMC8209549 DOI: 10.3389/fbioe.2021.654087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/10/2021] [Indexed: 01/21/2023] Open
Abstract
The growth plate (GP) is a cartilaginous region situated between the epiphysis and metaphysis at the end of the immature long bone, which is susceptible to mechanical damage because of its vulnerable structure. Due to the limited regeneration ability of the GP, current clinical treatment strategies (e.g., bone bridge resection and fat engraftment) always result in bone bridge formation, which will cause length discrepancy and angular deformity, thus making satisfactory outcomes difficult to achieve. The introduction of cartilage repair theory and cartilage tissue engineering technology may encourage novel therapeutic approaches for GP repair using tissue engineered GPs, including biocompatible scaffolds incorporated with appropriate seed cells and growth factors. In this review, we summarize the physiological structure of GPs, the pathological process, and repair phases of GP injuries, placing greater emphasis on advanced tissue engineering strategies for GP repair. Furthermore, we also propose that three-dimensional printing technology will play a significant role in this field in the future given its advantage of bionic replication of complex structures. We predict that tissue engineering strategies will offer a significant alternative to the management of GP injuries.
Collapse
Affiliation(s)
- Xianggang Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Zuhao Li
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Chenyu Wang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, China
| | - Haotian Bai
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Zhonghan Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Yuzhe Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Yirui Bao
- Department of Orthopedics, Chinese PLA 965 Hospital, Jilin, China
| | - Ming Ren
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - He Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Jincheng Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, China.,Orthopaedic Research Institute of Jilin Province, Changchun, China
| |
Collapse
|
14
|
Al-Shabrawey M, Hussein K, Wang F, Wan M, Elmasry K, Elsherbiny N, Saleh H, Yu PB, Tawfik A, Ibrahim AS. Bone Morphogenetic Protein-2 Induces Non-Canonical Inflammatory and Oxidative Pathways in Human Retinal Endothelial Cells. Front Immunol 2021; 11:568795. [PMID: 33584642 PMCID: PMC7878387 DOI: 10.3389/fimmu.2020.568795] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 12/08/2020] [Indexed: 11/13/2022] Open
Abstract
The mechanisms of diabetic retinopathy (DR), are not yet fully understood. We previously demonstrated an upregulation of retinal bone morphogenetic protein-2 (BMP2) in experimental diabetes and in retinas of diabetic human subjects. The purpose of current study was to investigate the role of non-canonical inflammatory pathway in BMP2-induced retinal endothelial cell (REC) barrier dysfunction. For this purpose, we used RT-PCR and western blotting to evaluate the levels of BMP2 signaling components (BMP2, BMP4, BMP receptors), VEGF, phosphorylated p38 MAPK and NFκB, and oxidative stress markers in cultured human retinal endothelial cells (HRECs) subjected to BMP2 (50ng/ml) for up to 24 h. Also, effect of high glucose (HG, 30mM D-glucose) on the expression of BMP2 and its downstream genes was examined in HRECs. H2-DCF is a fluorogenic dye that measures the levels of cellular reactive oxygen species (ROS) was used to measure the pro-oxidative effect of BMP2. Moreover, we evaluated the effect of inhibiting p38 and VEGF signaling on BMP2-induced HRECs barrier dysfunction by measuring the trans-endothelial cell electrical resistance (TER) using electric cell-substrate impedance sensing (ECIS). We also tested the effect of HG on the integrity of HRECs barrier in the presence or absence of inhibitors of BMP2 signaling. Our data reveals that BMP2 and high glucose upregulates BMP components of the BMP signaling pathway (SMAD effectors, BMP receptors, and TGFβ ligand itself) and induces phosphorylation of p38 MAPK and NFκB with nuclear translocation of NFκB. Inhibition of p38 or NFκB attenuated BMP2-induced VEGF expression and barrier dysfunction in HRECs. Also, inhibition of VEGFR2 attenuated BMP2-induced barrier dysfunction. Moreover, BMP2 induces generation of ROS and endothelial nitric oxide synthase (eNOS) expression and activity in HRECs. Finally, HG upregulated BMP2 and its downstream genes (SMAD, BMP4, ALKs, and TGF-β) in HRECs and BMP2 inhibitors attenuated HG-induced HRECs barrier dysfunction. Our results suggest that in addition to the regular canonical SMAD signaling BMP2 induces non-canonical inflammatory pathway in HRECs via activation of p38/NFκB pathway that causes the upregulation of VEGF and the disruption of HRECs. Inhibition of BMP2 signaling is a potential therapeutic intervention to preserve endothelial cell barrier function in DR.
Collapse
Affiliation(s)
- Mohamed Al-Shabrawey
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Ophthalmology and Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Khaled Hussein
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
- Department of Medicine and Surgery, Oral and Dental Research Division, National Research Centre, Cairo, Egypt
| | - Fang Wang
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
- Department of Traditional Chinese Medicine, School of Medicine, Jianghan University, Wuhan, China
| | - Ming Wan
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
- Department of Traditional Chinese Medicine, School of Medicine, Jianghan University, Wuhan, China
| | - Khaled Elmasry
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
- Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nehal Elsherbiny
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Heba Saleh
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Paul B. Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Amany Tawfik
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Ophthalmology and Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Ahmed S. Ibrahim
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Ophthalmology, Visual, and Anatomical Sciences, Department of Pharmacology, Wayne State University, Detroit, MI, United States
| |
Collapse
|
15
|
Fu R, Liu C, Yan Y, Li Q, Huang RL. Bone defect reconstruction via endochondral ossification: A developmental engineering strategy. J Tissue Eng 2021; 12:20417314211004211. [PMID: 33868628 PMCID: PMC8020769 DOI: 10.1177/20417314211004211] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/05/2023] Open
Abstract
Traditional bone tissue engineering (BTE) strategies induce direct bone-like matrix formation by mimicking the embryological process of intramembranous ossification. However, the clinical translation of these clinical strategies for bone repair is hampered by limited vascularization and poor bone regeneration after implantation in vivo. An alternative strategy for overcoming these drawbacks is engineering cartilaginous constructs by recapitulating the embryonic processes of endochondral ossification (ECO); these constructs have shown a unique ability to survive under hypoxic conditions as well as induce neovascularization and ossification. Such developmentally engineered constructs can act as transient biomimetic templates to facilitate bone regeneration in critical-sized defects. This review introduces the concept and mechanism of developmental BTE, explores the routes of endochondral bone graft engineering, highlights the current state of the art in large bone defect reconstruction via ECO-based strategies, and offers perspectives on the challenges and future directions of translating current knowledge from the bench to the bedside.
Collapse
Affiliation(s)
- Rao Fu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuanqi Liu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxin Yan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Chen Y, Ouyang X, Wu Y, Guo S, Xie Y, Wang G. Co-culture and Mechanical Stimulation on Mesenchymal Stem Cells and Chondrocytes for Cartilage Tissue Engineering. Curr Stem Cell Res Ther 2020; 15:54-60. [PMID: 31660820 DOI: 10.2174/1574888x14666191029104249] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/09/2019] [Accepted: 09/18/2019] [Indexed: 02/08/2023]
Abstract
Defects in articular cartilage injury and chronic osteoarthritis are very widespread and common, and the ability of injured cartilage to repair itself is limited. Stem cell-based cartilage tissue engineering provides a promising therapeutic option for articular cartilage damage. However, the application of the technique is limited by the number, source, proliferation, and differentiation of stem cells. The co-culture of mesenchymal stem cells and chondrocytes is available for cartilage tissue engineering, and mechanical stimulation is an important factor that should not be ignored. A combination of these two approaches, i.e., co-culture of mesenchymal stem cells and chondrocytes under mechanical stimulation, can provide sufficient quantity and quality of cells for cartilage tissue engineering, and when combined with scaffold materials and cytokines, this approach ultimately achieves the purpose of cartilage repair and reconstruction. In this review, we focus on the effects of co-culture and mechanical stimulation on mesenchymal stem cells and chondrocytes for articular cartilage tissue engineering. An in-depth understanding of the impact of co-culture and mechanical stimulation of mesenchymal stem cells and chondrocytes can facilitate the development of additional strategies for articular cartilage tissue engineering.
Collapse
Affiliation(s)
- Yawen Chen
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Xinli Ouyang
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Yide Wu
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Shaojia Guo
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Yongfang Xie
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| | - Guohui Wang
- Key Laboratory of Biological Medicines in Universities of Shandong Province, Weifang Medical University, Weifang, 261053, China
| |
Collapse
|
17
|
De Angelis E, Grolli S, Saleri R, Conti V, Andrani M, Berardi M, Cavalli V, Passeri B, Ravanetti F, Borghetti P. Platelet lysate reduces the chondrocyte dedifferentiation during in vitro expansion: Implications for cartilage tissue engineering. Res Vet Sci 2020; 133:98-105. [PMID: 32961475 DOI: 10.1016/j.rvsc.2020.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/08/2020] [Accepted: 08/30/2020] [Indexed: 12/13/2022]
Abstract
In vitro studies have demonstrated that platelet lysate (PL) can serve as an alternative to platelet-rich plasma (PRP) to sustain chondrocyte proliferation and production of extracellular matrix components in chondrocytes. The present study aimed to evaluate the direct effects of PL on equine articular chondrocytes in vitro in order to provide a rationale for in vivo use of PL. An in vitro cell proliferation and de-differentiation model was used: primary articular chondrocytes isolated from horse articular cartilage were cultured at low density under adherent conditions to promote cell proliferation. Chondrocytes were cultured in serum-free medium, 10% foetal bovine serum (FBS) supplemented medium, or in the presence of alginate beads containing 5%, 10% and 20% PL. Cell proliferation and gene expression of relevant chondrocyte differentiation markers were investigated. The proliferative capacity of cultured chondrocytes, was sustained more effectively at certain concentrations of PL as compared to that with FBS. In addition, as opposed to FBS, PL, particularly at percentages of 5% and 10%, could maintain the gene expression pattern of relevant chondrocyte differentiation markers. In particular, 5% PL supplementation showed the best compromise between chondrocyte proliferation capacity and maintenance of differentiation. The results of the present study provide a rationale for using PL as an alternative to FBS for in vitro expansion of chondrocytes for matrix-assisted chondrocyte implantation, construction of 3D scaffolds for tissue engineering, and treatment of damaged articular cartilage.
Collapse
Affiliation(s)
| | - Stefano Grolli
- Department of Veterinary Sciences, University of Parma, Italy
| | - Roberta Saleri
- Department of Veterinary Sciences, University of Parma, Italy
| | - Virna Conti
- Department of Veterinary Sciences, University of Parma, Italy
| | - Melania Andrani
- Department of Veterinary Sciences, University of Parma, Italy
| | - Martina Berardi
- Department of Veterinary Sciences, University of Parma, Italy
| | - Valeria Cavalli
- Department of Veterinary Sciences, University of Parma, Italy
| | | | | | - Paolo Borghetti
- Department of Veterinary Sciences, University of Parma, Italy
| |
Collapse
|
18
|
Dai G, Xiao H, Zhao C, Chen H, Liao J, Huang W. LncRNA H19 Regulates BMP2-Induced Hypertrophic Differentiation of Mesenchymal Stem Cells by Promoting Runx2 Phosphorylation. Front Cell Dev Biol 2020; 8:580. [PMID: 32903671 PMCID: PMC7438821 DOI: 10.3389/fcell.2020.00580] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
Objectives Bone morphogenetic protein 2 (BMP2) triggers hypertrophic differentiation after chondrogenic differentiation of mesenchymal stem cells (MSCs), which blocked the further application of BMP2-mediated cartilage tissue engineering. Here, we investigated the underlying mechanisms of BMP2-mediated hypertrophic differentiation of MSCs. Materials and Methods In vitro and in vivo chondrogenic differentiation models of MSCs were constructed. The expression of H19 in mouse limb was detected by fluorescence in situ hybridization (FISH) analysis. Transgenes BMP2, H19 silencing, and overexpression were expressed by adenoviral vectors. Gene expression was determined by reverse transcription and quantitative real-time PCR (RT-qPCR), Western blot, and immunohistochemistry. Correlations between H19 expressions and other parameters were calculated with Spearman’s correlation coefficients. The combination of H19 and Runx2 was identified by RNA immunoprecipitation (RIP) analysis. Results We identified that H19 expression level was highest in proliferative zone and decreased gradually from prehypertrophic zone to hypertrophic zone in mouse limbs. With the stimulation of BMP2, the highest expression level of H19 was followed after the peak expression level of Sox9; meanwhile, H19 expression levels were positively correlated with chondrogenic differentiation markers, especially in the late stage of BMP2 stimulation, and negatively correlated with hypertrophic differentiation markers. Our further experiments found that silencing H19 promoted BMP2-triggered hypertrophic differentiation through in vitro and in vivo tests, which indicated the essential role of H19 for maintaining the phenotype of BMP2-induced chondrocytes. In mechanism, we characterized that H19 regulated BMP2-mediated hypertrophic differentiation of MSCs by promoting the phosphorylation of Runx2. Conclusion These findings suggested that H19 regulates BMP2-induced hypertrophic differentiation of MSCs by promoting the phosphorylation of Runx2.
Collapse
Affiliation(s)
- Guangming Dai
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haozhuo Xiao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chen Zhao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junyi Liao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Huang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Bourebaba L, Röcken M, Marycz K. Osteochondritis dissecans (OCD) in Horses - Molecular Background of its Pathogenesis and Perspectives for Progenitor Stem Cell Therapy. Stem Cell Rev Rep 2020; 15:374-390. [PMID: 30796679 PMCID: PMC6534522 DOI: 10.1007/s12015-019-09875-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Osteochondrosis (osteochondrosis dissecans; OCD) is a disease syndrome of growing cartilage related to different clinical entities such as epiphysitis, subchondral cysts and angular carpal deformities, which occurs in growing animals of all species, including horses. Nowadays, these disorders are affecting increasing numbers of young horses worldwide. As a complex multifactorial disease, OCD is initiated when failure in cartilage canals because of existing ischemia, chondrocyte biogenesis impairment as well as biochemical and genetic disruptions occur. Recently, particular attention have been accorded to the definition of possible relations between OCD and some metabolic disorders; in this way, implication of mitochondrial dysfunctions, endoplasmic reticulum disruptions, oxidative stress or endocrinological affections are among the most considered axes for future researches. As one of the most frequent cause of impaired orthopaedic potential, which may result in a sharp decrease in athletic performances of the affected animals, and lead to the occurrence of complications such as joint fragility and laminitis, OCD remains as one of the primary causes of considerable economic losses in all sections of the equine industry. It would therefore be important to provide more information on the exact pathophysiological mechanism(s) underlying early OC(D) lesions, in order to implement innovative strategies involving the use of progenitor stem cells, which are considered nowadays as a promising approach to regenerative medicine, with the potential to treat numerous orthopaedic disorders, including osteo-degenerative diseases, for prevention and reduction of incidence of the disease, not only in horses, but also in human medicine, as the equine model is already widely accepted by the scientific community and approved by the FDA, for the research and application of cellular therapies in the treatment of human conditions.
Collapse
Affiliation(s)
- Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland. .,Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany.
| |
Collapse
|
20
|
Kronemberger GS, Dalmônico GML, Rossi AL, Leite PEC, Saraiva AM, Beatrici A, Silva KR, Granjeiro JM, Baptista LS. Scaffold- and serum-free hypertrophic cartilage tissue engineering as an alternative approach for bone repair. Artif Organs 2020; 44:E288-E299. [PMID: 31950507 DOI: 10.1111/aor.13637] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/25/2019] [Accepted: 01/10/2020] [Indexed: 12/12/2022]
Abstract
Human adipose stem/stromal cell (ASC) spheroids were used as a serum-free in vitro model to recapitulate the molecular events and extracellular matrix organization that orchestrate a hypertrophic cartilage phenotype. Induced-ASC spheroids (ø = 450 µm) showed high cell viability throughout the period of culture. The expression of collagen type X alpha 1 chain (COLXA1) and matrix metallopeptidase 13 (MMP-13) was upregulated at week 2 in induced-ASC spheroids compared with week 5 (P < .001) evaluated by quantitative real-time PCR. In accordance, secreted levels of IL-6 (P < .0001), IL-8 (P < .0001), IL-10 (P < .0001), bFGF (P < .001), VEGF (P < .0001), and RANTES (P < .0001) were the highest at week 2. Strong in situ staining for collagen type X and low staining for TSP-1 was associated with the increase of hypertrophic genes expression at week 2 in induced-ASC spheroids. Collagen type I, osteocalcin, biglycan, and tenascin C were detected at week 5 by in situ staining, in accordance with the highest expression of alkaline phosphatase (ALPL) gene and the presence of calcium deposits as evaluated by Alizarin Red O staining. Induced-ASC spheroids showed a higher force required to compression at week 2 (P < .0001). The human ASC spheroids under serum-free inducer medium and normoxic culture conditions were induced to a hypertrophic cartilage phenotype, opening a new perspective to recapitulate endochondral ossification in vivo.
Collapse
Affiliation(s)
- Gabriela S Kronemberger
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Duque de Caxias, Brazil.,Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil
| | | | | | - Paulo Emílio Correa Leite
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Antonio M Saraiva
- Laboratory of Macromolecules, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Anderson Beatrici
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Scientific and Technological Metrology Division (Dimci), National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Karina Ribeiro Silva
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - José Mauro Granjeiro
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Laboratory of Clinical Research in Odontology, Fluminense Federal University (UFF), Niterói, Brazil
| | - Leandra Santos Baptista
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Duque de Caxias, Brazil.,Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| |
Collapse
|
21
|
Dias RB, Guimarães JAM, Cury MB, Rocha LR, da Costa ES, Nogueira LP, Hochman-Mendez C, Fortuna-Costa A, Silva AKF, Cunha KS, de Souza SAL, Duarte MEL, Sartore RC, Bonfim DC. The Manufacture of GMP-Grade Bone Marrow Stromal Cells with Validated In Vivo Bone-Forming Potential in an Orthopedic Clinical Center in Brazil. Stem Cells Int 2019; 2019:2608482. [PMID: 31781235 PMCID: PMC6875385 DOI: 10.1155/2019/2608482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/26/2019] [Accepted: 09/18/2019] [Indexed: 12/30/2022] Open
Abstract
In vitro-expanded bone marrow stromal cells (BMSCs) have long been proposed for the treatment of complex bone-related injuries because of their inherent potential to differentiate into multiple skeletal cell types, modulate inflammatory responses, and support angiogenesis. Although a wide variety of methods have been used to expand BMSCs on a large scale by using good manufacturing practice (GMP), little attention has been paid to whether the expansion procedures indeed allow the maintenance of critical cell characteristics and potency, which are crucial for therapeutic effectiveness. Here, we described standard procedures adopted in our facility for the manufacture of clinical-grade BMSC products with a preserved capacity to generate bone in vivo in compliance with the Brazilian regulatory guidelines for cells intended for use in humans. Bone marrow samples were obtained from trabecular bone. After cell isolation in standard monolayer flasks, BMSC expansion was subsequently performed in two cycles, in 2- and 10-layer cell factories, respectively. The average cell yield per cell factory at passage 1 was of 21.93 ± 12.81 × 106 cells, while at passage 2, it was of 83.05 ± 114.72 × 106 cells. All final cellular products were free from contamination with aerobic/anaerobic pathogens, mycoplasma, and bacterial endotoxins. The expanded BMSCs expressed CD73, CD90, CD105, and CD146 and were able to differentiate into osteogenic, chondrogenic, and adipogenic lineages in vitro. Most importantly, nine out of 10 of the cell products formed bone when transplanted in vivo. These validated procedures will serve as the basis for in-house BMSC manufacturing for use in clinical applications in our center.
Collapse
Affiliation(s)
- Rhayra B. Dias
- Master Program in Musculoskeletal Sciences, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - João A. M. Guimarães
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
- Trauma Center, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Marco B. Cury
- Hip Surgery Center, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Leonardo R. Rocha
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
- Trauma Center, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Elaine S. da Costa
- Institute of Paediatrics and Puericulture Martagão Gesteira, Federal University of Rio de Janeiro, Rio de Janeiro 21941-912, Brazil
| | | | - Camila Hochman-Mendez
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Texas Heart Institute, Regenerative Medicine Research, Texas 77030, USA
| | - Anneliese Fortuna-Costa
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Anna Karoline F. Silva
- Graduate Program in Pathology, Fluminense Federal University, Rio de Janeiro 24030-215, Brazil
| | - Karin S. Cunha
- Graduate Program in Pathology, Fluminense Federal University, Rio de Janeiro 24030-215, Brazil
| | - Sergio A. L. de Souza
- Department of Radiology, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Maria Eugênia L. Duarte
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Rafaela C. Sartore
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| | - Danielle C. Bonfim
- Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940-070, Brazil
| |
Collapse
|
22
|
Nasiri N, Hosseini S, Alini M, Khademhosseini A, Baghaban Eslaminejad M. Targeted cell delivery for articular cartilage regeneration and osteoarthritis treatment. Drug Discov Today 2019; 24:2212-2224. [PMID: 31398399 DOI: 10.1016/j.drudis.2019.07.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 12/17/2022]
|
23
|
Li H, Yue B. Effects of various antimicrobial agents on multi-directional differentiation potential of bone marrow-derived mesenchymal stem cells. World J Stem Cells 2019; 11:322-336. [PMID: 31293715 PMCID: PMC6600849 DOI: 10.4252/wjsc.v11.i6.322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/30/2019] [Accepted: 05/23/2019] [Indexed: 02/06/2023] Open
Abstract
Antimicrobial drugs of several classes play an important role in the treatment of bone and joint infections. In addition to fighting pathogenic microorganisms, the effects of drugs on local tissues and cells are also related to the course and prognosis of bone and joint infections. The multi-directional differentiation potential of bone marrow-derived mesenchymal stem cells (MSCs) is essential for tissue repair after local injury, which is directly related to the recovery of bone, cartilage, and medullary adipose tissue. Our previous studies and the literature indicate that certain antimicrobial agents can regulate the differentiation potential of bone marrow-derived MSCs. Here, in order to systematically analyze the effects of various antimicrobial drugs on local tissue regeneration, we comprehensively review the studies on the effects of these drugs on MSC differentiation, and classify them according to the three differentiation directions (osteogenesis, chondrogenesis, and adipogenesis). Our review demonstrates the specific effects of different antimicrobial agents on bone marrow-derived MSCs and the range of concentrations at which they work, and provides a basis for drug selection at different sites of infection.
Collapse
Affiliation(s)
- Hui Li
- Department of Bone and Joint Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
24
|
Schliermann A, Nickel J. Unraveling the Connection between Fibroblast Growth Factor and Bone Morphogenetic Protein Signaling. Int J Mol Sci 2018; 19:ijms19103220. [PMID: 30340367 PMCID: PMC6214098 DOI: 10.3390/ijms19103220] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/07/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022] Open
Abstract
Ontogeny of higher organisms as well the regulation of tissue homeostasis in adult individuals requires a fine-balanced interplay of regulating factors that individually trigger the fate of particular cells to either stay undifferentiated or to differentiate towards distinct tissue specific lineages. In some cases, these factors act synergistically to promote certain cellular responses, whereas in other tissues the same factors antagonize each other. However, the molecular basis of this obvious dual signaling activity is still only poorly understood. Bone morphogenetic proteins (BMPs) and fibroblast growth factors (FGFs) are two major signal protein families that have a lot in common: They are both highly preserved between different species, involved in essential cellular functions, and their ligands vastly outnumber their receptors, making extensive signal regulation necessary. In this review we discuss where and how BMP and FGF signaling cross paths. The compiled data reflect that both factors synchronously act in many tissues, and that antagonism and synergism both exist in a context-dependent manner. Therefore, by challenging a generalization of the connection between these two pathways a new chapter in BMP FGF signaling research will be introduced.
Collapse
Affiliation(s)
- Anna Schliermann
- Lehrstuhl für Tissue Engineering und Regenerative Medizin, Universitätsklinikum Würzburg, Röntgenring 11, 97222 Würzburg, Germany.
| | - Joachim Nickel
- Lehrstuhl für Tissue Engineering und Regenerative Medizin, Universitätsklinikum Würzburg, Röntgenring 11, 97222 Würzburg, Germany.
- Fraunhofer Institut für Silicatforschung, Translationszentrum TLZ-RT, Röntgenring 11, 97222 Würzburg, Germany.
| |
Collapse
|