1
|
Semerci Sevimli T, Inan U, Mantar D, Guler K, Ahmadova Z, Gulec K, Topal AE. In vitro Chondrogenic Induction Promotes the Expression Level of IL-10 via the TGF-β/SMAD and Canonical Wnt/β-catenin Signaling Pathways in Exosomes Secreted by Human Adipose Tissue-derived Mesenchymal Stem Cells. Cell Biochem Biophys 2024; 82:3741-3750. [PMID: 39266872 DOI: 10.1007/s12013-024-01461-z] [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] [Accepted: 07/23/2024] [Indexed: 09/14/2024]
Abstract
Current treatment approaches cannot exactly regenerate cartilage tissue. Regarding some problems encountered with cell therapy, exosomes are advantageous because of their "cell-free" nature. This study examines the relationship between IL-10 and TGF-β and Canonical Wnt/β-catenin signal pathways in human adipose tissue-derived MSCs exosomes (hAT-MSCs-Exos) after in vitro chondrogenic differentiation. Human adipose tissue-derived mesenchymal stem cells (hAT-MSCs) and, as a control group, human fetal chondroblast cells (hfCCs) were differentiated chondrogenically in vitro. Exosome isolation and characterization analyses were performed. Chondrogenic differentiation was shown by Alcian Blue and Safranin O stainings. The expression levels of IL-10, TGF-β/SMAD signaling pathway genes, and Canonical Wnt/β-catenin signaling pathway genes, which play an essential role in chondrogenesis, were analyzed by RT-qPCR. Conditioned media cytokine levels were measured by using the TGF-β and IL-10 ELISA kits. IL-10 expression was upregulated in both chondrogenic differentiated hAT-MSC-Exos (dhAT-MSC-Exos) (p < 0.0001). In the TGF-β signaling pathway, TGF-β (p < 0.0001), SMAD2 (p < 0.0001), SMAD4 (p < 0.001), ACAN (p < 0.0001), SOX9 (p < 0.05) and COL1A2 (p < 0.0001) expressions were upregulated in dhAT-MSC-Exos. SMAD3 expression was upregulated in non-differentiated hAT-MSC-Exos. In the Canonical Wnt/β-catenin signaling pathway, WNT (p < 0.0001) and CTNNB1(p < 0.0001) expressions were upregulated in dhAT-MSC-Exos. AXIN (p < 0.0001) expression was upregulated in non-differentiated hAT-MSC-Exos. TGF-β and IL-10 levels were higher in dhAT-MSCs) (p < 0.0001). Related to these results, IL-10 may induce TGF-β/SMAD and Canonical Wnt/β-catenin signaling pathways in hAT-MSC exosomes obtained after chondrogenic differentiation. Therefore, using these exosomes for cartilage regeneration can lead to the development of treatment methods.
Collapse
Affiliation(s)
- Tugba Semerci Sevimli
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040, Eskisehir, Turkey.
| | - Ulukan Inan
- Department of Orthopedics and Traumatology, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | | | - Kubra Guler
- Department of Biochemistry, School of Pharmacy, Bahcesehir University, Istanbul, Turkey
| | - Zarifa Ahmadova
- Department of Surgery, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Kadri Gulec
- Department of Analytical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskisehir, Turkey
| | - Ahmet Emin Topal
- Department of Biochemistry, School of Pharmacy, Bahcesehir University, Istanbul, Turkey
| |
Collapse
|
2
|
Wu J, Wu J, Xiang W, Gong Y, Feng D, Fang S, Wu Y, Liu Z, Li Y, Chen R, Zhang X, Li B, Chen L, Jin R, Li S, Zhang B, Zhang T, Yin L, Zhou Y, Huang S, Liu N, Xu H, Lian J, Wang Y, Zhou S, Ni Z. Engineering exosomes derived from TNF-α preconditioned IPFP-MSCs enhance both yield and therapeutic efficacy for osteoarthritis. J Nanobiotechnology 2024; 22:555. [PMID: 39261846 PMCID: PMC11391629 DOI: 10.1186/s12951-024-02795-9] [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: 03/20/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND The pathogenesis of osteoarthritis (OA) involves the progressive degradation of articular cartilage. Exosomes derived from mesenchymal stem cells (MSC-EXOs) have been shown to mitigate joint pathological injury by attenuating cartilage destruction. Optimization the yield and therapeutic efficacy of exosomes derived from MSCs is crucial for promoting their clinical translation. The preconditioning of MSCs enhances the therapeutic potential of engineered exosomes, offering promising prospects for application by enabling controlled and quantifiable external stimulation. This study aims to address these issues by employing pro-inflammatory preconditioning of MSCs to enhance exosome production and augment their therapeutic efficacy for OA. METHODS The exosomes were isolated from the supernatant of infrapatellar fat pad (IPFP)-MSCs preconditioned with a pro-inflammatory factor, TNF-α, and their production was subsequently quantified. The exosome secretion-related pathways in IPFP-MSCs were evaluated through high-throughput transcriptome sequencing analysis, q-PCR and western blot analysis before and after TNF-α preconditioning. Furthermore, exosomes derived from TNF-α preconditioned IPFP-MSCs (IPFP-MSC-EXOsTNF-α) were administered intra-articularly in an OA mouse model, and subsequent evaluations were conducted to assess joint pathology and gait alterations. The expression of proteins involved in the maintenance of cartilage homeostasis within the exosomes was determined through proteomic analysis. RESULTS The preconditioning with TNF-α significantly enhanced the exosome secretion of IPFP-MSCs compared to unpreconditioned MSCs. The potential mechanism involved the activation of the PI3K/AKT signaling pathway in IPFP-MSCs by TNF-α precondition, leading to an up-regulation of autophagy-related protein 16 like 1(ATG16L1) levels, which subsequently facilitated exosome secretion. The intra-articular administration of IPFP-MSC-EXOsTNF-α demonstrated superior efficacy in ameliorating pathological changes in the joints of OA mice. The preconditioning of TNF-α enhanced the up-regulation of low-density lipoprotein receptor-related protein 1 (LRP1) levels in IPFP-MSC-EXOsTNF-α, thereby exerting chondroprotective effects. CONCLUSION TNF-α preconditioning constitutes an effective and promising method for optimizing the therapeutic effects of IPFP-MSCs derived exosomes in the treatment of OA.
Collapse
Affiliation(s)
- Jiangyi Wu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Jinhui Wu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Wei Xiang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Yunquan Gong
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
- Department of Orthopedics, Shanghai Hospital, Wanzhou District, Chongqing, 40400, China
| | - Daibo Feng
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Shunzheng Fang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Yaran Wu
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing, 400038, China
| | - Zheng Liu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Yang Li
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Ran Chen
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Xiaoqi Zhang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Bingfei Li
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Lifeng Chen
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Runze Jin
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Song Li
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400000, China
| | - Bin Zhang
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400000, China
- Rehabilitation Center, Strategic Support Force Xingcheng Special Duty Sanatorium, Xingcheng, 125105, China
| | - Tongyi Zhang
- Department of General Practice, Chinese PLA General Hospital of the Central Theater Command, Wuhan, 430012, China
| | - Lin Yin
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Yizhao Zhou
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Shu Huang
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Ningning Liu
- Department of Laboratory Medicine, The Fifth Clinical Medical College of, Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, 450003, China
| | - Hao Xu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Jiqin Lian
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing, 400038, China
| | - Yongqian Wang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China.
| | - Siru Zhou
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China.
| | - Zhenhong Ni
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China.
| |
Collapse
|
3
|
Lan X, Boluk Y, Adesida AB. 3D Bioprinting of Hyaline Cartilage Using Nasal Chondrocytes. Ann Biomed Eng 2024; 52:1816-1834. [PMID: 36952145 DOI: 10.1007/s10439-023-03176-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/22/2023] [Indexed: 03/24/2023]
Abstract
Due to the limited self-repair capacity of the hyaline cartilage, the repair of cartilage remains an unsolved clinical problem. Tissue engineering strategy with 3D bioprinting technique has emerged a new insight by providing patient's personalized cartilage grafts using autologous cells for hyaline cartilage repair and regeneration. In this review, we first summarized the intrinsic property of hyaline cartilage in both maxillofacial and orthopedic regions to establish the requirement for 3D bioprinting cartilage tissue. We then reviewed the literature and provided opinion pieces on the selection of bioprinters, bioink materials, and cell sources. This review aims to identify the current challenges for hyaline cartilage bioprinting and the directions for future clinical development in bioprinted hyaline cartilage.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Yaman Boluk
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada.
| | - Adetola B Adesida
- Department of Surgery, Divisions of Orthopedic Surgery & Surgical Research, Faculty of Medicine & Dentistry, Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, AB, Canada.
- Department of Surgery, Division of Otolaryngology, Faculty of Medicine & Dentistry, Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
4
|
Sirous S, Aghamohseni MM, Farhad SZ, Beigi M, Ostadsharif M. Mesenchymal stem cells in PRP and PRF containing poly(3-caprolactone)/gelatin Scaffold: a comparative in-vitro study. Cell Tissue Bank 2024; 25:559-570. [PMID: 38363442 DOI: 10.1007/s10561-023-10116-x] [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: 02/18/2023] [Accepted: 10/09/2023] [Indexed: 02/17/2024]
Abstract
Scaffold design is one of the three most essential parts of tissue engineering. Platelet-rich plasma (PRP) and platelet-rich fibrin (PRF) have been used in clinics and regenerative medicine for years. However, the temporal release of their growth factors limits their efficacy in tissue engineering. In the present study, we planned to synthesize nanofibrous scaffolds with the incorporation of PRP and PRF by electrospinning method to evaluate the effect of the release of PRP and PRF growth factors on osteogenic gene expression, calcification, proliferation, and cell adhesion of human bone marrow mesenchymal stem cell (h-BMSC) as they are part of scaffold structures. Therefore, we combined PRP/PRF, derived from the centrifugation of whole blood, with gelatin and Polycaprolactone (PCL) and produced nanofibrous electrospun PCL/Gel/PRP and PCL/Gel/PRF scaffolds. Three groups of scaffolds were fabricated, and h-BMSCs were seeded on them: (1) PCL/Gel; (2) PCL/Gel/PRP; (3) PCL/Gel/PRF. MTS assay was performed to assess cell proliferation and adhesion, and alizarin red staining confirmed the formation of bone minerals during the experiment. The result indicated that PCL/Gel did not have any better outcomes than the PRP and PRF group in any study variants after the first day of the experiment. PCL/gelatin/PRF was more successful regarding cell proliferation and adhesion. Although PCL/gelatin/PRP showed more promising results on the last day of the experiment in mineralization and osteogenic gene expression, except RUNX2, in which the difference with PCL/gelatin/PRF group was not significant.
Collapse
Affiliation(s)
- Samin Sirous
- Periodontics preceptor, UCLA School of Dentistry, Los Angeles, USA
- School of Dentistry, Islamic Azad University (Khorasgan branch), Isfahan, Iran
| | - Mohammad Mostafa Aghamohseni
- School of Dentistry, Islamic Azad University (Khorasgan branch), Isfahan, Iran.
- Chairman of Student Research Committee, Islamic Azad University (Khorasgan branch), Isfahan, Iran.
| | - Shirin Zahra Farhad
- Department of Periodontics, Faculty of Dentistry, Isfahan (Khorasgan) branch, Islamic Azad University, Isfahan, Iran
| | - Mohammadhossein Beigi
- Silicon Hall: Micro/Nano Manufacturing Facility, Faculty of Engineering and Applied Science, Ontario Tech University, Ontario, Canada
| | - Maryam Ostadsharif
- Department of Medical Basic Sciences, Isfahan(Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| |
Collapse
|
5
|
Wang Z, Lu J, Ge H, Li Z, Zhang M, Pan F, Wang R, Jin H, Yang G, Shen Z, Du G, Zhan H. Morphology and transverse alignment of the patella have no effect on knee gait characteristics in healthy Chinese adults over the age of 40 years. Front Bioeng Biotechnol 2024; 12:1319602. [PMID: 38562671 PMCID: PMC10982314 DOI: 10.3389/fbioe.2024.1319602] [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: 10/11/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Background: The influence of patella morphology and horizontal alignment on knee joint kinematics and kinetics remains uncertain. This study aimed to assess patella morphology and transverse alignment in relation to knee kinetics and kinematics in individuals without knee conditions. A secondary objective was to investigate the impact of femur and tibia alignment and shape on knee gait within this population. Patients and methods: We conducted a prospective collection of data, including full-leg anteroposterior and skyline X-ray views and three-dimensional gait data, from a cohort comprising 54 healthy individuals aged 40 years and older. Our study involved correlation and logistic regression analyses to examine the influence of patella, femur, and tibia morphology and alignment on knee gait. Results: The patellar tilt angle or the patella index did not show any significant relationships with different aspects of gait in the knee joint, such as velocity, angle, or moment (p > 0.05, respectively). Using multivariate logistic regression analysis, we found that the tibiofemoral angle and the Q angle both had a significant effect on the adduction angle (OR = 1.330, 95%CI 1.033-1.711, p = 0.027; OR = 0.475, 95%CI 0.285-0.792, p = 0.04; respectively). The primary variable influencing the knee adduction moment was the tibiofemoral angle (OR = 1.526, 95% CI 1.125-2.069, p = 0.007). Conclusion: In healthy Chinese individuals aged over 40, patella morphology and transverse alignment do not impact knee gait. However, the femoral-tibial angle has a big impact on the knee adduction moment.
Collapse
Affiliation(s)
- Zhengming Wang
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Jiehang Lu
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Haiya Ge
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Zhengyan Li
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Min Zhang
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Fuwei Pan
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Department of Massage, Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Rui Wang
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hengkai Jin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Guangyue Yang
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhibi Shen
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Guoqing Du
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongsheng Zhan
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Traumatology and Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Yang F, Xiong WQ, Li CZ, Wu MJ, Zhang XZ, Ran CX, Li ZH, Cui Y, Liu BY, Zhao DW. Extracellular vesicles derived from mesenchymal stem cells mediate extracellular matrix remodeling in osteoarthritis through the transport of microRNA-29a. World J Stem Cells 2024; 16:191-206. [PMID: 38455098 PMCID: PMC10915956 DOI: 10.4252/wjsc.v16.i2.191] [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: 10/26/2023] [Revised: 11/18/2023] [Accepted: 01/30/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is a common orthopedic condition with an uncertain etiology, possibly involving genetics and biomechanics. Factors like changes in chondrocyte microenvironment, oxidative stress, inflammation, and immune responses affect KOA development. Early-stage treatment options primarily target symptom relief. Mesenchymal stem cells (MSCs) show promise for treatment, despite challenges. Recent research highlights microRNAs (miRNAs) within MSC-released extracellular vesicles that can potentially promote cartilage regeneration and hinder KOA progression. This suggests exosomes (Exos) as a promising avenue for future treatment. While these findings emphasize the need for effective KOA progression management, further safety and efficacy validation for Exos is essential. AIM To explore miR-29a's role in KOA, we'll create miR-29a-loaded vesicles, testing for early treatment in rat models. METHODS Extraction of bone marrow MSC-derived extracellular vesicles, preparation of engineered vesicles loaded with miR-29a using ultrasonication, and identification using quantitative reverse transcription polymerase chain reaction; after establishing a rat model of KOA, rats were randomly divided into three groups: Blank control group injected with saline, normal extracellular vesicle group injected with normal extracellular vesicle suspension, and engineered extracellular vesicle group injected with engineered extracellular vesicle suspension. The three groups were subjected to general behavioral observation analysis, imaging evaluation, gross histological observation evaluation, histological detection, and immunohistochemical detection to compare and evaluate the progress of various forms of arthritis. RESULTS General behavioral observation results showed that the extracellular vesicle group and engineered extracellular vesicle group had better performance in all four indicators of pain, gait, joint mobility, and swelling compared to the blank control group. Additionally, the engineered extracellular vesicle group had better pain relief at 4 wk and better knee joint mobility at 8 wk compared to the normal extracellular vesicle group. Imaging examination results showed that the blank control group had the fastest progression of arthritis, the normal extracellular vesicle group had a relatively slower progression, and the engineered extracellular vesicle group had the slowest progression. Gross histological observation results showed that the blank control group had the most obvious signs of arthritis, the normal extracellular vesicle group showed signs of arthritis, and the engineered extracellular vesicle group showed no significant signs of arthritis. Using the Pelletier gross score evaluation, the engineered extracellular vesicle group had the slowest progression of arthritis. Results from two types of staining showed that the articular cartilage of rats in the normal extracellular vesicle and engineered extracellular vesicle groups was significantly better than that of the blank control group, and the engineered extracellular vesicle group had the best cartilage cell and joint surface condition. Immunohistochemical detection of type II collagen and proteoglycan showed that the extracellular matrix of cartilage cells in the normal extracellular vesicle and engineered extracellular vesicle groups was better than that of the blank control group. Compared to the normal extracellular vesicle group, the engineered extracellular vesicle group had a better regulatory effect on the extracellular matrix of cartilage cells. CONCLUSION Engineered Exos loaded with miR-29a can exert anti-inflammatory effects and maintain extracellular matrix stability, thereby protecting articular cartilage, and slowing the progression of KOA.
Collapse
Affiliation(s)
- Fan Yang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Wan-Qi Xiong
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Chen-Zhi Li
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Ming-Jian Wu
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Xiu-Zhi Zhang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Chun-Xiao Ran
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Zhen-Hao Li
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Yan Cui
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| | - Bao-Yi Liu
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China.
| | - De-Wei Zhao
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning Province, China
| |
Collapse
|
7
|
Kaonis S, Aboellail Z, Forman J, Ghosh S. High-Throughput Multiparametric Quantification of Mechanics Driven Heterogeneity in Mesenchymal Stromal Cell Population. Adv Biol (Weinh) 2024; 8:e2300318. [PMID: 37840408 DOI: 10.1002/adbi.202300318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Indexed: 10/17/2023]
Abstract
Mesenchymal stromal or stem cells (MSCs) are one of the most promising candidates for a myriad of cell therapy applications. Despite showing promise in numerous preclinical and clinical studies, MSC-based therapy is not yet a reality for regenerative medicine due to its suboptimal outcome at the clinical endpoint. The mechanical environment is a critical determinant of MSC gene expression and function. This study reports that MSC population becomes phenotypically heterogenous and commits to an unwanted osteoprogenitor pathway when it experiences an abnormal mechanically stiff environment, compared to its native softer environment. A method is developed to measure the heterogeneity using nuclear shape, chromatin state, and CD73 marker. Heterogeneity is shown to be associated with a larger spread in the nuclear shape parameters and a smaller spread in the chromatin openness. Subsequently, intervention strategies are investigated to create a more homogeneous MSC population. Culturing MSCs on soft surfaces or inhibiting actomyosin on stiff surfaces can make them more homogeneous, while inhibiting YAP, Runx2, and actin polymerization helps maintain but does not fully homogenize them. This study offers insights for cell and tissue engineers, aiding in the design of optimal conditions and materials for MSC culture, ultimately enhancing their therapeutic potential.
Collapse
Affiliation(s)
- Samantha Kaonis
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
| | - Zack Aboellail
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
| | - Jack Forman
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
| | - Soham Ghosh
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Mechanical Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
- Cell and Molecular Biology, Colorado State University, 1050 Libbie Coy Way, Fort Collins, CO, 80524, USA
| |
Collapse
|
8
|
Huang S, Liu Y, Wang C, Xiang W, Wang N, Peng L, Jiang X, Zhang X, Fu Z. Strategies for Cartilage Repair in Osteoarthritis Based on Diverse Mesenchymal Stem Cells-Derived Extracellular Vesicles. Orthop Surg 2023; 15:2749-2765. [PMID: 37620876 PMCID: PMC10622303 DOI: 10.1111/os.13848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023] Open
Abstract
Osteoarthritis (OA) causes disability and significant economic and social burden. Cartilage injury is one of the main pathological features of OA, and is often manifested by excessive chondrocyte death, inflammatory response, abnormal bone metabolism, imbalance of extracellular matrix (ECM) metabolism, and abnormal vascular or nerve growth. Regrettably, due to the avascular nature of cartilage, its capacity to repair is notably limited. Mesenchymal stem cells-derived extracellular vesicles (MSCs-EVs) play a pivotal role in intercellular communication, presenting promising potential not only as early diagnostic biomarkers in OA but also as efficacious therapeutic strategy. MSCs-EVs were confirmed to play a therapeutic role in the pathological process of cartilage injury mentioned above. This paper comprehensively provides the functions and mechanisms of MSCs-EVs in cartilage repair.
Collapse
Affiliation(s)
- Shanjun Huang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Yujiao Liu
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Chenglong Wang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Wei Xiang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Nianwu Wang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Li Peng
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Xuanang Jiang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Xiaomin Zhang
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| | - Zhijiang Fu
- Orthopedics DepartmentThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
9
|
Cong B, Sun T, Zhao Y, Chen M. Current and Novel Therapeutics for Articular Cartilage Repair and Regeneration. Ther Clin Risk Manag 2023; 19:485-502. [PMID: 37360195 PMCID: PMC10290456 DOI: 10.2147/tcrm.s410277] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/28/2023] [Indexed: 06/28/2023] Open
Abstract
Articular cartilage repair is a sophisticated process that has is being recently investigated. There are several different approaches that are currently reported to promote cartilage repair, like cell-based therapies, biologics, and physical therapy. Cell-based therapies involve the using stem cells or chondrocytes, which make up cartilage, to promote the growth of new cartilage. Biologics, like growth factors, are also being applied to enhance cartilage repair. Physical therapy, like exercise and weight-bearing activities, can also be used to promote cartilage repair by inducing new cartilage growth and improving joint function. Additionally, surgical options like osteochondral autograft, autologous chondrocyte implantation, microfracture, and others are also reported for cartilage regeneration. In the current literature review, we aim to provide an up-to-date discussion about these approaches and discuss the current research status.
Collapse
Affiliation(s)
- Bo Cong
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
- Yantai Key Laboratory for Repair and Reconstruction of Bone & Joint, Yantai, 264003, People’s Republic of China
| | - Tao Sun
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
- Yantai Key Laboratory for Repair and Reconstruction of Bone & Joint, Yantai, 264003, People’s Republic of China
| | - Yuchi Zhao
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
- Yantai Key Laboratory for Repair and Reconstruction of Bone & Joint, Yantai, 264003, People’s Republic of China
| | - Mingqi Chen
- Department of Orthopedics, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, 264003, People’s Republic of China
| |
Collapse
|
10
|
Taninaka A, Kabata T, Hayashi K, Kajino Y, Inoue D, Ohmori T, Ueoka K, Yamamuro Y, Kataoka T, Saiki Y, Yanagi Y, Ima M, Iyobe T, Tsuchiya H. Chondroprotective Effects of Chondrogenic Differentiated Adipose-Derived Mesenchymal Stem Cells Sheet on Degenerated Articular Cartilage in an Experimental Rabbit Model. Bioengineering (Basel) 2023; 10:bioengineering10050574. [PMID: 37237645 DOI: 10.3390/bioengineering10050574] [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: 03/30/2023] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) have been studied for many years as a therapeutic option for osteoarthritis (OA); however, their efficacy remains insufficient. Since platelet-rich plasma (PRP) induces chondrogenic differentiation in ADSCs and the formation of a sheet structure by ascorbic acid can increase the number of viable cells, we hypothesized that the injection of chondrogenic cell sheets combined with the effects of PRP and ascorbic acid may hinder the progression of OA. The effects of induction of differentiation by PRP and formation of sheet structure by ascorbic acid on changes in chondrocyte markers (collagen II, aggrecan, Sox9) in ADSCs were evaluated. Changes in mucopolysaccharide and VEGF-A secretion from cells injected intra-articularly in a rabbit OA model were also evaluated. ADSCs treated by PRP strongly chondrocyte markers, including type II collagen, Sox9, and aggrecan, and their gene expression was maintained even after sheet-like structure formation induced by ascorbic acid. In this rabbit OA model study, the inhibition of OA progression by intra-articular injection was improved by inducing chondrocyte differentiation with PRP and sheet structure formation with ascorbic acid in ADSCs.
Collapse
Affiliation(s)
- Atsushi Taninaka
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Tamon Kabata
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Katsuhiro Hayashi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Yoshitomo Kajino
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Daisuke Inoue
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Takaaki Ohmori
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Ken Ueoka
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Yuki Yamamuro
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Tomoyuki Kataoka
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Yoshitomo Saiki
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Yu Yanagi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Musashi Ima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Takahiro Iyobe
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8641, Japan
| |
Collapse
|
11
|
Chen Z, Ding W, Duan P, Lv X, Feng Y, Yin Z, Luo Z, Li Z, Zhang H, Zhou T, Tan H. HWJMSC-derived extracellular vesicles ameliorate IL-1β-induced chondrocyte injury through regulation of the BMP2/RUNX2 axis via up-regulation TFRC. Cell Signal 2023; 105:110604. [PMID: 36669606 DOI: 10.1016/j.cellsig.2023.110604] [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: 09/19/2022] [Revised: 12/29/2022] [Accepted: 01/15/2023] [Indexed: 01/19/2023]
Abstract
Articular osteochondral injury is a common and frequently occurring disease in orthopedics that is caused by aging, disease, and trauma. The cytokine interleukin-1β (IL-1β) is a crucial mediator of the inflammatory response, which exacerbates damage during chronic disease and acute tissue injury. Human Wharton's jelly mesenchymal stem cell (HWJMSC) extracellular vesicles (HWJMSC-EVs) have been shown to promote cartilage regeneration. The study aimed to investigate the influence and mechanisms of HWJMSC-EVs on the viability, apoptosis, and cell cycle of IL-1β-induced chondrocytes. HWJMSC-EVs were isolated by Ribo™ Exosome Isolation Reagent kit. Nanoparticle tracking analysis was used to determine the size and concentration of HWJMSC-EVs. We characterized HWJMSC-EVs by western blot and transmission electron microscope. The differentiation, viability, and protein level of chondrocytes were measured by Alcian blue staining, Cell Counting Kit-8, and western blot, respectively. Flow cytometer was used to determine apoptosis and cell cycle of chondrocytes. The results showed that HWJMSCs relieved IL-1β-induced chondrocyte injury by inhibiting apoptosis and elevating viability and cell cycle of chondrocyte, which was reversed with exosome inhibitor (GW4869). HWJMSC-EVs were successfully extracted and proven to be uptake by chondrocytes. HWJMSC-EVs ameliorate IL-1β-induced chondrocyte injury by inhibiting cell apoptosis and elevating viability and cycle of cell, but these effects were effectively reversed by knockdown of transferrin receptor (TFRC). Notably, using bone morphogenetic protein 2 (BMP2) pathway agonist and inhibitor suggested that HWJMSC-EVs ameliorate IL-1β-induced chondrocyte injury through activating the BMP2 pathway via up-regulation TFRC. Furthermore, over-expression of runt-related transcription factor 2 (RUNX2) reversed the effects of BMP2 pathway inhibitor promotion of IL-1β-induced chondrocyte injury. These results suggested that HWJMSC-EVs ameliorate IL-1β-induced chondrocyte injury by regulating the BMP2/RUNX2 axis via up-regulation TFRC. HWJMSC-EVs may play a new insight for early medical interventions in patients with articular osteochondral injury.
Collapse
Affiliation(s)
- Zhian Chen
- Graduate School, Kunming Medical University, Kunming City, Yunnan Province, China
| | - Wei Ding
- College of Medicine Technology, Yunnan Medical Health College, Kunming City, Yunnan Province, China
| | - Peiya Duan
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming City, Yunnan Province, China
| | - Xiaoyu Lv
- Graduate School, Kunming Medical University, Kunming City, Yunnan Province, China
| | - Yujiao Feng
- Graduate School, Kunming Medical University, Kunming City, Yunnan Province, China
| | - Zhengbo Yin
- Graduate School, Kunming Medical University, Kunming City, Yunnan Province, China
| | - Zhihong Luo
- Department of Orthopaedics, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China
| | - Zhigui Li
- Department of Orthopaedics, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China
| | - Hua Zhang
- Department of Orthopaedics, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China
| | - Tianhua Zhou
- Department of Orthopaedics, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China.
| | - Hongbo Tan
- Department of Orthopaedics, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China.
| |
Collapse
|
12
|
Chariyev-Prinz F, Szojka A, Neto N, Burdis R, Monaghan MG, Kelly DJ. An assessment of the response of human MSCs to hydrostatic pressure in environments supportive of differential chondrogenesis. J Biomech 2023; 154:111590. [PMID: 37163962 DOI: 10.1016/j.jbiomech.2023.111590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/31/2023] [Accepted: 04/11/2023] [Indexed: 05/12/2023]
Abstract
Mechanical stimulation can modulate the chondrogenic differentiation of stem/progenitor cells and potentially benefit tissue engineering (TE) of functional articular cartilage (AC). Mechanical cues like hydrostatic pressure (HP) are often applied to cell-laden scaffolds, with little optimization of other key parameters (e.g. cell density, biomaterial properties) known to effect lineage commitment. In this study, we first sought to establish cell seeding densities and fibrin concentrations supportive of robust chondrogenesis of human mesenchymal stem cells (hMSCs). High cell densities (15*106 cells/ml) were more supportive of sGAG deposition on a per cell basis, while collagen deposition was higher at lower seeding densities (5*106 cells/ml). Employment of lower fibrin (2.5 %) concentration hydrogels supported more robust chondrogenesis of hMSCs, with higher collagen type II and lower collagen type X deposition compared to 5 % hydrogels. The application of HP to hMSCs maintained in identified chondro-inductive culture conditions had little effect on overall levels of cartilage-specific matrix production. However, if hMSCs were first temporally primed with TGF-β3 before its withdrawal, they responded to HP by increased sGAG production. The response to HP in higher cell density cultures was also associated with a metabolic shift towards glycolysis, which has been linked with a mature chondrocyte-like phenotype. These results suggest that mechanical stimulation may not be necessary to engineer functional AC grafts using hMSCs if other culture conditions have been optimised. However, such bioreactor systems can potentially be employed to better understand how engineered tissues respond to mechanical loading in vivo once removed from in vitro culture environments.
Collapse
Affiliation(s)
- Farhad Chariyev-Prinz
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Alex Szojka
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Nuno Neto
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Ross Burdis
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Michael G Monaghan
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
13
|
Chen Z, Song X, Mu X, Zhang J, Cheang UK. 2D Magnetic Microswimmers for Targeted Cell Transport and 3D Cell Culture Structure Construction. ACS APPLIED MATERIALS & INTERFACES 2023; 15:8840-8853. [PMID: 36752406 DOI: 10.1021/acsami.2c18955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Cell delivery using magnetic microswimmers is a promising tool for targeted therapy. However, it remains challenging to rapidly and uniformly manufacture cell-loaded microswimmers that can be assembled into cell-supporting structures at diseased sites. Here, rapid and uniform manufacturable 2D magnetic achiral microswimmers with pores were fabricated to deliver bone marrow mesenchymal stem cells (BMSCs) to regenerate articular-damaged cartilage. Under actuation with magnetic fields, the BMSC-loaded microswimmers take advantage of the achiral structure to exhibit rolling or swimming motions to travel on smooth and rough surfaces, up inclined planes, or in the bulk fluid. Cell viability, proliferation, and differentiation tests performed days after cell seeding verified the microswimmers' biocompatibility. Long-distance targeting and in situ assemblies into 3D cell-supporting structures with BMSC-loaded microswimmers were demonstrated using a knee model and U-shaped wells. Overall, combining the advantages of preparing an achiral 2D structured microswimmer with magnetically driven motility results in a platform for cell transport and constructing 3D cell cultures that can improve cell delivery at lesion sites for biomedical applications.
Collapse
Affiliation(s)
- Zhi Chen
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaoxia Song
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xueliang Mu
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Junkai Zhang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - U Kei Cheang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen 518055, China
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Southern University of Science and Technology, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Human-Augmentation and Rehabilitation Robotics in Universities, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
14
|
Bone Marrow-Derived Mesenchymal Stem Cell Implants for the Treatment of Focal Chondral Defects of the Knee in Animal Models: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:ijms24043227. [PMID: 36834639 PMCID: PMC9958893 DOI: 10.3390/ijms24043227] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Osteoarthritis remains an unfortunate long-term consequence of focal cartilage defects of the knee. Associated with functional loss and pain, it has necessitated the exploration of new therapies to regenerate cartilage before significant deterioration and subsequent joint replacement take place. Recent studies have investigated a multitude of mesenchymal stem cell (MSC) sources and polymer scaffold compositions. It is uncertain how different combinations affect the extent of integration of native and implant cartilage and the quality of new cartilage formed. Implants seeded with bone marrow-derived MSCs (BMSCs) have demonstrated promising results in restoring these defects, largely through in vitro and animal studies. A PRISMA systematic review and meta-analysis was conducted using five databases (PubMed, MEDLINE, EMBASE, Web of Science, and CINAHL) to identify studies using BMSC-seeded implants in animal models of focal cartilage defects of the knee. Quantitative results from the histological assessment of integration quality were extracted. Repair cartilage morphology and staining characteristics were also recorded. Meta-analysis demonstrated that high-quality integration was achieved, exceeding that of cell-free comparators and control groups. This was associated with repair tissue morphology and staining properties which resembled those of native cartilage. Subgroup analysis showed better integration outcomes for studies using poly-glycolic acid-based scaffolds. In conclusion, BMSC-seeded implants represent promising strategies for the advancement of focal cartilage defect repair. While a greater number of studies treating human patients is necessary to realize the full clinical potential of BMSC therapy, high-quality integration scores suggest that these implants could generate repair cartilage of substantial longevity.
Collapse
|
15
|
Rigogliuso S, Campora S, Notarbartolo M, Ghersi G. Recovery of Bioactive Compounds from Marine Organisms: Focus on the Future Perspectives for Pharmacological, Biomedical and Regenerative Medicine Applications of Marine Collagen. Molecules 2023; 28:molecules28031152. [PMID: 36770818 PMCID: PMC9920902 DOI: 10.3390/molecules28031152] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/05/2023] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
Marine environments cover more than 70% of the Earth's surface and are among the richest and most complex ecosystems. In terms of biodiversity, the ocean represents an important source, still not widely exploited, of bioactive products derived from species of bacteria, plants, and animals. However, global warming, in combination with multiple anthropogenic practices, represents a serious environmental problem that has led to an increase in gelatinous zooplankton, a phenomenon referred to as jellyfish bloom. In recent years, the idea of "sustainable development" has emerged as one of the essential elements of green-economy initiatives; therefore, the marine environment has been re-evaluated and considered an important biological resource. Several bioactive compounds of marine origin are being studied, and among these, marine collagen represents one of the most attractive bio-resources, given its use in various disciplines, such as clinical applications, cosmetics, the food sector, and many other industrial applications. This review aims to provide a current overview of marine collagen applications in the pharmacological and biomedical fields, regenerative medicine, and cell therapy.
Collapse
Affiliation(s)
- Salvatrice Rigogliuso
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy
| | - Simona Campora
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy
- Correspondence: (S.C.); (M.N.); Tel.: +39-091-238-62813 (S.C.); +39-091-238-97426 (M.N.)
| | - Monica Notarbartolo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy
- Correspondence: (S.C.); (M.N.); Tel.: +39-091-238-62813 (S.C.); +39-091-238-97426 (M.N.)
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy
- Abiel s.r.l., c/o Department STEBICEF, University of Palermo, Viale delle Scienze, Ed. 16, 90128 Palermo, Italy
| |
Collapse
|
16
|
Exosomes treating osteoarthritis: hope with challenge. Heliyon 2023; 9:e13152. [PMID: 36711315 PMCID: PMC9880404 DOI: 10.1016/j.heliyon.2023.e13152] [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: 10/22/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/23/2023] Open
Abstract
Osteoarthritis (OA) has been proven as the second primary cause of pain and disability in the elderly population, impact patients both physically and mentally, as well as imposing a heavy burden on the global healthcare system. Current treatment methods, whether conservative or surgical, that aim at relieving symptoms can not delay or reverse the degenerative process in the structure. Scientists and clinicians are facing a revolution in OA treatment strategies. The emergence of exosomes brings hope for OA treatment based on pathology, which is attributed to its full potential in protecting chondrocytes from excessive death, alleviating inflammation, maintaining cartilage matrix metabolism, and regulating angiogenesis and subchondral bone remodeling. Therefore, we summarized the recent studies of exosomes in OA, aiming to comprehensively understand the functions and mechanisms of exosomes in OA treatment, which may provide direction and theoretical support for formulating therapeutic strategies in the future.
Collapse
|
17
|
Li DX, Ma Z, Szojka ARA, Lan X, Kunze M, Mulet-Sierra A, Westover L, Adesida AB. Non-hypertrophic chondrogenesis of mesenchymal stem cells through mechano-hypoxia programing. J Tissue Eng 2023; 14:20417314231172574. [PMID: 37216035 PMCID: PMC10192798 DOI: 10.1177/20417314231172574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/09/2023] [Indexed: 05/24/2023] Open
Abstract
Cartilage tissue engineering aims to generate functional replacements to treat cartilage defects from damage and osteoarthritis. Human bone marrow-derived mesenchymal stem cells (hBM-MSC) are a promising cell source for making cartilage, but current differentiation protocols require the supplementation of growth factors like TGF-β1 or -β3. This can lead to undesirable hypertrophic differentiation of hBM-MSC that progress to bone. We have found previously that exposing engineered human meniscus tissues to physiologically relevant conditions of the knee (mechanical loading and hypoxia; hence, mechano-hypoxia conditioning) increased the gene expression of hyaline cartilage markers, SOX9 and COL2A1, inhibited hypertrophic marker COL10A1, and promoted bulk mechanical property development. Adding further to this protocol, we hypothesize that combined mechano-hypoxia conditioning with TGF-β3 growth factor withdrawal will promote stable, non-hypertrophic chondrogenesis of hBM-MSC embedded in an HA-hydrogel. We found that the combined treatment upregulated many cartilage matrix- and development-related markers while suppressing many hypertrophic- and bone development-related markers. Tissue level assessments with biochemical assays, immunofluorescence, and histochemical staining confirmed the gene expression data. Further, mechanical property development in the dynamic compression treatment shows promise toward generating functional engineered cartilage through more optimized and longer culture conditions. In summary, this study introduced a novel protocol to differentiate hBM-MSC into stable, cartilage-forming cells.
Collapse
Affiliation(s)
- David Xinzheyang Li
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Civil and Environmental
Engineering, Faculty of Engineering, AB, University of Alberta, Edmonton, AB,
Canada
| | - Zhiyao Ma
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Alexander RA Szojka
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Xiaoyi Lan
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Civil and Environmental
Engineering, Faculty of Engineering, AB, University of Alberta, Edmonton, AB,
Canada
| | - Melanie Kunze
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Lindsey Westover
- Department of Mechanical Engineering,
Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Adetola B Adesida
- Department of Surgery, Faculty of
Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
18
|
Islam MS, Ebrahimi-Barough S, Al Mahtab M, Shirian S, Aghayan HR, Arjmand B, Allahverdi A, Ranjbar FE, Sadeg AB, Ai J. Encapsulation of rat bone marrow-derived mesenchymal stem cells (rBMMSCs) in collagen type I containing platelet-rich plasma for osteoarthritis treatment in rat model. Prog Biomater 2022; 11:385-396. [PMID: 36271317 DOI: 10.1007/s40204-022-00200-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/02/2022] [Indexed: 11/28/2022] Open
Abstract
Osteoarthritis (OA) is the most common form of degenerative joint disease, affecting more than 25% of the adults despite its prevalence in the elderly population. Most of the current therapeutic modalities aim at symptomatic treatment which lingers the disease progression. In recent years, regenerative medicine such as stem cell transplantation and tissue engineering has been suggested as a potential curative intervention for OA. The objective of this current study was to assess the safety and efficacy of an injectable tissue-engineered construct composed of rat bone marrow mesenchymal stem cells (rBMMSCs), platelet-rich plasma (PRP), and collagen type I in rat model of OA. To produce collagen type I, PRP and rBMMSCs, male Wistar rats were ethically euthanized. After isolation, culture, expansion and characterization of rBMMSCs, tissue-engineered construct was formed by a combination of appropriate amount of collagen type I, PRP and rBMMSCs. In vitro studies were conducted to evaluate the effect of PRP on chondrogenic differentiation capacity of encapsulated cells. In the following, the tissue-engineered construct was injected in knee joints of rat models of OA (24 rats in 4 groups: OA, OA + MSC, OA + collagen + MSC + PRP, OA + MSC + collagen). After 6 weeks, the animals were euthanized and knee joint histopathology examinations of knee joint samples were performed to evaluate the effect of each treatment on OA. Tissue-engineered construct was successfully manufactured and in vitro assays demonstrated the relevant chondrogenic genes and proteins expression were higher in PRP group than that of others. Histopathological findings of the knee joint samples showed favorable regenerative effect of rBMMSCs + PRP + collagen group compared to others. We introduced an injectable tissue-engineered product composed of rBMMSCs + PRP + collagen with potential regenerative effect on cartilage that has been damaged by OA.
Collapse
Affiliation(s)
- Md Shahidul Islam
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mamun Al Mahtab
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Dhaka, Bangladesh
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.,Shefa Neuroscience Research Center, Khatam-Alanbia Hospital, Tehran, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Allahverdi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Esmaeili Ranjbar
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Amin Bigham Sadeg
- Department of Clinical Sciences, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.,Shiraz Molecular Pathology Research Center, Dr Daneshbod Path Lab, Shiraz, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Molnar V, Pavelić E, Vrdoljak K, Čemerin M, Klarić E, Matišić V, Bjelica R, Brlek P, Kovačić I, Tremolada C, Primorac D. Mesenchymal Stem Cell Mechanisms of Action and Clinical Effects in Osteoarthritis: A Narrative Review. Genes (Basel) 2022; 13:genes13060949. [PMID: 35741711 PMCID: PMC9222975 DOI: 10.3390/genes13060949] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
With the insufficient satisfaction rates and high cost of operative treatment for osteoarthritis (OA), alternatives have been sought. Furthermore, the inability of current medications to arrest disease progression has led to rapidly growing clinical research relating to mesenchymal stem cells (MSCs). The availability and function of MSCs vary according to tissue source. The three primary sources include the placenta, bone marrow, and adipose tissue, all of which offer excellent safety profiles. The primary mechanisms of action are trophic and immunomodulatory effects, which prevent the further degradation of joints. However, the function and degree to which benefits are observed vary significantly based on the exosomes secreted by MSCs. Paracrine and autocrine mechanisms prevent cell apoptosis and tissue fibrosis, initiate angiogenesis, and stimulate mitosis via growth factors. MSCs have even been shown to exhibit antimicrobial effects. Clinical results incorporating clinical scores and objective radiological imaging have been promising, but a lack of standardization in isolating MSCs prevents their incorporation in current guidelines.
Collapse
Affiliation(s)
- Vilim Molnar
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Eduard Pavelić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
| | - Kristijan Vrdoljak
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (K.V.); (M.Č.)
| | - Martin Čemerin
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (K.V.); (M.Č.)
| | - Emil Klarić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
| | - Vid Matišić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
| | - Roko Bjelica
- Department of Oral Surgery, School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Petar Brlek
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
| | | | | | - Dragan Primorac
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (V.M.); (E.P.); (E.K.); (V.M.); (P.B.)
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Split, 21000 Split, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Medical School, University of Rijeka, 51000 Rijeka, Croatia
- Medical School REGIOMED, 96450 Coburg, Germany
- Eberly College of Science, The Pennsylvania State University, University Park, PA 16802, USA
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, West Haven, CT 06516, USA
- Correspondence:
| |
Collapse
|
20
|
Andersen C, Uvebrant K, Mori Y, Aarsvold S, Jacobsen S, Berg LC, Lundgren-Åkerlund E, Lindegaard C. Human integrin α10β1-selected mesenchymal stem cells home to cartilage defects in the rabbit knee and assume a chondrocyte-like phenotype. Stem Cell Res Ther 2022; 13:206. [PMID: 35578319 PMCID: PMC9109317 DOI: 10.1186/s13287-022-02884-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have shown promising results in stimulating cartilage repair and in the treatment of osteoarthritis (OA). However, the fate of the MSCs after intra-articular injection and their role in cartilage regeneration is not clear. To address these questions, this study investigated (1) homing of labeled human adipose tissue derived integrin α10β1-selected MSCs (integrin α10-MSCs) to a cartilage defect in a rabbit model and (2) the ability of the integrin α10-MSCs to differentiate to chondrocytes and to produce cartilage matrix molecules in vivo. DESIGN Integrin α10-MSCs were labeled with superparamagnetic iron oxide nanoparticles (SPIONs) co-conjugated with Rhodamine B to allow visualization by both MRI and fluorescence microscopy. A cartilage defect was created in the articular cartilage of the intertrochlear groove of the femur of rabbits. Seven days post-surgery, labeled integrin α10-MSCs or vehicle were injected into the joint. Migration and distribution of the SPION-labeled integrin α10-MSCs was evaluated by high-field 9.4 T MRI up to 10 days after injection. Tissue sections from the repair tissue in the defects were examined by fluorescence microscopy. RESULTS In vitro characterization of the labeled integrin α10-MSCs demonstrated maintained viability, proliferation rate and trilineage differentiation capacity compared to unlabeled MSCs. In vivo MRI analysis detected the labeled integrin α10-MSCs in the cartilage defects at all time points from 12 h after injection until day 10 with a peak concentration between day 1 and 4 after injection. The labeled MSCs were also detected lining the synovial membrane at the early time points. Fluorescence analysis confirmed the presence of the labeled integrin α10-MSCs in all layers of the cartilage repair tissue and showed co-localization between the labeled cells and the specific cartilage molecules aggrecan and collagen type II indicating in vivo differentiation of the MSCs to chondrocyte-like cells. No adverse effects of the α10-MSC treatment were detected during the study period. CONCLUSION Our results demonstrated migration and homing of human integrin α10β1-selected MSCs to cartilage defects in the rabbit knee after intra-articular administration as well as chondrogenic differentiation of the MSCs in the regenerated cartilage tissue.
Collapse
Affiliation(s)
- Camilla Andersen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark.
| | | | - Yuki Mori
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | | | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark
| | - Lise Charlotte Berg
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark
| | | | - Casper Lindegaard
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Højbakkegaard Allé 5, 2630, Taastrup, Denmark
| |
Collapse
|
21
|
Wang Z, Le H, Wang Y, Liu H, Li Z, Yang X, Wang C, Ding J, Chen X. Instructive cartilage regeneration modalities with advanced therapeutic implantations under abnormal conditions. Bioact Mater 2022; 11:317-338. [PMID: 34977434 PMCID: PMC8671106 DOI: 10.1016/j.bioactmat.2021.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/19/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
The development of interdisciplinary biomedical engineering brings significant breakthroughs to the field of cartilage regeneration. However, cartilage defects are considerably more complicated in clinical conditions, especially when injuries occur at specific sites (e.g., osteochondral tissue, growth plate, and weight-bearing area) or under inflammatory microenvironments (e.g., osteoarthritis and rheumatoid arthritis). Therapeutic implantations, including advanced scaffolds, developed growth factors, and various cells alone or in combination currently used to treat cartilage lesions, address cartilage regeneration under abnormal conditions. This review summarizes the strategies for cartilage regeneration at particular sites and pathological microenvironment regulation and discusses the challenges and opportunities for clinical transformation.
Collapse
Affiliation(s)
- Zhonghan Wang
- Department of Plastic and Reconstruct Surgery, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, PR China
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - Hanxiang Le
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - Yanbing Wang
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - He Liu
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - Zuhao Li
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - Xiaoyu Yang
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - Chenyu Wang
- Department of Plastic and Reconstruct Surgery, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, PR China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| |
Collapse
|
22
|
Xiang XN, Zhu SY, He HC, Yu X, Xu Y, He CQ. Mesenchymal stromal cell-based therapy for cartilage regeneration in knee osteoarthritis. Stem Cell Res Ther 2022; 13:14. [PMID: 35012666 PMCID: PMC8751117 DOI: 10.1186/s13287-021-02689-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/07/2021] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis, as a degenerative disease, is a common problem and results in high socioeconomic costs and rates of disability. The most commonly affected joint is the knee and characterized by progressive destruction of articular cartilage, loss of extracellular matrix, and progressive inflammation. Mesenchymal stromal cell (MSC)-based therapy has been explored as a new regenerative treatment for knee osteoarthritis in recent years. However, the detailed functions of MSC-based therapy and related mechanism, especially of cartilage regeneration, have not been explained. Hence, this review summarized how to choose, authenticate, and culture different origins of MSCs and derived exosomes. Moreover, clinical application and the latest mechanistical findings of MSC-based therapy in cartilage regeneration were also demonstrated.
Collapse
Affiliation(s)
- Xiao-Na Xiang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Si-Yi Zhu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Hong-Chen He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xi Yu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yang Xu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Cheng-Qi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China. .,School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China. .,Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China. .,Rehabilitation Medicine Centre, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
23
|
Phelps J, Leonard C, Shah S, Krawetz R, Hart DA, Duncan NA, Sen A. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:73-87. [PMID: 35641171 PMCID: PMC8895489 DOI: 10.1093/stcltm/szab008] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal progenitor cells (MPCs) have shown promise initiating articular cartilage repair, with benefits largely attributed to the trophic factors they secrete. These factors can be found in the conditioned medium (CM) collected from cell cultures, and it is believed that extracellular vesicles (EVs) within this CM are at least partially responsible for MPC therapeutic efficacy. This study aimed to examine the functionality of the EV fraction of CM compared to whole CM obtained from human adipose-derived MPCs in an in vivo murine cartilage defect model. Mice treated with whole CM or the EV fraction demonstrated an enhanced cartilage repair score and type II collagen deposition at the injury site compared to saline controls. We then developed a scalable bioprocess using stirred suspension bioreactors (SSBs) to generate clinically relevant quantities of MPC-EVs. Whereas static monolayer culture systems are simple to use and readily accessible, SSBs offer increased scalability and a more homogenous environment due to constant mixing. This study evaluated the biochemical and functional properties of MPCs and their EV fractions generated in static culture versus SSBs. Functionality was assessed using in vitro MPC chondrogenesis as an outcome measure. SSBs supported increased MPC expression of cartilage-specific genes, and EV fractions derived from both static and SSB culture systems upregulated type II collagen production by MPCs. These results suggest that SSBs are an effective platform for the generation of MPC-derived EVs with the potential to induce cartilage repair.
Collapse
Affiliation(s)
- Jolene Phelps
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Catherine Leonard
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Sophia Shah
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
| | - Roman Krawetz
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - David A Hart
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Neil A Duncan
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Musculoskeletal Mechanobiology and Multiscale Mechanics Bioengineering Lab, Department of Civil Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Arindom Sen
- Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Center for Bioengineering Research and Education, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Corresponding author: Arindom Sen, Pharmaceutical Production Research Facility, Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada. Tel: +403-210-9452; Fax: +403-220-8962;
| |
Collapse
|
24
|
Onoi Y, Hiranaka T, Hida Y, Fujishiro T, Okamoto K, Matsumoto T, Kuroda R. Second-look Arthroscopic Findings and Clinical Outcomes after Adipose-derived Regenerative Cell Injection in Knee Osteoarthritis. Clin Orthop Surg 2022; 14:377-385. [PMID: 36061847 PMCID: PMC9393284 DOI: 10.4055/cios20312] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 06/18/2021] [Accepted: 07/11/2021] [Indexed: 12/03/2022] Open
Abstract
Background To evaluate the clinical outcomes and second-look arthroscopic findings after intra-articular adipose-derived regenerative cell (ADRC) injection as treatment for knee osteoarthritis (OA). Methods ADRCs were administered to 11 patients (19 knees; mean age, 61.7 years) with knee OA. Subcutaneous adipose tissue was harvested by liposuction from both thighs, and arthroscopic lavage was performed, followed by ADRC injection (mean dose, 1.40 × 107 cells) into the synovial fluid. Outcome measures included the Knee Injury and Osteoarthritis Outcome Score, Lysholm score, and visual analog scale score. Arthroscopic examinations were performed to assess the International Cartilage Repair Society cartilage injury grade preoperatively and overall repair postoperatively. Noninvasive assessments were performed at baseline and at 1-, 3-, and 6-month follow-ups; arthroscopic assessments were performed at baseline and at 6 months. Results All outcome measures significantly improved after treatment. This improvement was evident 1 month after treatment and was sustained until the 6-month follow-up. Data from second-look arthroscopy showed better repair in low-grade cartilage lesions than in lesions with a greater degree of damage. No patients demonstrated worsening of Kellgren-Lawrence grade, and none underwent total knee arthroplasty during this period. Conclusions Clinical outcomes were improved in patients with knee OA after ADRC administration. Cartilage regeneration was more effective in smaller damaged lesions than in bigger lesions.
Collapse
Affiliation(s)
- Yuma Onoi
- Department of Orthopedic Surgery and Joint Surgery Center, Takatsuki General Hospital, Osaka, Japan
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takafumi Hiranaka
- Department of Orthopedic Surgery and Joint Surgery Center, Takatsuki General Hospital, Osaka, Japan
| | - Yuichi Hida
- Department of Orthopedic Surgery and Joint Surgery Center, Takatsuki General Hospital, Osaka, Japan
| | - Takaaki Fujishiro
- Department of Orthopedic Surgery and Joint Surgery Center, Takatsuki General Hospital, Osaka, Japan
| | - Koji Okamoto
- Department of Orthopedic Surgery and Joint Surgery Center, Takatsuki General Hospital, Osaka, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryosuke Kuroda
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
25
|
Cooper SM, Rainbow RS. The Developing Field of Scaffold-Free Tissue Engineering for Articular Cartilage Repair. TISSUE ENGINEERING. PART B, REVIEWS 2021; 28:995-1006. [PMID: 34605669 DOI: 10.1089/ten.teb.2021.0130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Articular cartilage is critical for proper joint mobility as it provides a smooth and lubricated surface between articulating bones and allows for transmission of load to underlying bones. Extended wear or injury of this tissue can result in osteoarthritis, a degenerative disease affecting millions across the globe. Because of its low regenerative capacity, articular cartilage cannot heal on its own and effective treatments for injured joint restoration remain a challenge. Strategies in tissue engineering have been demonstrated as potential therapeutic approaches to regenerate and repair damaged articular cartilage. Although many of these strategies rely on the use of an exogenous three-dimensional scaffolds to regenerate cartilage, scaffold-free tissue engineering provides numerous advantages over scaffold-based methods. This review highlights the latest advancements in scaffold-free tissue engineering for cartilage and the potential for clinical translation.
Collapse
Affiliation(s)
- Sarah M Cooper
- Department of Mechanical and Materials Engineering, Queen's University, Kingston, Canada
| | - Roshni S Rainbow
- Department of Mechanical and Materials Engineering, Queen's University, Kingston, Canada
| |
Collapse
|
26
|
Adipose Tissue-Derived Mesenchymal Stem Cells as a Potential Restorative Treatment for Cartilage Defects: A PRISMA Review and Meta-Analysis. Pharmaceuticals (Basel) 2021; 14:ph14121280. [PMID: 34959680 PMCID: PMC8705514 DOI: 10.3390/ph14121280] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/13/2022] Open
Abstract
Cartilage defects are a predisposing factor for osteoarthritis. Conventional therapies are mostly palliative and there is an interest in developing newer therapies that target the disease’s progression. Mesenchymal stem cells (MSCs) have been suggested as a promising therapy to restore hyaline cartilage to cartilage defects, though the optimal cell source has remained under investigation. A PRISMA systematic review was conducted utilising five databases (MEDLINE, EMBASE, Cochrane Library, Scopus, Web of Science) which identified nineteen human studies that used adipose tissue-derived MSC (AMSC)-based therapies, including culture-expanded AMSCs and stromal vascular fraction, to treat cartilage defects. Clinical, imaging and histological outcomes, as well as other relevant details pertaining to cartilage regeneration, were extracted from each study. Pooled analysis revealed a significant improvement in WOMAC scores (mean difference: −25.52; 95%CI (−30.93, −20.10); p < 0.001), VAS scores (mean difference: −3.30; 95%CI (−3.72, −2.89); p < 0.001), KOOS scores and end point MOCART score (mean: 68.12; 95%CI (62.18, 74.05)), thus showing improvement. The studies in this review demonstrate the safety and efficacy of AMSC-based therapies for cartilage defects. Establishing standardised methods for MSC extraction and delivery, and performing studies with long follow-up should enable future high-quality research to provide the evidence needed to bring AMSC-based therapies into the market.
Collapse
|
27
|
Rajagopal K, Arjunan P, Marepally S, Madhuri V. Controlled Differentiation of Mesenchymal Stem Cells into Hyaline Cartilage in miR-140-Activated Collagen Hydrogel. Cartilage 2021; 13:571S-581S. [PMID: 34581616 PMCID: PMC8804822 DOI: 10.1177/19476035211047627] [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: 11/16/2022] Open
Abstract
OBJECTIVE Hypertrophic cartilage formation is a major setback in mesenchymal stem cells (MSCs)-mediated cartilage repair, and overcoming it requires optimization of differentiation. Here, we tested the miR-140 activated collagen hydrogel for the chondrogenic differentiation of MSCs and to produce hyaline cartilage. METHODS Bone marrow MSCs isolated from 3 patients were pretreated with miR-140 and then chondrogenic differentiated. The 3-dimensional (3D) transfection potential of 5 different transfection reagents (Polyethylenimine, Lipofectamine, TransIT-X2, Amide:Cholesterol-based liposomes [AmC] and AmC pegylated with Tocofersolan [AmCTOC]) was compared and the reagent that showed higher green fluorescent protein (GFP) expression was selected. Finally, the collagen hydrogel was activated using miR-140-transfection complex and sustained delivered to MSCs during chondrogenic differentiation. After differentiation, the outcome was assessed by reverse transcription-polymerase chain reaction (RT-PCR), histology, immunohistochemistry, and compared with scrambled miRNA treated control. RESULTS Pretreatment of MSCs with miR-140 significantly increased the expression of cartilage-specific genes (COL2A1, SOX9, and ACAN) with reduced hypertrophic chondrocyte (COL10A1) marker expression and better safranin-O staining than the control. The AmCTOC liposome showed a significant increase in 3D transfection of GFP expressing plasmid than the others. Furthermore, the knockdown of GAPDH using siRNA in HEK cells and expression of GFP mRNA in human bone marrow MSCs confirmed the 3D-transfection efficiency of AmCTOC. The sustained delivery of miR-140 using activated matrix formed a hyaline cartilage-like tissue with minimal COL10A1 expression in RT-PCR and immunohistochemistry. CONCLUSION Our results demonstrated the therapeutic potential of miR-140-activated hydrogel for MSCs-based cartilage tissue engineering, which could also be used for endogenous stem cells-mediated cartilage repair.
Collapse
Affiliation(s)
- Karthikeyan Rajagopal
- Department of Paediatric
Orthopaedics, Centre for Stem Cell Research, Christian Medical College,
Vellore, Tamil Nadu, India,Department of Paed ortho and
Centre for stem cell research are two different departments in Christian
medical college, Vellore
| | - Porkizhi Arjunan
- Laboratory of Nanobioscience and
Nanobiotechnology, Centre for Stem Cell Research, Christian Medical College,
Vellore, Tamil Nadu, India
| | - Srujan Marepally
- Laboratory of Nanobioscience and
Nanobiotechnology, Centre for Stem Cell Research, Christian Medical College,
Vellore, Tamil Nadu, India
| | - Vrisha Madhuri
- Department of Paediatric
Orthopaedics, Centre for Stem Cell Research, Christian Medical College,
Vellore, Tamil Nadu, India,Department of Paed ortho and
Centre for stem cell research are two different departments in Christian
medical college, Vellore,Vrisha Madhuri, Department of
Paediatric Orthopaedics, Christian Medical College, First floor, Paul
Brand Building, Vellore, Tamil Nadu 632004, India.
| |
Collapse
|
28
|
Puzzitiello RN, Dubin J, Menendez ME, Moverman MA, Pagani NR, Drager J, Salzler MJ. Public Opinion and Expectations of Stem Cell Therapies in Orthopaedics. Arthroscopy 2021; 37:3510-3517.e2. [PMID: 34126222 DOI: 10.1016/j.arthro.2021.05.058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/03/2021] [Accepted: 05/28/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE To explore public opinion, understanding, and preferences regarding the use of stem cell therapies for the treatment of joint and tendon pathologies using online crowdsourcing. METHODS A 30-question survey was completed by 931 members of the public using Amazon Mechanical Turk, a validated crowdsourcing method. Outcomes included perceptions and preferences regarding the use of stem cells therapies for the nonsurgical treatment of orthopaedic conditions. Sociodemographic factors and a validated assessment of health literacy were collected. Inclusion criteria were adult participants 18 years or older, residence within the United States, and a valid Social Security number. Multivariable logistic regression modeling was used to determine population characteristics associated with the belief that stem cells represent the most effective treatment for long-standing joint or tendon disorders. RESULTS Most respondents reported that stem cell therapies have convincing evidence to support their use for orthopaedic conditions (84.5%) and are approved and regulated by the Food and Drug Administration (65%). About three-quarters of respondents reported that stem cells can stop the progression of and alleviate pain from arthritis or damaged tendons, and over half (53.5%) reported that stem cells can cure arthritis. Factors with the greatest influence on respondents' decision to receive stem cell therapies are research supporting their safety and effectiveness and doctor recommendation. However, 63.3% of respondents stated that they would consider stem cells if their doctor recommended it, regardless of evidence supporting their effectiveness, and over half would seek another doctor if their orthopaedic surgeon did not offer this treatment option. CONCLUSIONS The public's limited understanding regarding the current evidence associated with stem cell therapies for osteoarthritis and tendinous pathologies may contribute to unrealistic expectations and misinformed decisions. This study highlights the importance of patient education and expectation setting, as well as evidence transparency, as stem cell therapies become increasingly accessible. LEVEL OF EVIDENCE Level IV, case series.
Collapse
Affiliation(s)
- Richard N Puzzitiello
- Department of Orthopaedic Surgery, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, U.S.A.; New England Baptist Hospital, Boston, Massachusetts, U.S.A
| | | | - Mariano E Menendez
- Department of Orthopaedic Surgery, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, U.S.A.; New England Baptist Hospital, Boston, Massachusetts, U.S.A
| | - Michael A Moverman
- Department of Orthopaedic Surgery, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, U.S.A.; New England Baptist Hospital, Boston, Massachusetts, U.S.A
| | - Nicholas R Pagani
- Department of Orthopaedic Surgery, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, U.S.A.; New England Baptist Hospital, Boston, Massachusetts, U.S.A
| | - Justin Drager
- Department of Orthopaedic Surgery, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, U.S.A
| | - Matthew J Salzler
- Department of Orthopaedic Surgery, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, U.S.A..
| |
Collapse
|
29
|
BMSC-Derived Exosomes Ameliorate Osteoarthritis by Inhibiting Pyroptosis of Cartilage via Delivering miR-326 Targeting HDAC3 and STAT1//NF- κB p65 to Chondrocytes. Mediators Inflamm 2021; 2021:9972805. [PMID: 34764819 PMCID: PMC8577926 DOI: 10.1155/2021/9972805] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 09/04/2021] [Accepted: 09/25/2021] [Indexed: 02/07/2023] Open
Abstract
Background In the past decade, mesenchymal stem cells (MSCs) have been widely used for the treatment of osteoarthritis (OA), and noncoding RNAs in exosomes may play a major role. Aim The present study is aimed at exploring the effect and mechanism of miR-326 in exosomes secreted by bone marrow mesenchymal stem cells (BMSCs) on pyroptosis of cartilage and OA improvement. Methods Exosomes from BMSCs (BMSC-Exos) were isolated and identified to incubate with OA chondrocytes. Proliferation, migration, specific gene and miR-326 expression, and pyroptosis of chondrocytes were detected. BMSCs or chondrocytes were transfected with miR-326 mimics or inhibitors to investigate the effect of miR-326 in BMSC-Exos on pyroptosis of chondrocytes and the potential mechanism. Finally, a rat OA model was established to verify the effect and mechanism of miR-326 in BMSC-Exos on cartilage of pyroptosis. Results Incubation with BMSC-Exos could significantly improve the survival rate, migration ability, and chondrocyte-specific genes (COL2A1, SOX9, Agg, and Prg4) and miR-326 expression of OA chondrocytes and significantly inhibit pyroptosis of chondrocytes by downregulation of the levels of inflammatory cytokines, Caspase-1 activity, and pyroptosis-related proteins such as GSDMD, NLRP3, ASC, IL-1β, and IL-18 (P < 0.01). PKH26 labeling confirmed the uptake of BMSC-Exos by chondrocytes. Incubation with exosomes extracted from BMSCs overexpressing miR-326 can significantly repress the pyroptosis of chondrocytes, while knockdown of miR-326 had the opposite effect (P < 0.01). The same result was also demonstrated by direct interference with the expression level of miR-326 in chondrocytes (P < 0.01). In addition, we found that the overexpression of miR-326 significantly inhibited the expression of HDAC3 and NF-κB p65 and significantly promoted the expression of STAT1, acetylated STAT1, and acetylated NF-κB p65 in chondrocytes (P < 0.01). The targeted relationship between miR-326 and HDAC3 was verified by dual-luciferase reporter assay. Animal experiments confirmed the mechanism by which miR-326 delivered by BMSC-Exos inhibits pyroptosis of cartilage by targeting HDAC3 and STAT1/NF-κB p65 signaling pathway. Conclusion BMSC-Exos can deliver miR-326 to chondrocytes and cartilage and improve OA by targeting HDAC3 and STAT1//NF-κB p65 to inhibit pyroptosis of chondrocytes and cartilage. Our findings provide a new mechanism for BMSC-Exos to treat OA.
Collapse
|
30
|
Levato R, Lim KS, Li W, Asua AU, Peña LB, Wang M, Falandt M, Bernal PN, Gawlitta D, Zhang YS, Woodfield TBF, Malda J. High-resolution lithographic biofabrication of hydrogels with complex microchannels from low-temperature-soluble gelatin bioresins. Mater Today Bio 2021; 12:100162. [PMID: 34870141 PMCID: PMC8626672 DOI: 10.1016/j.mtbio.2021.100162] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/20/2022] Open
Abstract
Biofabrication via light-based 3D printing offers superior resolution and ability to generate free-form architectures, compared to conventional extrusion technologies. While extensive efforts in the design of new hydrogel bioinks lead to major advances in extrusion methods, the accessibility of lithographic bioprinting is still hampered by a limited choice of cell-friendly resins. Herein, we report the development of a novel set of photoresponsive bioresins derived from ichthyic-origin gelatin, designed to print high-resolution hydrogel constructs with embedded convoluted networks of vessel-mimetic channels. Unlike mammalian gelatins, these materials display thermal stability as pre-hydrogel solutions at room temperature, ideal for bioprinting on any easily-accessible lithographic printer. Norbornene- and methacryloyl-modification of the gelatin backbone, combined with a ruthenium-based visible light photoinitiator and new coccine as a cytocompatible photoabsorber, allowed to print structures resolving single-pixel features (∼50 μm) with high shape fidelity, even when using low stiffness gels, ideal for cell encapsulation (1-2 kPa). Moreover, aqueous two-phase emulsion bioresins allowed to modulate the permeability of the printed hydrogel bulk. Bioprinted mesenchymal stromal cells displayed high functionality over a month of culture, and underwent multi-lineage differentiation while colonizing the bioresin bulk with tissue-specific neo-deposited extracellular matrix. Importantly, printed hydrogels embedding complex channels with perfusable lumen (diameter <200 μm) were obtained, replicating anatomical 3D networks with out-of-plane branches (i.e. brain vessels) that cannot otherwise be reproduced by extrusion bioprinting. This versatile bioresin platform opens new avenues for the widespread adoption of lithographic biofabrication, and for bioprinting complex channel-laden constructs with envisioned applications in regenerative medicine and hydrogel-based organ-on-a-chip devices.
Collapse
Affiliation(s)
- Riccardo Levato
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands
- Department of Orthopaedics, University Medical Center Utrecht, the Netherlands
| | - Khoon S Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, the Netherlands
| | - Wanlu Li
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, USA
| | - Ane Urigoitia Asua
- Department of Orthopaedics, University Medical Center Utrecht, the Netherlands
| | - Laura Blanco Peña
- Department of Orthopaedics, University Medical Center Utrecht, the Netherlands
| | - Mian Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, USA
| | - Marc Falandt
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands
| | | | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, the Netherlands
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, USA
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, the Netherlands
| | - Jos Malda
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands
- Department of Orthopaedics, University Medical Center Utrecht, the Netherlands
| |
Collapse
|
31
|
Duan A, Shen K, Li B, Li C, Zhou H, Kong R, Shao Y, Qin J, Yuan T, Ji J, Guo W, Wang X, Xue T, Li L, Huang X, Sun Y, Cai Z, Liu W, Liu F. Extracellular vesicles derived from LPS-preconditioned human synovial mesenchymal stem cells inhibit extracellular matrix degradation and prevent osteoarthritis of the knee in a mouse model. Stem Cell Res Ther 2021; 12:427. [PMID: 34321073 PMCID: PMC8317426 DOI: 10.1186/s13287-021-02507-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/11/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Previous studies report that lipopolysaccharide (LPS)-preconditioned mesenchymal stem cells have enhanced trophic support and improved regenerative and repair properties. Extracellular vesicles secreted by synovial mesenchymal stem cells (EVs) can reduce cartilage damage caused by osteoarthritis (OA). Previous studies show that extracellular vesicles secreted by LPS-preconditioned synovial mesenchymal stem cells (LPS-pre EVs) can improve the response to treatment of osteoarthritis (OA). This study sought to explore effects of LPS-pre EVs on chondrocyte proliferation, migration, and chondrocyte apoptosis, as well as the protective effect of LPS-pre EVs on mouse articular cartilage. METHODS Chondrocytes were extracted to explore the effect of LPS-pre EVs on proliferation, migration, and apoptosis of chondrocytes. In addition, the effect of LPS-pre EVs on expression level of important proteins of chondrocytes was explored suing in vitro experiments. Further, intraarticular injection of LPS-pre EVs was performed on the destabilization of the medial meniscus (DMM)-induced mouse models of OA to explore the therapeutic effect of LPS-pre EVs on osteoarthritis in vivo. RESULTS Analysis showed that LPS-pre EVs significantly promoted proliferation and migration of chondrocytes and inhibited the apoptosis of chondrocytes compared with PBS and EVs. Moreover, LPS-pre EVs inhibited decrease of aggrecan and COL2A1 and increase of ADAMTS5 caused by IL-1β through let-7b. Furthermore, LPS-pre EVs significantly prevented development of OA in DMM-induced mouse models of OA. CONCLUSIONS LPS pretreatment is an effective and promising method to improve therapeutic effect of extracellular vesicles secreted from SMSCs on OA.
Collapse
Affiliation(s)
- Ao Duan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Kai Shen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Beichen Li
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210001, Jiangsu, China
| | - Cong Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Hao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Renyi Kong
- Department of Orthopedics, Xincheng Hospital of Traditional Chinese Medicine, Maanshan, 243131, Anhui, China
| | - Yuqi Shao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jian Qin
- Department of Orthopedics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, China
| | - Tangbo Yuan
- Department of Orthopedics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, China
| | - Juan Ji
- Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211100, China
| | - Wei Guo
- Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211100, China
| | - Xipeng Wang
- Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211100, China
| | - Tengfei Xue
- Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211100, China
| | - Lei Li
- Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211100, China
| | - Xinxin Huang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yuqin Sun
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Zhenyu Cai
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Wei Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Feng Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
32
|
Rahman S, Szojka ARA, Liang Y, Kunze M, Goncalves V, Mulet-Sierra A, Jomha NM, Adesida AB. Inability of Low Oxygen Tension to Induce Chondrogenesis in Human Infrapatellar Fat Pad Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:703038. [PMID: 34381784 PMCID: PMC8350173 DOI: 10.3389/fcell.2021.703038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Articular cartilage of the knee joint is avascular, exists under a low oxygen tension microenvironment, and does not self-heal when injured. Human infrapatellar fat pad-sourced mesenchymal stem cells (IFP-MSC) are an arthroscopically accessible source of mesenchymal stem cells (MSC) for the repair of articular cartilage defects. Human IFP-MSC exists physiologically under a low oxygen tension (i.e., 1-5%) microenvironment. Human bone marrow mesenchymal stem cells (BM-MSC) exist physiologically within a similar range of oxygen tension. A low oxygen tension of 2% spontaneously induced chondrogenesis in micromass pellets of human BM-MSC. However, this is yet to be demonstrated in human IFP-MSC or other adipose tissue-sourced MSC. In this study, we explored the potential of low oxygen tension at 2% to drive the in vitro chondrogenesis of IFP-MSC. We hypothesized that 2% O2 will induce stable chondrogenesis in human IFP-MSC without the risk of undergoing endochondral ossification at ectopic sites of implantation. METHODS Micromass pellets of human IFP-MSC were cultured under 2% O2 or 21% O2 (normal atmosphere O2) in the presence or absence of chondrogenic medium with transforming growth factor-β3 (TGFβ3) for 3 weeks. Following in vitro chondrogenesis, the resulting pellets were implanted in immunodeficient athymic nude mice for 3 weeks. RESULTS A low oxygen tension of 2% was unable to induce chondrogenesis in human IFP-MSC. In contrast, chondrogenic medium with TGFβ3 induced in vitro chondrogenesis. All pellets were devoid of any evidence of undergoing endochondral ossification after subcutaneous implantation in athymic mice.
Collapse
Affiliation(s)
- Samia Rahman
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Alexander R. A. Szojka
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Yan Liang
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Victoria Goncalves
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Nadr M. Jomha
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Adetola B. Adesida
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of Alberta Hospital, Edmonton, AB, Canada
| |
Collapse
|
33
|
Chijimatsu R, Miwa S, Okamura G, Miyahara J, Tachibana N, Ishikura H, Higuchi J, Maenohara Y, Tsuji S, Sameshima S, Takagi K, Nakazato K, Kawaguchi K, Yamagami R, Inui H, Taketomi S, Tanaka S, Saito T. Divergence in chondrogenic potential between in vitro and in vivo of adipose- and synovial-stem cells from mouse and human. Stem Cell Res Ther 2021; 12:405. [PMID: 34266496 PMCID: PMC8281654 DOI: 10.1186/s13287-021-02485-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Somatic stem cell transplantation has been performed for cartilage injury, but the reparative mechanisms are still conflicting. The chondrogenic potential of stem cells are thought as promising features for cartilage therapy; however, the correlation between their potential for chondrogenesis in vitro and in vivo remains undefined. The purpose of this study was to investigate the intrinsic chondrogenic condition depends on cell types and explore an indicator to select useful stem cells for cartilage regeneration. METHODS The chondrogenic potential of two different stem cell types derived from adipose tissue (ASCs) and synovium (SSCs) of mice and humans was assessed using bone morphogenic protein-2 (BMP2) and transforming growth factor-β1 (TGFβ1). Their in vivo chondrogenic potential was validated through transplantation into a mouse osteochondral defect model. RESULTS All cell types showed apparent chondrogenesis under the combination of BMP2 and TGFβ1 in vitro, as assessed by the formation of proteoglycan- and type 2 collagen (COL2)-rich tissues. However, our results vastly differed with those observed following single stimulation among species and cell types; apparent chondrogenesis of mouse SSCs was observed with supplementation of BMP2 or TGFβ1, whereas chondrogenesis of mouse ASCs and human SSCs was observed with supplementation of BMP2 not TGFβ1. Human ASCs showed no obvious chondrogenesis following single stimulation. Mouse SSCs showed the formation of hyaline-like cartilage which had less fibrous components (COL1/3) with supplementation of TGFβ1. However, human cells developed COL1/3+ tissues with all treatments. Transcriptomic analysis for TGFβ receptors and ligands of cells prior to chondrogenic induction did not indicate their distinct reactivity to the TGFβ1 or BMP2. In the transplanted site in vivo, mouse SSCs formed hyaline-like cartilage (proteoglycan+/COL2+/COL1-/COL3-) but other cell types mainly formed COL1/3-positive fibrous tissues in line with in vitro reactivity to TGFβ1. CONCLUSION Optimal chondrogenic factors driving chondrogenesis from somatic stem cells are intrinsically distinct among cell types and species. Among them, the response to TGFβ1 may possibly represent the fate of stem cells when locally transplanted into cartilage defects.
Collapse
Affiliation(s)
- Ryota Chijimatsu
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Satoshi Miwa
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Junya Miyahara
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naohiro Tachibana
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hisatoshi Ishikura
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junya Higuchi
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuji Maenohara
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Shin Sameshima
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kentaro Takagi
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keiu Nakazato
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kohei Kawaguchi
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryota Yamagami
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Inui
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shuji Taketomi
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sakae Tanaka
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taku Saito
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
34
|
Migliorini F, Berton A, Salvatore G, Candela V, Khan W, Longo UG, Denaro V. Autologous Chondrocyte Implantation and Mesenchymal Stem Cells for the Treatments of Chondral Defects of the Knee- A Systematic Review. Curr Stem Cell Res Ther 2021; 15:547-556. [PMID: 32081109 DOI: 10.2174/1574888x15666200221122834] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/23/2019] [Accepted: 01/09/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND There is still a lack of consensus about the best treatment of chondral defects of the knee. We conducted a systematic PRISMA review to evaluate clinical outcomes of Autologous Chondrocyte Implantation (ACI) and Mesenchymal Stem Cell (MSC) injections for the treatment of focal chondral defects of the knee. METHODS A systematic review of literature was performed according to the PRISMA guidelines. All the articles reporting data on ACI and MSC treatments for chondral defects of the knee were considered for inclusion. The main databases were accessed: PubMed, Medline, CINAHL, Cochrane, Embase and Google Scholar. The statistical analysis was performed using the Review Manager Software. RESULTS In the p-ACI group (987 knees), the Cincinnati Score improved by 18.94% (p=0.1), VAS by 38% (p=0.01), Tegner score by 19.11% (p=0.03), Lysholm score by 22.40% (p=0.01), IKCD by 27.36% (p=0.003). In the c-ACI group (444 knees), the Cincinnati Score improved by 23.80% (p=0.08), KOOS by 23.48% (p=0.03), VAS by 33.2% (p=0.005), IKDC by 33.30% (p=0.005). In the m-ACI group (599 knees), the Cincinnati Score improved by 26.80% (p=0.08), KOOS by 31.59% (p=0.1), VAS by 30.43% (p=0.4), Tegner score by 23.1% (p=0.002), Lysholm score by 31.14% (p=0.004), IKCD by 30.57% (p<0.001). In the MSCs group (291 knees), the KOOS improved by 29.7% (p=0.003), VAS by 41.89% (p<0.001), Tegner score by 25.81% (p=0.003), Lysholm score by 36.96% (p<0.001), IKCD by 30.57% (p=0.001). CONCLUSION Both ACI and MSC therapies can be considered as a concrete solution to treat focal chondral defects of the knee.
Collapse
Affiliation(s)
- Filippo Migliorini
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo, 200, 00128 Trigoria, Rome, Italy
| | - Alessandra Berton
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo, 200, 00128 Trigoria, Rome, Italy
| | - Giuseppe Salvatore
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo, 200, 00128 Trigoria, Rome, Italy
| | - Vincenzo Candela
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo, 200, 00128 Trigoria, Rome, Italy
| | - Wasim Khan
- Division of Trauma & Orthopaedic Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 2QQ, United Kingdom
| | - Umile G Longo
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo, 200, 00128 Trigoria, Rome, Italy
| | - Vincenzo Denaro
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo, 200, 00128 Trigoria, Rome, Italy
| |
Collapse
|
35
|
Liu C. Application of marine collagen for stem-cell-based therapy and tissue regeneration (Review). MEDICINE INTERNATIONAL 2021; 1:6. [PMID: 36698868 PMCID: PMC9855277 DOI: 10.3892/mi.2021.5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/22/2021] [Indexed: 01/28/2023]
Abstract
Tissue engineering and regenerative medicine is becoming an important component in modern biological scientific research. Tissue engineering, a branch of regenerative medicine, is a field that is actively developing to meet the challenges presented in biomedical applications. This particularly applies to the research area of stem cells and biomaterials, due to both being pivotal determinants for the successful restoration or regeneration of damaged tissues and organs. Recently, the development of innovative marine collagen-based biomaterials has attracted attention due to the reported environmentally friendly properties, the lack of zoonotic disease transmission, biocompatibility, bioactivity, the lack of ethics-related concerns and cost-effectiveness for manufacturing. The present review aimed to summarize the potential application and function of marine collagen in stem cell research in a medical and clinical setting. In addition, the present review cited recent studies regarding the latest research advances into using marine collagen for cartilage, bone, periodontal and corneal regeneration. It also characterized the distinct advantages of using marine collagen for stem cell-based tissue repair and regeneration. In addition, the present review comprehensively discussed the most up to date information on stem cell biology, particularly the possibility of treating stem cells with marine collagen to maximize their multi-directional differentiation capability, which highlights the potential use of marine collagen in regenerative medicine. Furthermore, recent research progress on the potential immunomodulatory capacity of mesenchymal stem cells following treatment with marine collagen to improve the understanding of cell-matrix interactions was investigated. Finally, perspectives on the possible future research directions for the application of marine collagen in the area of regenerative medicine are provided.
Collapse
Affiliation(s)
- Chao Liu
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
36
|
Zhou X, Xu W, Wang Y, Zhang H, Zhang L, Li C, Yao S, Huang Z, Huang L, Luo D. LncRNA DNM3OS regulates GREM2 via miR-127-5p to suppress early chondrogenic differentiation of rat mesenchymal stem cells under hypoxic conditions. Cell Mol Biol Lett 2021; 26:22. [PMID: 34049478 PMCID: PMC8161583 DOI: 10.1186/s11658-021-00269-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background Improved chondrogenic differentiation of mesenchymal stem cells (MSCs) by genetic regulation is a potential method for regenerating articular cartilage. MiR-127-5p has been reported to promote cartilage differentiation of rat bone marrow MSCs (rMSCs); however, the regulatory mechanisms underlying hypoxia-stimulated chondrogenic differentiation remain unknown. Methods rMSCs were induced to undergo chondrogenic differentiation under normoxic or hypoxic conditions. Expression of lncRNA DNM3OS, miR-127-5p, and GREM2 was detected by quantitative real-time PCR. Proteoglycans were detected by Alcian blue staining. Western blot assays were performed to examine the relative levels of GREM2 and chondrogenic differentiation related proteins. Luciferase reporter assays were performed to assess the association among DNM3OS, miR-127-5p, and GREM2. Results MiR-127-5p levels were upregulated, while DNM3OS and GREM2 levels were downregulated in rMSCs induced to undergo chondrogenic differentiation, and those changes were attenuated by hypoxic conditions (1% O2). Further in vitro experiments revealed that downregulation of miR-127-5p reduced the production of proteoglycans and expression of chondrogenic differentiation markers (COL1A1, COL2A1, SOX9, and ACAN) and osteo/chondrogenic markers (BMP-2, p-SMAD1/2). MiR-127-5p overexpression produced the opposite results in rMSCs induced to undergo chondrogenic differentiation under hypoxic conditions. GREM2 was found to be a direct target of miR-127-5p, which was suppressed in rMSCs undergoing chondrogenic differentiation. Moreover, DNM3OS could directly bind to miR-127-5p and inhibit chondrogenic differentiation of rMSCs via regulating GREM2. Conclusions Our study revealed a novel molecular pathway (DNM3OS/miR-127-5p/GREM2) that may be involved in hypoxic chondrogenic differentiation.
Collapse
Affiliation(s)
- Xiaozhong Zhou
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wangyang Xu
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Yeyang Wang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Hui Zhang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Li Zhang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Chao Li
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Shun Yao
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Zixiang Huang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Lishan Huang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Dixin Luo
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.
| |
Collapse
|
37
|
Tortorici M, Petersen A, Ehrhart K, Duda GN, Checa S. Scaffold-Dependent Mechanical and Architectural Cues Guide Osteochondral Defect Healing in silico. Front Bioeng Biotechnol 2021; 9:642217. [PMID: 33659244 PMCID: PMC7917217 DOI: 10.3389/fbioe.2021.642217] [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: 12/15/2020] [Accepted: 01/22/2021] [Indexed: 11/13/2022] Open
Abstract
Osteochondral defects in joints require surgical intervention to relieve pain and restore function. However, no current treatment enables a complete reconstitution of the articular surface. It is known that both mechanical and biological factors play a key role on osteochondral defect healing, however the underlying principles and how they can be used in the design of treatment strategies remain largely unknown. To unravel the underlying principles of mechanobiology in osteochondral defect healing, i.e., how mechanical stimuli can guide biological tissue formation, we employed a computational approach investigating the scaffold-associated mechanical and architectural properties that would enable a guided defect healing. A previous computer model of the knee joint was further developed to simulate healing of an empty osteochondral defect. Then, scaffolds were implanted in the defect and their architectures and material properties were systematically varied to identify their relevance in osteochondral defect healing. Scaffold mechanical and architectural properties were capable of influencing osteochondral defect healing. Specifically, scaffold material elastic modulus values in the range of cancellous bone (low GPa range) and a scaffold architecture that provided stability, i.e., resistance against displacement, in both the main loading direction and perpendicular to it supported the repair process. The here presented model, despite its simplifications, is regarded as a powerful tool to screen for promising properties of novel scaffold candidates fostering osteochondral defect regeneration prior to their implementation in vivo.
Collapse
Affiliation(s)
- Martina Tortorici
- Julius Wolff Institute, Charité Universitaetsmedizin Berlin, Berlin, Germany.,Berlin-Branderburg School for Regenerative Therapies, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Ansgar Petersen
- Julius Wolff Institute, Charité Universitaetsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Klara Ehrhart
- Julius Wolff Institute, Charité Universitaetsmedizin Berlin, Berlin, Germany.,Continuum Mechanics and Material Theory, Faculty V of Mechanical Engineering and Transport Systems, Institute of Mechanics, Technische Universtitaet Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute, Charité Universitaetsmedizin Berlin, Berlin, Germany.,Berlin-Branderburg School for Regenerative Therapies, Charité Universitaetsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute, Charité Universitaetsmedizin Berlin, Berlin, Germany.,Berlin-Branderburg School for Regenerative Therapies, Charité Universitaetsmedizin Berlin, Berlin, Germany
| |
Collapse
|
38
|
Wang Z, Yan K, Ge G, Zhang D, Bai J, Guo X, Zhou J, Xu T, Xu M, Long X, Hao Y, Geng D. Exosomes derived from miR-155-5p-overexpressing synovial mesenchymal stem cells prevent osteoarthritis via enhancing proliferation and migration, attenuating apoptosis, and modulating extracellular matrix secretion in chondrocytes. Cell Biol Toxicol 2021; 37:85-96. [PMID: 33099657 DOI: 10.1007/s10565-020-09559-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 09/23/2020] [Indexed: 11/29/2022]
Abstract
Synovial mesenchymal stem cells (SMSCs) have the potential to attenuate osteoarthritis (OA)-induced injury. The role and mechanism of SMSC-derived exosomes (SMSC-Exos), pivotal paracrine factors of stem cells, in OA-associated injury remain unclear. We aimed to confirm the effect of SMSC-Exos with specific modifications on OA-induced damage and to investigate the potential molecular mechanisms. Exosomes derived from miR-155-5p-overexpressing SMSCs (SMSC-155-5p-Exos) and SMSCs (SMSC-Exos) were isolated and characterized. CCK-8, Transwell, and Western blot analyses were used to detect proliferation, migration, extracellular matrix (ECM) secretion, and apoptosis of osteoarthritic chondrocytes. The therapeutic effect of exosomes in a mouse model of OA was examined using immunohistochemical staining and OARSI scores. SPSS 17.0 and GraphPad software were used for all statistical analyses in this study. The SMSC-Exos enhanced the proliferation and migration and inhibited the apoptosis of osteoarthritic chondrocytes but had no effect on ECM secretion. The miR-155-5p-overexpressing exosomes showed common characteristics of exosomes in vitro and further promoted ECM secretion by targeting Runx2. Thus, the SMSC-155-5p-Exos promoted proliferation and migration, suppressed apoptosis and enhanced ECM secretion of osteoarthritic chondrocytes, and effectively prevented OA in a mouse model. In addition, overexpression of Runx2 partially reversed the effect of the SMSC-155-5p-Exos on osteoarthritic chondrocytes. Given the insufficient effect of the SMSC-Exos on the ECM secretion of osteoarthritic chondrocytes, we modified the SMSM-Exos and demonstrated that the SMSC-155-5p-Exos could prevent OA. Exosomes derived from modified SMSCs may be a new treatment strategy to prevent OA. Graphical abstract.
Collapse
Affiliation(s)
- Zhirong Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shizi Road, 215006, Suzhou, People's Republic of China
| | - Kai Yan
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, 215006, Suzhou, People's Republic of China
| | - Gaoran Ge
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shizi Road, 215006, Suzhou, People's Republic of China
| | - Di Zhang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, 215006, Suzhou, People's Republic of China
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shizi Road, 215006, Suzhou, People's Republic of China
| | - Xiaobin Guo
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shizi Road, 215006, Suzhou, People's Republic of China
| | - Jing Zhou
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, 215006, Suzhou, People's Republic of China
| | - Tianpeng Xu
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, 215006, Suzhou, People's Republic of China
| | - Menglei Xu
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, 215006, Suzhou, People's Republic of China
| | - Xiao Long
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shizi Road, 215006, Suzhou, People's Republic of China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, 215006, Suzhou, People's Republic of China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188, Shizi Road, 215006, Suzhou, People's Republic of China.
| |
Collapse
|
39
|
Liu J, Lu Y, Xing F, Liang J, Wang Q, Fan Y, Zhang X. Cell-free scaffolds functionalized with bionic cartilage acellular matrix microspheres to enhance the microfracture treatment of articular cartilage defects. J Mater Chem B 2021; 9:1686-1697. [PMID: 33491727 DOI: 10.1039/d0tb02616f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Microfracture surgery remains the most popular treatment for articular cartilage lesions in the clinic, but often leads to the formation of inferior fibrocartilage tissue and damage to subchondral bone. To overcome these problems, extracellular matrix (ECM) scaffolds derived from decellularized natural cartilaginous tissues were introduced and showed excellent biological properties to direct the differentiation of bone marrow stem cells. However, besides the limited allogenic/allogenic supply and the risk of disease transfer from xenogeneic tissues, the effectiveness of ECM scaffolds always varied with a high variability of natural tissue quality. In this study, we developed composite scaffolds functionalized with a cell-derived ECM source, namely, bionic cartilage acellular matrix microspheres (BCAMMs), that support the chondrogenic differentiation of bone marrow cells released from microfracture. The scaffolds with BCAMMs at different developmental stages were investigated in articular cartilage regeneration and subchondral bone repair. Compared to microfracture, the addition of cell-free BCAMM scaffolds has demonstrated a great improvement of regenerated cartilage tissue quality in a rabbit model as characterized by a semi-quantitative analysis of cells, histology and biochemical assays as well as micro-CT images. Moreover, the variation in ECM properties was found to significantly affect the cartilage regeneration, highlighting the challenges of homogenous scaffolds in working with microfracture. Together, our results demonstrate that the biofunctionalized BCAMM scaffold with cell-derived ECM shows great potential to combine with microfracture for clinical translation to repair cartilage defects.
Collapse
Affiliation(s)
- Jun Liu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China. and State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yan Lu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Fei Xing
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, Sichuan, China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| |
Collapse
|
40
|
Cellular Technologies in Traumatology: from Cells to Tissue Engineering. ACTA BIOMEDICA SCIENTIFICA 2021. [DOI: 10.29413/abs.2020-5.6.8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
41
|
van Schaik TJA, Gaul F, Dorthé EW, Lee EE, Grogan SP, D’Lima DD. Development of an Ex Vivo Murine Osteochondral Repair Model. Cartilage 2021; 12:112-120. [PMID: 30373381 PMCID: PMC7755972 DOI: 10.1177/1947603518809402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE Mouse models are commonly used in research applications due to the relatively low cost, highly characterized strains, as well as the availability of many genetically modified phenotypes. In this study, we characterized an ex vivo murine osteochondral repair model using human infrapatellar fat pad (IPFP) progenitor cells. DESIGN Femurs from euthanized mice were removed and clamped in a custom multidirectional vise to create cylindrical osteochondral defects 0.5 mm in diameter and 0.5 mm deep in both condyles. The IPFP contains progenitors that are a promising cell source for the repair of osteochondral defects. For proof of concept, human IPFP-derived progenitor cells, from osteoarthritic (OA) patients, cultured as pellets, were implanted into the defects and cultured in serum-free medium with TGFβ3 for 3 weeks and then processed for histology and immunostaining. RESULTS The custom multidirectional vise enabled reproducible creation of osteochondral defects in murine femoral condyles. Implantation of IPFP-derived progenitor cells led to development of cartilaginous tissue with Safranin O staining and deposition of collagen type II in the extracellular matrix. CONCLUSIONS We showed feasibility in creating ex vivo osteochondral defects and demonstrated the regenerative potential of OA human IPFP-derived progenitors in mouse femurs. The murine model can be used to study the effects of aging and OA on tissue regeneration and to explore molecular mechanisms of cartilage repair using genetically modified mice.
Collapse
Affiliation(s)
- Thomas J. A. van Schaik
- Orthopaedic Research Laboratory, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Florian Gaul
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Erik W. Dorthé
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Emily E. Lee
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Shawn P. Grogan
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Darryl D. D’Lima
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA,Darryl D. D’Lima, Scripps Health, Shiley Center for Orthopaedic Research and Education at Scripps Clinic, 10666 North Torrey Pines Road, MS126, La Jolla, CA 92027, USA.
| |
Collapse
|
42
|
BMSC-derived exosomes from congenital polydactyly tissue alleviate osteoarthritis by promoting chondrocyte proliferation. Cell Death Discov 2020; 6:142. [PMID: 33303743 PMCID: PMC7730395 DOI: 10.1038/s41420-020-00374-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/12/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
In the past decade, mesenchymal stem cells (MSCs) have been widely used for the treatment of osteoarthritis (OA), and exosomes may play a major role. Here, we acquired a special kind of MSCs from the bone marrow of surgically resected tissue from the hand of a patient with polydactyly. Experiments were focused on the role of polydactyly bone marrow-derived MSCs (pBMSCs) in osteoarthritis. The results showed that the pBMSCs had a greater ability than the BMSCs to differentiate into chondrocytes. Mechanistically, the expression of BMP4 was significantly higher in the pBMSCs than it was in the BMSCs. Furthermore, we showed that the migration and proliferation of chondrocytes were stimulated by exosomes secreted by pBMSC (pBMSC-EXOs). Notably, the downregulation of BMP4 in pBMSCs by siRNA inhibited both the chondrogenic differentiation potential of the MSCs and the function of the chondrocytes. In addition, the injection of pBMSC-EXOs and BMSC-EXOs attenuated OA in an OA mouse model, but the pBMSC-EXOs had a superior therapeutic effect compared with that of the BMSC-EXOs. Taken together, the data indicate that pBMSCs have greater ability to differentiate into chondrocytes and regulate chondrocyte formation through BMP4 signaling. Therefore, pBMSC-EXOs may represent a novel treatment for OA.
Collapse
|
43
|
Li T, Chen S, Pei M. Contribution of neural crest-derived stem cells and nasal chondrocytes to articular cartilage regeneration. Cell Mol Life Sci 2020; 77:4847-4859. [PMID: 32504256 PMCID: PMC9150440 DOI: 10.1007/s00018-020-03567-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/06/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022]
Abstract
Due to poor self-regenerative potential of articular cartilage, stem cell-based regeneration becomes a hopeful approach for the treatment of articular cartilage defects. Recent studies indicate that neural crest-derived cells (NCDCs) have the potential for repairing articular cartilage with even greater chondrogenic capacity than mesoderm-derived cells (MDCs): a conventional stem cell source for cartilage regeneration. Given that NCDCs originate from a different germ layer in the early embryo compared with MDCs that give rise to articular cartilage, a mystery remains regarding their capacity for articular cartilage regeneration. In this review, we summarize the similarities and differences between MDCs and NCDCs including articular and nasal chondrocytes in cell origin, anatomy, and chondrogenic differentiation and propose that NCDCs might be promising cell origins for articular cartilage regeneration.
Collapse
Affiliation(s)
- Tianyou Li
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
- Department of Pediatric Orthopaedics, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Song Chen
- Department of Orthopaedics, The General Hospital of Western Theater Command, Chengdu, 610083, Sichuan, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA.
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
44
|
Mardones R, Giai Via A, Pipino G, Jofre CM, Muñoz S, Narvaez E, Maffulli N. BM-MSCs differentiated to chondrocytes for treatment of full-thickness cartilage defect of the knee. J Orthop Surg Res 2020; 15:455. [PMID: 33023626 PMCID: PMC7539404 DOI: 10.1186/s13018-020-01852-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/30/2020] [Indexed: 01/06/2023] Open
Abstract
Background Full-thickness articular cartilage injury of the knee is a major cause of disability. The aim of this study is to assess the outcome of patients treated with differentiated to chondrocytes bone marrow mesenchymal stem cells (BM-MSCs) cultured on a collagen type I/III (Chondro-Gide®) scaffold. The secondary aim was to confirm the absence of adverse events. Methods Fifteen patients (19 knees) with symptomatic full-thickness cartilage defects of the knee were enrolled. Bone marrow was harvested from the iliac crest, BM-MSCs were prepared, and expanded cells were grown in a standard medium or in a standard culture medium containing TGF-β. BM-MSCs differentiated to chondrocytes were seeded in a porcine collagen type I/III scaffold (Chondro-Gide®) and cultured in TGF-β containing media. After 4 weeks, the membrane was sutured on the cartilage defect. All patients underwent plain radiographs (antero-posterior, lateral, and axial view of the patella) and MRI of the affected knee. The Oxford knee score, the Lyhsolm scale, and the VAS score were administered to all patients. At final follow-up a MRI for the study of articular cartilage was undertaken. Results The mean size of the cartilage lesions was 20 × 17 mm (range, 15 × 10 mm–30 × 30 mm). At final follow-up, the median Oxford knee score and Lyhsolm scale scores significantly improved from 29 (range 12–39; SD 7.39) to 45 (range 24–48; SD 5.6) and from 55.5 (range 25–81; SD 17.7) to 94.5 (58–100; SD 10.8), respectively. Pain, according to the VAS score, significantly improved. Sixty percent of patients reported their satisfaction as excellent, 20% as good, 14% as fair, and 1 patient as poor. Conclusion The treatment of full-thickness chondral injuries of the knee with differentiated to chondrocytes BM-MSCs and Chondro-Gide® scaffold showed encouraging outcomes. Further studies involving more patients, and with longer follow-up, are required to evaluate the effectiveness of the treatment and the long-term results.
Collapse
Affiliation(s)
| | - Alessio Giai Via
- Department of Orthopaedic Surgery and Traumatology, San Camillo-Fortalini Hospital, Rome, Italy.
| | - Gennaro Pipino
- UCM Malta, Campus of Lugano, Lugano, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Villa Regina Hospital, Bologna, Italy.,Department of Orthopedics and Physiotherapy, UCM University, Msida, Malta
| | - Claudio M Jofre
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago, Chile
| | - Sara Muñoz
- Department of Radiology, Clinica Las Condes, Lo Fontecilla 441, Las Condes, Santiago de Chile, Chile
| | - Edgar Narvaez
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago, Chile
| | - Nicola Maffulli
- Institute of Science and Technology in Medicine, Keele University School of Medicine, Thornburrow Drive, Stoke on Trent, England.,Department of Orthopedics and Physiotherapy, UCM University, Msida, Malta
| |
Collapse
|
45
|
Chen X, Wang Y, Chen R, Qu N, Zhang B, Xia C. Suppressing PLCγ1 enhances osteogenic and chondrogenic potential of BMSCs. Biochem Biophys Res Commun 2020; 532:292-299. [PMID: 32868075 DOI: 10.1016/j.bbrc.2020.08.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/14/2020] [Indexed: 11/28/2022]
Abstract
Phosphatidylcholine-specific phospholipase Cγ1 (PLCγ1) is involved in regulating cell metabolism. However, little is known how PLCγ1 directs BMSC differentiation. Here, we investigated the role of PLCγ1 in rat BMSC differentiation into osteoblasts and chondrocytes. The results of Alizarin red and Alcian blue staining showed that PLCγ1 inhibitor U73122 significantly enhanced the mineralization capacity and proteoglycan deposition of BMSCs. The results of qPCR technique and Western blot analysis showed that long-term treatment of U73122 enhanced COL1A1 and OPG mRNA levels and Collagen 1A1, BMP2, and p-Smad1/5/9 protein levels and that short-term treatment of U73122 enhanced COL2A1 and SOX9 mRNA levels and Collagen 2, SOX9, Aggrecan, TGF-β3, and p-Smad2/3 protein levels. Decreased p-mTOR and p-P38 contributed to enhanced osteogenic potentials of BMSCs and increased p-P38 contributed to enhanced chondrogenic potentials of BMSCs. The scaffold transplantation with U73122+BMSC was more efficacious than BMSC alone for osteochondral defect repair in a rat model. Therefore, suppressing PLCγ1 could improve the capacity to effectively use BMSCs for cell therapy of osteochondral defect.
Collapse
Affiliation(s)
- Xiaolei Chen
- Bone & Joint Research Institute, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Yue Wang
- Bone & Joint Research Institute, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China
| | - Ri Chen
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ning Qu
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Bing Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Chun Xia
- Bone & Joint Research Institute, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, 361004, China.
| |
Collapse
|
46
|
Clinical Application Status of Articular Cartilage Regeneration Techniques: Tissue-Engineered Cartilage Brings New Hope. Stem Cells Int 2020; 2020:5690252. [PMID: 32676118 PMCID: PMC7345961 DOI: 10.1155/2020/5690252] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 05/22/2020] [Accepted: 05/28/2020] [Indexed: 12/16/2022] Open
Abstract
Hyaline articular cartilage lacks blood vessels, lymphatics, and nerves and is characterised by limited self-repair ability following injury. Traditional techniques of articular cartilage repair and regeneration all have certain limitations. The development of tissue engineering technology has brought hope to the regeneration of articular cartilage. The strategies of tissue-engineered articular cartilage can be divided into three types: “cell-scaffold construct,” cell-free, and scaffold-free. In “cell-scaffold construct” strategies, seed cells can be autologous chondrocytes or stem. Among them, some commercial products with autologous chondrocytes as seed cells, such as BioSeed®-C and CaReS®, have been put on the market and some products are undergoing clinical trials, such as NOVOCART® 3D. The stem cells are mainly pluripotent stem cells and mesenchymal stem cells from different sources. Cell-free strategies that indirectly utilize the repair and regeneration potential of stem cells have also been used in clinical settings, such as TruFit and MaioRegen. Finally, the scaffold-free strategy is also a new development direction, and the short-term repair results of related products, such as NOVOCART® 3D, are encouraging. In this paper, the commonly used techniques of articular cartilage regeneration in surgery are reviewed. By studying different strategies and different seed cells, the clinical application status of tissue-engineered articular cartilage is described in detail.
Collapse
|
47
|
Wu Y, Yang Z, Denslin V, Ren X, Lee CS, Yap FL, Lee EH. Repair of Osteochondral Defects With Predifferentiated Mesenchymal Stem Cells of Distinct Phenotypic Character Derived From a Nanotopographic Platform. Am J Sports Med 2020; 48:1735-1747. [PMID: 32191492 DOI: 10.1177/0363546520907137] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Articular cartilage has a zonal architecture and biphasic mechanical properties. The recapitulation of surface lubrication properties with high compressibility of the deeper layers of articular cartilage during regeneration is essential in achieving long-term cartilage integrity. Current clinical approaches for cartilage repair, especially with the use of mesenchymal stem cells (MSCs), have yet to restore the hierarchically organized architecture of articular cartilage. HYPOTHESIS MSCs predifferentiated on surfaces with specific nanotopographic patterns can provide phenotypically stable and defined chondrogenic cells and, when delivered as a bilayered stratified construct at the cartilage defect site, will facilitate the formation of functionally superior cartilage tissue in vivo. STUDY DESIGN Controlled laboratory study. METHODS MSCs were subjected to chondrogenic differentiation on specific nanopatterned surfaces. The phenotype of the differentiated cells was assessed by the expression of cartilage markers. The ability of the 2-dimensional nanopattern-generated chondrogenic cells to retain their phenotypic characteristics after removal from the patterned surface was tested by subjecting the enzymatically harvested cells to 3-dimensional fibrin hydrogel culture. The in vivo efficacy in cartilage repair was demonstrated in an osteochondral rabbit defect model. Repair by bilayered construct with specific nanopattern predifferentiated cells was compared with implantation with cell-free fibrin hydrogel, undifferentiated MSCs, and mixed-phenotype nanopattern predifferentiated MSCs. Cartilage repair was evaluated at 12 weeks after implantation. RESULTS Three weeks of predifferentiation on 2-dimensional nanotopographic patterns was able to generate phenotypically stable chondrogenic cells. Implantation of nanopatterned differentiated MSCs as stratified bilayered hydrogel constructs improved the repair quality of cartilage defects, as indicated by histological scoring, mechanical properties, and polarized microscopy analysis. CONCLUSION Our results indicate that with an appropriate period of differentiation, 2-dimensional nanotopographic patterns can be employed to generate phenotypically stable chondrogenic cells, which, when implanted as stratified bilayered hydrogel constructs, were able to form functionally superior cartilage tissue. CLINICAL RELEVANCE Our approach provides a relatively straightforward method of obtaining large quantities of zone-specific chondrocytes from MSCs to engineer a stratified cartilage construct that could recapitulate the zonal architecture of hyaline cartilage, and it represents a significant improvement in current MSC-based cartilage regeneration.
Collapse
Affiliation(s)
- Yingnan Wu
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zheng Yang
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Vinitha Denslin
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore
| | - XiaFei Ren
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chang Sheng Lee
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Fung Ling Yap
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), Singapore
| | - Eng Hin Lee
- Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
48
|
Increased Expression of Sox9 during Balance of BMSCs/Chondrocyte Bricks in Platelet-Rich Plasma Promotes Construction of a Stable 3-D Chondrogenesis Microenvironment for BMSCs. Stem Cells Int 2020; 2020:5492059. [PMID: 32565827 PMCID: PMC7271054 DOI: 10.1155/2020/5492059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/03/2020] [Accepted: 03/17/2020] [Indexed: 11/17/2022] Open
Abstract
Sox9 is an intrinsic transcription factor related to the determination and maintenance of chondrogenic lineage of bone marrow mesenchymal stem cells (BMSCs). In recent research, we have proved that fragmented chondrocyte aggregates (cell bricks) could promote chondrogenesis of BMSCs in vivo. However, it is still unknown whether the ratio of BMSCs/chondrocyte bricks has a significant influence on 3-D cartilage regeneration and related molecular mechanism. To address this issue, the current study subcutaneously injected three groups of cell complex with different rabbit BMSCs/chondrocyte bricks' ratios (1 : 2, 1 : 1, and 2 : 1) into nude mice. Gross morphology observation, histological and immunohistochemical assays, biochemical analysis, gene expression analysis, and western blot were used to compare the influence of different BMSCs/chondrocyte bricks' ratios on the properties of tissue-engineered cartilage and explore the related molecular mechanism. The constructs of 1 : 1 BMSCs/chondrocyte bricks, (B1CB1) group resulted in persistent chondrogenesis with appropriate morphology and adequate central nutritional perfusion without ossification. The related mechanism is that increased expression of Sox9 in the B1C1 group promoted chondrogenesis and inhibited the osteogenesis of BMSCs through upregulating Col-II as well as downregulating RUNX2 and downstream of Col-X and Col-I by upregulating Nkx3.2. This study demonstrated that BMSCs/chondrocyte bricks 1:1 should be a suitable ratio and the Sox9-Nkx3.2-RUNX2 pathway was a related mechanism which played an important role in the niche for stable chondrogenesis of BMSCs constructed by chondrocyte bricks and PRP.
Collapse
|
49
|
Connective Tissue Progenitor Analysis of Bone Marrow Aspirate Concentrate Harvested From the Body of the Ilium During Arthroscopic Acetabular Labral Repair. Arthroscopy 2020; 36:1311-1320. [PMID: 31958539 DOI: 10.1016/j.arthro.2019.11.125] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/24/2019] [Accepted: 11/24/2019] [Indexed: 02/02/2023]
Abstract
PURPOSE To evaluate the number and concentration of progenitors of the bone marrow aspirate (BMA) harvest from the body of the ilium in comparison with other established aspiration sites. METHODS The inclusion criteria consisted of primary hip arthroscopy for acetabular labral tear. BMA was performed by placing an aspiration needle into the body of ilium just proximal to the sourcil in 33 patients. The BMA was centrifuged and processed in the operating room, resulting in approximately 3 to 5 mL of bone marrow aspirate concentrate (BMAC). Samples of both BMA and BMAC sample were analyzed. RESULTS The cohort of 30 patients had a mean number of nucleated cells of 24.0 million nucleated cells/cc of BMA. The BMAC samples had a mean connective tissue progenitor (CTP) cell concentration of 879.3 stem cells/cc of BMAC, a mean CTP prevalence of 34.1 stem cells/million nucleated cells, and a mean number of days to form colonies of 2.97 days. All 4 metrics of CTP harvest did not vary significantly with age, body mass index, sex, or laterality. The nucleated cell count was significantly associated with both CTP prevalence, r2 = 0.287 (P = .002), and CTP concentration, r2 = 0.388 (P < .001). CONCLUSIONS BMAC harvested from the body of the ilium during concurrent hip arthroscopy is a technically and biologically feasible option. Furthermore, the harvest site was found to have a CTP concentration that is similar or exceeds other published harvest sites. Finally, BMAC processing and application to areas of articular cartilage wear was performed efficiently and safely with no increase in morbidity or complications. CLINICAL RELEVANCE The body of the ilium is a reliable and rich source of CTP cells. This study may assist orthopaedic surgeons interested in performing biologic augmentation during hip surgery in determining a harvest site.
Collapse
|
50
|
Go G, Jeong SG, Yoo A, Han J, Kang B, Kim S, Nguyen KT, Jin Z, Kim CS, Seo YR, Kang JY, Na JY, Song EK, Jeong Y, Seon JK, Park JO, Choi E. Human adipose–derived mesenchymal stem cell–based medical microrobot system for knee cartilage regeneration in vivo. Sci Robot 2020; 5:5/38/eaay6626. [DOI: 10.1126/scirobotics.aay6626] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022]
Abstract
Targeted cell delivery by a magnetically actuated microrobot with a porous structure is a promising technique to enhance the low targeting efficiency of mesenchymal stem cell (MSC) in tissue regeneration. However, the relevant research performed to date is only in its proof-of-concept stage. To use the microrobot in a clinical stage, biocompatibility and biodegradation materials should be considered in the microrobot, and its efficacy needs to be verified using an in vivo model. In this study, we propose a human adipose–derived MSC–based medical microrobot system for knee cartilage regeneration and present an in vivo trial to verify the efficacy of the microrobot using the cartilage defect model. The microrobot system consists of a microrobot body capable of supporting MSCs, an electromagnetic actuation system for three-dimensional targeting of the microrobot, and a magnet for fixation of the microrobot to the damaged cartilage. Each component was designed and fabricated considering the accessibility of the patient and medical staff, as well as clinical safety. The efficacy of the microrobot system was then assessed in the cartilage defect model of rabbit knee with the aim to obtain clinical trial approval.
Collapse
|