1
|
Shirzad M, Daraei A, Najafzadehvarzi H, Farnoush N, Parsian H. Co-culture system of breast cancer and normal cells to investigate inflammation: using doxorubicin encapsulated in adipose-derived exosomes. Med Oncol 2024; 42:21. [PMID: 39630192 DOI: 10.1007/s12032-024-02568-2] [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: 09/12/2024] [Accepted: 11/11/2024] [Indexed: 01/23/2025]
Abstract
Doxorubicin (DOX) chemotherapy for breast cancer is an effective treatment option, but it also has disadvantages. Exosomes (EXOs) have safely and successfully transported DOX and reduced its adverse effects; however, its use is still being explored. In this study, a co-culture system of malignant and non-malignant breast cells was used to generate an in vitro model reflecting the in vivo cellular microenvironment, and the effects of this treatment were investigated by examining inflammatory genes. Extracellular matrices (EXOs) were extracted from mesenchymal stem cells derived from human adipose tissue by ultracentrifugation. Later, Western blotting, dynamic light scattering (DLS) and transmission electron microscopy methods were used to examine the properties of the EXO. DOX was encapsulated in the EXOs by sonication and the loading rate was measured by spectrophotometry. In the current study, a co-culture system was used to investigate the cytotoxic effects of free DOX and DOX encapsulated in EXOs (EXO-DOX) on various breast cell lines, including MCF-7, MCF-10A, MDA-MB-231, and A-MSC. Additionally, the expression levels of inflammatory cytokines (IL-1β, IL-6, IL-10, and TNF-α) were examined. Methylthiazolyldiphenyl-tetrazolium bromide assay demonstrated that free DOX showed the highest cytotoxicity against MCF-10A cells, followed by MCF-7 cells. Conversely, EXO-DOX indicated a greater effect on MCF-7 cells and had a lower IC50 compared to MDA-MB-231 cells. Free DOX significantly downregulated the expression of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), particularly in MCF-7 and MCF-10A cells, while concurrently upregulating IL-10 expression. EXO-DOX induced a more significant alteration in cytokine expression than the control and free DOX treatment groups. The co-culture system revealed a synergistic effect of free DOX on cancer cells while simultaneously mitigating the toxic effects of DOX on normal cells. This study suggests that EXO-DOX has promising potential as a targeted drug delivery system that could potentially improve therapeutic efficacy and minimize off-target toxicity.
Collapse
Affiliation(s)
- Moein Shirzad
- Student Research Committee, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Hossein Najafzadehvarzi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Pharmacology Department, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Nazila Farnoush
- Department of Surgery, Babol University of Medical Sciences, Babol, Iran
| | - Hadi Parsian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
2
|
Gao M, Chen Y, Li X, Li D, Liu A, Gong L, Ning Z, Nie W, Guo Y, Lv Z. Methionine supplementation regulates eggshell quality and uterine transcriptome in late-stage broiler breeders. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 19:56-69. [PMID: 39628644 PMCID: PMC11612657 DOI: 10.1016/j.aninu.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/15/2024] [Accepted: 04/27/2024] [Indexed: 12/06/2024]
Abstract
This study aimed to compare the effects of dietary methionine (Met) and 2-hydroxy-4-(methylthio)-butanoate (HMTBA) on the eggshell quality of broiler breeder hens and elucidate the mechanism of Met in improving eggshell quality from the perspectives of eggshell microstructure and shell gland physiological function. A total of 720 WOD188 broiler breeder hens at 40 weeks old were assigned to 3 groups, with 8 replicates per group and 30 birds per replicate. Over 7 weeks, birds were fed a basal diet or the same diet supplemented with 0.15% Met or 0.17% HMTBA. Our findings revealed significant improvements in the Met group for egg shape index, shell thickness, breaking strength, and fracture toughness (P < 0.05), whereas the HMTBA group showed no significant improvements (P > 0.05). Met supplementation increased calcium and phosphorus levels in both serum and shell gland tissue (P < 0.05), and enhanced Ca2+ ATPase activity in shell gland tissue (P < 0.05). Histomorphological changes cluded enhanced mucosal fold dimensions and increased epithelial height in the shell gland (P < 0.05). Met also improved eggshell ultrastructure, resulting in a thicker effective layer and broader mammillae with fewer type B structures (P < 0.05). The mRNA levels for genes regulating eggshell ultrastructure, such as ovocleidin-116 (OC-116), calbindin 1 (CALB1), and integral membrane protein 2C (ITM2C), were significantly upregulated in the Met group (P < 0.05). Transcriptome analysis identified 248 differentially upregulated genes in the Met group, primarily linked to the non-canonical Wnt/Ca2+ signaling pathway, crucial for calcium ion transport and cellular proliferation. This research highlights that Met supplementation improves eggshell quality by enhancing calcium transport and cellular proliferation in uterine function, particularly through the modulation of Wnt family member 11 (WNT11) and CALB1, influencing calcium deposition and ultrastructural development.
Collapse
Affiliation(s)
- Mingkun Gao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Youying Chen
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiaomin Li
- Beijing Huadu Yukou Poultry Industry Co., Ltd., Beijing 101206, China
| | - Dongli Li
- Beijing Huadu Yukou Poultry Industry Co., Ltd., Beijing 101206, China
| | - Aiqiao Liu
- Beijing Huadu Yukou Poultry Industry Co., Ltd., Beijing 101206, China
| | - Lu Gong
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhonghua Ning
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Wei Nie
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zengpeng Lv
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
3
|
Xu T, Zhang K, Hu Y, Yang R, Tang J, Fu W. Comparison of the Therapeutic Efficacy and Autophagy-Mediated Mechanisms of Action of Urine-Derived and Adipose-Derived Stem Cells in Osteoarthritis. Am J Sports Med 2024; 52:3130-3146. [PMID: 39311500 DOI: 10.1177/03635465241277176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent and disabling disease that affects a significant proportion of the global population. Urine-derived stem cells (USCs) have shown great prospects in the treatment of OA, but there is no study that has compared them with traditional stem cells. PURPOSE This study aimed to compare the therapeutic efficacy and mechanisms of USCs and adipose-derived stem cells (ADSCs) for OA treatment. STUDY DESIGN Controlled laboratory study. METHODS We compared the biological properties of USCs and ADSCs using CCK-8, colony formation, EdU, adhesion, and apoptosis assays. We evaluated the protective effects of USCs and ADSCs on IL-1β-treated OA chondrocytes by chemical staining, immunofluorescence, and Western blotting. We assessed the effects of USCs and ADSCs on chondrocyte autophagy by transmission electron microscopy, immunofluorescence, and Western blotting. We also compared the therapeutic efficacy of intra-articular injections of USCs and ADSCs by gross, histological, micro-computed tomography, and immunohistochemical analyses in an OA rat model induced by anterior cruciate ligament transection. RESULTS USCs showed higher proliferation, colony formation, DNA synthesis, adhesion, and anti-apoptotic abilities than ADSCs. Both USCs and ADSCs increased the expression of cartilage-specific proteins and decreased the expression of matrix degradation-related proteins and inflammatory factors in OA chondrocytes. USCs had a greater advantage in suppressing MMP-13 and inflammatory factors than ADSCs. Both USCs and ADSCs enhanced autophagy in OA chondrocytes, with USCs being more effective than ADSCs. The autophagy inhibitor 3-MA reduced the enhanced autophagy and protective effects of USCs and ADSCs on OA chondrocytes. CONCLUSION To our knowledge, this is the first study to explore the efficacy of USCs in the treatment of knee OA and to compare them with ADSCs. Considering the superior properties of USCs in terms of noninvasive acquisition, a high cost-benefit ratio, and low ethical concerns, our study suggests that they may be a more promising therapeutic option than ADSCs for OA treatment under rigorous regulatory pathways. CLINICAL RELEVANCE USCs may be a superior cell source for stem cells to treat knee OA, and this study strengthens the evidence for the application of USCs.
Collapse
Affiliation(s)
- Tianhao Xu
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kaibo Zhang
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunan Hu
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Runze Yang
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiexi Tang
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weili Fu
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Chapman JH, Ghosh D, Attari S, Ude CC, Laurencin CT. Animal Models of Osteoarthritis: Updated Models and Outcome Measures 2016-2023. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:127-146. [PMID: 38983776 PMCID: PMC11233113 DOI: 10.1007/s40883-023-00309-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2024]
Abstract
Purpose Osteoarthritis (OA) is a global musculoskeletal disorder that affects primarily the knee and hip joints without any FDA-approved disease-modifying therapies. Animal models are essential research tools in developing therapies for OA; many animal studies have provided data for the initiation of human clinical trials. Despite this, there is still a need for strategies to recapitulate the human experience using animal models to better develop treatments and understand pathogenesis. Since our last review on animal models of osteoarthritis in 2016, there have been exciting updates in OA research and models. The main purpose of this review is to update the latest animal models and key features of studies in OA research. Method We used our existing classification method and screened articles in PubMed and bibliographic search for animal OA models between 2016 and 2023. Relevant and high-cited articles were chosen for inclusion in this narrative review. Results Recent studies were analyzed and classified. We also identified ex vivo models as an area of ongoing research. Each animal model offers its own benefit in the study of OA and there are a full range of outcome measures that can be assessed. Despite the vast number of models, each has its drawbacks that have limited translating approved therapies for human use. Conclusion Depending on the outcome measures and objective of the study, researchers should pick the best model for their work. There have been several exciting studies since 2016 that have taken advantage of regenerative engineering techniques to develop therapies and better understand OA. Lay Summary Osteoarthritis (OA) is a chronic debilitating disease without any cure that affects mostly the knee and hip joints and often results in surgical joint replacement. Cartilage protects the joint from mechanical forces and degrades with age or in response to injury. The many contributing causes of OA are still being investigated, and animals are used for preclinical research and to test potential new treatments. A single consensus OA animal model for preclinical studies is non-existent. In this article, we review the many animal models for OA and provide a much-needed update on studies and model development since 2016.
Collapse
Affiliation(s)
- James H. Chapman
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Debolina Ghosh
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Seyyedmorteza Attari
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Chinedu C. Ude
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical and Bimolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
5
|
Kennedy O, Kitson A, Okpara C, Chow LW, Gonzalez-Fernandez T. Immunomodulatory Strategies for Cartilage Regeneration in Osteoarthritis. Tissue Eng Part A 2024; 30:259-271. [PMID: 38126327 DOI: 10.1089/ten.tea.2023.0255] [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] [Indexed: 12/23/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent musculoskeletal disorder and a leading cause of disability globally. Although many efforts have been made to treat this condition, current tissue engineering (TE) and regenerative medicine strategies fail to address the inflammatory tissue environment that leads to the rapid progression of the disease and prevents cartilage tissue formation. First, this review addresses in detail the current anti-inflammatory therapies for OA with a special emphasis on pharmacological approaches, gene therapy, and mesenchymal stromal cell (MSC) intra-articular administration, and discusses the reasons behind the limited clinical success of these approaches at enabling cartilage regeneration. Then, we analyze the state-of-the-art TE strategies and how they can be improved by incorporating immunomodulatory capabilities such as the optimization of biomaterial composition, porosity and geometry, and the loading of anti-inflammatory molecules within an engineered structure. Finally, the review discusses the future directions for the new generation of TE strategies for OA treatment, specifically focusing on the spatiotemporal modulation of anti-inflammatory agent presentation to allow for tailored patient-specific therapies. Impact statement Osteoarthritis (OA) is a prevalent and debilitating musculoskeletal disorder affecting millions worldwide. Despite significant advancements in regenerative medicine and tissue engineering (TE), mitigating inflammation while simultaneously promoting cartilage tissue regeneration in OA remains elusive. In this review article, we discuss current anti-inflammatory therapies and explore their potential synergy with cutting-edge cartilage TE strategies, with a special focus on novel spatiotemporal and patient-specific anti-inflammatory strategies.
Collapse
Affiliation(s)
- Orlaith Kennedy
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
- Department of Biomedical Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
| | - Andrew Kitson
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Chiebuka Okpara
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Lesley W Chow
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | | |
Collapse
|
6
|
Liu L, Wang J, Liu L, Shi W, Gao H, Liu L. WITHDRAWN: The dysregulated autophagy in osteoarthritis: Revisiting molecular profile. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024:S0079-6107(24)00034-8. [PMID: 38531488 DOI: 10.1016/j.pbiomolbio.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/21/2024] [Accepted: 03/22/2024] [Indexed: 03/28/2024]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/policies/article-withdrawal.
Collapse
Affiliation(s)
- Liang Liu
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| | - Jie Wang
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| | - Lu Liu
- Department of Internal Medicine, Tianbao Central Health Hospital, Xintai City, Shandong Province, Shandong, Xintai, 271200, China
| | - Wenling Shi
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| | - Huajie Gao
- Operating Room of Qingdao University Affiliated Hospital, Qingdao, Pingdu, 266000, China
| | - Lun Liu
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| |
Collapse
|
7
|
Campbell TM, Trudel G. Protecting the regenerative environment: selecting the optimal delivery vehicle for cartilage repair-a narrative review. Front Bioeng Biotechnol 2024; 12:1283752. [PMID: 38333081 PMCID: PMC10850577 DOI: 10.3389/fbioe.2024.1283752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Focal cartilage defects are common in youth and older adults, cause significant morbidity and constitute a major risk factor for developing osteoarthritis (OA). OA is the most common musculoskeletal (MSK) disease worldwide, resulting in pain, stiffness, loss of function, and is currently irreversible. Research into the optimal regenerative approach and methods in the setting of either focal cartilage defects and/or OA holds to the ideal of resolving both diseases. The two fundamentals required for cartilage regenerative treatment are 1) the biological element contributing to the regeneration (e.g., direct application of stem cells, or of an exogenous secretome), and 2) the vehicle by which the biological element is suspended and delivered. The vehicle provides support to the regenerative process by providing a protective environment, a structure that allows cell adherence and migration, and a source of growth and regenerative factors that can activate and sustain regeneration. Models of cartilage diseases include osteochondral defect (OCD) (which usually involve one focal lesion), or OA (which involves a more diffuse articular cartilage loss). Given the differing nature of these models, the optimal regenerative strategy to treat different cartilage diseases may not be universal. This could potentially impact the translatability of a successful approach in one condition to that of the other. An analogy would be the repair of a pothole (OCD) versus repaving the entire road (OA). In this narrative review, we explore the existing literature evaluating cartilage regeneration approaches for OCD and OA in animal then in human studies and the vehicles used for each of these two conditions. We then highlight strengths and challenges faced by the different approaches presented and discuss what might constitute the optimal cartilage regenerative delivery vehicle for clinical cartilage regeneration.
Collapse
Affiliation(s)
- T. Mark Campbell
- Elisabeth Bruyère Hospital, Ottawa, ON, Canada
- Bone and Joint Research Laboratory, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Guy Trudel
- Bone and Joint Research Laboratory, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The Ottawa Hospital, Department of Medicine, Division of Physical Medicine and Rehabilitation, Ottawa, ON, Canada
| |
Collapse
|
8
|
Guo P, Li H, Wang X, Li X, Li X. PG545 Prevents Osteoarthritis Development by Regulating PI3K/AKT/mTOR Signaling and Activating Chondrocyte Autophagy. Pharmacology 2023; 108:576-588. [PMID: 37820587 DOI: 10.1159/000532078] [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: 04/26/2022] [Accepted: 02/16/2023] [Indexed: 10/13/2023]
Abstract
INTRODUCTION Osteoarthritis (OA) is a degenerative disease common in the elderly and is characterized by joint pain, swelling, and restricted movement. In recent years, heparanase has been reported to play an important role in the development of osteoarthritic cartilage. PG545 is a heparan sulfate mimetic with heparanase inhibitory activity. In this study, the therapeutic effects and possible mechanisms of PG545 were investigated in a chondrocyte injury model induced by interleukin-1β (IL -1β). METHODS Following treatment with PG545 or the autophagy inhibitor 3-methyladenine (3-MA), chondrocyte viability was detected using Cell Counting Kit-8 and fluorescein diacetate/propidium iodide double staining. The apoptosis rate of chondrocytes was determined by flow cytometry. Expression of light chain 3 and P62 was monitored by immunofluorescence labeling. Western blot, lentivirus infection with red fluorescent protein and green fluorescent protein, and quantitative real-time polymerase chain reaction were used to determine the expression levels of chondrocyte markers, apoptosis-related factors, autophagy proteins, and key proteins of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway. The expression and activity of stress-specific enzymes such as malondialdehyde, superoxide dismutase, and catalase (CAT) were investigated. Chondrocytes with ATG5 knockdown were used to investigate the relationship between the therapeutic effect of PG545 and autophagy. The therapeutic effect of PG545 was verified in vivo. RESULTS PG545 had a significant protective effect on chondrocytes by reducing oxidative stress, apoptosis, and degradation of chondrocytes and increasing chondrocyte proliferation. PG545 was effective in inducing autophagy in IL-1β-treated cells, while 3-MA attenuated the effect. The PI3K/Akt/mTOR pathway may be involved in the promotion of autophagy and OA treatment by PG545. CONCLUSION PG545 was able to restore impaired autophagy and autophagic flux via the PI3K/Akt/mTOR pathway, thereby delaying the progression of OA, suggesting that PG545 may be a novel therapeutic approach for OA.
Collapse
Affiliation(s)
- Peiyu Guo
- Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hua Li
- Department of Sport Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuming Wang
- Department of Respiratory Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xingguo Li
- Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xi Li
- Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
9
|
Liu Z, Wang T, Sun X, Nie M. Autophagy and apoptosis: regulatory factors of chondrocyte phenotype transition in osteoarthritis. Hum Cell 2023:10.1007/s13577-023-00926-2. [PMID: 37277675 DOI: 10.1007/s13577-023-00926-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/25/2023] [Indexed: 06/07/2023]
Abstract
Osteoarthritis (OA) is the main pathogenic factor in diseases that cause joint deformities. As the main manifestation of the progress of OA, cartilage degradation has been closely associated with the degeneration of chondrocytes, which is induced by inflammatory factors and other trauma factors. Autophagy and apoptosis are the main mechanisms for cells to maintain homeostasis and play crucial roles in OA. Under the influence of external environmental factors (such as aging and injury), the metabolism of cells can be altered, which may affect the extent of autophagy and apoptosis. With the progression of OA, these changes can alter the cell phenotypes, and the cells of different phenotypes display distinct differences in morphology and function. In this review, we have summarized the alteration in cell metabolism, autophagy, and the extent of apoptosis during OA progression and its effects on the cell phenotypes to provide new ideas for further research on the mechanisms of phenotypic transition and therapeutic strategies so as to reverse the cell phenotypes.
Collapse
Affiliation(s)
- Zhibo Liu
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, People's Republic of China
| | - Ting Wang
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, People's Republic of China
| | - Xianding Sun
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, People's Republic of China.
| | - Mao Nie
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing, People's Republic of China.
| |
Collapse
|
10
|
Zhao S, Liu Y, Wang J, Wen Y, Wu B, Yang D, Wang G, Xiu G, Ling B, Du D, Xu J. ADSCs increase the autophagy of chondrocytes through decreasing miR-7-5p in Osteoarthritis rats by targeting ATG4A. Int Immunopharmacol 2023; 120:110390. [PMID: 37262955 DOI: 10.1016/j.intimp.2023.110390] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 05/15/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is a highly degenerative joint disease, mainly companying with progressive destruction of articular cartilage. Adipose-derived stromal cells (ADSCs) therapy enhances articular cartilage repair, extracellular matrix (ECM) synthesis and attenuates joints inflammation, but specific mechanisms of therapeutic benefit remain poorly understood. This study aimed to clarify the therapeutic effects and mechanisms of ADSCs on cartilage damage in the keen joint of OA rat model. METHODS Destabilization of the medial meniscus (DMM) and anterior cruciate ligament transection (ACLT) surgery-induced OA rats were treated with allogeneic ADSCs by intra-articular injections for 6 weeks. The protective effect of ADSCs in vivo was measured using Safranin O and fast green staining, immunofluorescence and western blot analysis. Meanwhile, the miRNA-7-5p (miR-7-5p) expression was assessed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The mechanism of increased autophagy with ADSCs addition through decreasing miR-7-5p was revealed using oligonucleotides, and adenovirus in rat chondrocytes. The luciferase reporter assay revealed the molecular role of miR-7-5p and autophagy related 4A (ATG4A). The substrate of mTORC1 pathway: (p-)p70S6 and (p-)S6 in OA models with ADSCs addition were detected by western blotting. RESULTS The ADSCs treatment repaired the articular cartilage and maintained chondrocytes ECM homeostasis through modulating chondrocytes autophagy in the OA model, indicators of the change of autophagic proteins expression and autophagic flux. Meanwhile, the increased autophagy induced by ADSCs treatment was closely related to the decreased expression of host-derived miR-7-5p, a negative modulator of OA progression. Functional genomics (overexpression of genes) in vitro studies demonstrate the inhibition of host-derived miR-7-5p in mediating the benefit of ADSCs administration in OA model. Then ATG4A was defined as a target gene of miR-7-5p, and the negative relation between miR-7-5p and ATG4A was investigated in the OA model treated with ADSCs. Furthermore, miR-7-5p mediated chondrocyte autophagy by targeting ATG4A in the OA model treated with ADSCs was confirmed with the rescue trial of ATG4A/miR-7-5p overexpression on rat chondrocyte. Finally, the mTORC1 signaling pathways mediated by host-derived miR-7-5p with ADSCs treatment were decreased in OA rats. CONCLUSIONS ADSCs promote the chondrocytes autophagy by decreasing miR-7-5p in articular cartilage by targeting ATG4A and a potential role for ADSCs based therapeutics for preventing of articular cartilage destruction and extracellular matrix (ECM) degradation in OA.
Collapse
Affiliation(s)
- Shu Zhao
- East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu'e Liu
- East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Wang
- East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Wen
- East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Baitong Wu
- East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Danjing Yang
- East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guangming Wang
- East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guanghui Xiu
- Department of Intensive Care Unit, Affiliated Hospital of Yunnan University (The Second People's Hospital of Yunnan Province),Yunnan University, Kunming, China
| | - Bin Ling
- Department of Intensive Care Unit, Affiliated Hospital of Yunnan University (The Second People's Hospital of Yunnan Province),Yunnan University, Kunming, China
| | - Dajiang Du
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Jun Xu
- East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
11
|
Chae DS, Han S, Lee MK, Kim SW. BMP-2 Genome-Edited Human MSCs Protect against Cartilage Degeneration via Suppression of IL-34 in Collagen-Induced Arthritis. Int J Mol Sci 2023; 24:ijms24098223. [PMID: 37175932 PMCID: PMC10179718 DOI: 10.3390/ijms24098223] [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: 04/07/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Even though the regenerative potential of mesenchymal stem cells (MSCs) has been extensively studied, there is a debate regarding their minimal therapeutic properties. Bone morphogenetic proteins (BMP) are involved in cartilage metabolism, chondrogenesis, and bone healing. In this study, we aimed to analyze the role of genome-edited BMP-2 overexpressing amniotic mesenchymal stem cells (AMMs) in a mouse model of collagen-induced arthritis (CIA). The BMP-2 gene was synthesized and inserted into AMMs using transcription activator-like effector nucleases (TALENs), and BMP-2-overexpressing AMMs (AMM/B) were sorted and characterized using quantitative reverse transcription polymerase chain reaction (qRT-PCR). The co-culture of AMM/B with tumor necrosis factor (TNF)-α-treated synovial fibroblasts significantly decreased the levels of interleukin (IL)-34. The therapeutic properties of AMM/B were evaluated using the CIA mouse model. The injection of AMM/B attenuated CIA progression and inhibited T helper (Th)17 cell activation in CIA mice. In addition, the AMM/B injection increased proteoglycan expression in cartilage and decreased the infiltration of inflammatory cells and factors, including IL-1β, TNF-α, cyclooxygenase (COX)-2, and Nuclear factor kappa B (NF-kB) in the joint tissues. Therefore, editing the BMP-2 genome in MSCs might be an alternative strategy to enhance their therapeutic potential for treating cartilage degeneration in arthritic joints.
Collapse
Affiliation(s)
- Dong-Sik Chae
- Department of Orthopedic Surgery, Catholic Kwandong University College of Medicine, International St. Mary's Hospital, Incheon 22711, Republic of Korea
| | - Seongho Han
- Department of Family Medicine, Dong-A University College of Medicine, Dong-A University Medical Center, Busan 49201, Republic of Korea
| | - Min-Kyung Lee
- Department of Dental Hygiene, Dong-Eui University, Busan 47340, Republic of Korea
| | - Sung-Whan Kim
- Department Medicine, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea
| |
Collapse
|
12
|
Gong Y, Li S, Wu J, Zhang T, Fang S, Feng D, Luo X, Yuan J, Wu Y, Yan X, Zhang Y, Zhu J, Wu J, Lian J, Xiang W, Ni Z. Autophagy in the pathogenesis and therapeutic potential of post-traumatic osteoarthritis. BURNS & TRAUMA 2023; 11:tkac060. [PMID: 36733467 PMCID: PMC9887948 DOI: 10.1093/burnst/tkac060] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/29/2022] [Indexed: 02/04/2023]
Abstract
Autophagy, as a fundamental mechanism for cellular homeostasis, is generally involved in the occurrence and progression of various diseases. Osteoarthritis (OA) is the most common musculoskeletal disease that often leads to pain, disability and economic loss in patients. Post-traumatic OA (PTOA) is a subtype of OA, accounting for >12% of the overall burden of OA. PTOA is often caused by joint injuries including anterior cruciate ligament rupture, meniscus tear and intra-articular fracture. Although a variety of methods have been developed to treat acute joint injury, the current measures have limited success in effectively reducing the incidence and delaying the progression of PTOA. Therefore, the pathogenesis and intervention strategy of PTOA need further study. In the past decade, the roles and mechanisms of autophagy in PTOA have aroused great interest in the field. It was revealed that autophagy could maintain the homeostasis of chondrocytes, reduce joint inflammatory level, prevent chondrocyte death and matrix degradation, which accordingly improved joint symptoms and delayed the progression of PTOA. Moreover, many strategies that target PTOA have been revealed to promote autophagy. In this review, we summarize the roles and mechanisms of autophagy in PTOA and the current strategies for PTOA treatment that depend on autophagy regulation, which may be beneficial for PTOA patients in the future.
Collapse
Affiliation(s)
| | | | | | - Tongyi Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Changjiang Street, Yuzhong District, Chongqing 400042, China,Department of General practice, Chinese PLA General Hospital of the Central Theater Command, Wuluo Street, Wuchang District, Wuhan 430000, China
| | - Shunzheng Fang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Changjiang Street, Yuzhong District, Chongqing 400042, China
| | - Daibo Feng
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Changjiang Street, Yuzhong District, Chongqing 400042, China
| | - Xiaoqing Luo
- Department of Wound Repair and Rehabilitation Medicine, 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, Changjiang Street, Yuzhong District, Chongqing 400042, China
| | - Jing Yuan
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing 400038, China
| | - Yaran Wu
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing 400038, China
| | - Xiaojing Yan
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing 400038, China
| | - Yan Zhang
- Department of Pediatrics, People's Hospital Affiliated to Chongqing Three Gorges Medical College, Guoben Street, Wanzhou district, Chongqing 404000, China
| | - Jun Zhu
- Department of Cardiology, Shanghai Hospital, Shanghai Street, Wanzhou District, Chongqing 404000, China
| | - Jiangyi Wu
- Department of Sports Medicine and Rehabilitation, Shenzhen Hospital, Peking University, Lianhua Street, Futian District, Shenzhen 518034, China
| | - Jiqin Lian
- Correspondence. Zhenghong Ni, ; Wei Xiang, ; Jiqin Lian,
| | - Wei Xiang
- Correspondence. Zhenghong Ni, ; Wei Xiang, ; Jiqin Lian,
| | - Zhenhong Ni
- Correspondence. Zhenghong Ni, ; Wei Xiang, ; Jiqin Lian,
| |
Collapse
|
13
|
Perucca Orfei C, Boffa A, Sourugeon Y, Laver L, Magalon J, Sánchez M, Tischer T, Filardo G, de Girolamo L. Cell-based therapies have disease-modifying effects on osteoarthritis in animal models. A systematic review by the ESSKA Orthobiologic Initiative. Part 1: adipose tissue-derived cell-based injectable therapies. Knee Surg Sports Traumatol Arthrosc 2023; 31:641-655. [PMID: 36104484 PMCID: PMC9898370 DOI: 10.1007/s00167-022-07063-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE The aim of this systematic review was to determine if adipose tissue-derived cell-based injectable therapies can induce disease-modifying effects in joints affected by osteoarthritis (OA). METHODS A systematic review was performed on three electronic databases (PubMed, Web of Science, Embase) according to PRISMA guidelines. A synthesis of the results was performed investigating disease-modifying effects in preclinical studies comparing injectable adipose-derived products with OA controls or other products, different formulations or injection intervals, and the combination with other products. The risk of bias was assessed according to the SYRCLE's tool. RESULTS Seventy-one studies were included (2,086 animals) with an increasing publication trend over time. Expanded cells were used in 65 studies, 3 studies applied point of care products, and 3 studies investigated both approaches. Overall, 48 out of 51 studies (94%) reported better results with adipose-derived products compared to OA controls, with positive findings in 17 out of 20 studies (85%) in macroscopic, in 37 out of 40 studies (93%) in histological, and in 22 out of 23 studies (96%) in immunohistochemical evaluations. Clinical and biomarker evaluations showed positive results in 14 studies out of 18 (78%) and 12 studies out of 14 (86%), while only 9 studies out of 17 (53%) of the imaging evaluations were able to detect differences versus controls. The risk of bias was low in 38% of items, unclear in 51%, and high in (11%). CONCLUSION The current preclinical models document consistent evidence of disease-modifying effects of adipose-derived cell-based therapies for the treatment of OA. The high heterogeneity of the published studies highlights the need for further targeted research to provide recommendations on the optimal methodologies for a more effective application of these injective therapies for the treatment of OA in clinical practice. LEVEL OF EVIDENCE II.
Collapse
Affiliation(s)
- Carlotta Perucca Orfei
- grid.417776.4IRCCS Istituto Ortopedico Galeazzi, Laboratorio di Biotecnologie Applicate all’Ortopedia, Milan, Italy
| | - Angelo Boffa
- Applied and Translational Research Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - Yosef Sourugeon
- grid.413731.30000 0000 9950 8111Rambam Health Care Campus, Haifa, Israel
| | - Lior Laver
- grid.414084.d0000 0004 0470 6828Department of Orthopaedics, Hillel Yaffe Medical Center (HYMC), Hadera, Israel ,Arthrosport Clinic, Tel-Aviv, Israel ,grid.6451.60000000121102151Technion University Hospital (Israel Institute of Technology) - Rappaport Faculty of Medicine, Haifa, Israel
| | - Jérémy Magalon
- grid.414336.70000 0001 0407 1584Cell Therapy Laboratory, Hôpital De La Conception, AP-HM, Marseille, France ,grid.5399.60000 0001 2176 4817INSERM, NRA, C2VN, Aix Marseille Univ, Marseille, France ,SAS Remedex, Marseille, France
| | - Mikel Sánchez
- grid.473696.9Arthroscopic Surgery Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, Spain ,Advanced Biological Therapy Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, Spain
| | - Thomas Tischer
- grid.10493.3f0000000121858338Department of Orthopaedic Surgery, University of Rostock, Rostock, Germany
| | - Giuseppe Filardo
- grid.419038.70000 0001 2154 6641Applied and Translational Research Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy ,grid.469433.f0000 0004 0514 7845Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland ,grid.29078.340000 0001 2203 2861Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Laura de Girolamo
- grid.417776.4IRCCS Istituto Ortopedico Galeazzi, Laboratorio di Biotecnologie Applicate all’Ortopedia, Milan, Italy
| |
Collapse
|
14
|
Juskovic A, Nikolic M, Ljujic B, Matic A, Zivkovic V, Vucicevic K, Milosavljevic Z, Vojinovic R, Jovicic N, Zivanovic S, Milivojevic N, Jakovljevic V, Bolevich S, Miletic Kovacevic M. Effects of Combined Allogenic Adipose Stem Cells and Hyperbaric Oxygenation Treatment on Pathogenesis of Osteoarthritis in Knee Joint Induced by Monoiodoacetate. Int J Mol Sci 2022; 23:ijms23147695. [PMID: 35887046 PMCID: PMC9317268 DOI: 10.3390/ijms23147695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/04/2022] [Accepted: 07/09/2022] [Indexed: 01/07/2023] Open
Abstract
The beneficial effects of HBO in inflammatory processes make it an attractive type of treatment for chronic arthritis. In addition, the effects of combination therapy based on adipose stem cells and HBO on OA progression have not been fully investigated. The current study explored the efficacy of intra-articular injection of allogeneic adipose-derived mesenchymal stem cells (ADMSCs) combined with hyperbaric oxygenation treatment (HBO) in a rat osteoarthritis (OA) model. The rat OA model was induced by intra-articular injection of monoiodoacetate (MIA) and 7 days after application of MIA rats were divided into five groups: healthy control (CTRL), osteoarthritis (OA), ADMSCs (ADS), the HBO+ADS21day and HBO+ADS28day groups. A single dose of 1 × 106 allogeneic ADMSCs suspended in sterile saline was injected into the knee joint alone or in combination with HBO treatment. Rats were sacrificed at 3 or 4 weeks after MIA injection. Treatment outcomes were evaluated by radiographic, morphological and histological analysis and by specific staining of articular cartilage. We also measured the level of inflammatory and pro/antioxidative markers. We confirmed that combined treatment of ADMSCs and HBO significantly improved the regeneration of cartilage in the knee joint. Rtg score of knee joint damage was significantly decreased in the HBO+ADS21day and HBO+ADS28day groups compared to the OA. However, the positive effect in the HBO+ADS28day group was greater than the HBO+ADS21day group. The articular cartilage was relatively normal in the HBO+ADS28day group, but moderate degeneration was observed in the HBO+ADS21day compared to the OA group. These findings are in line with the histopathological results. A significantly lower level of O2−. was observed in the HBO+ADS28day group but a higher NO level compared to the HBO+ADS21day group. Moreover, in the HBO+ADS28day group significantly higher concentrations of IL-10 were observed but there was no significant difference in proinflammatory cytokine in serum samples. These results indicate that a single intra-articular injection of allogeneic ADMSCs combined with HBO efficiently attenuated OA progression after 28 days with greater therapeutic effect compared to alone ADMSCs or after 3 weeks of combined treatment. Combined treatment might be an effective treatment for OA in humans.
Collapse
Affiliation(s)
- Aleksandar Juskovic
- Department of Orthopaedic Surgery, Clinical Centre of Montenegro, 81110 Podgorica, Montenegro;
| | - Marina Nikolic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.N.); (V.Z.); (V.J.)
| | - Biljana Ljujic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Correspondence: ; Tel.: +381-343-06800
| | - Aleksandar Matic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
- University Clinical Center, 34000 Kragujevac, Serbia;
| | - Vladimir Zivkovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.N.); (V.Z.); (V.J.)
- Department of Pharmacology of the Institute of Biodesign and Complex System Modelling, First Moscow State Medical University I.M. Sechenov, 119991 Moscow, Russia
| | - Ksenija Vucicevic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Zoran Milosavljevic
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.M.); (N.J.); (M.M.K.)
| | - Radisa Vojinovic
- University Clinical Center, 34000 Kragujevac, Serbia;
- Department of Radiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Nemanja Jovicic
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.M.); (N.J.); (M.M.K.)
| | - Suzana Zivanovic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Nevena Milivojevic
- Laboratory for Bioengineering, Institute of Information Technologies Kragujevac, Department of Natural and Mathematical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.N.); (V.Z.); (V.J.)
- Department of Human Pathology, First Moscow State Medical University I.M. Sechenov, 119991 Moscow, Russia;
| | - Sergey Bolevich
- Department of Human Pathology, First Moscow State Medical University I.M. Sechenov, 119991 Moscow, Russia;
| | - Marina Miletic Kovacevic
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.M.); (N.J.); (M.M.K.)
| |
Collapse
|
15
|
Liu X, Liu Y, He H, Xiang W, He C. Human adipose and synovial mesenchymal stem cells improve osteoarthritis in rats by reducing chondrocyte reactive oxygen species and inhibiting inflammatory response. J Clin Lab Anal 2022; 36:e24353. [PMID: 35312120 PMCID: PMC9102617 DOI: 10.1002/jcla.24353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 12/01/2022] Open
Abstract
Background We explored the therapeutic effects of Adipose‐derived mesenchymal stem cells (ADMSCs) and Synovial‐derived mesenchymal stem cells (SDMSCs) on osteoarthritis (OA). Methods SDMSCs and ADMSCs were co‐cultured with chondrocytes and stimulated with interleukin (IL)‐1β. An OA model was established on rats by intra‐articular injection with ADMSCs and SDMSCs. After 8 weeks, the joint diameter difference was detected, and histological staining was used to observe the pathological changes in cartilage tissue. Enzyme‐linked immunosorbent assay (ELISA) was used to detect the expressions of IL‐6, tumor necrosis factor (TNF)‐α and IL‐1β in joint fluid. The expressions of COL2A1, Aggrecan, Matrix metalloproteinase (MMP)‐13, SOX9, IL‐6, TNF‐α and IL‐1β were detected by qRT‐PCR and Western blotting in cartilage tissue. Reactive oxygen species (ROS) content in cells and cartilage tissues was detected by ROS kit. Results SDMSCs and ADMSCs co‐cultured with chondrocytes could reduce MMP‐13 expression, increase the expressions of COL2A1, Aggrecan and SOX9, as well as reverse the effects of IL‐1β on promoting ROS content and inflammatory factors levels. After the OA model was established, the injection of ADMSCs and SDMSCs reduced the differences in joint diameter and tissue lesions in OA rats. The OA model led to increased levels of IL‐6, TNF‐α and IL‐1β in joint fluid and cartilage tissue, while the injection of ADMSCs and SDMSCs inhibited the inflammatory factor levels in OA rats, and increased the expressions of COL2A1, Aggrecan and SOX9 in OA rats. Conclusion ADMSCs and SDMSCs improve osteoarthritis in rats by reducing chondrocyte ROS and inhibiting inflammatory response.
Collapse
Affiliation(s)
- Xunzhi Liu
- Orthopedics Department First Affiliated Hospital of Gannan Medical University Ganzhou City China
| | - Yaqing Liu
- Pediatric Department First Affiliated Hospital of Gannan Medical University Ganzhou City China
| | - Huabin He
- Orthopedics Department First Affiliated Hospital of Gannan Medical University Ganzhou City China
| | - Weiwei Xiang
- Orthopedics Department First Affiliated Hospital of Gannan Medical University Ganzhou City China
| | - Cheng He
- Orthopedics Department First Affiliated Hospital of Gannan Medical University Ganzhou City China
| |
Collapse
|
16
|
Wang G, Xing D, Liu W, Zhu Y, Liu H, Yan L, Fan K, Liu P, Yu B, Li JJ, Wang B. Preclinical studies and clinical trials on mesenchymal stem cell therapy for knee osteoarthritis: A systematic review on models and cell doses. Int J Rheum Dis 2022; 25:532-562. [PMID: 35244339 DOI: 10.1111/1756-185x.14306] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/30/2022] [Accepted: 02/07/2022] [Indexed: 12/15/2022]
Abstract
AIM To provide a systematic analysis of the study design in knee osteoarthritis (OA) preclinical studies, focusing on the characteristics of animal models and cell doses, and to compare these to the characteristics of clinical trials using mesenchymal stem cells (MSCs) for the treatment of knee OA. METHOD A systematic and comprehensive search was conducted using the PubMed, Web of Science, Ovid, and Embase electronic databases for research papers published in 2009-2020 on testing MSC treatment in OA animal models. The PubMed database and ClinicalTrials.gov website were used to search for published studies reporting clinical trials of MSC therapy for knee OA. RESULTS In total, 9234 articles and two additional records were retrieved, of which 120 studies comprising preclinical and clinical studies were included for analysis. Among the preclinical studies, rats were the most commonly used species for modeling knee OA, and anterior cruciate ligament transection was the most commonly used method for inducing OA. There was a correlation between the cell dose and body weight of the animal. In clinical trials, there was large variation in the dose of MSCs used to treat knee OA, ranging from 1 × 106 to 200 × 106 cells with an average of 37.91 × 106 cells. CONCLUSION Mesenchymal stem cells have shown great potential in improving pain relief and tissue protection in both preclinical and clinical studies of knee OA. Further high-quality preclinical and clinical studies are needed to explore the dose effectiveness relationship of MSC therapy and to translate the findings from preclinical studies to humans.
Collapse
Affiliation(s)
- Guishan Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China.,Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Beijing, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, China
| | - Yuanyuan Zhu
- Department of Pharmacy, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Haifeng Liu
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Lei Yan
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Kenan Fan
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Peidong Liu
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Baofeng Yu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Jiao Jiao Li
- Faculty of Engineering and IT, School of Biomedical Engineering, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Bin Wang
- Department of Orthopedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China.,Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Carpintero-Fernández P, Varela-Eirín M, García-Yuste A, López-Díaz I, Caeiro JR, Mayán MD. Osteoarthritis: Mechanistic Insights, Senescence, and Novel Therapeutic Opportunities. Bioelectricity 2022; 4:39-47. [PMID: 39355566 PMCID: PMC11441363 DOI: 10.1089/bioe.2021.0039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disease. In the last years, the research community has focused on understanding the molecular mechanisms that led to the pathogenesis of the disease, trying to identify different molecular and clinical phenotypes along with the discovery of new therapeutic opportunities. Different types of cell-to-cell communication mechanisms have been proposed to contribute to OA progression, including mechanisms mediated by connexin43 (Cx43) channels or by small extracellular vesicles. Furthermore, changes in the chondrocyte phenotype such as cellular senescence have been proposed as new contributors of the OA progression, changing the paradigm of the disease. The use of different drugs able to restore chondrocyte phenotype, to reduce cellular senescence and senescence-associated secretory phenotype components, and to modulate ion channel activity or Cx43 appears to be promising therapeutic strategies for the different types of OA. In this review, we aim to summarize the current knowledge in OA phenotypes related with aging and tissue damage and the new therapeutic opportunities currently available.
Collapse
Affiliation(s)
- Paula Carpintero-Fernández
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - Marta Varela-Eirín
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Alejandro García-Yuste
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - Iñaki López-Díaz
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - José Ramón Caeiro
- Department of Orthopaedic Surgery and Traumatology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - María D Mayán
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| |
Collapse
|
18
|
All-Trans Retinoic Acid-Preconditioned Mesenchymal Stem Cells Improve Motor Function and Alleviate Tissue Damage After Spinal Cord Injury by Inhibition of HMGB1/NF-κB/NLRP3 Pathway Through Autophagy Activation. J Mol Neurosci 2022; 72:947-962. [PMID: 35147911 DOI: 10.1007/s12031-022-01977-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/20/2022] [Indexed: 12/19/2022]
Abstract
Spinal cord injury (SCI) is a significant public health issue that imposes numerous burdens on patients and society. Uncontrolled excessive inflammation in the second pathological phase of SCI can aggravate the injury. In this paper, we hypothesized that suppressing inflammatory pathways via autophagy could aid functional recovery, and prevent spinal cord tissue degeneration following SCI. To this end, we examined the effects of intrathecal injection of all-trans retinoic acid (ATRA)-preconditioned bone marrow mesenchymal stem cells (BM-MSCs) (ATRA-MSCs) on autophagy activity and the HMGB1/NF-κB/NLRP3 inflammatory pathway in an SCI rat model. This study demonstrated that SCI increased the expression of Beclin-1 (an autophagy-related gene) and NLRP3 inflammasome components such as NLRP3, ASC, Caspase-1, and pro-inflammatory cytokines IL-1β, IL-18, IL-6, and TNF-α. Additionally, following SCI, the protein levels of key autophagy factors (Beclin-1 and LC3-II) and HMGB1/NF-κB/NLRP3 pathway factors (HMGB1, p-NF-κB, NLRP3, IL-1β, and TNF-α) increased. Our findings indicated that ATRA-MSCs enhanced Beclin-1 and LC3-II levels, regulated the HMGB1/NF-κB/NLRP3 pathway, and inhibited pro-inflammatory cytokines. These factors improved hind limb motor activity and aided in the survival of neurons. Furthermore, ATRA-MSCs demonstrated greater beneficial effects than MSCs in treating spinal cord injury. Overall, ATRA-MSC treatment revealed beneficial effects on the damaged spinal cord by suppressing excessive inflammation and activating autophagy. Further research and investigation of the pathways involved in SCI and the use of amplified stem cells may be beneficial for future clinical use.
Collapse
|
19
|
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: 76] [Impact Index Per Article: 25.3] [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
|
20
|
Kim J. Dysregulated circular RNAs and their pathological implications in knee osteoarthritis: potential novel therapeutic targets and diagnostic biomarkers. ALL LIFE 2022. [DOI: 10.1080/26895293.2021.2020172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Jaehee Kim
- Department of Alternative Medicine, Graduate School of Alternative Medicine, Kyonggi University (Seoul Campus), Seoul, Republic of Korea
| |
Collapse
|
21
|
Cao J, Ding H, Shang J, Ma L, Wang Q, Feng S. Weighted gene co-expression network analysis reveals specific modules and hub genes related to immune infiltration of osteoarthritis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1525. [PMID: 34790731 PMCID: PMC8576690 DOI: 10.21037/atm-21-4566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/19/2021] [Indexed: 12/26/2022]
Abstract
Background The incidence of osteoarthritis (OA), a chronic degenerative disease, is increasing every year. There is no effective clinical treatment for OA and the pathological mechanism remains unclear. Early diagnosis is an effective strategy to control the progress of OA. In this study, we aimed to identify potential early diagnostic biomarkers. Methods We downloaded the gene expression profile dataset, GSE51588 and GSE55235, from the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO) public database. The differentially expressed genes (DEGs) were screened out using the “limma” R package. Weighted gene co-expression network analysis (WGCNA) was utilized to build the co-expression network between the normal and OA samples. A Venn diagram was constructed to detect the hub genes. Potential molecular mechanisms and signaling pathways were enriched by gene set variation analysis (GSVA). Single sample gene set enrichment analysis (ssGSEA) was used to identify the immune infiltration of OA. Results We screened out three hub genes based on WGCNA and DEGs in this study. GSVA results showed that nuclear factor interleukin-3 (NFIL3) was related to tumor necrosis factor alpha (TNF-α) signaling via nuclear factor kappa-B (NF-κB), the reactive oxygen species pathway, and myelocytomatosis (MYC) targets v2. Highly-expressed ADM (adrenomedullin) pathways included TNF-α signaling via NF-κB, the reactive oxygen species pathway, and ultraviolet (UV) response up. OGN (osteoglycin)-enriched pathways included epithelial mesenchymal transition, coagulation, and peroxisome. Conclusions We identified three hub genes (NFIL3, ADM, and OGN) that were correlated to the development and progression of OA, which may provide new biomarkers for early diagnosis.
Collapse
Affiliation(s)
- Jiangang Cao
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Han Ding
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Shang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Lei Ma
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Qi Wang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
22
|
Gu C, Feng J, Waqas A, Deng Y, Zhang Y, Chen W, Long J, Huang S, Chen L. Technological Advances of 3D Scaffold-Based Stem Cell/Exosome Therapy in Tissues and Organs. Front Cell Dev Biol 2021; 9:709204. [PMID: 34568322 PMCID: PMC8458970 DOI: 10.3389/fcell.2021.709204] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Recently, biomaterial scaffolds have been widely applied in the field of tissue engineering and regenerative medicine. Due to different production methods, unique types of three-dimensional (3D) scaffolds can be fabricated to meet the structural characteristics of tissues and organs, and provide suitable 3D microenvironments. The therapeutic effects of stem cell (SC) therapy in tissues and organs are considerable and have attracted the attention of academic researchers worldwide. However, due to the limitations and challenges of SC therapy, exosome therapy can be used for basic research and clinical translation. The review briefly introduces the materials (nature or polymer), shapes (hydrogels, particles and porous solids) and fabrication methods (crosslinking or bioprinting) of 3D scaffolds, and describes the recent progress in SC/exosome therapy with 3D scaffolds over the past 5 years (2016-2020). Normal SC/exosome therapy can improve the structure and function of diseased and damaged tissues and organs. In addition, 3D scaffold-based SC/exosome therapy can significantly improve the structure and function cardiac and neural tissues for the treatment of various refractory diseases. Besides, exosome therapy has the same therapeutic effects as SC therapy but without the disadvantages. Hence, 3D scaffold therapy provides an alternative strategy for treatment of refractory and incurable diseases and has entered a transformation period from basic research into clinical translation as a viable therapeutic option in the future.
Collapse
Affiliation(s)
- Chenyang Gu
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jia Feng
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- School of Medicine, Southeast University, Nanjing, China
| | - Ahmed Waqas
- School of Medicine, Southeast University, Nanjing, China
| | - Yushu Deng
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yifan Zhang
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wanghao Chen
- Department of Neurosurgery, Ninth People Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Long
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Shiying Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lukui Chen
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
23
|
Effect of Warm Acupuncture Combined with Bone Marrow Mesenchymal Stem Cells Transplantation on Cartilage Tissue in Rabbit Knee Osteoarthritis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5523726. [PMID: 34422071 PMCID: PMC8373500 DOI: 10.1155/2021/5523726] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 07/11/2021] [Accepted: 08/02/2021] [Indexed: 12/16/2022]
Abstract
The current study was designed to investigate the effect and underlying mechanism of warm acupuncture combined with bone marrow mesenchymal stem cells (BMSC) transplantation on cartilage tissue injury in rabbit knee osteoarthritis (KOA). In the study, 50 rabbits were randomly divided into 5 groups: blank group, KOA group, warm acupuncture group, BMSCs group, and warm acupuncture combined with BMSCs group. After warm acupuncture combined with BMSCs, the Modified Lequesne MG knee joint assessment scale was used to evaluate the degree of knee joint behavior, the Taiping Peng method generally observed the histomorphology changes of KOA rabbit cartilage, and hematoxylin-eosin staining, safranin O green staining, and toluidine blue staining were conducted to evaluate the extent of cartilage tissue pathology. Furthermore, transmission electron microscopy and TUNEL staining were used to observe cell apoptosis, and immunohistochemistry and qPCR analysis were used to detect the expression of apoptosis-related proteins and mRNA. Results showed that administration of warm acupuncture combined with BMSCs recovered the joint function and significantly decreased Lequesne MG score. The degree of cartilage tissue pathological damage has been improved, cartilage ultrastructure degeneration has recovered, peripheral blood vessels have mild edema, blood supply has gradually recovered, and even small amounts of red blood cells have appeared. In addition, warm acupuncture combined with BMSCs treatment suppressed chondrocyte apoptosis in rabbits with knee osteoarthritis by reduced TUNEL-positive chondrocytes and simultaneously reversed the mRNA expression of Bax, Bcl-2, and Caspase-3. These results indicate that warm acupuncture combined with BMSCs transplantation has a potential protective effect on rabbit KOA, which may be mediated by inhibiting chondrocyte apoptosis.
Collapse
|
24
|
Zhang C, Tian R, Dreifus EM, Hashemi Shahraki A, Holt G, Cai R, Griswold A, Bejarano P, Jackson R, V Schally A, Mirsaeidi M. Activity of the growth hormone-releasing hormone antagonist MIA602 and its underlying mechanisms of action in sarcoidosis-like granuloma. Clin Transl Immunology 2021; 10:e1310. [PMID: 34257968 PMCID: PMC8256670 DOI: 10.1002/cti2.1310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/09/2021] [Accepted: 06/15/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES Growth hormone-releasing hormone (GHRH) is a potent stimulator of growth hormone (GH) secretion from the pituitary gland. Although GHRH is essential for the growth of immune cells, the regulatory effects of its antagonist in granulomatous disease remain unknown. METHODS Here, we report expression of GHRH receptor (R) in human tissue with sarcoidosis granuloma and demonstrate the anti-inflammatory effects of MIA602 (a GHRH antagonist) in two in vitro human granuloma models and an in vivo granuloma model using different methods including ELISA, immunohistochemistry, RNA-seq analysis and flow cytometry. RESULTS MIA602 decreases the levels of IL-2, IL-2R, IL-7, IL-12, IL-17A and TNF-α in an in vitro granuloma model. Further, we show that the anti-inflammatory effect of MIA602 appears to be mediated by a reduction in CD45+CD68+ cells in granulomatous tissue and upregulation in PD-1 expression in macrophages. Analysis of the expression of proteins involved in the mitochondrial stage of apoptosis showed that MIA602 increases the levels of caspase-3, BCL-xL/BAK dimer and MCl-1/Bak dimer in the granuloma. These findings indicate that MIA602 may not induce apoptosis. CONCLUSIONS Our findings further suggest that GHRH-R is potentially a clinical target for the treatment of granulomatous disease and that MIA602 may be used as a novel therapeutic agent for sarcoidosis.
Collapse
Affiliation(s)
- Chongxu Zhang
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
| | - Runxia Tian
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
| | | | | | - Gregory Holt
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
- Division of Pulmonary and Critical CareUniversity of MiamiMiamiFLUSA
| | - Renzhi Cai
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
| | - Anthony Griswold
- School of MedicineJohn P. Hussman Institute for Human GenomicsUniversity of MiamiMiamiFLUSA
| | | | - Robert Jackson
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
- School of MedicineUniversity of MiamiMiamiFLUSA
| | - Andrew V Schally
- Polypeptide and Cancer InstituteVeterans Affairs Medical CenterMiamiFLUSA
- Department of PathologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Mehdi Mirsaeidi
- Section of PulmonaryMiami VA Healthcare SystemMiamiFLUSA
- Division of Pulmonary and Critical CareUniversity of MiamiMiamiFLUSA
| |
Collapse
|
25
|
Theeuwes WF, van den Bosch MHJ, Thurlings RM, Blom AB, van Lent PLEM. The role of inflammation in mesenchymal stromal cell therapy in osteoarthritis, perspectives for post-traumatic osteoarthritis: a review. Rheumatology (Oxford) 2021; 60:1042-1053. [PMID: 33410465 DOI: 10.1093/rheumatology/keaa910] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/26/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
OA is a complex and highly prevalent degenerative disease affecting the whole joint, in which factors like genetic predisposition, gender, age, obesity and traumas contribute to joint destruction. ∼50-80% of OA patients develop synovitis. OA-associated risk factors contribute to joint instability and the release of cartilage matrix fragments, activating the synovium to release pro-inflammatory factors and catabolic enzymes in turn damaging the cartilage and creating a vicious circle. Currently, no cure is available for OA. Mesenchymal stromal cells (MSCs) have been tested in OA for their chondrogenic and anti-inflammatory properties. Interestingly, MSCs are most effective when administered during synovitis. This review focusses on the interplay between joint inflammation and the immunomodulation by MSCs in OA. We discuss the potential of MSCs to break the vicious circle of inflammation and describe current perspectives and challenges for clinical application of MSCs in treatment and prevention of OA, focussing on preventing post-traumatic OA.
Collapse
Affiliation(s)
- Wessel F Theeuwes
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Rogier M Thurlings
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Arjen B Blom
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
26
|
Bryk M, Karnas E, Mlost J, Zuba-Surma E, Starowicz K. Mesenchymal stem cells and extracellular vesicles for the treatment of pain: Current status and perspectives. Br J Pharmacol 2021; 179:4281-4299. [PMID: 34028798 DOI: 10.1111/bph.15569] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/26/2021] [Accepted: 05/05/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are multipotent progenitor cells of mesodermal origin. Due to their capacity for self-renewal and differentiation into several cell types, MSCs have been extensively studied in experimental biology and regenerative medicine in recent years. Moreover, MSCs release extracellular vesicles (EVs), which might be partly responsible for their regenerative properties. MSCs regulate several processes in target cells via paracrine signalling, such as immunomodulation, anti-apoptotic signalling, tissue remodelling, angiogenesis and anti-fibrotic signalling. The aim of this review is to provide a detailed description of the functional properties of MSCs and EVs and their potential clinical applications, with a special focus on pain treatment. The analgesic, anti-inflammatory and regenerative properties of MSCs and EVs will be discussed for several diseases, such as neuropathic pain, osteoarthritis and spinal cord injury.
Collapse
Affiliation(s)
- Marta Bryk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Elżbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jakub Mlost
- Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | | |
Collapse
|
27
|
Yu C, Li D, Wang C, Xia K, Wang J, Zhou X, Ying L, Shu J, Huang X, Xu H, Han B, Chen Q, Li F, Tang J, Liang C, Slater N. Injectable kartogenin and apocynin loaded micelle enhances the alleviation of intervertebral disc degeneration by adipose-derived stem cell. Bioact Mater 2021; 6:3568-3579. [PMID: 33842742 PMCID: PMC8022109 DOI: 10.1016/j.bioactmat.2021.03.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/20/2021] [Accepted: 03/06/2021] [Indexed: 02/06/2023] Open
Abstract
Cell transplantation has been proved the promising therapeutic effects on intervertebral disc degeneration (IVDD). However, the increased levels of reactive oxygen species (ROS) in the degenerated region will impede the efficiency of human adipose-derived stem cells (human ADSCs) transplantation therapy. It inhibits human ADSCs proliferation, and increases human ADSCs apoptosis. Herein, we firstly devised a novel amphiphilic copolymer PEG-PAPO, which could self-assemble into a nanosized micelle and load lipophilic kartogenin (KGN), as a single complex (PAKM). It was an injectable esterase-responsive micelle, and showed controlled release ability of KGN and apocynin (APO). Oxidative stimulation promoted the esterase activity in human ADSCs, which accelerate degradation of esterase-responsive micelle. Compared its monomer, the PAKM micelle possessed better bioactivities, which were attributed to their synergistic effect. It enhanced the viability, autophagic activation (P62, LC3 II), ECM-related transcription factor (SOX9), and ECM (Collagen II, Aggrecan) maintenance in human ADSCs. Furthermore, it is demonstrated that the injection of PAKM with human ADSCs yielded higher disc height and water content in rats. Therefore, PAKM micelles perform promoting cell survival and differentiation effects, and may be a potential therapeutic agent for IVDD.
Collapse
Affiliation(s)
- Chao Yu
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Dongdong Li
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, and College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Chenggui Wang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Kaishun Xia
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Jingkai Wang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Xiaopeng Zhou
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Liwei Ying
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Jiawei Shu
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Xianpeng Huang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Haibin Xu
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Bin Han
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Qixin Chen
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Fangcai Li
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Jianbin Tang
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, and College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Chengzhen Liang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Nigel Slater
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
28
|
Lin Z, Miao J, Zhang T, He M, Zhou X, Zhang H, Gao Y, Bai L. d-Mannose suppresses osteoarthritis development in vivo and delays IL-1β-induced degeneration in vitro by enhancing autophagy activated via the AMPK pathway. Biomed Pharmacother 2021; 135:111199. [PMID: 33401221 DOI: 10.1016/j.biopha.2020.111199] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/13/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is a heterogeneous disease that is consistently difficult to treat due to the complexity of the regulatory network involved in OA pathogenesis, especially in terms of cartilage degeneration. As a C-2 epimer of glucose, d-mannose can alleviate bone loss and repress immunopathology by upregulating regulatory T cells; however, the role of d-mannose in OA-related cartilage degeneration remains unknown. In this study, we investigated the chondroprotective effect of d-mannose in vitro and in vivo on OA. We found that incubating interleukin (IL)-1β-treated rat chondrocytes with d-mannose restrained OA degeneration by elevating cell proliferation, strongly activating autophagy, reducing apoptosis, and downregulating catabolism. Additionally, oral gavage administration of d-mannose to monosodium iodoacetate (MIA)-treated rats revealed that a median (1.25 g/kg/day) rather than high or low dose of d-mannose suppressed OA progression and attenuated OA development based on lower macroscopic scores for cartilage, decreased histological scores for cartilage and synovium, strongly activated autophagy, and downregulated catabolism. In terms of a downstream mechanism, we showed that d-mannose might attenuate OA degeneration by activating autophagy in IL-1β-treated rat chondrocytes by promoting the phosphorylation of 5' AMP-activated protein kinase (AMPK). Our in vitro findings revealed that d-mannose delayed IL-1β-induced OA degeneration in rat chondrocytes by enhancing autophagy activation through the AMPK pathway. Furthermore, the in vivo results indicated that a median dose of d-mannose suppressed MIA-induced OA development. These results suggested that d-mannose exhibits chondroprotective effects and represents a potential disease-modifying drug and novel therapeutic agent for OA.
Collapse
Affiliation(s)
- Zhiming Lin
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Jianing Miao
- Medical Research Center/Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Shenyang, 110000, China
| | - Tao Zhang
- Department of Thoracic Surgery, Xiamen Branch of Zhongshan Hospital of Fudan University, Xiamen, 361000, China
| | - Ming He
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Xiaonan Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - He Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Yue Gao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Lunhao Bai
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110000, China.
| |
Collapse
|
29
|
Ji Q, Qiao X, Liu Y, Wang D. Expression of long-chain noncoding RNA GAS5 in osteoarthritis and its effect on apoptosis and autophagy of osteoarthritis chondrocytes. Histol Histopathol 2021; 36:475-484. [PMID: 33586778 DOI: 10.14670/hh-18-312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the expression of long-chain noncoding RNA GAS5 in osteoarthritis (OA) and the effect of silencing GAS5 on autophagy of osteoarthritis chondrocytes (OACs). METHODS OA rat models were constructed by cutting the anterior cruciate ligament, and the expressions of GAS5 in rat cartilage tissues at 4 weeks (early OA) and 12 weeks (late OA) after modeling were detected. The rat chondrocytes were isolated, cultured and transfected with si-GAS5 to silencing GAS5. Then, the changes of apoptosis and autophagy levels of OA chondrocytes were detected by transfection of GFP-LC3 and flow cytometry. Bioinformatic tools were used to analyze the miRNA binding to GAS5 and the downstream target genes, then luciferase reporter assay and GDC-0349 (inhibitor of mTOR) were used to verify their relationships. RESULTS The expression of GAS5 in cartilage tissue of OA rats was higher than control, which was higher in late OA than that in early OA. After silencing the GAS5, the autophagy ability of OACs was increased and the apoptosis rate was decreased. GAS5 was able to bind to miR-144 and regulate the expressin of mTOR. mTOR inhibitor GDC-0349 could reverse the inhibition of GAS5 on autophagy but could not reverse its effect on apoptosis. CONCLUSION GAS5 expresses highly in OA cartilage tissues and increases with the progression of OA. GAS5 inhibits autophagy and promotes the apoptosis of OACs, and the inhibition of autophagy may be related to its regulation of mTOR.
Collapse
Affiliation(s)
- Qinghui Ji
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, PR China. .,Department of Orthopedics, The First Affiliated Hospital of Jiamusi University, Jiamusi, PR China
| | - Xiaofeng Qiao
- Department of Orthopedics, The First Affiliated Hospital of Jiamusi University, Jiamusi, PR China
| | - Yongxiang Liu
- Department of Orthopedics, Hegang People's Hospital, Hegang, Heilongjiang Province, PR China
| | - Dawei Wang
- Department of Orthopedics, First Hospital of Zhangjiakou, Zhangjiakou, Hebei Province, PR China
| |
Collapse
|
30
|
Shi S, Tian T, Li Y, Xiao D, Zhang T, Gong P, Lin Y. Tetrahedral Framework Nucleic Acid Inhibits Chondrocyte Apoptosis and Oxidative Stress through Activation of Autophagy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:56782-56791. [PMID: 33289541 DOI: 10.1021/acsami.0c17307] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is a degenerative articular cartilage pathogenic process that is accompanied by excessive chondrocyte apoptosis. The occurrence of chondrocyte death and OA is related to decreased autophagy. Tetrahedral framework nucleic acid (TFNA), a potent bioactive DNA nanomaterial, exerts antiapoptotic and antioxidative effects in various diseases, resulting in autophagy promotion and inhibition of the Wnt/β-catenin-signaling pathway. Here, we aimed to elucidate the therapeutic effects of TFNA on OA and its potential molecular mechanism of action. TFNA was synthesized and characterized by established methods. An interleukin (IL)-1β stimulated OA cell model was established and treated with TFNA. Cellular uptake of TFNA and intracellular reactive oxygen species levels were examined via immunofluorescence and flow cytometry. Apoptotic cell death was documented by the Cell Counting Kit-8 (CCK8) assay and flow cytometry. Transmission electron microscopy was applied to view the autophagosomes. The expression of BCL2, BAX, caspase-3, Nrf2, HO-1, LC3-II, Beclin1, Atg7, β-catenin, Lef-1, and CyclinD1 was detected by immunofluorescence and western blotting. TFNA was successfully synthesized and effectively entered chondrocytes in the absence or presence of IL-1β without the help of transfection agents. TFNA treatment in IL-1β-induced chondrocytes reduced apoptosis by activating the BCL2/BAX/caspase-3 pathway, inhibited oxidative stress by regulating the Nrf2/HO-1-signaling pathway, and enhanced autophagy through upregulated LC3-II, Beclin1, and Atg7. Moreover, TFNA showed chondroprotective effects by regulating the Wnt/β-catenin-signaling pathway. Overall, TFNA may have utility as a therapeutic nanomedicine for OA.
Collapse
Affiliation(s)
- Sirong Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Taoran Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yanjing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.,College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| |
Collapse
|
31
|
Zhang J, Cheng F, Rong G, Tang Z, Gui B. Hsa_circ_0005567 Activates Autophagy and Suppresses IL-1β-Induced Chondrocyte Apoptosis by Regulating miR-495. Front Mol Biosci 2020; 7:216. [PMID: 33062640 PMCID: PMC7477291 DOI: 10.3389/fmolb.2020.00216] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
Excessive chondrocyte apoptosis is mostly responsible for the progression of osteoarthritis (OA). It has been shown that circular RNAs (circRNAs) are differentially expressed in OA cartilage and participate in various pathological processes during OA. Here, this study was designed to explore the effect and molecular mechanism of hsa_circ_0005567 on IL-1β-induced chondrocyte apoptosis. The results showed that hsa_circ_0005567 knockdown aggravated the IL-1β-induced chondrocyte apoptosis. In contrast, hsa_circ_0005567 overexpression attenuated the IL-1β-induced chondrocyte apoptosis, but this effect could be abrogated by 3-methyladenine (an inhibitor of autophagy), suggesting that hsa_circ_0005567 overexpression inhibited chondrocyte apoptosis by inducing autophagy. Furthermore, hsa_circ_0005567 competitively bound to miR-495 and derepressed the expression of ATG14, an early autophagy marker that was a direct target of miR-495. Moreover, both miR-495 mimic and ATG14 knockdown counteracted the autophagy-promoting and anti-apoptotic effects of hsa_circ_0005567 overexpression in IL-1β-treated chondrocytes. Taken together, hsa_circ_0005567 activates autophagy by regulating the miR-495/ATG14 axis and thereby suppresses IL-1β-induced chondrocyte apoptosis. These findings suggest that hsa_circ_0005567 may serve as a therapeutic target for the treatment of OA.
Collapse
Affiliation(s)
- Jinling Zhang
- Department of Orthopedic, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fangyue Cheng
- Department of Rheumatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Genxiang Rong
- Department of Orthopedic, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhi Tang
- Department of Orthopedic, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Binjie Gui
- Department of Orthopedic, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
32
|
Shi X, Han L, Sun T, Zhang F, Ji S, Zhang M, Wang X, Yang W. Silencing UHRF1 enhances cell autophagy to prevent articular chondrocytes from apoptosis in osteoarthritis through PI3K/AKT/mTOR signaling pathway. Biochem Biophys Res Commun 2020; 529:1018-1024. [PMID: 32819559 DOI: 10.1016/j.bbrc.2020.06.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/06/2020] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a common chronic degenerative joint disease, and chondrocyte apoptosis is one of most important pathological changes of OA pathogenesis. Growing studies have shown that Ubiquitin-like with PHD and RING finger domains 1 (UHRF1) is an important epigenetic regulatory factor that regulates cell proliferation and apoptosis of various tumors, but its role in OA remains ill-defined. In the present study, we found that UHRF1 expression was increased in human OA cartilage tissues, compared with normal cartilage tissues. Interleukin-1β (IL-1β), a major inflammatory cytokine that promotes cartilage degradation in OA, was used to stimulate primary human chondrocytes in vitro. The expression of UHRF1 was also enhanced in IL-1β-induced chondrocytes. Moreover, down-regulation of UHRF1 induced an increase on cell proliferation and autophagy, and a decrease on apoptosis of chondrocytes after IL-1β treatment. Further data indicated that silencing UHRF1 attenuated the up-regulation of IL-1β on phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway in chondrocytes. Then, an activator of PI3K weakened the effect of UHRF1 silencing on cell proliferation, autophagy, apoptosis of IL-1β-induced chondrocytes, and the cell autophagy special inhibitor 3-methyladenine (3-MA) also showed a same impact on UHRF1, hence suggesting that knockdown of UHRF1 enhances cell autophagy to protect chondrocytes from apoptosis in OA through PI3K/AKT/mTOR signaling pathway. In conclusion, our study suggests that UHRF1 may be a potential regulator of chondrocyte apoptosis in the pathogenesis of OA.
Collapse
Affiliation(s)
- Xiaojuan Shi
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lei Han
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Tianshu Sun
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Feng Zhang
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shiying Ji
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Min Zhang
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaoqing Wang
- Outpatient Department, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Weihong Yang
- Outpatient Department,No.986 Hospital,The Forth Military Medical University, Xi'an, 710054, China.
| |
Collapse
|
33
|
Snider C, Bellrichard M, Meyer A, Kannan R, Grant D, Grant S. A novel crosslinker-free technique toward the fabrication of collagen microspheres. J Biomed Mater Res B Appl Biomater 2020; 108:2789-2798. [PMID: 32190977 DOI: 10.1002/jbm.b.34608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/10/2020] [Accepted: 03/08/2020] [Indexed: 11/09/2022]
Abstract
Injectable collagen microspheres (CMs) have the potential to be an excellent tool to deliver various modulatory agents or to be used as a cellular transporter. A drawback has been the difficulty in producing reliable and spherical CMs. A crosslinker-free method to fabricate CMs was developed using liquid collagen (LC) in a water-in-oil emulsion process with varying concentrations of surfactant span-80. Different emulsion times of up to 16-hr were utilized to produce the CMs. Visual microscopy and scanning electron microscopy were utilized to determine the morphology of the CMs. To determine the fibril nature of the CMs, focus ion beam milling, energy dispersive spectroscopy, and Fourier Transformation-Infrared spectroscopy were performed. A cell biocompatibility study was performed to assess the biocompatibility of the CMs. The results demonstrated that consistent spherical CMs were achievable by changing the span-80 concentration. The CMs were fibrilized not only at the surface, but also at the core. Both the 1- and 16-hr emulsion time demonstrated biocompatibility and it appeared that the cells preferentially adhered to the CMs. This crosslinker-free method to fabricate CMs resulted in spherical, stable, biocompatible CMs, and could be an excellent technique for multiple tissue engineering applications.
Collapse
Affiliation(s)
- Colten Snider
- Department of Biomedical, Biological & Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| | - Mitch Bellrichard
- Department of Veterinary Pathology, University of Missouri, Columbia, Missouri, USA
| | - Amber Meyer
- Department of Materials Science and Engineering, Missouri University Science & Technology, Rolla, Missouri, USA
| | - Raghuraman Kannan
- Department of Biomedical, Biological & Chemical Engineering, University of Missouri, Columbia, Missouri, USA.,Department of Radiology, University of Missouri, Columbia, Missouri, USA
| | - Dave Grant
- Department of Biomedical, Biological & Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| | - Sheila Grant
- Department of Biomedical, Biological & Chemical Engineering, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
34
|
Sharaf K, Kleinsasser A, Schwenk-Zieger S, Gires O, Schinke H, Kohlbauer V, Jakob M, Canis M, Haubner F. Molecular Characterization of Lipoaspirates Used in Regenerative Head and Neck Surgery. JAMA FACIAL PLAST SU 2020; 21:526-534. [PMID: 31556908 DOI: 10.1001/jamafacial.2019.0851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Importance Adipose-derived mesenchymal stem cells (ASCs) have been used commonly in regenerative medicine and increasingly for head and neck surgical procedures. Lipoaspiration with centrifugation is purported to be a mild method for the extraction of ASCs used for autologous transplants to restore tissue defects or induce wound healing. The content of ASCs, their paracrine potential, and cellular potential in wound healing have not been explored for this method to our knowledge. Objective To evaluate the characteristics of lipoaspirates used in reconstructive head and neck surgical procedures with respect to wound healing. Design, Setting, and Participants This case series study included 15 patients who received autologous fat injections in the head and neck during surgical procedures at a tertiary referral center. The study was performed from October 2017 to November 2018, and data were analyzed from October 2017 to February 2019. Main Outcomes and Measures Excessive material of lipoaspirates from subcutaneous abdominal fatty tissue was examined. Cellular composition was analyzed using immunohistochemistry (IHC) and flow cytometry, and functionality was assessed through adipose, osteous, and chondral differentiation in vitro. Supernatants were tested for paracrine ASC functions in fibroblast wound-healing assays. Enzyme-linked immunosorbent assay measurement of tumor necrosis factor (TNF), vascular endothelial growth factor (VEGF), stromal-derived factor 1α (SDF-1α), and transforming growth factor β3 (TGF-β3) was performed. Results Among the 15 study patients (8 [53.3%] male; mean [SD] age at the time of surgery, 63.0 [2.8] years), the stromal vascular fraction (mean [SE], 53.3% [4.2%]) represented the largest fraction within the native lipoaspirates. The cultivated cells were positive for CD73 (mean [SE], 99.90% [0.07%]), CD90 (99.40% [0.32%]), and CD105 (88.54% [2.74%]); negative for CD34 (2.70% [0.45%]) and CD45 (1.74% [0.28%]) in flow cytometry; and negative for CD14 (10.56 [2.81] per 300 IHC score) and HLA-DR (6.89 [2.97] per 300 IHC score) in IHC staining; they differentiated into osteoblasts, adipocytes, and chondrocytes. The cultivated cells showed high expression of CD44 (mean [SE], 99.78% [0.08%]) and CD273 (82.56% [5.83%]). The supernatants were negative for TNF (not detectable) and SDF-1α (not detectable) and were positive for VEGF (mean [SE], 526.74 [149.84] pg/mL for explant supernatants; 528.26 [131.79] pg/106 per day for cell culture supernatants) and TGF-β3 (mean [SE], 22.79 [3.49] pg/mL for explant supernatants; 7.97 [3.15] pg/106 per day for cell culture supernatants). Compared with control (25% or 50% mesenchymal stem cell medium), fibroblasts treated with ASC supernatant healed the scratch-induced wound faster (mean [SE]: control, 1.000 [0.160]; explant supernatant, 1.369 [0.070]; and passage 6 supernatant, 1.492 [0.094]). Conclusions and Relevance The cells fulfilled the international accepted criteria for mesenchymal stem cells. The lipoaspirates contained ASCs that had the potential to multidifferentiate with proliferative and immune-modulating properties. The cytokine profile of the isolated ASCs had wound healing-promoting features. Lipoaspirates may have a regenerative potential and an application in head and neck surgery. Level of Evidence NA.
Collapse
Affiliation(s)
- Kariem Sharaf
- Department of Otolaryngology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Antonia Kleinsasser
- Department of Otolaryngology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Sabina Schwenk-Zieger
- Department of Otolaryngology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Olivier Gires
- Department of Otolaryngology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Henrik Schinke
- Department of Otolaryngology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Vera Kohlbauer
- Department of Otolaryngology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Mark Jakob
- Department of Otolaryngology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Martin Canis
- Department of Otolaryngology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Frank Haubner
- Department of Otolaryngology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW To review the current basic science and clinical literature on mesenchymal stem cell (MSC) therapy for articular cartilage defects and osteoarthritis of the knee. RECENT FINDINGS MSCs derived from bone marrow, adipose, and umbilical tissue have the capacity for self-renewal and differentiation into the chondrocyte lineage. In theory, MSC therapy may help restore cartilage focally or diffusely where nascent regenerative potential in the intra-articular environment is limited. Over the last several years, in vitro and animal studies have elucidated the use of MSCs in isolation as injectables, in combination with biological delivery media and scaffolding, and as surgical adjuvants for cartilage regeneration and treatment of knee degenerative conditions. More recently, clinical and translational literature has grown more convincing from early descriptive case series to randomized controlled trials showing promise in efficacy and safety. Studies describing MSC for knee cartilage regeneration applications are numerous and varied in quality. Future research directions should include work on elucidating optimal cell concentration and dosing, as well as standardization in methodology and reporting in prospective trials. Backed by promise from in vitro and animal studies, preliminary clinical evidence on MSC therapy shows promise as a nonoperative therapeutic option or an adjuvant to existing surgical cartilage restoration techniques. While higher quality evidence to support MSC therapy has emerged over the last several years, further refinement of methodology will be necessary to support its routine clinical use.
Collapse
|
36
|
Torres-Torrillas M, Rubio M, Damia E, Cuervo B, Del Romero A, Peláez P, Chicharro D, Miguel L, Sopena JJ. Adipose-Derived Mesenchymal Stem Cells: A Promising Tool in the Treatment of Musculoskeletal Diseases. Int J Mol Sci 2019; 20:ijms20123105. [PMID: 31242644 PMCID: PMC6627452 DOI: 10.3390/ijms20123105] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 02/08/2023] Open
Abstract
Chronic musculoskeletal (MSK) pain is one of the most common medical complaints worldwide and musculoskeletal injuries have an enormous social and economical impact. Current pharmacological and surgical treatments aim to relief pain and restore function; however, unsatiscactory outcomes are commonly reported. In order to find an accurate treatment to such pathologies, over the last years, there has been a significantly increasing interest in cellular therapies, such as adipose-derived mesenchymal stem cells (AMSCs). These cells represent a relatively new strategy in regenerative medicine, with many potential applications, especially regarding MSK disorders, and preclinical and clinical studies have demonstrated their efficacy in muscle, tendon, bone and cartilage regeneration. Nevertheless, several worries about their safety and side effects at long-term remain unsolved. This article aims to review the current state of AMSCs therapy in the treatment of several MSK diseases and their clinical applications in veterinary and human medicine.
Collapse
Affiliation(s)
- Marta Torres-Torrillas
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Monica Rubio
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Elena Damia
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Belen Cuervo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Ayla Del Romero
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Pau Peláez
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Deborah Chicharro
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Laura Miguel
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| | - Joaquin J Sopena
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain.
| |
Collapse
|
37
|
Protective effects of ten oligostilbenes from Paeonia suffruticosa seeds on interleukin-1β-induced rabbit osteoarthritis chondrocytes. BMC Chem 2019; 13:72. [PMID: 31384819 PMCID: PMC6661769 DOI: 10.1186/s13065-019-0589-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/07/2019] [Indexed: 11/10/2022] Open
Abstract
Background Paeonia suffruticosa is an important traditional Chinese herb used to treat osteoarthritis (OA) and oligostilbenes are the main active ingredient of the seeds of P. suffruticosa. The monomer trans-resveratrol of this species was demonstrated to have chondroprotective effects as a lead compound for the treatment of osteoarthritis, but it has not been applied due to its low efficacy. Methods Oligostilbenes were isolated by chromatography and were identified by NMR and HPLC. A rabbit osteoarthritis chondrocyte model was induced by interleukin-1β and was treated with individual drugs to systematically evaluate their effects. Cell Counting Kit 8 was used to test their effects on cell viability, calculate EC50 and plot a dose-response curve.Their effects on apoptosis were analyzed by Annexin V and PI staining, and the expression of chondrocyte-specific genes COL2A1, MMP13 and SOX9 was evaluated by real-time PCR. Results Paeonia suffruticosa seed extract could promote the cell viability of rabbit OA chondrocytes at low concentration and then ten oligostilbenes were isolated from it. Trans-oligostilbenes were better than their cis-forms, trimers and dimers were better than monomers for promoting the cell viability of rabbit osteoarthritis chondrocytes. None of the oligostilbenes was more effective than seed extract at the appropriate concentration; 1 μM oligostilbenes all showed various anti-apoptotic effects. Trans-gnetin H showed the best effect on proliferation and inhibition of MMP13 expression on OA chondrocytes, while trans-viniferin was most effective in promoting the expression of COL2A1 and SOX9. Conclusions Ten oligostilbenes from P. suffruticosa seed all have certain protective effects on OA chondrocytes at low concentration. The trans-viniferin and some trimers have the potential to be further developed for the treatment of osteoarthritis because they were more effective than resveratrol and diacerein. The synergistic effect that may exist between oligostilbenes also warrants further research.
Collapse
|
38
|
Wang AT, Feng Y, Jia HH, Zhao M, Yu H. Application of mesenchymal stem cell therapy for the treatment of osteoarthritis of the knee: A concise review. World J Stem Cells 2019; 11:222-235. [PMID: 31110603 PMCID: PMC6503460 DOI: 10.4252/wjsc.v11.i4.222] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/07/2019] [Accepted: 03/16/2019] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) refers to a chronic joint disease characterized by degenerative changes of articular cartilage and secondary bone hyperplasia. Since articular cartilage has a special structure, namely the absence of blood vessels as well as the low conversion rate of chondrocytes in the cartilage matrix, the treatment faces numerous clinical challenges. Traditional OA treatment (e.g., arthroscopic debridement, microfracture, autologous or allogeneic cartilage transplantation, chondrocyte transplantation) is primarily symptomatic treatment and pain management, which cannot contribute to regenerating degenerated cartilage or reducing joint inflammation. Also, the generated mixed fibrous cartilage tissue is not the same as natural hyaline cartilage. Mesenchymal stem cells (MSCs) have turned into the most extensively explored new therapeutic drugs in cell-based OA treatment as a result of their ability to differentiate into chondrocytes and their immunomodulatory properties. In this study, the preliminary results of preclinical (OA animal model)/clinical trials regarding the effects of MSCs on cartilage repair of knee joints are briefly summarized, which lay a solid application basis for more and deeper clinical studies on cell-based OA treatment.
Collapse
Affiliation(s)
- Ai-Tong Wang
- Cell Products of National Engineering Research Center, National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Ying Feng
- Cell Products of National Engineering Research Center, National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Hong-Hong Jia
- Cell Products of National Engineering Research Center, National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Meng Zhao
- Cell Products of National Engineering Research Center, National Stem Cell Engineering Research Center, Tianjin 300457, China
| | - Hao Yu
- Cell Products of National Engineering Research Center, National Stem Cell Engineering Research Center, Tianjin 300457, China
| |
Collapse
|