1
|
Lee S, Kim B, Lee MJ, Kim D, Park S, Kim J, Arai Y, Lee SH. Curcumin-PLGA NPs coated with targeting biomimetic personalized stem cell membranes for osteoarthritis therapy. J Control Release 2025; 381:113625. [PMID: 40081740 DOI: 10.1016/j.jconrel.2025.113625] [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: 10/24/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Traditional drug delivery systems for OA treatments face limitations due to rapid clearance within the joint and low biocompatibility. Moreover, the inflammation associated with OA exacerbates tissue damage and delays the regenerative capacity of therapeutics. To overcome these limitations, an OA-specific drug delivery system designated dCOL2-CM-Cur-PNPs is developed herein to target OA cartilage for anti-inflammatory and cartilage regeneration purposes. This system is constructed using cell membranes obtained from induced pluripotent stem cell -derived mesenchymal stem cells (iMSC-CMs), poly(D,l-lactide-co-glycolide) (PLGA) nanoparticles loaded with the well-known anti-inflammatory and cartilage-regenerating agent curcumin (Cur-PNPs), and damaged type II collagen (dCOL2)-targeting phospholipids. Coating the Cur-PNPs with iMSC-CMs enhances the sustained release of curcumin and improves its cellular uptake by OA-induced chondrocytes. The dCOL2-CM-Cur-PNPs restores the chondrogenic properties of the OA-induced chondrocytes, inhibit the pro-inflammatory function of M1 macrophages, and promote the anti-inflammatory function of M2 macrophages. The dCOL2-targeting phospholipids integrated on the surface of the iMSC-CMs facilitate specific binding to OA cartilage, as validated by in-vitro and in-vivo experiments. Additionally, the dCOL2-CM-Cur-PNPs alleviate OA progression in a DMM rat model. This drug delivery system based on iMSC-CMs modified with dCOL2-targeting phospholipids demonstrates significant potential as a next-generation platform for promoting cartilage regeneration through OA-specific therapy.
Collapse
Affiliation(s)
- Sunjun Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Bowon Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Min-Ju Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Deogil Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Sunghyun Park
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Jinsik Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea.
| |
Collapse
|
2
|
Wang Z, Li X, Jiang Y, Wu T, Guo S, Li T. Preparation of hydrogel microsphere and its application in articular cartilage injury. Mater Today Bio 2025; 31:101641. [PMID: 40130039 PMCID: PMC11931253 DOI: 10.1016/j.mtbio.2025.101641] [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: 11/26/2024] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
In recent years, hydrogel microspheres have garnered significant attention due to their unique structure and functionality, demonstrating substantial potential in articular cartilage injury repair. This paper provides a comprehensive overview of current strategies for cartilage injury repair and summarizes the materials and preparation methods of hydrogel microspheres. Furthermore, it highlights the multiple roles of hydrogel microspheres in cartilage repair, including inflammation control, regulation of chondrocyte metabolism, drug and cell delivery, lubrication improvement, and recruitment of endogenous stem cells. Finally, the paper discusses the application prospects of hydrogel microspheres, identifies current limitations and challenges, and offers insights to guide future research and practical applications in cartilage injury repair.
Collapse
Affiliation(s)
- Zehua Wang
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xiaoxia Li
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266000, China
| | - Yaping Jiang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Tingyu Wu
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Sijia Guo
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Tao Li
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| |
Collapse
|
3
|
Li Z, Lu H, Fan L, Ma X, Duan Z, Zhang Y, Fu Y, Wang S, Guan Y, Yang D, Chen Q, Xu T, Yang Y. Microneedle-Delivered PDA@Exo for Multifaceted Osteoarthritis Treatment via PI3K-Akt-mTOR Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406942. [PMID: 39206714 PMCID: PMC11558126 DOI: 10.1002/advs.202406942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Osteoarthritis (OA) is marked by cartilage deterioration, subchondral bone changes, and an inflammatory microenvironment. The study introduces the Microneedle-Delivered Polydopamine-Exosome (PDA@Exo MN), a therapeutic that not only preserves cartilage and promotes bone regeneration but also improves localized drug delivery through enhanced penetration capabilities. PDA@Exo MN shows strong reactive oxygen species (ROS) scavenging abilities and high biocompatibility, fostering osteogenesis and balancing anabolic and catabolic processes in cartilage. It directs macrophage polarization from M0 to the anti-inflammatory M2 phenotype. RNA sequencing of treated chondrocytes demonstrates restored cellular function and activated antioxidant responses, with modulated inflammatory pathways. The PI3K-AKT-mTOR pathway's activation, essential for PDA@Exo's effects, is confirmed via bioinformatics and Western blot. In vivo assessments robustly validate that PDA@Exo MN prevents cartilage degradation and OA progression, supported by histological assessments and micro-CT analysis, highlighting its disease-modifying impact. The excellent biocompatibility of PDA@Exo MN, verified through histological (H&E) and blood tests showing no organ damage, underscores its safety and efficacy for OA therapy, making it a novel and multifunctional nanomedical approach in orthopedics, characterized by organ-friendliness and biosecurity.
Collapse
Affiliation(s)
- Zihua Li
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Hengli Lu
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Limin Fan
- School of MedicineTongji UniversityShanghai200092P. R. China
| | - Xiaoyi Ma
- School of MedicineTongji UniversityShanghai200092P. R. China
| | - Zhengwei Duan
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Yiwei Zhang
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Yuesong Fu
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Sen Wang
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Yonghao Guan
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Dong Yang
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Qingjing Chen
- Southern Medical UniversityGuangzhou510515P. R. China
| | - Tianyang Xu
- Department of OrthopedicsShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072P. R. China
| | - Yunfeng Yang
- Department of OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| |
Collapse
|
4
|
Wu X, Fu Y, Ma J, Li C, He A, Zhang T. LGR5 Modulates Differentiated Phenotypes of Chondrocytes Through PI3K/AKT Signaling Pathway. Tissue Eng Regen Med 2024; 21:791-807. [PMID: 38771465 PMCID: PMC11187034 DOI: 10.1007/s13770-024-00645-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Tissue engineering is increasingly viewed as a promising avenue for functional cartilage reconstruction. However, chondrocyte dedifferentiation during in vitro culture remains an obstacle for clinical translation of tissue engineered cartilage. Re-differentiated induction have been employed to induce dedifferentiated chondrocytes back to their original phenotype. Regrettably, these strategies have been proven to be only moderately effective. METHODS To explore underlying mechanism, RNA transcriptome sequencing was conducted on primary chondrocytes (P0), dedifferentiated chondrocytes (P5), and redifferentiated chondrocytes (redifferentiation-induction of P5, P5.R). Based on multiple bioinformatics analysis, LGR5 was identified as a target gene. Subsequently, stable cell lines with LGR5 knocking-down and overexpression were established using P0 chondrocytes. The phenotypic changes in P1 and P5 chondrocytes with either LGR5 knockdown or overexpression were assessed to ascertain the potential influence of LGR5 dysregulation on chondrocyte phenotypes. Regulatory mechanism was then investigated using bioinformatic analysis, protein-protein docking, immunofluorescence co-localization and immunoprecipitation. RESULTS The current study found that dysregulation of LGR5 can significantly impact the dedifferentiated phenotypes of chondrocytes (P5). Upregulation of LGR5 appears to activate the PI3K/AKT signal via increasing the phosphorylation levels of AKT (p-AKT1). Moreover, the increase of p-AKT1 may stabilize β-catenin and enhance the intensity of Wnt/β-catenin signal, and help to restore the dedifferentated phenotype of chondrocytes. CONCLUSION LGR5 can modulate the phenotypes of chondrocytes in P5 passage through PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Xu Wu
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
- Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, ENT Institute, Fudan University, Shanghai, 200031, China
| | - Yaoyao Fu
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
- Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, ENT Institute, Fudan University, Shanghai, 200031, China
| | - Jing Ma
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
- Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, ENT Institute, Fudan University, Shanghai, 200031, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, China
| | - Chenlong Li
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
- Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, ENT Institute, Fudan University, Shanghai, 200031, China
| | - Aijuan He
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China.
| | - Tianyu Zhang
- Department of Facial Plastic and Reconstructive Surgery, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China.
- Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, ENT Institute, Fudan University, Shanghai, 200031, China.
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
5
|
Olate-Moya F, Rubí-Sans G, Engel E, Mateos-Timoneda MÁ, Palza H. 3D Bioprinting of Biomimetic Alginate/Gelatin/Chondroitin Sulfate Hydrogel Nanocomposites for Intrinsically Chondrogenic Differentiation of Human Mesenchymal Stem Cells. Biomacromolecules 2024; 25:3312-3324. [PMID: 38728671 DOI: 10.1021/acs.biomac.3c01444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
3D-printed hydrogel scaffolds biomimicking the extracellular matrix (ECM) are key in cartilage tissue engineering as they can enhance the chondrogenic differentiation of mesenchymal stem cells (MSCs) through the presence of active nanoparticles such as graphene oxide (GO). Here, biomimetic hydrogels were developed by cross-linking alginate, gelatin, and chondroitin sulfate biopolymers in the presence of GO as a bioactive filler, with excellent processability for developing bioactive 3D printed scaffolds and for the bioprinting process. A novel bioink based on our hydrogel with embedded human MSCs presented a cell survival rate near 100% after the 3D bioprinting process. The effects of processing and filler concentration on cell differentiation were further quantitatively evaluated. The nanocomposited hydrogels render high MSC proliferation and viability, exhibiting intrinsic chondroinductive capacity without any exogenous factor when used to print scaffolds or bioprint constructs. The bioactivity depended on the GO concentration, with the best performance at 0.1 mg mL-1. These results were explained by the rational combination of the three biopolymers, with GO nanoparticles having carboxylate and sulfate groups in their structures, therefore, biomimicking the highly negatively charged ECM of cartilage. The bioactivity of this biomaterial and its good processability for 3D printing scaffolds and 3D bioprinting techniques open up a new approach to developing novel biomimetic materials for cartilage repair.
Collapse
Affiliation(s)
- Felipe Olate-Moya
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Avenida Beauchef 851, 8370458 Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Avenida Monseñor Álvaro del Portillo 12455, 7620086 Las Condes, Chile
| | - Gerard Rubí-Sans
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri Reixac, 10, 12, 08028, 08019 Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 50018 Zaragoza, Spain
| | - Elisabeth Engel
- IMEM-BRT Group, Departament de Ciència i Enginyeria de Materials, EEBE, Universitat Politècnica de Catalunya (UPC), C/Eduard Maristany 10-14, 08019 Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri Reixac, 10, 12, 08028, 08019 Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 50018 Zaragoza, Spain
| | - Miguel Ángel Mateos-Timoneda
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Josep Trueta Street s/n, 08195 Sant Cugat del Vallès, Barcelona, Spain
- Department of Basic Sciences, Faculty of Medicine and Health Sciences, Univesitat Internacional de Catalunya, Josep Trueta Street s/n, 08195 Sant Cugat del Vallès, Barcelona, Spain
| | - Humberto Palza
- Departamento de Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Avenida Beauchef 851, 8370458 Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Avenida Monseñor Álvaro del Portillo 12455, 7620086 Las Condes, Chile
| |
Collapse
|
6
|
Yıldırım M, Ünsal N, Kabataş B, Eren O, Şahin F. Effect of Solanum lycopersicum and Citrus limon-Derived Exosome-Like Vesicles on Chondrogenic Differentiation of Adipose-Derived Stem Cells. Appl Biochem Biotechnol 2024; 196:203-219. [PMID: 37103740 DOI: 10.1007/s12010-023-04491-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 04/28/2023]
Abstract
Articular cartilage defect treatment is a very important problem because its therapeutic options are not successful enough. Due to the weak self-repairing capacity of the avascular cartilage, even minor damage can progress and cause joint damage leading to osteoarthritis. Although various treatment strategies have been developed to repair damaged cartilage, cell- and exosome-based therapies are promising. Plant extracts have been used for decades, and their effects on cartilage regeneration have been studied. Exosome-like vesicles, which are secreted by all living cells, are involved in cell-to-cell communication and cell homeostasis. The differentiation potential of exosome-like vesicles isolated from S. lycopersicum and C. limon, which are known to have anti-inflammatory and antioxidant properties, was investigated in the differentiation of human adipose-derived mesenchymal stem cells (hASCs) into chondrocytes. In order to obtain tomato-derived exosome-like vesicles (TELVs) and lemon-derived exosome-like vesicles (LELVs) Aquous Two- Phase system was performed. Characterisation of isolated vesicles based on size, shape were achived via Zetasizer, NTA FAME analysis, and SEM techniques. These results showed that TELVs and LELVs increased cell viability and did not show any toxic effects on stem cells. Although TELVs triggered chondrocyte formation, LELVs downregulated. The expression of ACAN, SOX9, and COMP, known as chondrocyte markers, was increased by TELV treatment. In addition, protein expression of the two most important proteins, COL2 and COLXI, found in the extracellular matrix of cartilage, increased. These findings suggest that TELVs can be used for cartilage regeneration, and may be a novel and promising treatment for osteoarthritis.
Collapse
Affiliation(s)
- Merve Yıldırım
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, TR-34755, Istanbul, Turkey
| | - Naz Ünsal
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, TR-34755, Istanbul, Turkey
| | - Bilge Kabataş
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, TR-34755, Istanbul, Turkey
| | - Olcay Eren
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, TR-34755, Istanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, TR-34755, Istanbul, Turkey.
| |
Collapse
|
7
|
Jurczak P, Lach S. Hydrogels as Scaffolds in Bone-Related Tissue Engineering and Regeneration. Macromol Biosci 2023; 23:e2300152. [PMID: 37276333 DOI: 10.1002/mabi.202300152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/22/2023] [Indexed: 06/07/2023]
Abstract
Several years have passed since the medical and scientific communities leaned toward tissue engineering as the most promising field to aid bone diseases and defects resulting from degenerative conditions or trauma. Owing to their histocompatibility and non-immunogenicity, bone grafts, precisely autografts, have long been the gold standard in bone tissue therapies. However, due to issues associated with grafting, especially the surgical risks and soaring prices of the procedures, alternatives are being extensively sought and researched. Fibrous and non-fibrous materials, synthetic substitutes, or cell-based products are just a few examples of research directions explored as potential solutions. A very promising subgroup of these replacements involves hydrogels. Biomaterials resembling the bone extracellular matrix and therefore acting as 3D scaffolds, providing the appropriate mechanical support and basis for cell growth and tissue regeneration. Additional possibility of using various stimuli in the form of growth factors, cells, etc., within the hydrogel structure, extends their use as bioactive agent delivery platforms and acts in favor of their further directed development. The aim of this review is to bring the reader closer to the fascinating subject of hydrogel scaffolds and present the potential of these materials, applied in bone and cartilage tissue engineering and regeneration.
Collapse
Affiliation(s)
- Przemyslaw Jurczak
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Centre Polish Academy of Sciences, Gdansk, 80-308, Poland
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk, 80-308, Poland
| | - Slawomir Lach
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk, 80-308, Poland
| |
Collapse
|
8
|
Baei P, Daemi H, Aramesh F, Baharvand H, Eslaminejad MB. Advances in mechanically robust and biomimetic polysaccharide-based constructs for cartilage tissue engineering. Carbohydr Polym 2023; 308:120650. [PMID: 36813342 DOI: 10.1016/j.carbpol.2023.120650] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
The purpose of cartilage tissue engineering is to provide artificial constructs with biological functions and mechanical features that resemble native tissue to improve tissue regeneration. Biochemical characteristics of the cartilage extracellular matrix (ECM) microenvironment provide a platform for researchers to develop biomimetic materials for optimal tissue repair. Due to the structural similarity of polysaccharides into physicochemical characteristics of cartilage ECM, these natural polymers capture special attention for developing biomimetic materials. The mechanical properties of constructs play a crucial influence in load-bearing cartilage tissues. Moreover, the addition of appropriate bioactive molecules to these constructs can promote chondrogenesis. Here, we discuss polysaccharide-based constructs that can be used to create substitutes for cartilage regeneration. We intend to focus on newly developed bioinspired materials, fine-tuning the mechanical properties of constructs, the design of carriers loaded by chondroinductive agents, and development of appropriate bioinks as a bioprinting approach for cartilage regeneration.
Collapse
Affiliation(s)
- Payam Baei
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran
| | - Hamed Daemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran.
| | - Fatemeh Aramesh
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University ofTehran, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
9
|
Jones CL, Penney BT, Theodossiou SK. Engineering Cell-ECM-Material Interactions for Musculoskeletal Regeneration. Bioengineering (Basel) 2023; 10:bioengineering10040453. [PMID: 37106640 PMCID: PMC10135874 DOI: 10.3390/bioengineering10040453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
The extracellular microenvironment regulates many of the mechanical and biochemical cues that direct musculoskeletal development and are involved in musculoskeletal disease. The extracellular matrix (ECM) is a main component of this microenvironment. Tissue engineered approaches towards regenerating muscle, cartilage, tendon, and bone target the ECM because it supplies critical signals for regenerating musculoskeletal tissues. Engineered ECM-material scaffolds that mimic key mechanical and biochemical components of the ECM are of particular interest in musculoskeletal tissue engineering. Such materials are biocompatible, can be fabricated to have desirable mechanical and biochemical properties, and can be further chemically or genetically modified to support cell differentiation or halt degenerative disease progression. In this review, we survey how engineered approaches using natural and ECM-derived materials and scaffold systems can harness the unique characteristics of the ECM to support musculoskeletal tissue regeneration, with a focus on skeletal muscle, cartilage, tendon, and bone. We summarize the strengths of current approaches and look towards a future of materials and culture systems with engineered and highly tailored cell-ECM-material interactions to drive musculoskeletal tissue restoration. The works highlighted in this review strongly support the continued exploration of ECM and other engineered materials as tools to control cell fate and make large-scale musculoskeletal regeneration a reality.
Collapse
Affiliation(s)
- Calvin L Jones
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| | - Brian T Penney
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| | - Sophia K Theodossiou
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Dr MS2085, Boise, ID 83725, USA
| |
Collapse
|
10
|
Huang L, Jin M, Gu R, Xiao K, Lu M, Huo X, Sun M, Yang Z, Wang Z, Zhang W, Zhi L, Meng Z, Ma J, Ma J, Zhang R. miR-199a-5p Reduces Chondrocyte Hypertrophy and Attenuates Osteoarthritis Progression via the Indian Hedgehog Signal Pathway. J Clin Med 2023; 12:jcm12041313. [PMID: 36835852 PMCID: PMC9959662 DOI: 10.3390/jcm12041313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Osteoarthritis (OA), the most common type of arthritis, is an age-associated disease, characterized by the progressive degradation of articular cartilage, synovial inflammation, and degeneration of subchondral bone. Chondrocyte proliferation is regulated by the Indian hedgehog (IHH in humans, Ihh in animals) signaling molecule, which regulates hypertrophy and endochondral ossification in the development of the skeletal system. microRNAs (miRNAs, miRs) are a family of about 22-nucleotide endogenous non-coding RNAs, which negatively regulate gene expression. In this study, the expression level of IHH was upregulated in the damaged articular cartilage tissues among OA patients and OA cell cultures, while that of miR-199a-5p was the opposite. Further investigations demonstrated that miR-199a-5p could directly regulate IHH expression and reduce chondrocyte hypertrophy and matrix degradation via the IHH signal pathway in the primary human chondrocytes. The intra-articular injection of synthetic miR-199a-5p agomir attenuated OA symptoms in rats, including the alleviation of articular cartilage destruction, subchondral bone degradation, and synovial inflammation. The miR-199a-5p agomir could also inhibit the Ihh signaling pathway in vivo. This study might help in understanding the role of miR-199a-5p in the pathophysiology and molecular mechanisms of OA and indicate a potential novel therapeutic strategy for OA patients.
Collapse
Affiliation(s)
- Lei Huang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Meng Jin
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Ruiying Gu
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Kunlin Xiao
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Mengnan Lu
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Xinyu Huo
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Mengyao Sun
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Zhi Yang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Zhiyuan Wang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Weijie Zhang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Liqiang Zhi
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Ziang Meng
- Department of Mathematics and Computing Science, Simon Fraser University, Vancouver, BC V6B 5K3, Canada
| | - Jie Ma
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Jianbing Ma
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Correspondence: (J.M.); (R.Z.)
| | - Rui Zhang
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Correspondence: (J.M.); (R.Z.)
| |
Collapse
|
11
|
Talebi Jouybari M, Fani N, Jahangir S, Bagheri F, Golru R, Taghiyar L. Validation of Tissue-Engineered Constructs: Preclinical and Clinical Studies. CARTILAGE: FROM BIOLOGY TO BIOFABRICATION 2023:491-527. [DOI: 10.1007/978-981-99-2452-3_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
Lin J, Chen L, Yang J, Li X, Wang J, Zhu Y, Xu X, Cui W. Injectable Double Positively Charged Hydrogel Microspheres for Targeting-Penetration-Phagocytosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2202156. [PMID: 36056898 DOI: 10.1002/smll.202202156] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/27/2022] [Indexed: 06/15/2023]
Abstract
The localization and accumulation of drugs in the body determine their therapeutic effects; however, the specific microstructure of damaged tissues hinders drug delivery. Currently, there is a shortage of effective drug carriers to breach these barriers and achieve efficient tissue and cellular delivery of drugs. In this study, an injectable double positively charged functional hydrogel microsphere with "targeting cartilage extracellular matrix", "cartilage penetration", and "cellular phagocytosis" is designed for matching the structural characteristics of joints, addressing the difficulties of drug delivery in joints. The microspheres could be adsorbed on the negatively charged cartilage surface because of their positively charged poly-lysine surface. Furthermore, the internally loaded positively charged polyamidoamine contained kartogenin, which helped further the penetration of the cartilage under the guidance of electrical charge. The microspheres could release kartogenin for more than 21 days. In in vivo experiments, the microspheres effectively improve the efficiency of drug delivery, inhibit the degradation of cartilage matrix and subchondral bone, and delay the development of osteoarthritis. As a double positively charged drug delivery system, the versatile microsphere has great potential for treating osteoarthritis and other diseases.
Collapse
Affiliation(s)
- Jiawei Lin
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Liang Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Jielai Yang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Xingchen Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Juan Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yuan Zhu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Xiangyang Xu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
13
|
Targeted mesenchymal stem cell therapy equipped with a cell-tissue nanomatchmaker attenuates osteoarthritis progression. Sci Rep 2022; 12:4015. [PMID: 35256711 PMCID: PMC8901617 DOI: 10.1038/s41598-022-07969-9] [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: 10/16/2021] [Accepted: 02/22/2022] [Indexed: 11/08/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are at the forefront of research for a wide range of diseases, including osteoarthritis (OA). Despite having attracted the attention of orthopedists, current MSC therapy techniques are limited by poor MSC implantation in tissue defects and lack of lateral tissue integration, which has restricted the efficacy of cell therapy to alleviate OA symptoms only. Here, we developed targeted MSC therapy for OA cartilage using a cell-tissue matchmaking nanoconstruct (C-TMN). C-TMN, as an MSC vehicle, consists of a central iron oxide nanoparticle armed with two types of antibodies, one directed at the MSC surface and the other against articular cartilage. We treated rat OA articular cartilage with intra-articular injections of C-TMN with and without exogenous MSCs. We observed substantial improvements in both symptomatic and radiographic OA caused by C-TMN, which was independent of exogenous MSCs. This new approach could predict a promising future for OA management.
Collapse
|
14
|
Jelodari S, Ebrahimi Sadrabadi A, Zarei F, Jahangir S, Azami M, Sheykhhasan M, Hosseini S. New Insights into Cartilage Tissue Engineering: Improvement of Tissue-Scaffold Integration to Enhance Cartilage Regeneration. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7638245. [PMID: 35118158 PMCID: PMC8807044 DOI: 10.1155/2022/7638245] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023]
Abstract
Distinctive characteristics of articular cartilage such as avascularity and low chondrocyte conversion rate present numerous challenges for orthopedists. Tissue engineering is a novel approach that ameliorates the regeneration process by exploiting the potential of cells, biodegradable materials, and growth factors. However, problems exist with the use of tissue-engineered construct, the most important of which is scaffold-cartilage integration. Recently, many attempts have been made to address this challenge via manipulation of cellular, material, and biomolecular composition of engineered tissue. Hence, in this review, we highlight strategies that facilitate cartilage-scaffold integration. Recent advances in where efficient integration between a scaffold and native cartilage could be achieved are emphasized, in addition to the positive aspects and remaining problems that will drive future research.
Collapse
Affiliation(s)
- Sahar Jelodari
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amin Ebrahimi Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fatemeh Zarei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom, Iran
| | - Samaneh Hosseini
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
15
|
Yao X, Ma Y, Zhou W, Liao Y, Jiang Z, Lin J, He Q, Wu H, Wei W, Wang X, Björklund M, Ouyang H. In-cytoplasm mitochondrial transplantation for mesenchymal stem cells engineering and tissue regeneration. Bioeng Transl Med 2022; 7:e10250. [PMID: 35111950 PMCID: PMC8780934 DOI: 10.1002/btm2.10250] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 12/15/2022] Open
Abstract
Stem cell therapies are unsatisfactory due to poor cell survival and engraftment. Stem cell used for therapy must be properly "tuned" for a harsh in vivo environment. Herein, we report that transfer of exogenous mitochondria (mito) to adipose-derived mesenchymal stem cells (ADSCs) can effectively boost their energy levels, enabling efficient cell engraftment. Importantly, the entire process of exogeneous mitochondrial endocytosis is captured by high-content live-cell imaging. Mitochondrial transfer leads to acutely enhanced bioenergetics, with nearly 17% of higher adenosine 5'-triphosphate (ATP) levels in ADSCs treated with high mitochondrial dosage and further results in altered secretome profiles of ADSCs. Mitochondrial transfer also induced the expression of 334 mRNAs in ADSCs, which are mainly linked to signaling pathways associated with DNA replication and cell division. We hypothesize that increase in ATP and cyclin-dependent kinase 1 and 2 expression might be responsible for promoting enhanced proliferation, migration, and differentiation of ADSCs in vitro. More importantly, mito-transferred ADSCs display prolonged cell survival, engraftment and horizontal transfer of exogenous mitochondria to surrounding cells in a full-thickness skin defect rat model with improved skin repair compared with nontreated ADSCs. These results demonstrate that intracellular mitochondrial transplantation is a promising strategy to engineer stem cells for tissue regeneration.
Collapse
Affiliation(s)
- Xudong Yao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
- The Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Yuanzhu Ma
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Wenyan Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Youguo Liao
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Zongsheng Jiang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Junxin Lin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Qiulin He
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Hongwei Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Wei Wei
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
- The Fourth Affiliated HospitalZhejiang University School of MedicineYiwuChina
| | - Xiaozhao Wang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Mikael Björklund
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang UniversityHainingChina
- Department of Sports MedicineZhejiang University School of MedicineHangzhouChina
- China Orthopedic Regenerative Medicine Group (CORMed)HangzhouChina
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of MedicineHangzhouChina
| |
Collapse
|
16
|
Higher Chondrogenic Potential of Extracellular Vesicles Derived from Mesenchymal Stem Cells Compared to Chondrocytes-EVs In Vitro. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9011548. [PMID: 34938811 PMCID: PMC8687842 DOI: 10.1155/2021/9011548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/09/2021] [Accepted: 11/16/2021] [Indexed: 12/21/2022]
Abstract
The inability of cartilage to self-repair necessitates an effective therapeutic approach to restore damaged tissues. Extracellular vesicles (EVs) are attractive options because of their roles in cellular communication and tissue repair where they regulate the cellular processes of proliferation, differentiation, and recruitment. However, it is a challenge to determine the relevant cell sources for isolation of EVs with high chondrogenic potential. The current study aims to evaluate the chondrogenic potential of EVs derived from chondrocytes (Cho-EV) and mesenchymal stem cells (MSC-EV). The EVs were separately isolated from conditioned media of both rabbit bone marrow MSCs and chondrocyte cultures. The isolated vesicles were assessed in terms of size, morphology, and surface marker expression. The chondrogenic potential of MSCs in the presence of different concentrations of EVs (50, 100, and 150 μg/ml) was evaluated during 21 days, and chondrogenic surface marker expressions were checked by qRT-PCR and histologic assays. The extracted vesicles had a spherical morphology and a size of 44.25 ± 8.89 nm for Cho-EVs and 112.1 ± 10.10 nm for MSC-EVs. Both groups expressed the EV-specific surface markers CD9 and CD81. Higher expression of chondrogenic specified markers, especially collagen type II (COL II), and secretion of glycosaminoglycans (GAGs) and proteoglycans were observed in MSCs treated with 50 and 100 μg/ml MSC-EVs compared to the Cho-EVs. The results from the use of EVs, particularly MSC-EVs, with high chondrogenic ability will provide a basis for developing therapeutic agents for cartilage repair.
Collapse
|
17
|
Enhancing Stem Cell Therapy for Cartilage Repair in Osteoarthritis-A Hydrogel Focused Approach. Gels 2021; 7:gels7040263. [PMID: 34940323 PMCID: PMC8701810 DOI: 10.3390/gels7040263] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/05/2021] [Accepted: 12/09/2021] [Indexed: 12/19/2022] Open
Abstract
Sem cells hold tremendous promise for the treatment of cartilage repair in osteoarthritis. In addition to their multipotency, stem cells possess immunomodulatory effects that can alleviate inflammation and enhance cartilage repair. However, the widely clinical application of stem cell therapy to cartilage repair and osteoarthritis has proven difficult due to challenges in large-scale production, viability maintenance in pathological tissue site and limited therapeutic biological activity. This review aims to provide a perspective from hydrogel-focused approach to address few key challenges in stem cell-based therapy for cartilage repair and highlight recent progress in advanced hydrogels, particularly microgels and dynamic hydrogels systems for improving stem cell survival, retention and regulation of stem cell fate. Finally, progress in hydrogel-assisted gene delivery and genome editing approaches for the development of next generation of stem cell therapy for cartilage repair in osteoarthritis are highlighted.
Collapse
|
18
|
Heydari Nasrabadi M, Parsivand M, Mohammadi N, Asghari Moghaddam N. Comparison of Elaeagnus angustifolia L. extract and quercetin on mouse model of knee osteoarthritis. J Ayurveda Integr Med 2021; 13:100529. [PMID: 34862093 PMCID: PMC8728052 DOI: 10.1016/j.jaim.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 01/21/2023] Open
Abstract
Osteoarthritis (OA) is the most commonly observed arthritic disease causing severe pain and impairing patient's quality of life. This study aimed to investigate and compare the effect of Elaeagnus angustifolia extract and quercetin on the mouse model of knee osteoarthritis (OA). Sixty Balb-C mice were used to establish the monosodium iodoacetate (MIA) model of OA. Then, they were randomized into untreated OA group (normal nutrition), E. angustifolia extract-treated group (32 mg/kg by gavage), quercetin-treated group (20 mg/kg by gavage) and ibuprofen- treated group (20 mg/kg). Fifteen mice with no MIA treatment were considered as the normal controls. The mice were treated for 28 days. The histopathological analysis was performed on knee joints. Expression levels of matrix metalloproteinase 3 and 13 (MMP-1 and MMP-13) in serum were assessed in addition. Histopathological study indicated that in the quercetin-treated group, the thickness of femur and tibia were significantly increased (P < 0.05). Among groups treated by E. angustifolia extract, quercetin and ibuprofen, the concentration of MMP-3 was 5.47 ± 1.75 ng/ml, 4.38 ± 1.78 ng/ml and 4.86 ± 1.40 ng/ml, respectively. The level of MMP-13 in sera was 3.32 ± 1.64 ng/ml, 2.67 ± 1.73 ng/ml and 5.31 ± 1.68 ng/ml in the same order (P < 0.05). The results of this study suggest that the quercetin was useful in the reduction of symptoms of OA and raised the improvement of damaged cartilage. Hence, it can be a beneficial medical supplement in OA treatment. Besides, E. angustifolia extract and quercetin significantly reduced the serum MMP-3 and MMP-13 concentrations. It could be one of the mechanisms through that E. angustifolia plays a role in remission of OA.
Collapse
Affiliation(s)
| | - Malahat Parsivand
- Department of Biology, Parand Branch, Islamic Azad University, Tehran, Iran
| | - Narges Mohammadi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
19
|
Han X, Wu Y, Shan Y, Zhang X, Liao J. Effect of Micro-/Nanoparticle Hybrid Hydrogel Platform on the Treatment of Articular Cartilage-Related Diseases. Gels 2021; 7:gels7040155. [PMID: 34698122 PMCID: PMC8544595 DOI: 10.3390/gels7040155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/18/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023] Open
Abstract
Joint diseases that mainly lead to articular cartilage injury with prolonged severe pain as well as dysfunction have remained unexplained for many years. One of the main reasons is that damaged articular cartilage is unable to repair and regenerate by itself. Furthermore, current therapy, including drug therapy and operative treatment, cannot solve the problem. Fortunately, the micro-/nanoparticle hybrid hydrogel platform provides a new strategy for the treatment of articular cartilage-related diseases, owing to its outstanding biocompatibility, high loading capability, and controlled release effect. The hybrid platform is effective for controlling symptoms of pain, inflammation and dysfunction, and cartilage repair and regeneration. In this review, we attempt to summarize recent studies on the latest development of micro-/nanoparticle hybrid hydrogel for the treatment of articular cartilage-related diseases. Furthermore, some prospects are proposed, aiming to improve the properties of the micro-/nanoparticle hybrid hydrogel platform so as to offer useful new ideas for the effective and accurate treatment of articular cartilage-related diseases.
Collapse
|
20
|
Hyaluronan and Derivatives: An In Vitro Multilevel Assessment of Their Potential in Viscosupplementation. Polymers (Basel) 2021; 13:polym13193208. [PMID: 34641024 PMCID: PMC8512809 DOI: 10.3390/polym13193208] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/08/2021] [Accepted: 09/12/2021] [Indexed: 12/12/2022] Open
Abstract
In this research work, viscosupplements based on linear, derivatized, crosslinked and complexed HA forms were extensively examined, providing data on the hydrodynamic parameters for the water-soluble-HA-fraction, rheology, sensitivity to enzymatic hydrolysis and capacity to modulate specific biomarkers’ expression in human pathological chondrocytes and synoviocytes. Soluble HA ranged from 0 to 32 mg/mL and from 150 to 1330 kDa MW. The rheological behavior spanned from purely elastic to viscoelastic, suggesting the diversity of the categories that are suitable for restoring specific/different features of the healthy synovial fluid. The rheological parameters were reduced in a diverse manner upon dilution and hyaluronidases action, indicating different durations of the viscosupplementation effect. Bioactivity was found for all the samples, increasing the expression of different matrix markers (e.g., hyaluronan-synthase); however, the hybrid cooperative complexes performed better in most of the experiments. Hybrid cooperative complexes improved COLII mRNA expression (~12-fold increase vs. CTR), proved the most effective at preserving cell phenotype. In addition, in these models, the HA samples reduced inflammation. IL-6 was down-regulated vs. CTR by linear and chemically modified HA, and especially by hybrid complexes. The results represent the first comprehensive panel of data directly comparing the diverse HA forms for intra-articular injections and provide valuable information for tailoring products’ clinical use as well as for designing new, highly performing HA-formulations that can address specific needs.
Collapse
|
21
|
Employing Extracellular Matrix-Based Tissue Engineering Strategies for Age-Dependent Tissue Degenerations. Int J Mol Sci 2021; 22:ijms22179367. [PMID: 34502277 PMCID: PMC8431718 DOI: 10.3390/ijms22179367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 01/10/2023] Open
Abstract
Tissues and organs are not composed of solely cellular components; instead, they converge with an extracellular matrix (ECM). The composition and function of the ECM differ depending on tissue types. The ECM provides a microenvironment that is essential for cellular functionality and regulation. However, during aging, the ECM undergoes significant changes along with the cellular components. The ECM constituents are over- or down-expressed, degraded, and deformed in senescence cells. ECM aging contributes to tissue dysfunction and failure of stem cell maintenance. Aging is the primary risk factor for prevalent diseases, and ECM aging is directly or indirectly correlated to it. Hence, rejuvenation strategies are necessitated to treat various age-associated symptoms. Recent rejuvenation strategies focus on the ECM as the basic biomaterial for regenerative therapies, such as tissue engineering. Modified and decellularized ECMs can be used to substitute aged ECMs and cell niches for culturing engineered tissues. Various tissue engineering approaches, including three-dimensional bioprinting, enable cell delivery and the fabrication of transplantable engineered tissues by employing ECM-based biomaterials.
Collapse
|
22
|
Li Z, Li M, Xu P, Ma J, Zhang R. Compositional Variation and Functional Mechanism of Exosomes in the Articular Microenvironment in Knee Osteoarthritis. Cell Transplant 2021; 29:963689720968495. [PMID: 33086893 PMCID: PMC7784575 DOI: 10.1177/0963689720968495] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a major cause of disability worldwide with increasing age. Knee OA (KOA) is the most prevalent type of OA. Recently, it is considered that KOA is a whole joint disease, including articular cartilage, subchondral bone, synovium, ligaments, joint capsules, and muscles around the joint. Exosomes in knee joint are mainly secreted by articular chondrocytes and synoviocytes. They participate in cell and tissue cross-talk by carrying a complex cargo of proteins, lipids, nucleic acids, etc. Under normal conditions, exosomes maintain the microenvironmental homeostasis of the joint cavity. Under pathological conditions, the composition and function of exosomes changes, which in turn, disrupts the balance of anabolism and catabolism of articular chondrocyte and facilitates inflammatory responses, thus accelerating KOA progression. As a regenerative medicine, mesenchymal stem cells (MSCs) are promised to facilitate repair of degenerated cartilage and decelerate OA process. The therapeutic function of MSC mainly depends on MSC-derived exosomes, which can restore the homeostasis of the articular microenvironment. In the future, the specific mechanism of exosomes for OA treatment needs further elucidation, and the treatment effect of exosomes for long-term and/or severe OA needs further exploration.
Collapse
Affiliation(s)
- Zheng Li
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.,Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Manling Li
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Gui Yang, China
| | - Peng Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jie Ma
- Medical Research Center, Xi'an No. 3 Hospital, Xi'an, China.,School of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Rui Zhang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
23
|
Gellan gum/alginate-based Ca-enriched acellular bilayer hydrogel with robust interface bonding for effective osteochondral repair. Carbohydr Polym 2021; 270:118382. [PMID: 34364624 DOI: 10.1016/j.carbpol.2021.118382] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/14/2021] [Accepted: 06/24/2021] [Indexed: 02/05/2023]
Abstract
The treatment of osteochondral (OC) defects remains challenging because of the lack of economical and feasible therapeutic strategies for OC repair and reconstruction. In this study, we report an integrated bilayer hydrogel with robust interface binding force (40 kPa) by facilitating the diffusion of calcium ions to the secondary crosslink of the bilayer hydrogel, in which gellan gum and sodium alginate acted as the chondral layer, gellan gum and hydroxyapatite acted as subchondral layer. This integrated construct has high cytocompatibility, and can seed with mesenchymal stem cells (MSCs) related to different functional protein expression for cartilage and bone formation, respectively. Furthermore, in the rabbit critical-sized osteochondral defect model (4.0 mm in diameter and 8.0 mm in depth), the calcium enriched hydrogel act as a calcium reservoir, promote neovascularization at week 4, and repair the critical defect at week 8, demonstrating the feasible preparation of an acellular hydrogel for OC repair.
Collapse
|
24
|
Baer K, Kieser S, Schon B, Rajendran K, Ten Harkel T, Ramyar M, Löbker C, Bateman C, Butler A, Raja A, Hooper G, Anderson N, Woodfield T. Spectral CT imaging of human osteoarthritic cartilage via quantitative assessment of glycosaminoglycan content using multiple contrast agents. APL Bioeng 2021; 5:026101. [PMID: 33834156 PMCID: PMC8018795 DOI: 10.1063/5.0035312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/22/2021] [Indexed: 01/06/2023] Open
Abstract
Detection of early osteoarthritis to stabilize or reverse the damage to articular cartilage would improve patient function, reduce disability, and limit the need for joint replacement. In this study, we investigated nondestructive photon-processing spectral computed tomography (CT) for the quantitative measurement of the glycosaminoglycan (GAG) content compared to destructive histological and biochemical assay techniques in normal and osteoarthritic tissues. Cartilage-bone cores from healthy bovine stifles were incubated in 50% ioxaglate (Hexabrix®) or 100% gadobenate dimeglumine (MultiHance®). A photon-processing spectral CT (MARS) scanner with a CdTe-Medipix3RX detector imaged samples. Calibration phantoms of ioxaglate and gadobenate dimeglumine were used to determine iodine and gadolinium concentrations from photon-processing spectral CT images to correlate with the GAG content measured using a dimethylmethylene blue assay. The zonal distribution of GAG was compared between photon-processing spectral CT images and histological sections. Furthermore, discrimination and quantification of GAG in osteoarthritic human tibial plateau tissue using the same contrast agents were demonstrated. Contrast agent concentrations were inversely related to the GAG content. The GAG concentration increased from 25 μg/ml (85 mg/ml iodine or 43 mg/ml gadolinium) in the superficial layer to 75 μg/ml (65 mg/ml iodine or 37 mg/ml gadolinium) in the deep layer of healthy bovine cartilage. Deep zone articular cartilage could be distinguished from subchondral bone by utilizing the material decomposition technique. Photon-processing spectral CT images correlated with histological sections in healthy and osteoarthritic tissues. Post-imaging material decomposition was able to quantify the GAG content and distribution throughout healthy and osteoarthritic cartilage using Hexabrix® and MultiHance® while differentiating the underlying subchondral bone.
Collapse
Affiliation(s)
| | - Sandra Kieser
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE), Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch 8011, New Zealand
| | | | | | | | - Mohsen Ramyar
- Department of Radiology, University of Otago Christchurch, Christchurch 8011, New Zealand
| | | | - Christopher Bateman
- Department of Radiology, University of Otago Christchurch, Christchurch 8011, New Zealand
| | | | | | | | - Nigel Anderson
- Department of Radiology, University of Otago Christchurch, Christchurch 8011, New Zealand
| | | |
Collapse
|
25
|
Ravari MK, Mashayekhan S, Zarei F, Sayyahpour FA, Taghiyar L, Baghban Eslaminejad M. Fabrication and characterization of an injectable reinforced composite scaffold for cartilage tissue engineering: an in vitro study. Biomed Mater 2021; 16:045007. [PMID: 33784250 DOI: 10.1088/1748-605x/abed97] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There are limitations in current medications of articular cartilage injuries. Although injectable bioactive hydrogels are promising options, they have decreased biomechanical performance. Researchers should consider many factors when providing solutions to overcome these challenges. In this study, we created an injectable composite hydrogel from chitosan and human acellular cartilage extracellular matrix (ECM) particles. In order to enhance its mechanical properties, we reinforced this hydrogel with microporous microspheres composed of the same materials as the structural building blocks of the scaffold. Articular cartilage from human donors was decellularized by a combination of physical, chemical, and enzymatic methods. The decellularization efficiency was assessed by histological analysis and assessment of DNA content. We characterized the composite constructs in terms of storage modulus, gelation time, biocompatibility, and differentiation potential. The results showed that mechanical behavior increased with an increase in microsphere content. The sample that contained 10% microsphere had an enhanced storage modulus of up to 90 kPa. Biocompatibility and preliminary differentiation investigations revealed that this composite hydrogel might have potential benefits for cartilage tissue engineering.
Collapse
Affiliation(s)
- Mojtaba Khozaei Ravari
- Department of chemical and petroleum engineering, Sharif University of Technology, Tehran 11365-8639, Iran. Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | | | | | | | | | | |
Collapse
|
26
|
Xue S, Zhou X, Sang W, Wang C, Lu H, Xu Y, Zhong Y, Zhu L, He C, Ma J. Cartilage-targeting peptide-modified dual-drug delivery nanoplatform with NIR laser response for osteoarthritis therapy. Bioact Mater 2021; 6:2372-2389. [PMID: 33553822 PMCID: PMC7844135 DOI: 10.1016/j.bioactmat.2021.01.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/12/2021] [Accepted: 01/12/2021] [Indexed: 12/14/2022] Open
Abstract
Cartilage-targeting delivery of therapeutic agents is still an effective strategy for osteoarthritis (OA) therapy. Recently, scavenging for reactive oxygen species (ROS) and activating autophagy have been increasingly reported to treat OA effectively. In this study, we designed, for the first time, a dual-drug delivery system based on metal organic framework (MOF)-decorated mesoporous polydopamine (MPDA) which composed of rapamycin (Rap) loaded into the mesopores and bilirubin (Br) loaded onto the shell of MOF. The collagen II-targeting peptide (WYRGRL) was then conjugated on the surface of above nanocarrier to develop a cartilage-targeting dual-drug delivery nanoplatform (RB@MPMW). Our results indicated the sequential release of two agents from RB@MPMW could be achieved via near-infrared (NIR) laser irritation. Briefly, the rapid release of Br from the MOF shell exhibited excellent ROS scavenging ability and anti-apoptosis effects, however responsively reduced autophagy activity, to a certain extent. Meanwhile, following the NIR irradiation, Rap was rapidly released from MPDA core and further enhanced autophagy activation and chondrocyte protection. RB@MPMW continuously phosphorylated AMPK and further rescued mitochondrial energy metabolism of chondrocytes following IL-1β stimulation via activating SIRT1-PGC-1α signaling pathway. Additionally, the cartilage-targeting property of peptide-modified nanocarrier could be monitored via Magnetic Resonance (MR) and IVIS imaging. More significantly, RB@MPMW effectively delayed cartilage degeneration in ACLT rat model. Overall, our findings indicated that the as-prepared dual-drug delivery nanoplatform exerted potent anti-inflammation and anti-apoptotic effects, rescued energy metabolism of chondrocytes in vitro and prevented cartilage degeneration in vivo, which thereby showed positive performance for OA therapy. Collagen type II-targeting peptide and positive surface potential endow RB@MPMW with a fine cartilage affinity ability. RB@MPMW possess superb biological functions of scavenging free radicals and autophagy induction. RB@MPMW effectively promotes chondrocyte mitochondrial energy metabolism in the inflammatory microenvironment. RB@MPMW has a good MR imaging ability, which could monitor its therapeutic effects in vivo.
Collapse
Affiliation(s)
- Song Xue
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Weilin Sang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Cong Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Haiming Lu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yiming Xu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yiming Zhong
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Libo Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Jinzhong Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
27
|
Taghiyar L, Jahangir S, Khozaei Ravari M, Shamekhi MA, Eslaminejad MB. Cartilage Repair by Mesenchymal Stem Cell-Derived Exosomes: Preclinical and Clinical Trial Update and Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1326:73-93. [PMID: 33629260 DOI: 10.1007/5584_2021_625] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) and other degenerative joint diseases are characterized by articular cartilage destruction, synovial inflammation, sclerosis of subchondral bone, and loss of extracellular matrix (ECM). Worldwide, these diseases are major causes of disability. Cell therapies have been considered to be the best therapeutic strategies for long-term treatment of articular cartilage diseases. It has been suggested that the mechanism of stem cell-based therapy is related to paracrine secretion of extracellular vesicles (EVs), which are recognized as the main secretion factors of stem cells. EVs, and in particular the subclass exosomes (Exos), are novel therapeutic approaches for treatment of cartilage lesions and OA. The results of recent studies have shown that EVs isolated from mesenchymal stem cells (MSCs) could inhibit OA progression. EVs isolated from various stem cell sources, such as MSCs, may contribute to tissue regeneration of the limbs, skin, heart, and other tissues. Here, we summarize recent findings of preclinical and clinical studies on different MSC-derived EVs and their effectiveness as a treatment for damaged cartilage. The Exos isolation techniques in OA treatment are also highlighted.
Collapse
Affiliation(s)
- Leila Taghiyar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahrbano Jahangir
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mojtaba Khozaei Ravari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
28
|
Mesenchymal Stem Cell Therapy for Osteoarthritis: Practice and Possible Promises. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1387:107-125. [DOI: 10.1007/5584_2021_695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
Wang M, Luo Y, Yu Y, Chen F. Bioengineering Approaches to Accelerate Clinical Translation of Stem Cell Therapies Treating Osteochondral Diseases. Stem Cells Int 2020; 2020:8874742. [PMID: 33424981 PMCID: PMC7775142 DOI: 10.1155/2020/8874742] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/17/2020] [Accepted: 12/12/2020] [Indexed: 12/15/2022] Open
Abstract
The osteochondral tissue is an interface between articular cartilage and bone. The diverse composition, mechanical properties, and cell phenotype in these two tissues pose a big challenge for the reconstruction of the defected interface. Due to the availability and inherent regenerative therapeutic properties, stem cells provide tremendous promise to repair osteochondral defect. This review is aimed at highlighting recent progress in utilizing bioengineering approaches to improve stem cell therapies for osteochondral diseases, which include microgel encapsulation, adhesive bioinks, and bioprinting to control the administration and distribution. We will also explore utilizing synthetic biology tools to control the differentiation fate and deliver therapeutic biomolecules to modulate the immune response. Finally, future directions and opportunities in the development of more potent and predictable stem cell therapies for osteochondral repair are discussed.
Collapse
Affiliation(s)
- Meng Wang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yixuan Luo
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yin Yu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fei Chen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
30
|
Intra-articular injection of anti-inflammatory peptide-loaded glycol chitosan/fucoidan nanogels to inhibit inflammation and attenuate osteoarthritis progression. Int J Biol Macromol 2020; 170:469-478. [PMID: 33359610 DOI: 10.1016/j.ijbiomac.2020.12.158] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/14/2020] [Accepted: 12/20/2020] [Indexed: 01/20/2023]
Abstract
Glycol chitosan/fucoidan nanogels loaded with anti-inflammatory peptide KAFAK (GC/Fu@KAFAK NGs) were fabricated based on the electrostatic interaction and genipin cross-linking methods. The prepared NGs had an average size of 286.3 ± 5.0 nm and positive surface charge of 14.0 ± 0.2 mV. The anti-inflammatory and chondro-protective effects of GC/Fu@KAFAK NGs were evaluated on interlecukin-1β (IL-1β)-stimulated rat chondrocytes. We found that GC/Fu@KAFAK NGs not only inhibited the expression of inflammatory factors interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), but also enhanced the expression of chondrogenic markers type II collagen, aggrecan, and Sox9. More importantly, in rat osteoarthritis (OA) model, the intra-articular (IA) injection of GC/Fu@KAFAK NGs reduced glycosaminoglycan loss and diminished inflammatory cytokine release. In addition, GC/Fu@KAFAK NGs showed good biocompatibility both in vitro and in vivo. In conclusion, IA inject-able GC/Fu@KAFAK NGs might have great potential in OA treatment.
Collapse
|
31
|
Peng Z, Sun H, Bunpetch V, Koh Y, Wen Y, Wu D, Ouyang H. The regulation of cartilage extracellular matrix homeostasis in joint cartilage degeneration and regeneration. Biomaterials 2020; 268:120555. [PMID: 33285440 DOI: 10.1016/j.biomaterials.2020.120555] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 11/05/2020] [Accepted: 11/18/2020] [Indexed: 12/31/2022]
Abstract
Osteoarthritis (OA) is a major cause of disability and socioeconomic loss worldwide. However, the current pharmacological approaches used to treat OA are largely palliative. Being the hallmark of OA, the cartilage extracellular matrix (ECM) destruction and abnormal homeostasis is gaining more attention as a therapeutic target in cartilage regeneration. Moreover, during the progression of OA, the cartilage ECM shows significant pathological alternations, which can be promising biomarkers in identifying the pathological stages of OA. In this review, we summarize the role of abnormal ECM homeostasis in the joint cartilage during OA. Furthermore, we provide an update on the cartilage ECM derived biomarkers and regenerative medicine therapies targeting cartilage ECM which includes preclinical animal models study and clinical trials.
Collapse
Affiliation(s)
- Zhi Peng
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, And Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, And Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Sun
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, And Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, And Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Varitsara Bunpetch
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, And Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, And Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiwen Koh
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, And Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Ya Wen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, And Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, And Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongmei Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, And Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, And Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, And Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, And Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
32
|
More N, Srivastava A, Kapusetti G. Graphene Oxide Reinforcement Enhances the Piezoelectric and Mechanical Properties of Poly(3-hydroxybutyrate- co-3-hydroxy valerate)-Based Nanofibrous Scaffolds for Improved Proliferation of Chondrocytes and ECM Production. ACS APPLIED BIO MATERIALS 2020; 3:6823-6835. [PMID: 35019345 DOI: 10.1021/acsabm.0c00765] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The discovery of piezoelectricity in natural cartilage has inspired the development of piezoelectric biomaterials for its repair and regeneration using tissue engineering approaches. In the present work, piezoelectric scaffolds composed of poly(3-hydroxybutyrate-co-3-hydroxy valerate) (PB) and graphene oxide (GO) have been successfully fabricated by the electrospinning technology. The fabricated scaffolds were examined for their morphological, physical, chemical, piezoelectric, and biological characterizations. The fiber diameter was found to be in the range of 600-800 nm appropriate for chondrogenic growth. Reinforcement of 1.5% GO enhanced the tensile strength of PB to 2.08 ± 0.33 MPa compared to PB alone (0.59 ± 0.12). Reinforcement of GO significantly enhances the piezoelectric coefficient (d33), and for 0.5, 1, and 1.5% GO in PB, it was found to be 0.12 ± 0.015, 0.57 ± 0.19, and 0.94 ± 0.03 pC/N, respectively, and corresponding voltages of 11.84 ± 1.4, 54.69 ± 18.29, and 100.2 ± 3.2 mV, respectively, were generated. The biological activity of the smart piezo scaffolds was also evaluated on freshly isolated goat chondrocytes. The GO-reinforced scaffold showed higher cell proliferation and cell adhesion as confirmed by alamarBlue assay and field emission scanning electron microscopy imaging. The GO-reinforced scaffold has demonstrated significantly higher extracellular matrix production compared to PB as confirmed by histochemistry and real-time polymerase chain reaction. Hence, the GO-based piezoelectric PB electrospun scaffold can be a better alternative for cell-free and growth factor-free approach for cartilage tissue engineering.
Collapse
Affiliation(s)
- Namdev More
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Akshay Srivastava
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Govinda Kapusetti
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar 382355, Gujarat, India
| |
Collapse
|
33
|
Zhang G, Zhou Y, Su M, Yang X, Zeng B. Inhibition of microRNA-27b-3p relieves osteoarthritis pain via regulation of KDM4B-dependent DLX5. Biofactors 2020; 46:788-802. [PMID: 32856377 DOI: 10.1002/biof.1670] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/12/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) represents a progressive degenerative disorder that predominantly affects the synovial membranes of joints. Recent studies have highlighted the significant role played by microRNAs (miRNAs) in OA development. The current study aimed to elucidate the underlying modulatory role of miR-27b-3p in the development of OA. The expression of miR-27b-3p in the OA patients and rat models post anterior cruciate ligament transection operation was measured using reverse transcription quantitative polymerase chain reaction, through which overexpressed miR-27b-3p was found in both of the samples. To further explore the miR-27b-3p functions in OA, western blot analysis, enzyme-linked immunosorbent assay, and β-galactosidase activity assay were conducted with the results showing that knockdown of miR-27b-3p promoted expression of the osteogenic differentiation markers while inhibiting expression of the adipogenic differentiation markers, inflammatory factors, and cellular senescence of bone marrow mesenchymal stem cells (BMSCs). After that, the interactions between miR-27b-3p, lysine Demethylase 4B (KDM4B), and Distal-Less Homeobox 5 (DLX5) identified using dual-luciferase reporter gene assay and ChIP assay revealed that miR-27b-3p inhibited KDM4B and further reduced expression of DLX5. Finally, the paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) were assessed in rat models, and increased PWT and PWL were detected after miR-27b-3p silencing. In conclusion, suppression of miR-27b-3p could enhance KDM4B and DLX5 to alleviate OA pain, shedding light on a new potential therapeutic target for OA.
Collapse
Affiliation(s)
- Guixiang Zhang
- Operating Room, Xiangtan Central Hospital, Xiangtan, China
| | - Yang Zhou
- Department of Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Manman Su
- Department of Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Xucheng Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Biyun Zeng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
34
|
Chen L, Liu J, Guan M, Zhou T, Duan X, Xiang Z. Growth Factor and Its Polymer Scaffold-Based Delivery System for Cartilage Tissue Engineering. Int J Nanomedicine 2020; 15:6097-6111. [PMID: 32884266 PMCID: PMC7434569 DOI: 10.2147/ijn.s249829] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 07/02/2020] [Indexed: 02/05/2023] Open
Abstract
The development of biomaterials, stem cells and bioactive factors has led to cartilage tissue engineering becoming a promising tactic to repair cartilage defects. Various polymer three-dimensional scaffolds that provide an extracellular matrix (ECM) mimicking environment play an important role in promoting cartilage regeneration. In addition, numerous growth factors have been found in the regenerative process. However, it has been elucidated that the uncontrolled delivery of these factors cannot fully exert regenerative potential and can also elicit undesired side effects. Considering the complexity of the ECM, neither scaffolds nor growth factors can independently obtain successful outcomes in cartilage tissue engineering. Therefore, collectively, an appropriate combination of growth factors and scaffolds have great potential to promote cartilage repair effectively; this approach has become an area of considerable interest in recent investigations. Of late, an increasing trend was observed in cartilage tissue engineering towards this combination to develop a controlled delivery system that provides adequate physical support for neo-cartilage formation and also enables spatiotemporally delivery of growth factors to precisely and fully exert their chondrogenic potential. This review will discuss the role of polymer scaffolds and various growth factors involved in cartilage tissue engineering. Several growth factor delivery strategies based on the polymer scaffolds will also be discussed, with examples from recent studies highlighting the importance of spatiotemporal strategies for the controlled delivery of single or multiple growth factors in cartilage tissue engineering applications.
Collapse
Affiliation(s)
- Li Chen
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China.,School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jiaxin Liu
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Ming Guan
- School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Tongqing Zhou
- School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xin Duan
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Zhou Xiang
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|