1
|
Thompson CL, Bonassar LJ. Timing of cartilage articulation following impact injury affects the response of surface zone chondrocytes. J Orthop Res 2024. [PMID: 39482253 DOI: 10.1002/jor.26002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/09/2024] [Accepted: 10/09/2024] [Indexed: 11/03/2024]
Abstract
Post-traumatic osteoarthritis develops following an inciting injury to a joint and results in cartilage degeneration. Mechanical loading, including articulation, drives anabolic responses in cartilage clinically, in vivo, and in vitro. Tribological articulation, or sliding of cartilage on a glass counterface, has long been used as an in vitro tool to study cartilage tissue behavior. However, it is unclear if tribological articulation affects chondrocyte fate following injury, and if the timing of articulation impacts the resultant effect. The goal of this study was to investigate the effect of tribological articulation on injured cartilage tissue at two time points: (i) performed immediately after injury and (ii) 24 h after injury. Neonatal bovine femoral cartilage explants were injured using a rapid spring-loaded impactor and subsequently subjected to tribological articulation. Cell death due to impact injury was highest near the articular surface, suggesting a strain-dependent mechanism. Immediate articulation following injury mitigated cell death compared to injury alone or delayed articulation; markers for both general cell death and early-stage apoptosis were markedly decreased in the explants that were immediately slid. Interestingly, mitigation of cell death due to sliding was most predominant at the cartilage surface. Tribological articulation is known to create fluid flow within the tissue, predominantly at the articular surface, which could drive the protective response seen here. Altogether, this work shows that perturbations to the cellular environment immediately following cartilage injury significantly impact chondrocyte fate.
Collapse
Affiliation(s)
| | - Lawrence J Bonassar
- Meinig of Biomedical Engineering, Cornell University, Ithaca, New York, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
2
|
Guo L, Li P, Rong X, Wei X. Key roles of the superficial zone in articular cartilage physiology, pathology, and regeneration. Chin Med J (Engl) 2024:00029330-990000000-01274. [PMID: 39439390 DOI: 10.1097/cm9.0000000000003319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Indexed: 10/25/2024] Open
Abstract
ABSTRACT The superficial zone (SFZ) of articular cartilage is an important interface that isolates deeper zones from the microenvironment of the articular cavity and is directly exposed to various biological and mechanical stimuli. The SFZ is not only a crucial structure for maintaining the normal physiological function of articular cartilage but also the earliest site of osteoarthritis (OA) cartilage degeneration and a major site of cartilage progenitor cells, suggesting that the SFZ might represent a key target for the early diagnosis and treatment of OA. However, to date, SFZ research has not received sufficient attention, accounting for only about 0.58% of cartilage tissue research. The structure, biological composition, function, and related mechanisms of the SFZ in the physiological and pathological processes of articular cartilage remain unclear. This article reviews the key role of the SFZ in articular cartilage physiology and pathology and focuses on the characteristics of SFZ in articular cartilage degeneration and regeneration in OA, aiming to provide researchers with a systematic understanding of the current research status of the SFZ of articular cartilage, hoping that scholars will give more attention to the SFZ of articular cartilage in the future.
Collapse
Affiliation(s)
- Li Guo
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Pengcui Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xueqin Rong
- Department of Pain Medicine Center, Central Hospital of Sanya, Sanya, Hainan 572000, China
| | - Xiaochun Wei
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| |
Collapse
|
3
|
Guo J, Su K, Wang L, Feng B, You X, Deng M, Toh WS, Wu J, Cheng B, Xia J. Poly( p-coumaric acid) nanoparticles alleviate temporomandibular joint osteoarthritis by inhibiting chondrocyte ferroptosis. Bioact Mater 2024; 40:212-226. [PMID: 38973989 PMCID: PMC11224931 DOI: 10.1016/j.bioactmat.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024] Open
Abstract
Oxidative stress and inflammation are key drivers of osteoarthritis (OA) pathogenesis and disease progression. Herein we report the synthesis of poly(p-coumaric) nanoparticles (PCA NPs) from p-courmaic acid (p-CA), a naturally occurring phytophenolic acid, to be a multifunctional and drug-free therapeutic for temporomandibular joint osteoarthritis (TMJOA). Compared to hyaluronic acid (HA) that is clinically given as viscosupplementation, PCA NPs exhibited long-term efficacy, superior anti-oxidant and anti-inflammatory properties in alleviating TMJOA and repairing the TMJ cartilage and subchondral bone in a rat model of TMJOA. Notably, TMJ repair mediated by PCA NPs could be attributed to their anti-oxidant and anti-inflammatory properties in enhancing cell proliferation and matrix synthesis, while reducing inflammation, oxidative stress, matrix degradation, and chondrocyte ferroptosis. Overall, our study demonstrates a multifunctional nanoparticle, synthesized from natural p-coumaric acid, that is stable and possess potent antioxidant, anti-inflammatory properties and ferroptosis inhibition, beneficial for treatment of TMJOA.
Collapse
Affiliation(s)
- Jiaxin Guo
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Kai Su
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Liying Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, PR China
| | - Bingyu Feng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Xinru You
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Miao Deng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Wei Seong Toh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore
- Faculty of Dentistry, National University of Singapore, 119085, Singapore
| | - Jun Wu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511455, PR China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| |
Collapse
|
4
|
Zhang CM, Kang YX, Zheng SY, Liu Y, Liu MR, Wu GG, Guo L, Li PC, Zhao RP, Duan QQ, Wang SW, Wei XC. A pH-responsive novel delivery system utilizing carbon quantum dots loaded with PT2385 for targeted inhibition of HIF-2α in the treatment of osteoarthritis. Int J Pharm 2024; 666:124752. [PMID: 39321898 DOI: 10.1016/j.ijpharm.2024.124752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/02/2024] [Accepted: 09/22/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a progressive joint disorder marked by the degradation of cartilage. Elevated concentrations of hypoxia-inducible factor-2α (HIF-2α) are intricately linked to the pathological development of OA. PT2385 has demonstrated effective inhibition of HIF-2α, thereby potentially impeding the initial advancement of OA. Nevertheless, challenges persist, including limited penetration into the deeper layers of cartilage, issues related to charge rejection, and a heightened rate of clearance from the joint. These constraints necessitate further consideration and exploration. METHODS It has been demonstrated that PT2385 exhibits efficient inhibition of HIF-2α expression, thereby contributing to the delay in the progression of osteoarthritis. The pH-responsive attributes of carbon quantum dots, specifically those employing m-phenylenediamine (m-CQDs) coated with bovine serum albumin (BSA), have been systematically evaluated. In both in vitro settings involving cartilage explants and in vivo experiments, the efficacy of BSA-m-CQDs-PT2385 (BCP) has been confirmed in facilitating the transport of PT2385 to the middle and deep layers of cartilage. Furthermore, the BCP system demonstrates controlled drug release contingent upon alterations in environmental pH. RESULTS While the use of PT2385 alone provides protective effects on chondrocytes within an inflamed environment, there exists an opportunity for further enhancement in its efficacy when administered via intra-articular injection. The BCP formulation, characterized by appropriate particle size and charge, facilitates seamless penetration into cartilage tissue. Additionally, BCP demonstrates the capability to release drugs in response to changes in environmental pH. In vitro experiments reveal that BCP effectively inhibits Hif-2α expression and catabolic factors in chondrocytes. Notably, cartilage explants and in vivo experiments indicate that BCP surpasses PT2385 alone in inhibiting the expression of HIF-2α and matrix metalloproteinase 13, particularly in the middle and deep layers. CONCLUSIONS The BCP drug delivery system exhibits selective release of PT2385 in response to pH changes occurring during the progression of osteoarthritis (OA), thereby inhibiting HIF-2α expression deep within the cartilage. The use of BCP significantly augments the capacity of PT2385 to retard both cartilage degeneration and the progression of osteoarthritis. Consequently, BCP as an innovative approach utilizing m-CQDs to deliver PT2385 into articular cartilage, shows potential for treating osteoarthritis.This strategy opens new avenues for osteoarthritis treatment.
Collapse
Affiliation(s)
- Cheng-Ming Zhang
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Yu-Xin Kang
- State Key Laboratory of Advanced Stainless Steels, Taiyuan Iron and Steel (Group) Co., Ltd., Taiyuan 030003, PR China
| | - Si-Yu Zheng
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Yang Liu
- Department of Laboratory Medicine, Handan Second Hospital, Hebei University of Engineering, Handan 056000, PR China
| | - Meng-Rou Liu
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Gai-Ge Wu
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Li Guo
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Peng-Cui Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Rui-Peng Zhao
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Qian-Qian Duan
- College of Information and Computer & Key Laboratory of Advanced Transducers and Intelligent Control System of the Ministry of Education and Shanxi Province, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Shao-Wei Wang
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Xiao-Chun Wei
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| |
Collapse
|
5
|
Zhao X, Huang H, Jiang X, Zheng S, Qiu C, Cheng Y, Lin Y, Wang Y, Yan Y, Di X, Hu M, Zhu W, Wu F, Shi X, Chen R, Kou L. Supramolecular nanoparticle loaded with bilirubin enhances cartilage protection and alleviates osteoarthritis via modulating oxidative stress and inflammatory responses. Colloids Surf B Biointerfaces 2024; 245:114243. [PMID: 39288548 DOI: 10.1016/j.colsurfb.2024.114243] [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: 07/27/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
Osteoarthritis (OA) is a chronic inflammation that gradually leads to cartilage degradation. Prolonged chondrocyte oxidative stress contributes to the development of diseases, including chondrocyte apoptosis, cartilage matrix degradation, and aggravation of articular cartilage damage. Bilirubin (BR) possesses strong antioxidant properties by scavenging reactive oxygen species (ROS) and potent protection effects against inflammation. However, its insolubility and short half-life limit its clinical use. Therefore, we developed a supramolecular system of ε-polylysine (EPL) conjugated by β-cyclodextrin (β-CD) on the side chain, and bilirubin was loaded via host-guest interactions, which resulted in the self-assemble of this system into bilirubin-loaded polylysine-β-cyclodextrin nanoparticle (PB) with improving solubility while reducing toxicity and prolonging medication action time. To explore PB's potential pharmacological mechanisms on OA, we established in vitro and in vivo OA models. PB exerted ROS-scavenging proficiency and anti-apoptotic effects on rat chondrocytes by activating the Nrf2-HO-1/GPX4 signaling pathway. Additionally, PB reprogrammed the cartilage microenvironment by regulating the NF-κB signaling pathway to maintain chondrocyte function. Animal experiments further confirmed that PB had excellent scavenging ability for ROS and inflammatory factors related to charge adsorption with cartilage as well as long retention ability. Together, this work suggests that PB has superior protective abilities with beneficial effects on OA, indicating its great potential for intervention therapy targeting chondrocytes.
Collapse
Affiliation(s)
- Xinyu Zhao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Huirong Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China
| | - Xinyu Jiang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China
| | - Shimin Zheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China
| | - Chenyu Qiu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China
| | - Yingfeng Cheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China
| | - Yinhao Lin
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China
| | - Yunzhi Wang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China
| | - Yuqi Yan
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China
| | - Xinyu Di
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China
| | - Miyun Hu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Wanling Zhu
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Fugen Wu
- Department of Pediatric, The First People's Hospital of Wenling, Taizhou, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China.
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China.
| |
Collapse
|
6
|
Sundar S, Koopman A, Manzoni TJ, Xie W, Bhatti QUA, Lo CY, Damani VS, Yang AN, Pochan D, Parreno J, Engiles JB, Kayser LV, Dhong C. Kinetics and Retention of Polystyrenesulfonate for Proteoglycan Replacement in Cartilage. Biomacromolecules 2024; 25:5819-5833. [PMID: 39142342 PMCID: PMC11389691 DOI: 10.1021/acs.biomac.4c00479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
Tissue hydration provides articular cartilage with dynamic viscoelastic properties. Early stage osteoarthritis (OA) is marked by loss of proteoglycans and glycosaminoglycans (GAG), lowering fixed charge density, and impairing tissue osmotic function. The most common GAG replacement, chondroitin sulfate (CS), has failed to show effectiveness. Here, we investigated a synthetic polyelectrolyte, poly(styrenesulfonate) (PSS), both as a model compound to investigate polyelectrolyte transport in cartilage, and as a potential candidate to restore bulk fixed charge density in cartilage with GAG loss. Through bovine explants and histology, we determined zonal-based effective diffusion coefficients for three different molecular weights of PSS. Compared to CS, PSS was retained longer in GAG-depleted cartilage in static and compression-based desorption experiments. We explained enhanced solute performance of PSS by its more compact morphology and higher charge density by small-angle X-ray scattering. This study may improve design of GAG mimetic molecules for repairing osmotic function in OA cartilage.
Collapse
Affiliation(s)
- Shalini Sundar
- Department
of Biomedical Engineering, University of
Delaware, Newark 19716, Delaware, United States
| | - Allison Koopman
- Department
of Materials Science and Engineering, University
of Delaware, Newark 19716, Delaware, United States
| | - Thomas J. Manzoni
- Department
of Biological Sciences, University of Delaware, Newark 19716, Delaware, United States
| | - Weiran Xie
- Department
of Materials Science and Engineering, University
of Delaware, Newark 19716, Delaware, United States
| | - Qurat-Ul-Ain Bhatti
- Department
of Materials Science and Engineering, University
of Delaware, Newark 19716, Delaware, United States
| | - Chun-Yuan Lo
- Department
of Chemistry and Biochemistry, University
of Delaware, Newark 19716, Delaware, United States
| | - Vidhika S. Damani
- Department
of Materials Science and Engineering, University
of Delaware, Newark 19716, Delaware, United States
| | - Ai Nin Yang
- Department
of Chemistry and Biochemistry, University
of Delaware, Newark 19716, Delaware, United States
| | - Darrin Pochan
- Department
of Materials Science and Engineering, University
of Delaware, Newark 19716, Delaware, United States
| | - Justin Parreno
- Department
of Biomedical Engineering, University of
Delaware, Newark 19716, Delaware, United States
- Department
of Biological Sciences, University of Delaware, Newark 19716, Delaware, United States
| | - Julie B. Engiles
- Department
of Clinical Studies, University of Pennsylvania
School of Veterinary Medicine, Kennett Square 19348, Pennsylvania, United States
- Department
of Pathobiology, University of Pennsylvania
School of Veterinary Medicine, Kennett Square, Pennsylvania 19348, United States
| | - Laure V. Kayser
- Department
of Materials Science and Engineering, University
of Delaware, Newark 19716, Delaware, United States
- Department
of Chemistry and Biochemistry, University
of Delaware, Newark 19716, Delaware, United States
| | - Charles Dhong
- Department
of Biomedical Engineering, University of
Delaware, Newark 19716, Delaware, United States
- Department
of Materials Science and Engineering, University
of Delaware, Newark 19716, Delaware, United States
| |
Collapse
|
7
|
Li X, Sheng S, Li G, Hu Y, Zhou F, Geng Z, Su J. Research Progress in Hydrogels for Cartilage Organoids. Adv Healthc Mater 2024; 13:e2400431. [PMID: 38768997 DOI: 10.1002/adhm.202400431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/29/2024] [Indexed: 05/22/2024]
Abstract
The repair and regeneration of cartilage has always been a hot topic in medical research. Cartilage organoids (CORGs) are special cartilage tissue created using tissue engineering techniques outside the body. These engineered organoids tissues provide models that simulate the complex biological functions of cartilage, opening new possibilities for cartilage regenerative medicine and treatment strategies. However, it is crucial to establish suitable matrix scaffolds for the cultivation of CORGs. In recent years, utilizing hydrogel to culture stem cells and induce their differentiation into chondrocytes has emerged as a promising method for the in vitro construction of CORGs. In this review, the methods for establishing CORGs are summarized and an overview of the advantages and limitations of using matrigel in the cultivation of such organoids is provided. Furthermore, the importance of cartilage tissue ECM and alternative hydrogel substitutes for Matrigel, such as alginate, peptides, silk fibroin, and DNA derivatives is discussed, and the pros and cons of using these hydrogels for the cultivation of CORGs are outlined. Finally, the challenges and future directions in hydrogel research for CORGs are discussed. It is hoped that this article provides valuable references for the design and development of hydrogels for CORGs.
Collapse
Affiliation(s)
- Xiaolong Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics and Traumatology, Nanning Hospital of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530000, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Yan Hu
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
8
|
Tuppurainen J, Paakkari P, Jäntti J, Nissinen MT, Fugazzola MC, van Weeren R, Ylisiurua S, Nieminen MT, Kröger H, Snyder BD, Joenathan A, Grinstaff MW, Matikka H, Korhonen RK, Mäkelä JTA. Revealing Detailed Cartilage Function Through Nanoparticle Diffusion Imaging: A Computed Tomography & Finite Element Study. Ann Biomed Eng 2024; 52:2584-2595. [PMID: 39012563 PMCID: PMC11329549 DOI: 10.1007/s10439-024-03552-7] [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: 01/12/2024] [Accepted: 05/23/2024] [Indexed: 07/17/2024]
Abstract
The ability of articular cartilage to withstand significant mechanical stresses during activities, such as walking or running, relies on its distinctive structure. Integrating detailed tissue properties into subject-specific biomechanical models is challenging due to the complexity of analyzing these characteristics. This limitation compromises the accuracy of models in replicating cartilage function and impacts predictive capabilities. To address this, methods revealing cartilage function at the constituent-specific level are essential. In this study, we demonstrated that computational modeling derived individual constituent-specific biomechanical properties could be predicted by a novel nanoparticle contrast-enhanced computer tomography (CECT) method. We imaged articular cartilage samples collected from the equine stifle joint (n = 60) using contrast-enhanced micro-computed tomography (µCECT) to determine contrast agents' intake within the samples, and compared those to cartilage functional properties, derived from a fibril-reinforced poroelastic finite element model. Two distinct imaging techniques were investigated: conventional energy-integrating µCECT employing a cationic tantalum oxide nanoparticle (Ta2O5-cNP) contrast agent and novel photon-counting µCECT utilizing a dual-contrast agent, comprising Ta2O5-cNP and neutral iodixanol. The results demonstrate the capacity to evaluate fibrillar and non-fibrillar functionality of cartilage, along with permeability-affected fluid flow in cartilage. This finding indicates the feasibility of incorporating these specific functional properties into biomechanical computational models, holding potential for personalized approaches to cartilage diagnostics and treatment.
Collapse
Affiliation(s)
- Juuso Tuppurainen
- Department of Technical Physics, University of Eastern Finland, POB 1627, 70211, Kuopio, Finland.
- Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland.
| | - Petri Paakkari
- Department of Technical Physics, University of Eastern Finland, POB 1627, 70211, Kuopio, Finland
- Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Jiri Jäntti
- Department of Technical Physics, University of Eastern Finland, POB 1627, 70211, Kuopio, Finland
- Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Mikko T Nissinen
- Department of Technical Physics, University of Eastern Finland, POB 1627, 70211, Kuopio, Finland
| | - Maria C Fugazzola
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - René van Weeren
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Sampo Ylisiurua
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Miika T Nieminen
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Heikki Kröger
- Department of Orthopaedics and Traumatology, Kuopio University Hospital, Kuopio, Finland
- Kuopio Musculoskeletal Research Unit, University of Eastern Finland, Kuopio, Finland
| | - Brian D Snyder
- Department of Orthopedic Surgery, Boston Children's Hospital, Boston, USA
| | - Anisha Joenathan
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, USA
| | - Mark W Grinstaff
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, USA
| | - Hanna Matikka
- Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Rami K Korhonen
- Department of Technical Physics, University of Eastern Finland, POB 1627, 70211, Kuopio, Finland
| | - Janne T A Mäkelä
- Department of Technical Physics, University of Eastern Finland, POB 1627, 70211, Kuopio, Finland
- Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
9
|
Mei H, Sha C, Lv Q, Liu H, Jiang L, Song Q, Zeng Y, Zhou J, Zheng Y, Zhong W, Zhou J, Li J. Multifunctional polymeric nanocapsules with enhanced cartilage penetration and retention for osteoarthritis treatment. J Control Release 2024; 374:466-477. [PMID: 39179111 DOI: 10.1016/j.jconrel.2024.08.031] [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: 05/28/2024] [Revised: 08/10/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024]
Abstract
Osteoarthritis (OA) is a prevalent joint disease characterized by cartilage degeneration and subchondral bone homeostasis imbalance. Effective topical OA therapy is challenging, as therapeutic drugs often suffer from insufficient penetration and rapid clearance. We develop miniature polydopamine (PDA) nanocapsules (sub-60 nm), which are conjugated with collagen-binding polypeptide (CBP) and loaded with an anabolic drug (i.e., parathyroid hormone 1-34, PTH 1-34) for efficient OA treatment. Such multifunctional polymeric nanocapsules, denoted as PDA@CBP-PTH, possess deformability when interacting with the dense collagen fiber networks, enabling the efficient penetration into 1 mm cartilage in 4 h and prolonged retention within the joints up to 28 days. Moreover, PDA@CBP-PTH nanocapsules exhibit excellent reactive oxygen species scavenging property in chondrocytes and enhance the anabolism in subchondral bone. The nanosystem, as dual-mode treatment for OA, demonstrates rapid penetration, long-lasting effects, and combinational therapeutic impact, paving the way for reversing the progression of OA for joint health care.
Collapse
Affiliation(s)
- Hongxiang Mei
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chuanlu Sha
- College of Biomass Science and Engineering, Key Laboratory of Leather Chemistry, Engineering of Ministry of Education, National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu 610065, China
| | - Qinyi Lv
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hai Liu
- College of Biomass Science and Engineering, Key Laboratory of Leather Chemistry, Engineering of Ministry of Education, National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu 610065, China
| | - Linli Jiang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qiantao Song
- College of Biomass Science and Engineering, Key Laboratory of Leather Chemistry, Engineering of Ministry of Education, National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu 610065, China
| | - Yiwei Zeng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jiawei Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yule Zheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenbin Zhong
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Jiajing Zhou
- College of Biomass Science and Engineering, Key Laboratory of Leather Chemistry, Engineering of Ministry of Education, National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu 610065, China.
| | - Juan Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
10
|
Millán Cotto HA, Pathrikar TV, Hakim B, Baby HM, Zhang H, Zhao P, Ansaripour R, Amini R, Carrier RL, Bajpayee AG. Cationic-motif-modified exosomes for mRNA delivery to retinal photoreceptors. J Mater Chem B 2024; 12:7384-7400. [PMID: 38946491 PMCID: PMC11323772 DOI: 10.1039/d4tb00849a] [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] [Indexed: 07/02/2024]
Abstract
Topical treatment of vitreoretinal diseases remains a challenge due to slow corneal uptake and systemic clearance. Exosomes are emerging nanocarriers for drug delivery due to biocompatibility and cellular targeting properties. To apply them for retinal targeting via the topical route, exosomes must traverse various ocular barriers including the cornea, lens, vitreous humor (VH), and the retina itself. Here we engineered high-purity milk-derived exosomes by anchoring arginine-rich cationic motifs via PEG2000 lipid insertion on their surface. Modification enabled exosomes to use weak-reversible electrostatic interactions with anionic glycosaminoglycan (GAG) and water content of the tissue to enhance their transport rate and retention. Addition of cationic motifs neutralized the anionic surface charge of exosomes (-24 to -2 mV) without impacting size or morphology. Cationic-motif-modified exosomes exhibited two-fold faster steady state diffusivity through bovine corneas compared to unmodified exosomes. Fluorescence recovery after photobleaching confirmed that cationic-motif-modified exosomes can diffuse through VH without steric hindrance. In healthy VH, cationic-motif-modified exosomes demonstrated stronger binding resulting in three-fold lower average diffusivity that enhanced by six-fold in 50% GAG-depleted VH recapitulating advanced liquefaction. Cationic-motif-modified exosomes penetrated through the full-thickness of porcine retinal explants resulting in ten-fold higher uptake in photoreceptors and three-fold greater transfection with encapsulated eGFP mRNA compared to unmodified exosomes. Cationic-motif-modified exosomes are safe to use as they did not adversely affect the mechanical swelling properties of the cornea or lens nor impact retinal cell viability. Cationic-motif-modified exosomes, therefore, offer themselves as a cell-free nanocarrier platform for gene delivery to retinal photoreceptors potentially via the topical route.
Collapse
Affiliation(s)
| | | | - Bill Hakim
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Helna M Baby
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Peng Zhao
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | - Ronak Ansaripour
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| | - Rouzbeh Amini
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, USA
| | - Rebecca L Carrier
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
11
|
Weber P, Asadikorayem M, Zenobi-Wong M. Zwitterionic Poly-Carboxybetaine Polymers Restore Lubrication of Inflamed Articular Cartilage. Adv Healthc Mater 2024:e2401623. [PMID: 39007282 DOI: 10.1002/adhm.202401623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/02/2024] [Indexed: 07/16/2024]
Abstract
Osteoarthritis is a degenerative joint disease that is associated with decreased synovial fluid viscosity and increased cartilage friction. Though viscosupplements are available for decades, their clinical efficacy is limited and there is ample need for more effective joint lubricants. This study first evaluates the tribological and biochemical properties of bovine articular cartilage explants after stimulation with the inflammatory cytokine interleukin-1β. This model is then used to investigate the tribological potential of carboxybetaine (CBAA)-based zwitterionic polymers of linear and bottlebrush architecture. Due to their affinity for cartilage tissue, these polymers form a highly hydrated surface layer that decreases friction under high load in the boundary lubrication regime. For linear pCBAA, these benefits are retained over several weeks and the relaxation time of cartilage explants under compression is furthermore decreased, thereby potentially boosting the weeping lubrication mechanism. Bottlebrush bb-pCBAA shows smaller benefits under boundary lubrication but is more viscous than linear pCBAA, therefore providing better lubrication under low load in the fluid-film regime and enabling a longer residence time to bind to the cartilage surface. Showing how CBAA-based polymers restore the lost lubrication mechanisms during inflammation can inspire the next steps toward more effective joint lubricants in the future.
Collapse
Affiliation(s)
- Patrick Weber
- Tissue Engineering + Biofabrication Laboratory, ETH Zurich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Maryam Asadikorayem
- Tissue Engineering + Biofabrication Laboratory, ETH Zurich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, ETH Zurich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| |
Collapse
|
12
|
Yi X, Leng P, Wang S, Liu L, Xie B. Functional Nanomaterials for the Treatment of Osteoarthritis. Int J Nanomedicine 2024; 19:6731-6756. [PMID: 38979531 PMCID: PMC11230134 DOI: 10.2147/ijn.s465243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/15/2024] [Indexed: 07/10/2024] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease, affecting more than 595 million people worldwide. Nanomaterials possess superior physicochemical properties and can influence pathological processes due to their unique structural features, such as size, surface interface, and photoelectromagnetic thermal effects. Unlike traditional OA treatments, which suffer from short half-life, low stability, poor bioavailability, and high systemic toxicity, nanotherapeutic strategies for OA offer longer half-life, enhanced targeting, improved bioavailability, and reduced systemic toxicity. These advantages effectively address the limitations of traditional therapies. This review aims to inspire researchers to develop more multifunctional nanomaterials and promote their practical application in OA treatment.
Collapse
Affiliation(s)
- Xinyue Yi
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, People’s Republic of China
| | - Pengyuan Leng
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| | - Supeng Wang
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, People’s Republic of China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| | - Bingju Xie
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
13
|
Saeedi T, Prokopovich P. Screening of poly-beta amino ester coated emulsion of ketorolac for cartilage delivery. J Mater Chem B 2024; 12:5930-5939. [PMID: 38804519 PMCID: PMC11186506 DOI: 10.1039/d4tb00313f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Osteoarthritis (OA) is a prevalent chronic health condition necessitating effective treatment strategies. Globally, there were 86 million people with incident knee osteoarthritis in 2020. Pain management remains the primary approach to OA as the nature of cartilage poses challenges for drug delivery. An emulsion-based delivery system, using a class of positively charged and hydrolysable polymers (poly-beta-amino-esters) to coat oil droplets containing drugs, has been shown to enhance and prolong drug localization in ex vivo cartilage models. As the properties of the polymers used in this technology strongly depend on the monomers used in the synthesis, this study presents the screening of a wide range of PBAEs as droplet coating agents and using ketorolac as a model of nonsteroidal anti-inflammatory drugs. The emulsions prepared with this PBAE library were characterized, and drug localisation and retention were evaluated in both native and glycosaminoglycan (GAG) depleted cartilage ex vivo models. Optimal candidates were identified and tested in an ex vivo model showing the ability to protect chondrocyte cell viability and increase both GAG and collagen contents in cartilage exposed to cytokine (IL-1α) simulating acute cartilage damage. This work demonstrates the potential of PBAE coated emulsion as a delivery system for effective drug delivery in OA treatment.
Collapse
Affiliation(s)
- Tahani Saeedi
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff, UK.
| | | |
Collapse
|
14
|
Ma T, Xu G, Gao T, Zhao G, Huang G, Shi J, Chen J, Song J, Xia J, Ma X. Engineered Exosomes with ATF5-Modified mRNA Loaded in Injectable Thermogels Alleviate Osteoarthritis by Targeting the Mitochondrial Unfolded Protein Response. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21383-21399. [PMID: 38626424 DOI: 10.1021/acsami.3c17209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Osteoarthritis (OA) progression is highly associated with chondrocyte mitochondrial dysfunction and disorders of catabolism and anabolism of the extracellular matrix (ECM) in the articular cartilage. The mitochondrial unfolded protein response (UPRmt), which is an integral component of the mitochondrial quality control (MQC) system, is essential for maintaining chondrocyte homeostasis. We successfully validated the pivotal role of activating transcription factor 5 (ATF5) in upregulating the UPRmt, mitigating IL-1β-induced inflammation and mitochondrial dysfunction, and promoting balanced metabolism in articular cartilage ECM, proving its potential as a promising therapeutic target for OA. Modified mRNAs (modRNAs) have emerged as novel and efficient gene delivery vectors for nucleic acid therapeutic approaches. In this study, we combined Atf5-modRNA (modAtf5) with engineered exosomes derived from bone mesenchymal stem cells (ExmodAtf5) to exert cytoprotective effects on chondrocytes in articular cartilage via Atf5. However, the rapid localized metabolization of ExmodAtf5 limits its application. PLGA-PEG-PLGA (Gel), an injectable thermosensitive hydrogel, was used as a carrier of ExmodAtf5 (Gel@ExmodAtf5) to achieve a sustained release of ExmodAtf5. In vitro and in vivo, the use of Gel@ExmodAtf5 was shown to be a highly effective strategy for OA treatment. The in vivo therapeutic effect of Gel@ExmodAtf5 was evidenced by the preservation of the intact cartilage surface, low OARSI scores, fewer osteophytes, and mild subchondral bone sclerosis and cystic degeneration. Consequently, the combination of ExmodAtf5 and PLGA-PEG-PLGA could significantly enhance the therapeutic efficacy and prolong the exosome release. In addition, the mitochondrial protease ClpP enhanced chondrocyte autophagy by modulating the mTOR/Ulk1 pathway. As a result of our research, Gel@ExmodAtf5 can be considered to be effective at alleviating the progression of OA.
Collapse
Affiliation(s)
- Tiancong Ma
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Guangyu Xu
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Tian Gao
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Guanglei Zhao
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Gangyong Huang
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jingsheng Shi
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jie Chen
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jian Song
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jun Xia
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Xiaosheng Ma
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| |
Collapse
|
15
|
Yang R, Chu H, Yue H, Mishina Y, Zhang Z, Liu H, Li B. BMP signaling maintains auricular chondrocyte identity and prevents microtia development by inhibiting protein kinase A. eLife 2024; 12:RP91883. [PMID: 38690987 PMCID: PMC11062634 DOI: 10.7554/elife.91883] [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] [Indexed: 05/03/2024] Open
Abstract
Elastic cartilage constitutes a major component of the external ear, which functions to guide sound to the middle and inner ears. Defects in auricle development cause congenital microtia, which affects hearing and appearance in patients. Mutations in several genes have been implicated in microtia development, yet, the pathogenesis of this disorder remains incompletely understood. Here, we show that Prrx1 genetically marks auricular chondrocytes in adult mice. Interestingly, BMP-Smad1/5/9 signaling in chondrocytes is increasingly activated from the proximal to distal segments of the ear, which is associated with a decrease in chondrocyte regenerative activity. Ablation of Bmpr1a in auricular chondrocytes led to chondrocyte atrophy and microtia development at the distal part. Transcriptome analysis revealed that Bmpr1a deficiency caused a switch from the chondrogenic program to the osteogenic program, accompanied by enhanced protein kinase A activation, likely through increased expression of Adcy5/8. Inhibition of PKA blocked chondrocyte-to-osteoblast transformation and microtia development. Moreover, analysis of single-cell RNA-seq of human microtia samples uncovered enriched gene expression in the PKA pathway and chondrocyte-to-osteoblast transformation process. These findings suggest that auricle cartilage is actively maintained by BMP signaling, which maintains chondrocyte identity by suppressing osteogenic differentiation.
Collapse
Affiliation(s)
- Ruichen Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong UniversityShanghaiChina
| | - Hongshang Chu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong UniversityShanghaiChina
| | - Hua Yue
- Department of Osteoporosis and Bone Diseases, Shanghai Clinical Research Center of Bone Disease, Shanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Yuji Mishina
- Department of Biologic and Materials & Prosthodontics, University of Michigan School of DentistryAnn ArborUnited States
| | - Zhenlin Zhang
- Department of Osteoporosis and Bone Diseases, Shanghai Clinical Research Center of Bone Disease, Shanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong UniversityShanghaiChina
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong UniversityShanghaiChina
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghaiChina
| |
Collapse
|
16
|
Zhou H, Zhang Z, Mu Y, Yao H, Zhang Y, Wang DA. Harnessing Nanomedicine for Cartilage Repair: Design Considerations and Recent Advances in Biomaterials. ACS NANO 2024; 18:10667-10687. [PMID: 38592060 DOI: 10.1021/acsnano.4c00780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Cartilage injuries are escalating worldwide, particularly in aging society. Given its limited self-healing ability, the repair and regeneration of damaged articular cartilage remain formidable challenges. To address this issue, nanomaterials are leveraged to achieve desirable repair outcomes by enhancing mechanical properties, optimizing drug loading and bioavailability, enabling site-specific and targeted delivery, and orchestrating cell activities at the nanoscale. This review presents a comprehensive survey of recent research in nanomedicine for cartilage repair, with a primary focus on biomaterial design considerations and recent advances. The review commences with an introductory overview of the intricate cartilage microenvironment and further delves into key biomaterial design parameters crucial for treating cartilage damage, including microstructure, surface charge, and active targeting. The focal point of this review lies in recent advances in nano drug delivery systems and nanotechnology-enabled 3D matrices for cartilage repair. We discuss the compositions and properties of these nanomaterials and elucidate how these materials impact the regeneration of damaged cartilage. This review underscores the pivotal role of nanotechnology in improving the efficacy of biomaterials utilized for the treatment of cartilage damage.
Collapse
Affiliation(s)
- Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| | - Yi Zhang
- School of Integrated Circuit Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
- Center for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, Hong Kong SAR 999077, China
| |
Collapse
|
17
|
Schwartz G, Best TM, Chen CB, Travascio F, Jackson AR. Assessing the role of surface layer and molecular probe size in diffusion within meniscus tissue. PLoS One 2024; 19:e0301432. [PMID: 38626169 PMCID: PMC11020779 DOI: 10.1371/journal.pone.0301432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/15/2024] [Indexed: 04/18/2024] Open
Abstract
Diffusion within extracellular matrix is essential to deliver nutrients and larger metabolites to the avascular region of the meniscus. It is well known that both structure and composition of the meniscus vary across its regions; therefore, it is crucial to fully understand how the heterogenous meniscal architecture affects its diffusive properties. The objective of this study was to investigate the effect of meniscal region (core tissue, femoral, and tibial surface layers) and molecular weight on the diffusivity of several molecules in porcine meniscus. Tissue samples were harvested from the central area of porcine lateral menisci. Diffusivity of fluorescein (MW 332 Da) and three fluorescence-labeled dextrans (MW 3k, 40k, and 150k Da) was measured via fluorescence recovery after photobleaching. Diffusivity was affected by molecular size, decreasing as the Stokes' radius of the solute increased. There was no significant effect of meniscal region on diffusivity for fluorescein, 3k and 40k dextrans (p>0.05). However, region did significantly affect the diffusivity of 150k Dextran, with that in the tibial surface layer being larger than in the core region (p = 0.001). Our findings contribute novel knowledge concerning the transport properties of the meniscus fibrocartilage. This data can be used to advance the understanding of tissue pathophysiology and explore effective approaches for tissue restoration.
Collapse
Affiliation(s)
- Gabi Schwartz
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States of America
| | - Thomas M. Best
- Department of Orthopaedic Surgery, University of Miami, Miami, FL, United States of America
- UHealth Sports Medicine Institute, Coral Gables, FL, United States of America
| | - Cheng-Bang Chen
- Department of Industrial and Systems Engineering, University of Miami, Coral Gables, FL, United States of America
| | - Francesco Travascio
- Department of Orthopaedic Surgery, University of Miami, Miami, FL, United States of America
- Department of Mechanical and Aerospace Engineering, University of Miami, Coral Gables, FL, United States of America
- Max Biedermann Institute for Biomechanics at Mount Sinai Medical Center, Miami Beach, FL, United States of America
| | - Alicia R. Jackson
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States of America
| |
Collapse
|
18
|
Chen M, Lu Y, Liu Y, Liu Q, Deng S, Liu Y, Cui X, Liang J, Zhang X, Fan Y, Wang Q. Injectable Microgels with Hybrid Exosomes of Chondrocyte-Targeted FGF18 Gene-Editing and Self-Renewable Lubrication for Osteoarthritis Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312559. [PMID: 38266145 DOI: 10.1002/adma.202312559] [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: 11/22/2023] [Revised: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Abnormal silencing of fibroblast growth factor (FGF) signaling significantly contributes to joint dysplasia and osteoarthritis (OA); However, the clinical translation of FGF18-based protein drugs is hindered by their short half-life, low delivery efficiency and the need for repeated articular injections. This study proposes a CRISPR/Cas9-based approach to effectively activate the FGF18 gene of OA chondrocytes at the genome level in vivo, using chondrocyte-affinity peptide (CAP) incorporated hybrid exosomes (CAP/FGF18-hyEXO) loaded with an FGF18-targeted gene-editing tool. Furthermore, CAP/FGF18-hyEXO are encapsulated in methacrylic anhydride-modified hyaluronic (HAMA) hydrogel microspheres via microfluidics and photopolymerization to create an injectable microgel system (CAP/FGF18-hyEXO@HMs) with self-renewable hydration layers to provide persistent lubrication in response to frictional wear. Together, the injectable CAP/FGF18-hyEXO@HMs, combined with in vivo FGF18 gene editing and continuous lubrication, have demonstrated their capacity to synergistically promote cartilage regeneration, decrease inflammation, and prevent ECM degradation both in vitro and in vivo, holding great potential for clinical translation.
Collapse
Affiliation(s)
- Manyu Chen
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yan Lu
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yuhan Liu
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, P. R. China
| | - Quanying Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Siyan Deng
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yuan Liu
- Orthopedics Research Institute, Department of Orthopedics, West China Hospital Sichuan University, Chengdu, 610041, P. R. China
| | - Xiaolin Cui
- School of medicine the Chinese University of Hong Kong, Shenzhen, 518172, P. R. China
- Department of Orthopedic Surgery & Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine University of Otago, Christchurch, 8140, New Zealand
| | - Jie Liang
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
- College of Biomedical Engineering Sichuan University, 29 Wangjiang Road, Chengdu, 610064, P. R. China
| |
Collapse
|
19
|
Nhan J, Strebel N, Virah Sawmy K, Yin J, St-Pierre JP. Characterization of Calcium- and Strontium-Polyphosphate Particles Toward Drug Delivery into Articular Cartilage. Macromol Biosci 2024; 24:e2300345. [PMID: 37777870 DOI: 10.1002/mabi.202300345] [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: 07/27/2023] [Revised: 09/21/2023] [Indexed: 10/02/2023]
Abstract
Drug delivery into articular cartilage poses many challenges due in part to its lack of vasculature. While intra-articular injections are effective for the local administration of drugs, small molecules are rapidly cleared from the synovial fluid. As such, there is a need to develop effective drug delivery strategies to improve the residence times of bioactive molecules in the joint and elicit a sustained therapeutic effect. In this study, calcium- and strontium-polyphosphate particles are synthesized and characterized as potential drug carriers into articular cartilage. Physicochemical characterization reveals that the particles exhibit a spherical morphology, have a negative zeta potential, and are nanoscale in size. Biological characterization in chondrocytes confirms cellular uptake of the particles and demonstrates both size and concentration-dependent cytotoxicity at high concentrations. Furthermore, treatment of chondrocytes with these particles results in a reduction in cell proliferation and metabolic activity, confirming biological effects. Finally, incubation with cartilage tissue explants suggests successful uptake, despite the particles exhibiting a negative surface charge. Therefore, from the results of this study, these polyphosphate-based particles have potential as a drug carrier into articular cartilage and warrant further development.
Collapse
Affiliation(s)
- Jordan Nhan
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Nicolas Strebel
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Khushnouma Virah Sawmy
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Jordan Yin
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| | - Jean-Philippe St-Pierre
- Department of Chemical and Biological Engineering, Faculty of Engineering, University of Ottawa, 161 Louis-Pasteur Pvt., Ottawa, Ontario, K1N 6N5, Canada
| |
Collapse
|
20
|
Deng R, Zhao R, Zhang Z, Chen Y, Yang M, Lin Y, Ye J, Li N, Qin H, Yan X, Shi J, Yuan F, Song S, Xu Z, Song Y, Fu J, Xu B, Nie G, Yu JK. Chondrocyte membrane-coated nanoparticles promote drug retention and halt cartilage damage in rat and canine osteoarthritis. Sci Transl Med 2024; 16:eadh9751. [PMID: 38381849 DOI: 10.1126/scitranslmed.adh9751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024]
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by progressive degeneration of articular cartilage. A challenge in the development of disease-modifying drugs is effective delivery to chondrocytes. The unique structure of the joint promotes rapid clearance of drugs through synovial fluid, and the dense and avascular cartilage extracellular matrix (ECM) limits drug penetration. Here, we show that poly(lactide-co-glycolic acid) nanoparticles coated in chondrocyte membranes (CM-NPs) were preferentially taken up by rat chondrocytes ex vivo compared with uncoated nanoparticles. Internalization of the CM-NPs was mediated primarily by E-cadherin, clathrin-mediated endocytosis, and micropinocytosis. These CM-NPs adhered to the cartilage ECM in rat knee joints in vivo and penetrated deeply into the cartilage matrix with a residence time of more than 34 days. Simulated synovial fluid clearance studies showed that CM-NPs loaded with a Wnt pathway inhibitor, adavivint (CM-NPs-Ada), delayed the catabolic metabolism of rat and human chondrocytes and cartilage explants under inflammatory conditions. In a surgical model of rat OA, drug-loaded CM-NPs effectively restored gait, attenuated periarticular bone remodeling, and provided chondroprotection against cartilage degeneration. OA progression was also mitigated by CM-NPs-Ada in a canine model of anterior cruciate ligament transection. These results demonstrate the feasibility of using chondrocyte membrane-coated nanoparticles to improve the pharmacokinetics and efficacy of anti-OA drugs.
Collapse
Affiliation(s)
- Ronghui Deng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zining Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yang Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Meng Yang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yixuan Lin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jing Ye
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Nan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xin Yan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Fuzhen Yuan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Shitang Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Zijie Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yifan Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jiangnan Fu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Bingbing Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jia-Kuo Yu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
- Orthopedic Sports Medicine Center, Beijing Tsinghua Changgung Hospital, Affiliated Hospital of Tsinghua University, Beijing 102218, P. R. China
| |
Collapse
|
21
|
Zuo G, Zhuang P, Yang X, Jia Q, Cai Z, Qi J, Deng L, Zhou Z, Cui W, Xiao J. Regulating Chondro-Bone Metabolism for Treatment of Osteoarthritis via High-Permeability Micro/Nano Hydrogel Microspheres. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305023. [PMID: 38084002 PMCID: PMC10837371 DOI: 10.1002/advs.202305023] [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: 07/22/2023] [Revised: 11/13/2023] [Indexed: 02/04/2024]
Abstract
Destruction of cartilage due to the abnormal remodeling of subchondral bone (SB) leads to osteoarthritis (OA), and restoring chondro-bone metabolic homeostasis is the key to the treatment of OA. However, traditional intra-articular injections for the treatment of OA cannot directly break through the cartilage barrier to reach SB. In this study, the hydrothermal method is used to synthesize ultra-small size (≈5 nm) selenium-doped carbon quantum dots (Se-CQDs, SC), which conjugated with triphenylphosphine (TPP) to create TPP-Se-CQDs (SCT). Further, SCT is dynamically complexed with hyaluronic acid modified with aldehyde and methacrylic anhydride (AHAMA) to construct highly permeable micro/nano hydrogel microspheres (SCT@AHAMA) for restoring chondro-bone metabolic homeostasis. In vitro experiments confirmed that the selenium atoms scavenged reactive oxygen species (ROS) from the mitochondria of mononuclear macrophages, inhibited osteoclast differentiation and function, and suppressed early chondrocyte apoptosis to maintain a balance between cartilage matrix synthesis and catabolism. In vivo experiments further demonstrated that the delivery system inhibited osteoclastogenesis and H-vessel invasion, thereby regulating the initiation and process of abnormal bone remodeling and inhibiting cartilage degeneration in SB. In conclusion, the micro/nano hydrogel microspheres based on ultra-small quantum dots facilitate the efficient penetration of articular SB and regulate chondro-bone metabolism for OA treatment.
Collapse
Affiliation(s)
- Guilai Zuo
- School of Health Science and EngineeringUniversity of Shanghai for Science and TechnologyShanghai200093P. R. China
- Department of Orthopaedic OncologyChangzheng HospitalNaval Military Medical UniversityShanghai200003P. R. China
- Department of Bone TumorThe Affiliated Hospital of Qingdao UniversityNo. 59, Haier RoadQingdaoShandong266000P. R. China
| | - Pengzhen Zhuang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
- Pharmaceutical Sciences LaboratoryFaculty of Science and EngineeringÅbo Akademi UniversityTurku20520Finland
| | - Xinghai Yang
- Department of Orthopaedic OncologyChangzheng HospitalNaval Military Medical UniversityShanghai200003P. R. China
| | - Qi Jia
- Department of Orthopaedic OncologyChangzheng HospitalNaval Military Medical UniversityShanghai200003P. R. China
| | - Zhengwei Cai
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Jin Qi
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Lianfu Deng
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Zhenhua Zhou
- Department of Orthopaedic OncologyChangzheng HospitalNaval Military Medical UniversityShanghai200003P. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Jianru Xiao
- School of Health Science and EngineeringUniversity of Shanghai for Science and TechnologyShanghai200093P. R. China
- Department of Orthopaedic OncologyChangzheng HospitalNaval Military Medical UniversityShanghai200003P. R. China
| |
Collapse
|
22
|
Mailhiot SE, Tolkkinen K, Henschel H, Mareš J, Hanni M, Nieminen MT, Telkki VV. Melting of aqueous NaCl solutions in porous materials: shifted phase transition distribution (SIDI) approach for determining NMR cryoporometry pore size distributions. Phys Chem Chem Phys 2024; 26:3441-3450. [PMID: 38205817 DOI: 10.1039/d3cp04029a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Nuclear magnetic resonance cryoporometry (NMRC) and differential scanning calorimetry thermoporometry (DSC-TPM) are powerful methods for measuring mesopore size distributions. The methods are based on the fact that, according to the Gibbs-Thomson equation, the melting point depression of a liquid confined to a pore is inversely proportional to the pore size. However, aqueous salt solutions, which inherently exist in a broad range of biological porous materials as well as technological applications such as electrolytes, do not melt at a single temperature. This causes artefacts in the pore size distributions extracted by traditional Gibbs-Thomson analysis of NMRC and DSC-TPM data. Bulk aqueous NaCl solutions are known to have a broad distribution of melting points between the eutectic and pure water phase transition points (252-273 K). Here, we hypothesize that, when aqueous NaCl solution (saline) is confined to a small pore, the whole melting point distribution is shifted toward lower temperatures by the value predicted by the Gibbs-Thomson equation. We show that this so-called shifted phase transition distribution (SIDI) approach removes the artefacts arising from the traditional Gibbs-Thomson analysis and gives correct pore size distributions for saline saturated mesoporous silica gel and controlled pore materials analyzed by NMR cryoporometry. Furthermore, we demonstrate that the method can be used for determining pore sizes in collagen-chondroitin sulphate hydrogels resembling the composition of the extracellular matrix of articular cartilage. It is straightforward to apply the SIDI analysis for DSC-TMP data as well.
Collapse
Affiliation(s)
| | | | - Henning Henschel
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jiří Mareš
- NMR Research Unit, University of Oulu, Oulu, Finland.
| | - Matti Hanni
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Miika T Nieminen
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | | |
Collapse
|
23
|
Brackin RB, McColgan GE, Pucha SA, Kowalski MA, Drissi H, Doan TN, Patel JM. Improved Cartilage Protection with Low Molecular Weight Hyaluronic Acid Hydrogel. Bioengineering (Basel) 2023; 10:1013. [PMID: 37760116 PMCID: PMC10525634 DOI: 10.3390/bioengineering10091013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/13/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
Traumatic joint injuries are common, leading to progressive tissue degeneration and the development of osteoarthritis. The post-traumatic joint experiences a pro-inflammatory milieu, initiating a subtle but deteriorative process in cartilage tissue. To prevent or even reverse this process, our group previously developed a tissue-penetrating methacrylated hyaluronic acid (MeHA) hydrogel system, crosslinked within cartilage to restore and/or protect the tissue. In the current study, we further optimized this approach by investigating the impact of biomaterial molecular weight (MW; 20, 75, 100 kDa) on its integration within and reinforcement of cartilage, as well as its ability to protect tissue degradation in a catabolic state. Indeed, the low MW MeHA integrated and reinforced cartilage tissue better than the high MW counterparts. Furthermore, in a 2 week IL-1β explant culture model, the 20 kDa MeHA demonstrated the most protection from biphasic mechanical loss, best retention of proteoglycans (Safranin O staining), and least aggrecan breakdown (NITEGE). Thus, the lower MW MeHA gels integrated better into the tissue and provided the greatest protection of the cartilage matrix. Future work will test this formulation in a preclinical model, with the goal of translating this therapeutic approach for cartilage preservation.
Collapse
Affiliation(s)
- Riley B. Brackin
- Atlanta VA Medical Center, Decatur, GA 30033, USA
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Gail E. McColgan
- Atlanta VA Medical Center, Decatur, GA 30033, USA
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Saitheja A. Pucha
- Atlanta VA Medical Center, Decatur, GA 30033, USA
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Michael A. Kowalski
- Atlanta VA Medical Center, Decatur, GA 30033, USA
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Hicham Drissi
- Atlanta VA Medical Center, Decatur, GA 30033, USA
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Thanh N. Doan
- Atlanta VA Medical Center, Decatur, GA 30033, USA
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Jay M. Patel
- Atlanta VA Medical Center, Decatur, GA 30033, USA
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30329, USA
| |
Collapse
|
24
|
Aldrich JL, Panicker A, Ovalle R, Sharma B. Drug Delivery Strategies and Nanozyme Technologies to Overcome Limitations for Targeting Oxidative Stress in Osteoarthritis. Pharmaceuticals (Basel) 2023; 16:1044. [PMID: 37513955 PMCID: PMC10383173 DOI: 10.3390/ph16071044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/26/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Oxidative stress is an important, but elusive, therapeutic target for osteoarthritis (OA). Antioxidant strategies that target oxidative stress through the elimination of reactive oxygen species (ROS) have been widely evaluated for OA but are limited by the physiological characteristics of the joint. Current hallmarks in antioxidant treatment strategies include poor bioavailability, poor stability, and poor retention in the joint. For example, oral intake of exogenous antioxidants has limited access to the joint space, and intra-articular injections require frequent dosing to provide therapeutic effects. Advancements in ROS-scavenging nanomaterials, also known as nanozymes, leverage bioactive material properties to improve delivery and retention. Material properties of nanozymes can be tuned to overcome physiological barriers in the knee. However, the clinical application of these nanozymes is still limited, and studies to understand their utility in treating OA are still in their infancy. The objective of this review is to evaluate current antioxidant treatment strategies and the development of nanozymes as a potential alternative to conventional small molecules and enzymes.
Collapse
Affiliation(s)
| | | | | | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; (J.L.A.)
| |
Collapse
|
25
|
Morejon A, Schwartz G, Best TM, Travascio F, Jackson AR. Effect of molecular weight and tissue layer on solute partitioning in the knee meniscus. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100360. [PMID: 37122844 PMCID: PMC10133802 DOI: 10.1016/j.ocarto.2023.100360] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
Objective Knee meniscus tissue is partly vascularized, meaning that nutrients must be transported through the extracellular matrix of the avascular portion to reach resident cells. Similarly, drugs used as therapeutic agents to treat meniscal pathologies rely on transport through the tissue. The driving force of diffusive transport is the gradient of concentration, which depends on molecular solubility. The meniscus is organized into a core region sandwiched between the tibial and femoral superficial layers. Structural differences exist across meniscal regions; therefore, regional differences in solubility are also hypothesized. Methods Samples from the core, tibial and femoral layers were obtained from 5 medial and 5 lateral porcine menisci. The partition coefficient (K) of fluorescein, 3 kDa and 40 kDa dextrans in the layers of the meniscus was measured using an equilibration experiment. The effect of meniscal compartment, layer, and solute molecular weight on K was analyzed using a three-way ANOVA. Results K ranged from a high of ∼2.9 in fluorescein to a low of ∼0.1 in 40 kDa dextran and was inversely related to the solute molecular weight across all tissue regions. Tissue layer only had a significant effect on partitioning of 40k Dex solute, which was lower in the tibial surface layer relative to the core (p = 0.032). Conclusion This study provides insight into depth-dependent partitioning in the meniscus, indicating the limiting effect of the meniscus superficial layer on solubility increases with solute molecular size. This illustrates how the surface layers could potentially reduce the effectiveness of drug delivery therapies incorporating large molecules (>40 kDa).
Collapse
Affiliation(s)
- Andy Morejon
- Department of Mechanical and Aerospace Engineering, University of Miami, Coral Gables, FL, USA
| | - Gabi Schwartz
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
| | - Thomas M. Best
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
- Department of Orthopedic Surgery, University of Miami, Coral Gables, FL, USA
- UHealth Sports Medicine Institute, Coral Gables, FL, USA
| | - Francesco Travascio
- Department of Mechanical and Aerospace Engineering, University of Miami, Coral Gables, FL, USA
- Department of Orthopedic Surgery, University of Miami, Coral Gables, FL, USA
- Max Biedermann Institute for Biomechanics at Mount Sinai Medical Center, Miami Beach, FL, USA
- Corresponding author. College of Engineering, University of Miami, 1251 Memorial Drive, MEB 276, Coral Gables, FL 33146, USA.
| | - Alicia R. Jackson
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
- Corresponding author. College of Engineering, University of Miami, 1251 Memorial Drive, MEA 219, Coral Gables, FL 33146 USA.
| |
Collapse
|
26
|
Debnath K, Heras KL, Rivera A, Lenzini S, Shin JW. Extracellular vesicle-matrix interactions. NATURE REVIEWS. MATERIALS 2023; 8:390-402. [PMID: 38463907 PMCID: PMC10919209 DOI: 10.1038/s41578-023-00551-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/24/2023] [Indexed: 03/12/2024]
Abstract
The extracellular matrix in microenvironments harbors a variety of signals to control cellular functions and the materiality of tissues. Most efforts to synthetically reconstitute the matrix by biomaterial design have focused on decoupling cell-secreted and polymer-based cues. Cells package molecules into nanoscale lipid membrane-bound extracellular vesicles and secrete them. Thus, extracellular vesicles inherently interact with the meshwork of the extracellular matrix. In this Review, we discuss various aspects of extracellular vesicle-matrix interactions. Cells receive feedback from the extracellular matrix and leverage intracellular processes to control the biogenesis of extracellular vesicles. Once secreted, various biomolecular and biophysical factors determine whether extracellular vesicles are locally incorporated into the matrix or transported out of the matrix to be taken up by other cells or deposited into tissues at a distal location. These insights can be utilized to develop engineered biomaterials where EV release and retention can be precisely controlled in host tissue to elicit various biological and therapeutic outcomes.
Collapse
Affiliation(s)
- Koushik Debnath
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Kevin Las Heras
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy (UPV/EHU)
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Ambar Rivera
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60608, USA
| | - Stephen Lenzini
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Jae-Won Shin
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
27
|
Ajayi TO, Liu S, Rosen C, Rinaldi-Ramos CM, Allen KD, Sharma B. Application of magnetic particle imaging to evaluate nanoparticle fate in rodent joints. J Control Release 2023; 356:347-359. [PMID: 36868518 DOI: 10.1016/j.jconrel.2023.02.038] [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: 10/20/2022] [Revised: 02/16/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023]
Abstract
Nanoparticles are a promising approach for improving intra-articular drug delivery and tissue targeting. However, techniques to non-invasively track and quantify their concentration in vivo are limited, resulting in an inadequate understanding of their retention, clearance, and biodistribution in the joint. Currently, fluorescence imaging is often used to track nanoparticle fate in animal models; however, this approach has limitations that impede long-term quantitative assessment of nanoparticles over time. The goal of this work was to evaluate an emerging imaging modality, magnetic particle imaging (MPI), for intra-articular tracking of nanoparticles. MPI provides 3D visualization and depth-independent quantification of superparamagnetic iron oxide nanoparticle (SPION) tracers. Here, we developed and characterized a polymer-based magnetic nanoparticle system incorporated with SPION tracers and cartilage targeting properties. MPI was then used to longitudinally assess nanoparticle fate after intra-articular injection. Magnetic nanoparticles were injected into the joints of healthy mice, and evaluated for nanoparticle retention, biodistribution, and clearance over 6 weeks using MPI. In parallel, the fate of fluorescently tagged nanoparticles was tracked using in vivo fluorescence imaging. The study was concluded at day 42, and MPI and fluorescence imaging demonstrated different profiles in nanoparticle retention and clearance from the joint. MPI signal was persistent over the study duration, suggesting NP retention of at least 42 days, much longer than the 14 days observed based on fluorescence signal. These data suggest that the type of tracer - SPIONs or fluorophores - and modality of imaging can affect interpretation of nanoparticle fate in the joint. Given that understanding particle fate over time is paramount for attaining insights about therapeutic profiles in vivo, our data suggest MPI may yield a quantitative and robust method to non-invasively track nanoparticles following intra-articular injection on an extended timeline.
Collapse
Affiliation(s)
- Tolulope O Ajayi
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Sitong Liu
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - Chelsea Rosen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Carlos M Rinaldi-Ramos
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - Kyle D Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
28
|
Karami P, Stampoultzis T, Guo Y, Pioletti DP. A guide to preclinical evaluation of hydrogel-based devices for treatment of cartilage lesions. Acta Biomater 2023; 158:12-31. [PMID: 36638938 DOI: 10.1016/j.actbio.2023.01.015] [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: 08/30/2022] [Revised: 12/19/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023]
Abstract
The drive to develop cartilage implants for the treatment of major defects in the musculoskeletal system has resulted in a major research thrust towards developing biomaterial devices for cartilage repair. Investigational devices for the restoration of articular cartilage are considered as significant risk materials by regulatory bodies and therefore proof of efficacy and safety prior to clinical testing represents a critical phase of the multidisciplinary effort to bridge the gap between bench and bedside. To date, review articles have thoroughly covered different scientific facets of cartilage engineering paradigm, but surprisingly, little attention has been given to the preclinical considerations revolving around the validation of a biomaterial implant. Considering hydrogel-based cartilage products as an example, the present review endeavors to provide a summary of the critical prerequisites that such devices should meet for cartilage repair, for successful implantation and subsequent preclinical validation prior to clinical trials. Considerations pertaining to the choice of appropriate animal model, characterization techniques for the quantitative and qualitative outcome measures, as well as concerns with respect to GLP practices are also extensively discussed. This article is not meant to provide a systematic review, but rather to introduce a device validation-based roadmap to the academic investigator, in anticipation of future healthcare commercialization. STATEMENT OF SIGNIFICANCE: There are significant challenges around translation of in vitro cartilage repair strategies to approved therapies. New biomaterial-based devices must undergo exhaustive investigations to ensure their safety and efficacy prior to clinical trials. These considerations are required to be applied from early developmental stages. Although there are numerous research works on cartilage devices and their in vivo evaluations, little attention has been given into the preclinical pathway and the corresponding approval processes. With a focus on hydrogel devices to concretely illustrate the preclinical path, this review paper intends to highlight the various considerations regarding the preclinical validation of hydrogel devices for cartilage repair, from regulatory considerations, to implantation strategies, device performance aspects and characterizations.
Collapse
Affiliation(s)
- Peyman Karami
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
| | - Theofanis Stampoultzis
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
| | - Yanheng Guo
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
| | - Dominique P Pioletti
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland.
| |
Collapse
|
29
|
Fahey M, Bennett M, Thomas M, Montney K, Vivancos-Koopman I, Pugliese B, Browning L, Bonassar LJ, Delco M. Mesenchymal stromal cells donate mitochondria to articular chondrocytes exposed to mitochondrial, environmental, and mechanical stress. Sci Rep 2022; 12:21525. [PMID: 36513773 PMCID: PMC9747781 DOI: 10.1038/s41598-022-25844-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
Abstract
Articular cartilage has limited healing capacity and no drugs are available that can prevent or slow the development of osteoarthritis (OA) after joint injury. Mesenchymal stromal cell (MSC)-based regenerative therapies for OA are increasingly common, but questions regarding their mechanisms of action remain. Our group recently reported that although cartilage is avascular and relatively metabolically quiescent, injury induces chondrocyte mitochondrial dysfunction, driving cartilage degradation and OA. MSCs are known to rescue injured cells and improve healing by donating healthy mitochondria in highly metabolic tissues, but mitochondrial transfer has not been investigated in cartilage. Here, we demonstrate that MSCs transfer mitochondria to stressed chondrocytes in cell culture and in injured cartilage tissue. Conditions known to induce chondrocyte mitochondrial dysfunction, including stimulation with rotenone/antimycin and hyperoxia, increased transfer. MSC-chondrocyte mitochondrial transfer was blocked by non-specific and specific (connexin-43) gap-junction inhibition. When exposed to mechanically injured cartilage, MSCs localized to areas of matrix damage and extended cellular processes deep into microcracks, delivering mitochondria to chondrocytes. This work provides insights into the chemical, environmental, and mechanical conditions that can elicit MSC-chondrocyte mitochondrial transfer in vitro and in situ, and our findings suggest a new potential role for MSC-based therapeutics after cartilage injury.
Collapse
Affiliation(s)
- Megan Fahey
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Maureen Bennett
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Matthew Thomas
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Kaylee Montney
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Irene Vivancos-Koopman
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Brenna Pugliese
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Lindsay Browning
- Meinig School of Biomedical Engineering, Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Michelle Delco
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
30
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
31
|
Wang Y, Liu L, Le Z, Tay A. Analysis of Nanomedicine Efficacy for Osteoarthritis. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Yuwen Wang
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Ling Liu
- Institute of Health Innovation and Technology National University of Singapore Singapore 117599 Singapore
| | - Zhicheng Le
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Andy Tay
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
- Institute of Health Innovation and Technology National University of Singapore Singapore 117599 Singapore
- Tissue Engineering Programme National University of Singapore Singapore 117510 Singapore
| |
Collapse
|
32
|
Carboxymethyl chitosan-assisted MnOx nanoparticles: Synthesis, characterization, detection and cartilage repair in early osteoarthritis. Carbohydr Polym 2022; 294:119821. [DOI: 10.1016/j.carbpol.2022.119821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 12/24/2022]
|
33
|
Warren MR, Vedadghavami A, Bhagavatula S, Bajpayee AG. Effects of polycationic drug carriers on the electromechanical and swelling properties of cartilage. Biophys J 2022; 121:3542-3561. [PMID: 35765244 PMCID: PMC9515003 DOI: 10.1016/j.bpj.2022.06.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 11/15/2022] Open
Abstract
Cationic nanocarriers offer a promising solution to challenges in delivering drugs to negatively charged connective tissues, such as to articular cartilage for the treatment of osteoarthritis (OA). However, little is known about the effects that cationic macromolecules may have on the mechanical properties of cartilage at high interstitial concentrations. We utilized arginine-rich cationic peptide carriers (CPCs) with varying net charge (from +8 to +20) to investigate the biophysical mechanisms of nanocarrier-induced alterations to cartilage biomechanical properties. We observed that CPCs increased the compressive modulus of healthy bovine cartilage explants by up to 70% and decreased the stiffness of glycosaminoglycan-depleted tissues (modeling OA) by 69%; in both cases, the magnitude of the change in stiffness correlated with the uptake of CPC charge variants. Next, we directly measured CPC-induced osmotic deswelling in cartilage tissue due to shielding of charge repulsions between anionic extracellular matrix constituents, with magnitudes of reductions between 36 and 64 kPa. We then demonstrated that electrostatic interactions were required for CPC-induced stiffening to occur, evidenced by no observed increase in tissue stiffness when measured in hypertonic bathing salinity. We applied a non-ideal Donnan osmotic model (under triphasic theory) to separate bulk modulus measurements into Donnan and non-Donnan components, which further demonstrated the conflicting charge-shielding and matrix-stiffening effects of CPCs. These results show that cationic drug carriers can alter tissue mechanical properties via multiple mechanisms, including the expected charge shielding as well as a novel stiffening phenomenon mediated by physical linkages. We introduce a model for how the magnitudes of these mechanical changes depend on tunable physical properties of the drug carrier, including net charge, size, and spatial charge distribution. We envision that the results and theory presented herein will inform the design of future cationic drug-delivery systems intended to treat diseases in a wide range of connective tissues.
Collapse
Affiliation(s)
- Matthew R Warren
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Armin Vedadghavami
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Sanjana Bhagavatula
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Ambika G Bajpayee
- Department of Bioengineering, Northeastern University, Boston, Massachusetts; Department of Mechanical Engineering, Northeastern University, Boston, Massachusetts.
| |
Collapse
|
34
|
Kwak JS, Lee Y, Yang J, Kim SK, Shin Y, Kim HJ, Choi JH, Im YJ, Kim MJ, Lee Yu K, Chang You J, Chun JS. Characterization of rhodanine derivatives as potential disease-modifying drugs for experimental mouse osteoarthritis. Osteoarthritis Cartilage 2022; 30:1210-1221. [PMID: 35513246 DOI: 10.1016/j.joca.2022.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study was performed to characterize selected rhodanine derivatives as potential preclinical disease-modifying drugs for experimental osteoarthritis (OA) in mice. METHODS Three rhodanine derivatives, designated rhodanine (R)-501, R-502, and R-503, were selected as candidate OA disease-modifying drugs. Their effects were evaluated by intra-articular (IA) injection in OA mouse models induced by DMM (destabilization of the medial meniscus) or adenoviral overexpression in joint tissues of hypoxia-inducible factor (HIF)-2α or zinc importer ZIP8. The regulatory mechanisms impacted by the rhodanine derivatives were examined in primary-culture chondrocytes and fibroblast-like synoviocytes (FLS). RESULTS All three rhodanine derivatives inhibited OA development caused by DMM or overexpression of HIF-2α or ZIP8. Compared to vehicle-treated group, for example, IA injection of R-501 in DMM-operated mice reduced median OARSI grade from 3.78 (IQR 3.00-5.00) to 1.89 (IQR 0.94-2.00, P = 0.0001). R-502 and R-503 also reduced from 3.67 (IQR 2.11-4.56) to 2.00 (IQR 1.00-2.00, P = 0.0030) and 2.00 (IQR 1.83-2.67, P = 0.0378), respectively. Mechanistically, the rhodanine derivatives inhibited the nuclear localization and transcriptional activity of HIF-2α in chondrocytes and FLS. They did not bind to Zn2+ or modulate Zn2+ homeostasis in chondrocytes or FLS; instead, they inhibited the nuclear localization and transcriptional activity of the Zn2+-dependent transcription factor, MTF1. HIF-2α, ZIP8, and interleukin-1β could upregulate matrix-degrading enzymes in chondrocytes and FLS, and the rhodanine derivatives inhibited these effects. CONCLUSION IA administration of rhodanine derivatives significantly reduced OA pathogenesis in various mouse models, demonstrating that these derivatives have disease-modifying therapeutic potential against OA pathogenesis.
Collapse
Affiliation(s)
- J-S Kwak
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Y Lee
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - J Yang
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - S K Kim
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Y Shin
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - H-J Kim
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - J H Choi
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Y J Im
- College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - M-J Kim
- Avixgen Inc., Seoul, 06649, Republic of Korea
| | - K Lee Yu
- National Research Laboratory for Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - J Chang You
- Avixgen Inc., Seoul, 06649, Republic of Korea; National Research Laboratory for Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - J-S Chun
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
35
|
Li Y, Wu H, Li Z, Li B, Zhu M, Chen D, Ye F, Yu B, Huang Y. Species variation in the cartilaginous endplate of the lumbar intervertebral disc. JOR Spine 2022; 5:e1218. [PMID: 36203863 PMCID: PMC9520767 DOI: 10.1002/jsp2.1218] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/06/2022] Open
Abstract
Backgrounds Cartilaginous endplate (CEP) plays an essential role in intervertebral disc (IVD) health and disease. The aim was to compare the CEP structure of lumbar IVD and to reveal the detailed pattern of integration between the CEP and bony endplate (BEP) from different species. Methods A total of 34 IVDs (5 human, 5 goat, 8 pig, 8 rabbit, and 8 rat IVDs) were collected, fixed and midsagittally cut; in each IVD, one-half was used for histological staining to observe the CEP morphology, and the other half was used for scanning electron microscopy (SEM) analysis to measure the diameters and distributions of collagen fibers in the central and peripheral CEP areas and to observe the pattern of CEP-BEP integration from different species. Results The human, pig, goat, and rabbit IVDs had the typical BEP-CEP structure, but the rat CEP was directly connected with the growth plate. Human CEP was the thickest (896.95 ± 87.71 μm) among these species, followed by pig, goat, rat, and rabbit CEPs. Additionally, the mean cellular density of the rabbit CEP was the highest, which was 930 ± 202 per mm2, followed by the rat, goat, pig, and human CEPs. In all the species, the collagen fiber diameter in the peripheral area was much bigger than that in the central area. The collagen fiber diameters of CEP from the human, pig, goat, and rat were distributed between 35 nm and 65 nm. The BEP and CEP were connected by the collagen from the CEP, aggregating into bundles or cross links with each other to form a network, and anchored to BEP. Conclusions Significant differences in the thickness, cellular density, and collagen characterization of CEPs from different species were demonstrated; the integration of BEP-CEP in humans, pigs, goats, and rabbits was mainly achieved by the collagen bundles anchoring system, while the typical BEP-CEP interface did not exist in rats.
Collapse
Affiliation(s)
- Yun‐He Li
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine SurgeryPeking University Shenzhen HospitalShenzhenChina
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic BiomaterialsPeking University Shenzhen HospitalShenzhenChina
| | - Hai‐Long Wu
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine SurgeryPeking University Shenzhen HospitalShenzhenChina
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic BiomaterialsPeking University Shenzhen HospitalShenzhenChina
| | - Zhen Li
- AO Research Institute DavosDavosSwitzerland
| | - Bin‐Bin Li
- Department of Human Anatomy & HistoembryologyHangzhou Normal UniversityHangzhouChina
| | - Man Zhu
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine SurgeryPeking University Shenzhen HospitalShenzhenChina
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic BiomaterialsPeking University Shenzhen HospitalShenzhenChina
| | - Di Chen
- Research Center for Computer‐aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Fei‐Hong Ye
- Hangzhou Zhigu Research Center for Tissue Engineering and Regenerative MedicineHangzhouChina
| | - Bin‐Sheng Yu
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine SurgeryPeking University Shenzhen HospitalShenzhenChina
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic BiomaterialsPeking University Shenzhen HospitalShenzhenChina
- Institute of Orthopaedics, Peking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenChina
| | - Yong‐Can Huang
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine SurgeryPeking University Shenzhen HospitalShenzhenChina
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic BiomaterialsPeking University Shenzhen HospitalShenzhenChina
- Institute of Orthopaedics, Peking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenChina
| |
Collapse
|
36
|
Hall ME, Wang AS, Gold GE, Levenston ME. Contrast solution properties and scan parameters influence the apparent diffusivity of computed tomography contrast agents in articular cartilage. J R Soc Interface 2022; 19:20220403. [PMID: 35919981 PMCID: PMC9346352 DOI: 10.1098/rsif.2022.0403] [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: 05/27/2022] [Accepted: 07/08/2022] [Indexed: 11/12/2022] Open
Abstract
The inability to detect early degenerative changes to the articular cartilage surface that commonly precede bulk osteoarthritic degradation is an obstacle to early disease detection for research or clinical diagnosis. Leveraging a known artefact that blurs tissue boundaries in clinical arthrograms, contrast agent (CA) diffusivity can be derived from computed tomography arthrography (CTa) scans. We combined experimental and computational approaches to study protocol variations that may alter the CTa-derived apparent diffusivity. In experimental studies on bovine cartilage explants, we examined how CA dilution and transport direction (absorption versus desorption) influence the apparent diffusivity of untreated and enzymatically digested cartilage. Using multiphysics simulations, we examined mechanisms underlying experimental observations and the effects of image resolution, scan interval and early scan termination. The apparent diffusivity during absorption decreased with increasing CA concentration by an amount similar to the increase induced by tissue digestion. Models indicated that osmotically-induced fluid efflux strongly contributed to the concentration effect. Simulated changes to spatial resolution, scan spacing and total scan time all influenced the apparent diffusivity, indicating the importance of consistent protocols. With careful control of imaging protocols and interpretations guided by transport models, CTa-derived diffusivity offers promise as a biomarker for early degenerative changes.
Collapse
Affiliation(s)
- Mary E. Hall
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Adam S. Wang
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Garry E. Gold
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Marc E. Levenston
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| |
Collapse
|
37
|
Siefen T, Bjerregaard S, Borglin C, Lamprecht A. Assessment of joint pharmacokinetics and consequences for the intraarticular delivery of biologics. J Control Release 2022; 348:745-759. [PMID: 35714731 DOI: 10.1016/j.jconrel.2022.06.015] [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: 04/05/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 01/15/2023]
Abstract
Intraarticular (IA) injections provide the opportunity to deliver biologics directly to their site of action for a local and efficient treatment of osteoarthritis. However, the synovial joint is a challenging site of administration since the drug is rapidly eliminated across the synovial membrane and has limited distribution into cartilage, resulting in unsatisfactory therapeutic efficacy. In order to rationally develop appropriate drug delivery systems, it is essential to thoroughly understand the unique biopharmaceutical environments and kinetics in the joint to adequately simulate them in relevant experimental models. This review presents a detailed view on articular kinetics and drug-tissue interplay of IA administered drugs and summarizes how these can be translated into reasonable formulation strategies by identification of key factors through which the joint residence time can be prolonged and specific structures can be targeted. In this way, pros and cons of the delivery approaches for biologics will be evaluated and the extent to which biorelevant models are applicable to gain mechanistic insights and ameliorate formulation design is discussed.
Collapse
Affiliation(s)
- Tobias Siefen
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany
| | | | | | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; PEPITE (EA4267), University of Burgundy/Franche-Comté, Besançon, France.
| |
Collapse
|
38
|
Housmans BAC, Neefjes M, Surtel DAM, Vitík M, Cremers A, van Rhijn LW, van der Kraan PM, van den Akker GGH, Welting TJM. Synovial fluid from end-stage osteoarthritis induces proliferation and fibrosis of articular chondrocytes via MAPK and RhoGTPase signaling. Osteoarthritis Cartilage 2022; 30:862-874. [PMID: 35176481 DOI: 10.1016/j.joca.2021.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Alterations in the composition of synovial fluid have been associated with adverse effects on cartilage integrity and function. Here, we examined the phenotypic and proliferative behavior of human articular chondrocytes when cultured in vitro for 13 days with synovial fluid derived from end-stage osteoarthritis patients. MATERIALS AND METHODS Chondrocyte proliferation and phenotypical changes induced by osteoarthritic synovial fluid were analyzed using DNA staining, RT-qPCR, immunostainings, and immunoblotting. The molecular mechanisms by which osteoarthritic synovial fluid induced fibrosis and proliferation were studied using a phospho-protein antibody array and luciferase-based transcription factor activity assays. Specific pathway inhibitors were used to probe the involvement of pathways in fibrosis and proliferation. RESULTS Prolonged stimulation with osteoarthritic synovial fluid sustained chondrocyte proliferation and induced profound phenotypic changes, favoring a fibrotic over a chondrogenic or hypertrophic phenotype. A clear loss of chondrogenic markers at both the transcriptional and protein level was observed, while expression of several fibrosis-associated markers were upregulated over time. Phospho-kinase analysis revealed activation of MAPK and RhoGTPase signaling pathways by osteoarthritic synovial fluid, which was confirmed by elevated transcriptional activity of Elk-1 and SRF. Inhibitor studies revealed that ERK played a central role in the loss of chondrocyte phenotype, while EGFR and downstream mediators p38, JNK and Rac/Cdc42 were essential for fibrosis-associated collagen expression. Finally, we identified EGF signaling as a key activator of chondrocyte proliferation. CONCLUSIONS Osteoarthritic synovial fluid promoted chondrocyte fibrosis and proliferation through EGF receptor activation and downstream MAPK and RhoGTPase signaling.
Collapse
Affiliation(s)
- B A C Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - M Neefjes
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - D A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - M Vitík
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - A Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - L W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - P M van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - G G H van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - T J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands; Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
39
|
Monteagudo S, Cornelis FMF, Wang X, de Roover A, Peeters T, Quintiens J, Sermon A, de Almeida RC, Meulenbelt I, Lories RJ. ANP32A represses Wnt signaling across tissues thereby protecting against osteoarthritis and heart disease. Osteoarthritis Cartilage 2022; 30:724-734. [PMID: 35227892 DOI: 10.1016/j.joca.2022.02.615] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/24/2022] [Accepted: 02/20/2022] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To investigate how ANP32A, previously linked to the antioxidant response, regulates Wnt signaling as unraveled by transcriptome analysis of Anp32a-deficient mouse articular cartilage, and its implications for osteoarthritis (OA) and diseases beyond the joint. METHODS Anp32a knockdown chondrogenic ATDC5 cells were cultured in micromasses. Wnt target genes, differentiation markers and matrix deposition were quantified. Wnt target genes were determined in articular cartilage from Anp32a-deficient mice and primary human articular chondrocytes upon ANP32A silencing, using qPCR, luciferase assays and immunohistochemistry. Co-immunoprecipitation, immunofluorescence and chromatin-immunoprecipitation quantitative PCR probed the molecular mechanism via which ANP32A regulates Wnt signaling. Anp32a-deficient mice were subjected to the destabilization of the medial meniscus (DMM) OA model and treated with a Wnt inhibitor and an antioxidant. Severity of OA was assessed by cartilage damage and osteophyte formation. Human Protein Atlas data analysis identified additional organs where ANP32A may regulate Wnt signaling. Wnt target genes were determined in heart and hippocampus from Anp32a-deficient mice, and cardiac hypertrophy and fibrosis quantified. RESULTS Anp32a loss triggered Wnt signaling hyper-activation in articular cartilage. Mechanistically, ANP32A inhibited target gene expression via histone acetylation masking. Wnt antagonist treatment reduced OA severity in Anp32a-deficient mice by preventing osteophyte formation but not cartilage degradation, contrasting with antioxidant treatment. Dual therapy ameliorated more OA features than individual treatments. Anp32a-deficient mice also showed Wnt hyper-activation in the heart, potentially explaining the cardiac hypertrophy phenotype found. CONCLUSIONS ANP32A is a novel translationally relevant repressor of Wnt signaling impacting osteoarthritis and cardiac disease.
Collapse
Affiliation(s)
- S Monteagudo
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| | - F M F Cornelis
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| | - X Wang
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| | - A de Roover
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| | - T Peeters
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| | - J Quintiens
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| | - A Sermon
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| | - R C de Almeida
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, RC Leiden, the Netherlands.
| | - I Meulenbelt
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, RC Leiden, the Netherlands; Integrated Research on Developmental Determinants of Ageing and Longevity (IDEAL), RC Leiden, the Netherlands.
| | - R J Lories
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium; Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
40
|
Tang T, Landis W, Raguin E, Werner P, Bertinetti L, Dean M, Wagermaier W, Fratzl P. A 3D Network of Nanochannels for Possible Ion and Molecule Transit in Mineralizing Bone and Cartilage. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202100162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Tengteng Tang
- Department of Biomaterials Max Planck Institute of Colloids and Interfaces Am Mühlenberg 1 14476 Potsdam Germany
| | - William Landis
- Department of Preventive and Restorative Dental Sciences University of California at San Francisco 707 Parnassus Avenue San Francisco CA 94143 USA
| | - Emeline Raguin
- Department of Biomaterials Max Planck Institute of Colloids and Interfaces Am Mühlenberg 1 14476 Potsdam Germany
| | - Peter Werner
- Department of Biomaterials Max Planck Institute of Colloids and Interfaces Am Mühlenberg 1 14476 Potsdam Germany
| | - Luca Bertinetti
- Center for Molecular Bioengineering TU Dresden Tatzberg 41 01307 Dresden Germany
| | - Mason Dean
- Department of Infectious Diseases and Public Health City University of Hong Kong 31 To Yuen Street, Tat Chee Avenue Kowloon Hong Kong
| | - Wolfgang Wagermaier
- Department of Biomaterials Max Planck Institute of Colloids and Interfaces Am Mühlenberg 1 14476 Potsdam Germany
| | - Peter Fratzl
- Department of Biomaterials Max Planck Institute of Colloids and Interfaces Am Mühlenberg 1 14476 Potsdam Germany
| |
Collapse
|
41
|
Ding X, Gao J, Yu X, Shi J, Chen J, Yu L, Chen S, Ding J. 3D-Printed Porous Scaffolds of Hydrogels Modified with TGF-β1 Binding Peptides to Promote In Vivo Cartilage Regeneration and Animal Gait Restoration. ACS APPLIED MATERIALS & INTERFACES 2022; 14:15982-15995. [PMID: 35363484 DOI: 10.1021/acsami.2c00761] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The treatment of cartilage injury and osteoarthritis has been a classic problem for many years. The idea of in situ tissue regeneration paves a way for osteochondral repair in vivo. Herein, a hydrogel scaffold linked with bioactive peptides that can selectively adsorb transforming growth factor β1 (TGF-β1) was hypothesized to not only afford cell ingrowth space but also induce the endogenous TGF-β1 recruitment for chondrogenesis promotion. In this study, bilayered porous scaffolds with gelatin methacryloyl (GelMA) hydrogels as a matrix were constructed via three-dimensional (3D) printing, of which the upper layer was covalently bound with bioactive peptides that can adsorb TGF-β1 for cartilage repair and the lower layer was blended with hydroxyapatite for subchondral regeneration. The scaffolds showed promising therapeutic efficacy proved by cartilage and osteogenic induction in vitro and osteochondral repair of rats in vivo. In particular, the animal gait behavior was recovered after the in situ tissue regeneration, and the corresponding gait analysis demonstrated the promotion of tissue regeneration induced by the porous hydrogels with the binding peptides.
Collapse
Affiliation(s)
- Xiaoquan Ding
- Department of Sports Medicine, Huashan Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200040, China
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jingming Gao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Xiaoye Yu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Jiayue Shi
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Jun Chen
- Department of Sports Medicine, Huashan Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200040, China
| | - Lin Yu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200040, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200438, China
| |
Collapse
|
42
|
Huang H, Lou Z, Zheng S, Wu J, Yao Q, Chen R, Kou L, Chen D. Intra-articular drug delivery systems for osteoarthritis therapy: shifting from sustained release to enhancing penetration into cartilage. Drug Deliv 2022; 29:767-791. [PMID: 35261301 PMCID: PMC8920370 DOI: 10.1080/10717544.2022.2048130] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a progressive chronic inflammation that leads to cartilage degeneration. OA Patients are commonly given pharmacological treatment, but the available treatments are not sufficiently effective. The development of sustained-release drug delivery systems (DDSs) for OA may be an attractive strategy to prevent rapid drug clearance and improve the half-life of a drug at the joint cavity. Such delivery systems will improve the therapeutic effects of anti-inflammatory effects in the joint cavity. Whereas, for disease-modifying OA drugs (DMOADs) which target chondrocytes or act on mesenchymal stem cells (MSCs), the cartilage-permeable DDSs are required to maximize their efficacy. This review provides an overview of joint structure in healthy and pathological conditions, introduces the advances of the sustained-release DDSs and the permeable DDSs, and discusses the rational design of the permeable DDSs for OA treatment. We hope that the ideas generated in this review will promote the development of effective OA drugs in the future.
Collapse
Affiliation(s)
- Huirong Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zijian Lou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shimin Zheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianing Wu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Daosen Chen
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
43
|
Structural Characterization Analyses of Low Brass Filler Biomaterial for Hard Tissue Implanted Scaffold Applications. MATERIALS 2022; 15:ma15041421. [PMID: 35207962 PMCID: PMC8875846 DOI: 10.3390/ma15041421] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/30/2022] [Accepted: 02/07/2022] [Indexed: 01/27/2023]
Abstract
A biomaterial was created for hard tissue implanted scaffolds as a translational therapeutic approach. The existing biomaterials containing titanium dioxide filler posed a risk of oxygen gas vacancy. This will block the canaliculars, leading to a limit on the nutrient fluid supply. To overcome this problem, low brass was used as an alternative filler to eliminate the gas vacancy. Low brass with composition percentages of 0%, 2%, 5%, 15%, and 30% was filled into the polyester urethane liquidusing the metallic filler polymer reinforced method. The structural characterizations of the low brass filler biomaterial were investigated by Field Emission Scanning Electron Microscopy. The results showed the surface membrane strength was higher than the side and cross-section. The composition shapes found were hexagon for polyester urethane and peanut for low brass. Low brass stabilised polyester urethane in biomaterials by the formation of two 5-ringed tetrahedral crystal structures. The average pore diameter was 308.9 nm, which is suitable for articular cartilage cells. The pore distribution was quite dispersed, and its curve had a linear relationship between area and diameter, suggestive of the sphere-shaped pores. The average porosities were different between using FESEM results of 6.04% and the calculated result of 3.28%. In conclusion, this biomaterial had a higher surface membrane strength and rather homogeneous dispersed pore structures.
Collapse
|
44
|
Rohanifar M, Johnston BB, Davis AL, Guang Y, Nommensen K, Fitzpatrick JA, Pham CN, Setton LA. Hydraulic permeability and compressive properties of porcine and human synovium. Biophys J 2022; 121:575-581. [PMID: 35032457 PMCID: PMC8874024 DOI: 10.1016/j.bpj.2022.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/21/2021] [Accepted: 01/11/2022] [Indexed: 11/02/2022] Open
Abstract
The synovium is a multilayer connective tissue separating the intra-articular spaces of the diarthrodial joint from the extra-synovial vascular and lymphatic supply. Synovium regulates drug transport into and out of the joint, yet its material properties remain poorly characterized. Here, we measured the compressive properties (aggregate modulus, Young's modulus, and Poisson's ratio) and hydraulic permeability of synovium with a combined experimental-computational approach. A compressive aggregate modulus and Young's modulus for the solid phase of synovium were quantified from linear regression of the equilibrium confined and unconfined compressive stress upon strain, respectively (HA = 4.3 ± 2.0 kPa, Es = 2.1 ± 0.75, porcine; HA = 3.1 ± 2.0 kPa, Es = 2.8 ± 1.7, human). Poisson's ratio was estimated to be 0.39 and 0.40 for porcine and human tissue, respectively, from moduli values in a Monte Carlo simulation. To calculate hydraulic permeability, a biphasic finite element model's predictions were numerically matched to experimental data for the time-varying ramp and hold phase of a single increment of applied strain (k = 7.4 ± 4.1 × 10-15 m4/N.s, porcine; k = 7.4 ± 4.3 × 10-15 m4/N.s, human). We can use these newly measured properties to predict fluid flow gradients across the tissue in response to previously reported intra-articular pressures. These values for material constants are to our knowledge the first available measurements in synovium that are necessary to better understand drug transport in both healthy and pathological joints.
Collapse
Affiliation(s)
- Milad Rohanifar
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Benjamin B. Johnston
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Alexandra L. Davis
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Young Guang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Kayla Nommensen
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - James A.J. Fitzpatrick
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri,Department of Neuroscience and Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Christine N. Pham
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, Missouri
| | - Lori A. Setton
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri,Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri,Corresponding author
| |
Collapse
|
45
|
Kupratis ME, Gure AE, Benson JM, Ortved KF, Burris DL, Price C. Comparative tribology II-Measurable biphasic tissue properties have predictable impacts on cartilage rehydration and lubricity. Acta Biomater 2022; 138:375-389. [PMID: 34728427 DOI: 10.1016/j.actbio.2021.10.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022]
Abstract
Healthy articular cartilage supports load bearing and frictional properties unmatched among biological tissues and man-made bearing materials. Balancing fluid exudation and recovery under loaded and articulated conditions is essential to the tissue's biological and mechanical longevity. Our prior tribological investigations, which leveraged the convergent stationary contact area (cSCA) configuration, revealed that sliding alone can modulate cartilage interstitial fluid pressurization and the recovery and maintenance of lubrication under load through a mechanism termed 'tribological rehydration.' Our recent comparative assessment of tribological rehydration revealed remarkably consistent sliding speed-dependent fluid recovery and lubrication behaviors across femoral condyle cartilage from five mammalian species (equine/horse, bovine/cow, porcine/pig, ovine/sheep, and caprine/goat). In the present study, we identified and characterized key predictive relationships among tissue properties, sliding-induced tribological rehydration, and the modulation/recovery of lubrication within healthy articular cartilage. Using correlational analysis, we linked observed speed-dependent tribological rehydration behaviors to cartilage's geometry and biphasic properties (tensile and compressive moduli, and permeability). Together, these findings demonstrate that easily measurable biphasic tissue characteristics (e.g., bulk tissue material properties, compressive strain magnitude, and strain rates) can be used to predict cartilage's rehydration and lubricating abilities, and ultimately its function in vivo. STATEMENT OF SIGNIFICANCE: In healthy cartilage, articulation recovers fluid lost to static loading thereby sustaining tissue lubricity. Osteoarthritis causes changes to cartilage composition, stiffness, and permeability associated with faster fluid exudation and presumably poorer frictional outcomes. Yet, the relationship between mechanical properties and fluid recovery during articulation/sliding remains unclear. Through innovative, high-speed benchtop sliding and indentation experiments, we found that cartilage's tissue properties regulate its exudation/hydration under slow sliding speeds but have minimal effect at high sliding speeds. In fact, cartilage rehydration appears insensitive to permeability and stiffness under high fluid load support conditions. This new understanding of the balance of cartilage exudation and rehydration during activity, based upon comparative tribology studies, may improve prevention and rehabilitation strategies for joint injuries and osteoarthritis.
Collapse
Affiliation(s)
- Meghan E Kupratis
- Biomedical Engineering, University of Delaware, Newark, Delaware, USA
| | - Ahmed E Gure
- Biomedical Engineering, University of Delaware, Newark, Delaware, USA
| | - Jamie M Benson
- Biomedical Engineering, University of Delaware, Newark, Delaware, USA
| | - Kyla F Ortved
- Clinical Studies, New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania, USA
| | - David L Burris
- Biomedical Engineering, University of Delaware, Newark, Delaware, USA; Mechanical Engineering, University of Delaware, Newark, Delaware, USA
| | - Christopher Price
- Biomedical Engineering, University of Delaware, Newark, Delaware, USA; Mechanical Engineering, University of Delaware, Newark, Delaware, USA.
| |
Collapse
|
46
|
Chen P, Liu X, Gu C, Zhong P, Song N, Li M, Dai Z, Fang X, Liu Z, Zhang J, Tang R, Fan S, Lin X. A plant-derived natural photosynthetic system for improving cell anabolism. Nature 2022; 612:546-554. [PMID: 36477541 PMCID: PMC9750875 DOI: 10.1038/s41586-022-05499-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 10/31/2022] [Indexed: 12/12/2022]
Abstract
Insufficient intracellular anabolism is a crucial factor involved in many pathological processes in the body1,2. The anabolism of intracellular substances requires the consumption of sufficient intracellular energy and the production of reducing equivalents. ATP acts as an 'energy currency' for biological processes in cells3,4, and the reduced form of NADPH is a key electron donor that provides reducing power for anabolism5. Under pathological conditions, it is difficult to correct impaired anabolism and to increase insufficient levels of ATP and NADPH to optimum concentrations1,4,6-8. Here we develop an independent and controllable nanosized plant-derived photosynthetic system based on nanothylakoid units (NTUs). To enable cross-species applications, we use a specific mature cell membrane (the chondrocyte membrane (CM)) for camouflage encapsulation. As proof of concept, we demonstrate that these CM-NTUs enter chondrocytes through membrane fusion, avoid lysosome degradation and achieve rapid penetration. Moreover, the CM-NTUs increase intracellular ATP and NADPH levels in situ following exposure to light and improve anabolism in degenerated chondrocytes. They can also systemically correct energy imbalance and restore cellular metabolism to improve cartilage homeostasis and protect against pathological progression of osteoarthritis. Our therapeutic strategy for degenerative diseases is based on a natural photosynthetic system that can controllably enhance cell anabolism by independently providing key energy and metabolic carriers. This study also provides an enhanced understanding of the preparation and application of bioorganisms and composite biomaterials for the treatment of disease.
Collapse
Affiliation(s)
- Pengfei Chen
- grid.13402.340000 0004 1759 700XDepartment of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xin Liu
- grid.13402.340000 0004 1759 700XDepartment of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Chenhui Gu
- grid.13402.340000 0004 1759 700XDepartment of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Peiyu Zhong
- grid.13402.340000 0004 1759 700XDepartment of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Nan Song
- grid.13402.340000 0004 1759 700XDepartment of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Mobai Li
- grid.13402.340000 0004 1759 700XDepartment of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhanqiu Dai
- grid.13402.340000 0004 1759 700XDepartment of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangqian Fang
- grid.13402.340000 0004 1759 700XDepartment of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhaoming Liu
- grid.13402.340000 0004 1759 700XDepartment of Chemistry, Zhejiang University, Hangzhou, China
| | - Jianfeng Zhang
- grid.13402.340000 0004 1759 700XDepartment of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, China.
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Xianfeng Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
47
|
Travascio F, Valladares-Prieto S, Jackson AR. EFFECTS OF SOLUTE SIZE AND TISSUE COMPOSITION ON MOLECULAR AND MACROMOLECULAR DIFFUSIVITY IN HUMAN KNEE CARTILAGE. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 2. [PMID: 34611626 DOI: 10.1016/j.ocarto.2020.100087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Objective Articular cartilage is an avascular tissue. Accordingly, diffusivity represents a fundamental transport mechanism for nutrients and other molecular signals regulating its cell metabolism and maintenance of the extracellular matrix. Understanding how solutes spread into articular cartilage is crucial to elucidating its pathologies, and to designing treatments for repair and restoration of its extracellular matrix. As in other connective tissues, diffusivity in articular cartilage may vary depending both its composition and the specific diffusing solute. Hence, this study investigated the roles of solute size and tissue composition on molecular diffusion in knee articular cartilage. Design FRAP tests were conducted to measure diffusivity of five molecular probes, with size ranging from ~332Da to 70,000Da, in human knee articular cartilage. The measured diffusion coefficients were related to molecular size, as well as water and glycosaminoglycan (GAG) content of femoral and tibial condyle cartilage. Results Diffusivity was affected by molecular size, with the magnitude of the diffusion coefficients decreasing as the Stokes radius of the probe increased. The values of diffusion coefficients in tibial and femoral samples were not significantly different from one another, despite the fact that tibial samples exhibited significantly higher water content and lower GAG content of the femoral specimens. Water content did not affect diffusivity. In contrast, diffusivities of large molecules were sensitive to GAG content. Conclusions This study provides new knowledge on the mechanisms of diffusion in articular cartilage. Our findings can be leveraged to further investigate osteoarthritis and to design treatments for cartilage restoration or replacement.
Collapse
Affiliation(s)
- Francesco Travascio
- Department of Mechanical and Aerospace Engineering, University of Miami, Coral Gables, FL.,Department of Orthopaedic Surgery, University of Miami, Miami, FL.,Max Biedermann Institute for Biomechanics at Mount Sinai Medical Center, Miami Beach, FL
| | | | - Alicia R Jackson
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL
| |
Collapse
|
48
|
Singh YP, Moses JC, Bhardwaj N, Mandal BB. Overcoming the Dependence on Animal Models for Osteoarthritis Therapeutics - The Promises and Prospects of In Vitro Models. Adv Healthc Mater 2021; 10:e2100961. [PMID: 34302436 DOI: 10.1002/adhm.202100961] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/10/2021] [Indexed: 12/19/2022]
Abstract
Osteoarthritis (OA) is a musculoskeletal disease characterized by progressive degeneration of osteochondral tissues. Current treatment is restricted to the reduction of pain and loss of function of the joint. To better comprehend the OA pathophysiological conditions, several models are employed, however; there is no consensus on a suitable model. In this review, different in vitro models being developed for possible therapeutic intervention of OA are outlined. Herein, various in vitro OA models starting from 2D model, co-culture model, 3D models, dynamic culture model to advanced technologies-based models such as 3D bioprinting, bioassembly, organoids, and organ-on-chip-based models are discussed with their advantages and disadvantages. Besides, different growth factors, cytokines, and chemicals being utilized for induction of OA condition are reviewed in detail. Furthermore, there is focus on scrutinizing different molecular and possible therapeutic targets for better understanding the mechanisms and OA therapeutics. Finally, the underlying challenges associated with in vitro models are discussed followed by future prospective. Taken together, a comprehensive overview of in vitro OA models, factors to induce OA-like conditions, and intricate molecular targets with the potential to develop personalized osteoarthritis therapeutics in the future with clinical translation is provided.
Collapse
Affiliation(s)
- Yogendra Pratap Singh
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Joseph Christakiran Moses
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Nandana Bhardwaj
- Department of Science and Mathematics Indian Institute of Information Technology Guwahati Bongora Guwahati Assam 781015 India
| | - Biman B. Mandal
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- Centre for Nanotechnology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- School of Health Sciences and Technology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| |
Collapse
|
49
|
Vernerey FJ, Lalitha Sridhar S, Muralidharan A, Bryant SJ. Mechanics of 3D Cell-Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem Rev 2021; 121:11085-11148. [PMID: 34473466 DOI: 10.1021/acs.chemrev.1c00046] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hydrogels are highly water-swollen molecular networks that are ideal platforms to create tissue mimetics owing to their vast and tunable properties. As such, hydrogels are promising cell-delivery vehicles for applications in tissue engineering and have also emerged as an important base for ex vivo models to study healthy and pathophysiological events in a carefully controlled three-dimensional environment. Cells are readily encapsulated in hydrogels resulting in a plethora of biochemical and mechanical communication mechanisms, which recapitulates the natural cell and extracellular matrix interaction in tissues. These interactions are complex, with multiple events that are invariably coupled and spanning multiple length and time scales. To study and identify the underlying mechanisms involved, an integrated experimental and computational approach is ideally needed. This review discusses the state of our knowledge on cell-hydrogel interactions, with a focus on mechanics and transport, and in this context, highlights recent advancements in experiments, mathematical and computational modeling. The review begins with a background on the thermodynamics and physics fundamentals that govern hydrogel mechanics and transport. The review focuses on two main classes of hydrogels, described as semiflexible polymer networks that represent physically cross-linked fibrous hydrogels and flexible polymer networks representing the chemically cross-linked synthetic and natural hydrogels. In this review, we highlight five main cell-hydrogel interactions that involve key cellular functions related to communication, mechanosensing, migration, growth, and tissue deposition and elaboration. For each of these cellular functions, recent experiments and the most up to date modeling strategies are discussed and then followed by a summary of how to tune hydrogel properties to achieve a desired functional cellular outcome. We conclude with a summary linking these advancements and make the case for the need to integrate experiments and modeling to advance our fundamental understanding of cell-matrix interactions that will ultimately help identify new therapeutic approaches and enable successful tissue engineering.
Collapse
Affiliation(s)
- Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States.,Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Shankar Lalitha Sridhar
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States
| | - Archish Muralidharan
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Stephanie J Bryant
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States.,Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States.,BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States
| |
Collapse
|
50
|
Keppie SJ, Mansfield JC, Tang X, Philp CJ, Graham HK, Önnerfjord P, Wall A, McLean C, Winlove CP, Sherratt MJ, Pavlovskaya GE, Vincent TL. Matrix-Bound Growth Factors are Released upon Cartilage Compression by an Aggrecan-Dependent Sodium Flux that is Lost in Osteoarthritis. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab037. [PMID: 34423304 PMCID: PMC8374957 DOI: 10.1093/function/zqab037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/25/2021] [Accepted: 07/30/2021] [Indexed: 01/07/2023]
Abstract
Articular cartilage is a dense extracellular matrix-rich tissue that degrades following chronic mechanical stress, resulting in osteoarthritis (OA). The tissue has low intrinsic repair especially in aged and osteoarthritic joints. Here, we describe three pro-regenerative factors; fibroblast growth factor 2 (FGF2), connective tissue growth factor, bound to transforming growth factor-beta (CTGF-TGFβ), and hepatoma-derived growth factor (HDGF), that are rapidly released from the pericellular matrix (PCM) of articular cartilage upon mechanical injury. All three growth factors bound heparan sulfate, and were displaced by exogenous NaCl. We hypothesised that sodium, sequestered within the aggrecan-rich matrix, was freed by injurious compression, thereby enhancing the bioavailability of pericellular growth factors. Indeed, growth factor release was abrogated when cartilage aggrecan was depleted by IL-1 treatment, and in severely damaged human osteoarthritic cartilage. A flux in free matrix sodium upon mechanical compression of cartilage was visualised by 23Na -MRI just below the articular surface. This corresponded to a region of reduced tissue stiffness, measured by scanning acoustic microscopy and second harmonic generation microscopy, and where Smad2/3 was phosphorylated upon cyclic compression. Our results describe a novel intrinsic repair mechanism, controlled by matrix stiffness and mediated by the free sodium concentration, in which heparan sulfate-bound growth factors are released from cartilage upon injurious load. They identify aggrecan as a depot for sequestered sodium, explaining why osteoarthritic tissue loses its ability to repair. Treatments that restore matrix sodium to allow appropriate release of growth factors upon load are predicted to enable intrinsic cartilage repair in OA. SIGNIFICANCE STATEMENT Osteoarthritis is the most prevalent musculoskeletal disease, affecting 250 million people worldwide.1 We identify a novel intrinsic repair response in cartilage, mediated by aggrecan-dependent sodium flux, and dependent upon matrix stiffness, which results in the release of a cocktail of pro-regenerative growth factors after injury. Loss of aggrecan in late-stage osteoarthritis prevents growth factor release and likely contributes to disease progression. Treatments that restore matrix sodium in osteoarthritis may recover the intrinsic repair response to improve disease outcome.
Collapse
Affiliation(s)
- Stuart J Keppie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | | | - Xiaodi Tang
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - Christopher J Philp
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, NG7 2QX, UK
| | - Helen K Graham
- School of Biological Sciences, The University of Manchester, Manchester, M13 9PT, UK
| | - Patrik Önnerfjord
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Alanna Wall
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - Celia McLean
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - C Peter Winlove
- School of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK
| | - Michael J Sherratt
- School of Biological Sciences, The University of Manchester, Manchester, M13 9PT, UK
| | - Galina E Pavlovskaya
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, NG7 2QX, UK
| | | |
Collapse
|