1
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2024:e2402737. [PMID: 39506433 DOI: 10.1002/adhm.202402737] [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: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| |
Collapse
|
2
|
Li H, Zhao T, Yuan Z, Gao T, Yang Y, Li R, Tian Q, Tang P, Guo Q, Zhang L. Cartilage lacuna-biomimetic hydrogel microspheres endowed with integrated biological signal boost endogenous articular cartilage regeneration. Bioact Mater 2024; 41:61-82. [PMID: 39104774 PMCID: PMC11299526 DOI: 10.1016/j.bioactmat.2024.06.037] [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: 11/16/2023] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 08/07/2024] Open
Abstract
Despite numerous studies on chondrogenesis, the repair of cartilage-particularly the reconstruction of cartilage lacunae through an all-in-one advanced drug delivery system remains limited. In this study, we developed a cartilage lacuna-like hydrogel microsphere system endowed with integrated biological signals, enabling sequential immunomodulation and endogenous articular cartilage regeneration. We first integrated the chondrogenic growth factor transforming growth factor-β3 (TGF-β3) into mesoporous silica nanoparticles (MSNs). Then, TGF-β3@MSNs and insulin-like growth factor 1 (IGF-1) were encapsulated within microspheres made of polydopamine (pDA). In the final step, growth factor-loaded MSN@pDA and a chitosan (CS) hydrogel containing platelet-derived growth factor-BB (PDGF-BB) were blended to produce growth factors loaded composite microspheres (GFs@μS) using microfluidic technology. The presence of pDA reduced the initial acute inflammatory response, and the early, robust release of PDGF-BB aided in attracting endogenous stem cells. Over the subsequent weeks, the continuous release of IGF-1 and TGF-β3 amplified chondrogenesis and matrix formation. μS were incorporated into an acellular cartilage extracellular matrix (ACECM) and combined with a polydopamine-modified polycaprolactone (PCL) structure to produce a tissue-engineered scaffold that mimicked the structure of the cartilage lacunae evenly distributed in the cartilage matrix, resulting in enhanced cartilage repair and patellar cartilage protection. This research provides a strategic pathway for optimizing growth factor delivery and ensuring prolonged microenvironmental remodeling, leading to efficient articular cartilage regeneration.
Collapse
Affiliation(s)
- Hao Li
- School of Medicine, Nankai University, Tianjin, China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Tianyuan Zhao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
- Department of Orthopaedics, Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
| | - Zhiguo Yuan
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tianze Gao
- School of Medicine, Nankai University, Tianjin, China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
| | - Yongkang Yang
- School of Medicine, Nankai University, Tianjin, China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
| | - Runmeng Li
- School of Medicine, Nankai University, Tianjin, China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
| | - Qinyu Tian
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
| | - Peifu Tang
- School of Medicine, Nankai University, Tianjin, China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Quanyi Guo
- School of Medicine, Nankai University, Tianjin, China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
| | - Licheng Zhang
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| |
Collapse
|
3
|
Semitela A, Marques PAAP, Completo A. Strategies to engineer articular cartilage with biomimetic zonal features: a review. Biomater Sci 2024. [PMID: 39463257 DOI: 10.1039/d4bm00579a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Articular cartilage (AC) is a highly specialized tissue with restricted ability for self-regeneration, given its avascular and acellular nature. Although a considerable number of surgical treatments is available for the repair, reconstruction, and regeneration of AC defects, most of them do not prioritize the development of engineered cartilage with zonal stratification derived from biomimetic biochemical, biomechanical and topographic cues. In the absence of these zonal elements, engineered cartilage will exhibit increased susceptibility to failure and will neither be able to withstand the mechanical loading to which AC is subjected nor will it integrate well with the surrounding tissue. In this regard, new breakthroughs in the development of hierarchical stratified engineered cartilage are highly sought after. Initially, this review provides a comprehensive analysis of the composition and zonal organization of AC, aiming to enhance our understanding of the significance of the structure of AC for its function. Next, we direct our attention towards the existing in vitro and in vivo studies that introduce zonal elements in engineered cartilage to elicit appropriate AC regeneration by employing tissue engineering strategies. Finally, the advantages, challenges, and future perspectives of these approaches are presented.
Collapse
Affiliation(s)
- Angela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Paula A A P Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
4
|
Jacinto JGP, Ogundipe TG, Benazzi C, Häfliger IM, Muscatello LV, Bolcato M, Rinnovati R, Gentile A, Drögemüller C. Familial osteochondrodysplastic and cardiomyopathic syndrome in Chianina cattle. J Vet Intern Med 2024. [PMID: 39460958 DOI: 10.1111/jvim.17221] [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: 02/02/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Skeletal dysplasia encompasses a heterogeneous group of genetic disorders characterized by an abnormal development of bones, joints, and cartilage. Two Chianina half-sibling calves from consanguineous mating with congenital skeletal malformations and cardiac abnormalities were identified. HYPOTHESIS/OBJECTIVES To characterize the disease phenotype, to evaluate its genetic cause, and to determine the prevalence of the deleterious alleles in the Chianina population. ANIMALS Two affected calves, their parents and 332 Chianina bulls. METHODS The affected animals underwent clinicopathological investigation. Whole-genome sequencing trio-approach and PCR-based assessment of the frequency of TDP-glucose 4,6-dehydratase (TGDS) and laminin subunit alpha 4 (LAMA4) alleles were performed. RESULTS The cases presented with retarded growth, poor nutritional status associated with muscular atrophy and angular deformities of the hindlimbs. Radiologic examination identified generalized osteopenia and shortening of the limb long bones. Necropsy showed osteochondrodysplastic limbs and dilatation of the heart right ventricle. On histological examination, the physeal cartilages were characterized by multifocal mild to moderate loss of the normal columnar arrangement of chondrocytes. Osteopenia also was observed. Genetic analysis identified a missense variant in TGDS and a splice-site variant in LAMA4, both of which were homozygous in the 2 cases. Parents were heterozygous and allele frequency in the Chianina population for the TGDS variant was 5% and for the LAMA4 variant was 2%. CONCLUSIONS AND CLINICAL IMPORTANCE Genetic findings identified 2 potentially pathogenic alleles in TGDS and LAMA4, but no clear mode of inheritance could be determined.
Collapse
Affiliation(s)
- Joana G P Jacinto
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
- Institute of Genetics, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Tolulope G Ogundipe
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Cinzia Benazzi
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Irene M Häfliger
- Institute of Genetics, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Luisa V Muscatello
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Marilena Bolcato
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Riccardo Rinnovati
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Arcangelo Gentile
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Cord Drögemüller
- Institute of Genetics, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Melrose J, Guilak F. Diverse and multifunctional roles for perlecan ( HSPG2) in repair of the intervertebral disc. JOR Spine 2024; 7:e1362. [PMID: 39081381 PMCID: PMC11286675 DOI: 10.1002/jsp2.1362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 06/11/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Perlecan is a widely distributed, modular, and multifunctional heparan sulfate proteoglycan, which facilitates cellular communication with the extracellular environment to promote tissue development, tissue homeostasis, and optimization of biomechanical tissue functions. Perlecan-mediated osmotic mechanotransduction serves to regulate the metabolic activity of cells in tissues subjected to tension, compression, or shear. Perlecan interacts with a vast array of extracellular matrix (ECM) proteins through which it stabilizes tissues and regulates the proliferation or differentiation of resident cell populations. Here we examine the roles of the HS-proteoglycan perlecan in the normal and destabilized intervertebral disc. The intervertebral disc cell has evolved to survive in a hostile weight bearing, acidic, low oxygen tension, and low nutrition environment, and perlecan provides cytoprotection, shields disc cells from excessive compressive forces, and sequesters a range of growth factors in the disc cell environment where they aid in cellular survival, proliferation, and differentiation. The cells in mechanically destabilized connective tissues attempt to re-establish optimal tissue composition and tissue functional properties by changing the properties of their ECM, in the process of chondroid metaplasia. We explore the possibility that perlecan assists in these cell-mediated tissue remodeling responses by regulating disc cell anabolism. Perlecan's mechano-osmotic transductive property may be of potential therapeutic application.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling InstituteNorthern Sydney Local Health DistrictSt. LeonardsNew South WalesAustralia
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydneyNew South WalesAustralia
- Sydney Medical School, NorthernThe University of SydneySt. LeonardsNew South WalesAustralia
- Faculty of Medicine and HealthThe University of Sydney, Royal North Shore HospitalSt. LeonardsNew South WalesAustralia
| | - Farshid Guilak
- Department of Orthopaedic SurgeryWashington UniversitySt. LouisMissouriUSA
- Department of OrthopaedicsShriners Hospitals for ChildrenSt. LouisMissouriUSA
| |
Collapse
|
6
|
Li T, Liu J, Guo M, Bin FC, Duan Q, Dong XZ, Jin F, Fujita K, Zheng ML. Femtosecond Laser Maskless Optical Projection Lithography of Cartilage PCM Inspired 3D Protein Matrix to Chondrocyte Phenotype. Adv Healthc Mater 2024; 13:e2400849. [PMID: 38687974 DOI: 10.1002/adhm.202400849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/28/2024] [Indexed: 05/02/2024]
Abstract
Hydrogels containing chondrocytes have exhibited excellent potential in regenerating hyaline cartilage. However, chondrocytes are vulnerable to dedifferentiation during in vitro culture, leading to fibrosis and mechanical degradation of newly formed cartilage. It is proposed to modulate cartilage formation via the developed chondrocyte pericellular matrix (PCM) -like scaffolds for the first time, in which the S, M, and L-sized scaffolds are fabricated by femtosecond laser maskless optical projection lithography (FL-MOPL) of bovine serum albumin-glyceryl methacrylate hydrogel. Chondrocytes on the M PCM-like scaffold can maintain round morphology and synthesize extracellular matrix (ECM) to induce regeneration of hyaline cartilage microtissues by geometrical restriction. A series of M PCM-like scaffolds is fabricated with different stiffness and those with a high Young's modulus are more effective in maintaining the chondrocyte phenotype. The proposed PCM-like scaffolds are effective in modulating cartilage formation influenced by pore size, depth, and stiffness, which will pave the way for a better understanding of the geometric cues of mechanotransduction interactions in regulating cell fate and open up new avenues for tissue engineering.
Collapse
Affiliation(s)
- Teng Li
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Jie Liu
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| | - Min Guo
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Fan-Chun Bin
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Qi Duan
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
- School of Future Technologies, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, China
| | - Xian-Zi Dong
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| | - Feng Jin
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| | - Katsumasa Fujita
- Department of Applied Physics, Osaka University, 2-1Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mei-Ling Zheng
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, China
| |
Collapse
|
7
|
Huffer A, Mao M, Ballard K, Ozdemir T. Biomimetic Hyaluronan Binding Biomaterials to Capture the Complex Regulation of Hyaluronan in Tissue Development and Function. Biomimetics (Basel) 2024; 9:499. [PMID: 39194478 DOI: 10.3390/biomimetics9080499] [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: 06/24/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Within native ECM, Hyaluronan (HA) undergoes remarkable structural remodeling through its binding receptors and proteins called hyaladherins. Hyaladherins contain a group of tandem repeat sequences, such as LINK domains, BxB7 homologous sequences, or 20-50 amino acid long short peptide sequences that have high affinity towards side chains of HA. The HA binding sequences are critical players in HA distribution and regulation within tissues and potentially attractive therapeutic targets to regulate HA synthesis and organization. While HA is a versatile and successful biopolymer, most HA-based therapeutics have major differences from a native HA molecule, such as molecular weight discrepancies, crosslinking state, and remodeling with other HA binding proteins. Recent studies showed the promise of HA binding domains being used as therapeutic biomaterials for osteoarthritic, ocular, or cardiovascular therapeutic products. However, we propose that there is a significant potential for HA binding materials to reveal the physiological functions of HA in a more realistic setting. This review is focused on giving a comprehensive overview of the connections between HA's role in the body and the potential of HA binding material applications in therapeutics and regenerative medicine. We begin with an introduction to HA then discuss HA binding molecules and the process of HA binding. Finally, we discuss HA binding materials anf the future prospects of potential HA binding biomaterials systems in the field of biomaterials and tissue engineering.
Collapse
Affiliation(s)
- Amelia Huffer
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines, Rapid City, SD 57701, USA
| | - Mingyang Mao
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines, Rapid City, SD 57701, USA
| | - Katherine Ballard
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines, Rapid City, SD 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines, Rapid City, SD 57701, USA
| |
Collapse
|
8
|
van Mourik M, Tiemeijer BM, van Zon M, Abinzano F, Tel J, Foolen J, Ito K. Cartilage-derived cells display heterogeneous pericellular matrix synthesis in agarose microgels. Matrix Biol Plus 2024; 23:100157. [PMID: 39139760 PMCID: PMC11321428 DOI: 10.1016/j.mbplus.2024.100157] [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: 04/17/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 08/15/2024] Open
Abstract
The pericellular matrix (PCM) surrounding chondrocytes is essential for articular cartilage tissue engineering. As the current isolation methods to obtain chondrocytes with their PCM (chondrons) result in a heterogeneous mixture of chondrocytes and chondrons, regenerating the PCM using a tissue engineering approach could prove beneficial. In this study, we aimed to discern the behavior of articular chondrocytes (ACs) in regenerating the PCM in such an approach and whether this would also be true for articular cartilage-derived progenitor cells (ACPCs), as an alternative cell source. Bovine ACs and ACPCs were encapsulated in agarose microgels using droplet-based microfluidics. ACs were stimulated with TGF-β1 and dexamethasone and ACPCs were sequentially stimulated with BMP-9 followed by TGF-β1 and dexamethasone. After 0, 3, 5, and 10 days of culture, PCM components, type-VI collagen and perlecan, and ECM component, type-II collagen, were assessed using flow cytometry and fluorescence microscopy. Both ACs and ACPCs synthesized the PCM before the ECM. It was seen for the first time that synthesis of type-VI collagen always preceded perlecan. While the PCM synthesized by ACs resembled native chondrons after only 5 days of culture, ACPCs often made less well-structured PCMs. Both cell types showed variations between individual cells and donors. On one hand, this was more prominent in ACPCs, but also a subset of ACPCs showed superior PCM and ECM regeneration, suggesting that isolating these cells may potentially improve cartilage repair strategies.
Collapse
Affiliation(s)
- Marloes van Mourik
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Bart M. Tiemeijer
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Maarten van Zon
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
| | - Florencia Abinzano
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Jurjen Tel
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Jasper Foolen
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| |
Collapse
|
9
|
Miao MZ, Lee JS, Yamada KM, Loeser RF. Integrin signalling in joint development, homeostasis and osteoarthritis. Nat Rev Rheumatol 2024; 20:492-509. [PMID: 39014254 DOI: 10.1038/s41584-024-01130-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2024] [Indexed: 07/18/2024]
Abstract
Integrins are key regulators of cell-matrix interactions during joint development and joint tissue homeostasis, as well as in the development of osteoarthritis (OA). The signalling cascades initiated by the interactions of integrins with a complex network of extracellular matrix (ECM) components and intracellular adaptor proteins orchestrate cellular responses necessary for maintaining joint tissue integrity. Dysregulated integrin signalling, triggered by matrix degradation products such as matrikines, disrupts this delicate balance, tipping the scales towards an environment conducive to OA pathogenesis. The interplay between integrin signalling and growth factor pathways further underscores the multifaceted nature of OA. Moreover, emerging insights into the role of endocytic trafficking in regulating integrin signalling add a new layer of complexity to the understanding of OA development. To harness the therapeutic potential of targeting integrins for mitigation of OA, comprehensive understanding of their molecular mechanisms across joint tissues is imperative. Ultimately, deciphering the complexities of integrin signalling will advance the ability to treat OA and alleviate its global burden.
Collapse
Affiliation(s)
- Michael Z Miao
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Craniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Division of Rheumatology, Allergy, and Immunology and the Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Janice S Lee
- Craniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Office of the Clinical Director, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| | - Richard F Loeser
- Division of Rheumatology, Allergy, and Immunology and the Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
10
|
Bergstrom AR, Glimm MG, Houske EA, Cooper G, Viles E, Chapman M, Bourekis K, Welhaven HD, Brahmachary PP, Hahn AK, June RK. Metabolic Profiles of Encapsulated Chondrocytes Exposed to Short-Term Simulated Microgravity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601604. [PMID: 39005264 PMCID: PMC11245029 DOI: 10.1101/2024.07.01.601604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The mechanism by which chondrocytes respond to reduced mechanical loading environments and the subsequent risk of developing osteoarthritis remains unclear. This is of particular concern for astronauts. In space the reduced joint loading forces during prolonged microgravity (10-6 g) exposure could lead to osteoarthritis (OA), compromising quality of life post-spaceflight. In this study, we encapsulated human chondrocytes in an agarose gel of similar stiffness to the pericellular matrix to mimic the cartilage microenvironment. We then exposed agarose-chondrocyte constructs to simulated microgravity (SM) using a rotating wall vessel (RWV) bioreactor to better assess the cartilage health risks associated with spaceflight. Global metabolomic profiling detected a total of 1205 metabolite features across all samples, with 497 significant metabolite features identified by ANOVA (FDR-corrected p-value < 0.05). Specific metabolic shifts detected in response to SM exposure resulted in clusters of co-regulated metabolites, as well as key metabolites identified by variable importance in projection scores. Microgravity-induced metabolic shifts in gel constructs and media were indicative of protein synthesis, energy metabolism, nucleotide metabolism, and oxidative catabolism. The microgravity associated-metabolic shifts were consistent with early osteoarthritic metabolomic profiles in human synovial fluid, which suggests that even short-term exposure to microgravity (or other reduced mechanical loading environments) may lead to the development of OA.
Collapse
Affiliation(s)
- Annika R. Bergstrom
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
- Department of Chemical & Biological Engineering, Villanova University, Villanova, PA, USA, 19085
| | - Matthew G. Glimm
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
| | - Eden A. Houske
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
| | - Gwendolyn Cooper
- Molecular Biosciences Program, Montana State University, Bozeman, MT, USA, 59717
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, MT, USA, 59717
| | - Ethan Viles
- Molecular Biosciences Program, Montana State University, Bozeman, MT, USA, 59717
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT, USA, 59717
| | - Marrin Chapman
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
| | - Katherine Bourekis
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
| | - Hope D. Welhaven
- Molecular Biosciences Program, Montana State University, Bozeman, MT, USA, 59717
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, MT, USA, 59717
| | - Priyanka P. Brahmachary
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT, USA, 59717
| | - Alyssa K. Hahn
- Department of Biological & Environmental Science, Carroll College, Helena, MT, USA, 59625
| | - Ronald K. June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT, USA, 59717
| |
Collapse
|
11
|
Wang C, Fan M, Heo SJ, Adams SM, Li T, Liu Y, Li Q, Loebel C, Alisafaei F, Burdick JA, Lu XL, Birk DE, Mauck RL, Han L. Structure-Mechanics Principles and Mechanobiology of Fibrocartilage Pericellular Matrix: A Pivotal Role of Type V Collagen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600498. [PMID: 38979323 PMCID: PMC11230444 DOI: 10.1101/2024.06.26.600498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding resident cells in various tissue types, regulating matrix turnover, cell-matrix cross-talk and disease initiation. This study elucidated the structure-mechanical properties and mechanobiological functions of the PCM in fibrocartilage, a family of connective tissues that sustain complex tensile and compressive loads in vivo. Studying the murine meniscus as the model tissue, we showed that fibrocartilage PCM contains thinner, random collagen fibrillar networks that entrap proteoglycans, a structure distinct from the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). In comparison to the ECM, the PCM has a lower modulus and greater isotropy, but similar relative viscoelastic properties. In Col5a1 +/- menisci, the reduction of collagen V, a minor collagen localized in the PCM, resulted in aberrant fibril thickening with increased heterogeneity. Consequently, the PCM exhibited a reduced modulus, loss of isotropy and faster viscoelastic relaxation. This disrupted PCM contributes to perturbed mechanotransduction of resident meniscal cells, as illustrated by reduced intracellular calcium signaling, as well as upregulated biosynthesis of lysyl oxidase and tenascin C. When cultured in vitro, Col5a1 +/- meniscal cells synthesized a weakened nascent PCM, which had inferior properties towards protecting resident cells against applied tensile stretch. These findings underscore the PCM as a distinctive microstructure that governs fibrocartilage mechanobiology, and highlight the pivotal role of collagen V in PCM function. Targeting the PCM or its molecular constituents holds promise for enhancing not only meniscus regeneration and osteoarthritis intervention, but also addressing diseases across various fibrocartilaginous tissues.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Sheila M Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Claudia Loebel
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, United States
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| |
Collapse
|
12
|
Liu Y, Du L, Zhang H, Li G, Luo Y, Hu Z, Xu R, Yao J, Shi Z, Chen Y, Zhang C, Jin Z, Zhang C, Xie C, Fu J, Zhu Y, Zhu Y. Bioprinted biomimetic hydrogel matrices guiding stem cell aggregates for enhanced chondrogenesis and cartilage regeneration. J Mater Chem B 2024; 12:5360-5376. [PMID: 38700242 DOI: 10.1039/d4tb00323c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Articular cartilage tissue has limited self-repair capabilities, with damage frequently progressing to irreversible degeneration. Engineered tissues constructed through bioprinting and embedded with stem cell aggregates offer promising therapeutic alternatives. Aggregates of bone marrow mesenchymal stromal cells (BMSCs) demonstrate enhanced and more rapid chondrogenic differentiation than isolated cells, thus facilitating cartilage repair. However, it remains a key challenge to precisely control biochemical microenvironments to regulate cellular adhesion and cohesion within bioprinted matrices simultaneously. Herein, this work reports a bioprintable hydrogel matrix with high cellular adhesion and aggregation properties for cartilage repair. The hydrogel comprises an enhanced cell-adhesive gelatin methacrylate and a cell-cohesive chitosan methacrylate (CHMA), both of which are subjected to photo-initiated crosslinking. By precisely adjusting the CHMA content, the mechanical stability and biochemical cues of the hydrogels are finely tuned to promote cellular aggregation, chondrogenic differentiation and cartilage repair implantation. Multi-layer constructs encapsulated with BMSCs, with high cell viability reaching 91.1%, are bioprinted and photo-crosslinked to support chondrogenic differentiation for 21 days. BMSCs rapidly form aggregates and display efficient chondrogenic differentiation both on the hydrogels and within bioprinted constructs, as evidenced by the upregulated expression of Sox9, Aggrecan and Collagen 2a1 genes, along with high protein levels. Transplantation of these BMSC-laden bioprinted hydrogels into cartilaginous defects demonstrates effective hyaline cartilage repair. Overall, this cell-responsive hydrogel scaffold holds immense promise for applications in cartilage tissue engineering.
Collapse
Affiliation(s)
- Yuetian Liu
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
- Research Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Lijuan Du
- Research Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Hua Zhang
- Research Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
- State Key Laboratory of Molecular Engineering of Polymers (Fudan University), Shanghai 200438, China
| | - Guanrong Li
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
- Research Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Yang Luo
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
- Research Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Zeming Hu
- Research Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Rong Xu
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
- Research Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Jie Yao
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
| | - Zheyuan Shi
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
- Research Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Yige Chen
- Research Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Chi Zhang
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
| | - Zhanping Jin
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
| | - Caihua Zhang
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
| | - Chanchan Xie
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
| | - Jun Fu
- Key Laboratory of Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China.
| | - Yabin Zhu
- Research Institute of Smart Medicine and Biological Engineering, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Yingchun Zhu
- The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
| |
Collapse
|
13
|
van der Kraan PM, van Caam AP, Blaney Davidson EN, van den Bosch MH, van de Loo FA. Growth factors that drive aggrecan synthesis in healthy articular cartilage. Role for transforming growth factor-β? OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100459. [PMID: 38486843 PMCID: PMC10938168 DOI: 10.1016/j.ocarto.2024.100459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/04/2024] [Indexed: 03/17/2024] Open
Abstract
Introduction Articular cartilage makes smooth movement possible and destruction of this tissue leads to loss of joint function. An important biomolecule that determines this function is the large aggregating proteoglycan of cartilage, aggrecan. Aggrecan has a relatively short half-life in cartilage and therefore continuous production of this molecule is essential. Methods In this narrative review we discuss what is the role of growth factors in driving the synthesis of aggrecan in articular cartilage. A literature search has been done using the search items; cartilage, aggrecan, explant, Transforming Growth factor-β (TGF-β), Insulin-like Growth Factor (IGF), Bone Morphogenetic Protein (BMP) and the generic term "growth factors". Focus has been on studies using healthy cartilage and models of cartilage regeneration have been excluded. Results In healthy adult articular cartilage IGF is the main factor that drives aggrecan synthesis and maintains adequate levels of production. BMP's and TGF-β have a very limited role but appear to be more important during chondrogenesis and cartilage development. The major role of TGF-β is not stimulation of aggrecan synthesis but maintenance of the differentiated articular cartilage chondrocyte phenotype. Conclusion TGF-β is a factor that is generally considered as an important factor in stimulating aggrecan synthesis in cartilage but its role in this might be very restrained in healthy, adult articular cartilage.
Collapse
Affiliation(s)
| | - Arjan P.M. van Caam
- Radboudumc, Experimental Rheumatology, Department of Rheumatology, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Esmeralda N. Blaney Davidson
- Radboudumc, Experimental Rheumatology, Department of Rheumatology, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Martijn H.J. van den Bosch
- Radboudumc, Experimental Rheumatology, Department of Rheumatology, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Fons A.J. van de Loo
- Radboudumc, Experimental Rheumatology, Department of Rheumatology, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| |
Collapse
|
14
|
Du G, Zhang J, Shuai Q, Li L, Zhang Q, Shi R. Development of alginate-collagen interpenetrating network for osteoarthritic cartilage by in situ softening. Int J Biol Macromol 2024; 266:131259. [PMID: 38574937 DOI: 10.1016/j.ijbiomac.2024.131259] [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: 11/18/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
This study presents an alginate-collagen interpenetrating network (IPN) matrix of incorporating collagen fibrils into an alginate hydrogel by physical mixing and controlled gelation. The resulting matrix closely mimics the physiological and pathological stiffness range of the chondrocyte pericellular matrix (PCM). Chondrocytes were cultured within three-dimensional (3D) alginate-collagen IPN matrices with varying stiffness, namely Firm, Medium, and Soft. Alginate lyase was introduced to study the effects of the changes in stiffness of the Firm on chondrocyte response by in situ softening. The developed alginate-collagen IPN matrix displayed good cell-biocompatibility. Compared with stiffer tissue culture plastic (TCP), chondrocytes grown within Firm displayed a stabilized differentiated phenotype characterized by higher expression levels of aggrecan, collagen II, and SOX-9. Moreover, the developed alginate-collagen IPN matrix exhibited a gradually increased percentage of propidium iodide (PI)-positive dead cells with decreasing stiffness. Softer matrices directed cells towards higher proliferation rates and spherical morphologies while stimulating chondrocyte cluster formation. Furthermore, reducing Firm stiffness by in situ softening decreased aggrecan expression, contributing to matrix degradation similar to that seen in osteoarthritis (OA). Hence, the 3D alginate-collagen IPN constructs hold significant potential for in vitro replicating PCM stiffness changes observed in OA cartilage.
Collapse
Affiliation(s)
- Genlai Du
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Taiyuan 030001, China.
| | - Jiaqi Zhang
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Taiyuan 030001, China
| | - Qizhi Shuai
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Taiyuan 030001, China
| | - Li Li
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Taiyuan 030001, China
| | - Quanyou Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; Department of Orthopaedics, the Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Ruyi Shi
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Taiyuan 030001, China.
| |
Collapse
|
15
|
Rathnayake MSB, Boos MA, Farrugia BL, van Osch GJVM, Stok KS. Glycosaminoglycan-Mediated Interactions in Articular, Auricular, Meniscal, and Nasal Cartilage. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 38613808 DOI: 10.1089/ten.teb.2023.0346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2024]
Abstract
Glycosaminoglycans (GAGs) are ubiquitous components in the cartilage extracellular matrix (ECM). Ultrastructural arrangement of ECM and GAG-mediated interactions with collagen are known to govern the mechanics in articular cartilage, but these interactions are less clear in other cartilage types. Therefore, this article reviews the current literature on ultrastructure of articular, auricular, meniscal, and nasal septal cartilage, seeking insight into GAG-mediated interactions influencing mechanics. Ultrastructural features of these cartilages are discussed to highlight differences between them. GAG-mediated interactions are reviewed under two categories: interactions with chondrocytes and interactions with other fibrillar macromolecules of the ECM. Moreover, efforts to replicate GAG-mediated interactions to improve mechanical integrity of tissue-engineered cartilage constructs are discussed. In conclusion, studies exploring cartilage specific GAGs are poorly represented in the literature, and the ultrastructure of nasal septal and auricular cartilage is less studied compared with articular and meniscal cartilages. Understanding the contribution of GAGs in cartilage mechanics at the ultrastructural level and translating that knowledge to engineered cartilage will facilitate improvement of cartilage tissue engineering approaches.
Collapse
Affiliation(s)
- Manula S B Rathnayake
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Australia
| | - Manuela A Boos
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Australia
| | - Brooke L Farrugia
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Australia
- Graeme Clark Institute for Biomedical Engineering, The University of Melbourne, Parkville, Australia
| | - Gerjo J V M van Osch
- Department of Otorhinolaryngology, Head and Neck Surgery and Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Delft, The Netherlands
| | - Kathryn S Stok
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Australia
| |
Collapse
|
16
|
Nudelman H, Lőrincz A, Lamberti AG, Varga M, Kassai T, Józsa G. Management of Juvenile Osteochondral Fractures Utilising Absorbable PLGA Implants. J Clin Med 2024; 13:375. [PMID: 38256509 PMCID: PMC10816157 DOI: 10.3390/jcm13020375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
The incidence of articular injury, particularly osteochondral fractures (OCFs), has seen a cinnotable increase in recent years. Regardless of their location, fragments can be overlooked by plain radiographs, which might lead to osteoarthritis in the long run. Diagnostic imaging has a pivotal role in the assessment and classification of the fracture severity, as well as the presence of any associated dislocations. These fractures require surgical intervention for the restoration of joint function and the reduction of long-term complications. This paper aims to present the surgical correction and post-operative treatment of osteochondral fractures with absorbable implants in four children. The following affected areas are discussed: lateral condyle of the femur, patella and radial head. Utilising absorbable implants for the management of OCFs provides numerous advantages, including the elimination of the need for re-anaesthesia and reoperation, reduction of complications and facilitation of early rehabilitation. This approach also minimises the period of hospitalisation and proves effective in pediatric OCF treatment.
Collapse
Affiliation(s)
- Hermann Nudelman
- Division of Surgery, Traumatology and Otorhinolaryngology, Department of Paediatrics, Clinical Complex, University of Pécs, 7 József Attila Street, 7623 Pécs, Hungary; (H.N.)
| | - Aba Lőrincz
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti Street, 7624 Pécs, Hungary;
| | - Anna Gabriella Lamberti
- Division of Surgery, Traumatology and Otorhinolaryngology, Department of Paediatrics, Clinical Complex, University of Pécs, 7 József Attila Street, 7623 Pécs, Hungary; (H.N.)
| | - Marcell Varga
- Department of Pediatric Traumatology, Péterfy Hospital, Manninger Jenő National Trauma Center, 17 Fiumei Street, 1081 Budapest, Hungary
| | - Tamás Kassai
- Department of Pediatric Traumatology, Péterfy Hospital, Manninger Jenő National Trauma Center, 17 Fiumei Street, 1081 Budapest, Hungary
| | - Gergő Józsa
- Division of Surgery, Traumatology and Otorhinolaryngology, Department of Paediatrics, Clinical Complex, University of Pécs, 7 József Attila Street, 7623 Pécs, Hungary; (H.N.)
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti Street, 7624 Pécs, Hungary;
| |
Collapse
|
17
|
Fredrikson JP, Brahmachary PP, June RK, Cox LM, Chang CB. Pericellular Matrix Formation and Atomic Force Microscopy of Single Primary Human Chondrocytes Cultured in Alginate Microgels. Adv Biol (Weinh) 2024; 8:e2300268. [PMID: 37688354 PMCID: PMC10843004 DOI: 10.1002/adbi.202300268] [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: 06/30/2023] [Revised: 08/21/2023] [Indexed: 09/10/2023]
Abstract
One of the main components of articular cartilage is the chondrocyte's pericellular matrix (PCM), which is critical for regulating mechanotransduction, biochemical cues, and healthy cartilage development. Here, individual primary human chondrocytes (PHC) are encapsulated and cultured in 50 µm diameter alginate microgels using drop-based microfluidics. This unique culturing method enables PCM formation and manipulation of individual cells. Over ten days, matrix formation is observed using autofluorescence imaging, and the elastic moduli of isolated cells are measured using AFM. Matrix production and elastic modulus increase are observed for the chondrons cultured in microgels. Furthermore, the elastic modulus of cells grown in microgels increases ≈ten-fold over ten days, nearly reaching the elastic modulus of in vivo PCM. The AFM data is further analyzed using a Gaussian mixture model and shows that the population of PHCs grown in microgels exhibit two distinct populations with elastic moduli averaging 9.0 and 38.0 kPa. Overall, this work shows that microgels provide an excellent culture platform for the growth and isolation of PHCs, enabling PCM formation that is mechanically similar to native PCM. The microgel culture platform presented here has the potential to revolutionize cartilage regeneration procedures through the inclusion of in vitro developed PCM.
Collapse
Affiliation(s)
- Jacob P Fredrikson
- Department of Chemical & Biological Engineering, Montana State University, P.O. Box 173920, Bozeman, MT, 59717, USA
- Center for Biofilm Engineering, Montana State University, P.O. Box 173980, Bozeman, MT, 59717, USA
| | - Priyanka P Brahmachary
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT, 59717, USA
| | - Ronald K June
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT, 59717, USA
- Department of Microbiology & Cell Biology, Montana State University, P.O. Box 173520, Bozeman, MT, 59717, USA
| | - Lewis M Cox
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT, 59717, USA
| | - Connie B Chang
- Department of Chemical & Biological Engineering, Montana State University, P.O. Box 173920, Bozeman, MT, 59717, USA
- Center for Biofilm Engineering, Montana State University, P.O. Box 173980, Bozeman, MT, 59717, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| |
Collapse
|
18
|
Semitela Â, Pinto SC, Capitão A, Marques PAAP, Completo A. Fabrication of Customizable and Reproducible 3D Chondrocyte-Laden Nanofibrous Architectures: Effect of Specific Fiber Alignments and Porosities on Chondrocyte Response under Cyclic Compression. ACS APPLIED BIO MATERIALS 2023; 6:5541-5554. [PMID: 37947854 DOI: 10.1021/acsabm.3c00737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Electrospinning has been widely employed to fabricate complex extracellular matrix-like microenvironments for tissue engineering due to its ability to replicate structurally biomimetic micro- and nanotopographic cues. Nevertheless, these nanofibrous structures are typically either confined to bidimensional systems or confined to three-dimensional ones that are unable to provide controlled multiscale patterns. Thus, an electrospinning modality was used in this work to fabricate chondrocyte-laden nanofibrous scaffolds with highly customizable three-dimensional (3D) architectures in an automated manner, with the ultimate goal of recreating a suitable 3D scaffold for articular cartilage tissue engineering. Three distinct architectures were designed and fabricated by combining multiple nanofibrous and chondrocyte-laden hydrogel layers and tested in vitro in a compression bioreactor system. Results demonstrated that it was possible to precisely control the placement and alignment of electrospun polycaprolactone and gelatin nanofibers, generating three unique architectures with distinctive macroscale porosity, water absorption capacity, and mechanical properties. The architecture organized in a lattice-like fashion was highly porous with substantial pore interconnectivity, resulting in a high-water absorption capacity but a poor compression modulus and relatively weaker energy dissipation capacity. The donut-like 3D geometry was the densest, with lower swelling, but the highest compression modulus and improved energy dissipation ability. The third architecture combined a lattice and donut-like fibrous arrangement, exhibiting intermediary behavior in terms of porosity, water absorption, compression modulus, and energy dissipation capacity. The properties of the donut-like 3D architecture demonstrated great potential for articular cartilage tissue engineering, as it mimicked key topographic, chemical, and mechanical characteristics of chondrocytes' surrounding environment. In fact, the combination of these architectural features with a dynamically compressive mechanical stimulus triggered the best in vitro results in terms of viability and biosynthetic production.
Collapse
Affiliation(s)
- Ângela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Susana C Pinto
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana Capitão
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Paula A A P Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
19
|
Mao J, Huang L, Ding Y, Ma X, Wang Q, Ding L. Insufficiency of collagenases in establishment of primary chondrocyte culture from cartilage of elderly patients receiving total joint replacement. Cell Tissue Bank 2023; 24:759-768. [PMID: 37138136 DOI: 10.1007/s10561-023-10094-0] [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: 02/09/2023] [Accepted: 04/18/2023] [Indexed: 05/05/2023]
Abstract
Background Collagenases are frequently used in chondrocyte isolation from articular cartilage. However, the sufficiency of this enzyme in establishing primary human chondrocyte culture remains unknown. Methods Cartilage slices shaved from femoral head or tibial plateau of patients receiving total joint replacement surgery (16 hips, 8 knees) were subjected to 0.02% collagenase IA digestion for 16 h with (N = 19) or without (N = 5) the pre-treatment of 0.4% pronase E for 1.5 h. Chondrocyte yield and viability were compared between two groups. Chondrocyte phenotype was determined by the expression ratio of collagen type II to I. The morphology of cultured chondrocytes was monitored with a light microscope.Results Cartilage with pronase E pre-treatment yielded significantly higher chondrocytes than that without the pre-treatment (3,399 ± 1,637 cells/mg wet cartilage vs. 1,895 ± 688 cells/mg wet cartilage; P = 0.0067). Cell viability in the former group was also significantly higher than that in the latter (94% ± 2% vs. 86% ± 6%; P = 0.03). When cultured in monolayers, cells from cartilage with pronase E pre-treatment grew in a single plane showing rounded shape while cells from the other group grew in multi-planes and exhibited irregular shape. The mRNA expression ratio of collagen type II to I was 13.2 ± 7.5 in cells isolated from cartilage pre-treated with pronase E, indicating a typical chondrocyte phenotype. Conclusions Collagenase IA was not sufficient in establishing primary human chondrocyte culture. Cartilage must be treated with pronase E prior to collagenase IA application.
Collapse
Affiliation(s)
- Jiamin Mao
- Department of Basic Medical Sciences, Jiangnan University Wuxi College of Medicine, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu, China
| | - Lexi Huang
- Department of Basic Medical Sciences, Jiangnan University Wuxi College of Medicine, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu, China
| | - Yiyang Ding
- Department of Basic Medical Sciences, Jiangnan University Wuxi College of Medicine, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu, China
| | - Xiaoyu Ma
- Department of Basic Medical Sciences, Jiangnan University Wuxi College of Medicine, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu, China
| | - Quanming Wang
- Department of Orthopaedic Surgery, Jiangnan University Affiliated Hospital, Wuxi, Jiangsu, China
| | - Lei Ding
- Department of Basic Medical Sciences, Jiangnan University Wuxi College of Medicine, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
20
|
Lee CY, Nedunchezian S, Lin SY, Su YF, Wu CW, Wu SC, Chen CH, Wang CK. Bilayer osteochondral graft in rabbit xenogeneic transplantation model comprising sintered 3D-printed bioceramic and human adipose-derived stem cells laden biohydrogel. J Biol Eng 2023; 17:74. [PMID: 38012588 PMCID: PMC10680339 DOI: 10.1186/s13036-023-00389-x] [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: 08/08/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023] Open
Abstract
Reconstruction of severe osteochondral defects in articular cartilage and subchondral trabecular bone remains a challenging problem. The well-integrated bilayer osteochondral graft design expects to be guided the chondrogenic and osteogenic differentiation for stem cells and provides a promising solution for osteochondral tissue repair in this study. The subchondral bone scaffold approach is based on the developed finer and denser 3D β-tricalcium phosphate (β-TCP) bioceramic scaffold process, which is made using a digital light processing (DLP) technology and the novel photocurable negative thermo-responsive (NTR) bioceramic slurry. Then, the concave-top disc sintered 3D-printed bioceramic incorporates the human adipose-derived stem cells (hADSCs) laden photo-cured hybrid biohydrogel (HG + 0.5AFnSi) comprised of hyaluronic acid methacryloyl (HAMA), gelatin methacryloyl (GelMA), and 0.5% (w/v) acrylate-functionalized nano-silica (AFnSi) crosslinker. The 3D β-TCP bioceramic compartment is used to provide essential mechanical support for cartilage regeneration in the long term and slow biodegradation. However, the apparent density and compressive strength of the 3D β-TCP bioceramics can be obtained for ~ 94.8% theoretical density and 11.38 ± 1.72 MPa, respectively. In addition, the in vivo results demonstrated that the hADSC + HG + 0.5AFnSi/3D β-TCP of the bilayer osteochondral graft showed a much better osteochondral defect repair outcome in a rabbit model. The other word, the subchondral bone scaffold of 3D β-TCP bioceramic could accelerate the bone formation and integration with the adjacent host cancellous tissue at 12 weeks after surgery. And then, a thicker cartilage layer with a smooth surface and uniformly aligned chondrocytes were observed by providing enough steady mechanical support of the 3D β-TCP bioceramic scaffold.
Collapse
Affiliation(s)
- Chih-Yun Lee
- Ph.D. Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Swathi Nedunchezian
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Sung-Yen Lin
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Departments of Orthopaedics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Orthopaedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Orthopaedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, 80145, Taiwan
| | - Yu-Feng Su
- Faculty of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80756, Taiwan
- Department of Surgery, Division of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Che-Wei Wu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Shun-Cheng Wu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Nursing, Asia University, Taichung, 41354, Taiwan
| | - Chung-Hwan Chen
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Departments of Orthopaedics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Orthopaedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Orthopaedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, 80145, Taiwan
- Ph.D. Program in Biomedical Engineering, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chih-Kuang Wang
- Ph.D. Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
21
|
Yan W, Maimaitimin M, Wu Y, Fan Y, Ren S, Zhao F, Cao C, Hu X, Cheng J, Ao Y. Meniscal fibrocartilage regeneration inspired by meniscal maturational and regenerative process. SCIENCE ADVANCES 2023; 9:eadg8138. [PMID: 37939174 PMCID: PMC10631723 DOI: 10.1126/sciadv.adg8138] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023]
Abstract
Meniscus is a complex and crucial fibrocartilaginous tissue within the knee joint. Meniscal regeneration remains to be a scientific and translational challenge. We clarified that mesenchymal stem cells (MSCs) participated in meniscal maturation and regeneration using MSC-tracing transgenic mice model. Here, inspired by meniscal natural maturational and regenerative process, we developed an effective and translational strategy to facilitate meniscal regeneration by three-dimensionally printing biomimetic meniscal scaffold combining autologous synovium transplant, which contained abundant intrinsic MSCs. We verified that this facilitated anisotropic meniscus-like tissue regeneration and protected cartilage from degeneration in large animal model. Mechanistically, the biomechanics and matrix stiffness up-regulated Piezo1 expression, facilitating concerted activation of calcineurin and NFATc1, further activated YAP-pSmad2/3-SOX9 axis, and consequently facilitated fibrochondrogenesis of MSCs during meniscal regeneration. In addition, Piezo1 induced by biomechanics and matrix stiffness up-regulated collagen cross-link enzyme expression, which catalyzed collagen cross-link and thereby enhanced mechanical properties of regenerated tissue.
Collapse
Affiliation(s)
- Wenqiang Yan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Maihemuti Maimaitimin
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yue Wu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yifei Fan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Shuang Ren
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Fengyuan Zhao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Chenxi Cao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Jin Cheng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yingfang Ao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| |
Collapse
|
22
|
Peng X, Huang Y, Genin GM. The fibrous character of pericellular matrix mediates cell mechanotransduction. JOURNAL OF THE MECHANICS AND PHYSICS OF SOLIDS 2023; 180:105423. [PMID: 38559448 PMCID: PMC10978028 DOI: 10.1016/j.jmps.2023.105423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Cells in solid tissues sense and respond to mechanical signals that are transmitted through extracellular matrix (ECM) over distances that are many times their size. This long-range force transmission is known to arise from strain-stiffening and buckling in the collagen fiber ECM network, but must also pass through the denser pericellular matrix (PCM) that cells form by secreting and compacting nearby collagen. However, the role of the PCM in the transmission of mechanical signals is still unclear. We therefore studied an idealized computational model of cells embedded within fibrous collagen ECM and PCM. Our results suggest that the smaller network pore sizes associated with PCM attenuates tension-driven collagen-fiber alignment, undermining long-range force transmission and shielding cells from mechanical stress. However, elongation of the cell body or anisotropic cell contraction can compensate for these effects to enable long distance force transmission. Results are consistent with recent experiments that highlight an effect of PCM on shielding cells from high stresses. Results have implications for the transmission of mechanical signaling in development, wound healing, and fibrosis.
Collapse
Affiliation(s)
- Xiangjun Peng
- U.S. National Science Foundation Science and Technology Center for Engineering Mechanobiology, and Department of Biomedical Engineering, Washington University, St. Louis, MO 63130 United States
| | - Yuxuan Huang
- U.S. National Science Foundation Science and Technology Center for Engineering Mechanobiology, and Department of Biomedical Engineering, Washington University, St. Louis, MO 63130 United States
| | - Guy M. Genin
- U.S. National Science Foundation Science and Technology Center for Engineering Mechanobiology, and Department of Biomedical Engineering, Washington University, St. Louis, MO 63130 United States
| |
Collapse
|
23
|
Mariadoss AVA, Wang CZ. Exploring the Cellular and Molecular Mechanism of Discoidin Domain Receptors (DDR1 and DDR2) in Bone Formation, Regeneration, and Its Associated Disease Conditions. Int J Mol Sci 2023; 24:14895. [PMID: 37834343 PMCID: PMC10573612 DOI: 10.3390/ijms241914895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
The tyrosine kinase family receptor of discoidin domain receptors (DDR1 and DDR2) is known to be activated by extracellular matrix collagen catalytic binding protein receptors. They play a remarkable role in cell proliferation, differentiation, migration, and cell survival. DDR1 of the DDR family regulates matrix-metalloproteinase, which causes extracellular matrix (ECM) remodeling and reconstruction during unbalanced homeostasis. Collagenous-rich DDR1 triggers the ECM of cartilage to regenerate the cartilage tissue in osteoarthritis (OA) and temporomandibular disorder (TMD). Moreover, DDR2 is prominently present in the fibroblasts, smooth muscle cells, myofibroblasts, and chondrocytes. It is crucial in generating and breaking collagen vital cellular activities like proliferation, differentiation, and adhesion mechanisms. However, the deficiency of DDR1 rather than DDR2 was detrimental in cases of OA and TMDs. DDR1 stimulated the ECM cartilage and improved bone regeneration. Based on the above information, we made an effort to outline the advancement of the utmost promising DDR1 and DDR2 regulation in bone and cartilage, also summarizing their structural, biological activity, and selectivity.
Collapse
Affiliation(s)
| | - Chau-Zen Wang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung 912301, Taiwan
| |
Collapse
|
24
|
Brown WE, Lavernia L, Bielajew BJ, Hu JC, Athanasiou KA. Human nasal cartilage: Functional properties and structure-function relationships for the development of tissue engineering design criteria. Acta Biomater 2023; 168:113-124. [PMID: 37454708 DOI: 10.1016/j.actbio.2023.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Nose reconstruction often requires scarce cartilage grafts. Nasal cartilage properties must be determined to serve as design criteria for engineering grafts. Thus, mechanical and biochemical properties were obtained in multiple locations of human nasal septum, upper lateral cartilage (ULC), and lower lateral cartilage (LLC). Within each region, no statistical differences among locations were detected, but anisotropy at some septum locations was noted. In the LLC, the tensile modulus and ultimate tensile strength (UTS) in the inferior-superior direction were statistically greater than in the anterior-posterior direction. Cartilage from all regions exhibited hyperelasticity in tension, but regions varied in degree of hyalinicity (i.e., Col II:Col I ratio). The septum contained the most collagen II and least collagen I and III, making it more hyaline than the ULC and LLC. The septum had a greater aggregate modulus, UTS, and lower total collagen/wet weight (Col/WW) than the ULC and LLC. The ULC had greater tensile modulus, DNA/WW, and lower glycosaminoglycan/WW than the septum and LLC. The ULC had a greater pyridinoline/Col than the septum. Histological staining suggested the presence of chondrons in all regions. In the ULC and LLC, tensile modulus correlated with total collagen content, while aggregate modulus correlated with pyridinoline content and weakly with pentosidine content. However, future studies should be performed to validate these proposed structure-function relationships. This study of human nasal cartilage provides 1) crucial design criteria for nasal cartilage tissue engineering efforts, 2) quantification of major and minor collagen subtypes and crosslinks, and 3) structure-function relationships. Surprisingly, the large mechanical properties found, particularly in the septum, suggests that nasal cartilage may experience higher-than-expected mechanical loads. STATEMENT OF SIGNIFICANCE: While tissue engineering holds promise to generate much-needed cartilage grafts for nasal reconstruction, little is known about nasal cartilage from an engineering perspective. In this study, the mechanical and biochemical properties of the septum, upper lateral cartilage (ULC), and lower lateral cartilage (LLC) were evaluated using cartilage-specific methods. For the first time in this tissue, all major and minor collagens and collagen crosslinks were measured, demonstrating that the septum was more hyaline than the ULC and LLC. Additionally, new structure-function relationships in the ULC and LLC were identified. This study greatly expands upon the quantitative understanding of human nasal cartilage and provides crucial engineering design criteria for much-needed nasal cartilage tissue engineering efforts.
Collapse
Affiliation(s)
- Wendy E Brown
- Department of Biomedical Engineering, University of California Irvine, 3120 Natural Sciences II, Irvine, CA, 92697, USA
| | - Laura Lavernia
- Department of Biomedical Engineering, University of California Irvine, 3120 Natural Sciences II, Irvine, CA, 92697, USA
| | - Benjamin J Bielajew
- Department of Biomedical Engineering, University of California Irvine, 3120 Natural Sciences II, Irvine, CA, 92697, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Irvine, 3120 Natural Sciences II, Irvine, CA, 92697, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California Irvine, 3120 Natural Sciences II, Irvine, CA, 92697, USA.
| |
Collapse
|
25
|
Kwok B, Chandrasekaran P, Wang C, He L, Mauck RL, Dyment NA, Koyama E, Han L. Rapid specialization and stiffening of the primitive matrix in developing articular cartilage and meniscus. Acta Biomater 2023; 168:235-251. [PMID: 37414114 PMCID: PMC10529006 DOI: 10.1016/j.actbio.2023.06.047] [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: 02/20/2023] [Revised: 06/02/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023]
Abstract
Understanding early patterning events in the extracellular matrix (ECM) formation can provide a blueprint for regenerative strategies to better recapitulate the function of native tissues. Currently, there is little knowledge on the initial, incipient ECM of articular cartilage and meniscus, two load-bearing counterparts of the knee joint. This study elucidated distinctive traits of their developing ECMs by studying the composition and biomechanics of these two tissues in mice from mid-gestation (embryonic day 15.5) to neo-natal (post-natal day 7) stages. We show that articular cartilage initiates with the formation of a pericellular matrix (PCM)-like primitive matrix, followed by the separation into distinct PCM and territorial/interterritorial (T/IT)-ECM domains, and then, further expansion of the T/IT-ECM through maturity. In this process, the primitive matrix undergoes a rapid, exponential stiffening, with a daily modulus increase rate of 35.7% [31.9 39.6]% (mean [95% CI]). Meanwhile, the matrix becomes more heterogeneous in the spatial distribution of properties, with concurrent exponential increases in the standard deviation of micromodulus and the slope correlating local micromodulus with the distance from cell surface. In comparison to articular cartilage, the primitive matrix of meniscus also exhibits exponential stiffening and an increase in heterogeneity, albeit with a much slower daily stiffening rate of 19.8% [14.9 24.9]% and a delayed separation of PCM and T/IT-ECM. These contrasts underscore distinct development paths of hyaline versus fibrocartilage. Collectively, these findings provide new insights into how knee joint tissues form to better guide cell- and biomaterial-based repair of articular cartilage, meniscus and potentially other load-bearing cartilaginous tissues. STATEMENT OF SIGNIFICANCE: Successful regeneration of articular cartilage and meniscus is challenged by incomplete knowledge of early events that drive the initial formation of the tissues' extracellular matrix in vivo. This study shows that articular cartilage initiates with a pericellular matrix (PCM)-like primitive matrix during embryonic development. This primitive matrix then separates into distinct PCM and territorial/interterritorial domains, undergoes an exponential daily stiffening of ≈36% and an increase in micromechanical heterogeneity. At this early stage, the meniscus primitive matrix shows differential molecular traits and exhibits a slower daily stiffening of ≈20%, underscoring distinct matrix development between these two tissues. Our findings thus establish a new blueprint to guide the design of regenerative strategies to recapitulate the key developmental steps in vivo.
Collapse
Affiliation(s)
- Bryan Kwok
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Lan He
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Nathaniel A Dyment
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
26
|
Zheng S, An S, Luo Y, Vithran DTA, Yang S, Lu B, Deng Z, Li Y. HYBID in osteoarthritis: Potential target for disease progression. Biomed Pharmacother 2023; 165:115043. [PMID: 37364478 DOI: 10.1016/j.biopha.2023.115043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
HYBID is a new hyaluronan-degrading enzyme and exists in various cells of the human body. Recently, HYBID was found to over-express in the osteoarthritic chondrocytes and fibroblast-like synoviocytes. According to these researches, high level of HYBID is significantly correlated with cartilage degeneration in joints and hyaluronic acid degradation in synovial fluid. In addition, HYBID can affect inflammatory cytokine secretion, cartilage and synovium fibrosis, synovial hyperplasia via multiple signaling pathways, thereby exacerbating osteoarthritis. Based on the existing research of HYBID in osteoarthritis, HYBID can break the metabolic balance of HA in joints through the degradation ability independent of HYALs/CD44 system and furthermore affect cartilage structure and mechanotransduction of chondrocytes. In particular, in addition to HYBID itself being able to trigger some signaling pathways, we believe that low-molecular-weight hyaluronan produced by excess degradation can also stimulate some disease-promoting signaling pathways by replacing high-molecular-weight hyaluronan in joints. The specific role of HYBID in osteoarthritis is gradually revealed, and the discovery of HYBID raises the new way to treat osteoarthritis. In this review, the expression and basic functions of HYBID in joints were summarized, and reveal potential role of HYBID as a key target in treatment for osteoarthritis.
Collapse
Affiliation(s)
- Shengyuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Senbo An
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yan Luo
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Djandan Tadum Arthur Vithran
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shaoqu Yang
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Bangbao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
Zhu Y, Zhang M, Sun Q, Wang X, Li X, Li Q. Advanced Mechanical Testing Technologies at the Cellular Level: The Mechanisms and Application in Tissue Engineering. Polymers (Basel) 2023; 15:3255. [PMID: 37571149 PMCID: PMC10422338 DOI: 10.3390/polym15153255] [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: 07/11/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Mechanics, as a key physical factor which affects cell function and tissue regeneration, is attracting the attention of researchers in the fields of biomaterials, biomechanics, and tissue engineering. The macroscopic mechanical properties of tissue engineering scaffolds have been studied and optimized based on different applications. However, the mechanical properties of the overall scaffold materials are not enough to reveal the mechanical mechanism of the cell-matrix interaction. Hence, the mechanical detection of cell mechanics and cellular-scale microenvironments has become crucial for unraveling the mechanisms which underly cell activities and which are affected by physical factors. This review mainly focuses on the advanced technologies and applications of cell-scale mechanical detection. It summarizes the techniques used in micromechanical performance analysis, including atomic force microscope (AFM), optical tweezer (OT), magnetic tweezer (MT), and traction force microscope (TFM), and analyzes their testing mechanisms. In addition, the application of mechanical testing techniques to cell mechanics and tissue engineering scaffolds, such as hydrogels and porous scaffolds, is summarized and discussed. Finally, it highlights the challenges and prospects of this field. This review is believed to provide valuable insights into micromechanics in tissue engineering.
Collapse
Affiliation(s)
- Yingxuan Zhu
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Mengqi Zhang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qingqing Sun
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
28
|
Upadhyay U, Kolla S, Chelluri LK. Extracellular matrix composition analysis of human articular cartilage for the development of organ-on-a-chip. Biochem Biophys Res Commun 2023; 667:81-88. [PMID: 37209566 DOI: 10.1016/j.bbrc.2023.04.117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 04/18/2023] [Accepted: 04/29/2023] [Indexed: 05/22/2023]
Abstract
INTRODUCTION Articular cartilage has a complex extracellular matrix (ECM) that provides it a defined architecture for its load-bearing properties. The complete understanding of ECM components is imperative for developing biomimetic organ-on-a-chip tissue construct. OBJECTIVE This study aimed to decellularize and characterize the ECM for its protein profiling to generate a niche for enhanced chondrocyte proliferation. METHODS Articular cartilage scrapings were subjected to mechanical and collagenase digestion, followed by sodium dodecyl sulfate (SDS) treatment for 8 h and 16 h. The de-cellularization efficiency was confirmed by hematoxylin & eosin, alcian blue, masson's trichrome staining, and scanning electron microscopy (SEM). The ECM protein profile was quantified by liquid chromatography tandem mass spectrometry (LC-MS/MS) using a bottom-up approach. RESULTS Histological characterization revealed void lacunae that lacked staining for cellular components. The ECM, sulfated glycosaminoglycan content, and collagen fibers were preserved after 8 h and 16 h of de-cellularization. The SEM ultrastructure images showed that few chondrocytes adhered to the ECM after 8 h and cell-free ECM after 16 h of de-cellularization. LC-MS/MS analysis identified 66 proteins with heterotypic collagen types COL1A1-COL6A1, COL14A1, COL22A1 and COL25A1 showed moderate fold change and expression levels, while COL18A1, COL26A1, chondroitin sulfate, matrix metalloproteinase-9 (MMP9), fibronectin, platelet glycoprotein 1 beta alpha (GP1BA), vimentin, bone morphogenetic protein 6 (BMP6), fibroblast growth factor 4 (FGF4) and growth hormone receptor (GHR) showed maximum fold change and expression levels. CONCLUSIONS The standardized de-cellularization process could preserve majority of ECM components, providing structural integrity and architecture to the ECM. The Identified proteins quantified for their expression levels provided insight into engineering the ECM composition for developing cartilage-on-a-chip.
Collapse
Affiliation(s)
- Upasna Upadhyay
- Stem Cell Unit, Global Medical Education and Research Foundation, Lakdi-ka-pul, Hyderabad, Telangana, 500004, India.
| | - Saketh Kolla
- Department of Orthopedics, Gleneagles Global Hospitals, Lakdi-ka-pul, Hyderabad, Telangana, 500004, India.
| | - Lakshmi Kiran Chelluri
- Stem Cell Unit, Global Medical Education and Research Foundation, Lakdi-ka-pul, Hyderabad, Telangana, 500004, India; Advanced Diagnostics & Therapeutics, Gleneagles Global Hospitals, Hyderabad, 500004, India.
| |
Collapse
|
29
|
Pinto-Cardoso R, Bessa-Andrês C, Correia-de-Sá P, Bernardo Noronha-Matos J. Could hypoxia rehabilitate the osteochondral diseased interface? Lessons from the interplay of hypoxia and purinergic signals elsewhere. Biochem Pharmacol 2023:115646. [PMID: 37321413 DOI: 10.1016/j.bcp.2023.115646] [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: 04/07/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
The osteochondral unit comprises the articular cartilage (90%), subchondral bone (5%) and calcified cartilage (5%). All cells present at the osteochondral unit that is ultimately responsible for matrix production and osteochondral homeostasis, such as chondrocytes, osteoblasts, osteoclasts and osteocytes, can release adenine and/or uracil nucleotides to the local microenvironment. Nucleotides are released by these cells either constitutively or upon plasma membrane damage, mechanical stress or hypoxia conditions. Once in the extracellular space, endogenously released nucleotides can activate membrane-bound purinoceptors. Activation of these receptors is fine-tuning regulated by nucleotides' breakdown by enzymes of the ecto-nucleotidase cascade. Depending on the pathophysiological conditions, both the avascular cartilage and the subchondral bone subsist to significant changes in oxygen tension, which has a tremendous impact on tissue homeostasis. Cell stress due to hypoxic conditions directly influences the expression and activity of several purinergic signalling players, namely nucleotide release channels (e.g. Cx43), NTPDase enzymes and purinoceptors. This review gathers experimental evidence concerning the interplay between hypoxia and the purinergic signalling cascade contributing to osteochondral unit homeostasis. Reporting deviations to this relationship resulting from pathological alterations of articular joints may ultimately unravel novel therapeutic targets for osteochondral rehabilitation. At this point, one can only hypothesize how hypoxia mimetic conditions can be beneficial to the ex vivo expansion and differentiation of osteo- and chondro-progenitors for auto-transplantation and tissue regenerative purposes.
Collapse
Affiliation(s)
- Rui Pinto-Cardoso
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Catarina Bessa-Andrês
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia; Center for Drug Discovery and Innovative Medicines (MedInUP), Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP).
| |
Collapse
|
30
|
Ouyang Z, Dong L, Yao F, Wang K, Chen Y, Li S, Zhou R, Zhao Y, Hu W. Cartilage-Related Collagens in Osteoarthritis and Rheumatoid Arthritis: From Pathogenesis to Therapeutics. Int J Mol Sci 2023; 24:9841. [PMID: 37372989 DOI: 10.3390/ijms24129841] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Collagens serve essential mechanical functions throughout the body, particularly in the connective tissues. In articular cartilage, collagens provide most of the biomechanical properties of the extracellular matrix essential for its function. Collagen plays a very important role in maintaining the mechanical properties of articular cartilage and the stability of the ECM. Noteworthily, many pathogenic factors in the course of osteoarthritis and rheumatoid arthritis, such as mechanical injury, inflammation, and senescence, are involved in the irreversible degradation of collagen, leading to the progressive destruction of cartilage. The degradation of collagen can generate new biochemical markers with the ability to monitor disease progression and facilitate drug development. In addition, collagen can also be used as a biomaterial with excellent properties such as low immunogenicity, biodegradability, biocompatibility, and hydrophilicity. This review not only provides a systematic description of collagen and analyzes the structural characteristics of articular cartilage and the mechanisms of cartilage damage in disease states but also provides a detailed characterization of the biomarkers of collagen production and the role of collagen in cartilage repair, providing ideas and techniques for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Ziwei Ouyang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| | - Lei Dong
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| | - Feng Yao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Ke Wang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Shufang Li
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Renpeng Zhou
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Heifei 230601, China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Heifei 230032, China
| |
Collapse
|
31
|
Kroupa KR, Gangi LR, Zimmerman BK, Hung CT, Ateshian GA. Superficial zone chondrocytes can get compacted under physiological loading: A multiscale finite element analysis. Acta Biomater 2023; 163:248-258. [PMID: 36243365 PMCID: PMC10324087 DOI: 10.1016/j.actbio.2022.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/27/2022] [Accepted: 10/05/2022] [Indexed: 11/01/2022]
Abstract
Recent in vivo and in vitro studies have demonstrated that superficial zone (SZ) chondrocytes within articular layers of diarthrodial joints die under normal physiologic loading conditions. In order to further explore the implications of this observation in future investigations, we first needed to understand the mechanical environment of SZ chondrocytes that might cause them to die under physiological sliding contact conditions. In this study we performed a multiscale finite element analysis of articular contact to track the temporal evolution of a SZ chondrocyte's interstitial fluid pressure, hydraulic permeability, and volume under physiologic loading conditions. The effect of the pericellular matrix modulus and permeability was parametrically investigated. Results showed that SZ chondrocytes can lose ninety percent of their intracellular fluid after several hours of intermittent or continuous contact loading, resulting in a reduction of intracellular hydraulic permeability by more than three orders of magnitude. These findings are consistent with loss of cell viability due to the impediment of cellular metabolic pathways induced by the loss of fluid. They suggest that there is a simple mechanical explanation for the vulnerability of SZ chondrocytes to sustained physiological loading conditions. Future studies will focus on validating these specific findings experimentally. STATEMENT OF SIGNIFICANCE: As with any mechanical system, normal 'wear and tear' of cartilage tissue lining joints is expected. Yet incidences of osteoarthritis are uncommon in individuals younger than 45. This counter-intuitive observation suggests there must be an intrinsic repair mechanism compensating for this wear and tear over many decades of life. Recent experimental studies have shown superficial zone chondrocytes die under physiologic loading conditions, suggesting that this repair mechanism may involve cell replenishment. To better understand the mechanical environment of these cells, we performed a multiscale computational analysis of articular contact under loading. Results indicated that normal activities like walking or standing can induce significant loss of intracellular fluid volume, potentially hindering metabolic activity and fluid transport properties, and causing cell death.
Collapse
Affiliation(s)
- Kimberly R Kroupa
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, 220 S.W. Mudd, New York, NY 10027, USA
| | - Lianna R Gangi
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Brandon K Zimmerman
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, 220 S.W. Mudd, New York, NY 10027, USA
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA; Department of Orthopedic Surgery, Columbia University, 622 West 168th Street PH 11 - Center, New York, NY 10032, USA
| | - Gerard A Ateshian
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, 220 S.W. Mudd, New York, NY 10027, USA; Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA.
| |
Collapse
|
32
|
Hallström GF, Jones DL, Locke RC, Bonnevie ED, Kim SY, Laforest L, Garcia DC, Mauck RL. Microenvironmental mechanoactivation through Yap/Taz suppresses chondrogenic gene expression. Mol Biol Cell 2023; 34:ar73. [PMID: 37043309 PMCID: PMC10295477 DOI: 10.1091/mbc.e22-12-0543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/23/2023] [Accepted: 04/06/2023] [Indexed: 04/13/2023] Open
Abstract
Chondrocyte phenotype is preserved when cells are round and the actin cytoskeleton is cortical. Conversely, these cells rapidly dedifferentiate in vitro with increased mechanoactive Rho signaling, which increases cell size and causes large actin stress fiber to form. While the effects of Rho on chondrocyte phenotype are well established, the molecular mechanism is not yet fully elucidated. Yap, a transcriptional coregulator, is regulated by Rho in a mechanotransductive manner and can suppress chondrogenesis in vivo. Here, we sought to elucidate the relationship between mechanoactive Rho and Yap on chondrogenic gene expression. We first show that decreasing mechanoactive state through Rho inhibition results in a broad increase in chondrogenic gene expression. Next, we show that Yap and its coregulator Taz are negative regulators of chondrogenic gene expression, and removal of these factors promotes chondrogenesis even in environments that promote cell spreading. Finally, we establish that Yap/Taz is essential for translating Rho-mediated signals to negatively regulate chondrogenic gene expression, and that its removal negates the effects of increased Rho signaling. Together, these data indicate that Rho is a mechanoregulator of chondrogenic differentiation, and that its impact on chondrogenic expression is exerted principally through mechanically induced translocation and activity of Yap and Taz.
Collapse
Affiliation(s)
- Grey F. Hallström
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Dakota L. Jones
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
| | - Ryan C. Locke
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Edward D. Bonnevie
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Sung Yeon Kim
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Lorielle Laforest
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
| | - Diana Cruz Garcia
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104
| |
Collapse
|
33
|
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
|
34
|
Experimental Study on the Biological Outcome of Auricular Cartilage and Costal Cartilage at Different Time Periods After Autologous Cartilage Rhinoplasty. J Craniofac Surg 2023; 34:785-789. [PMID: 36168117 DOI: 10.1097/scs.0000000000009043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/04/2022] [Indexed: 11/26/2022] Open
Abstract
Since autologous cartilage is a good transplant material, it is widely used in various fields of clinical medicine. In this study, we collected clinical specimens obtained at different numbers of years after transplantation and used histologic staining to explore the post-transplantation changes in auricular cartilage and costal cartilage. A retrospective analysis was performed on patients who underwent primary autologous cartilage rhinoplasty and secondary rhinoplasty from 2017 to 2021, and the remaining autologous cartilage tissue after surgery was used for histologic testing. As time progressed after transplantation, the density of costal chondrocytes decreased first and then increased, while the secretion of type II collagen and extracellular matrix both decreased slightly. There was a clear boundary between the cartilage tissue and the surrounding connective tissue, and there was no ingrowth of blood vessels in the cartilage. Auricular cartilage showed a decrease in the integrity of the matrix edge. Moreover, local fibrosis was visible, and vascular ingrowth was observed at the edge of the cartilage. The content of type II collagen first increased and then decreased, and the cell secretion function was lower than that of normal chondrocytes. The results of the study suggest that the histologic outcome of elastic cartilage after transplantation is significantly different from that of hyaline cartilage. Moreover, costal cartilage was more stable than auricular cartilage after transplantation.
Collapse
|
35
|
Ultrashort echo time magnetization transfer imaging of knee cartilage and meniscus after long-distance running. Eur Radiol 2023:10.1007/s00330-023-09462-x. [PMID: 36814033 DOI: 10.1007/s00330-023-09462-x] [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: 07/26/2022] [Revised: 11/20/2022] [Accepted: 01/22/2023] [Indexed: 02/24/2023]
Abstract
OBJECTIVE To assess the detection of changes in knee cartilage and meniscus of amateur marathon runners before and after long-distance running using a 3D ultrashort echo time MRI sequence with magnetization transfer preparation (UTE-MT). METHODS We recruited 23 amateur marathon runners (46 knees) in this prospective cohort study. MRI scans using UTE-MT and UTE-T2* sequences were performed pre-race, 2 days post-race, and 4 weeks post-race. UTE-MT ratio (UTE-MTR) and UTE-T2* were measured for knee cartilage (eight subregions) and meniscus (four subregions). The sequence reproducibility and inter-rater reliability were also investigated. RESULTS Both the UTE-MTR and UTE-T2* measurements showed good reproducibility and inter-rater reliability. For most subregions of cartilage and meniscus, the UTE-MTR values decreased 2 days post-race and increased after 4 weeks of rest. Conversely, the UTE-T2* values increased 2 days post-race and decreased after 4 weeks. The UTE-MTR values in lateral tibial plateau, central medial femoral condyle, and medial tibial plateau showed a significant decrease at 2 days post-race compared to the other two time points (p < 0.05). By comparison, no significant UTE-T2* changes were found for any cartilage subregions. For meniscus, the UTE-MTR values in medial posterior horn and lateral posterior horn regions at 2 days post-race were significantly lower than those at pre-race and 4 weeks post-race (p < 0.05). By comparison, only the UTE-T2* values in medial posterior horn showed a significant difference. CONCLUSIONS UTE-MTR is a promising method for the detection of dynamic changes in knee cartilage and meniscus after long-distance running. KEY POINTS • Long-distance running causes changes in the knee cartilage and meniscus. • UTE-MT monitors dynamic changes of knee cartilage and meniscal non-invasively. • UTE-MT is superior to UTE-T2* in monitoring dynamic changes in knee cartilage and meniscus.
Collapse
|
36
|
Semenistaja S, Skuja S, Kadisa A, Groma V. Healthy and Osteoarthritis-Affected Joints Facing the Cellular Crosstalk. Int J Mol Sci 2023; 24:4120. [PMID: 36835530 PMCID: PMC9964755 DOI: 10.3390/ijms24044120] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Osteoarthritis (OA) is a chronic, progressive, severely debilitating, and multifactorial joint disease that is recognized as the most common type of arthritis. During the last decade, it shows an incremental global rise in prevalence and incidence. The interaction between etiologic factors that mediate joint degradation has been explored in numerous studies. However, the underlying processes that induce OA remain obscure, largely due to the variety and complexity of these mechanisms. During synovial joint dysfunction, the osteochondral unit undergoes cellular phenotypic and functional alterations. At the cellular level, the synovial membrane is influenced by cartilage and subchondral bone cleavage fragments and extracellular matrix (ECM) degradation products from apoptotic and necrotic cells. These "foreign bodies" serve as danger-associated molecular patterns (DAMPs) that trigger innate immunity, eliciting and sustaining low-grade inflammation in the synovium. In this review, we explore the cellular and molecular communication networks established between the major joint compartments-the synovial membrane, cartilage, and subchondral bone of normal and OA-affected joints.
Collapse
Affiliation(s)
- Sofija Semenistaja
- Department of Doctoral Studies, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Sandra Skuja
- Joint Laboratory of Electron Microscopy, Institute of Anatomy and Anthropology, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Anda Kadisa
- Department of Internal Diseases, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Valerija Groma
- Joint Laboratory of Electron Microscopy, Institute of Anatomy and Anthropology, Rīga Stradiņš University, LV-1007 Riga, Latvia
| |
Collapse
|
37
|
Sánchez-Porras D, Durand-Herrera D, Carmona R, Blanco-Elices C, Garzón I, Pozzobon M, San Martín S, Alaminos M, García-García ÓD, Chato-Astrain J, Carriel V. Expression of Basement Membrane Molecules by Wharton Jelly Stem Cells (WJSC) in Full-Term Human Umbilical Cords, Cell Cultures and Microtissues. Cells 2023; 12:cells12040629. [PMID: 36831296 PMCID: PMC9954414 DOI: 10.3390/cells12040629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
Wharton's jelly stem cells (WJSC) from the human umbilical cord (UC) are one of the most promising mesenchymal stem cells (MSC) in tissue engineering (TE) and advanced therapies. The cell niche is a key element for both, MSC and fully differentiated tissues, to preserve their unique features. The basement membrane (BM) is an essential structure during embryonic development and in adult tissues. Epithelial BMs are well-known, but similar structures are present in other histological structures, such as in peripheral nerve fibers, myocytes or chondrocytes. Previous studies suggest the expression of some BM molecules within the Wharton's Jelly (WJ) of UC, but the distribution pattern and full expression profile of these molecules have not been yet elucidated. In this sense, the aim of this histological study was to evaluate the expression of main BM molecules within the WJ, cultured WJSC and during WJSC microtissue (WJSC-MT) formation process. Results confirmed the presence of a pericellular matrix composed by the main BM molecules-collagens (IV, VII), HSPG2, agrin, laminin and nidogen-around the WJSC within UC. Additionally, ex vivo studies demonstrated the synthesis of these BM molecules, except agrin, especially during WJSC-MT formation process. The WJSC capability to synthesize main BM molecules could offer new alternatives for the generation of biomimetic-engineered substitutes where these molecules are particularly needed.
Collapse
Affiliation(s)
- David Sánchez-Porras
- Tissue Engineering Group, Department of Histology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Doctoral Program in Biomedicine, Doctoral School, Universidad de Granada, 18016 Granada, Spain
| | - Daniel Durand-Herrera
- Tissue Engineering Group, Department of Histology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain
- Facultad de Odontología, Universidad Michoacana de San Nicolás de Hidalgo (UMSNH), Morelia 58010, Mexico
| | - Ramón Carmona
- Department of Cell Biology, Faculty of Sciences, Universidad de Granada, 18071 Granada, Spain
| | - Cristina Blanco-Elices
- Tissue Engineering Group, Department of Histology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Ingrid Garzón
- Tissue Engineering Group, Department of Histology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Michela Pozzobon
- Department of Women and Children’s Health, University of Padova, 35129 Padova, Italy
- Corso Stati Uniti 4, Institute of Pediatric Research Città della Speranza, 35127 Padova, Italy
| | - Sebastián San Martín
- Centro de Investigaciones Biomédicas, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2520000, Chile
| | - Miguel Alaminos
- Tissue Engineering Group, Department of Histology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Óscar Darío García-García
- Tissue Engineering Group, Department of Histology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Correspondence: (Ó.D.G.-G.); (J.C.-A.)
| | - Jesús Chato-Astrain
- Tissue Engineering Group, Department of Histology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Correspondence: (Ó.D.G.-G.); (J.C.-A.)
| | - Víctor Carriel
- Tissue Engineering Group, Department of Histology, Faculty of Medicine, Universidad de Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| |
Collapse
|
38
|
Zhu D, Trinh P, Liu E, Yang F. Cell-Cell Interactions Enhance Cartilage Zonal Development in 3D Gradient Hydrogels. ACS Biomater Sci Eng 2023; 9:831-843. [PMID: 36629329 DOI: 10.1021/acsbiomaterials.2c00469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cartilage tissue is characterized by zonal organization with gradual transitions of biochemical and mechanical cues from superficial to deep zones. We previously reported that 3D gradient hydrogels made of polyethylene glycol and chondroitin sulfate can induce zonal-specific responses of chondrocytes, resulting in zonal cartilage formation that mimics native tissues. While the role of cell-matrix interactions has been studied extensively, how cell-cell interactions across different zones influence cartilage zonal development remains unknown. The goal of this study is to harness gradient hydrogels as a tool to elucidate the role of cell-cell interactions in driving cartilage zonal development. When encapsulated in intact gradient hydrogels, chondrocytes exhibited strong zonal-specific responses that mimic native cartilage zonal organization. However, the separate culture of each zone of gradient hydrogels resulted in a significant decrease in cell proliferation and cartilage matrix deposition across all zones, while the trend of zonal dependence remains. Unexpectedly, mixing the coculture of all five zones of hydrogels in the same culture well largely abolished the zonal differences, with all zones behaving similarly to the softest zone. These results suggest that paracrine signal exchange among cells in different zones is essential in driving cartilage zonal development, and a spatial organization of zones is required for proper tissue zonal development. Intact, separate, or coculture groups resulted in distinct gene expression patterns in mechanosensing and cartilage-specific markers, suggesting that cell-cell interactions can also modulate mechanosensing. We further showed that 7 days of priming in intact gradient culture was sufficient to instruct the cells to complete the zonal development, and the separate or mixed coculture after 7 days of intact culture had minimal effects on cartilage formation. This study highlights the important role of cell-cell interactions in driving cartilage zonal development and validates gradient hydrogels as a useful tool to elucidate the role of cell-matrix and cell-cell interactions in driving zonal development during tissue morphogenesis and regeneration.
Collapse
Affiliation(s)
- Danqing Zhu
- Department of Bioengineering, Stanford University, Palo Alto, California 94305, United States
| | - Pavin Trinh
- Department of Bioengineering, Stanford University, Palo Alto, California 94305, United States
| | - Elisa Liu
- Department of Bioengineering, Stanford University, Palo Alto, California 94305, United States
| | - Fan Yang
- Department of Bioengineering, Stanford University, Palo Alto, California 94305, United States.,Department of Orthopaedic Surgery, Stanford University, Palo Alto, California 94305, United States
| |
Collapse
|
39
|
Arnold KM, Sicard D, Tschumperlin DJ, Westendorf JJ. Atomic Force Microscopy Micro-Indentation Methods for Determining the Elastic Modulus of Murine Articular Cartilage. SENSORS (BASEL, SWITZERLAND) 2023; 23:1835. [PMID: 36850434 PMCID: PMC9967621 DOI: 10.3390/s23041835] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
The mechanical properties of biological tissues influence their function and can predict degenerative conditions before gross histological or physiological changes are detectable. This is especially true for structural tissues such as articular cartilage, which has a primarily mechanical function that declines after injury and in the early stages of osteoarthritis. While atomic force microscopy (AFM) has been used to test the elastic modulus of articular cartilage before, there is no agreement or consistency in methodologies reported. For murine articular cartilage, methods differ in two major ways: experimental parameter selection and sample preparation. Experimental parameters that affect AFM results include indentation force and cantilever stiffness; these are dependent on the tip, sample, and instrument used. The aim of this project was to optimize these experimental parameters to measure murine articular cartilage elastic modulus by AFM micro-indentation. We first investigated the effects of experimental parameters on a control material, polydimethylsiloxane gel (PDMS), which has an elastic modulus on the same order of magnitude as articular cartilage. Experimental parameters were narrowed on this control material, and then finalized on wildtype C57BL/6J murine articular cartilage samples that were prepared with a novel technique that allows for cryosectioning of epiphyseal segments of articular cartilage and long bones without decalcification. This technique facilitates precise localization of AFM measurements on the murine articular cartilage matrix and eliminates the need to separate cartilage from underlying bone tissues, which can be challenging in murine bones because of their small size. Together, the new sample preparation method and optimized experimental parameters provide a reliable standard operating procedure to measure microscale variations in the elastic modulus of murine articular cartilage.
Collapse
Affiliation(s)
- Katherine M. Arnold
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Delphine Sicard
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel J. Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
40
|
Wang C, Kahle ER, Li Q, Han L. Nanomechanics of Aggrecan: A New Perspective on Cartilage Biomechanics, Disease and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:69-82. [PMID: 37052847 DOI: 10.1007/978-3-031-25588-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Articular cartilage is a hydrated macromolecular composite mainly composed of type II collagen fibrils and the large proteoglycan, aggrecan. Aggrecan is a key determinant of the load bearing and energy dissipation functions of cartilage. Previously, studies of cartilage biomechanics have been primarily focusing on the macroscopic, tissue-level properties, which failed to elucidate the molecular-level activities that govern cartilage development, function, and disease. This chapter provides a brief summary of Dr. Alan J. Grodzinsky's seminal contribution to the understanding of aggrecan molecular mechanics at the nanoscopic level. By developing and applying a series of atomic force microscopy (AFM)-based nanomechanical tools, Grodzinsky and colleagues revealed the unique structural and mechanical characteristics of aggrecan at unprecedented resolutions. In this body of work, the "bottle-brush"-like ultrastructure of aggrecan was directly visualized for the first time. Meanwhile, molecular mechanics of aggrecan was studied using a physiological-like 2D biomimetic assembly of aggrecan on multiple fronts, including compression, dynamic loading, shear, and adhesion. These studies not only generated new insights into the development, aging, and disease of cartilage, but established a foundation for designing and evaluating novel cartilage regeneration strategies. For example, building on the scientific foundation and methodology infrastructure established by Dr. Grodzinsky, recent studies have elucidated the roles of other proteoglycans in mediating cartilage integrity, such as decorin and perlecan, and evaluated the therapeutic potential of biomimetic proteoglycans in improving cartilage regeneration.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Elizabeth R Kahle
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
41
|
Chen L, Li C, He A, Tong H, Lu X, Yang R, Chen X, Wu X, Wang X, Wang S, Ma J, Fu Y, Zhang T. Changes of Age-related Auricular Cartilage Plasticity and Biomechanical Property in a Rabbit Model. Laryngoscope 2023; 133:88-94. [PMID: 35385162 DOI: 10.1002/lary.30124] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/18/2022] [Accepted: 03/15/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Ear molding is an emerging technique that can correct auricular deformities. Treatment initiation time is the most important prognostic determinant of ear molding. Here, we aimed to examine why auricular cartilage plasticity appeared to diminish with age. Thus, we characterized age-related changes in the biomechanical, biochemical, and morphological properties of auricular cartilage. METHODS New Zealand rabbits were used as the experimental animal. We examined immature [postnatal 0 day (P0), 5 days (P5), 15 days (P15)], young [2 months (2M)], and mature [6 months (6M)] rabbits. Rabbits' ears were splinted and folded using adhesive fixation strips. Folding duration ranged from 1 day to 5 days to 10 days. Photographs were taken to calculate the retained fold angle. Cartilage morphology and extracellular matrix (ECM) content were examined histologically (using hematoxylin-eosin, Safranin O, elastic Van Gieson, and Masson's trichrome). Water content, DNA content, and cell density were also analyzed. Biomechanical properties were measured using a Nano indenter. RESULTS Immature ears had smaller angles after strip removal, and the angled deformation lasted a longer time. Cartilage matrix compositions, including glycosaminoglycan (GAG), elastin fiber, and collagen, increased over development. The water content, DNA content, and cell density decreased with age. Young's modulus was significantly higher in mature cartilage. CONCLUSIONS Here, we successfully established an animal model of ear molding and demonstrated that immature cartilage was associated with better plasticity. We also found that the cartilage's biomechanical property increased with the accumulation of ECM. The biomechanical change could underlie age-related shape plasticity. LEVEL OF EVIDENCE NA Laryngoscope, 133:88-94, 2023.
Collapse
Affiliation(s)
- Lili Chen
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Chenlong Li
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Aijuan He
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Hua Tong
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Xinyu Lu
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Run Yang
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Xin Chen
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Xu Wu
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Xin Wang
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Shuqi Wang
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Jing Ma
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Yaoyao Fu
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Tianyu Zhang
- ENT institute, Eye & ENT Hospital, Fudan University, Shanghai, China.,Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China.,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, China
| |
Collapse
|
42
|
He W, Wu Y, Luo Z, Yang G, Ye W, Chen X, Ren J, Liang T, Liao Z, Jiang S, Wang K. Injectable Decorin/Gellan Gum Hydrogel Encapsulating Adipose-Derived Stem Cells Enhances Anti-Inflammatory Effect in Cartilage Injury via Autophagy Signaling. Cell Transplant 2023; 32:9636897231196493. [PMID: 37688441 PMCID: PMC10493051 DOI: 10.1177/09636897231196493] [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: 06/15/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 09/10/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) are employed as a promising alternative in treating cartilage injury. Regulating the inflammatory "fingerprint" of ADSCs to improve their anti-inflammatory properties could enhance therapy efficiency. Herein, a novel injectable decorin/gellan gum hydrogel combined with ADSCs encapsulation for arthritis cartilage treatment is proposed. Decorin/gellan gum hydrogel was prepared according to the previous manufacturing protocol. The liquid-solid form transition of gellan gum hydrogel is perfectly suitable for intra-articular injection. Decorin-enriched matrix showing an immunomodulatory ability to enhance ADSCs anti-inflammatory phenotype under inflammation microenvironment by regulating autophagy signaling. This decorin/gellan gum/ADSCs hydrogel efficiently reverses interleukin-1β-induced cellular injury in chondrocytes. Through a mono-iodoacetate-induced arthritis mice model, the synergistic therapeutic effect of this ADSCs-loaded hydrogel, including inflammation attenuation and cartilage protection, is demonstrated. These results make the decorin/gellan gum hydrogel laden with ADSCs an ideal candidate for treating inflammatory joint disorders.
Collapse
Affiliation(s)
- Weiping He
- Department of Orthopedic Surgery, Dongguan Hospital of Integrated Traditional Chinese and Western Medicine, Dongguan, China
| | - Yu Wu
- Department of Plastic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhihong Luo
- Department of Orthopedic Surgery, Dongguan Hospital of Integrated Traditional Chinese and Western Medicine, Dongguan, China
| | - Genghua Yang
- Department of Orthopedic Surgery, Dongguan Hospital of Integrated Traditional Chinese and Western Medicine, Dongguan, China
| | - Woquan Ye
- Department of Orthopedic Surgery, Dongguan Hospital of Integrated Traditional Chinese and Western Medicine, Dongguan, China
| | - Xi Chen
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jianhua Ren
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Tangzhao Liang
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhiqiang Liao
- Department of Orthopedic Surgery, Dongguan Hospital of Integrated Traditional Chinese and Western Medicine, Dongguan, China
| | - Shihai Jiang
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Kun Wang
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
43
|
Ahmadian E, Eftekhari A, Janas D, Vahedi P. Nanofiber scaffolds based on extracellular matrix for articular cartilage engineering: A perspective. Nanotheranostics 2023; 7:61-69. [PMID: 36593799 PMCID: PMC9760364 DOI: 10.7150/ntno.78611] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/27/2022] [Indexed: 11/24/2022] Open
Abstract
Articular cartilage has a low self-repair capacity due to the lack of vessels and nerves. In recent times, nanofiber scaffolds have been widely used for this purpose. The optimum nanofiber scaffold should stimulate new tissue's growth and mimic the articular cartilage nature. Furthermore, the characteristics of the scaffold should match those of the cellular matrix components of the native tissue to best merge with the target tissue. Therefore, selective modification of prefabricated scaffolds based on the structure of the repaired tissues is commonly conducted to promote restoring the tissue. A thorough analysis is required to find out the architectural features of scaffolds that are essential to make the treatment successful. The current review aims to target this challenge. The article highlights different optimization approaches of nanofibrous scaffolds for improved cartilage tissue engineering. In this context, the influence of the architecture of nanoscaffolds on performance is discussed in detail. Finally, based on the gathered information, a future outlook is provided to catalyze development in this promising field.
Collapse
Affiliation(s)
- Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Eftekhari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran,✉ Corresponding authors: Aziz Eftekhari (), Dawid Janas (), Parviz Vahedi ()
| | - Dawid Janas
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100, Gliwice, Poland,✉ Corresponding authors: Aziz Eftekhari (), Dawid Janas (), Parviz Vahedi ()
| | - Parviz Vahedi
- Department of Anatomical Sciences, Maragheh University of Medical Sciences, Maragheh 78151-55158, Iran,✉ Corresponding authors: Aziz Eftekhari (), Dawid Janas (), Parviz Vahedi ()
| |
Collapse
|
44
|
Mucopolysaccharidoses: Cellular Consequences of Glycosaminoglycans Accumulation and Potential Targets. Int J Mol Sci 2022; 24:ijms24010477. [PMID: 36613919 PMCID: PMC9820209 DOI: 10.3390/ijms24010477] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/09/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022] Open
Abstract
Mucopolysaccharidoses (MPSs) constitute a heterogeneous group of lysosomal storage disorders characterized by the lysosomal accumulation of glycosaminoglycans (GAGs). Although lysosomal dysfunction is mainly affected, several cellular organelles such as mitochondria, endoplasmic reticulum, Golgi apparatus, and their related process are also impaired, leading to the activation of pathophysiological cascades. While supplying missing enzymes is the mainstream for the treatment of MPS, including enzyme replacement therapy (ERT), hematopoietic stem cell transplantation (HSCT), or gene therapy (GT), the use of modulators available to restore affected organelles for recovering cell homeostasis may be a simultaneous approach. This review summarizes the current knowledge about the cellular consequences of the lysosomal GAGs accumulation and discusses the use of potential modulators that can reestablish normal cell function beyond ERT-, HSCT-, or GT-based alternatives.
Collapse
|
45
|
Höflsauer S, Bonnaire FC, Bamberger CE, Danalache M, Feierabend M, Hofmann UK. Changes in stiffness of the extracellular and pericellular matrix in the anulus fibrosus of lumbar intervertebral discs over the course of degeneration. Front Bioeng Biotechnol 2022; 10:1006615. [PMID: 36619385 PMCID: PMC9816436 DOI: 10.3389/fbioe.2022.1006615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/08/2022] [Indexed: 12/24/2022] Open
Abstract
Analogous to articular cartilage, changes in spatial chondrocyte organisation have been proposed to be a strong indicator for local tissue degeneration in the intervertebral disc (IVD). While a progressive structural and functional degradation of the extracellular (ECM) and pericellular (PCM) matrix occurs in osteoarthritic cartilage, these processes have not yet been biomechanically elucidated in the IVD. We aimed to evaluate the local stiffness of the ECM and PCM in the anulus fibrosus of the IVD on the basis of local chondrocyte spatial organisation. Using atomic force microscopy, we measured the Young's modulus of the local ECM and PCM in human and bovine disc samples using the spatial chondrocyte patterns as an image-based biomarker. By measuring tissue from 31 patients and six bovine samples, we found a significant difference in the elastic moduli (E) of the PCM in clusters when compared to the healthy patterns single cells (p = 0.029), pairs (p = 0.016), and string-formations (p = 0.010). The ECM/PCM ratio ranged from 0.62-0.89. Interestingly, in the bovine IVD, the ECM/PCM ratio of the E significantly varied (p = 0.002) depending on the tissue origin. Overall the reduced E in clusters demonstrates that cluster formation is not only a morphological phenomenon describing disc degeneration, but it marks a compromised biomechanical functioning. Immunohistochemical analyses indicate that collagen type III degradation might be involved. This study is the first to describe and quantify the differences in the E of the ECM in relation to the PCM in the anulus fibrosus of the IVD by means of atomic force microscopy on the basis of spatial chondrocyte organisation.
Collapse
Affiliation(s)
- Sebastian Höflsauer
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, Tübingen, Germany,Medical Faculty of the University of Tübingen, Tübingen, Germany
| | - Florian Christof Bonnaire
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, Tübingen, Germany,Department of Orthopaedic Surgery, University Hospital of Tübingen, Tübingen, Germany
| | - Charlotte Emma Bamberger
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, Tübingen, Germany,Medical Faculty of the University of Tübingen, Tübingen, Germany
| | - Marina Danalache
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, Tübingen, Germany,Medical Faculty of the University of Tübingen, Tübingen, Germany
| | - Martina Feierabend
- Institute for Bioinformatics and Medical Informatics, Faculty of Science of the University of Tübingen, Tübingen, Germany,*Correspondence: Martina Feierabend,
| | - Ulf Krister Hofmann
- Department of Orthopaedic Surgery, University Hospital of Tübingen, Tübingen, Germany,Department of Orthopaedic Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
46
|
Al-Maslamani NA, Oldershaw R, Tew S, Curran J, D’Hooghe P, Yamamoto K, Horn HF. Chondrocyte De-Differentiation: Biophysical Cues to Nuclear Alterations. Cells 2022; 11:cells11244011. [PMID: 36552775 PMCID: PMC9777101 DOI: 10.3390/cells11244011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Autologous chondrocyte implantation (ACI) is a cell therapy to repair cartilage defects. In ACI a biopsy is taken from a non-load bearing area of the knee and expanded in-vitro. The expansion process provides the benefit of generating a large number of cells required for implantation; however, during the expansion these cells de-differentiate and lose their chondrocyte phenotype. In this review we focus on examining the de-differentiation phenotype from a mechanobiology and biophysical perspective, highlighting some of the nuclear mechanics and chromatin changes in chondrocytes seen during the expansion process and how this relates to the gene expression profile. We propose that manipulating chondrocyte nuclear architecture and chromatin organization will highlight mechanisms that will help to preserve the chondrocyte phenotype.
Collapse
Affiliation(s)
- Noor A. Al-Maslamani
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Correspondence:
| | - Rachel Oldershaw
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Simon Tew
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Jude Curran
- Department of Mechanical, Materials and Aerospace Engineering, School of Engineering, University of Liverpool, Liverpool L69 3GH, UK
| | - Pieter D’Hooghe
- Department of Orthopaedic Surgery, Aspetar Orthopaedic and Sports Medicine Hospital, Doha P.O. Box 29222, Qatar
| | - Kazuhiro Yamamoto
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Henning F. Horn
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
| |
Collapse
|
47
|
Kahle ER, Patel N, Sreenivasappa HB, Marcolongo MS, Han L. Targeting cell-matrix interface mechanobiology by integrating AFM with fluorescence microscopy. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 176:67-81. [PMID: 36055517 PMCID: PMC9691605 DOI: 10.1016/j.pbiomolbio.2022.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/14/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Mechanosensing at the interface of a cell and its surrounding microenvironment is an essential driving force of physiological processes. Understanding molecular activities at the cell-matrix interface has the potential to provide novel targets for improving tissue regeneration and early disease intervention. In the past few decades, the advancement of atomic force microscopy (AFM) has offered a unique platform for probing mechanobiology at this crucial microdomain. In this review, we describe key advances under this topic through the use of an integrated system of AFM (as a biomechanical testing tool) with complementary immunofluorescence (IF) imaging (as an in situ navigation system). We first describe the body of work investigating the micromechanics of the pericellular matrix (PCM), the immediate cell micro-niche, in healthy, diseased, and genetically modified tissues, with a focus on articular cartilage. We then summarize the key findings in understanding cellular biomechanics and mechanotransduction, in which, molecular mechanisms governing transmembrane ion channel-mediated mechanosensing, cytoskeleton remodeling, and nucleus remodeling have been studied in various cell and tissue types. Lastly, we provide an overview of major technical advances that have enabled more in-depth studies of mechanobiology, including the integration of AFM with a side-view microscope, multiple optomicroscopy, a fluorescence recovery after photobleaching (FRAP) module, and a tensile stretching device. The innovations described here have contributed greatly to advancing the fundamental knowledge of extracellular matrix biomechanics and cell mechanobiology for improved understanding, detection, and intervention of various diseases.
Collapse
Affiliation(s)
- Elizabeth R Kahle
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Neil Patel
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Harini B Sreenivasappa
- Cell Imaging Center, Office of Research and Innovation, Drexel University, PA 19104, United States
| | - Michele S Marcolongo
- Department of Mechanical Engineering, Villanova University, Villanova, PA 19085, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
48
|
Pei S, Zhou Y, Li Y, Azar T, Wang W, Kim DG, Liu XS. Instrumented nanoindentation in musculoskeletal research. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 176:38-51. [PMID: 35660010 DOI: 10.1016/j.pbiomolbio.2022.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Musculoskeletal tissues, such as bone, cartilage, and muscle, are natural composite materials that are constructed with a hierarchical structure ranging from the cell to tissue level. The component differences and structural complexity, together, require comprehensive multiscale mechanical characterization. In this review, we focus on nanoindentation testing, which is used for nanometer to sub-micrometer length scale mechanical characterization. In the following context, we will summarize studies of nanoindentation in musculoskeletal research, examine the critical factors that affect nanoindentation testing results, and briefly summarize other commonly used techniques that can be conjoined with nanoindentation for synchronized imaging and colocalized characterization.
Collapse
Affiliation(s)
- Shaopeng Pei
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Yilu Zhou
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Yihan Li
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Tala Azar
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Wenzheng Wang
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States; Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Do-Gyoon Kim
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA
| | - X Sherry Liu
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States.
| |
Collapse
|
49
|
Zhang H, Li Q, Xu X, Zhang S, Chen Y, Yuan T, Zeng Z, Zhang Y, Mei Z, Yan S, Zhang L, Wei S. Functionalized Microscaffold-Hydrogel Composites Accelerating Osteochondral Repair through Endochondral Ossification. ACS APPLIED MATERIALS & INTERFACES 2022; 14:52599-52617. [PMID: 36394998 DOI: 10.1021/acsami.2c12694] [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: 06/16/2023]
Abstract
Osteochondral regeneration remains a key challenge because of the limited self-healing ability of the bone and its complex structure and composition. Biomaterials based on endochondral ossification (ECO) are considered an attractive candidate to promote bone repair because they can effectively address the difficulties in establishing vascularization and poor bone regeneration via intramembranous ossification (IMO). However, its clinical application is limited by the complex cellular behavior of ECO and the long time required for induction of the cell cycle. Herein, functionalized microscaffold-hydrogel composites are developed to accelerate the developmental bone growth process via recapitulating ECO. The design comprises arginine-glycine-aspartic acid (RGD)-peptide-modified microscaffolds loaded with kartogenin (KGN) and wrapped with a layer of RGD- and QK-peptide-comodified alginate hydrogel. These microscaffolds enhance the proliferation and aggregation behavior of the human bone marrow mesenchymal stem cells (hBMSCs); the controlled release of kartogenin induces the differentiation of hBMSCs into chondrocytes; and the hydrogel grafted with RGD and QK peptide facilitates chondrocyte hypertrophy, which creates a vascularized niche for osteogenesis and finally accelerates osteochondral repair in vivo. The findings provide an efficient bioengineering approach by sequentially modulating cellular ECO behavior for osteochondral defect repair.
Collapse
Affiliation(s)
- He Zhang
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Qian Li
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| | - Xiangliang Xu
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Siqi Zhang
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| | - Yang Chen
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| | - Tao Yuan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Tumor Biology, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Ziqian Zeng
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Yifei Zhang
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Zi Mei
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Shuang Yan
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Lei Zhang
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Shicheng Wei
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| |
Collapse
|
50
|
Zhao XX, Xie WQ, Xiao WF, Li HZ, Naranmandakh S, Bruyere O, Reginster JY, Li YS. Perlecan: Roles in osteoarthritis and potential treating target. Life Sci 2022; 312:121190. [PMID: 36379311 DOI: 10.1016/j.lfs.2022.121190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
Osteoarthritis (OA) is the most common joint disease, affecting hundreds of millions of people globally, which leads to a high cost of treatment and further medical care and an apparent decrease in patient prognosis. The recent view of OA pathogenesis is that increased vascularity, bone remodeling, and disordered turnover are influenced by multivariate risk factors, such as age, obesity, and overloading. The view also reveals the gap between the development of these processes and early stage risk factors. This review presents the latest research on OA-related signaling pathways and analyzes the potential roles of perlecan, a typical component of the well-known protective structure against osteoarthritic pericellular matrix (PCM). Based on the experimental results observed in end-stage OA models, we summarized and analyzed the role of perlecan in the development of OA. In normal cartilage, it plays a protective role by maintaining the integrin of PCM and sequesters growth factors. Second, perlecan in cartilage is required to not only activate vascular epithelium growth factor receptor (VEGFR) signaling of endothelial cells for vascular invasion and catabolic autophagy, but also for different signaling pathways for the catabolic and anabolic actions of chondrocytes. Finally, perlecan may participate in pain sensitization pathways.
Collapse
Affiliation(s)
- Xiao-Xuan Zhao
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya School of Medicine, Central South University, Changsha 410083, Hunan, China
| | - Wen-Qing Xie
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Wen-Feng Xiao
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Heng-Zhen Li
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Shinen Naranmandakh
- School of Arts and Sciences, National University of Mongolia, Sukhbaatar district, 14201 Ulaanbaatar, Mongolia
| | - Olivier Bruyere
- Department of Public Health, Epidemiology and Health Economics, University of Liège, CHU Sart Tilman B23, 4000 Liège, Belgium
| | - Jean-Yves Reginster
- Department of Public Health, Epidemiology and Health Economics, University of Liège, CHU Sart Tilman B23, 4000 Liège, Belgium.
| | - Yu-Sheng Li
- Deparment of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|