101
|
Abstract
I review experimental developments in the growth and application of surface-grafted weak polyelectrolytes (brushes), concentrating on their surface, tribological, and adhesive and bioadhesive properties, and their role as actuators.
Collapse
Affiliation(s)
- Mark Geoghegan
- School of Engineering, Newcastle University, Merz Court, Newcastle-upon-Tyne NE1 7RU, UK.
| |
Collapse
|
102
|
Cui X, Alcala-Orozco CR, Baer K, Li J, Murphy C, Durham M, Lindberg G, Hooper GJ, Lim K, Woodfield TBF. 3D bioassembly of cell-instructive chondrogenic and osteogenic hydrogel microspheres containing allogeneic stem cells for hybrid biofabrication of osteochondral constructs. Biofabrication 2022; 14. [PMID: 35344942 DOI: 10.1088/1758-5090/ac61a3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/28/2022] [Indexed: 12/21/2022]
Abstract
Recently developed modular bioassembly techniques hold tremendous potential in tissue engineering and regenerative medicine, due to their ability to recreate the complex microarchitecture of native tissue. Here, we developed a novel approach to fabricate hybrid tissue-engineered constructs adopting high-throughput microfluidic and 3D bioassembly strategies. Osteochondral tissue fabrication was adopted as an example in this study, because of the challenges in fabricating load bearing osteochondral tissue constructs with phenotypically distinct zonal architecture. By developing cell-instructive chondrogenic and osteogenic bioink microsphere modules in high-throughput, together with precise manipulation of the 3D bioassembly process, we successfully fabricated hybrid engineered osteochondral tissue in vitro with integrated but distinct cartilage and bone layers. Furthermore, by encapsulating allogeneic umbilical cord blood-derived mesenchymal stromal cells (UCB-MSCs), and demonstrating chondrogenic and osteogenic differentiation, the hybrid biofabrication of hydrogel microspheres in this 3D bioassembly model offers potential for an off-the-shelf, single-surgery strategy for osteochondral tissue repair.
Collapse
Affiliation(s)
- Xiaolin Cui
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Cesar R Alcala-Orozco
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Kenzie Baer
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Jun Li
- Dept. of Orthopaedic Surgery , University of Otago, 2 Riccarton Avenue, Christchurch, Christchurch, Canterbury, 8011, NEW ZEALAND
| | - Caroline Murphy
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Mitch Durham
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Gabriella Lindberg
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| | - Gary J Hooper
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8041, NEW ZEALAND
| | - Khoon Lim
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Avenue, Christchurch, 8140, NEW ZEALAND
| | - Tim B F Woodfield
- Department of Orthopaedic Surgery, Centre for Bioengineering & Nanomedicine, University of Otago Christchurch, 2 Riccarton Ave, Christchurch, 8140, NEW ZEALAND
| |
Collapse
|
103
|
Yin LP, Zheng HX, Zhu H. Short stature associated with a novel mutation in the aggrecan gene: A case report and literature review. World J Clin Cases 2022; 10:2811-2817. [PMID: 35434101 PMCID: PMC8968812 DOI: 10.12998/wjcc.v10.i9.2811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/02/2021] [Accepted: 02/20/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mutations in the aggrecan (ACAN) gene are identified in patients with: spondyloepiphyseal dysplasia, Kimberley type; short stature with advanced bone age (BA); in the presence or absence of heterozygous ACAN mutation-induced early-onset osteoarthritis and/or osteochondritis dissecans; and spondyloepimetaphyseal dysplasia, ACAN type. Heterozygous mutations contribute to spondyloepiphyseal dysplasia, Kimberley type (MIM#608361), which is a milder skeletal dysplasia. In contrast, homozygous mutations cause a critical skeletal dysplasia, which is called spondyloepimetaphyseal dysplasia, ACAN type (MIM#612813). Lately, investigations on exome and genome sequencing have shown that ACAN mutations can also lead to idiopathic short stature with or without an advanced BA, in the presence or absence of early-onset osteoarthritis and/or osteochondritis dissecans (MIM#165800). We herein reported a heterozygous defect of ACAN in a family with autosomal dominant short stature, BA acceleration, and premature growth cessation.
CASE SUMMARY A 2-year-old male patient visited us due to growth retardation. The patient presented symmetrical short stature (height 79 cm, < -2 SD) without facial features and other congenital abnormalities. Whole-exome sequencing revealed a heterozygous pathogenic variant c. 871C>T (p. Gln291*) of ACAN, which was not yet reported in cases of short stature. This mutation was also detected in his father and paternal grandmother. According to the Human Gene Mutation Database, 67 ACAN mutations are registered. Most of these mutations are genetically inheritable, and very few children with short stature are associated with ACAN mutations. To date, heterozygous ACAN mutations have been reported in approximately 40 families worldwide, including a few individuals with a decelerated BA.
CONCLUSION Heterozygous c. 871C>T (p. Gln291*) variation of the ACAN gene was the disease-causing variant in this family. Collectively, our newly discovered mutation expanded the spectrum of ACAN gene mutations.
Collapse
Affiliation(s)
- Li-Ping Yin
- Department of Paediatrics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou 213000, Jiangsu Province, China
| | - Hong-Xue Zheng
- Department of Paediatrics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou 213000, Jiangsu Province, China
| | - Hong Zhu
- Department of Paediatrics, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou 213000, Jiangsu Province, China
| |
Collapse
|
104
|
Immunohistochemical Detection of Various Proteoglycans in the Extracellular Matrix of Zebra Mussels. FISHES 2022. [DOI: 10.3390/fishes7020074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Mussels have been used as animal models for studying ecotoxicology, biomineralization, and bio-adhesion for many years. Despite a wealth of studies on their shell matrix and byssus proteins, few studies have focused on the extracellular matrix molecules in mussel soft tissues. Extracellular matrix molecules may play important roles in biomineralization, immune reaction, and tissue homeostasis. In the present study, extracellular matrix and mineralization-related molecules in zebra mussel soft tissue were immunolocalized using well-characterized monoclonal antibodies. Our results demonstrate specific immunolocalization for collagen IV, fibronectin, and keratan sulfate in hemocytes; collagen IV in peripheral nerves; and aggrecan, link protein, and collagen XVIII in foot tissue. Laminin, decorin, and osteonectin were also broadly immunolocalized in mussel soft tissues. The distributions of these extracellular matrix molecules in mussel tissues are in line with the cell-mediated shell mineralization hypothesis, providing evidence for the molecules involved in the peripheral nervous system and byssus formation, and explaining the conservation of extracellular matrix molecules during evolution. These results further contribute to establishing zebra mussels as an attractive animal model in biomedical research.
Collapse
|
105
|
Histone Extraction from Human Articular Cartilage for the Study of Epigenetic Regulation in Osteoarthritis. Int J Mol Sci 2022; 23:ijms23063355. [PMID: 35328777 PMCID: PMC8955682 DOI: 10.3390/ijms23063355] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
Osteoarthritis (OA) is a chronic disease that affects articular cartilage, causing its degeneration. Although OA is one of the most prevalent pathologies globally, there are no definitive treatments available. Recently, research has focused on elucidating the complex interplay that takes place between inflammatory processes and epigenetic regulation, showing that histone post-translational modifications (PTMs) can exert a pronounced effect on the expression of OA-related genes. OA chondrocytes enhance the production of interleukin 1β (IL-1β) and interleukin 8 (IL-8), which are epigenetically regulated. These cytokines upregulate the synthesis of matrix metalloproteinases (MMPs) and aggrecanases, which promote the extracellular matrix (ECM) destruction. This motivates the study of histone PTMs to investigate the epigenetic regulation of proinflammatory molecules, but the absence of specific protocols to extract histones from human articular cartilage has complicated this task. The lack of effective methods can be explained by the structural complexity and low cellularity of this tissue, which are responsible for the biomechanical properties that allow the movement of the joint but also complicate histone isolation. Here, we provide a histone extraction procedure specifically adapted for cryopreserved human articular cartilage that can be useful to understand epigenetic regulation in OA and accelerate the search for novel strategies.
Collapse
|
106
|
Kakizaki I, Kato Y. A simple quality evaluation method for proteoglycan after addition to beverages. J Appl Glycosci (1999) 2022; 69:45-48. [PMID: 35891900 PMCID: PMC9276523 DOI: 10.5458/jag.jag.jag-2021_0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/15/2022] [Indexed: 10/25/2022] Open
Affiliation(s)
- Ikuko Kakizaki
- Department of Glycotechnology, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine
| | | |
Collapse
|
107
|
Aidos L, Modina SC, Millar VRH, Peretti GM, Mangiavini L, Ferroni M, Boschetti F, Di Giancamillo A. Meniscus Matrix Structural and Biomechanical Evaluation: Age-Dependent Properties in a Swine Model. Bioengineering (Basel) 2022; 9:bioengineering9030117. [PMID: 35324808 PMCID: PMC8945511 DOI: 10.3390/bioengineering9030117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 11/16/2022] Open
Abstract
The analysis of the morphological, structural, biochemical, and mechanical changes of the Extracellular Matrix (ECM), which occur during meniscus development, represents the goal of the present study. Medial fully developed menisci (FD, 9-month-old pigs), partially developed menisci (PD, 1-month-old piglets), and not developed menisci (ND, from stillbirths) were collected. Cellularity and glycosaminoglycans (GAGs) deposition were evaluated by ELISA, while Collagen 1 and aggrecan were investigated by immunohistochemistry and Western blot analyses in order to be compared to the biomechanical properties of traction and compression tensile forces, respectively. Cellularity decreased from ND to FD and GAGs showed the opposite trend (p < 0.01 both). Collagen 1 decreased from ND to FD, as well as the ability to resist to tensile traction forces (p < 0.01), while aggrecan showed the opposite trend, in accordance with the biomechanics: compression test showed that FD meniscus greatly resists to deformation (p < 0.01). This study demonstrated that in swine meniscus, clear morphological and biomechanical changes follow the meniscal maturation and specialization during growth, starting with an immature pattern (ND) to the mature organized meniscus of the FD, and they could be useful to understand the behavior of this structure in the light of its tissue bioengineering.
Collapse
Affiliation(s)
- Lucia Aidos
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (L.A.); (V.R.H.M.); (G.M.P.); (L.M.)
| | - Silvia Clotilde Modina
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 26900 Lodi, Italy;
| | - Valentina Rafaela Herrera Millar
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (L.A.); (V.R.H.M.); (G.M.P.); (L.M.)
| | - Giuseppe Maria Peretti
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (L.A.); (V.R.H.M.); (G.M.P.); (L.M.)
- IRCCS, Istituto Ortopedico Galeazzi, 20161 Milano, Italy;
| | - Laura Mangiavini
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (L.A.); (V.R.H.M.); (G.M.P.); (L.M.)
- IRCCS, Istituto Ortopedico Galeazzi, 20161 Milano, Italy;
| | - Marco Ferroni
- Department of Chemistry, Material and Chemical Engineering “Giulio Natta”, Politecnico di Milano, 20133 Milano, Italy;
| | - Federica Boschetti
- IRCCS, Istituto Ortopedico Galeazzi, 20161 Milano, Italy;
- Department of Chemistry, Material and Chemical Engineering “Giulio Natta”, Politecnico di Milano, 20133 Milano, Italy;
| | - Alessia Di Giancamillo
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (L.A.); (V.R.H.M.); (G.M.P.); (L.M.)
- Correspondence: ; Tel.: +39-02503-34606
| |
Collapse
|
108
|
Chremos A, Horkay F, Douglas JF. Influence of network defects on the conformational structure of nanogel particles: From "closed compact" to "open fractal" nanogel particles. J Chem Phys 2022; 156:094903. [PMID: 35259888 PMCID: PMC8898093 DOI: 10.1063/5.0072274] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/30/2021] [Indexed: 12/11/2022] Open
Abstract
We propose an approach to generate a wide range of randomly branched polymeric structures to gain general insights into how polymer topology encodes a configurational structure in solution. Nanogel particles can take forms ranging from relatively symmetric sponge-like compact structures to relatively anisotropic open fractal structures observed in some nanogel clusters and in some self-associating polymers in solutions, such as aggrecan solutions under physiologically relevant conditions. We hypothesize that this broad "spectrum" of branched polymer structures derives from the degree of regularity of bonding in the network defining these structures. Accordingly, we systematically introduce bonding defects in an initially perfect network having a lattice structure in three and two topological dimensions corresponding to "sponge" and "sheet" structures, respectively. The introduction of bonding defects causes these "closed" and relatively compact nanogel particles to transform near a well-defined bond percolation threshold into "open" fractal objects with the inherent anisotropy of randomly branched polymers. Moreover, with increasing network decimation, the network structure of these polymers acquires other configurational properties similar to those of randomly branched polymers. In particular, the mass scaling of the radius of gyration and its eigenvalues, as well as hydrodynamic radius, intrinsic viscosity, and form factor for scattering, all undergo abrupt changes that accompany these topological transitions. Our findings support the idea that randomly branched polymers can be considered to be equivalent to perforated sheets from a "universality class" standpoint. We utilize our model to gain insight into scattering measurements made on aggrecan solutions.
Collapse
Affiliation(s)
- Alexandros Chremos
- Section on Quantitative Imaging and Tissue Sciences, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ferenc Horkay
- Section on Quantitative Imaging and Tissue Sciences, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jack F. Douglas
- Materials Science and Engineering Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| |
Collapse
|
109
|
Nguyen M, Panitch A. Proteoglycans and proteoglycan mimetics for tissue engineering. Am J Physiol Cell Physiol 2022; 322:C754-C761. [PMID: 35235426 PMCID: PMC8993519 DOI: 10.1152/ajpcell.00442.2021] [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: 11/22/2022]
Abstract
Proteoglycans play a crucial role in proper tissue morphology and function throughout the body that is defined by a combination of their core protein and the attached glycosaminoglycan chains. Although they serve a myriad of roles, the functions of extracellular proteoglycans can be generally sorted into four categories: modulation of tissue mechanical properties, regulation and protection of the extracellular matrix, sequestering of proteins, and regulation of cell signaling. The loss of proteoglycans can result in significant tissue disfunction, ranging from poor mechanical properties to uncontrolled inflammation. Because of the key roles they play in proper tissue function and due to their complex synthesis, the past two decades have seen significant research into the development of proteoglycan mimetic molecules to recapitulate the function of proteoglycans for therapeutic and tissue engineering applications. These strategies have ranged from semisynthetic graft copolymers to recombinant proteoglycan domains synthesized by genetically engineered cells. In this review, we highlight some of the important functions of extracellular proteoglycans, as well as the strategies developed to recapitulate these functions.
Collapse
Affiliation(s)
- Michael Nguyen
- Department of Biomedical Engineering, University of California, Davis, CA, United States
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, CA, United States.,Department of Surgery, UC Davis Health, University of California, Sacramento, CA, United States
| |
Collapse
|
110
|
De Kinderen P, Meester J, Loeys B, Peeters S, Gouze E, Woods S, Mortier G, Verstraeten A. Differentiation of Induced Pluripotent Stem Cells Into Chondrocytes: Methods and Applications for Disease Modeling and Drug Discovery. J Bone Miner Res 2022; 37:397-410. [PMID: 35124831 DOI: 10.1002/jbmr.4524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/25/2022] [Accepted: 02/01/2022] [Indexed: 11/11/2022]
Abstract
Induced pluripotent stem cell (iPSC) technology allows pathomechanistic and therapeutic investigation of human heritable disorders affecting tissue types whose collection from patients is difficult or even impossible. Among them are cartilage diseases. Over the past decade, iPSC-chondrocyte disease models have been shown to exhibit several key aspects of known disease mechanisms. Concurrently, an increasing number of protocols to differentiate iPSCs into chondrocytes have been published, each with its respective (dis)advantages. In this review we provide a comprehensive overview of the different differentiation approaches, the hitherto described iPSC-chondrocyte disease models and mechanistic and/or therapeutic insights that have been derived from their investigation, and the current model limitations. Key lessons are that the most appropriate differentiation approach is dependent upon the cartilage disease under investigation and that further optimization is still required to recapitulate the in vivo cartilage. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Pauline De Kinderen
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Josephina Meester
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Bart Loeys
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.,Department of Human Genetics, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Silke Peeters
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Elvire Gouze
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | - Steven Woods
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Geert Mortier
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Aline Verstraeten
- Centre of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
111
|
Ma Z, Li DX, Kunze M, Mulet-Sierra A, Westover L, Adesida AB. Engineered Human Meniscus in Modeling Sex Differences of Knee Osteoarthritis in Vitro. Front Bioeng Biotechnol 2022; 10:823679. [PMID: 35284415 PMCID: PMC8904202 DOI: 10.3389/fbioe.2022.823679] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/21/2022] [Indexed: 12/30/2022] Open
Abstract
Background: Osteoarthritis (OA) primarily affects mechanical load-bearing joints. The knee joint is the most impacted by OA. Knee OA (KOA) occurs in almost all demographic groups, but the prevalence and severity are disproportionately higher in females. The molecular mechanism underlying the pathogenesis and progression of KOA is unknown. The molecular basis of biological sex matters of KOA is not fully understood. Mechanical stimulation plays a vital role in modulating OA-related responses of load-bearing tissues. Mechanical unloading by simulated microgravity (SMG) induced OA-like gene expression in engineered cartilage, while mechanical loading by cyclic hydrostatic pressure (CHP), on the other hand, exerted a pro-chondrogenic effect. This study aimed to evaluate the effects of mechanical loading and unloading via CHP and SMG, respectively, on the OA-related profile changes of engineered meniscus tissues and explore biological sex-related differences.Methods: Tissue-engineered menisci were made from female and male meniscus fibrochondrocytes (MFCs) under static conditions of normal gravity in chondrogenic media and subjected to SMG and CHP culture. Constructs were assayed via histology, immunofluorescence, GAG/DNA assays, RNA sequencing, and testing of mechanical properties.Results: The mRNA expression of ACAN and COL2A1, was upregulated by CHP but downregulated by SMG. COL10A1, a marker for chondrocyte hypertrophy, was downregulated by CHP compared to SMG. Furthermore, CHP increased GAG/DNA levels and wet weight in both female and male donors, but only significantly in females. From the transcriptomics, CHP and SMG significantly modulated genes related to the ossification, regulation of ossification, extracellular matrix, and angiogenesis Gene Ontology (GO) terms. A clear difference in fold-change magnitude and direction was seen between the two treatments for many of the genes. Furthermore, differences in fold-change magnitudes were seen between male and female donors within each treatment. SMG and CHP also significantly modulated genes in OA-related KEGG pathways, such as mineral absorption, Wnt signalling pathway, and HIF-1 signalling pathway.Conclusion: Engineered menisci responded to CHP and SMG in a sex-dependent manner. SMG may induce an OA-like profile, while CHP promotes chondrogenesis. The combination of SMG and CHP could serve as a model to study the early molecular events of KOA and potential drug-targetable pathways.
Collapse
Affiliation(s)
- Zhiyao Ma
- Department of Surgery, Divisions of Orthopaedic Surgery, Surgical Research and Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - David Xinzheyang Li
- Department of Surgery, Divisions of Orthopaedic Surgery, Surgical Research and Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Civil and Environmental Engineering, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Department of Surgery, Divisions of Orthopaedic Surgery, Surgical Research and Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Department of Surgery, Divisions of Orthopaedic Surgery, Surgical Research and Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Lindsey Westover
- Department of Mechanical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Adetola B. Adesida
- Department of Surgery, Divisions of Orthopaedic Surgery, Surgical Research and Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Adetola B. Adesida,
| |
Collapse
|
112
|
Zhulina EB, Borisov OV. Bottlebrush polymer gels: architectural control over swelling and osmotic bulk modulus. SOFT MATTER 2022; 18:1239-1246. [PMID: 35043819 DOI: 10.1039/d1sm01575c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Swelling behaviour and bulk moduli of polymer gels comprising of crosslinked bottlebrush subchains enable fine tuning by varying polymerization degrees of the main and side chains of the bottlebrush strands as well as their grafting densities. By using scaling approach we predict power law dependences of structural and elastic properties of swollen bottlebrush gels on the set of relevant architectural parameters and construct phase diagrams consisting of regions corresponding to different power law asymptotics for these dependences. In particular, our theory predict that bulk elastic modulus of the gel exhibits non-monotonous dependence on the degree of polymerization of side chains of the bottlebrush strands.
Collapse
Affiliation(s)
- Ekaterina B Zhulina
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Oleg V Borisov
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, St. Petersburg, Russia
- Institut des Sciences Analytiques et de Physico-Chimie pour l'Environnement et les Matériaux, UMR 5254 CNRS UPPA, Pau, France.
| |
Collapse
|
113
|
Sim HJ, Cho C, Kim HE, Hong JY, Song EK, Kwon KY, Jang DG, Kim SJ, Lee HS, Lee C, Kwon T, Yang S, Park TJ. Augmented ERAD (ER-associated degradation) activity in chondrocytes is necessary for cartilage development and maintenance. SCIENCE ADVANCES 2022; 8:eabl4222. [PMID: 35061535 PMCID: PMC8782459 DOI: 10.1126/sciadv.abl4222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/29/2021] [Indexed: 05/28/2023]
Abstract
Chondrocytes secrete massive extracellular matrix (ECM) molecules that are produced, folded, and modified in the endoplasmic reticulum (ER). Thus, the ER-associated degradation (ERAD) complex-which removes misfolded and unfolded proteins to maintain proteostasis in the ER- plays an indispensable role in building and maintaining cartilage. Here, we examined the necessity of the ERAD complex in chondrocytes for cartilage formation and maintenance. We show that ERAD gene expression is exponentially increased during chondrogenesis, and disruption of ERAD function causes severe chondrodysplasia in developing embryos and loss of adult articular cartilage. ERAD complex malfunction also causes abnormal accumulation of cartilage ECM molecules and subsequent chondrodysplasia. ERAD gene expression is decreased in damaged cartilage from patients with osteoarthritis (OA), and disruption of ERAD function in articular cartilage leads to cartilage destruction in a mouse OA model.
Collapse
Affiliation(s)
- Hyo Jung Sim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Chanmi Cho
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
- Department of Biomedical Sciences, Graduate School, Ajou University, Suwon 16499, Korea
- CIRNO, Sungkyunkwan University, Suwon 16419, Korea
- Degenerative Inter Diseases Research Center, Ajou University School of Medicine, Suwon 16499, Korea
| | - Ha Eun Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Ju Yeon Hong
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Eun Kyung Song
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Keun Yeong Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Dong Gil Jang
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Seok-Jung Kim
- Department of Orthopaedic Surgery, Uijeongbu St. Mary’s Hospital, Catholic University of Korea College of Medicine, Uijeongbu 11765, Korea
| | - Hyun-Shik Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Changwook Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Taejoon Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Korea
| | - Siyoung Yang
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
- Department of Biomedical Sciences, Graduate School, Ajou University, Suwon 16499, Korea
- CIRNO, Sungkyunkwan University, Suwon 16419, Korea
- Degenerative Inter Diseases Research Center, Ajou University School of Medicine, Suwon 16499, Korea
| | - Tae Joo Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Korea
| |
Collapse
|
114
|
Nonsuwan P, Nishijima N, Sakakibara K, Nakaji-Hirabayashi T, Yoshikawa C. Concentrated polymer brush-modified cellulose nanofibers promote chondrogenic differentiation of human mesenchymal stem cells by controlling self-assembly. J Mater Chem B 2022; 10:2444-2453. [PMID: 35045146 DOI: 10.1039/d1tb02307a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In order to develop new three-dimensional (3D) cell culture systems for articular cartilage regeneration, concentrated poly(styrene sulfonate sodium salt) brush-modified cellulose nanofibers were employed as building blocks for the self-assembly of human mesenchymal stem cells (hMSCs). Unique 3D cellular structures, such as giant spheres and sheets, were formed by controlling hMSC self-assembly.
Collapse
Affiliation(s)
- Punnida Nonsuwan
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki 305-0047, Japan.
| | - Nanami Nishijima
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki 305-0047, Japan. .,Faculty of Engineering, Academic Assembly, University of Toyama, 3190 Gofuku, Toyama, Toyama 930-8555, Japan
| | - Keita Sakakibara
- Research Institute for Sustainable Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), 3-11-32 Kagamiyama, Higashi-hiroshima, Hiroshima 739-0046, Japan
| | - Tadashi Nakaji-Hirabayashi
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki 305-0047, Japan. .,Faculty of Engineering, Academic Assembly, University of Toyama, 3190 Gofuku, Toyama, Toyama 930-8555, Japan.,Graduate School of Innovative Life Science, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Chiaki Yoshikawa
- Research Center for Functional Materials, National Institute for Materials Science (NIMS), Tsukuba, Ibaraki 305-0047, Japan.
| |
Collapse
|
115
|
Alexandrou E, Dauber A, Tyzinski L, Hwa V, Andrew M, Kim H, Elangovan S, Gubanich P, Taylor-Haas JA, Paterno M, Backeljauw P. Clinical phenotype and musculoskeletal characteristics of patients with aggrecan deficiency. Am J Med Genet A 2022; 188:1193-1203. [PMID: 35001504 DOI: 10.1002/ajmg.a.62639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/21/2021] [Accepted: 11/28/2021] [Indexed: 11/10/2022]
Abstract
Aggrecan is a proteoglycan within the physeal and articular cartilage. Aggrecan deficiency, due to heterozygous mutations in the ACAN gene, causes dominantly inherited short stature and, in many patients, early-onset osteoarthritis and degenerative disc disease. We aimed to further characterize this phenotypic spectrum with an emphasis on musculoskeletal health. Twenty-two individuals from nine families were enrolled. Histories and examinations focused on joint health, gait analysis, joint specific patient reported outcomes, and imaging studies were performed. All patients had dominantly inherited short stature, with the exception of a de novo mutation. Short stature was worse in adults versus children (median height -3.05 SD vs. -2.25 SD). ACAN mutations were not always associated with bone age advancement (median advancement +1.1 years, range 0 to +2 years). Children had subtle disproportionality and clinically silent joint disease-25% with osteochondritis dissecans (OD). Adults had a high prevalence of joint symptomatology-decline in knee function, disability from spinal complaints, and lower physical activity on outcome measures. Osteoarthritis (OA) and OD was detected in 90% of adults, and orthopedic surgeries were reported in 60%. Aggrecan deficiency leads to short stature with progressive decline in height SD, mild skeletal dysplasia, and increasing prevalence of joint pathology over time. Optimal musculoskeletal health and quality of life can be attained with timely identification of pathology and intervention.
Collapse
Affiliation(s)
- Eirene Alexandrou
- Division of Endocrinology, University of Iowa Stead Family Children's Hospital, Iowa City, Iowa, USA.,Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Andrew Dauber
- Division of Endocrinology, Children's National Hospital, Washington, District of Columbia, USA.,Department of Pediatrics, George Washington School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Leah Tyzinski
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Vivian Hwa
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Melissa Andrew
- Division of Endocrinology, Children's National Hospital, Washington, District of Columbia, USA
| | - Hee Kim
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stacey Elangovan
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Paul Gubanich
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - Mark Paterno
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Philippe Backeljauw
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
116
|
Bay-Jensen AC, Mobasheri A, Thudium CS, Kraus VB, Karsdal MA. Blood and urine biomarkers in osteoarthritis - an update on cartilage associated type II collagen and aggrecan markers. Curr Opin Rheumatol 2022; 34:54-60. [PMID: 34652292 PMCID: PMC8635261 DOI: 10.1097/bor.0000000000000845] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW Osteoarthritis (OA) is a painful disease for which drug development has proven difficult. One major reason for this is the heterogeneity of the disease and the current lack of operationalized means to distinguish various disease endotypes (molecular subtypes). Biomarkers measured in blood or urine, reflecting joint tissue turnover, have been developed and tested during the last decades. In this narrative review, we provide highlights on biomarkers derived from the two most studied and abundant cartilage proteins - type II collagen and aggrecan. RECENT FINDINGS Multiple biomarkers assessing type II collagen degradation and formation, and aggrecan turnover have been developed. Several markers, such as uCTX-II, have been validated for their association with disease severity and prognosis, as well as pharmacodynamically used to describe the mode of action and efficacy of drugs in development. There is a great need for biomarkers for subdividing patients (i.e., endotyping) and recent scientific advances have not yet come closer to achieving this goal. SUMMARY There is strong support for using biomarkers for understanding OA, reflecting degradation and formation of the joint tissues, focused on type II collagen and aggrecan. There is still a lack of in vitro diagnostics, in all contexts of use.
Collapse
Affiliation(s)
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- University Medical Center Utrecht, Department of Orthopedics, Rheumatology and Clinical Immunology, Utrecht, The Netherlands
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, University of Liege, Liege, Belgium
| | | | - Virginia B. Kraus
- Duke Molecular Physiology Institute and Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | |
Collapse
|
117
|
Hellicar J, Stevenson NL, Stephens DJ, Lowe M. Supply chain logistics - the role of the Golgi complex in extracellular matrix production and maintenance. J Cell Sci 2022; 135:273996. [PMID: 35023559 PMCID: PMC8767278 DOI: 10.1242/jcs.258879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The biomechanical and biochemical properties of connective tissues are determined by the composition and quality of their extracellular matrix. This, in turn, is highly dependent on the function and organisation of the secretory pathway. The Golgi complex plays a vital role in directing matrix output by co-ordinating the post-translational modification and proteolytic processing of matrix components prior to their secretion. These modifications have broad impacts on the secretion and subsequent assembly of matrix components, as well as their function in the extracellular environment. In this Review, we highlight the role of the Golgi in the formation of an adaptable, healthy matrix, with a focus on proteoglycan and procollagen secretion as example cargoes. We then discuss the impact of Golgi dysfunction on connective tissue in the context of human disease and ageing.
Collapse
Affiliation(s)
- John Hellicar
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.,Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673
| | - Nicola L Stevenson
- Cell Biology Laboratories, School of Biochemistry, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - David J Stephens
- Cell Biology Laboratories, School of Biochemistry, Faculty of Life Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - Martin Lowe
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, The Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|
118
|
Ali N, Turkiewicz A, Hughes V, Folkesson E, Tjörnstand J, Neuman P, Önnerfjord P, Englund M. Proteomics profiling of human synovial fluid suggests increased protein interplay in early-osteoarthritis (OA) that is lost in late-stage OA. Mol Cell Proteomics 2022; 21:100200. [PMID: 35074580 PMCID: PMC8941261 DOI: 10.1016/j.mcpro.2022.100200] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 11/04/2021] [Accepted: 01/15/2022] [Indexed: 11/28/2022] Open
Abstract
The underlying molecular mechanisms in osteoarthritis (OA) development are largely unknown. This study explores the proteome and the pairwise interplay of proteins in synovial fluid from patients with late-stage knee OA (arthroplasty), early knee OA (arthroscopy due to degenerative meniscal tear), and from deceased controls without knee OA. Synovial fluid samples were analyzed using state-of-the-art mass spectrometry with data-independent acquisition. The differential expression of the proteins detected was clustered and evaluated with data mining strategies and a multilevel model. Group-specific slopes of associations were estimated between expressions of each pair of identified proteins to assess the co-expression (i.e., interplay) between the proteins in each group. More proteins were increased in early-OA versus controls than late-stage OA versus controls. For most of these proteins, the fold changes between late-stage OA versus controls and early-stage OA versus controls were remarkably similar suggesting potential involvement in the OA process. Further, for the first time, this study illustrated distinct patterns in protein co-expression suggesting that the interplay between the protein machinery is increased in early-OA and lost in late-stage OA. Further efforts should focus on earlier stages of the disease than previously considered. Synovial fluid proteomics study of different stages of osteoarthritis (OA). Higher catabolic activity is found in both early- and late-stage OA. Imbalance of the metabolic homeostasis in late-stage OA. Understanding early-stage OA may lead to finding better effective therapies.
Collapse
|
119
|
Rajesh N, Moudgil-Joshi J, Kaliaperumal C. Smoking and degenerative spinal disease: A systematic review. BRAIN AND SPINE 2022; 2:100916. [PMID: 36248118 PMCID: PMC9560562 DOI: 10.1016/j.bas.2022.100916] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/28/2022] [Accepted: 07/11/2022] [Indexed: 11/29/2022]
Abstract
Smoking is a major cause of morbidity and mortality worldwide and is responsible for the death of more than 8 million people per year globally. Through a systematic literature review, we aim to review the harmful effects of tobacco smoking on degenerative spinal diseases (DSD). DSD is a debilitating disease and there is a need to identify if smoking can be an attributable contender for the occurrence of this disease, as it can open up avenues for therapeutic options. Sources such as PubMed and Embase were used to review literature, maintaining tobacco smoking and spinal diseases as inclusion factors, excluding any article that did not explore this relationship. Risk of bias was assessed using analysis of results, sample size and methods and limitations. Upon review of the literature, tobacco smoking was found to be a major risk factor for the occurrence of DSDs, particularly lumbar spinal diseases. Smokers also experienced a greater need for surgery and greater postoperative wound healing complications, increased pain perception, delay in recovery and decreased satisfaction after receiving surgery. These effects were noted along the entire spine. Many mechanisms of action have been identified in the literature that provide plausible pictures of how smoking leads to spinal degeneration, exploring possible primary targets which can open up opportunities to develop potential therapeutic agents. More studies on cervical and thoracic spinal degeneration would be beneficial in identifying the effect of nicotine on these spinal levels. Some limitations included insufficient sample size, inconclusive evidence and lack of sufficient repeat studies. However, there appears to be a sufficient amount of research on smoking directly contributing to lumbar spinal pathology. Smoking is a risk factor for the occurence of degenerative spinal disease (DSD). There are numerous pathological mechanisms attributed to spinal pathology by smoking. Smoking appears to be a significant risk factor for lumbar DSDs, with smoke studies also suggesting its role in cervical DSDs. There is insufficient research on the effect of smoking on the thoracic spine. Smoking leads to worse outcomes and potential complications post-surgery, as well as increased pain perception and poorer subjective response post-surgery.
Collapse
|
120
|
Mechanical Cues: Bidirectional Reciprocity in the Extracellular Matrix Drives Mechano-Signalling in Articular Cartilage. Int J Mol Sci 2021; 22:ijms222413595. [PMID: 34948394 PMCID: PMC8707858 DOI: 10.3390/ijms222413595] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 12/29/2022] Open
Abstract
The composition and organisation of the extracellular matrix (ECM), particularly the pericellular matrix (PCM), in articular cartilage is critical to its biomechanical functionality; the presence of proteoglycans such as aggrecan, entrapped within a type II collagen fibrillar network, confers mechanical resilience underweight-bearing. Furthermore, components of the PCM including type VI collagen, perlecan, small leucine-rich proteoglycans—decorin and biglycan—and fibronectin facilitate the transduction of both biomechanical and biochemical signals to the residing chondrocytes, thereby regulating the process of mechanotransduction in cartilage. In this review, we summarise the literature reporting on the bidirectional reciprocity of the ECM in chondrocyte mechano-signalling and articular cartilage homeostasis. Specifically, we discuss studies that have characterised the response of articular cartilage to mechanical perturbations in the local tissue environment and how the magnitude or type of loading applied elicits cellular behaviours to effect change. In vivo, including transgenic approaches, and in vitro studies have illustrated how physiological loading maintains a homeostatic balance of anabolic and catabolic activities, involving the direct engagement of many PCM molecules in orchestrating this slow but consistent turnover of the cartilage matrix. Furthermore, we document studies characterising how abnormal, non-physiological loading including excessive loading or joint trauma negatively impacts matrix molecule biosynthesis and/or organisation, affecting PCM mechanical properties and reducing the tissue’s ability to withstand load. We present compelling evidence showing that reciprocal engagement of the cells with this altered ECM environment can thus impact tissue homeostasis and, if sustained, can result in cartilage degradation and onset of osteoarthritis pathology. Enhanced dysregulation of PCM/ECM turnover is partially driven by mechanically mediated proteolytic degradation of cartilage ECM components. This generates bioactive breakdown fragments such as fibronectin, biglycan and lumican fragments, which can subsequently activate or inhibit additional signalling pathways including those involved in inflammation. Finally, we discuss how bidirectionality within the ECM is critically important in enabling the chondrocytes to synthesise and release PCM/ECM molecules, growth factors, pro-inflammatory cytokines and proteolytic enzymes, under a specified load, to influence PCM/ECM composition and mechanical properties in cartilage health and disease.
Collapse
|
121
|
Denis A, Chergui S, Basalom S, Campeau PM, Janelle C, Pauyo T. Variable expressivity in a family with an aggrecanopathy. Mol Genet Genomic Med 2021; 10:e1773. [PMID: 34894100 PMCID: PMC8801139 DOI: 10.1002/mgg3.1773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/13/2021] [Accepted: 07/08/2021] [Indexed: 12/22/2022] Open
Abstract
Background Osteochondritis dissecans is a condition wherein there is a subchondral bone lesion that causes pain, inflammation, and cartilage damage. Dominant Familial Osteochondritis Dissecans is a rare and severe form of osteochondritis dissecans (OCD). It is caused by heterozygous pathogenic variants in the gene encoding Aggrecan; ACAN. Aggrecan, a proteoglycan, is an essential component of the articular and growth plate cartilage. Methods Herein, we report three individuals from one family; the proband who presented with short stature, a lower limb bone exostosis, and bilateral knee and elbow OCD at the age of 13 years old. His twin brother presented with isolated short stature and his father with short stature and lumbar disc herniation. Results Next‐generation sequencing of the ACAN gene in the proband identified a frameshift variant which is also present in the brother and father with short stature. The proband was treated surgically with bilateral elbow microfracture, after the failure of conservative therapy. Conclusion To the best of our knowledge, this is the first patient with an aggrecanopathy who presents with osteochondritis dissecans due to a frameshift variant. This family presents with variable expressivity which might be attributed to modifier genes.
Collapse
Affiliation(s)
- Antoine Denis
- Shriners Hospitals for Children, Montreal, Quebec, Canada
| | - Sami Chergui
- Shriners Hospitals for Children, Montreal, Quebec, Canada
| | - Shuaa Basalom
- Shriners Hospitals for Children, Montreal, Quebec, Canada
| | | | | | - Thierry Pauyo
- Shriners Hospitals for Children, Montreal, Quebec, Canada
| |
Collapse
|
122
|
Tran DT, Juang YC, Tsai L. Contrary response of porcine articular cartilage below and over 1000 s -1. Clin Biomech (Bristol, Avon) 2021; 90:105506. [PMID: 34610506 DOI: 10.1016/j.clinbiomech.2021.105506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/31/2021] [Accepted: 09/27/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Knee joints experience excessive loads quite frequently during sports activities, and these shocks could accelerate progressive degeneration in articular cartilage. METHODS Quasi-static and dynamic response of porcine knee articular cartilages were investigated in this research. Split Hopkinson Pressure Bars (SHPB) were utilized to examine the articular cartilage properties at strain rates between 0.01-2000 s-1. FINDINGS The results showed that strain rate is an important factor for articular cartilages, distinctively divided into above and below 1000 s-1. The articular cartilages exhibit a strain hardening phenomenon when shock loaded at strain rates under 1000 s-1. When loaded at strain rates over 1000 s-1, their ultimate strength and elastic modulus decreased with increasing strain rates. INTERPRETATION The biphasic structure of the cartilage explained the change of modulus. At the lower strain rates, fibers realigned and solidified the structure, while at higher strain rates, there is not enough time for the tissue fluid to move inside the cartilage, leading to a reduction in the deformability of the specimen and raising of Young's modulus. The results can be utilized to provide some useful data for biomaterial and computational works in the future.
Collapse
Affiliation(s)
- D T Tran
- National Kaohsiung University of Science and Technology, Department of Mechanical Engineering, No. 415, Jiangong rd., Kaohsiung, Taiwan
| | - Y C Juang
- National Kaohsiung University of Science and Technology, Department of Mechanical Engineering, No. 415, Jiangong rd., Kaohsiung, Taiwan
| | - L Tsai
- National Kaohsiung University of Science and Technology, Department of Mechanical Engineering, No. 415, Jiangong rd., Kaohsiung, Taiwan.
| |
Collapse
|
123
|
Gatenholm B, Lindahl C, Brittberg M, Simonsson S. Collagen 2A Type B Induction after 3D Bioprinting Chondrocytes In Situ into Osteoarthritic Chondral Tibial Lesion. Cartilage 2021; 13:1755S-1769S. [PMID: 32070108 PMCID: PMC8721610 DOI: 10.1177/1947603520903788] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE Large cartilage defects and osteoarthritis (OA) cause cartilage loss and remain a therapeutic challenge. Three-dimensional (3D) bioprinting with autologous cells using a computer-aided design (CAD) model generated from 3D imaging has the potential to reconstruct patient-specific features that match an articular joint lesion. DESIGN To scan a human OA tibial plateau with a cartilage defect, retrieved after total knee arthroplasty, following clinical imaging techniques were used: (1) computed tomography (CT), (2) magnetic resonance imaging (MRI), and (3) a 3D scanner. From such a scan, a CAD file was obtained to generate G-code to control 3D bioprinting in situ directly into the tibial plateau lesion. RESULTS Highest resolution was obtained using the 3D scanner (2.77 times more points/mm2 than CT), and of the 3 devices tested, only the 3D scanner was able to detect the actual OA defect area. Human chondrocytes included in 3D bioprinted constructs produced extracellular matrix and formed cartilage tissue fragments after 2 weeks of differentiation and high levels of a mature splice version of collagen type II (Col IIA type B), characteristic of native articular cartilage and aggrecan (ACAN). Chondrocytes had a mean viability of 81% in prints after day 5 of differentiation toward cartilage and similar viability was detected in control 3D pellet differentiation of chondrocytes (mean viability 72%). CONCLUSION Articular cartilage can be formed in 3D bioprints. Thus, this 3D bioprinting system with chondrocytes simulating a patient-specific 3D model provides an attractive strategy for future treatments of cartilage defects or early OA.
Collapse
Affiliation(s)
- Birgitta Gatenholm
- Department of Orthopaedics, Institute of
Clinical Sciences, the Sahlgrenska Academy at the University of Gothenburg,
Gothenburg, Sweden,Sahlgrenska University Hospital,
Mölndal, Sweden
| | - Carl Lindahl
- Institute of Biomedicine, Department of
Clinical Chemistry and Transfusion Medicine, University of Gothenburg, Gothenburg,
Sweden
| | - Mats Brittberg
- Cartilage Repair Unit, University of
Gothenburg, Region Halland Orthopaedics, Kungsbacka Hospital, Kungsbacka,
Sweden
| | - Stina Simonsson
- Institute of Biomedicine, Department of
Clinical Chemistry and Transfusion Medicine, University of Gothenburg, Gothenburg,
Sweden,Stina Simonsson, Department of Clinical
Chemistry and Transfusion Medicine, Institute of Biomedicine, University of
Gothenburg, Sahlgrenska University Hospital, Bruna Straket 16, Gothenburg, 413
45, Sweden.
| |
Collapse
|
124
|
Aghlara-Fotovat S, Nash A, Kim B, Krencik R, Veiseh O. Targeting the extracellular matrix for immunomodulation: applications in drug delivery and cell therapies. Drug Deliv Transl Res 2021; 11:2394-2413. [PMID: 34176099 DOI: 10.1007/s13346-021-01018-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 12/12/2022]
Abstract
Host immune cells interact bi-directionally with their extracellular matrix (ECM) to receive and deposit molecular signals, which orchestrate cellular activation, proliferation, differentiation, and function to maintain healthy tissue homeostasis. In response to pathogens or damage, immune cells infiltrate diseased sites and synthesize critical ECM molecules such as glycoproteins, proteoglycans, and glycosaminoglycans to promote healing. When the immune system misidentifies pathogens or fails to survey damaged cells effectively, maladies such as chronic inflammation, autoimmune diseases, and cancer can develop. In these conditions, it is essential to restore balance to the body through modulation of the immune system and the ECM. This review details the components of dysregulated ECM implicated in pathogenic environments and therapeutic approaches to restore tissue homeostasis. We evaluate emerging strategies to overcome inflamed, immune inhibitory, and otherwise diseased microenvironments, including mechanical stimulation, targeted proteases, adoptive cell therapy, mechanomedicine, and biomaterial-based cell therapeutics. We highlight various strategies that have produced efficacious responses in both pre-clinical and human trials and identify additional opportunities to develop next-generation interventions. Significantly, we identify a need for therapies to address dense or fibrotic tissue for the treatment of organ tissue damage and various cancer subtypes. Finally, we conclude that therapeutic techniques that disrupt, evade, or specifically target the pathogenic microenvironment have a high potential for improving therapeutic outcomes and should be considered a priority for immediate exploration. A schematic showing the various methods of extracellular matrix disruption/targeting in both fibrotic and cancerous environments. a Biomaterial-based cell therapy can be used to deliver anti-inflammatory cytokines, chemotherapeutics, or other factors for localized, slow release of therapeutics. b Mechanotherapeutics can be used to inhibit the deposition of molecules such as collagen that affect stiffness. c Ablation of the ECM and target tissue can be accomplished via mechanical degradation such as focused ultrasound. d Proteases can be used to improve the distribution of therapies such as oncolytic virus. e Localization of therapeutics such as checkpoint inhibitors can be improved with the targeting of specific ECM components, reducing off-target effects and toxicity.
Collapse
Affiliation(s)
| | - Amanda Nash
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Boram Kim
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Robert Krencik
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| |
Collapse
|
125
|
Tamiasso NV, Silva CMO, Reis AMS, Ocarino NM, Serakides R. Ethanol Alters Phenotype and Synthesis Activity of Rat Neonatal Articular Chondrocytes Grown in 2- and 3-Dimensional Culture. Cartilage 2021; 13:839S-846S. [PMID: 31441318 PMCID: PMC8804855 DOI: 10.1177/1947603519870862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE We sought to evaluate the effect of different concentrations of ethanol on phenotype and activity of articular chondrocyte synthesis of neonatal rats in 2-dimensional (2D) and 3-dimensional (3D) culture. METHODS Chondrocytes were cultured in chondrogenic medium with different concentrations of ethanol: 0.0% v/v (control); 0.05% v/v (8.6 mM); 0.25% v/v (42.9 mM), and 0.5% v/v (85.7 mM). Chondrocytes under 2D culture were subjected to MTT assay, while chondrocytes under 3D culture were processed for paraffin inclusion and stained by periodic acid Schiff (PAS) to evaluate mean chondrocyte diameter and percentages of cells, nucleus, cytoplasm, well-differentiated matrix, and PAS+ areas. The expression of gene transcripts for aggrecan, Sox9, and type II collagen was evaluated by real-time quantitative polymerase chain reaction. RESULTS There was no difference between groups by the MTT assay. PAS staining revealed that chondrocytes treated with 0.5% v/v ethanol had higher percentages of cytoplasm and nuclear areas, but with a reduction in PAS+ matrix area. The mean diameter of chondrocytes was similar between groups. The expression of aggrecan in the group treated with 0.5% v/v ethanol was lower in comparison to that in the control. In the groups treated with 0.25% v/v and 0.5% v/v ethanol, the percentage of differentiated cartilage was lower in comparison with that in the control. The group treated with 0.05% v/v ethanol was similar to the control in all parameters. CONCLUSIONS Ethanol acted directly on in vitro cultured articular chondrocytes of newborn rats, altering the chondrocyte phenotype and its synthesis activity, and these effects were dose dependent.
Collapse
Affiliation(s)
- Natalia Viana Tamiasso
- Núcleo de Células Tronco e Terapia
Celular Animal (NCT-TCA) da Escola de Veterinária da Universidade Federal de Minas
Gerais Belo Horizonte, Minas Gerais, Brazil
| | - Carla Maria Osório Silva
- Núcleo de Células Tronco e Terapia
Celular Animal (NCT-TCA) da Escola de Veterinária da Universidade Federal de Minas
Gerais Belo Horizonte, Minas Gerais, Brazil
| | | | - Natália Melo Ocarino
- Núcleo de Células Tronco e Terapia
Celular Animal (NCT-TCA) da Escola de Veterinária da Universidade Federal de Minas
Gerais Belo Horizonte, Minas Gerais, Brazil
| | - Rogéria Serakides
- Núcleo de Células Tronco e Terapia
Celular Animal (NCT-TCA) da Escola de Veterinária da Universidade Federal de Minas
Gerais Belo Horizonte, Minas Gerais, Brazil,Rogéria Serakides, Núcleo de Células Tronco
e Terapia Celular Animal (NCT-TCA) da Escola de Veterinária da Universidade
Federal de Minas Gerais Belo Horizonte, Av. Antônio Carlos 6627, Caixa Postal
567, campus Pampulha da UFMG, Belo Horizonte, MG CEP 30123-970, Brazil.
| |
Collapse
|
126
|
Lin JZ, Duan MR, Lin N, Zhao WJ. The emerging role of the chondroitin sulfate proteoglycan family in neurodegenerative diseases. Rev Neurosci 2021; 32:737-750. [PMID: 33655733 DOI: 10.1515/revneuro-2020-0146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/07/2021] [Indexed: 02/05/2023]
Abstract
Chondroitin sulfate (CS) is a kind of linear polysaccharide that is covalently linked to proteins to form proteoglycans. Chondroitin sulfate proteoglycans (CSPGs) consist of a core protein, with one or more CS chains covalently attached. CSPGs are precisely regulated and they exert a variety of physiological functions by binding to adhesion molecules and growth factors. Widely distributed in the nervous system in human body, CSPGs contribute to the major component of extracellular matrix (ECM), where they play an important role in the development and maturation of the nervous system, as well as in the pathophysiological response to damage to the central nervous system (CNS). While there are more than 30 types of CSPGs, this review covers the roles of the most important ones, including versican, aggrecan, neurocan and NG2 in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. The updated reports of the treatment of neurodegenerative diseases are involving CSPGs.
Collapse
Affiliation(s)
- Jia-Zhe Lin
- Neurosurgical Department, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
- Center for Neuroscience, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Ming-Rui Duan
- Center for Neuroscience, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Nuan Lin
- Obstetrics and Gynecology Department, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Wei-Jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou 515041, Guangdong, China
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
127
|
Disease-specific glycosaminoglycan patterns in the extracellular matrix of human lung and brain. Carbohydr Res 2021; 511:108480. [PMID: 34837849 DOI: 10.1016/j.carres.2021.108480] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 11/24/2022]
Abstract
A wide variety of diseases throughout the mammalian organism is characterized by abnormal deposition of various components of the extracellular matrix (ECM), including the heterogeneous family of glycosaminoglycans (GAGs), which contribute considerably to the ECM architecture as part of the so-called proteoglycans. The GAG's unique sulfation pattern, derived from highly dynamic and specific modification processes, has a massive impact on critical mediators such as cytokines and growth factors. Due to the strong connection between the specific sulfation pattern and GAG function, slight alterations of this pattern are often associated with enormous changes at the cell as well as at the organ level. This review aims to investigate the connection between modifications of GAG sulfation patterns and the wide range of pathological conditions, mainly focusing on a range of chronic diseases of the central nervous system (CNS) as well as the respiratory tract.
Collapse
|
128
|
Feng K, Xie X, Yuan J, Gong L, Zhu Z, Zhang J, Li H, Yang Y, Wang Y. Reversing the surface charge of MSC-derived small extracellular vesicles by εPL-PEG-DSPE for enhanced osteoarthritis treatment. J Extracell Vesicles 2021; 10:e12160. [PMID: 34724347 PMCID: PMC8559985 DOI: 10.1002/jev2.12160] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/27/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) possess a great therapeutical potential for osteoarthritis (OA) treatment. However, the steric and electrostatic hindrance of cartilage matrix leads to very limited distribution of MSC-sEVs in cartilage and low bioavailability of MSC-sEVs after intra-articular injection. To overcome this, a strategy to reverse the surface charge of MSC-sEVs by modifying the MSC-sEVs with a novel cationic amphiphilic macromolecule namely ε-polylysine-polyethylene-distearyl phosphatidylethanolamine (PPD) was developed in this study. Through incubation with 100 μg/ml PPD, positively charged MSC-sEVs (PPD-sEVs) were obtained, and the modification process showed nearly no disturbance to the integrity and contents of sEVs and exhibited good stability under the interference of anionic macromolecules. A more effective cellular uptake and homeostasis modulation ability of PPD-sEVs than unmodified MSC-sEVs to chondrocytes was demonstrated. More importantly, PPD-sEVs demonstrated significantly enhanced cartilage uptake, cartilage penetration, and joint retention capacity as compared to MSC-sEVs. Intra-articular injection of PPD-sEVs into a mouse OA model showed significantly improved bioavailability than MSC-sEVs, which resulted in enhanced therapeutic efficacy with reduced injection frequency. In general, this study provides a facile and effective strategy to improve the intra-articular bioavailability of MSC-sEVs and has a great potential to accelerate the clinical practice of MSC-sEVs based OA therapy.
Collapse
Affiliation(s)
- Kai Feng
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Xuetao Xie
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Ji Yuan
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Liangzhi Gong
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Zhaochen Zhu
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Juntao Zhang
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Haiyan Li
- Chemical and Environmental EngineeringSchool of EngineeringRMIT UniversityMelbourneAustralia
| | - Yunlong Yang
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Yang Wang
- Institute of Microsurgery on ExtremitiesDepartment of Orthopedic SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| |
Collapse
|
129
|
Thomas J, Chopra V, Sharma A, Panwar V, Kaushik S, Rajput S, Mittal M, Guha R, Chattopadhyay N, Ghosh D. An injectable hydrogel having proteoglycan-like hierarchical structure supports chondrocytes delivery and chondrogenesis. Int J Biol Macromol 2021; 190:474-486. [PMID: 34508717 DOI: 10.1016/j.ijbiomac.2021.08.226] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/18/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
The ECM of cartilage is composed of proteoglycans (PG) that contain glycosaminoglycan (GAG), aggrecan, hyaluronic acid (HA) and other molecular components which play an important role in regulating chondrocyte functions via cell-matrix interactions, integrin-mediated signalling etc. Implantation of chondrocytes encapsulated in scaffolds that mimic the micro-architecture of proteoglycan, is expected to enhance cartilage repair. With an aim to create a hydrogel having macromolecular structure that resembles the cartilage-specific ECM, we constructed a hierarchal structure that mimic the PG. The bottle brush structure of the aggrecan was obtained using chondroitin sulphate and carboxymethyl cellulose which served as GAG and core protein mimic respectively. A proteoglycan-like structure was obtained by cross-linking it with modified chitosan that served as a HA substitute. The physico-chemical characteristics of the above cross-linked injectable hydrogel supported long term human articular chondrocyte subsistence and excellent post-injection viability. The chondrocytes encapsulated in the PMH expressed significant levels of articular cartilage specific markers like collagen II, aggrecan, GAGs etc., indicating the ability of the hydrogel to support chondrocyte differentiation. The biocompatibility and biodegradability of the hydrogels was confirmed using suitable in vivo studies. The results revealed that the PG-mimetic hydrogel could serve as a promising scaffold for chondrocyte implantation.
Collapse
Affiliation(s)
- Jijo Thomas
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Vianni Chopra
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Anjana Sharma
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Vineeta Panwar
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Swati Kaushik
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Swati Rajput
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, U.P., India
| | - Monika Mittal
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, U.P., India
| | - Rajdeep Guha
- Laboratory Animal Facility, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, U.P., India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, U.P., India
| | - Deepa Ghosh
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India.
| |
Collapse
|
130
|
Nikhil A, Kumar A. Evaluating potential of tissue-engineered cryogels and chondrocyte derived exosomes in articular cartilage repair. Biotechnol Bioeng 2021; 119:605-625. [PMID: 34723385 DOI: 10.1002/bit.27982] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/18/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022]
Abstract
Treatment of articular cartilage injuries especially osteochondral tissue requires intervention of bioengineered scaffold. In this study, we investigated the potential of the tissue-engineered cryogel scaffold fabricated using cryogelation technology. Two types of cryogels viz. chitosan-gelatin-chondroitin sulfate (CGC) for articular cartilage and nano-hydroxyapatite-gelatin (HG) for subchondral bone were fabricated. Further, novel bilayer cryogel designed using single process fabrication of two layers (CGC as top layer and HG as the lower layer) was designed to mimic osteochondral unit. CGC cryogel was tested for their biocompatibility using the enzymatically isolated chondrcoytes from goat articular cartilage while HG cryogel was tested using pre-osteoblast cell line. Extracellular vesicles, specifically exosomes were isolated from the spent media of chondrocytes to validate their effect over cell proliferation and migration which are required for defect healing and infiltration respectively. These isolated exosomes were characterized and analyzed for confirming their size distribution profile and visualized morphologically using advanced microscopy techniques. For cartilage part, CGC cryogels were examined as delivery system for delivering exosomes at defect site, where 80% of release was observed in 72 h. Release of 18.7 µg chondroitin sulfate/mg cryogel was obtained in a period of one week from CGC cryogel (termed cryogel extract) which has chondroprotective effect. Further, effect of exosome concentration (10 and 20 µg/ml), CGC extract and combination of exosome and CGC extract (Exo-Ex) were assessed over the chondrocytes. In addition, in vitro scratch wound assay was performed to analyse the migration capacity over the micro-injury when treated with exosomes, cryogel extract and Exo-Ex. The overall results thus answer key questions of therapeutic potential of chondrocyte exosomes, cryogel extract in addition to potential of CGC and HG cryogel for osteochondral repair.
Collapse
Affiliation(s)
- Aman Nikhil
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India.,Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India.,Centre for Nanosciences, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India.,The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| |
Collapse
|
131
|
Lee MH, Kim HM, Chung HC, Kim DU, Lee JH. Low-Molecular-Weight Collagen Peptide Ameliorates Osteoarthritis Progression through Promoting Extracellular Matrix Synthesis by Chondrocytes in a Rabbit Anterior Cruciate Ligament Transection Model. J Microbiol Biotechnol 2021; 31:1401-1408. [PMID: 34528913 PMCID: PMC9705828 DOI: 10.4014/jmb.2108.08027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/15/2022]
Abstract
This study examined whether the oral administration of low-molecular-weight collagen peptide (LMCP) containing 3% Gly-Pro-Hyp with >15% tripeptide (Gly-X-Y) content could ameliorate osteoarthritis (OA) progression using a rabbit anterior cruciate ligament transection (ACLT) model of induced OA and chondrocytes isolated from a patient with OA. Oral LMCP administration (100 or 200 mg/kg/day) for 12 weeks ameliorated cartilage damage and reduced the loss of proteoglycan compared to the findings in the ACLT control group, resulting in dose-dependent (p < 0.05) improvements of the OARSI score in hematoxylin & eosin (H&E) and Safranin O staining. In microcomputed tomography analysis, LMCP also significantly (p < 0.05) suppressed the deterioration of the microstructure in tibial subchondral bone during OA progression. The elevation of IL-1βand IL-6 concentrations in synovial fluid following OA induction was dose-dependently (p < 0.05) reduced by LMCP treatment. Furthermore, immunohistochemistry illustrated that LMCP significantly (p < 0.05) upregulated type II collagen and downregulated matrix metalloproteinase-13 in cartilage tissue. Consistent with the in vivo results, LMCP significantly (p < 0.05) increased the mRNA expression of COL2A1 and ACAN in chondrocytes isolated from a patient with OA regardless of the conditions for IL-1βinduction. These findings suggest that LMCP has potential as a therapeutic treatment for OA that stimulates cartilage regeneration.
Collapse
Affiliation(s)
- Mun-Hoe Lee
- Health Food Research and Development, NEWTREE Co., Ltd., Seoul 05604, Republic of Korea
| | - Hyeong-Min Kim
- Health Food Research and Development, NEWTREE Co., Ltd., Seoul 05604, Republic of Korea
| | - Hee-Chul Chung
- Health Food Research and Development, NEWTREE Co., Ltd., Seoul 05604, Republic of Korea
| | - Do-Un Kim
- Health Food Research and Development, NEWTREE Co., Ltd., Seoul 05604, Republic of Korea
| | - Jin-Hee Lee
- Health Food Research and Development, NEWTREE Co., Ltd., Seoul 05604, Republic of Korea,Corresponding author Phone: +82-70-8015-8518 Fax: +82-2-6949-1293 E-mail:
| |
Collapse
|
132
|
Gögele C, Wiltzsch S, Lenhart A, Civilleri A, Weiger TM, Schäfer-Eckart K, Minnich B, Forchheimer L, Hornfeck M, Schulze-Tanzil G. Highly porous novel chondro-instructive bioactive glass scaffolds tailored for cartilage tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 130:112421. [PMID: 34702508 DOI: 10.1016/j.msec.2021.112421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/23/2021] [Accepted: 09/06/2021] [Indexed: 12/11/2022]
Abstract
Cartilage injuries remain challenging since the regenerative capacity of cartilage is extremely low. The aim was to design a novel type of bioactive glass (BG) scaffold with suitable topology that allows the formation of cartilage-specific extracellular matrix (ECM) after colonization with chondrogenic cells for cartilage repair. Highly porous scaffolds with interconnecting pores consisting of 100 % BG were manufactured using a melting, milling, sintering and leaching technique. Scaffolds were colonized with porcine articular chondrocytes (pAC) and undifferentiated human mesenchymal stromal cells (hMSC) for up to 35 days. Scaffolds displayed high cytocompatibility with no major pH shift. Scanning electron microscopy revealed the intimate pAC-scaffold interaction with typical cell morphology. After 14 days MSCs formed cell clusters but still expressed cartilage markers. Both cell types showed aggrecan, SOX9 gene and protein expression, cartilage proteoglycan and sulfated glycosaminoglycan synthesis for the whole culture time. Despite type II collagen gene expression could not anymore be detected at day 35, protein synthesis was visualized for both cell types during the whole culturing period, increasing in pAC and declining after day 14 in hMSC cultures. The novel BG scaffold was stable, cytocompatible and cartilage-specific protein synthesis indicated maintenance of pAC's differentiated phenotype and chondro-instructive effects on hMSCs.
Collapse
Affiliation(s)
- Clemens Gögele
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst-Nathan Str. 1, 90419 Nuremberg, Germany; Department of Biosciences, Paris Lodron University Salzburg, Hellbrunnerstraße 34, 5020 Salzburg, Austria.
| | - Sven Wiltzsch
- Faculty of Material Engineering, Nuremberg, Institute of Technology Georg Simon Ohm, Nuremberg, Germany.
| | - Armin Lenhart
- Faculty of Material Engineering, Nuremberg, Institute of Technology Georg Simon Ohm, Nuremberg, Germany.
| | - Aurelio Civilleri
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst-Nathan Str. 1, 90419 Nuremberg, Germany; Department of Civil, Environmental, Aerospace, Materials Engineering, Universita' di Palermo, Palermo, Italy.
| | - Thomas Martin Weiger
- Department of Biosciences, Paris Lodron University Salzburg, Hellbrunnerstraße 34, 5020 Salzburg, Austria.
| | - Kerstin Schäfer-Eckart
- Bone marrow Transplantation Unit, Medizinische Klinik 5, Klinikum Nürnberg, Paracelsus Medizinische Privatuniversität, Nuremberg, Germany.
| | - Bernd Minnich
- Department of Biosciences, Paris Lodron University Salzburg, Hellbrunnerstraße 34, 5020 Salzburg, Austria.
| | - Lukas Forchheimer
- Faculty of Material Engineering, Nuremberg, Institute of Technology Georg Simon Ohm, Nuremberg, Germany
| | - Markus Hornfeck
- Faculty of Material Engineering, Nuremberg, Institute of Technology Georg Simon Ohm, Nuremberg, Germany.
| | - Gundula Schulze-Tanzil
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, Prof. Ernst-Nathan Str. 1, 90419 Nuremberg, Germany.
| |
Collapse
|
133
|
Camacho P, Behre A, Fainor M, Seims KB, Chow LW. Spatial organization of biochemical cues in 3D-printed scaffolds to guide osteochondral tissue engineering. Biomater Sci 2021; 9:6813-6829. [PMID: 34473149 DOI: 10.1039/d1bm00859e] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Functional repair of osteochondral (OC) tissue remains challenging because the transition from bone to cartilage presents gradients in biochemical and physical properties necessary for joint function. Osteochondral regeneration requires strategies that restore the spatial composition and organization found in the native tissue. Several biomaterial approaches have been developed to guide chondrogenic and osteogenic differentiation of human mesenchymal stem cells (hMSCs). These strategies can be combined with 3D printing, which has emerged as a useful tool to produce tunable, continuous scaffolds functionalized with bioactive cues. However, functionalization often includes one or more post-fabrication processing steps, which can lead to unwanted side effects and often produce biomaterials with homogeneously distributed chemistries. To address these challenges, surface functionalization can be achieved in a single step by solvent-cast 3D printing peptide-functionalized polymers. Peptide-poly(caprolactone) (PCL) conjugates were synthesized bearing hyaluronic acid (HA)-binding (HAbind-PCL) or mineralizing (E3-PCL) peptides, which have been shown to promote hMSC chondrogenesis or osteogenesis, respectively. This 3D printing strategy enables unprecedented control of surface peptide presentation and spatial organization within a continuous construct. Scaffolds presenting both cartilage-promoting and bone-promoting peptides had a synergistic effect that enhanced hMSC chondrogenic and osteogenic differentiation in the absence of differentiation factors compared to scaffolds without peptides or only one peptide. Furthermore, multi-peptide organization significantly influenced hMSC response. Scaffolds presenting HAbind and E3 peptides in discrete opposing zones promoted hMSC osteogenic behavior. In contrast, presenting both peptides homogeneously throughout the scaffolds drove hMSC differentiation towards a mixed population of articular and hypertrophic chondrocytes. These significant results indicated that hMSC behavior was driven by dual-peptide presentation and organization. The downstream potential of this platform is the ability to fabricate biomaterials with spatially controlled biochemical cues to guide functional tissue regeneration without the need for differentiation factors.
Collapse
Affiliation(s)
- Paula Camacho
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Anne Behre
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Matthew Fainor
- Integrated Degree in Engineering, Arts, and Sciences Program, Lehigh University, Bethlehem, PA, USA
| | - Kelly B Seims
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, PA, USA.
| | - Lesley W Chow
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA.,Department of Materials Science and Engineering, Lehigh University, Bethlehem, PA, USA.
| |
Collapse
|
134
|
Li Y, Yuan Z, Yang H, Zhong H, Peng W, Xie R. Recent Advances in Understanding the Role of Cartilage Lubrication in Osteoarthritis. Molecules 2021; 26:6122. [PMID: 34684706 PMCID: PMC8540456 DOI: 10.3390/molecules26206122] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 01/15/2023] Open
Abstract
The remarkable lubrication properties of normal articular cartilage play an essential role in daily life, providing almost frictionless movements of joints. Alterations of cartilage surface or degradation of biomacromolecules within synovial fluid increase the wear and tear of the cartilage and hence determining the onset of the most common joint disease, osteoarthritis (OA). The irreversible and progressive degradation of articular cartilage is the hallmark of OA. Considering the absence of effective options to treat OA, the mechanosensitivity of chondrocytes has captured attention. As the only embedded cells in cartilage, the metabolism of chondrocytes is essential in maintaining homeostasis of cartilage, which triggers motivations to understand what is behind the low friction of cartilage and develop biolubrication-based strategies to postpone or even possibly heal OA. This review firstly focuses on the mechanism of cartilage lubrication, particularly on boundary lubrication. Then the mechanotransduction (especially shear stress) of chondrocytes is discussed. The following summarizes the recent development of cartilage-inspired biolubricants to highlight the correlation between cartilage lubrication and OA. One might expect that the restoration of cartilage lubrication at the early stage of OA could potentially promote the regeneration of cartilage and reverse its pathology to cure OA.
Collapse
Affiliation(s)
- Yumei Li
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; (Y.L.); (H.Y.); (H.Z.)
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Zhongrun Yuan
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China;
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Hui Yang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; (Y.L.); (H.Y.); (H.Z.)
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Haijian Zhong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; (Y.L.); (H.Y.); (H.Z.)
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Weijie Peng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; (Y.L.); (H.Y.); (H.Z.)
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Renjian Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; (Y.L.); (H.Y.); (H.Z.)
- Jiangxi Province Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
135
|
Lai YY, Li D, Chang SW. Computational insights into the substrate recognition mechanism of cartilage extracellular matrix degradation. Comput Struct Biotechnol J 2021; 19:5535-5545. [PMID: 34712398 PMCID: PMC8526910 DOI: 10.1016/j.csbj.2021.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 11/23/2022] Open
Abstract
Articular cartilage is connective tissue that forms a slippery load-bearing joint surface between bones. With outstanding mechanical properties, it plays an essential role in cushioning impact and protecting the ends of bones. Abnormal mechanical stimulation, such as repetitive overloading or chondral injury, induces excessive cartilage extracellular matrix (ECM) degradation, leading to osteoarthritis and other joint disorders. A disintegrin and metalloproteinase with thrombospondin motifs-5 (ADAMTS-5) is an aggrecanase that dominates the catalysis of aggrecan, the major proteoglycan in the cartilage ECM. Intriguingly, unlike its critical cleavage site Glu373-374Ala, another potential cleavage site, Glu419-420Ala, composed of the same amino acids in the aggrecan interglobular domain, is not a major cleavage site. It remains unclear how ADAMTS-5 distinguishes between them and hydrolyzes the correct scissile bonds. This research introduces a bottom-up in silico approach to reveal the molecular mechanism by which ADAMTS-5 recognizes the cleavage site on aggrecan. It is hypothesized that the sequence in the vicinity assists ADAMTS-5 in positioning the cleavage site. Specific residues were found to serve as binding sites, helping aggrecan bind more stably and fit into the enzyme better. The findings provide insight into the substrate binding and recognition mechanism for cartilage ECM degradation from a brand new atomic-scale perspective, laying the foundation for prophylaxis and treatment of related joint diseases.
Collapse
Affiliation(s)
- Yen-Yu Lai
- National Taiwan University, Department of Civil Engineering, Taipei 10617, Taiwan
| | - Deng Li
- National Taiwan University, Department of Civil Engineering, Taipei 10617, Taiwan
| | - Shu-Wei Chang
- National Taiwan University, Department of Civil Engineering, Taipei 10617, Taiwan
- National Taiwan University, Department of Biomedical Engineering, Taipei 10617, Taiwan
| |
Collapse
|
136
|
Xu Q, Torres JE, Hakim M, Babiak PM, Pal P, Battistoni CM, Nguyen M, Panitch A, Solorio L, Liu JC. Collagen- and hyaluronic acid-based hydrogels and their biomedical applications. MATERIALS SCIENCE & ENGINEERING. R, REPORTS : A REVIEW JOURNAL 2021; 146:100641. [PMID: 34483486 PMCID: PMC8409465 DOI: 10.1016/j.mser.2021.100641] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Hydrogels have been widely investigated in biomedical fields due to their similar physical and biochemical properties to the extracellular matrix (ECM). Collagen and hyaluronic acid (HA) are the main components of the ECM in many tissues. As a result, hydrogels prepared from collagen and HA hold inherent advantages in mimicking the structure and function of the native ECM. Numerous studies have focused on the development of collagen and HA hydrogels and their biomedical applications. In this extensive review, we provide a summary and analysis of the sources, features, and modifications of collagen and HA. Specifically, we highlight the fabrication, properties, and potential biomedical applications as well as promising commercialization of hydrogels based on these two natural polymers.
Collapse
Affiliation(s)
- Qinghua Xu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jessica E Torres
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Mazin Hakim
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Paulina M Babiak
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Pallabi Pal
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Carly M Battistoni
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Michael Nguyen
- Department of Biomedical Engineering, University of California Davis, Davis, California 95616, United States
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California Davis, Davis, California 95616, United States
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Julie C Liu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| |
Collapse
|
137
|
Wu CC, Tarng YW, Hsu DZ, Srinivasan P, Yeh YC, Lai YP, Hsieh DJ. Supercritical carbon dioxide decellularized porcine cartilage graft with PRP attenuated OA progression and regenerated articular cartilage in ACLT-induced OA rats. J Tissue Eng Regen Med 2021; 15:1118-1130. [PMID: 34581513 DOI: 10.1002/term.3252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 07/09/2021] [Accepted: 09/04/2021] [Indexed: 11/06/2022]
Abstract
Knee osteoarthritis (OA) is a common degenerative articular disorder and considered one of the primary causes of pain and functional disability. Knee OA is prevalent in 10% of men and 13% of women aged 60 years above. The study aims to use cartilage tissue engineering that combines the triads of decellularized porcine cartilage graft as "scaffold," plasma rich platelet (PRP) as "signal" and chondrocytes from rat as "cell" to attenuate ACLT-induced OA progression and regenerate the knee cartilage in rats. Decellularization of the porcine cartilage was characterized by hematoxylin and eosin, 4,6-Diamidino-2-phenylindole staining, scanning electron microscopy and residual DNA quantification. The protective effect of decellularized porcine cartilage graft (dPCG) was evaluated by intra-articular administration in surgically induced anterior cruciate ligament transection (ACLT) rat osteoarthritis (OA) model. Supercritical carbon dioxide technology completely decellularized the porcine cartilage. Intra-articular administration of dPCG with or without PRP significantly reduced the ACLT-induced OA symptoms and attenuated the OA progression. Pain-relief by dPCG with or without PRP was assessed by capacitance meter and improved articular cartilage damage in the rat knee was characterized by X-ray and micro-CT. Besides, the histological analysis depicted cartilage protection by dPCG with or without PRP. The repairation and attenuation effect by dPCG with or without PRP in the articular knee cartilage damage were also explored by safranin-O, type II collagen, aggrecan and SOX-9 immuno-staining. To conclude, intra-articular administration of dPCG with or without PRP is efficient in repairing the damaged cartilage in the experimental OA model.
Collapse
Affiliation(s)
- Chia-Chun Wu
- Department of Orthopedics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yih-Wen Tarng
- Department of Orthopedic, Kaohsiung Veterans General Hospital, Kaohsiung city, Taiwan, ROC
| | - Dur-Zong Hsu
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | | | - Yi-Chun Yeh
- R&D Center, ACRO Biomedical Co., Ltd., Kaohsiung City, Taiwan, ROC
| | - Yi-Ping Lai
- R&D Center, ACRO Biomedical Co., Ltd., Kaohsiung City, Taiwan, ROC
| | - Dar-Jen Hsieh
- R&D Center, ACRO Biomedical Co., Ltd., Kaohsiung City, Taiwan, ROC
| |
Collapse
|
138
|
Eckersley A, Ozols M, Chen P, Tam V, Hoyland JA, Trafford A, Chan D, Sherratt MJ. Peptide Location Fingerprinting Reveals Tissue Region-Specific Differences in Protein Structures in an Ageing Human Organ. Int J Mol Sci 2021; 22:10408. [PMID: 34638745 PMCID: PMC8509034 DOI: 10.3390/ijms221910408] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/18/2022] Open
Abstract
In ageing tissues, long-lived extracellular matrix (ECM) proteins are susceptible to the accumulation of structural damage due to diverse mechanisms including glycation, oxidation and protease cleavage. Peptide location fingerprinting (PLF) is a new mass spectrometry (MS) analysis technique capable of identifying proteins exhibiting structural differences in complex proteomes. PLF applied to published young and aged intervertebral disc (IVD) MS datasets (posterior, lateral and anterior regions of the annulus fibrosus) identified 268 proteins with age-associated structural differences. For several ECM assemblies (collagens I, II and V and aggrecan), these differences were markedly conserved between degeneration-prone (posterior and lateral) and -resistant (anterior) regions. Significant differences in peptide yields, observed within collagen I α2, collagen II α1 and collagen V α1, were located within their triple-helical regions and/or cleaved C-terminal propeptides, indicating potential accumulation of damage and impaired maintenance. Several proteins (collagen V α1, collagen II α1 and aggrecan) also exhibited tissue region (lateral)-specific differences in structure between aged and young samples, suggesting that some ageing mechanisms may act locally within tissues. This study not only reveals possible age-associated differences in ECM protein structures which are tissue-region specific, but also highlights the ability of PLF as a proteomic tool to aid in biomarker discovery.
Collapse
Affiliation(s)
- Alexander Eckersley
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK; (M.O.); (J.A.H.)
| | - Matiss Ozols
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK; (M.O.); (J.A.H.)
- Department of Human Genetics, Wellcome Sanger Institute, Genome Campus, Hinxton CB10 1SA, UK
| | - Peikai Chen
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China; (P.C.); (V.T.); (D.C.)
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen 518053, China
| | - Vivian Tam
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China; (P.C.); (V.T.); (D.C.)
| | - Judith A. Hoyland
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK; (M.O.); (J.A.H.)
- NIHR Manchester Biomedical Research Centre, Central Manchester Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9PL, UK
| | - Andrew Trafford
- Manchester Academic Health Science Centre, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK;
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China; (P.C.); (V.T.); (D.C.)
| | - Michael J. Sherratt
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK; (M.O.); (J.A.H.)
| |
Collapse
|
139
|
Wang ST, Neo BH, Betts RJ. Glycosaminoglycans: Sweet as Sugar Targets for Topical Skin Anti-Aging. Clin Cosmet Investig Dermatol 2021; 14:1227-1246. [PMID: 34548803 PMCID: PMC8449875 DOI: 10.2147/ccid.s328671] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/19/2021] [Indexed: 12/23/2022]
Abstract
Glycosaminoglycans (GAGs) are long, linear polysaccharides comprised of repeating disaccharide units with pleiotropic biological functions, with the non-sulfated GAG hyaluronic acid (HA), and sulfated GAGs dermatan sulfate, chondroitin sulfate, heparan sulfate, keratan sulfate, and to a lesser extent heparin all being expressed in skin. Their ability to regulate keratinocyte proliferation and differentiation, inflammatory processes and extracellular matrix composition and quality demonstrates their critical role in regulating skin physiology. Similarly, the water-binding properties of GAGs and structural qualities, particularly for HA, are crucial for maintaining proper skin form and hydration. The biological importance of GAGs, as well as extensive evidence that their properties and functions are altered in both chronological and extrinsic skin aging, makes them highly promising targets to improve cosmetic skin quality. Within the present review, we examine the cutaneous biological activity of GAGs alongside the protein complexes they form called proteoglycans and summarize the age-related changes of these molecules in skin. We also examine current topical interventional approaches to modulate GAGs for improved skin quality such as direct exogenous administration of GAGs, with a particular interest in strategies targeted at potentiating GAG levels in skin through either attenuating GAG degradation or increasing GAG production.
Collapse
Affiliation(s)
- Siew Tein Wang
- L'Oréal Research & Innovation, L'Oréal Singapore, Singapore
| | - Boon Hoe Neo
- L'Oréal Research & Innovation, L'Oréal Singapore, Singapore
| | | |
Collapse
|
140
|
Vernerey FJ, Lalitha Sridhar S, Muralidharan A, Bryant SJ. Mechanics of 3D Cell-Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem Rev 2021; 121:11085-11148. [PMID: 34473466 DOI: 10.1021/acs.chemrev.1c00046] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hydrogels are highly water-swollen molecular networks that are ideal platforms to create tissue mimetics owing to their vast and tunable properties. As such, hydrogels are promising cell-delivery vehicles for applications in tissue engineering and have also emerged as an important base for ex vivo models to study healthy and pathophysiological events in a carefully controlled three-dimensional environment. Cells are readily encapsulated in hydrogels resulting in a plethora of biochemical and mechanical communication mechanisms, which recapitulates the natural cell and extracellular matrix interaction in tissues. These interactions are complex, with multiple events that are invariably coupled and spanning multiple length and time scales. To study and identify the underlying mechanisms involved, an integrated experimental and computational approach is ideally needed. This review discusses the state of our knowledge on cell-hydrogel interactions, with a focus on mechanics and transport, and in this context, highlights recent advancements in experiments, mathematical and computational modeling. The review begins with a background on the thermodynamics and physics fundamentals that govern hydrogel mechanics and transport. The review focuses on two main classes of hydrogels, described as semiflexible polymer networks that represent physically cross-linked fibrous hydrogels and flexible polymer networks representing the chemically cross-linked synthetic and natural hydrogels. In this review, we highlight five main cell-hydrogel interactions that involve key cellular functions related to communication, mechanosensing, migration, growth, and tissue deposition and elaboration. For each of these cellular functions, recent experiments and the most up to date modeling strategies are discussed and then followed by a summary of how to tune hydrogel properties to achieve a desired functional cellular outcome. We conclude with a summary linking these advancements and make the case for the need to integrate experiments and modeling to advance our fundamental understanding of cell-matrix interactions that will ultimately help identify new therapeutic approaches and enable successful tissue engineering.
Collapse
Affiliation(s)
- Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States.,Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Shankar Lalitha Sridhar
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States
| | - Archish Muralidharan
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Stephanie J Bryant
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States.,Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States.,BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States
| |
Collapse
|
141
|
Ryan C, Pugliese E, Shologu N, Gaspar D, Rooney P, Islam MN, O'Riordan A, Biggs M, Griffin M, Zeugolis D. A combined physicochemical approach towards human tenocyte phenotype maintenance. Mater Today Bio 2021; 12:100130. [PMID: 34632361 PMCID: PMC8488312 DOI: 10.1016/j.mtbio.2021.100130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 02/08/2023] Open
Abstract
During in vitro culture, bereft of their optimal tissue context, tenocytes lose their phenotype and function. Considering that tenocytes in their native tissue milieu are exposed simultaneously to manifold signals, combination approaches (e.g. growth factor supplementation and mechanical stimulation) are continuously gaining pace to control cell fate during in vitro expansion, albeit with limited success due to the literally infinite number of possible permutations. In this work, we assessed the potential of scalable and potent physicochemical approaches that control cell fate (substrate stiffness, anisotropic surface topography, collagen type I coating) and enhance extracellular matrix deposition (macromolecular crowding) in maintaining human tenocyte phenotype in culture. Cell morphology was primarily responsive to surface topography. The tissue culture plastic induced the largest nuclei area, the lowest aspect ratio, and the highest focal adhesion kinase. Collagen type I coating increased cell number and metabolic activity. Cell viability was not affected by any of the variables assessed. Macromolecular crowding intensely enhanced and accelerated native extracellular matrix deposition, albeit not in an aligned fashion, even on the grooved substrates. Gene analysis at day 14 revealed that the 130 kPa grooved substrate without collagen type I coating and under macromolecular crowding conditions positively regulated human tenocyte phenotype. Collectively, this work illustrates the beneficial effects of combined physicochemical approaches in controlling cell fate during in vitro expansion.
Collapse
Affiliation(s)
- C.N.M. Ryan
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - E. Pugliese
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - N. Shologu
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - D. Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - P. Rooney
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Md N. Islam
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Discipline of Biochemistry, School of Natural Sciences, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - A. O'Riordan
- Tyndall National Institute, University College Cork (UCC), Cork, Ireland
| | - M.J. Biggs
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - M.D. Griffin
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - D.I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
142
|
Jiang L, Lin J, Zhao S, Wu J, Jin Y, Yu L, Wu N, Wu Z, Wang Y, Lin M. ADAMTS5 in Osteoarthritis: Biological Functions, Regulatory Network, and Potential Targeting Therapies. Front Mol Biosci 2021; 8:703110. [PMID: 34434966 PMCID: PMC8381022 DOI: 10.3389/fmolb.2021.703110] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/05/2021] [Indexed: 01/16/2023] Open
Abstract
ADAMTS5 is involved in the pathogenesis of OA. As the major aggrecanase-degrading articular cartilage matrix, ADAMTS5, has been regarded as a potential target for OA treatment. We here provide an updated insight on the regulation of ADAMTS5 and newly discovered therapeutic strategies for OA. Pathophysiological and molecular mechanisms underlying articular inflammation and mechanotransduction, as well as chondrocyte hypertrophy were discussed, and the role of ADAMTS5 in each biological process was reviewed, respectively. Senescence, inheritance, inflammation, and mechanical stress are involved in the overactivation of ADAMTS5, contributing to the pathogenesis of OA. Multiple molecular signaling pathways were observed to modulate ADAMTS5 expression, namely, Runx2, Fgf2, Notch, Wnt, NF-κB, YAP/TAZ, and the other inflammatory signaling pathways. Based on the fundamental understanding of ADAMTS5 in OA pathogenesis, monoclonal antibodies and small molecule inhibitors against ADAMTS5 were developed and proved to be beneficial pre-clinically both in vitro and in vivo. Recent novel RNA therapies demonstrated potentials in OA animal models. To sum up, ADAMTS5 inhibition and its signaling pathway–based modulations showed great potential in future therapeutic strategies for OA.
Collapse
Affiliation(s)
- Lejian Jiang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Spine Lab, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiachen Lin
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Sen Zhao
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaqian Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongming Jin
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Yu
- Department of Operating Room, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nan Wu
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhihong Wu
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China
| | - Yue Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Spine Lab, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mao Lin
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Spine Lab, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
143
|
Phenotypic Characterization of Immortalized Chondrocytes from a Desbuquois Dysplasia Type 1 Mouse Model: A Tool for Studying Defects in Glycosaminoglycan Biosynthesis. Int J Mol Sci 2021; 22:ijms22179304. [PMID: 34502207 PMCID: PMC8431031 DOI: 10.3390/ijms22179304] [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: 06/25/2021] [Revised: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 11/17/2022] Open
Abstract
The complexity of skeletal pathologies makes use of in vivo models essential to elucidate the pathogenesis of the diseases; nevertheless, chondrocyte and osteoblast cell lines provide relevant information on the underlying disease mechanisms. Due to the limitations of primary chondrocytes, immortalized cells represent a unique tool to overcome this problem since they grow very easily for several passages. However, in the immortalization procedure the cells might lose the original phenotype; thus, these cell lines should be deeply characterized before their use. We immortalized primary chondrocytes from a Cant1 knock-out mouse, an animal model of Desbuquois dysplasia type 1, with a plasmid expressing the SV40 large and small T antigen. This cell line, based on morphological and biochemical parameters, showed preservation of the chondrocyte phenotype. In addition reduced proteoglycan synthesis and oversulfation of glycosaminoglycan chains were demonstrated, as already observed in primary chondrocytes from the Cant1 knock-out mouse. In conclusion, immortalized Cant1 knock-out chondrocytes maintained the disease phenotype observed in primary cells validating the in vitro model and providing an additional tool to further study the proteoglycan biosynthesis defect. The same approach might be extended to other cartilage disorders.
Collapse
|
144
|
Højland AT, Tavernier LJM, Schrauwen I, Sommen M, Topsakal V, Schatteman I, Dhooge I, Huber A, Zanetti D, Kunst HPM, Hoischen A, Petersen MB, Van Camp G, Fransen E. A wide range of protective and predisposing variants in aggrecan influence the susceptibility for otosclerosis. Hum Genet 2021; 141:951-963. [PMID: 34410490 DOI: 10.1007/s00439-021-02334-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/04/2021] [Indexed: 12/16/2022]
Abstract
In this study, we investigated the association of ACAN variants with otosclerosis, a frequent cause of hearing loss among young adults. We sequenced the coding, 5'-UTR and 3'-UTR regions of ACAN in 1497 unrelated otosclerosis cases and 1437 matched controls from six different subpopulations. The association between variants in ACAN and the disease risk was tested through single variant and gene-based association tests. After correction for multiple testing, 14 variants were significantly associated with otosclerosis, ten of which represented independent association signals. Eight variants showed a consistent association across all subpopulations. Allelic odds ratios of the variants identified four predisposing and ten protective variants. Gene-based tests showed an association of very rare variants in the 3'-UTR with the phenotype. The associated exonic variants are all located in the CS domain of ACAN and include both protective and predisposing variants with a broad spectrum of effect sizes and population frequencies. This includes variants with strong effect size and low frequency, typical for monogenic diseases, to low effect size variants with high frequency, characteristic for common complex traits. This single-gene allelic spectrum with both protective and predisposing alleles is unique in the field of complex diseases. In conclusion, these findings are a significant advancement to the understanding of the etiology of otosclerosis.
Collapse
Affiliation(s)
- Allan Thomas Højland
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Research and Knowledge Center in Sensory Genetics, Department of Clinical Genetics, Aalborg University Hospital, Aalborg, Denmark
| | - Lisse J M Tavernier
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Isabelle Schrauwen
- Center for Statistical Genetics, Department of Neurology, Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY, USA
| | - Manou Sommen
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Vedat Topsakal
- Department of ORL and Head and Neck Surgery, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | - Isabelle Schatteman
- European Institute for ORL, St-Augustinus Hospital Antwerp, Antwerp, Belgium
| | - Ingeborg Dhooge
- Department of Otolaryngology, Ghent University Hospital, Ghent, Belgium
| | - Alex Huber
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Diego Zanetti
- Department of Clinical Sciences and Community Health, Audiology Unit, University of Milan, I.R.C.C.S. Fondazione "Cà Granda", Osp.Le Maggiore Policlinico, Milano, Italy
| | - Henricus P M Kunst
- Department of Otorhinolaryngology, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands.,Department of Otorhinolaryngology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Alexander Hoischen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Otorhinolaryngology, Hearing and Genes, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michael B Petersen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.,Research and Knowledge Center in Sensory Genetics, Department of Clinical Genetics, Aalborg University Hospital, Aalborg, Denmark
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.
| | - Erik Fransen
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium. .,StatUa Center for Statistics, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
145
|
Kim W, Choi JH, Kim P, Youn J, Song JE, Motta A, Migliaresi C, Khang G. Preparation and evaluation of gellan gum hydrogel reinforced with silk fibers with enhanced mechanical and biological properties for cartilage tissue engineering. J Tissue Eng Regen Med 2021; 15:936-947. [PMID: 34388313 DOI: 10.1002/term.3237] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 11/10/2022]
Abstract
Various research about cartilage regeneration using biomaterials has been done recently. Particularly, gellan gum hydrogel (GG) is reported to be suitable as a biomaterial for cartilage tissue engineering (TE) for its water uptaking ability, producibility, and environmental resemblance of native cartilage. Despite these advantages, mechanical and cell adhesion properties are still difficult to modulate. Reinforcement is essential to overcome these problems. Herein, GG was modified by physically blending with different lengths of silk fiber (SF). As SF is expected to improve such disadvantages of GG, mechanical and biological properties were characterized to confirm its reinforcement ability. Mechanical properties such as degradation rate, swelling rate, compression strength, and viscosity were studied and it was confirmed that SF significantly reinforces the mechanical properties of GG. Furthermore, in vitro study was carried out to confirm morphology, biocompatibility, proliferation, and chondrogenesis of chondrocytes encapsulated in the hydrogels. Overall, chondrocytes in the GG blended with SF (SF/GG) showed enhanced cell viability and growth. According to this study, SF/GG can be a promising biomaterial for cartilage TE biomaterial.
Collapse
Affiliation(s)
- Wooyoup Kim
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju-si, Korea
| | - Joo Hee Choi
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju-si, Korea
| | - Pilyun Kim
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju-si, Korea
| | - Jina Youn
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju-si, Korea
| | - Jeong Eun Song
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju-si, Korea
| | - Antonella Motta
- Department of Industrial Engineering and BIOtech Research Center, University of Trento, Trento, Italy
| | - Claudio Migliaresi
- Department of Industrial Engineering and BIOtech Research Center, University of Trento, Trento, Italy
| | - Gilson Khang
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, Jeonju-si, Korea.,Department of PolymerNano Science & Technology and Polymer Materials Fusion Research Center, Jeonbuk National University, Jeonju-si, Korea
| |
Collapse
|
146
|
Hayes AJ, Melrose J. Neural Tissue Homeostasis and Repair Is Regulated via CS and DS Proteoglycan Motifs. Front Cell Dev Biol 2021; 9:696640. [PMID: 34409033 PMCID: PMC8365427 DOI: 10.3389/fcell.2021.696640] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/13/2021] [Indexed: 01/04/2023] Open
Abstract
Chondroitin sulfate (CS) is the most abundant and widely distributed glycosaminoglycan (GAG) in the human body. As a component of proteoglycans (PGs) it has numerous roles in matrix stabilization and cellular regulation. This chapter highlights the roles of CS and CS-PGs in the central and peripheral nervous systems (CNS/PNS). CS has specific cell regulatory roles that control tissue function and homeostasis. The CNS/PNS contains a diverse range of CS-PGs which direct the development of embryonic neural axonal networks, and the responses of neural cell populations in mature tissues to traumatic injury. Following brain trauma and spinal cord injury, a stabilizing CS-PG-rich scar tissue is laid down at the defect site to protect neural tissues, which are amongst the softest tissues of the human body. Unfortunately, the CS concentrated in gliotic scars also inhibits neural outgrowth and functional recovery. CS has well known inhibitory properties over neural behavior, and animal models of CNS/PNS injury have demonstrated that selective degradation of CS using chondroitinase improves neuronal functional recovery. CS-PGs are present diffusely in the CNS but also form denser regions of extracellular matrix termed perineuronal nets which surround neurons. Hyaluronan is immobilized in hyalectan CS-PG aggregates in these perineural structures, which provide neural protection, synapse, and neural plasticity, and have roles in memory and cognitive learning. Despite the generally inhibitory cues delivered by CS-A and CS-C, some CS-PGs containing highly charged CS disaccharides (CS-D, CS-E) or dermatan sulfate (DS) disaccharides that promote neural outgrowth and functional recovery. CS/DS thus has varied cell regulatory properties and structural ECM supportive roles in the CNS/PNS depending on the glycoform present and its location in tissue niches and specific cellular contexts. Studies on the fruit fly, Drosophila melanogaster and the nematode Caenorhabditis elegans have provided insightful information on neural interconnectivity and the role of the ECM and its PGs in neural development and in tissue morphogenesis in a whole organism environment.
Collapse
Affiliation(s)
- Anthony J. Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Wales, United Kingdom
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Royal North Shore Hospital and The Faculty of Medicine and Health, The University of Sydney, St. Leonard’s, NSW, Australia
| |
Collapse
|
147
|
Keppie SJ, Mansfield JC, Tang X, Philp CJ, Graham HK, Önnerfjord P, Wall A, McLean C, Winlove CP, Sherratt MJ, Pavlovskaya GE, Vincent TL. Matrix-Bound Growth Factors are Released upon Cartilage Compression by an Aggrecan-Dependent Sodium Flux that is Lost in Osteoarthritis. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab037. [PMID: 34423304 PMCID: PMC8374957 DOI: 10.1093/function/zqab037] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/25/2021] [Accepted: 07/30/2021] [Indexed: 01/07/2023]
Abstract
Articular cartilage is a dense extracellular matrix-rich tissue that degrades following chronic mechanical stress, resulting in osteoarthritis (OA). The tissue has low intrinsic repair especially in aged and osteoarthritic joints. Here, we describe three pro-regenerative factors; fibroblast growth factor 2 (FGF2), connective tissue growth factor, bound to transforming growth factor-beta (CTGF-TGFβ), and hepatoma-derived growth factor (HDGF), that are rapidly released from the pericellular matrix (PCM) of articular cartilage upon mechanical injury. All three growth factors bound heparan sulfate, and were displaced by exogenous NaCl. We hypothesised that sodium, sequestered within the aggrecan-rich matrix, was freed by injurious compression, thereby enhancing the bioavailability of pericellular growth factors. Indeed, growth factor release was abrogated when cartilage aggrecan was depleted by IL-1 treatment, and in severely damaged human osteoarthritic cartilage. A flux in free matrix sodium upon mechanical compression of cartilage was visualised by 23Na -MRI just below the articular surface. This corresponded to a region of reduced tissue stiffness, measured by scanning acoustic microscopy and second harmonic generation microscopy, and where Smad2/3 was phosphorylated upon cyclic compression. Our results describe a novel intrinsic repair mechanism, controlled by matrix stiffness and mediated by the free sodium concentration, in which heparan sulfate-bound growth factors are released from cartilage upon injurious load. They identify aggrecan as a depot for sequestered sodium, explaining why osteoarthritic tissue loses its ability to repair. Treatments that restore matrix sodium to allow appropriate release of growth factors upon load are predicted to enable intrinsic cartilage repair in OA. SIGNIFICANCE STATEMENT Osteoarthritis is the most prevalent musculoskeletal disease, affecting 250 million people worldwide.1 We identify a novel intrinsic repair response in cartilage, mediated by aggrecan-dependent sodium flux, and dependent upon matrix stiffness, which results in the release of a cocktail of pro-regenerative growth factors after injury. Loss of aggrecan in late-stage osteoarthritis prevents growth factor release and likely contributes to disease progression. Treatments that restore matrix sodium in osteoarthritis may recover the intrinsic repair response to improve disease outcome.
Collapse
Affiliation(s)
- Stuart J Keppie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | | | - Xiaodi Tang
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - Christopher J Philp
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, NG7 2QX, UK
| | - Helen K Graham
- School of Biological Sciences, The University of Manchester, Manchester, M13 9PT, UK
| | - Patrik Önnerfjord
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Alanna Wall
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - Celia McLean
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - C Peter Winlove
- School of Physics and Astronomy, University of Exeter, Exeter, EX4 4QL, UK
| | - Michael J Sherratt
- School of Biological Sciences, The University of Manchester, Manchester, M13 9PT, UK
| | - Galina E Pavlovskaya
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, NG7 2QX, UK
| | | |
Collapse
|
148
|
Rios JJ, Denton K, Russell J, Kozlitina J, Ferreira CR, Lewanda AF, Mayfield JE, Moresco E, Ludwig S, Tang M, Li X, Lyon S, Khanshour A, Paria N, Khalid A, Li Y, Xie X, Feng JQ, Xu Q, Lu Y, Hammer RE, Wise CA, Beutler B. Germline Saturation Mutagenesis Induces Skeletal Phenotypes in Mice. J Bone Miner Res 2021; 36:1548-1565. [PMID: 33905568 PMCID: PMC8862308 DOI: 10.1002/jbmr.4323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 04/07/2021] [Accepted: 04/21/2021] [Indexed: 12/28/2022]
Abstract
Proper embryonic and postnatal skeletal development require coordination of myriad complex molecular mechanisms. Disruption of these processes, through genetic mutation, contributes to variation in skeletal development. We developed a high-throughput N-ethyl-N-nitrosourea (ENU)-induced saturation mutagenesis skeletal screening approach in mice to identify genes required for proper skeletal development. Here, we report initial results from live-animal X-ray and dual-energy X-ray absorptiometry (DXA) imaging of 27,607 G3 mice from 806 pedigrees, testing the effects of 32,198 coding/splicing mutations in 13,020 genes. A total of 39.7% of all autosomal genes were severely damaged or destroyed by mutations tested twice or more in the homozygous state. Results from our study demonstrate the feasibility of in vivo mutagenesis to identify mouse models of skeletal disease. Furthermore, our study demonstrates how ENU mutagenesis provides opportunities to create and characterize putative hypomorphic mutations in developmentally essential genes. Finally, we present a viable mouse model and case report of recessive skeletal disease caused by mutations in FAM20B. Results from this study, including engineered mouse models, are made publicly available via the online Mutagenetix database. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Jonathan J Rios
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA.,Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA.,McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA.,Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, TX, USA.,Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Kristin Denton
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Jamie Russell
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Julia Kozlitina
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
| | - Carlos R Ferreira
- Skeletal Genomics Unit, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy F Lewanda
- Rare Disease Institute, Children's National Hospital, Washington, DC, USA
| | - Joshua E Mayfield
- Department of Pharmacology, University of California, San Diego, CA, USA
| | - Eva Moresco
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sara Ludwig
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Miao Tang
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Xiaohong Li
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Stephen Lyon
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| | - Anas Khanshour
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Nandina Paria
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Aysha Khalid
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Yang Li
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Xudong Xie
- Department of Restorative Sciences, School of Dentistry, Texas A&M University, Dallas, TX, USA
| | - Jian Q Feng
- Department of Restorative Sciences, School of Dentistry, Texas A&M University, Dallas, TX, USA
| | - Qian Xu
- Department of Restorative Sciences, School of Dentistry, Texas A&M University, Dallas, TX, USA
| | - Yongbo Lu
- Department of Restorative Sciences, School of Dentistry, Texas A&M University, Dallas, TX, USA
| | - Robert E Hammer
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Carol A Wise
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA.,Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA.,McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA.,Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bruce Beutler
- Center for Genetics of Host Defense, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
149
|
Foster NC, Allen P, El Haj AJ, Grover LM, Moakes RJA. Tailoring Therapeutic Responses via Engineering Microenvironments with a Novel Synthetic Fluid Gel. Adv Healthc Mater 2021; 10:e2100622. [PMID: 34160135 PMCID: PMC11468753 DOI: 10.1002/adhm.202100622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/31/2021] [Indexed: 12/28/2022]
Abstract
This study reports the first fully synthetic fluid gel (SyMGels) using a simple poly(ethylene glycol) polymer. Fluid gels are an interesting class of materials: structured during gelation via shear-confinement to form microparticulate suspensions, through a bottom-up approach. Structuring in this way, when compared to first forming a gel and subsequently breaking it down, results in the formation of a particulate dispersion with particles "grown" in the shear flow. Resultantly, systems form a complex microstructure, where gelled particles concentrate remaining non-gelled polymer within the continuous phase, creating an amorphous-like interstitial phase. As such, these materials demonstrate mechanical characteristics typical of colloidal glasses, presenting solid-like behaviors at rest with defined yielding; likely through intrinsic particle-particle and particle-polymer interactions. To date, fluid gels have been fabricated using polysaccharides with relatively complex chemistries, making further modifications challenging. SyMGels are easily functionalised, using simple click-chemistry. This chemical flexibility, allows the creation of microenvironments with discrete biological decoration. Cellular control is demonstrated using MSC (mesenchymal stem cells)/chondrocytes and enables the regulation of key biomarkers such as aggrecan and SOX9. These potential therapeutic platforms demonstrate an important advancement in the biomaterial field, underpinning the mechanisms which drive their mechanical properties, and providing a versatile delivery system for advanced therapeutics.
Collapse
Affiliation(s)
- Nicola C. Foster
- Healthcare Technologies InstituteUniversity of BirminghamBirminghamB15 2TTUK
| | - Piers Allen
- Healthcare Technologies InstituteUniversity of BirminghamBirminghamB15 2TTUK
| | - Alicia J. El Haj
- Healthcare Technologies InstituteUniversity of BirminghamBirminghamB15 2TTUK
| | - Liam M. Grover
- Healthcare Technologies InstituteUniversity of BirminghamBirminghamB15 2TTUK
| | | |
Collapse
|
150
|
The Hexosamine Biosynthetic Pathway as a Therapeutic Target after Cartilage Trauma: Modification of Chondrocyte Survival and Metabolism by Glucosamine Derivatives and PUGNAc in an Ex Vivo Model. Int J Mol Sci 2021; 22:ijms22147247. [PMID: 34298867 PMCID: PMC8305151 DOI: 10.3390/ijms22147247] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 06/29/2021] [Accepted: 07/02/2021] [Indexed: 11/22/2022] Open
Abstract
The hexosamine biosynthetic pathway (HBP) is essential for the production of uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), the building block of glycosaminoglycans, thus playing a crucial role in cartilage anabolism. Although O-GlcNAcylation represents a protective regulatory mechanism in cellular processes, it has been associated with degenerative diseases, including osteoarthritis (OA). The present study focuses on HBP-related processes as potential therapeutic targets after cartilage trauma. Human cartilage explants were traumatized and treated with GlcNAc or glucosamine sulfate (GS); PUGNAc, an inhibitor of O-GlcNAcase; or azaserine (AZA), an inhibitor of GFAT-1. After 7 days, cell viability and gene expression analysis of anabolic and catabolic markers, as well as HBP-related enzymes, were performed. Moreover, expression of catabolic enzymes and type II collagen (COL2) biosynthesis were determined. Proteoglycan content was assessed after 14 days. Cartilage trauma led to a dysbalanced expression of different HBP-related enzymes, comparable to the situation in highly degenerated tissue. While GlcNAc and PUGNAc resulted in significant cell protection after trauma, only PUGNAc increased COL2 biosynthesis. Moreover, PUGNAc and both glucosamine derivatives had anti-catabolic effects. In contrast, AZA increased catabolic processes. Overall, “fueling” the HBP by means of glucosamine derivatives or inhibition of deglycosylation turned out as cells and chondroprotectives after cartilage trauma.
Collapse
|