1
|
Troop LD, Puetzer JL. Intermittent cyclic stretch of engineered ligaments drives hierarchical collagen fiber maturation in a dose- and organizational-dependent manner. Acta Biomater 2024; 185:296-311. [PMID: 39025395 PMCID: PMC11381169 DOI: 10.1016/j.actbio.2024.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Hierarchical collagen fibers are the primary source of strength in tendons and ligaments; however, these fibers largely do not regenerate after injury or with repair, resulting in limited treatment options. We previously developed a static culture system that guides ACL fibroblasts to produce native-sized fibers and early fascicles by 6 weeks. These constructs are promising ligament replacements, but further maturation is needed. Mechanical cues are critical for development in vivo and in engineered tissues; however, the effect on larger fiber and fascicle formation is largely unknown. Our objective was to investigate whether intermittent cyclic stretch, mimicking rapid muscle activity, drives further maturation in our system to create stronger engineered replacements and to explore whether cyclic loading has differential effects on cells at different degrees of collagen organization to better inform engineered tissue maturation protocols. Constructs were loaded with an established intermittent cyclic loading regime at 5 or 10 % strain for up to 6 weeks and compared to static controls. Cyclic loading drove cells to increase hierarchical collagen organization, collagen crimp, and tissue tensile properties, ultimately producing constructs that matched or exceeded immature ACL properties. Further, the effect of loading on cells varied depending on degree of organization. Specifically, 10 % load drove early improvements in tensile properties and composition, while 5 % load was more beneficial later in culture, suggesting a shift in mechanotransduction. This study provides new insight into how cyclic loading affects cell-driven hierarchical fiber formation and maturation, which will help to develop better rehabilitation protocols and engineer stronger replacements. STATEMENT OF SIGNIFICANCE: Collagen fibers are the primary source of strength and function in tendons and ligaments throughout the body. These fibers have limited regenerate after injury, with repair, and in engineered replacements, reducing treatment options. Cyclic load has been shown to improve fibril level alignment, but its effect at the larger fiber and fascicle length-scale is largely unknown. Here, we demonstrate intermittent cyclic loading increases cell-driven hierarchical fiber formation and tissue mechanics, producing engineered replacements with similar organization and mechanics as immature ACLs. This study provides new insight into how cyclic loading affects cell-driven fiber maturation. A better understanding of how mechanical cues regulate fiber formation will help to develop better engineered replacements and rehabilitation protocols to drive repair after injury.
Collapse
Affiliation(s)
- Leia D Troop
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Jennifer L Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, United States; Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA 23284, United States.
| |
Collapse
|
2
|
Şahin R, Aslan MG. Corneal aberrations are associated with low-energy meniscus injuries. BMC Ophthalmol 2024; 24:328. [PMID: 39107739 PMCID: PMC11302090 DOI: 10.1186/s12886-024-03601-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Visual impairment can cause balance problems. Therefore, visual impairment caused by an increase in corneal deviations can lead to sudden and unstable loads in the lower extremities. We aimed to investigate the possible relationship between low-energy meniscal injuries and corneal structural measures. METHODS This prospective, observational study included individuals aged between 18-40 years with a normal body-mass index. The study group consisted of 54 patients with grade 2 or 3 meniscus injuries after low-energy activity. The control group consisted of 54 healthy individuals without any complaints in the knee joint. The corneal parameters of all participants were evaluated with a Scheimpflug corneal topography and specular microscopy device. Simulated keratometry (SimK), minimum central corneal thickness (MCCT), cylindrical diopter (ClyD), corneal volume (CVol) spheric aberrations (SphAbb), high-order aberration (HOA), coma values, and endothelial parameters were recorded. RESULTS The research and control groups were similar in terms of age, body mass index, and gender distribution. There was no significant difference between the groups in the corneal SimK and CylD, parameters. However, HOA, Coma, SphAbb, and cell variability (Cv) values were significantly higher in the study group, and contrarily MCCT, CVol, and endothelial count (Cd) values were significantly lower. CONCLUSIONS Our findings suggest that individuals with relatively lower MCCT values tend to develop meniscal damage after low-energy activity. Hence, the loss of corneal strength in these patients may be a sign of possible weakness in the meniscus. The HOA value above 0.26, the coma value above 0.16, and the SphAbb value above 0.1 may significantly increase the possible meniscus injury.
Collapse
Affiliation(s)
- Rıfat Şahin
- Department of Orthopaedia and Traumatology, Faculty of Medicine, Recep Tayyip Erdogan University, İslampaşa Mah., Şehitler Cad., No:74, Zipcode: 53020, Merkez, Rize, Turkey.
| | - Mehmet Gökhan Aslan
- Department of Ophthalmology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
3
|
Wang T, Dogru S, Dai Z, Kim SY, Vickers NA, Albro MB. Physiologic Doses of Transforming Growth Factor-β Improve the Composition of Engineered Articular Cartilage. Tissue Eng Part A 2024. [PMID: 38874527 DOI: 10.1089/ten.tea.2023.0360] [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: 06/15/2024] Open
Abstract
Conventionally, for cartilage tissue engineering applications, transforming growth factor beta (TGF-β) is administered at doses that are several orders of magnitude higher than those present during native cartilage development. While these doses accelerate extracellular matrix (ECM) biosynthesis, they may also contribute to features detrimental to hyaline cartilage function, including tissue swelling, type I collagen (COL-I) deposition, cellular hypertrophy, and cellular hyperplasia. In contrast, during native cartilage development, chondrocytes are exposed to moderate TGF-β levels, which serve to promote strong biosynthetic enhancements while mitigating risks of pathology associated with TGF-β excesses. Here, we examine the hypothesis that physiologic doses of TGF-β can yield neocartilage with a more hyaline cartilage-like composition and structure relative to conventionally administered supraphysiologic doses. This hypothesis was examined on a model system of reduced-size constructs (∅2 × 2 mm or ∅3 × 2 mm) comprised of bovine chondrocytes encapsulated in agarose, which exhibit mitigated TGF-β spatial gradients allowing for an evaluation of the intrinsic effect of TGF-β doses on tissue development. Reduced-size (∅2 × 2 mm or ∅3 × 2 mm) and conventional-size constructs (∅4-∅6 mm × 2 mm) were subjected to a range of physiologic (0.1, 0.3, 1 ng/mL) and supraphysiologic (3, 10 ng/mL) TGF-β doses. At day 56, the physiologic 0.3 ng/mL dose yielded reduced-size constructs with native cartilage-matched Young's modulus (EY) (630 ± 58 kPa) and sulfated glycosaminoglycan (sGAG) content (5.9 ± 0.6%) while significantly increasing the sGAG-to-collagen ratio, leading to significantly reduced tissue swelling relative to constructs exposed to the supraphysiologic 10 ng/mL TGF-β dose. Furthermore, reduced-size constructs exposed to the 0.3 ng/mL dose exhibited a significant reduction in fibrocartilage-associated COL-I and a 77% reduction in the fraction of chondrocytes present in a clustered morphology, relative to the supraphysiologic 10 ng/mL dose (p < 0.001). EY was significantly lower for conventional-size constructs exposed to physiologic doses due to TGF-β transport limitations in these larger tissues (p < 0.001). Overall, physiologic TGF-β appears to achieve an important balance of promoting requisite ECM biosynthesis, while mitigating features detrimental to hyaline cartilage function. While reduced-size constructs are not suitable for the repair of clinical-size cartilage lesions, insights from this work can inform TGF-β dosing requirements for emerging scaffold release or nutrient channel delivery platforms capable of achieving uniform delivery of physiologic TGF-β doses to larger constructs required for clinical cartilage repair.
Collapse
Affiliation(s)
- Tianbai Wang
- Division of Materials Science & Engineering, Boston University, Boston MA, USA
| | - Sedat Dogru
- Department of Mechanical Engineering, Boston University, Boston MA, USA
| | - Zhonghao Dai
- Department of Biomedical Engineering, Boston University, Boston MA, USA
| | - Sung Yeon Kim
- Department of Biomedical Engineering, Boston University, Boston MA, USA
| | | | - Michael B Albro
- Department of Mechanical Engineering, Boston University, Boston MA, USA
| |
Collapse
|
4
|
Pancheri NM, Ellingson AJ, Marchus CR, Durgesh V, Verhage T, Yensen N, Schiele NR. Lysyl Oxidase Production by Murine C3H10T1/2 Mesenchymal Stem Cells Is Increased by TGFβs and Differentially Modulated by Mechanical Stimuli. Stem Cells Dev 2024; 33:355-364. [PMID: 38770821 DOI: 10.1089/scd.2023.0295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
Tendons are frequently injured and have limited regenerative capacity. This motivates tissue engineering efforts aimed at restoring tendon function through strategies to direct functional tendon formation. Generation of a crosslinked collagen matrix is paramount to forming mechanically functional tendon. However, it is unknown how lysyl oxidase (LOX), the primary mediator of enzymatic collagen crosslinking, is regulated by stem cells. This study investigates how multiple factors previously identified to promote tendon formation and healing (transforming growth factor [TGF]β1 and TGFβ2, mechanical stimuli, and hypoxia-inducible factor [HIF]-1α) regulate LOX production in the murine C3H10T1/2 mesenchymal stem cell (MSC) line. We hypothesized that TGFβ signaling promotes LOX activity in C3H10T1/2 MSCs, which is regulated by both mechanical stimuli and HIF-1α activation. TGFβ1 and TGFβ2 increased LOX levels as a function of concentration and time. Inhibiting the TGFβ type I receptor (TGFβRI) decreased TGFβ2-induced LOX production by C3H10T1/2 MSCs. Low (5 mPa) and high (150 mPa) magnitudes of fluid shear stress were applied to test impacts of mechanical stimuli, but without TGFβ2, loading alone did not alter LOX levels. Low loading (5 mPa) with TGFβ2 increased LOX at 7 days greater than TGFβ2 treatment alone. Neither HIF-1α knockdown (siRNA) nor activation (CoCl2) affected LOX levels. Ultimately, results suggest that TGFβ2 and appropriate loading magnitudes contribute to LOX production by C3H10T1/2 MSCs. Potential application of these findings includes treatment with TGFβ2 and appropriate mechanical stimuli to modulate LOX production by stem cells to ultimately control collagen matrix stiffening and support functional tendon formation.
Collapse
Affiliation(s)
- Nicholas M Pancheri
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Allison J Ellingson
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Colin R Marchus
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Vibhav Durgesh
- Department of Mechanical Engineering, University of Idaho, Moscow, Idaho, USA
| | - Tabitha Verhage
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Nicholas Yensen
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| | - Nathan R Schiele
- Department of Chemical & Biological Engineering, University of Idaho, Moscow, Idaho, USA
| |
Collapse
|
5
|
Puiggalí-Jou A, Rizzo R, Bonato A, Fisch P, Ponta S, Weber DM, Zenobi-Wong M. FLight Biofabrication Supports Maturation of Articular Cartilage with Anisotropic Properties. Adv Healthc Mater 2024; 13:e2302179. [PMID: 37867457 DOI: 10.1002/adhm.202302179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Indexed: 10/24/2023]
Abstract
Tissue engineering approaches that recapitulate cartilage biomechanical properties are emerging as promising methods to restore the function of injured or degenerated tissue. However, despite significant progress in this research area, the generation of engineered cartilage constructs akin to native counterparts still represents an unmet challenge. In particular, the inability to accurately reproduce cartilage zonal architecture with different collagen fibril orientations is a significant limitation. The arrangement of the extracellular matrix (ECM) plays a fundamental role in determining the mechanical and biological functions of the tissue. In this study, it is shown that a novel light-based approach, Filamented Light (FLight) biofabrication, can be used to generate highly porous, 3D cell-instructive anisotropic constructs that lead to directional collagen deposition. Using a photoclick-based photoresin optimized for cartilage tissue engineering, a significantly improved maturation of the cartilaginous tissues with zonal architecture and remarkable native-like mechanical properties is demonstrated.
Collapse
Affiliation(s)
- Anna Puiggalí-Jou
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Riccardo Rizzo
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, 52 Oxford Street, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Angela Bonato
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Philipp Fisch
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Simone Ponta
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| | - Daniel M Weber
- Division of Hand Surgery, University Children's Hospital Zürich, University of Zürich, Zürich, 8032, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich, 8093, Switzerland
| |
Collapse
|
6
|
Troop LD, Puetzer JL. Intermittent Cyclic Stretch of Engineered Ligaments Drives Hierarchical Collagen Fiber Maturation in a Dose- and Organizational-Dependent Manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.06.588420. [PMID: 38645097 PMCID: PMC11030411 DOI: 10.1101/2024.04.06.588420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Hierarchical collagen fibers are the primary source of strength in tendons and ligaments, however these fibers do not regenerate after injury or with repair, resulting in limited treatment options. We previously developed a culture system that guides ACL fibroblasts to produce native-sized fibers and fascicles by 6 weeks. These constructs are promising ligament replacements, but further maturation is needed. Mechanical cues are critical for development in vivo and in engineered tissues; however, the effect on larger fiber and fascicle formation is largely unknown. Our objective was to investigate whether intermittent cyclic stretch, mimicking rapid muscle activity, drives further maturation in our system to create stronger engineered replacements and to explore whether cyclic loading has differential effects on cells at different degrees of collagen organization to better inform engineered tissue maturation protocols. Constructs were loaded with an established intermittent cyclic loading regime at 5 or 10% strain for up to 6 weeks and compared to static controls. Cyclic loading drove cells to increase hierarchical collagen organization, collagen crimp, and tissue mechanics, ultimately producing constructs that matched or exceeded immature ACL properties. Further, the effect of loading on cells varied depending on degree of organization. Specifically, 10% load drove early improvements in mechanics and composition, while 5% load was more beneficial later in culture, suggesting a cellular threshold response and a shift in mechanotransduction. This study provides new insight into how cyclic loading affects cell-driven hierarchical fiber formation and maturation, which will help to develop better rehabilitation protocols and engineer stronger replacements.
Collapse
Affiliation(s)
- Leia D. Troop
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, United States
| | - Jennifer L. Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284, United States
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, 23284, United States
| |
Collapse
|
7
|
Ferreira SA, Tallia F, Heyraud A, Walker SA, Salzlechner C, Jones JR, Rankin SM. 3D printed hybrid scaffolds do not induce adverse inflammation in mice and direct human BM-MSC chondrogenesis in vitro. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100087. [PMID: 38312434 PMCID: PMC10835132 DOI: 10.1016/j.bbiosy.2024.100087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
Biomaterials that can improve the healing of articular cartilage lesions are needed. To address this unmet need, we developed novel 3D printed silica/poly(tetrahydrofuran)/poly(ε-caprolactone) (SiO2/PTHF/PCL-diCOOH) hybrid scaffolds. Our aim was to carry out essential studies to advance this medical device towards functional validation in pre-clinical trials. First, we show that the chemical composition, microarchitecture and mechanical properties of these scaffolds were not affected by sterilisation with gamma irradiation. To evaluate the systemic and local immunogenic reactivity of the sterilised 3D printed hybrid scaffolds, they were implanted subcutaneously into Balb/c mice. The scaffolds did not trigger a systemic inflammatory response over one week of implantation. The interaction between the host immune system and the implanted scaffold elicited a local physiological reaction with infiltration of mononuclear cells without any signs of a chronic inflammatory response. Then, we investigated how these 3D printed hybrid scaffolds direct chondrogenesis in vitro. Human bone marrow-derived mesenchymal stem/stromal cells (hBM-MSCs) seeded within the 3D printed hybrid scaffolds were cultured under normoxic or hypoxic conditions, with or without chondrogenic supplements. Chondrogenic differentiation assessed by both gene expression and protein production analyses showed that 3D printed hybrid scaffolds support hBM-MSC chondrogenesis. Articular cartilage-specific extracellular matrix deposition within these scaffolds was enhanced under hypoxic conditions (1.7 or 3.7 fold increase in the median of aggrecan production in basal or chondrogenic differentiation media). Our findings show that 3D printed SiO2/PTHF/PCL-diCOOH hybrid scaffolds have the potential to support the regeneration of cartilage tissue.
Collapse
Affiliation(s)
| | | | - Agathe Heyraud
- Department of Materials, Imperial College London, London, UK
| | - Simone A. Walker
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | - Julian R. Jones
- Department of Materials, Imperial College London, London, UK
| | - Sara M. Rankin
- National Heart & Lung Institute, Imperial College London, London, UK
| |
Collapse
|
8
|
Bernabei I, Faure E, Romani M, Wegrzyn J, Brinckmann J, Chobaz V, So A, Hugle T, Busso N, Nasi S. Inhibiting Lysyl Oxidases prevents pathologic cartilage calcification. Biomed Pharmacother 2024; 171:116075. [PMID: 38183742 DOI: 10.1016/j.biopha.2023.116075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024] Open
Abstract
Lysyl oxidases (LOX(L)) are enzymes that catalyze the formation of cross-links in collagen and elastin fibers during physiologic calcification of bone. However, it remains unknown whether they may promote pathologic calcification of articular cartilage, an important hallmark of debilitating arthropathies. Here, we have studied the possible roles of LOX(L) in cartilage calcification, related and not related to their cross-linking activity. We first demonstrated that inhibition of LOX(L) by β-aminoproprionitrile (BAPN) significantly reduced calcification in murine and human chondrocytes, and in joint of meniscectomized mice. These BAPN's effects on calcification were accounted for by different LOX(L) roles. Firstly, reduced LOX(L)-mediated extracellular matrix cross-links downregulated Anx5, Pit1 and Pit2 calcification genes. Secondly, BAPN reduced collagen fibrotic markers Col1 and Col3. Additionally, LOX(L) inhibition blocked chondrocytes hypertrophic differentiation (Runx2 and COL10), pro-inflammatory IL-6 release and reactive oxygen species (ROS) production, all triggers of chondrocyte calcification. Through unbiased transcriptomic analysis we confirmed a positive correlation between LOX(L) genes and genes for calcification, hypertrophy and extracellular matrix catabolism. This association was conserved throughout species (mouse, human) and tissues that can undergo pathologic calcification (kidney, arteries, skin). Overall, LOX(L) play a critical role in the process of chondrocyte calcification and may be therapeutic targets to treat cartilage calcification in arthropathies.
Collapse
Affiliation(s)
- Ilaria Bernabei
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital and University of Lausanne; Lausanne, Switzerland
| | - Elodie Faure
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital and University of Lausanne; Lausanne, Switzerland
| | - Mario Romani
- Aging and Bone Metabolism Laboratory, Service of Geriatric Medicine & Geriatric Rehabilitation, Department of Medicine, Lausanne University Hospital and University of Lausanne; Lausanne, Switzerland
| | - Julien Wegrzyn
- Department of Orthopedic Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jürgen Brinckmann
- Department of Dermatology and Institute of Virology and Cell Biology, University of Lübeck, Lübeck, Germany
| | - Véronique Chobaz
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital and University of Lausanne; Lausanne, Switzerland
| | - Alexander So
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital and University of Lausanne; Lausanne, Switzerland
| | - Thomas Hugle
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital and University of Lausanne; Lausanne, Switzerland
| | - Nathalie Busso
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital and University of Lausanne; Lausanne, Switzerland
| | - Sonia Nasi
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital and University of Lausanne; Lausanne, Switzerland.
| |
Collapse
|
9
|
Arnhold J. Inflammation-Associated Cytotoxic Agents in Tumorigenesis. Cancers (Basel) 2023; 16:81. [PMID: 38201509 PMCID: PMC10778456 DOI: 10.3390/cancers16010081] [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: 12/03/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic inflammatory processes are related to all stages of tumorigenesis. As inflammation is closely associated with the activation and release of different cytotoxic agents, the interplay between cytotoxic agents and antagonizing principles is highlighted in this review to address the question of how tumor cells overcome the enhanced values of cytotoxic agents in tumors. In tumor cells, the enhanced formation of mitochondrial-derived reactive species and elevated values of iron ions and free heme are antagonized by an overexpression of enzymes and proteins, contributing to the antioxidative defense and maintenance of redox homeostasis. Through these mechanisms, tumor cells can even survive additional stress caused by radio- and chemotherapy. Through the secretion of active agents from tumor cells, immune cells are suppressed in the tumor microenvironment and an enhanced formation of extracellular matrix components is induced. Different oxidant- and protease-based cytotoxic agents are involved in tumor-mediated immunosuppression, tumor growth, tumor cell invasion, and metastasis. Considering the special metabolic conditions in tumors, the main focus here was directed on the disturbed balance between the cytotoxic agents and protective mechanisms in late-stage tumors. This knowledge is mandatory for the implementation of novel anti-cancerous therapeutic approaches.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
10
|
He S, Fu X, Wang L, Xue Y, Zhou L, Qiao S, An J, Xia T. Self-Assemble Silk Fibroin Microcapsules for Cartilage Regeneration through Gene Delivery and Immune Regulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302799. [PMID: 37264755 DOI: 10.1002/smll.202302799] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/22/2023] [Indexed: 06/03/2023]
Abstract
Effective treatments for cartilage defects are currently lacking. Gene delivery using proper delivery systems has shown great potential in cartilage regeneration. However, the inflammatory microenvironment generated by the defected cartilage severely affects the system's delivery efficiency. Therefore, this study reports a silk fibroin microcapsule (SFM) structure based on layer-by-layer self-assembly, in which interleukin-4 (IL-4) is modified on silk by click chemistry and loaded with lysyl oxidase plasmid DNA (LOX pDNA). The silk microcapsules display good biocompatibility and the release rate of genes can be adjusted by controlling the number of self-assembled layers. Moreover, the functionalized SFMs mixed with methacrylated gelatin (GelMA) exhibit good injectability. The IL-4 on the outer layer of the SFM can regulate macrophages to polarize toward the M2 type, thereby promoting cartilage matrix repair and inhibiting inflammation. The LOX pDNA loaded inside can be effectively delivered into cells to promote extracellular matrix generation, significantly promoting cartilage regeneration. The results of this study provide a promising biomaterial for cartilage repair, and this novel silk-based microcapsule delivery system can also provide strategies for the treatment of other diseases.
Collapse
Affiliation(s)
- Shuangjian He
- Department of orthopedics, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Xuejie Fu
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Liang Wang
- Department of orthopedics, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Yangyang Xue
- Department of orthopedics, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Long Zhou
- Department of orthopedics, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Shigang Qiao
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Jianzhong An
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Tingting Xia
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| |
Collapse
|
11
|
Wang T, Dogru S, Dai Z, Kim SY, Vickers NA, Albro MB. Physiologic Doses of TGF-β Improve the Composition of Engineered Articular Cartilage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559554. [PMID: 37808691 PMCID: PMC10557735 DOI: 10.1101/2023.09.27.559554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
For cartilage regeneration applications, transforming growth factor beta (TGF-β) is conventionally administered at highly supraphysiologic doses (10-10,000 ng/mL) in an attempt to cue cells to fabricate neocartilage that matches the composition, structure, and functional properties of native hyaline cartilage. While supraphysiologic doses enhance ECM biosynthesis, they are also associated with inducing detrimental tissue features, such as fibrocartilage matrix deposition, pathologic-like chondrocyte clustering, and tissue swelling. Here we investigate the hypothesis that moderated TGF-β doses (0.1-1 ng/mL), akin to those present during physiological cartilage development, can improve neocartilage composition. Variable doses of media-supplemented TGF-β were administered to a model system of reduced-size cylindrical constructs (Ø2-Ø3 mm), which mitigate the TGF-β spatial gradients observed in conventional-size constructs (Ø4-Ø6 mm), allowing for a novel assessment of the intrinsic effect of TGF-β doses on macroscale neocartilage properties and composition. The administration of physiologic TGF-β to reduced-size constructs yields neocartilage with native-matched sGAG content and mechanical properties while providing a more hyaline cartilage-like composition, marked by: 1) reduced fibrocartilage-associated type I collagen, 2) 77% reduction in the fraction of cells present in a clustered morphology, and 3) 45% reduction in the degree of tissue swelling. Physiologic TGF-β appears to achieve an important balance of promoting requisite ECM biosynthesis, while mitigating hyaline cartilage compositional deficits. These results can guide the development of novel physiologic TGF-β-delivering scaffolds to improve the regeneration clinical-sized neocartilage tissues.
Collapse
|
12
|
Stampoultzis T, Guo Y, Nasrollahzadeh N, Rana VK, Karami P, Pioletti DP. Low-oxygen tension augments chondrocyte sensitivity to biomimetic thermomechanical cues in cartilage-engineered constructs. iScience 2023; 26:107491. [PMID: 37599834 PMCID: PMC10432199 DOI: 10.1016/j.isci.2023.107491] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/14/2023] [Accepted: 07/23/2023] [Indexed: 08/22/2023] Open
Abstract
Chondrocytes respond to various biophysical cues, including oxygen tension, transient thermal signals, and mechanical stimuli. However, understanding how these factors interact to establish a unique regulatory microenvironment for chondrocyte function remains unclear. Herein, we explore these interactions using a joint-simulating bioreactor that independently controls the culture's oxygen concentration, evolution of temperature, and mechanical loading. Our analysis revealed significant coupling between these signals, resulting in a remarkable ∼14-fold increase in collagen type II (COL2a) and aggrecan (ACAN) mRNA expression. Furthermore, dynamic thermomechanical stimulation enhanced glycosaminoglycan and COL2a protein synthesis, with the magnitude of the biosynthetic changes being oxygen dependent. Additionally, our mechanistic study highlighted the crucial role of SRY-box transcription factor 9 (SOX9) as a major regulator of chondrogenic response, specifically expressed in response to combined biophysical signals. These findings illuminate the integration of various mechanobiological cues by chondrocytes and provide valuable insights for improving the extracellular matrix content in cartilage-engineered constructs.
Collapse
Affiliation(s)
- Theofanis Stampoultzis
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Yanheng Guo
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Naser Nasrollahzadeh
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Vijay Kumar Rana
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Peyman Karami
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Dominique P. Pioletti
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Lausanne, Switzerland
| |
Collapse
|
13
|
Kimura M, Nakase J, Ishikawa T, Asai K, Yoshimizu R, Kanayama T, Yanatori Y, Ozaki N, Tsuchiya H. Growth-related changes in the ultrastructure of the quadriceps tendon. Knee 2023; 42:357-363. [PMID: 37150023 DOI: 10.1016/j.knee.2023.04.014] [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: 12/09/2022] [Revised: 03/16/2023] [Accepted: 04/12/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND The purpose of this study was to investigate the effect of growth on the ultrastructural characteristics of the quadriceps tendon (QT). METHODS Eighteen included patients were classified into three groups based on age and epiphyseal plate condition: the 'immature group' consisted of patients with open epiphyseal plates (11.5 ± 1.6 years old; mean ± standard deviation), the 'young group' consisted of patients aged <20 years with closed epiphyseal plates (15.8 ± 1.0 years), and the 'adult group' consisted of all patients aged >20 years (29.8 ± 11.3 years) irrespective of epiphyseal plate condition. Tendon tissue samples were used for ultrastructural analysis by transmission electron microscopy. Minimum collagen fibril diameters were measured from the cross sections of collagen fibril images using Image J software. The average number of collagen fibers per sample was 797 ± 109, and the average collagen fibril diameter of each sample was compared using one-way analysis of variance. RESULTS The mean collagen fibril diameter was 89.7 ± 14.4 nm in the immature group, 94.8 ± 16.4 nm in the young group, and 107.2 ± 12.1 nm in the adult group, with significant differences between the immature and adult groups, and between the young and adult groups (P = 0.001 and P = 0.021, respectively); however, no significant differences were observed between the immature and young adult groups (P = 0.49). CONCLUSIONS The collagen fibril diameter of the QT was found to have increased with growth. The study provided insights into graft selection.
Collapse
Affiliation(s)
- Mitsuhiro Kimura
- Department of Orthopedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan; Department of Orthopedic Surgery, National Hospital Organization, Kanazawa Medical Center, Kanazawa, Japan
| | - Junsuke Nakase
- Department of Orthopedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan.
| | - Tatsuya Ishikawa
- Department of Functional Anatomy, Graduate School of Medical Science Kanazawa University, Kanazawa, Japan
| | - Kazuki Asai
- Department of Orthopedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan; Department of Orthopedic Surgery, National Hospital Organization Hokuriku Hospital, Kanazawa, Japan
| | - Rikuto Yoshimizu
- Department of Orthopedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Tomoyuki Kanayama
- Department of Orthopedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Yusuke Yanatori
- Department of Orthopedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Noriyuki Ozaki
- Department of Functional Anatomy, Graduate School of Medical Science Kanazawa University, Kanazawa, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
14
|
Bates ME, Troop L, Brown ME, Puetzer JL. Temporal application of lysyl oxidase during hierarchical collagen fiber formation differentially effects tissue mechanics. Acta Biomater 2023; 160:98-111. [PMID: 36822485 PMCID: PMC10064799 DOI: 10.1016/j.actbio.2023.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/06/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023]
Abstract
The primary source of strength in menisci, tendons, and ligaments are hierarchical collagen fibers; however, these fibers are not regenerated after injury nor in engineered replacements, resulting in limited repair options. Collagen strength is reliant on fiber alignment, density, diameter, and crosslinking. Recently, we developed a culture system which guides cells in high-density collagen gels to develop native-like hierarchically organized collagen fibers, which match native fiber alignment and diameters by 6 weeks. However, tensile moduli plateau at 1MPa, suggesting crosslinking may be lacking. Collagen crosslinking is regulated by lysyl oxidase (LOX) which forms immature crosslinks that condense into mature trivalent crosslinks. Trivalent crosslinks are thought to be the primarily source of strength in fibers, but it's not well understood how they form. The objective of this study was to evaluate the effect of exogenous LOX in our culture system at different stages of hierarchical fiber formation to produce stronger replacements and to better understand factors affecting crosslink maturation. We found treatment with LOX isoform LOXL2 did not restrict hierarchical fiber formation, with constructs still forming aligned collagen fibrils by 2 weeks, larger fibers by 4 weeks, and early fascicles by 6 weeks. However, LOXL2 treatment did significantly increase mature pyridinium (PYD) crosslink accumulation and tissue mechanics, with timing of LOXL2 supplementation during fiber formation having a significant effect. Overall, we found one week of LOXL2 supplementation at 4 weeks produced constructs with native-like fiber organization, increased PYD accumulation, and increased mechanics, ultimately matching the tensile modulus of immature bovine menisci. STATEMENT OF SIGNIFICANCE: Collagen fibers are the primary source of strength and function in connective tissues throughout the body, however it remains a challenge to develop these fibers in engineered replacements, greatly reducing treatment options. Here we demonstrate lysyl oxidase like 2 (LOXL2) can be used to significantly improve the mechanics of tissue engineered constructs, but timing of application is important and will most likely depend on degree of collagen organization or maturation. Currently there is limited understanding of how collagen crosslinking is regulated, and this system is a promising platform to further investigate cellular regulation of LOX crosslinking. Understanding the mechanism that regulates LOX production and activity is needed to ultimately regenerate functional repair or replacements for connective tissues throughout the body.
Collapse
Affiliation(s)
- Madison E Bates
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284
| | - Leia Troop
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284
| | - M Ethan Brown
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284
| | - Jennifer L Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, 23284; Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, 23284, United States.
| |
Collapse
|
15
|
Merryweather DJ, Weston N, Roe J, Parmenter C, Lewis MP, Roach P. Exploring the microstructure of hydrated collagen hydrogels under scanning electron microscopy. J Microsc 2023; 290:40-52. [PMID: 36718074 DOI: 10.1111/jmi.13174] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/05/2023] [Accepted: 01/25/2023] [Indexed: 02/01/2023]
Abstract
Collagen hydrogels are a rapidly expanding platform in bioengineering and soft materials engineering for novel applications focused on medical therapeutics, medical devices and biosensors. Observations linking microstructure to material properties and function enables rational design strategies to control this space. Visualisation of the microscale organisation of these soft hydrated materials presents unique technical challenges due to the relationship between hydration and the molecular organisation of a collagen gel. Scanning electron microscopy is a robust tool widely employed to visualise and explore materials on the microscale. However, investigation of collagen gel microstructure is difficult without imparting structural changes during preparation and/or observation. Electrons are poorly propagated within liquid-phase materials, limiting the ability of electron microscopy to interrogate hydrated gels. Sample preparation techniques to remove water induce artefactual changes in material microstructure particularly in complex materials such as collagen, highlighting a critical need to develop robust material handling protocols for the imaging of collagen hydrogels. Here a collagen hydrogel is fabricated, and the gel state explored under high-vacuum (10-6 Pa) and low-vacuum (80-120 Pa) conditions, and in an environmental SEM chamber. Visualisation of collagen fibres is found to be dependent on the degree of sample hydration, with higher imaging chamber pressures and humidity resulting in decreased feature fidelity. Reduction of imaging chamber pressure is used to induce evaporation of gel water content, revealing collagen fibres of significantly larger diameter than observed in samples dehydrated prior to imaging. Rapid freezing and cryogenic handling of the gel material is found to retain a porous 3D structure following sublimation of the gel water content. Comparative analysis of collagen hydrogel materials demonstrates the care needed when preparing hydrogel samples for electron microscopy.
Collapse
Affiliation(s)
- Daniel J Merryweather
- Department of Chemistry, School of Science, Loughborough University, Leicestershire, UK
| | - Nicola Weston
- Nanoscale and Microscale Research Centre, University of Nottingham, Nottingham, UK
| | - Jordan Roe
- Department of Chemistry, School of Science, Loughborough University, Leicestershire, UK.,Department of Materials, Loughborough University, Leicestershire, UK
| | | | - Mark P Lewis
- National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University, Leicestershire, UK
| | - Paul Roach
- Department of Chemistry, School of Science, Loughborough University, Leicestershire, UK
| |
Collapse
|
16
|
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22:48. [PMID: 36906534 PMCID: PMC10007858 DOI: 10.1186/s12943-023-01744-8] [Citation(s) in RCA: 154] [Impact Index Per Article: 154.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2023] [Indexed: 03/13/2023] Open
Abstract
The malignant tumor is a multi-etiological, systemic and complex disease characterized by uncontrolled cell proliferation and distant metastasis. Anticancer treatments including adjuvant therapies and targeted therapies are effective in eliminating cancer cells but in a limited number of patients. Increasing evidence suggests that the extracellular matrix (ECM) plays an important role in tumor development through changes in macromolecule components, degradation enzymes and stiffness. These variations are under the control of cellular components in tumor tissue via the aberrant activation of signaling pathways, the interaction of the ECM components to multiple surface receptors, and mechanical impact. Additionally, the ECM shaped by cancer regulates immune cells which results in an immune suppressive microenvironment and hinders the efficacy of immunotherapies. Thus, the ECM acts as a barrier to protect cancer from treatments and supports tumor progression. Nevertheless, the profound regulatory network of the ECM remodeling hampers the design of individualized antitumor treatment. Here, we elaborate on the composition of the malignant ECM, and discuss the specific mechanisms of the ECM remodeling. Precisely, we highlight the impact of the ECM remodeling on tumor development, including proliferation, anoikis, metastasis, angiogenesis, lymphangiogenesis, and immune escape. Finally, we emphasize ECM "normalization" as a potential strategy for anti-malignant treatment.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - He Dou
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Yu
- Department of Gynecological Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, 150001, China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150000, China.
| | - Min Xiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
17
|
Barceló X, Eichholz KF, Gonçalves IF, Garcia O, Kelly DJ. Bioprinting of structurally organized meniscal tissue within anisotropic melt electrowritten scaffolds. Acta Biomater 2023; 158:216-227. [PMID: 36638941 DOI: 10.1016/j.actbio.2022.12.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/15/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023]
Abstract
The meniscus is characterised by an anisotropic collagen fibre network which is integral to its biomechanical functionality. The engineering of structurally organized meniscal grafts that mimic the anisotropy of the native tissue remains a significant challenge. In this study, inkjet bioprinting was used to deposit a cell-laden bioink into additively manufactured scaffolds of differing architectures to engineer fibrocartilage grafts with user defined collagen architectures. Polymeric scaffolds consisting of guiding fibre networks with varying aspect ratios (1:1; 1:4; 1:16) were produced using either fused deposition modelling (FDM) or melt electrowriting (MEW), resulting in scaffolds with different internal architectures and fibre diameters. Scaffold architecture was found to influence the spatial organization of the collagen network laid down by the jetted cells, with higher aspect ratios (1:4 and 1:16) supporting the formation of structurally anisotropic tissues. The MEW scaffolds supported the development of a fibrocartilaginous tissue with compressive mechanical properties similar to that of native meniscus, while the anisotropic tensile properties of these constructs could be tuned by altering the fibre network aspect ratio. This MEW framework was then used to generate scaffolds with spatially distinct fibre patterns, which in turn supported the development of heterogenous tissues consisting of isotropic and anisotropic collagen networks. Such bioprinted tissues could potentially form the basis of new treatment options for damaged and diseased meniscal tissue. STATEMENT OF SIGNIFICANCE: This study describes a multiple tool biofabrication strategy which enables the engineering of spatially organized fibrocartilage tissues. The architecture of MEW scaffolds can be tailored to not only modulate the directionality of the collagen fibres laid down by cells, but also to tune the anisotropic tensile mechanical properties of the resulting constructs, thereby enabling the engineering of biomimetic meniscal-like tissues. Furthermore, the inherent flexibility of MEW enables the development of zonally defined and potentially patient-specific implants.
Collapse
Affiliation(s)
- Xavier Barceló
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, D02 R590, Ireland; Department of Mechanical, Manufacturing, & Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, D02 R590, Ireland; Advanced Materials & Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland & Trinity College Dublin, Dublin, D02 F6N2, Ireland
| | - Kian F Eichholz
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, D02 R590, Ireland; Department of Mechanical, Manufacturing, & Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, D02 R590, Ireland; Advanced Materials & Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland & Trinity College Dublin, Dublin, D02 F6N2, Ireland
| | - Inês F Gonçalves
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, D02 R590, Ireland; Department of Mechanical, Manufacturing, & Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, D02 R590, Ireland; Advanced Materials & Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland & Trinity College Dublin, Dublin, D02 F6N2, Ireland
| | - Orquidea Garcia
- Johnson & Johnson 3D Printing Innovation & Customer Solutions, Johnson & Johnson Services, Inc., Irvine, CA, USA
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, D02 R590, Ireland; Department of Mechanical, Manufacturing, & Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, D02 R590, Ireland; Advanced Materials & Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland & Trinity College Dublin, Dublin, D02 F6N2, Ireland; Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland.
| |
Collapse
|
18
|
Otarola GA, Hu JC, Athanasiou KA. Ion modulatory treatments toward functional self-assembled neocartilage. Acta Biomater 2022; 153:85-96. [PMID: 36113725 DOI: 10.1016/j.actbio.2022.09.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022]
Abstract
Signals that recapitulate in vitro the conditions found in vivo, such as hypoxia or mechanical forces, contribute to the generation of tissue-engineered hyaline-like tissues. The cell regulatory processes behind hypoxic and mechanical stimuli rely on ion concentration; iron is required to degrade the hypoxia inducible factor 1a (HIF1α) under normoxia, whereas the initiation of mechanotransduction requires the cytoplasmic increase of calcium concentration. In this work, we propose that ion modulation can be used to improve the biomechanical properties of self-assembled neocartilage constructs derived from rejuvenated expanded minipig rib chondrocytes. The objectives of this work were 1) to determine the effects of iron sequestration on self-assembled neocartilage constructs using two doses of the iron chelator deferoxamine (DFO), and 2) to evaluate the performance of the combined treatment of DFO and ionomycin, a calcium ionophore that triggers cytoplasmic calcium accumulation. This study employed a two-phase approach. In Phase I, constructs treated with a high dose of DFO (100 µM) exhibited an 87% increase in pyridinoline crosslinks, a 57% increase in the Young's modulus, and a 112% increase in the ultimate tensile strength (UTS) of the neotissue. In Phase II, the combined use of both ion modulators resulted in 150% and 176% significant increases in the Young's modulus and UTS of neocartilage constructs, respectively; for the first time, neocartilage constructs achieved a Young's modulus of 11.76±3.29 MPa and UTS of 4.20±1.24 MPa. The results of this work provide evidence that ion modulation can be employed to improve the biomechanical properties in engineered neotissues. STATEMENT OF SIGNIFICANCE: The translation of tissue-engineered products requires the development of strategies capable of producing biomimetic neotissues in a replicable, controllable, and cost-effective manner. Among other functions, Fe2+ and Ca2+ are involved in the control of the hypoxic response and mechanotransduction, respectively. Both stimuli, hypoxia and mechanical forces, are known to favor chondrogenesis. This study utilized ion modulators to improve the mechanical properties self-assembled neocartilage constructs derived from expanded and rejuvenated costal chondrocytes via Fe2+ sequestration and Ca2+ influx, alone or in combination. The results indicate that ion modulation induced tissue maturation and a significant improvement of the mechanical properties, and holds potential as a tool to mitigate the need for bioreactors and engineer hyaline-like tissues.
Collapse
Affiliation(s)
- Gaston A Otarola
- 3131 Engineering Hall, Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA
| | - Jerry C Hu
- 3131 Engineering Hall, Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA
| | - Kyriacos A Athanasiou
- 3131 Engineering Hall, Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA.
| |
Collapse
|
19
|
Dooling LJ, Saini K, Anlaş AA, Discher DE. Tissue mechanics coevolves with fibrillar matrisomes in healthy and fibrotic tissues. Matrix Biol 2022; 111:153-188. [PMID: 35764212 PMCID: PMC9990088 DOI: 10.1016/j.matbio.2022.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/16/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022]
Abstract
Fibrillar proteins are principal components of extracellular matrix (ECM) that confer mechanical properties to tissues. Fibrosis can result from wound repair in nearly every tissue in adults, and it associates with increased ECM density and crosslinking as well as increased tissue stiffness. Such fibrotic tissues are a major biomedical challenge, and an emerging view posits that the altered mechanical environment supports both synthetic and contractile myofibroblasts in a state of persistent activation. Here, we review the matrisome in several fibrotic diseases, as well as normal tissues, with a focus on physicochemical properties. Stiffness generally increases with the abundance of fibrillar collagens, the major constituent of ECM, with similar mathematical trends for fibrosis as well as adult tissues from soft brain to stiff bone and heart development. Changes in expression of other core matrisome and matrisome-associated proteins or proteoglycans contribute to tissue stiffening in fibrosis by organizing collagen, crosslinking ECM, and facilitating adhesion of myofibroblasts. Understanding how ECM composition and mechanics coevolve during fibrosis can lead to better models and help with antifibrotic therapies.
Collapse
Affiliation(s)
- Lawrence J Dooling
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Karanvir Saini
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Alişya A Anlaş
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Dennis E Discher
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Noori-Dokht H, Joukar A, Karnik S, Williams T, Trippel SB, Wagner DR. A Photochemical Crosslinking Approach to Enhance Resistance to Mechanical Wear and Biochemical Degradation of Articular Cartilage. Cartilage 2022; 13:19476035221093064. [PMID: 35819016 PMCID: PMC9280829 DOI: 10.1177/19476035221093064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The objective of this study was to evaluate photochemical crosslinking using Al(III) phthalocyanine chloride tetrasulfonic acid (CASPc) and light with a wavelength of 670 nm as a potential therapy to strengthen articular cartilage and prevent tissue degradation. DESIGN Changes in viscoelastic properties with indentation were used to identify 2 crosslinking protocols for further testing. Crosslinked cartilage was subjected to an in vitro, accelerated wear test. The ability of the crosslinked tissue to resist biochemical degradation via collagenase was also measured. To better understand how photochemical crosslinking with CASPc varies through the depth of the tissue, the distribution of photo-initiator and penetration of light through the tissue depth was characterized. Finally, the effect of CASPc on chondrocyte viability and of co-treatment with an antioxidant was evaluated. RESULTS The equilibrium modulus was the most sensitive viscoelastic measure of crosslinking. Crosslinking decreased both mechanical wear and collagenase digestion compared with control cartilage. These beneficial effects were realized despite the fact that crosslinking appeared to be localized to a region near the articular surface. In addition, chondrocyte viability was maintained in crosslinked tissue treated with antioxidants. CONCLUSION These results suggest that photochemical crosslinking with CASPc and 670 nm light holds promise as a potential therapy to prevent cartilage degeneration by protecting cartilage from mechanical wear and biochemical degradation. Limitations were also evident, however, as an antioxidant treatment was necessary to maintain chondrocyte viability in crosslinked tissue.
Collapse
Affiliation(s)
- Hessam Noori-Dokht
- Department of Mechanical & Energy Engineering, Indiana University–Purdue University Indianapolis, Indianapolis, IN, USA,School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Amin Joukar
- Department of Mechanical & Energy Engineering, Indiana University–Purdue University Indianapolis, Indianapolis, IN, USA,School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Sonali Karnik
- Department of Mechanical & Energy Engineering, Indiana University–Purdue University Indianapolis, Indianapolis, IN, USA
| | - Taylor Williams
- Department of Biomedical Engineering, Indiana University–Purdue University Indianapolis, Indianapolis, IN, USA
| | - Stephen B. Trippel
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Diane R. Wagner
- Department of Mechanical & Energy Engineering, Indiana University–Purdue University Indianapolis, Indianapolis, IN, USA,Department of Biomedical Engineering, Indiana University–Purdue University Indianapolis, Indianapolis, IN, USA,Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA,Diane R. Wagner, Department of Mechanical & Energy Engineering, Indiana University–Purdue University Indianapolis, 723 W. Michigan Street, SL 260, Indianapolis, IN 46220, USA.
| |
Collapse
|
21
|
Merrild NG, Holzmann V, Ariosa-Morejon Y, Faull PA, Coleman J, Barrell WB, Young G, Fischer R, Kelly DJ, Addison O, Vincent TL, Grigoriadis AE, Gentleman E. Local depletion of proteoglycans mediates cartilage tissue repair in an ex vivo integration model. Acta Biomater 2022; 149:179-188. [PMID: 35779773 DOI: 10.1016/j.actbio.2022.06.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/25/2022] [Accepted: 06/17/2022] [Indexed: 11/28/2022]
Abstract
Successfully replacing damaged cartilage with tissue-engineered constructs requires integration with the host tissue and could benefit from leveraging the native tissue's intrinsic healing capacity; however, efforts are limited by a poor understanding of how cartilage repairs minor defects. Here, we investigated the conditions that foster natural cartilage tissue repair to identify strategies that might be exploited to enhance the integration of engineered/grafted cartilage with host tissue. We damaged porcine articular cartilage explants and using a combination of pulsed SILAC-based proteomics, ultrastructural imaging, and catabolic enzyme blocking strategies reveal that integration of damaged cartilage surfaces is not driven by neo-matrix synthesis, but rather local depletion of proteoglycans. ADAMTS4 expression and activity are upregulated in injured cartilage explants, but integration could be reduced by inhibiting metalloproteinase activity with TIMP3. These observations suggest that catabolic enzyme-mediated proteoglycan depletion likely allows existing collagen fibrils to undergo cross-linking, fibrillogenesis, or entanglement, driving integration. Catabolic enzymes are often considered pathophysiological markers of osteoarthritis. Our findings suggest that damage-induced upregulation of metalloproteinase activity may be a part of a healing response that tips towards tissue destruction under pathological conditions and in osteoarthritis, but could also be harnessed in tissue engineering strategies to mediate repair. STATEMENT OF SIGNIFICANCE: Cartilage tissue engineering strategies require graft integration with the surrounding tissue; however, how the native tissue repairs minor injuries is poorly understood. We applied pulsed SILAC-based proteomics, ultrastructural imaging, and catabolic enzyme blocking strategies to a porcine cartilage explant model and found that integration of damaged cartilage surfaces is driven by catabolic enzyme-mediated local depletion of proteoglycans. Although catabolic enzymes have been implicated in cartilage destruction in osteoarthritis, our findings suggest that damage-induced upregulation of metalloproteinase activity may be a part of a healing response that tips towards tissue destruction under pathological conditions. They also suggest that this natural cartilage tissue repair process could be harnessed in tissue engineering strategies to enhance the integration of engineered cartilage with host tissue.
Collapse
Affiliation(s)
- Nicholas Groth Merrild
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Viktoria Holzmann
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Yoanna Ariosa-Morejon
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Peter A Faull
- College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Jennifer Coleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - William B Barrell
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Gloria Young
- Department of Materials, Imperial College London, London SW7 2AZ, UK
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Owen Addison
- Centre for Oral, Clinical and Translational Sciences, King's College London, London SE1 9RT, UK
| | - Tonia L Vincent
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | | | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK.
| |
Collapse
|
22
|
Abstract
Tendons are collagen-rich musculoskeletal tissues that possess the mechanical strength needed to transfer forces between muscles and bones. The mechanical development and function of tendons are impacted by collagen crosslinks. However, there is a limited understanding of how collagen crosslinking is regulated in tendon during development and aging. Therefore, the objective of the present review was to highlight potential regulators of enzymatic and non-enzymatic collagen crosslinking and how they impact tendon function. The main collagen crosslinking enzymes include lysyl oxidase (LOX) and the lysyl oxidase-like isoforms (LOXL), whereas non-enzymatic crosslinking is mainly mediated by the formation of advanced glycation end products (AGEs). Regulators of the LOX and LOXL enzymes may include mechanical stimuli, mechanotransducive cell signaling pathways, sex hormones, transforming growth factor (TGF)β family, hypoxia, and interactions with intracellular or extracellular proteins. AGE accumulation in tendon is due to diabetic conditions and aging, and can be mediated by diet and mechanical stimuli. The formation of these enzymatic and non-enzymatic collagen crosslinks plays a major role in tendon biomechanics and in the mechanisms of force transfer. A more complete understanding of how enzymatic and non-enzymatic collagen crosslinking is regulated in tendon will better inform tissue engineering and regenerative therapies aimed at restoring the mechanical function of damaged tendons.
Collapse
Affiliation(s)
- A.J. Ellingson
- Chemical and Biological Engineering, University of Idaho, Moscow, ID, USA
| | - N.M. Pancheri
- Chemical and Biological Engineering, University of Idaho, Moscow, ID, USA
| | - N.R. Schiele
- Chemical and Biological Engineering, University of Idaho, Moscow, ID, USA,Address for correspondence: Nathan R. Schiele, Chemical and Biological Engineering, University of Idaho, 875 Perimeter Dr. MS 0904, Moscow, ID, USA. Telephone number: 208 8859063
| |
Collapse
|
23
|
Marchus CR, Knudson JA, Morrison AE, Strawn IK, Hartman AJ, Shrestha D, Pancheri NM, Glasgow I, Schiele NR. Low-cost, open-source cell culture chamber for regulating physiologic oxygen levels. HARDWAREX 2022; 11:e00253. [PMID: 35509920 PMCID: PMC9058583 DOI: 10.1016/j.ohx.2021.e00253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
The physiological oxygen levels for several mammalian cell types in vivo are considered to be hypoxic (low oxygen tension), but the vast majority of in vitro mammalian cell culture is conducted at atmospheric oxygen levels of around 21%. In order to understand the impact of low oxygen environments on cells, oxygen levels need to be regulated during in vitro culture. Two common methods for simulating a hypoxic environment are through the regulation of gas composition or chemical induction. Chemically mimicking hypoxia can have adverse effects such as reducing cell viability, making oxygen regulation in cell culture chambers crucial for long-term culture. However, oxygen-regulating cell culture incubators and commercial hypoxia chambers may not always be a viable option due to cost and limited customization. Other low-cost chambers have been developed, but they tend to lack control systems or are fairly small scale. Thus, the objective of this project was to design and develop a low-cost, open-source, controllable, and reproducible hypoxia chamber that can fit inside a standard cell culture incubator. This design allows for the control of O2 between 1 and 21%, while maintaining CO2 levels at 5%, as well as monitoring of temperature, pressure, and relative humidity. Testing showed our hypoxia chamber was able to maintain CO2 levels at 5% and hypoxic O2 levels at 1% and 5% for long-term cell culture. This simple and easy-to-manufacture design uses off the shelf components, and the total material cost was $832.47 (USD).
Collapse
Affiliation(s)
- Colin R.N. Marchus
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| | - Jacob A. Knudson
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| | - Alexandra E. Morrison
- University of Idaho, Department of Electrical and Computer Engineering, Moscow, ID, United States
| | - Isabell K. Strawn
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| | - Andrew J. Hartman
- University of Idaho, Department of Electrical and Computer Engineering, Moscow, ID, United States
| | - Dev Shrestha
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| | - Nicholas M. Pancheri
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| | - Ian Glasgow
- University of Idaho, Department of Mechanical Engineering, Moscow, ID, United States
| | - Nathan R. Schiele
- University of Idaho, Department of Chemical & Biological Engineering, Moscow, ID, United States
| |
Collapse
|
24
|
Lan X, Liang Y, Vyhlidal M, Erkut EJN, Kunze M, Mulet-Sierra A, Osswald M, Ansari K, Seikaly H, Boluk Y, Adesida AB. In vitro maturation and in vivo stability of bioprinted human nasal cartilage. J Tissue Eng 2022; 13:20417314221086368. [PMID: 35599742 PMCID: PMC9122109 DOI: 10.1177/20417314221086368] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/23/2022] [Indexed: 11/25/2022] Open
Abstract
The removal of skin cancer lesions on the nose often results in the loss of nasal
cartilage. The cartilage loss is either surgically replaced with autologous
cartilage or synthetic grafts. However, these replacement options come with
donor-site morbidity and resorption issues. 3-dimensional (3D) bioprinting
technology offers the opportunity to engineer anatomical-shaped autologous nasal
cartilage grafts. The 3D bioprinted cartilage grafts need to embody a
mechanically competent extracellular matrix (ECM) to allow for surgical suturing
and resistance to contraction during scar tissue formation. We investigated the
effect of culture period on ECM formation and mechanical properties of 3D
bioprinted constructs of human nasal chondrocytes (hNC)-laden type I collagen
hydrogel in vitro and in vivo. Tissue-engineered nasal cartilage constructs
developed from hNC culture in clinically approved collagen type I and type III
semi-permeable membrane scaffold served as control. The resulting 3D bioprinted
engineered nasal cartilage constructs were comparable or better than the
controls both in vitro and in vivo. This study demonstrates that 3D bioprinted
constructs of engineered nasal cartilage are feasible options in nasal cartilage
reconstructive surgeries.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Yan Liang
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Margaret Vyhlidal
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Esra JN Erkut
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Martin Osswald
- Institute for Reconstructive Sciences in Medicine, Misericordia Community Hospital, Edmonton, AB, Canada
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Khalid Ansari
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Hadi Seikaly
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Yaman Boluk
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Adetola B Adesida
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
25
|
Yuan Z, Long T, Zhang J, Lyu Z, Zhang W, Meng X, Qi J, Wang Y. 3D printed porous sulfonated polyetheretherketone scaffold for cartilage repair: Potential and limitation. J Orthop Translat 2022; 33:90-106. [PMID: 35330941 PMCID: PMC8913250 DOI: 10.1016/j.jot.2022.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 01/04/2023] Open
Abstract
Objective The treatment of cartilage lesions has always been a difficult problem. Although cartilage tissue engineering provides alternative treatment options for cartilage lesions, biodegradable tissue engineering scaffolds have limitations. Methods In this study, we constructed a porous PEEK scaffold via 3D printing, surface-engineered with concentrated sulfuric acid for 15 s (SPK-15), 30 s (SPK-30), and 60 s (SPK-60). We systematically evaluated the physical and chemical characteristics and biofunctionalities of the scaffolds, and then evaluated the macrophage polarization modulating ability and anti-inflammatory effects of the sulfonated PEEK, and observed the cartilage-protective effect of SPK using a co-culture study. We further evaluated the repair effect of PEEK and SPK by implanting the prosthetic scaffold into a cartilage defect in a rabbit model. Results Compared to the PEEK, SPK-15 and SPK-60 scaffolds, SPK-30 has a good micro/nanostructure, appropriate biomechanical properties (compressive modulus, 43 ± 5 MPa; Shaw hardness, 20.6 ± 1.3 HD; close to native cartilage, 30 ± 8 MPa, 17.8 ± 0.8 HD), and superior biofunctionalities. Compared to PEEK, sulfonated PEEK can favor macrophage polarization to the M2 phenotype, which increases anti-inflammatory cytokine secretion. Furthermore, SPK can also prevent macrophage-induced cartilage degeneration. The in-vivo animal experiment demonstrates that SPK can favor new tissue ingrowth and integration, prevent peri-scaffold cartilage degeneration and patellar cartilage degeneration, inhibit inflammatory cytokine secretion, and promote cartilage function restoration. Conclusion The present study confirmed that the 3D printed porous sulfonated PEEK scaffold could promote cartilage functional repair, and suggests a new promising strategy for treating cartilage defects with a functional prosthesis that spontaneously inhibits nearby cartilage degeneration. Translational potential of this article In the present study, we propose a new cartilage repair strategy based on a porous, non-biodegradable polyetheretherketone (PEEK) scaffold, which may bring up a new treatment route for elderly patients with cartilage lesions in the future.
Collapse
Affiliation(s)
- Zhiguo Yuan
- Department of Bone and Joint Surgery, Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Teng Long
- Department of Bone and Joint Surgery, Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jue Zhang
- Department of Bone and Joint Surgery, Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhuocheng Lyu
- Department of Bone and Joint Surgery, Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei Zhang
- Department of Bone and Joint Surgery, Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiangchao Meng
- Department of Bone and Joint Surgery, Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jin Qi
- Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - You Wang
- Department of Bone and Joint Surgery, Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- Corresponding author.
| |
Collapse
|
26
|
Tsutsumi H, Kurimoto R, Nakamichi R, Chiba T, Matsushima T, Fujii Y, Sanada R, Kato T, Shishido K, Sakamaki Y, Kimura T, Kishida A, Asahara H. Generation of a tendon-like tissue from human iPS cells. J Tissue Eng 2022; 13:20417314221074018. [PMID: 35083031 PMCID: PMC8785341 DOI: 10.1177/20417314221074018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/01/2022] [Indexed: 12/29/2022] Open
Abstract
Tendons and ligaments are essential connective tissues that connect the muscle and bone. Their recovery from injuries is known to be poor, highlighting the crucial need for an effective therapy. A few reports have described the development of artificial ligaments with sufficient strength from human cells. In this study, we successfully generated a tendon-like tissue (bio-tendon) using human induced pluripotent stem cells (iPSCs). We first differentiated human iPSCs into mesenchymal stem cells (iPSC-MSCs) and transfected them with Mohawk (Mkx) to obtain Mkx-iPSC-MSCs, which were applied to a newly designed chamber with a mechanical stretch incubation system. The embedded Mkx-iPSC-MSCs created bio-tendons and exhibited an aligned extracellular matrix structure. Transplantation of the bio-tendons into a mouse Achilles tendon rupture model showed host-derived cell infiltration with improved histological score and biomechanical properties. Taken together, the bio-tendon generated in this study has potential clinical applications for tendon/ligament-related injuries and diseases.
Collapse
Affiliation(s)
- Hiroki Tsutsumi
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Ryota Kurimoto
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Ryo Nakamichi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Tomoki Chiba
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Takahide Matsushima
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Yuta Fujii
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Risa Sanada
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Tomomi Kato
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Kana Shishido
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Yuriko Sakamaki
- Research Core, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Tsuyoshi Kimura
- Department of Material-Based Medical Engineering, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Akio Kishida
- Department of Material-Based Medical Engineering, Tokyo Medical and Dental University, Bunkyo City, Japan
| | - Hiroshi Asahara
- Department of Systems BioMedicine, Tokyo Medical and Dental University, Bunkyo City, Japan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
27
|
Cooper SM, Rainbow RS. The Developing Field of Scaffold-Free Tissue Engineering for Articular Cartilage Repair. TISSUE ENGINEERING. PART B, REVIEWS 2021; 28:995-1006. [PMID: 34605669 DOI: 10.1089/ten.teb.2021.0130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Articular cartilage is critical for proper joint mobility as it provides a smooth and lubricated surface between articulating bones and allows for transmission of load to underlying bones. Extended wear or injury of this tissue can result in osteoarthritis, a degenerative disease affecting millions across the globe. Because of its low regenerative capacity, articular cartilage cannot heal on its own and effective treatments for injured joint restoration remain a challenge. Strategies in tissue engineering have been demonstrated as potential therapeutic approaches to regenerate and repair damaged articular cartilage. Although many of these strategies rely on the use of an exogenous three-dimensional scaffolds to regenerate cartilage, scaffold-free tissue engineering provides numerous advantages over scaffold-based methods. This review highlights the latest advancements in scaffold-free tissue engineering for cartilage and the potential for clinical translation.
Collapse
Affiliation(s)
- Sarah M Cooper
- Department of Mechanical and Materials Engineering, Queen's University, Kingston, Canada
| | - Roshni S Rainbow
- Department of Mechanical and Materials Engineering, Queen's University, Kingston, Canada
| |
Collapse
|
28
|
Link JM, Hu JC, Athanasiou KA. Chondroitinase ABC Enhances Integration of Self-Assembled Articular Cartilage, but Its Dosage Needs to Be Moderated Based on Neocartilage Maturity. Cartilage 2021; 13:672S-683S. [PMID: 32441107 PMCID: PMC8804832 DOI: 10.1177/1947603520918653] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE To enhance the in vitro integration of self-assembled articular cartilage to native articular cartilage using chondroitinase ABC. DESIGN To examine the hypothesis that chondroitinase ABC (C-ABC) integration treatment (C-ABCint) would enhance integration of neocartilage of different maturity levels, this study was conducted in 2 phases. In phase I, the impact on integration of 2 treatments, TCL (TGF-β1, C-ABC, and lysyl oxidase like 2) and C-ABCint, was examined via a 2-factor, full factorial design. In phase II, construct maturity (2 levels) and C-ABCint concentration (3 levels) were the factors in a full factorial design to determine whether the effective C-ABCint dose was dependent on neocartilage maturity level. Neocartilages formed or treated per the factors above were placed into native cartilage rings, cultured for 2 weeks, and, then, integration was studied histologically and mechanically. Prior to integration, in phase II, a set of treated constructs were also assayed to provide a baseline of properties. RESULTS In phase I, C-ABCint and TCL treatments synergistically enhanced interface Young's modulus by 6.2-fold (P = 0.004) and increased interface tensile strength by 3.8-fold (P = 0.02) compared with control. In phase II, the interaction of the factors C-ABCint and construct maturity was significant (P = 0.0004), indicating that the effective C-ABCint dose to improve interface Young's modulus is dependent on construct maturity. Construct mechanical properties were preserved regardless of C-ABCint dose. CONCLUSIONS Applying C-ABCint to neocartilage is an effective integration strategy with translational potential, provided its dose is calibrated appropriately based on implant maturity, that also preserves implant biomechanical properties.
Collapse
Affiliation(s)
- Jarrett M. Link
- Department of Biomedical Engineering,
University of California, Irvine, CA, USA
| | - Jerry C. Hu
- Department of Biomedical Engineering,
University of California, Irvine, CA, USA
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering,
University of California, Irvine, CA, USA,Kyriacos A. Athanasiou, Distinguished
Professor Henry Samueli Chair, Director, DELTAi (Driving
Engineering and Life-science Translational Advances @ Irvine), Department of
Biomedical Engineering, Henry Samueli School of Engineering, University of
California, 3418 Engineering Hall, Irvine, CA 92697, USA.
| |
Collapse
|
29
|
Leighton MP, Rutenberg AD, Kreplak L. D-band strain underestimates fibril strain for twisted collagen fibrils at low strains. J Mech Behav Biomed Mater 2021; 124:104854. [PMID: 34601435 DOI: 10.1016/j.jmbbm.2021.104854] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/01/2021] [Accepted: 09/19/2021] [Indexed: 11/29/2022]
Abstract
Collagen fibrils are the main structural component of load-bearing tissues such as tendons, ligaments, skin, the cornea of the eye, and the heart. The D-band of collagen fibrils is an axial periodic density modulation that can be easily characterized by tissue-level X-ray scattering. During mechanical testing, D-band strain is often used as a proxy for fibril strain. However, this approach ignores the coupling between strain and molecular tilt. We examine the validity of this approximation using an elastomeric collagen fibril model that includes both the D-band and a molecular tilt field. In the low strain regime, we show that the D-band strain substantially underestimates fibril strain for strongly twisted collagen fibrils - such as fibrils from skin or corneal tissue.
Collapse
Affiliation(s)
- Matthew P Leighton
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, B3H 4R2, Nova Scotia, Canada; Department of Physics, Simon Fraser University, Burnaby, V5A 1S6, British Columbia, Canada
| | - Andrew D Rutenberg
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, B3H 4R2, Nova Scotia, Canada.
| | - Laurent Kreplak
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, B3H 4R2, Nova Scotia, Canada
| |
Collapse
|
30
|
Kim H, Shin YM, Chung S, Kim D, Park DB, Baek S, Park J, Kim SY, Kim D, Yi SW, Lee S, Lee JB, Ko J, Im G, Kang M, Sung H. Cell-Membrane-Derived Nanoparticles with Notch-1 Suppressor Delivery Promote Hypoxic Cell-Cell Packing and Inhibit Angiogenesis Acting as a Two-Edged Sword. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2101558. [PMID: 34431568 PMCID: PMC11468545 DOI: 10.1002/adma.202101558] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/02/2021] [Indexed: 06/13/2023]
Abstract
Cell-cell interactions regulate intracellular signaling via reciprocal contacts of cell membranes in tissue regeneration and cancer growth, indicating a critical need of membrane-derived tools in studying these processes. Hence, cell-membrane-derived nanoparticles (CMNPs) are produced using tonsil-derived mesenchymal stem cells (TMSCs) from children owing to their short doubling time. As target cell types, laryngeal cancer cells are compared to bone-marrow-derived MSCs (BMSCs) because of their cartilage damaging and chondrogenic characteristics, respectively. Treating spheroids of these cell types with CMNPs exacerbates interspheroid hypoxia with robust maintenance of the cell-cell interaction signature for 7 days. Both cell types prefer a hypoxic environment, as opposed to blood vessel formation that is absent in cartilage but is required for cancer growth. Hence, angiogenesis is inhibited by displaying the Notch-1 aptamer on CMNPs. Consequently, laryngeal cancer growth is suppressed efficiently in contrast to improved chondroprotection observed in a series of cell and animal experiments using a xenograft mouse model of laryngeal cancer. Altogether, CMNPs execute a two-edged sword function of inducing hypoxic cell-cell packing, followed by suppressing angiogenesis to promote laryngeal cancer death and chondrogenesis simultaneously. This study presents a previously unexplored therapeutic strategy for anti-cancer and chondroprotective treatment using CMNPs.
Collapse
Affiliation(s)
- Hye‐Seon Kim
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Young Min Shin
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Seyong Chung
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Dahee Kim
- Department of OtorhinolaryngologyYonsei University College of MedicineSeoul03722Republic of Korea
| | - Dan Bi Park
- TMD LAB Co., Ltd6th Floor, 31, Gwangnaru‐ro 8‐gilSeongdong‐guSeoul04799Republic of Korea
| | - Sewoom Baek
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Jeongeun Park
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Si Yeong Kim
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Dae‐Hyun Kim
- Department of Veterinary SurgeryCollege of Veterinary MedicineChungnam National University99, Daehak‐roYuseong‐guDaejeon34134Republic of Korea
| | - Se Won Yi
- TMD LAB Co., Ltd6th Floor, 31, Gwangnaru‐ro 8‐gilSeongdong‐guSeoul04799Republic of Korea
| | - Songhyun Lee
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Jung Bok Lee
- Department of Biological ScienceSookmyung Women's UniversityCheongpa‐ro 47‐gil 100, Yongsan‐guSeoul04310Republic of Korea
| | - Ji‐Yun Ko
- Department of Veterinary SurgeryCollege of Veterinary MedicineChungnam National University99, Daehak‐roYuseong‐guDaejeon34134Republic of Korea
- Research Institute of Convergence Life ScienceDongguk UniversityGoyang10326Republic of Korea
| | - Gun‐Il Im
- Department of OrthopedicsDongguk University Ilsan HospitalGoyang10326Republic of Korea
| | - Mi‐Lan Kang
- TMD LAB Co., Ltd6th Floor, 31, Gwangnaru‐ro 8‐gilSeongdong‐guSeoul04799Republic of Korea
| | - Hak‐Joon Sung
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| |
Collapse
|
31
|
Peterson BE, Rolfe RA, Kunselman A, Murphy P, Szczesny SE. Mechanical Stimulation via Muscle Activity Is Necessary for the Maturation of Tendon Multiscale Mechanics During Embryonic Development. Front Cell Dev Biol 2021; 9:725563. [PMID: 34540841 PMCID: PMC8446456 DOI: 10.3389/fcell.2021.725563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/16/2021] [Indexed: 11/17/2022] Open
Abstract
During embryonic development, tendons transform into a hypocellular tissue with robust tensile load-bearing capabilities. Previous work suggests that this mechanical transformation is due to increases in collagen fibril length and is dependent on mechanical stimulation via muscle activity. However, the relationship between changes in the microscale tissue structure and changes in macroscale tendon mechanics is still unclear. Additionally, the specific effect of mechanical stimulation on the multiscale structure-function relationships of developing tendons is also unknown. Therefore, the objective of this study was to measure the changes in tendon mechanics and structure at multiple length scales during embryonic development with and without skeletal muscle paralysis. Tensile testing of tendons from chick embryos was performed to determine the macroscale tensile modulus as well as the magnitude of the fibril strains and interfibrillar sliding with applied tissue strain. Embryos were also treated with either decamethonium bromide or pancuronium bromide to produce rigid or flaccid paralysis. Histology was performed to assess changes in tendon size, spacing between tendon subunits, and collagen fiber diameter. We found that the increase in the macroscale modulus observed with development is accompanied by an increase in the fibril:tissue strain ratio, which is consistent with an increase in collagen fibril length. Additionally, we found that flaccid paralysis reduced the macroscale tendon modulus and the fibril:tissue strain ratio, whereas less pronounced effects that were not statistically significant were observed with rigid paralysis. Finally, skeletal paralysis also reduced the size of collagen fibril bundles (i.e., fibers). Together, these data suggest that more of the applied tissue strain is transmitted to the collagen fibrils at later embryonic ages, which leads to an increase in the tendon macroscale tensile mechanics. Furthermore, our data suggest that mechanical stimulation during development is necessary to induce structural and mechanical changes at multiple physical length scales. This information provides valuable insight into the multiscale structure-function relationships of developing tendons and the importance of mechanical stimulation in producing a robust tensile load-bearing soft tissue.
Collapse
Affiliation(s)
- Benjamin E Peterson
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, United States
| | - Rebecca A Rolfe
- Department of Zoology, School of Natural Sciences, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Allen Kunselman
- Department of Public Health Science, Division of Biostatistics and Bioinformatics, Pennsylvania State University, Hershey, PA, United States
| | - Paula Murphy
- Department of Zoology, School of Natural Sciences, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Spencer E Szczesny
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, United States.,Department of Orthopaedics and Rehabilitation, Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
32
|
Choi YR, Collins KH, Springer LE, Pferdehirt L, Ross AK, Wu CL, Moutos FT, Harasymowicz NS, Brunger JM, Pham CTN, Guilak F. A genome-engineered bioartificial implant for autoregulated anticytokine drug delivery. SCIENCE ADVANCES 2021; 7:eabj1414. [PMID: 34516920 PMCID: PMC8442875 DOI: 10.1126/sciadv.abj1414] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/19/2021] [Indexed: 05/28/2023]
Abstract
Biologic drug therapies are increasingly used for inflammatory diseases such as rheumatoid arthritis but may cause significant adverse effects when delivered continuously at high doses. We used CRISPR-Cas9 genome editing of iPSCs to create a synthetic gene circuit that senses changing levels of endogenous inflammatory cytokines to trigger a proportional therapeutic response. Cells were engineered into cartilaginous constructs that showed rapid activation and recovery in response to inflammation in vitro or in vivo. In the murine K/BxN model of inflammatory arthritis, bioengineered implants significantly mitigated disease severity as measured by joint pain, structural damage, and systemic and local inflammation. Therapeutic implants completely prevented increased pain sensitivity and bone erosions, a feat not achievable by current clinically available disease-modifying drugs. Combination tissue engineering and synthetic biology promises a range of potential applications for treating chronic diseases via custom-designed cells that express therapeutic transgenes in response to dynamically changing biological signals.
Collapse
Affiliation(s)
- Yun-Rak Choi
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Luke E. Springer
- Division of Rheumatology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Lara Pferdehirt
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Alison K. Ross
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | | | - Natalia S. Harasymowicz
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jonathan M. Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Christine T. N. Pham
- Division of Rheumatology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA
- Shriners Hospitals for Children, St. Louis, MO 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
- Cytex Therapeutics Inc., Durham, NC 27704, USA
| |
Collapse
|
33
|
Engineering large, anatomically shaped osteochondral constructs with robust interfacial shear properties. NPJ Regen Med 2021; 6:42. [PMID: 34362933 PMCID: PMC8346478 DOI: 10.1038/s41536-021-00152-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/07/2021] [Indexed: 11/10/2022] Open
Abstract
Despite the prevalence of large (>5 cm2) articular cartilage defects involving underlying bone, current tissue-engineered therapies only address small defects. Tissue-engineered, anatomically shaped, native-like implants may address the need for off-the-shelf, tissue-repairing therapies for large cartilage lesions. This study fabricated an osteochondral construct of translationally relevant geometry with robust functional properties. Scaffold-free, self-assembled neocartilage served as the chondral phase, and porous hydroxyapatite served as the osseous phase of the osteochondral constructs. Constructs in the shape and size of an ovine femoral condyle (31 × 14 mm) were assembled at day 4 (early) or day 10 (late) of neocartilage maturation. Early osteochondral assembly increased the interfacial interdigitation depth by 244%, interdigitation frequency by 438%, interfacial shear modulus by 243-fold, and ultimate interfacial shear strength by 4.9-fold, compared to late assembly. Toward the development of a bioprosthesis for the repair of cartilage lesions encompassing up to an entire condylar surface, this study generated a large, anatomically shaped osteochondral construct with robust interfacial mechanical properties and native-like neocartilage interdigitation.
Collapse
|
34
|
Rahman S, Szojka ARA, Liang Y, Kunze M, Goncalves V, Mulet-Sierra A, Jomha NM, Adesida AB. Inability of Low Oxygen Tension to Induce Chondrogenesis in Human Infrapatellar Fat Pad Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:703038. [PMID: 34381784 PMCID: PMC8350173 DOI: 10.3389/fcell.2021.703038] [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: 04/30/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Articular cartilage of the knee joint is avascular, exists under a low oxygen tension microenvironment, and does not self-heal when injured. Human infrapatellar fat pad-sourced mesenchymal stem cells (IFP-MSC) are an arthroscopically accessible source of mesenchymal stem cells (MSC) for the repair of articular cartilage defects. Human IFP-MSC exists physiologically under a low oxygen tension (i.e., 1-5%) microenvironment. Human bone marrow mesenchymal stem cells (BM-MSC) exist physiologically within a similar range of oxygen tension. A low oxygen tension of 2% spontaneously induced chondrogenesis in micromass pellets of human BM-MSC. However, this is yet to be demonstrated in human IFP-MSC or other adipose tissue-sourced MSC. In this study, we explored the potential of low oxygen tension at 2% to drive the in vitro chondrogenesis of IFP-MSC. We hypothesized that 2% O2 will induce stable chondrogenesis in human IFP-MSC without the risk of undergoing endochondral ossification at ectopic sites of implantation. METHODS Micromass pellets of human IFP-MSC were cultured under 2% O2 or 21% O2 (normal atmosphere O2) in the presence or absence of chondrogenic medium with transforming growth factor-β3 (TGFβ3) for 3 weeks. Following in vitro chondrogenesis, the resulting pellets were implanted in immunodeficient athymic nude mice for 3 weeks. RESULTS A low oxygen tension of 2% was unable to induce chondrogenesis in human IFP-MSC. In contrast, chondrogenic medium with TGFβ3 induced in vitro chondrogenesis. All pellets were devoid of any evidence of undergoing endochondral ossification after subcutaneous implantation in athymic mice.
Collapse
Affiliation(s)
- Samia Rahman
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Alexander R. A. Szojka
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Yan Liang
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Victoria Goncalves
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Nadr M. Jomha
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Adetola B. Adesida
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of Alberta Hospital, Edmonton, AB, Canada
| |
Collapse
|
35
|
Atelocollagen-Embedded Chondrocyte Precursors as a Treatment for Grade-4 Cartilage Defects of the Femoral Condyle: A Case Series with up to 9-Year Follow-Up. Biomolecules 2021; 11:biom11070942. [PMID: 34202002 PMCID: PMC8301975 DOI: 10.3390/biom11070942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 12/22/2022] Open
Abstract
We demonstrated the safety and efficacy of autologous chondrocyte precursor (CP) cell therapy in repairing Grade 4 cartilage defects of medial femoral condyles. The autologous bone marrow mesenchymal stem cells of each participant were isolated, amplified, and then differentiated into CPs in atelocollagen. Neotissues made of CPs were implanted into cartilage defects with an average cell density of 4.9 ± 2.1 × 106 cells/cm2 through arthrotomy. The knee function was evaluated with the International Knee Documentation Committee (IKDC) subjective knee form. Patients' knee functions significantly improved by the 28th week (IKDC score = 68.3 ± 12.1), relative to the initial functionality before the CP therapy (IKDC score = 46.1 ± 16.4, p-value = 0.0014). Nine of these twelve patients maintained good knee functions for 9 years post-implantation (IKDC score = 69.8 ± 12.3) at levels higher than the pre-implantation values (p-value = 0.0018). Patients were evaluated with MRI and arthroscopy, and the defective sites exhibited a smooth surface without a gap between the implant and host tissue. This study demonstrates that autologous CPs successfully engraft into the host tissue and result in the re-formation of hyaline-like cartilage, thereby improving the impaired knee functions. Most importantly, no adverse event was reported during this long-term follow-up period.
Collapse
|
36
|
Wang D, Zhang X, Huang S, Liu Y, Fu BSC, Mak KKL, Blocki AM, Yung PSH, Tuan RS, Ker DFE. Engineering multi-tissue units for regenerative Medicine: Bone-tendon-muscle units of the rotator cuff. Biomaterials 2021; 272:120789. [PMID: 33845368 DOI: 10.1016/j.biomaterials.2021.120789] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022]
Abstract
Our body systems are comprised of numerous multi-tissue units. For the musculoskeletal system, one of the predominant functional units is comprised of bone, tendon/ligament, and muscle tissues working in tandem to facilitate locomotion. To successfully treat musculoskeletal injuries and diseases, critical consideration and thoughtful integration of clinical, biological, and engineering aspects are necessary to achieve translational bench-to-bedside research. In particular, identifying ideal biomaterial design specifications, understanding prior and recent tissue engineering advances, and judicious application of biomaterial and fabrication technologies will be crucial for addressing current clinical challenges in engineering multi-tissue units. Using rotator cuff tears as an example, insights relevant for engineering a bone-tendon-muscle multi-tissue unit are presented. This review highlights the tissue engineering strategies for musculoskeletal repair and regeneration with implications for other bone-tendon-muscle units, their derivatives, and analogous non-musculoskeletal tissue structures.
Collapse
Affiliation(s)
- Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Shuting Huang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Yang Liu
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Bruma Sai-Chuen Fu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | | | - Anna Maria Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Patrick Shu-Hang Yung
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
37
|
Leighton MP, Kreplak L, Rutenberg AD. Non-equilibrium growth and twist of cross-linked collagen fibrils. SOFT MATTER 2021; 17:1415-1427. [PMID: 33325971 DOI: 10.1039/d0sm01830a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The lysyl oxidase (LOX) enzyme that catalyses cross-link formation during the assembly of collagen fibrils in vivo is too large to diffuse within assembled fibrils, and so is incompatible with a fully equilibrium mechanism for fibril formation. We propose that enzymatic cross-links are formed at the fibril surface during the growth of collagen fibrils; as a consequence no significant reorientation of previously cross-linked collagen molecules occurs inside collagen fibrils during fibril growth in vivo. By imposing local equilibrium only at the fibril surface, we develop a coarse-grained quantitative model of in vivo fibril structure that incorporates a double-twist orientation of collagen molecules and a periodic D-band density modulation along the fibril axis. Radial growth is controlled by the concentration of available collagen molecules outside the fibril. In contrast with earlier equilibrium models of fibril structure, we find that all fibrils can exhibit a core-shell structure that is controlled only by the fibril radius. At small radii a core is developed with a linear double-twist structure as a function of radius. Within the core the double-twist structure is largely independent of the D-band. Within the shell at larger radii, the structure approaches a constant twist configuration that is strongly coupled with the D-band. We suggest a stable radius control mechanism that corneal fibrils can exploit near the edge of the linear core regime; while larger tendon fibrils use a cruder version of growth control that does not select a preferred radius.
Collapse
Affiliation(s)
- Matthew P Leighton
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| | - Laurent Kreplak
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| | - Andrew D Rutenberg
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| |
Collapse
|
38
|
Puetzer JL, Ma T, Sallent I, Gelmi A, Stevens MM. Driving Hierarchical Collagen Fiber Formation for Functional Tendon, Ligament, and Meniscus Replacement. Biomaterials 2021; 269:120527. [PMID: 33246739 PMCID: PMC7883218 DOI: 10.1016/j.biomaterials.2020.120527] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/09/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022]
Abstract
Hierarchical collagen fibers are the primary source of strength in musculoskeletal tendons, ligaments, and menisci. It has remained a challenge to develop these large fibers in engineered replacements or in vivo after injury. The objective of this study was to investigate the ability of restrained cell-seeded high density collagen gels to drive hierarchical fiber formation for multiple musculoskeletal tissues. We found boundary conditions applied to high density collagen gels were capable of driving tenocytes, ligament fibroblasts, and meniscal fibrochondrocytes to develop native-sized hierarchical collagen fibers 20-40 μm in diameter. The fibers organize similar to bovine juvenile collagen with native fibril banding patterns and hierarchical fiber bundles 50-350 μm in diameter by 6 weeks. Mirroring fiber organization, tensile properties of restrained samples improved significantly with time, reaching ~1 MPa. Additionally, tendon, ligament, and meniscal cells produced significantly different sized fibers, different degrees of crimp, and different GAG concentrations, which corresponded with respective juvenile tissue. To our knowledge, these are some of the largest, most organized fibers produced to date in vitro. Further, cells produced tissue specific hierarchical fibers, suggesting this system is a promising tool to better understand cellular regulation of fiber formation to better stimulate it in vivo after injury.
Collapse
Affiliation(s)
- Jennifer L Puetzer
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ; Department of Biomedical Engineering and Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States, 23284.
| | - Tianchi Ma
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Ignacio Sallent
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Amy Gelmi
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, And Institute for Biomedical Engineering, Imperial College London, London, United Kingdom, SW7 2AZ.
| |
Collapse
|
39
|
Sorushanova A, Skoufos I, Tzora A, Mullen AM, Zeugolis DI. The influence of animal species, gender and tissue on the structural, biophysical, biochemical and biological properties of collagen sponges. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:12. [PMID: 33475864 PMCID: PMC7819930 DOI: 10.1007/s10856-020-06485-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/18/2020] [Indexed: 05/03/2023]
Abstract
Although collagen type I is extensively used in biomedicine, no study to-date has assessed how the properties of the produced scaffolds are affected as a function of species, gender and tissue from which the collagen was extracted. Herein, we extracted and characterised collagen from porcine and bovine, male and female and skin and tendon tissues and we subsequently fabricated and assessed the structural, biophysical, biochemical and biological properties of collagen sponges. All collagen preparations were of similar purity and free-amine content (p > 0.05). In general, the porcine groups yielded more collagen; had higher (p < 0.05) denaturation temperature and resistance to enzymatic degradation; and lower (p < 0.05) swelling ratio and compression stress and modulus than the bovine groups of the same gender and tissue. All collagen preparations supported growth of human dermal fibroblasts and exhibited similar biological response to human THP-1 monocytes. These results further illustrate the need for standardisation of collagen preparations for the development of reproducible collagen-based devices. Assessment of the physicochemical and biological properties of collagen sponges as a function of animal species (bovine versus porcine), gender (male versus female) and tissue (skin versus tendon).
Collapse
Affiliation(s)
- Anna Sorushanova
- 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
| | - Ioannis Skoufos
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, Arta, Greece
| | - Athina Tzora
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, Arta, Greece
| | | | - Dimitrios 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), Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland.
| |
Collapse
|
40
|
Kwon H, Brown WE, O'Leary SA, Hu JC, Athanasiou KA. Rejuvenation of extensively passaged human chondrocytes to engineer functional articular cartilage. Biofabrication 2021; 13. [PMID: 33418542 DOI: 10.1088/1758-5090/abd9d9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/08/2021] [Indexed: 11/11/2022]
Abstract
Human articular chondrocytes (hACs) are scarce and lose their chondrogenic potential during monolayer passaging, impeding their therapeutic use. This study investigated i) the translatability of conservative chondrogenic passaging and aggregate rejuvenation on restoring chondrogenic properties of hACs passaged up to P9; and ii) the efficacy of a combined treatment of TGF-β1 (T), chondroitinase-ABC (C), and lysyl oxidase-like 2 (L), collectively termed TCL, on engineering functional human neocartilage via the self-assembling process, as a function of passage number up to P11. Here, we show that aggregate rejuvenation enhanced glycosaminoglycan (GAG) content and type II collagen staining at all passages and yielded human neocartilage with chondrogenic phenotype present up to P7. Addition of TCL extended the chondrogenic phenotype to P11 and significantly enhanced GAG content and type II collagen staining at all passages. Human neocartilage derived from high passages, treated with TCL, displayed mechanical properties that were on par with or greater than those derived from low passages. Conservative chondrogenic passaging and aggregate rejuvenation may be a viable new strategy 1) to address the perennial problem of chondrocyte scarcity and 2) to successfully rejuvenate the chondrogenic phenotype of extensively passaged cells (up to P11). Furthermore, tissue engineering human neocartilage via self-assembly in conjunction with TCL treatment advances the clinical use of extensively passaged human chondrocytes for cartilage repair.
Collapse
Affiliation(s)
- Heenam Kwon
- Biomedical Engineering, University of California Irvine, University of California Irvine, Irvine, California, 92697, UNITED STATES
| | - Wendy E Brown
- Biomedical Engineering, University of California Irvine, University of California Irvine, Irvine, California, 92697, UNITED STATES
| | - Siobhan A O'Leary
- Align Technology Inc, 2820 Orchard Pkwy, San Jose, California, 95134, UNITED STATES
| | - Jerry C Hu
- University of California Irvine, University of California Irvine, Irvine, California, 92697, UNITED STATES
| | - Kyriacos A Athanasiou
- Biomedical Engineering, University of California Irvine, University of California Irvine, Irvine, California, 92697, UNITED STATES
| |
Collapse
|
41
|
Bioengineering and Enabling Technologies: ABME Special Issue Editorial. Ann Biomed Eng 2020; 48:1445-1450. [PMID: 32232693 DOI: 10.1007/s10439-020-02485-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
42
|
Boyle ST, Johan MZ, Samuel MS. Tumour-directed microenvironment remodelling at a glance. J Cell Sci 2020; 133:133/24/jcs247783. [PMID: 33443095 DOI: 10.1242/jcs.247783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The tissue microenvironment supports normal tissue function and regulates the behaviour of parenchymal cells. Tumour cell behaviour, on the other hand, diverges significantly from that of their normal counterparts, rendering the microenvironment hostile to tumour cells. To overcome this problem, tumours can co-opt and remodel the microenvironment to facilitate their growth and spread. This involves modifying both the biochemistry and the biophysics of the normal microenvironment to produce a tumour microenvironment. In this Cell Science at a Glance article and accompanying poster, we outline the key processes by which epithelial tumours influence the establishment of the tumour microenvironment. As the microenvironment is populated by genetically normal cells, we discuss how controlling the microenvironment is both a significant challenge and a key vulnerability for tumours. Finally, we review how new insights into tumour-microenvironment interactions has led to the current consensus on how these processes may be targeted as novel anti-cancer therapies.
Collapse
Affiliation(s)
- Sarah T Boyle
- Centre for Cancer Biology, An Alliance between SA Pathology and the University of South Australia, Adelaide, SA 5001, Australia
| | - M Zahied Johan
- Centre for Cancer Biology, An Alliance between SA Pathology and the University of South Australia, Adelaide, SA 5001, Australia
| | - Michael S Samuel
- Centre for Cancer Biology, An Alliance between SA Pathology and the University of South Australia, Adelaide, SA 5001, Australia .,Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
43
|
The Effects of Hypoxia-Reoxygenation in Mouse Digital Flexor Tendon-Derived Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7305392. [PMID: 33456674 PMCID: PMC7787768 DOI: 10.1155/2020/7305392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/12/2020] [Accepted: 11/25/2020] [Indexed: 11/24/2022]
Abstract
Objective Ischemia-reperfusion injury refers to the exacerbated and irreversible tissue damage caused by blood flow restoration after a period of ischemia. The hypoxia-reoxygenation (H/R) model in vitro is ideal for studying ischemia-reperfusion injury at the cellular level. We employed this model and investigated the effects of cobalt chloride- (CoCl2-) induced H/R in cells derived from mouse digital flexor tendons. Materials and Methods Various H/R conditions were simulated via treatment of tendon-derived cells with different concentrations of CoCl2 for 24 h, followed by removal of CoCl2 to restore a normal oxygen state for up to 96 h. Cell viability was measured using the Cell Counting Kit-8 (CCK-8) assay. Cell growth was determined via observation of cell morphology and proliferation. Oxidative stress markers and mitochondrial activity were detected. The expression levels of hypoxia-inducible factor- (HIF-) 1α, vascular endothelial growth factor-A (VEGF-A), collagen I, and collagen III were determined using Western blot (WB), real-time PCR, and immunofluorescence staining. Cellular apoptosis was analyzed via flow cytometry, and the expression of apoptosis-related proteins Bax and bcl-2 was examined using WB. Results The cells treated with low concentrations of CoCl2 showed significantly increased cell viability after reoxygenation. The increase in cell viability was even more pronounced in cells that had been treated with high concentrations of CoCl2. Under H/R conditions, cell morphology and growth were unchanged, while oxidative stress reaction was induced and mitochondrial activity was increased. H/R exerted opposite effects on the expression of HIF-1α mRNA and protein. Meanwhile, the expression of VEGF-A was upregulated, whereas collagen type I and type III were significantly downregulated. The level of cellular apoptosis did not show significant changes during H/R, despite the significantly increased Bax protein and reduced bcl-2 protein levels that led to an increase in the Bax/bcl-2 ratio during reoxygenation. Conclusions Tendon-derived cells were highly tolerant to the hypoxic environments induced by CoCl2. Reoxygenation after hypoxia preconditioning promoted cell viability, especially in cells treated with high concentrations of CoCl2. H/R conditions caused oxidative stress responses but did not affect cell growth. The H/R process had a notable impact on collagen production and expression of apoptosis-related proteins by tendon-derived cells, while the level of cellular apoptosis remained unchanged.
Collapse
|
44
|
LOXL2 promotes aggrecan and gender-specific anabolic differences to TMJ cartilage. Sci Rep 2020; 10:20179. [PMID: 33214607 PMCID: PMC7678826 DOI: 10.1038/s41598-020-77178-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 11/05/2020] [Indexed: 12/24/2022] Open
Abstract
In the United States, 5–12% of adults have at least one symptom of temporomandibular joint (TMJ) disorders, including TMJ osteoarthritis (TMJ-OA). However, there is no chondroprotective agent that is approved for clinical application. We showed that LOXL2 is elevated in the regenerative response during fracture healing in mice and has a critical role in chondrogenic differentiation. Indeed, LOXL2 is an anabolic effector that attenuates pro-inflammatory signaling in OA cartilage of the TMJ and knee joint, induces chondroprotective and regenerative responses, and attenuates NF-kB signaling. The specific goal of the study was to evaluate if adenoviral delivery of LOXL2 is anabolic to human and mouse TMJ condylar cartilage in vivo and evaluate the protective and anabolic effect on cartilage-specific factors. We employed two different models to assess TMJ-OA. In one model, clinical TMJ-OA cartilage from 5 different samples in TMJ-OA cartilage plugs were implanted subcutaneously in nude mice. Adenovirus LOXL2 -treated implants showed higher mRNA levels of LOXL2, ACAN, and other anabolic genes compared to the adenovirus-Empty-treated implants. Further characterization by RNA-seq analysis showed LOXL2 promotes proteoglycan networks and extracellular matrix in human TMJ-OA cartilage implants in vivo. In order to evaluate if LOXL2-induced functional and sex-linked differences, both male and female four-month-old chondrodysplasia (Cho/+) mice, which develop progressive TMJ-OA due to a point mutation in the Col11a1 gene, were subjected to intraperitoneal injection with Adv-RFP-LOXL2 every 2 weeks for 12 weeks. The data showed that adenovirus delivery of LOXL2 upregulated LOXL2 and aggrecan (Acan), whereas MMP13 expression was slightly downregulated. The fold change expression of Acan and Runx2 induced by Adv-RFP-LOXL2 was higher in females compared to males. Interestingly, Adv-RFP-LOXL2 injection significantly increased Rankl expression in male but there was no change in females, whereas VegfB gene expression was increased in females, but not in males, as compared to those injected with Adv-RFP-Empty in respective groups. Our findings indicate that LOXL2 can induce specifically the expression of Acan and other anabolic genes in two preclinical models in vivo. Further, LOXL2 has beneficial functions in human TMJ-OA cartilage implants and promotes gender-specific anabolic responses in Cho/+ mice with progressive TMJ-OA, suggesting its merit for further study as an anabolic therapy for TMJ-OA.
Collapse
|
45
|
Bielajew BJ, Hu JC, Athanasiou KA. Collagen: quantification, biomechanics, and role of minor subtypes in cartilage. NATURE REVIEWS. MATERIALS 2020; 5:730-747. [PMID: 33996147 PMCID: PMC8114887 DOI: 10.1038/s41578-020-0213-1] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/28/2020] [Indexed: 05/02/2023]
Abstract
Collagen is a ubiquitous biomaterial in vertebrate animals. Although each of its 28 subtypes contributes to the functions of many different tissues in the body, most studies on collagen or collagenous tissues have focussed on only one or two subtypes. With recent developments in analytical chemistry, especially mass spectrometry, significant advances have been made toward quantifying the different collagen subtypes in various tissues; however, high-throughput and low-cost methods for collagen subtype quantification do not yet exist. In this Review, we introduce the roles of collagen subtypes and crosslinks, and describe modern assays that enable a deep understanding of tissue physiology and disease states. Using cartilage as a model tissue, we describe the roles of major and minor collagen subtypes in detail; discuss known and unknown structure-function relationships; and show how tissue engineers may harness the functional characteristics of collagen to engineer robust neotissues.
Collapse
Affiliation(s)
- Benjamin J. Bielajew
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Jerry C. Hu
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| |
Collapse
|
46
|
Wang C, Sha Y, Wang S, Chi Q, Sung KP, Xu K, Yang L. Lysyl oxidase suppresses the inflammatory response in anterior cruciate ligament fibroblasts and promotes tissue regeneration by targeting myotrophin via the nuclear factor‐kappa B pathway. J Tissue Eng Regen Med 2020; 14:1063-1076. [DOI: 10.1002/term.3077] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Chunli Wang
- National Innovation and Attracting Talents “111” base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing University Chongqing China
| | - Yongqiang Sha
- Center for Precision Medicine, School of Medicine and School of Biomedical SciencesHuaqiao University Xiamen China
| | - Sixiang Wang
- National Innovation and Attracting Talents “111” base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing University Chongqing China
| | - Qingjia Chi
- Department of Mechanics and Engineering Structure, Hubei Key Laboratory of Theory and Application of Advanced Materials MechanicsWuhan University of Technology Wuhan China
| | - K.L. Paul Sung
- National Innovation and Attracting Talents “111” base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing University Chongqing China
| | - Kang Xu
- National Innovation and Attracting Talents “111” base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing University Chongqing China
| | - Li Yang
- National Innovation and Attracting Talents “111” base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing University Chongqing China
| |
Collapse
|
47
|
Gonzalez-Leon EA, Bielajew BJ, Hu JC, Athanasiou KA. Engineering self-assembled neomenisci through combination of matrix augmentation and directional remodeling. Acta Biomater 2020; 109:73-81. [PMID: 32344175 DOI: 10.1016/j.actbio.2020.04.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/09/2020] [Accepted: 04/09/2020] [Indexed: 02/07/2023]
Abstract
Knee meniscus injury is frequent, resulting in over 1 million surgeries annually in the United States and Europe. Because of the near-avascularity of this fibrocartilaginous tissue and its intrinsic lack of healing, tissue engineering has been proposed as a solution for meniscus repair and replacement. This study describes an approach employing bioactive stimuli to enhance both extracellular matrix content and organization of neomenisci toward augmenting their mechanical properties. Self-assembled fibrocartilages were treated with TGF-β1, chondroitinase ABC, and lysyl oxidase-like 2 (collectively termed TCL) in addition to lysophosphatidic acid (LPA). TCL + LPA treatment synergistically improved circumferential tensile stiffness and strength, significantly enhanced collagen and pyridinoline crosslink content per dry weight, and achieved tensile anisotropy (circumferential/radial) values of neomenisci close to 4. This study utilizes a combination of bioactive stimuli for use in tissue engineering studies, providing a promising path toward deploying these neomenisci as functional repair and replacement tissues. STATEMENT OF SIGNIFICANCE: This study utilizes a scaffold-free approach, which strays from the tissue engineering paradigm of using scaffolds with cells and bioactive factors to engineer neotissue. While self-assembled neomenisci have attained compressive properties akin to native tissue, tensile properties still require improvement before being able to deploy engineered neomenisci as functional tissue repair or replacement options. In order to augment tensile properties, this study utilized bioactive factors known to augment matrix content in combination with a soluble factor that enhances matrix organization and anisotropy via cell traction forces. Using a bioactive factor to enhance matrix organization mitigates the need for bioreactors used to apply mechanical stimuli or scaffolds to induce proper fiber alignment.
Collapse
Affiliation(s)
- Erik A Gonzalez-Leon
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697.
| | - Benjamin J Bielajew
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697.
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697.
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697.
| |
Collapse
|
48
|
Lin W, Xu L, Li G. Molecular Insights Into Lysyl Oxidases in Cartilage Regeneration and Rejuvenation. Front Bioeng Biotechnol 2020; 8:359. [PMID: 32426343 PMCID: PMC7204390 DOI: 10.3389/fbioe.2020.00359] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Articular cartilage remains among the most difficult tissues to regenerate due to its poor self-repair capacity. The lysyl oxidase family (LOX; also termed as protein-lysine 6-oxidase), mainly consists of lysyl oxidase (LO) and lysyl oxidase-like 1-4 (LOXL1-LOXL4), has been traditionally defined as cuproenzymes that are essential for stabilization of extracellular matrix, particularly cross-linking of collagen and elastin. LOX is essential in the musculoskeletal system, particularly cartilage. LOXs-mediated collagen cross-links are essential for the functional integrity of articular cartilage. Appropriate modulation of the expression or activity of certain LOX members selectively may become potential promising strategy for cartilage repair. In the current review, we summarized the advances of LOX in cartilage homeostasis and functioning, as well as copper-mediated activation of LOX through hypoxia-responsive signaling axis during recent decades. Also, the molecular signaling network governing LOX expression has been summarized, indicating that appropriate modulation of hypoxia-responsive-signaling-directed LOX expression through manipulation of bioavailability of copper and oxygen is promising for further clinical implications of cartilage regeneration, which has emerged as a potential therapeutic approach for cartilage rejuvenation in tissue engineering and regenerative medicine. Therefore, targeted regulation of copper-mediated hypoxia-responsive signalling axis for selective modulation of LOX expression may become potential effective therapeutics for enhanced cartilage regeneration and rejuvenation in future clinical implications.
Collapse
Affiliation(s)
- Weiping Lin
- Department of Orthopaedics and Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Liangliang Xu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.,MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
49
|
Taheem DK, Jell G, Gentleman E. Hypoxia Inducible Factor-1α in Osteochondral Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:105-115. [PMID: 31774026 PMCID: PMC7166133 DOI: 10.1089/ten.teb.2019.0283] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/19/2019] [Indexed: 12/19/2022]
Abstract
Damage to osteochondral (OC) tissues can lead to pain, loss of motility, and progress to osteoarthritis. Tissue engineering approaches offer the possibility of replacing damaged tissues and restoring joint function; however, replicating the spatial and functional heterogeneity of native OC tissue remains a pressing challenge. Chondrocytes in healthy cartilage exist in relatively low-oxygen conditions, while osteoblasts in the underlying bone experience higher oxygen pressures. Such oxygen gradients also exist in the limb bud, where they influence OC tissue development. The cellular response to these spatial variations in oxygen pressure, which is mediated by the hypoxia inducible factor (HIF) pathway, plays a central role in regulating osteo- and chondrogenesis by directing progenitor cell differentiation and promoting and maintaining appropriate extracellular matrix production. Understanding the role of the HIF pathway in OC tissue development may enable new approaches to engineer OC tissue. In this review, we discuss strategies to spatially and temporarily regulate the HIF pathway in progenitor cells to create functional OC tissue for regenerative therapies. Impact statement Strategies to engineer osteochondral (OC) tissue are limited by the complex and varying microenvironmental conditions in native bone and cartilage. Indeed, native cartilage experiences low-oxygen conditions, while the underlying bone is relatively normoxic. The cellular response to these low-oxygen conditions, which is mediated through the hypoxia inducible factor (HIF) pathway, is known to promote and maintain the chondrocyte phenotype. By using tissue engineering scaffolds to spatially and temporally harness the HIF pathway, it may be possible to improve OC tissue engineering strategies for the regeneration of damaged cartilage and its underlying subchondral bone.
Collapse
Affiliation(s)
- Dheraj K. Taheem
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Gavin Jell
- Division of Surgery and Interventional Sciences, University College London, London, United Kingdom
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| |
Collapse
|
50
|
Engineered cartilage utilizing fetal cartilage-derived progenitor cells for cartilage repair. Sci Rep 2020; 10:5722. [PMID: 32235934 PMCID: PMC7109068 DOI: 10.1038/s41598-020-62580-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 01/29/2020] [Indexed: 01/01/2023] Open
Abstract
The aim of this study was to develop a fetal cartilage-derived progenitor cell (FCPC) based cartilage gel through self-assembly for cartilage repair surgery, with clinically useful properties including adhesiveness, plasticity, and continued chondrogenic remodeling after transplantation. Characterization of the gels according to in vitro self-assembly period resulted in increased chondrogenic features over time. Adhesion strength of the cartilage gels were significantly higher compared to alginate gel, with the 2-wk group showing a near 20-fold higher strength (1.8 ± 0.15 kPa vs. 0.09 ± 0.01 kPa, p < 0.001). The in vivo remodeling process analysis of the 2 wk cultured gels showed increased cartilage repair characteristics and stiffness over time, with higher integration-failure stress compared to osteochondral autograft controls at 4 weeks (p < 0.01). In the nonhuman primate investigation, cartilage repair scores were significantly better in the gel group compared to defects alone after 24 weeks (p < 0.001). Cell distribution analysis at 24 weeks showed that human cells remained within the transplanted defects only. A self-assembled, FCPC-based cartilage gel showed chondrogenic repair potential as well as adhesive properties, beneficial for cartilage repair.
Collapse
|