scholarly journals Integrating melt-electrowriting and inkjet bioprinting for engineering structurally organized articular cartilage

2021 ◽  
Author(s):  
Alexandre DUFOUR ◽  
Xavier Barceló Gallostra ◽  
Conor OKeeffe ◽  
Kian F Eichholz ◽  
Stanislas Von Euw ◽  
...  

Successful cartilage engineering requires the generation of biological grafts mimicking the structure, composition and mechanical behaviour of the native tissue. Here melt-electrowriting (MEW) was used to produce arrays of polymeric structures whose function was to orient the growth of cellular aggregates spontaneously generated within these structures, and to provide tensile reinforcement to the resulting tissues. Inkjeting was used to deposit defined numbers of cells into MEW structures, which self-assembled into an organized array of spheroids within hours, ultimately generating a hybrid tissue that was hyaline-like in composition. Structurally, the engineered cartilage mimicked the histotypical organization observed in skeletally immature synovial joints. This biofabrication framework was then used to generate scaled-up (50mm x 50mm) cartilage implants containing over 3,500 cellular aggregates in under 15 minutes. After 8 weeks in culture, a 50-fold increase in the compressive properties of these MEW reinforced tissues were observed, while the tensile properties were still dominated by the polymer network, resulting in a composite construct demonstrating tension-compression nonlinearity mimetic of the native tissue. Helium ion microscopy further demonstrated the development of an arcading collagen network within the engineered tissue. This hybrid bioprinting strategy provides a versatile and scalable approach to engineer cartilage biomimetic grafts for biological joint resurfacing.

2000 ◽  
Vol 122 (3) ◽  
pp. 252-260 ◽  
Author(s):  
Robert L. Mauck ◽  
Michael A. Soltz ◽  
Christopher C. B. Wang ◽  
Dennis D. Wong ◽  
Pen-Hsiu Grace Chao ◽  
...  

Due to its avascular nature, articular cartilage exhibits a very limited capacity to regenerate and to repair. Although much of the tissue-engineered cartilage in existence has been successful in mimicking the morphological and biochemical appearance of hyaline cartilage, it is generally mechanically inferior to the natural tissue. In this study, we tested the hypothesis that the application of dynamic deformational loading at physiological strain levels enhances chondrocyte matrix elaboration in cell-seeded agarose scaffolds to produce a more functional engineered tissue construct than in free swelling controls. A custom-designed bioreactor was used to load cell-seeded agarose disks dynamically in unconfined compression with a peak-to-peak compressive strain amplitude of 10 percent, at a frequency of 1 Hz, 3× (1 hour on, 1 hour off)/day, 5 days/week for 4 weeks. Results demonstrated that dynamically loaded disks yielded a sixfold increase in the equilibrium aggregate modulus over free swelling controls after 28 days of loading (100±16 kPa versus 15±8 kPa,p<0.0001). This represented a 21-fold increase over the equilibrium modulus of day 0 4.8±2.3 kPa. Sulfated glycosaminoglycan content and hydroxyproline content was also found to be greater in dynamically loaded disks compared to free swelling controls at day 21 (p<0.0001 and p=0.002, respectively). [S0148-0731(00)00703-2]


2012 ◽  
Vol 6 (2) ◽  
Author(s):  
Jenni R. Popp ◽  
Justine J. Roberts ◽  
Doug V. Gallagher ◽  
Kristi S. Anseth ◽  
Stephanie J. Bryant ◽  
...  

Mechanical stimulation is essential for chondrocyte metabolism and cartilage matrix deposition. Traditional methods for evaluating developing tissue in vitro are destructive, time consuming, and expensive. Nondestructive evaluation of engineered tissue is promising for the development of replacement tissues. Here we present a novel instrumented bioreactor for dynamic mechanical stimulation and nondestructive evaluation of tissue mechanical properties and extracellular matrix (ECM) content. The bioreactor is instrumented with a video microscope and load cells in each well to measure tissue stiffness and an ultrasonic transducer for evaluating ECM content. Chondrocyte-laden hydrogel constructs were placed in the bioreactor and subjected to dynamic intermittent compression at 1 Hz and 10% strain for 1 h, twice per day for 7 days. Compressive modulus of the constructs, measured online in the bioreactor and offline on a mechanical testing machine, did not significantly change over time. Deposition of sulfated glycosaminoglycan (sGAG) increased significantly after 7 days, independent of loading. Furthermore, the relative reflection amplitude of the loaded constructs decreased significantly after 7 days, consistent with an increase in sGAG content. This preliminary work with our novel bioreactor demonstrates its capabilities for dynamic culture and nondestructive evaluation.


2008 ◽  
Vol 5 (25) ◽  
pp. 929-939 ◽  
Author(s):  
Alexander Augst ◽  
Darja Marolt ◽  
Lisa E Freed ◽  
Charu Vepari ◽  
Lorenz Meinel ◽  
...  

Human mesenchymal stem cells (hMSCs) isolated from bone marrow aspirates were cultured on silk scaffolds in rotating bioreactors for three weeks with either chondrogenic or osteogenic medium supplements to engineer cartilage- or bone-like tissue constructs. Osteochondral composites formed from these cartilage and bone constructs were cultured for an additional three weeks in culture medium that was supplemented with chondrogenic factors, supplemented with osteogenic factors or unsupplemented. Progression of cartilage and bone formation and the integration between the two regions were assessed by medical imaging (magnetic resonance imaging and micro-computerized tomography imaging), and by biochemical, histological and mechanical assays. During composite culture (three to six weeks), bone-like tissue formation progressed in all three media to a markedly larger extent than cartilage-like tissue formation. The integration of the constructs was most enhanced in composites cultured in chondrogenic medium. The results suggest that tissue composites with well-mineralized regions and substantially less developed cartilage regions can be generated in vitro by culturing hMSCs on silk scaffolds in bioreactors, that hMSCs have markedly higher capacity for producing engineered bone than engineered cartilage, and that chondrogenic factors play major roles at early stages of bone formation by hMSCs and in the integration of the two tissue constructs into a tissue composite.


2021 ◽  
Vol 14 (1) ◽  
pp. 015008
Author(s):  
Ross Burdis ◽  
Farhad Chariyev-Prinz ◽  
Daniel J Kelly

Abstract Despite sustained efforts, engineering truly biomimetic articular cartilage (AC) via traditional top-down approaches remains challenging. Emerging biofabrication strategies, from 3D bioprinting to scaffold-free approaches that leverage principles of cellular self-organisation, are generating significant interest in the field of cartilage tissue engineering as a means of developing biomimetic tissue analogues in vitro. Although such strategies have advanced the quality of engineered cartilage, recapitulation of many key structural features of native AC, in particular a collagen network mimicking the tissue’s ‘Benninghoff arcade’, remains elusive. Additionally, a complete solution to fixating engineered cartilages in situ within damaged synovial joints has yet to be identified. This study sought to address both of these key challenges by engineering biomimetic AC within a device designed to anchor the tissue within a synovial joint defect. We first designed and fabricated a fixation device capable of anchoring engineered cartilage into the subchondral bone. Next, we developed a strategy for inkjet printing porcine mesenchymal stem/stromal cells (MSCs) into this supporting fixation device, which was also designed to provide instructive cues to direct the self-organisation of MSC condensations towards a stratified engineered AC. We found that a higher starting cell-density supported the development of a more zonally defined collagen network within the engineered tissue. Dynamic culture was implemented to further enhance the quality of this engineered tissue, resulting in an approximate 3 fold increase in glycosaminoglycan and collagen accumulation. Ultimately this strategy supported the development of AC that exhibited near-native levels of glycosaminoglycan accumulation (>5% WW), as well as a biomimetic collagen network organisation with a perpendicular to a parallel fibre arrangement (relative to the tissue surface) from the deep to superficial zones via arcading fibres within the middle zone of the engineered tissue. Collectively, this work demonstrates the successful convergence of novel biofabrication methods, bioprinting strategies and culture regimes to engineer a hybrid implant suited to resurfacing AC defects.


2007 ◽  
Vol 35 (4) ◽  
pp. 677-679 ◽  
Author(s):  
M. Plainfossé ◽  
P.V. Hatton ◽  
A. Crawford ◽  
Z.M. Jin ◽  
J. Fisher

Low-friction surfaces are critical for efficient joint articulation. The tribological properties of articular cartilage have been studied extensively in native tissue and joints. Despite their importance, very few studies have examined the frictional properties of tissue-engineered cartilage. We have therefore reviewed the relationship between composition, structure and friction in tissue-engineered cartilage.


Author(s):  
Andrea R. Tan ◽  
Eric G. Lima ◽  
Kacey G. Marra ◽  
Clark T. Hung

Tissue-engineering has great potential for treating cartilage pathologies such as osteoarthritis by replacing degraded tissue with newly developed engineered tissue. However proinflammatory cytokines such as interleukin-1α (IL-1α) are a confounding issue as they are often present in high concentrations as part of the chronic pathology or as a result of the surgical intervention itself(1). The catabolic effects of these mediators may be especially pronounced in engineered tissues whose cells are not yet fully embedded in the potentially chondroprotective enclosure of a cartilaginous extracellular matrix(2). One method to protecting initially fragile constructs from degradation may be through the use of non-toxic cross-linking agents. Genipin is a naturally occurring crosslinking agent that reacts with amino acids or amine groups and leads to the formation of stable crosslinked products that are identifiable by a dark blue pigment (Figure 1). Cartilage cross-linked with genipin has been shown to be more resistant to collagenase digestion(3) and to injection of chondroitinase-ABC(4). In this study, we examined whether engineered constructs pre-treated with genipin would better resist IL-1α induced catabolic degradation.


Author(s):  
Sonal R. Sampat ◽  
Drew A. Robinson ◽  
George P. Ackerman ◽  
Matthew V. Dermksian ◽  
Gerard A. Ateshian ◽  
...  

The avascular nature of cartilage and the harsh joint loading environment lead to a poor intrinsic healing capacity after injury, motivating the development of cell-based therapies for repair. Synovium-derived stem cells (SDSCs) have the potential for differentiating down a chondrogenic lineage and are thought to aid in articular cartilage repair after damage in vivo1. In the present study, we adopt a two-pronged strategy for growing clinically relevant cartilage grafts. Firstly, we compare the potential of SDSCs versus chondrocytes for engineering functional constructs. Secondly, we investigate the effect of extracellular osmolarity on mechanical and biochemical properties of SDSCs and similarly passaged chondrocytes in 3D culture. This approach is motivated by the fact that the in situ osmotic environment of chondrocytes varies with proteoglycan content and tissue deformation, altering the regulation of chondrocyte activity through mechanotransduction pathways2. We test the hypothesis that application of a hypertonic, more physiologic osmotic environment (created by addition of NaCl and KCl) relative to hypotonic media (300 mOsm), during 3D culture of SDSCs or chondrocytes in agarose hydrogels, improves the biochemical composition and mechanical properties of engineered tissue constructs.


Author(s):  
Linda M. Kock ◽  
Corrinus C. van Donkelaar ◽  
Keita Ito

High prevalence of osteoarthritis and poor intrinsic healing capacity of articular cartilage create a demand for cell-based strategies for cartilage repair. It is possible to tissue engineer cartilage with almost native proteoglycan content, but collagen reaches only 15% to 35% of the native content. Also its natural structural organization is not reproduced. These drawbacks contribute to its insufficient load-bearing properties.


2010 ◽  
Vol 132 (9) ◽  
Author(s):  
Cevat Erisken ◽  
Dilhan M. Kalyon ◽  
Hongjun Wang

The complex micro-/nanostructure of native cartilage-to-bone insertion exhibits gradations in extracellular matrix components, leading to variations in the viscoelastic and biomechanical properties along its thickness to allow for smooth transition of loads under physiological movements. Engineering a realistic tissue for osteochondral interface would, therefore, depend on the ability to develop scaffolds with properly graded physical and chemical properties to facilitate the mimicry of the complex elegance of native tissue. In this study, polycaprolactone nanofiber scaffolds with spatially controlled concentrations of β-tricalcium phosphate nanoparticles were fabricated using twin-screw extrusion-electrospinning process and seeded with MC3T3-E1 cells to form osteochondral tissue constructs. The objective of the study was to evaluate the linear viscoelastic and compressive properties of the native bovine osteochondral tissue and the tissue constructs formed in terms of their small-amplitude oscillatory shear, unconfined compression, and stress relaxation behavior. The native tissue, engineered tissue constructs, and unseeded scaffolds exhibited linear viscoelastic behavior for strain amplitudes less than 0.1%. Both native tissue and engineered tissue constructs demonstrated qualitatively similar gel-like behavior as determined using linear viscoelastic material functions. The normal stresses in compression determined at 10% strain for the unseeded scaffold, the tissue constructs cultured for four weeks, and the native tissue were 0.87±0.08 kPa, 3.59±0.34 kPa, and 210.80±8.93 kPa, respectively. Viscoelastic and biomechanical properties of the engineered tissue constructs were observed to increase with culture time reflecting the development of a tissuelike structure. These experimental findings suggest that viscoelastic material functions of the tissue constructs can provide valuable inputs for the stages of in vitro tissue development.


Author(s):  
Terri-Ann N. Kelly ◽  
Brendan L. Roach ◽  
Charles R. Mackenzie-Smith ◽  
Adam B. Nover ◽  
Eben G. Estell ◽  
...  

Native articular cartilage exhibits tension-compression nonlinearity (TCN), where the compressive modulus is lower than its relatively high tensile modulus [1–2]. TCN produces in restricted lateral expansion of the tissue upon axial compression. We previously demnostrated that osmotic swelling can be used to measure the TCN of engineered cartilage by placing the tissue in an initial state of tensile strain. Incremental application of compression can be used to study the tissue’s mechanical properties as it transitions from tension to compression [3]. Although engineered cartilage is able to achieve the Young’s modulus (E Y) and glycosaminoglycan (GAG) content of native tissue, the collagen content and dynamic modulus (G*) consistently underperform the native tissue. Removing GAG with chondroitinase ABC (cABC) has been shown to significantly decrease the tissue properties immediately after digestion but the properties rebound, with improved collagen content and G* compared to undigested controls [4]. Furthermore, we have previously shown that cABC digestion significantly increases TCN in engineered cartilage [3]. Dynamic loading (DL) has been shown to significantly increase the mechanical properties without significantly altering biochemical composition of engineered cartilage, however the mechanism through which DL modulates the mechanical strength of engineered cartilage may be due in part to improved extracellular matrix (ECM) organization [5]. We therefore hypothesize that cABC digestion and DL will improve the tensile properties of engineered cartilage.


Sign in / Sign up

Export Citation Format

Share Document