Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4361, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778053

ABSTRACT

Oxygen plays a crucial role in human embryogenesis, homeostasis, and tissue regeneration. Emerging engineered regenerative solutions call for novel oxygen delivery systems. To become a reality, these systems must consider physiological processes, oxygen release mechanisms and the target application. In this review, we explore the biological relevance of oxygen at both a cellular and tissue level, and the importance of its controlled delivery via engineered biomaterials and devices. Recent advances and upcoming trends in the field are also discussed with a focus on tissue-engineered constructs that could meet metabolic demands to facilitate regeneration.


Subject(s)
Oxygen , Regeneration , Tissue Engineering , Humans , Oxygen/metabolism , Tissue Engineering/methods , Regeneration/physiology , Animals , Biocompatible Materials/chemistry
2.
Tissue Eng Part A ; 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38517083

ABSTRACT

In the realm of in situ cartilage engineering, the targeted delivery of both cells and hydrogel materials to the site of a defect serves to directly stimulate chondral repair. Although the in situ application of stem cell-laden soft hydrogels to tissue defects holds great promise for cartilage regeneration, a significant challenge lies in overcoming the inherent limitation of these soft hydrogels, which must attain mechanical properties akin to the native tissue to withstand physiological loading. We therefore developed a system where a gelatin methacryloyl hydrogel laden with human adipose-derived mesenchymal stem cells is combined with a secondary structure to provide bulk mechanical reinforcement. In this study, we used the negative embodied sacrificial template 3D printing technique to generate eight different lattice-based reinforcement structures made of polycaprolactone, which ranged in porosity from 80% to 90% with stiffnesses from 28 ± 5 kPa to 2853 ± 236 kPa. The most promising of these designs, the hex prism edge, was combined with the cellular hydrogel and retained a stable stiffness over 41 days of chondrogenic differentiation. There was no significant difference between the hydrogel-only and hydrogel scaffold group in the sulfated glycosaminoglycan production (340.46 ± 13.32 µg and 338.92 ± 47.33 µg, respectively) or Type II Collagen gene expression. As such, the use of negative printing represents a promising solution for the integration of bulk reinforcement without losing the ability to produce new chondrogenic matrix.

3.
Front Cell Dev Biol ; 12: 1353154, 2024.
Article in English | MEDLINE | ID: mdl-38516128

ABSTRACT

Tissue-engineered implants for bone regeneration require consideration regarding their mineralization and vascularization capacity. Different geometries, such as biomimetic designs and lattices, can influence the mechanical properties and the vascularization capacity of bone-mimicking implants. Negative Embodied Sacrificial Template 3D (NEST3D) printing is a versatile technique across a wide range of materials that enables the production of bone-mimicking scaffolds. In this study, different scaffold motifs (logpile, Voronoi, and trabecular bone) were fabricated via NEST3D printing in polycaprolactone to determine the effect of geometrical design on stiffness (10.44 ± 6.71, 12.61 ± 5.71, and 25.93 ± 4.16 MPa, respectively) and vascularization. The same designs, in a polycaprolactone scaffold only, or when combined with gelatin methacryloyl, were then assessed for their ability to allow the infiltration of blood vessels in a chick chorioallantoic membrane (CAM) assay, a cost-effective and time-efficient in ovo assay to assess vascularization. Our findings showed that gelatin methacrylolyl alone did not allow new chorioallantoic membrane tissue or blood vessels to infiltrate within its structure. However, polycaprolactone on its own or when combined with gelatin methacrylolyl allowed tissue and vessel infiltration in all scaffold designs. The trabecular bone design showed the greatest mineralized matrix production over the three designs tested. This reinforces our hypothesis that both biomaterial choice and scaffold motifs are crucial components for a bone-mimicking scaffold.

4.
Curr Opin Biotechnol ; 86: 103065, 2024 04.
Article in English | MEDLINE | ID: mdl-38301593

ABSTRACT

With little to no ability to self-regenerate, human cartilage defects of the knee remain a major clinical challenge. Tissue engineering strategies include delivering specific types of cells and biomaterials to the injured cartilage for restoration of architecture and function. Pre-clinical models to test the efficacy of the therapies come with high costs and ethical issues, and imperfect prediction of performance in humans. Ex vivo models represent an alternative avenue to trial cartilage tissue engineering. Defined as viable explanted cartilage samples, ex vivo models can be cultured with a cell-laden biomaterial or tissue-engineered construct to evaluate cartilage repair. Though human and animal ex vivo models are currently used in the field, there is a need for alternative methods to assess the strength of integration, to increase throughput and manage variability and to optimise and standardise culture conditions, enhancing the utility of these models overall.


Subject(s)
Cartilage, Articular , Animals , Humans , Cartilage, Articular/surgery , Tissue Engineering , Biocompatible Materials
5.
Tissue Eng Regen Med ; 20(1): 25-47, 2023 02.
Article in English | MEDLINE | ID: mdl-36244053

ABSTRACT

BACKGROUND: Articular cartilage repair using implantable photocrosslinkable hydrogels laden with chondrogenic cells, represents a promising in situ cartilage engineering approach for surgical treatment. The development of a surgical procedure requires a minimal viable product optimized for the clinical scenario. In our previous work we demonstrated how gelatin based photocrosslinkable hydrogels in combination with infrapatellar derived stem cells allow the production of neocartilage in vitro. In this study, we aim to optimize the critical facets of the in situ cartilage engineering therapy: the cell source, the cell isolation methodology, the cell expansion protocol, the cell number, and the delivery approach. METHODS: We evaluated the impact of the critical facets of the cell-laden hydrogel therapy in vitro to define an optimized protocol that was then used in a rabbit model of cartilage repair. We performed cells counting and immunophenotype analyses, chondrogenic potential evaluation via immunostaining and gene expression, extrusion test analysis of the photocrosslinkable hydrogel, and clinical assessment of cartilage repair using macroscopic and microscopic scores. RESULTS: We identified the adipose derived stem cells as the most chondrogenic cells source within the knee joint. We then devised a minimally manipulated stem cell isolation procedure that allows a chondrogenic population to be obtained in only 85 minutes. We found that cell expansion prior to chondrogenesis can be reduced to 5 days after the isolation procedure. We characterized that at least 5 million of cells/ml is needed in the photocrosslinkable hydrogel to successfully trigger the production of neocartilage. The maximum repairable defect was calculated based on the correlation between the number of cells retrievable with the rapid isolation followed by 5-day non-passaged expansion phase, and the minimum chondrogenic concentration in photocrosslinkable hydrogel. We next optimized the delivery parameters of the cell-laden hydrogel therapy. Finally, using the optimized procedure for in situ tissue engineering, we scored superior cartilage repair when compared to the gold standard microfracture approach. CONCLUSION: This study demonstrates the possibility to repair a critical size articular cartilage defect by means of a surgical streamlined procedure with optimized conditions.


Subject(s)
Cartilage, Articular , Hydrogels , Animals , Rabbits , Tissue Engineering/methods , Bone and Bones , Stem Cells
6.
Adv Healthc Mater ; 11(24): e2201305, 2022 12.
Article in English | MEDLINE | ID: mdl-36541723

ABSTRACT

Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.


Subject(s)
Cartilage, Articular , Humans , Cartilage, Articular/metabolism , Tissue Engineering/methods , Prospective Studies , Biocompatible Materials/metabolism , Regeneration , Chondrogenesis , Tissue Scaffolds
7.
Acta Biomater ; 147: 129-146, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35643197

ABSTRACT

In this work we present a standardised quantitative ultrasound imaging (SQUI) approach for the non-destructive three-dimensional imaging and quantification of cartilage formation in hydrogel based bioscaffolds. The standardised concept involves the processing of ultrasound backscatter data with respect to an acellular phantom in combination with the compensation of sound speed mismatch diffraction effects between the bioscaffold and the phantom. As a proof-of-concept, the SQUI approach was tested on a variety of bioscaffolds with varying degree of neocartilage formation. These were composed of Gelatine Methacryloyl (GelMA) hydrogels laden with human adipose-derived stem cells (hADSCs). These were cultured under chondrogenic stimulation following a previously established protocol, where the degree of the neocartilage formation was modulated using different GelMA network densities (6, 8, 10 % w/v) and culture time (0, 14, 28 days). Using the SQUI approach we were able to detect marked acoustic and morphological changes occurring in the bioscaffolds a result of their different chondrogenic outcome. We defined an acoustic neocartilage indicator, the sonomarker, for the selective imaging and quantification of neocartilage formation. The sonomarker, of backscatter intensity logIBC -2.4, was found to correlate with data obtained via standard destructive bioassays. The ultrasonic evaluation of human specimens confirmed the sonomarker as a relevant intensity, although it was found to shift to higher intensity values in proportion to the cartilage condition as inferred from sound speed measurements. This study demonstrates the potential of the SQUI approach for the realization of non-destructive analysis of cartilage regeneration over-time. STATEMENT OF SIGNIFICANCE: As tissue engineering strategies for neocartilage regeneration evolve towards clinical implementation, alternative characterisation approaches that allow the non-destructive monitoring of extracellular matrix formation in implantable hydrogel based bioscaffolds are needed. In this work we present an innovative standardized quantitative ultrasound imaging (SQUI) approach that allows the non-destructive, volumetric, and quantitative evaluation of neocartilage formation in hydrogel based bioscaffolds. The standardised concept aims to provide a robust approach that accounts for the dynamic changes occurring during the conversion from a cellular bioscaffold towards the formation of a neocartilage construct. We believe that the SQUI approach will be of great benefit for the evaluation of constructs developing neocartilage, not only for in-vitro applications but also potentially applicable to in-vivo applications.


Subject(s)
Chondrogenesis , Hydrogels , Cartilage/diagnostic imaging , Cartilage/physiology , Humans , Hydrogels/pharmacology , Tissue Engineering/methods , Ultrasonography
8.
Mar Drugs ; 20(6)2022 May 30.
Article in English | MEDLINE | ID: mdl-35736169

ABSTRACT

Collagens from a wide array of animals have been explored for use in tissue engineering in an effort to replicate the native extracellular environment of the body. Marine-derived biomaterials offer promise over their conventional mammalian counterparts due to lower risk of disease transfer as well as being compatible with more religious and ethical groups within society. Here, collagen type I derived from a marine source (Macruronus novaezelandiae, Blue Grenadier) is compared with the more established porcine collagen type I and its potential in tissue engineering examined. Both collagens were methacrylated, to allow for UV crosslinking during extrusion 3D printing. The materials were shown to be highly cytocompatible with L929 fibroblasts. The mechanical properties of the marine-derived collagen were generally lower than those of the porcine-derived collagen; however, the Young's modulus for both collagens was shown to be tunable over a wide range. The marine-derived collagen was seen to be a potential biomaterial in tissue engineering; however, this may be limited due to its lower thermal stability at which point it degrades to gelatin.


Subject(s)
Bioprinting , Animals , Biocompatible Materials , Collagen , Collagen Type I , Gelatin , Hydrogels , Mammals , Swine , Tissue Engineering , Tissue Scaffolds
9.
Int J Mol Sci ; 22(22)2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34830302

ABSTRACT

Osteochondral (OC) defects are debilitating joint injuries characterized by the loss of full thickness articular cartilage along with the underlying calcified cartilage through to the subchondral bone. While current surgical treatments can provide some relief from pain, none can fully repair all the components of the OC unit and restore its native function. Engineering OC tissue is challenging due to the presence of the three distinct tissue regions. Recent advances in additive manufacturing provide unprecedented control over the internal microstructure of bioscaffolds, the patterning of growth factors and the encapsulation of potentially regenerative cells. These developments are ushering in a new paradigm of 'multiphasic' scaffold designs in which the optimal micro-environment for each tissue region is individually crafted. Although the adoption of these techniques provides new opportunities in OC research, it also introduces challenges, such as creating tissue interfaces, integrating multiple fabrication techniques and co-culturing different cells within the same construct. This review captures the considerations and capabilities in developing 3D printed OC scaffolds, including materials, fabrication techniques, mechanical function, biological components and design.


Subject(s)
Cartilage Diseases/surgery , Mesenchymal Stem Cell Transplantation/methods , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Absorbable Implants , Animals , Biocompatible Materials , Bone and Bones , Cartilage, Articular , Humans , Tissue Transplantation/methods
10.
MethodsX ; 8: 101324, 2021.
Article in English | MEDLINE | ID: mdl-34434839

ABSTRACT

The encapsulation of growth factors is an important component of tissue engineer- ing. Using microspheres is a convenient approach in which the dose of factors can be regulated by increasing or decreasing the number of encapsulated microspheres. Moreover, microspheres offer the possibility of delivering the growth factors directly to the target site. However, the fabrication of microspheres by traditional emulsion methods is largely variable due to the experimental procedure. We have developed a protocol using a commercially available microfluidic system that allows formation of tunable particle-size droplets loaded with growth factors. The methodology includes a guide for preparing an alginate-growth factors solution followed by the specific set-up needed for using the microfluidic system to form the microspheres. The pro- cedure also includes a unique post-crosslinking process without pH modification. These methods allow the preservation of integrity and bioactivity of the growth factors tested (BMP-6 and TGFß -3) and their subsequent sustained delivery.•The protocol can be tuned to form particles of various sizes.•The gentle post-crosslinking process allows conformational integrity of various bioactive molecules.

11.
Acta Biomater ; 129: 110-121, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34010693

ABSTRACT

Mesenchymal stem cell therapies show great promise in regenerative medicine. However, to generate clinically relevant numbers of these stem cells, significant in vitro expansion of the cells is required before transplantation into the affected wound or defect. The current gold standard protocol for recovering in vitro cultured cells involves treatment with enzymes such as trypsin which can affect the cell phenotype and ability to interact with the environment. Alternative enzyme free methods of adherent cell recovery have been investigated, but none match the convenience and performance of enzymatic detachment. In this work we have developed a synthetically simple, low cost cell culture substrate functionalized with gold nanorods that can support cell proliferation and detachment. When these nanorods are irradiated with biocompatible low intensity near infrared radiation (785 nm, 560 mWcm-2) they generate localized surface plasmon resonance induced nanoscale heating effects which trigger detachment of adherent mesenchymal stem cells. Through simulations and thermometry experiments we show that this localized heating is concentrated at the cell-nanorod interface, and that the stem cells detached using this technique show either similar or improved multipotency, viability and ability to differentiate into clinically desirable osteo and adipocytes, compared to enzymatically harvested cells. This proof-of-principle work shows that photothermally mediated cell detachment is a promising method for recovering mesenchymal stem cells from in vitro culture substrates, and paves the way for further studies to scale up this process and facilitate its clinical translation. STATEMENT OF SIGNIFICANCE: New non-enzymatic methods of harvesting adherent cells without damaging or killing them are highly desirable in fields such as regenerative medicine. Here, we present a synthetically simple, non-toxic, infra-red induced method of harvesting mesenchymal stem cells from gold nanorod functionalized substrates. The detached cells retain their ability to differentiate into therapeutically valuable osteo and adipocytes. This work represents a significant improvement on similar cell harvesting studies due to: its simplicity; the use of clinically valuable stem cells as oppose to immortalized cell lines; and the extensive cellular characterization performed. Understanding, not just if cells live or die but how they proliferate and differentiate after photothermal detachment will be essential for the translation of this and similar techniques into commercial devices.


Subject(s)
Mesenchymal Stem Cells , Nanotubes , Infrared Rays , Surface Plasmon Resonance
12.
Polymers (Basel) ; 13(2)2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33477660

ABSTRACT

Degradable bone implants are designed to foster the complete regeneration of natural tissue after large-scale loss trauma. Polycaprolactone (PCL) and hydroxyapatite (HA) composites are promising scaffold materials with superior mechanical and osteoinductive properties compared to the single materials. However, producing three-dimensional (3D) structures with high HA content as well as tuneable degradability remains a challenge. To address this issue and create homogeneously distributed PCL-nanoHA (nHA) scaffolds with tuneable degradation rates through both PCL molecular weight and nHA concentration, we conducted a detailed characterisation and comparison of a range of PCL-nHA composites across three molecular weight PCLs (14, 45, and 80 kDa) and with nHA content up to 30% w/w. In general, the addition of nHA results in an increase of viscosity for the PCL-nHA composites but has little effect on their compressive modulus. Importantly, we observe that the addition of nHA increases the rate of degradation compared to PCL alone. We show that the 45 and 80 kDa PCL-nHA groups can be fabricated via indirect 3D printing and have homogenously distributed nHA even after fabrication. Finally, the cytocompatibility of the composite materials is evaluated for the 45 and 80 kDa groups, with the results showing no significant change in cell number compared to the control. In conclusion, our analyses unveil several features that are crucial for processing the composite material into a tissue engineered implant.

13.
J Colloid Interface Sci ; 587: 240-251, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33360897

ABSTRACT

Drug delivery systems such as microspheres have shown potential in releasing biologicals effectively for tissue engineering applications. Microfluidic systems are especially attractive for generating microspheres as they produce microspheres of controlled-size and in low volumes, using micro-emulsion processes. However, the flow rate dependency on the encapsulation of molecules at a microscale is poorly understood. In particular, the flow rate and pressure parameters might influence the droplet formation and drug encapsulation efficiency. We evaluated the parameters within a two-reagent flow focusing microfluidic chip under continuous formation of hydrogel particles using a flourinated oil and an ionic crosslinkable alginate hydrogel. Fluorescein isothiocyanate-dextran sulfate (FITC-dextran sulfate MW: 40 kDa) was used to evaluate the variation of the encapsulation efficiency with the flow parameters, optimizing droplets and microsphere formation. The ideal flow rates allowing for maximum encapsulation efficiency, were utilised to form bioactive microspheres by delivering transforming growth factor beta-3 (TGFß-3) in cell culture media. Finally, we evaluated the potential of microfluidic-formed microspheres to be included within biological environments. The biocompatibility of the microspheres was tested over 28 days using adult human mesenchymal stem cells (hMSCs). The release profile of the growth factors from microspheres showed a sustained release in media, after an initial burst, up to 30 days. The metabolic activity of the cells cultured in the presence of the microspheres was similar to controls, supporting the biocompatibility of this approach. The fine-tuned parameters for alginate hydrogel to form microspheres have potential in encapsulating and preserving functional structure of bioactive agents for future tissue engineering applications.


Subject(s)
Alginates , Microfluidics , Humans , Hydrogels , Microspheres , Tissue Engineering
14.
Front Med Technol ; 3: 773673, 2021.
Article in English | MEDLINE | ID: mdl-35047967

ABSTRACT

Current surgical techniques to treat articular cartilage defects fail to produce a satisfactory long-term repair of the tissue. Regenerative approaches show promise in their ability to generate hyaline cartilage using biomaterials in combination with stem cells. However, the difficulty of seamlessly integrating the newly generated cartilage with the surrounding tissue remains a likely cause of long-term failure. To begin to address this integration issue, our strategy exploits a biological enzyme (microbial transglutaminase) to effect bioadhesion of a gelatin methacryloyl implant to host tissue. Mechanical characterization of the bioadhesive material shows that enzymatic crosslinking is compatible with photocrosslinking, allowing for a dual-crosslinked system with improved mechanical properties, and a slower degradation rate. Biocompatibility is illustrated with a 3D study of the metabolic activity of encapsulated human adipose derived stem cells. Furthermore, enzymatic crosslinking induced by transglutaminase is not prevented by the presence of cells, as measured by the bulk modulus of the material. Adhesion to human cartilage is demonstrated ex vivo with a significant increase in adhesive strength (5.82 ± 1.4 kPa as compared to 2.87 ± 0.9 kPa, p < 0.01) due to the addition of transglutaminase. For the first time, we have characterized a bioadhesive material composed of microbial transglutaminase and GelMA that can encapsulate cells, be photo crosslinked, and bond to host cartilage, taking a step toward the integration of regenerative implants.

15.
Biomaterials ; 264: 120383, 2021 01.
Article in English | MEDLINE | ID: mdl-33099133

ABSTRACT

Regenerative therapies based on photocrosslinkable hydrogels and stem cells are of growing interest in the field of cartilage repair. Cell-mediated degradation is critical for the successful clinical translation of implanted hydrogels. However, characterising cell-mediated degradation, while simultaneously monitoring the deposition of a distinct new matrix, remains a major challenge. In this study we generated a Fluorescently LAbelled Sensitive Hydrogel (FLASH) to correlate the degradation of a hydrogel bioscaffold with neocartilage formation. Gelatine Methacryloyl (GelMA) was covalently bound to the FITC fluorophore to generate FLASH and bioscaffolds were produced by casting different concentrations of FLASH GelMA, with and without human adipose-derived stem cells (hADSCs) undergoing chondrogenesis. The loss of fluorescence from FLASH bioscaffolds was correlated with changes in mechanical properties, expression of chondrogenic markers and accumulation of a cartilaginous extracellular matrix. The ability of the system to be used as a sensor to monitor bioscaffold degradability during chondrogenesis was evaluated in vitro, in a human ex vivo model of cartilage repair and in a full chondral defect in vivo rabbit model. This study represents a step towards the generation of a high throughput monitoring system to evaluate de novo cartilage formation in tissue engineering therapies.


Subject(s)
Chondrogenesis , Hydrogels , Animals , Cartilage , Extracellular Matrix , Rabbits , Tissue Engineering
16.
Cancers (Basel) ; 12(12)2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33255756

ABSTRACT

Dyskerin is a nucleolar protein involved in the small nucleolar RNA (snoRNA)-guided pseudouridylation of specific uridines on ribosomal RNA (rRNA), and in the stabilization of the telomerase RNA component (hTR). Loss of function mutations in DKC1 causes X-linked dyskeratosis congenita, which is characterized by a failure of proliferating tissues and increased susceptibility to cancer. However, several tumors show dyskerin overexpression. We observed that patients with primary breast cancers with high dyskerin levels are more frequently characterized by shorter survival rates and positive lymph node status than those with tumors with a lower dyskerin expression. To functionally characterize the effects of high dyskerin expression, we generated stably overexpressing DKC1 models finding that increased dyskerin levels conferred a more aggressive cellular phenotype in untransformed immortalized MCF10A cells. Contextually, DKC1 overexpression led to an upregulation of some snoRNAs, including SNORA67 and a significantly increased U1445 modification on 18S rRNA, the known target of SNORA67. Lastly, we found that dyskerin overexpression strongly enhanced the synthetic activity of ribosomes increasing translational efficiency in MCF10A. Altogether, our results indicate that dyskerin may sustain the neoplastic phenotype from an early stage in breast cancer endowing ribosomes with an augmented translation efficiency.

17.
Methods Mol Biol ; 2140: 111-133, 2020.
Article in English | MEDLINE | ID: mdl-32207108

ABSTRACT

In recent years, new technologies based on 3D bioprinting have emerged as ideal tools with which to arrange cells and biomaterials in three dimensions and so achieve tissue engineering's original goals. The simplest and most widely used form of bioprinting is based on pneumatic extrusion, where 3D structures are built up by drawing patterns of cell-laden or non-cell-laden material through a robotically manipulated syringe. Developing and characterizing new biomaterials for 3D bioprinting (i.e., bioinks) is critical for the progress of the field. This chapter describes a series of protocols for developing, optimizing, and testing new bioinks for extrusion-based 3D bioprinting.


Subject(s)
Biocompatible Materials , Bioprinting , Hydrogels , Printing, Three-Dimensional , Tissue Engineering/methods , Biocompatible Materials/chemistry , Bioprinting/methods , Equipment Design , Materials Testing , Pressure , Rheology , Robotics , Software , Syringes
18.
Methods Mol Biol ; 2140: 145-157, 2020.
Article in English | MEDLINE | ID: mdl-32207110

ABSTRACT

Three-dimensional (3D) bioprinting is driving major innovations in the area of cartilage tissue engineering. As an alternative to computer-aided 3D printing, in situ additive manufacturing has the advantage of matching the geometry of the defect to be repaired without specific preliminary image analysis, shaping the bioscaffold within the defect, and achieving the best possible contact between the bioscaffold and the host tissue. Here, we describe an in situ approach that allows 3D bioprinting of human adipose-derived stem cells (hADSCs) laden in 10%GelMa/2%HAMa (GelMa/HAMa) hydrogel. We use coaxial extrusion to obtain a core/shell bioscaffold with high cell viability, as well as adequate mechanical properties for articular cartilage regeneration and repair.


Subject(s)
Biocompatible Materials , Bioprinting/methods , Cartilage, Articular/surgery , Guided Tissue Regeneration/methods , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Printing, Three-Dimensional , Cell Survival , Humans , Hydrogels/radiation effects , Mesenchymal Stem Cells/cytology , Methacrylates , Photochemistry , Tissue Engineering , Tissue Scaffolds
20.
Stem Cells Transl Med ; 9(3): 302-315, 2020 03.
Article in English | MEDLINE | ID: mdl-31769213

ABSTRACT

Three-dimensional biofabrication using photo-crosslinkable hydrogel bioscaffolds has the potential to revolutionize the need for transplants and implants in joints, with articular cartilage being an early target tissue. However, to successfully translate these approaches to clinical practice, several barriers must be overcome. In particular, the photo-crosslinking process may impact on cell viability and DNA integrity, and consequently on chondrogenic differentiation. In this review, we primarily explore the specific sources of cellular cytotoxicity and genotoxicity inherent to the photo-crosslinking reaction, the methods to analyze cell death, cell metabolism, and DNA damage within the bioscaffolds, and the possible strategies to overcome these detrimental effects.


Subject(s)
Cartilage, Articular/metabolism , Cytotoxicity Tests, Immunologic/methods , Hydrogels/metabolism , Mutagenicity Tests/methods , Tissue Scaffolds/standards , Cell Differentiation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...