Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters











Publication year range
1.
Front Bioeng Biotechnol ; 12: 1425073, 2024.
Article in English | MEDLINE | ID: mdl-39355277

ABSTRACT

Melt electrowriting (MEW) is an electric-field-assisted additive biofabrication technique that has brought significant advancements to bioinspired scaffold design for soft tissue engineering and beyond. Owing to its targeted microfiber placement, MEW has become a powerful platform technology for the fabrication of in vitro disease models up to functional biohybrid constructs that are investigated in vivo to reach clinical translation soon. This work provides a concise overview of this rapidly evolving field by highlighting the key contributions of MEW to cardiovascular tissue engineering. Specifically, we i) pinpoint the methods to introduce microvascular networks in thick 3D constructs benefitting from (sacrificial) MEW microfibers, ii) report MEW-based concepts for small-diameter vascular grafts and stents, iii) showcase how contracting cardiac tissues can profit from the tunable structure-property relationship of MEW scaffolds, and iv) address how complete regenerative heart valves can be built on complex fiber scaffold architectures that recapitulate J-shaped tensile properties and tissue heterogeneity. Lastly, we touch on novel biomaterial advancements and discuss the technological challenges of MEW to unlock the full potential of this transformative technology.

2.
Adv Sci (Weinh) ; : e2405004, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39297316

ABSTRACT

Pelvic organ prolapse (POP) is a highly prevalent yet neglected health burden for women. Strengthening thepelvic floor with bioactive tissue-engineered meshes is an emerging concept. The study investigates tissue regenerative design parameters, including degradability, porosity, and angulation, to develop alternative degradable melt electrowritten (MEW) constructs for surgical applications of POP. MEW constructs are fabricated in hierarchical geometries by two-way stacking of the fibers with three different inter layer angles of 90°, 45°, or 22.5°. Implants printed at 22.5° have higher tensile strength under dry conditions and show better vaginal fibroblast (VF) attachment in vitro. In vivo assessment using preclinical mouse and ovine models demonstrates more effective degradation and improved tissue integration in 22.5° angular meshes compared to 90° and 45° meshes, with evidence of neo-collagen deposition within implants at 6 weeks. The pattern and geometry of the layered MEW implants also influence the foreign body response, where in the anti-inflammatory phenotype shows a greater ratio of anti-inflammatory CD206+ M2 macrophages/pro-inflammatory CCR7+ M1 macrophages. This presents an attractive strategy for improving the design and fabrication of next-generation vaginal implants for pelvic reconstructive surgery.

3.
Small ; : e2403729, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39246220

ABSTRACT

Skin equivalents (SE) that recapitulate biological and mechanical characteristics of the native tissue are promising platforms for assessing cosmetics and studying fundamental biological processes. Methods to achieve SEs with well-organized structure, and ideal biological and mechanical properties are limited. Here, the combination of melt electrowritten PCL scaffolds and cell-laden Matrigel to fabricate SE is described. The PCL scaffold provides ideal structural and mechanical properties, preventing deformation of the model. The model consists of a top layer for seeding keratinocytes to mimic the epidermis, and a bottom layer of Matrigel-based dermal compartment with fibroblasts. The compressive modulus and the biological properties after 3-day coculture indicate a close resemblance with the native skin. Using the SE, a testing system to study the damage caused by UVA irradiation and evaluate antioxidant efficacy is established. The effectiveness of Tea polyphenols (TPs) and L-ascorbic acid (Laa) is compared based on free radical generation. TPs are demonstrated to be more effective in downregulating free radical generation. Further, T1 relaxometry is used to detect the generation of free radicals at a single-cell level, which allows tracking of the same cell before and after UVA treatment.

4.
Polymers (Basel) ; 16(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125188

ABSTRACT

The human trabecular meshwork (HTM) is responsible for regulating intraocular pressure (IOP) by means of gradient porosity. Changes in its physical properties, like increases in stiffness or alterations in the extracellular matrix (ECM), are associated with increases in the IOP, which is the primary cause of glaucoma. The complexity of its structure limits the engineered models to one-layered and simple approaches, which do not accurately replicate the biological and physiological cues related to glaucoma. Here, a combination of melt electrowriting (MEW) and solution electrospinning (SE) is explored as a biofabrication technique used to produce a gradient porous scaffold that mimics the multi-layered structure of the native HTM. Polycaprolactone (PCL) constructs with a height of 20-710 µm and fiber diameters of 0.7-37.5 µm were fabricated. After mechanical characterization, primary human trabecular meshwork cells (HTMCs) were seeded over the scaffolds within the subsequent 14-21 days. In order to validate the system's responsiveness, cells were treated with dexamethasone (Dex) and the rho inhibitor Netarsudil (Net). Scanning electron microscopy and immunochemistry staining were performed to evaluate the expected morphological changes caused by the drugs. Cells in the engineered membranes exhibited an HTMC-like morphology and a correct drug response. Although this work demonstrates the utility of combining MEW and SE in reconstructing complex morphological features like the HTM, new geometries and dimensions should be tested, and future works need to be directed towards perfusion studies.

5.
ACS Appl Mater Interfaces ; 16(29): 37707-37721, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39001812

ABSTRACT

The utilization of micronano composite scaffolds has been extensively demonstrated to confer the superior advantages in bone repair compared to single nano- or micron-sized scaffolds. Nevertheless, the enhancement of bioactivities within these composite scaffolds remains challenging. In this study, we propose a novel approach to combine melt electrowriting (MEW) and solution electrospinning (SES) techniques for the fabrication of a composite scaffold incorporating hydroxyapatite (HAP), an osteogenic component, and roxithromycin (ROX), an antibacterial active component. Scanning electron microscopy (SEM) and Fourier-transform infrared spectroscopy (FTIR) confirmed the hierarchical architecture of the nanofiber-microgrid within the scaffold, as well as the successful loading of HAP and ROX. The incorporation of HAP enhanced the water absorption capacity of the composite scaffold, thus promoting cell adhesion and proliferation, as well as osteogenic differentiation. Furthermore, ROX resulted in effective antibacterial capability without any observable cytotoxicity. Finally, the scaffolds were applied to a rat calvarial defect model, and the results demonstrated that the 20% HAP group exhibited superior new bone formation without causing adverse reactions. Therefore, our findings present a promising strategy for designing and fabricating bioactive scaffolds for bone regeneration.


Subject(s)
Anti-Bacterial Agents , Durapatite , Osteogenesis , Tissue Engineering , Tissue Scaffolds , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Animals , Tissue Scaffolds/chemistry , Osteogenesis/drug effects , Rats , Durapatite/chemistry , Durapatite/pharmacology , Bone Regeneration/drug effects , Rats, Sprague-Dawley , Roxithromycin/chemistry , Roxithromycin/pharmacology , Nanofibers/chemistry , Staphylococcus aureus/drug effects , Bone and Bones/drug effects , Cell Proliferation/drug effects , Mice
6.
Nano Lett ; 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833276

ABSTRACT

Inspired by the imbalance between extrinsic and intrinsic tendon healing, this study fabricated a new biofilter scaffold with a hierarchical structure based on a melt electrowriting technique. The outer multilayered fibrous structure with connected porous characteristics provides a novel passageway for vascularization and isolates the penetration of scar fibers, which can be referred to as a biofilter process. In vitro experiments found that the porous architecture in the outer layer can effectively prevent cell infiltration, whereas the aligned fibers in the inner layer can promote cell recruitment and growth, as well as the expression of tendon-associated proteins in a simulated friction condition. It was shown in vivo that the biofilter process could promote tendon healing and reduce scar invasion. Herein, this novel strategy indicates great potential to design new biomaterials for balancing extrinsic and intrinsic healing and realizing scarless tendon healing.

7.
J Nanobiotechnology ; 22(1): 378, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38943185

ABSTRACT

Tissue engineered heart valves (TEHVs) demonstrates the potential for tissue growth and remodel, offering particular benefit for pediatric patients. A significant challenge in designing functional TEHV lies in replicating the anisotropic mechanical properties of native valve leaflets. To establish a biomimetic TEHV model, we employed melt-electrowriting (MEW) technology to fabricate an anisotropic PCL scaffold. By integrating the anisotropic MEW-PCL scaffold with bioactive hydrogels (GelMA/ChsMA), we successfully crafted an elastic scaffold with tunable mechanical properties closely mirroring the structure and mechanical characteristics of natural heart valves. This scaffold not only supports the growth of valvular interstitial cells (VICs) within a 3D culture but also fosters the remodeling of extracellular matrix of VICs. The in vitro experiments demonstrated that the introduction of ChsMA improved the hemocompatibility and endothelialization of TEHV scaffold. The in vivo experiments revealed that, compared to their non-hydrogel counterparts, the PCL-GelMA/ChsMA scaffold, when implanted into SD rats, significantly suppressed immune reactions and calcification. In comparison with the PCL scaffold, the PCL-GelMA/ChsMA scaffold exhibited higher bioactivity and superior biocompatibility. The amalgamation of MEW technology and biomimetic design approaches provides a new paradigm for manufacturing scaffolds with highly controllable microstructures, biocompatibility, and anisotropic mechanical properties required for the fabrication of TEHVs.


Subject(s)
Heart Valves , Hydrogels , Rats, Sprague-Dawley , Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Animals , Tissue Scaffolds/chemistry , Anisotropy , Rats , Hydrogels/chemistry , Biocompatible Materials/chemistry , Heart Valve Prosthesis , Polyesters/chemistry , Cells, Cultured , Humans , Extracellular Matrix/chemistry , Male
8.
Biomed Mater ; 19(5)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38914083

ABSTRACT

Melt electrowriting (MEW) is an additive manufacturing technique that harnesses electro-hydrodynamic phenomena to produce 3D-printed fibres with diameters on the scale of 10s of microns. The ability to print at this small scale provides opportunities to create structures with incredibly fine resolution and highly defined morphology. The current gold standard material for MEW is poly(ϵ-caprolactone) (PCL), a polymer with excellent biocompatibility but lacking in chemical groups that can allow intrinsic additional functionality. To provide this functionality while maintaining PCL's positive attributes, blending was performed with a Poly(Ethylene Glycol) (PEG)-based Acrylate endcapped Urethane-based Precursor (AUP). AUPs are a group of polymers, built on a backbone of existing polymers, which introduce additional functionality by the addition of one or more acrylate groups that terminate the polymer chain of a backbone polymer. By blending with a 20kDa AUP-PEG in small amounts, it is shown that MEW attributes are preserved, producing high-quality meshes. Blends were produced in various PCL:AUP weight ratios (100:0, 90:10 and 0:100) and processed into both solvent-cast films and MEW meshes that were used to characterise the properties of the blends. It was found that the addition of AUP-PEG to PCL significantly increases the hydrophilicity of structures produced with these polymers, and adds swelling capability compared to the non-swelling PCL. The developed blend (90:10) is shown to be processable using MEW, and the quality of manufactured scaffolds is evaluated against pure PCL scaffolds by performing scanning electron microscopy image analysis, with the quality of the novel MEW blend scaffolds showing comparable quality to that of pure PCL. The presence of the functionalisable AUP material on the surface of the developed scaffolds is also confirmed using fluorescence labelling of the acrylate groups. Biocompatibility of the MEW-processable blend was confirmed through a cell viability study, which found a high degree of cytocompatibility.


Subject(s)
Biocompatible Materials , Hydrophobic and Hydrophilic Interactions , Materials Testing , Polyesters , Polyethylene Glycols , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Polyethylene Glycols/chemistry , Polyesters/chemistry , Tissue Scaffolds/chemistry , Biocompatible Materials/chemistry , Tissue Engineering/methods , Humans , Polymers/chemistry , Cell Survival
9.
Polymers (Basel) ; 16(12)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38931980

ABSTRACT

As microfiber-based additive manufacturing (AM) technologies, melt electrowriting (MEW) and solution electrowriting (SEW) have demonstrated efficacy with more biomedically relevant materials. By processing SU-8 resin using MEW and SEW techniques, a material with substantially different mechanical, thermal, and optical properties than that typically processed is introduced. SU-8 polymer is temperature sensitive and requires the devising of a specific heating protocol to be properly processed. Smooth-surfaced microfibers resulted from MEW of SU8 for a short period (from 30 to 90 min), which provides the greatest control and, thus, reproducibility of the printed microfibers. This investigation explores various parameters influencing the electrowriting process, printing conditions, and post-processing to optimize the fabrication of intricate 3D structures. This work demonstrates the controlled generation of straight filaments and complex multi-layered architectures, which were characterized by brightfield, darkfield, and scanning electron microscopy (SEM). This research opens new avenues for the design and development of 3D-printed photonic systems by leveraging the properties of SU-8 after both MEW and SEW processing.

10.
Mater Today Bio ; 26: 101112, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38873104

ABSTRACT

Multifunctional wound dressings, enriched with biologically active agents for preventing or treating infections and promoting wound healing, along with cell delivery capability, are highly needed. To address this issue, composite scaffolds with potential in wound dressing applications were fabricated in this study. The poly-lactic acid/nanodiamonds (PLA/ND) scaffolds were first printed using melt electrowriting (MEW) and then coated with quaternized ß-chitin (QßC). The NDs were well-dispersed in the printed filaments and worked as fillers and bioactive additions to PLA material. Additionally, they improved coating effectiveness due to the interaction between their negative charges (from NDs) and positive charges (from QßC). NDs not only increased the thermal stability of PLA but also benefitted cellular behavior and inhibited the growth of bacteria. Scaffolds coated with QßC increased the effect of bacteria growth inhibition and facilitated the proliferation of human dermal fibroblasts. Additionally, we have observed rapid extracellular matrix (ECM) remodeling on QßC-coated PLA/NDs scaffolds. The scaffolds provided support for cell adhesion and could serve as a valuable tool for delivering cells to chronic wound sites. The proposed PLA/ND scaffold coated with QßC holds great potential for achieving fast healing in various types of wounds.

11.
Acta Biomater ; 180: 337-357, 2024 05.
Article in English | MEDLINE | ID: mdl-38583749

ABSTRACT

Periodontal regeneration requires the re-attachment of oblique and perpendicular periodontal ligament (PDL) fibres to newly formed cementum and alveolar bone, which has proven elusive with existing approaches. In this study, multiple fibre-guiding biphasic tissue engineered constructs were fabricated by melt electrowriting. The biphasic scaffolds were 95 % porous and consisted of a pore size gradient bone compartment and periodontal compartment made of fibre-guiding channels with micro-architectural features ranging from 100 to 60 µm aimed to direct PDL fibre alignment and attachment. In vitro evaluations over 3 and 7 days demonstrated a marked improvement in collagen fibre orientation (over 60 % fully aligned) for scaffolds with micro-architecture ≤100 µm. The biphasic scaffolds were placed on a dentine slice and implanted ectopically, and this demonstrated that all micro-channels groups facilitated oblique and perpendicular alignment and attachment on the dentine with a mean nuclei angle and mean collagen fibre angle of approximately 60° resembling the native periodontal ligament attachment. A further in vivo testing using a surgically created rodent periodontal model highlighted the 80 µm micro-channel group's effectiveness, showing a significant increase in oblique PDL fibre attachment (72 %) and periodontal regeneration (56 %) when compared to all other groups onto the tooth root compared to control groups. Further to this, immunohistochemistry demonstrated the presence of periostin in the newly formed ligament indicating that functional regeneration occurred These findings suggest that scaffold micro-architectures of 100 µm or below can play a crucial role in directing periodontal tissue regeneration, potentially addressing a critical gap in periodontal therapy. STATEMENT OF SIGNIFICANCE: Periodontal regeneration remains a significant clinical challenge. Essential to restoring dental health and function is the proper attachment of the periodontal ligament, which is functionally oriented, to regenerated bone and cementum. Our research presents an innovative biphasic scaffold, utilizing Melt Electrowriting to systematically guide tissue growth. Distinct from existing methods, our scaffold is highly porous, adaptable, and precisely guides periodontal ligament fibre attachment to the opposing tooth root and alveolar bone interfaces, a critical step for achieving periodontal functional regeneration. Our findings not only bridge a significant gap in biomaterial driven tissue guidance but also promise more predictable outcomes for patients, marking a transformative advancement in the field.


Subject(s)
Periodontal Ligament , Tissue Scaffolds , Tissue Scaffolds/chemistry , Periodontal Ligament/physiology , Animals , Tissue Engineering/methods , Male , Humans , Dentin/chemistry , Regeneration
12.
Adv Healthc Mater ; 13(19): e2400426, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38607966

ABSTRACT

Melt Electrowriting (MEW) is a continuously growing manufacturing platform. Its advantage is the consistent production of micro- to nanometer fibers, that stack intricately, forming complex geometrical shapes. MEW allows tuning of the mechanical properties of constructs via the geometry of deposited fibers. Due to this, MEW can create complex mechanics only seen in multi-material compounds and serve as guiding structures for cellular alignment. The advantage of MEW is also shown in combination with other biotechnological manufacturing methods to create multilayered constructs that increase mechanical approximation to native tissues, biocompatibility, and cellular response. These features make MEW constructs a perfect candidate for small-diameter vascular graft structures. Recently, studies have presented fascinating results in this regard, but is this truly the direction that tubular MEW will follow or are there also other options on the horizon? This perspective will explore the origins and developments of tubular MEW and present its growing importance in the field of artificial small-diameter vascular grafts with mechanical modulation and improved biomimicry and the impact of it in convergence with other manufacturing methods and how future technologies like AI may influence its progress.


Subject(s)
Blood Vessel Prosthesis , Tissue Engineering , Humans , Tissue Engineering/methods , Blood Vessels/physiology , Biocompatible Materials/chemistry , Animals , Tissue Scaffolds/chemistry
13.
Adv Mater ; 36(24): e2312084, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38447132

ABSTRACT

Melt electrowriting (MEW) is an emerging additive manufacturing (AM) technology that enables the precise deposition of continuous polymeric microfibers, allowing for the creation of high-resolution constructs. In recent years, MEW has undergone a revolution, with the introduction of active properties or additional functionalities through novel polymer processing strategies, the incorporation of functional fillers, postprocessing, or the combination with other techniques. While extensively explored in biomedical applications, MEW's potential in other fields remains untapped. Thus, this review explores MEW's characteristics from a materials science perspective, emphasizing the diverse range of materials and composites processed by this technique and their current and potential applications. Additionally, the prospects offered by postprinting processing techniques are explored, together with the synergy achieved by combining melt electrowriting with other manufacturing methods. By highlighting the untapped potentials of MEW, this review aims to inspire research groups across various fields to leverage this technology for innovative endeavors.

14.
Macromol Rapid Commun ; 45(11): e2400010, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38458610

ABSTRACT

This work presents a new method for 4D fabrication of two-way shape memory materials that are capable of reversible shapeshifting right after manufacturing, upon application of proper heating and cooling cycles. The innovative solution presented here consists in the combination of highly stretched electrospun shape memory polymer (SMP) nanofibers with a melt electrowritten elastomer. More specifically, the stretched nanofibers are made of a biocompatible thermoplastic polyurethane (TPU) with crystallizable soft segments, undergoing melt-induced contraction and crystallization-induced elongation upon heating and cooling, respectively. Reversible actuation during crystallization becomes possible due to the elastic recovery of the elastomer component, obtained by melt electrowriting of a commercial TPU filament. Thanks to the design freedom offered by additive manufacturing, the elastomer structure also has the role of guiding the shape transformation. Electrospinning and melt electrowriting process parameters are set up so to obtain smart 4D objects capable of two-way shape memory effect (SME), and the possibility of reversible and repeatable actuation is demonstrated. The two components are then combined in different proportions with the aim of tailoring the two-way SME, taking into account the effect of design parameters such as the SMP content, the elastomer pattern, and the composite thickness.


Subject(s)
Nanofibers , Polymers , Polymers/chemistry , Nanofibers/chemistry , Polyurethanes/chemistry , Elastomers/chemistry , Smart Materials/chemistry , Biocompatible Materials/chemistry
15.
Biofabrication ; 16(2)2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38447217

ABSTRACT

As bioprinting advances into clinical relevance with patient-specific tissue and organ constructs, it must be capable of multi-material fabrication at high resolutions to accurately mimick the complex tissue structures found in the body. One of the most fundamental structures to regenerative medicine is microvasculature. Its continuous hierarchical branching vessel networks bridge surgically manipulatable arteries (∼1-6 mm) to capillary beds (∼10µm). Microvascular perfusion must be established quickly for autologous, allogeneic, or tissue engineered grafts to survive implantation and heal in place. However, traditional syringe-based bioprinting techniques have struggled to produce perfusable constructs with hierarchical branching at the resolution of the arterioles (∼100-10µm) found in microvascular tissues. This study introduces the novel CEVIC bioprinting device (i.e.ContinuouslyExtrudedVariableInternalChanneling), a multi-material technology that breaks the current extrusion-based bioprinting paradigm of pushing cell-laden hydrogels through a nozzle as filaments, instead, in the version explored here, extruding thin, wide cell-laden hydrogel sheets. The CEVIC device adapts the chaotic printing approach to control the width and number of microchannels within the construct as it is extruded (i.e. on-the-fly). Utilizing novel flow valve designs, this strategy can produce continuous gradients varying geometry and materials across the construct and hierarchical branching channels with average widths ranging from 621.5 ± 42.92%µm to 11.67 ± 14.99%µm, respectively, encompassing the resolution range of microvascular vessels. These constructs can also include fugitive/sacrificial ink that vacates to leave demonstrably perfusable channels. In a proof-of-concept experiment, a co-culture of two microvascular cell types, endothelial cells and pericytes, sustained over 90% viability throughout 1 week in microchannels within CEVIC-produced gelatin methacryloyl-sodium alginate hydrogel constructs. These results justify further exploration of generating CEVIC-bioprinted microvasculature, such as pre-culturing and implantation studies.


Subject(s)
Bioprinting , Endothelial Cells , Humans , Bioprinting/methods , Tissue Engineering/methods , Hydrogels/chemistry , Printing, Three-Dimensional , Tissue Scaffolds/chemistry
16.
Int J Biol Macromol ; 265(Pt 2): 130938, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38493814

ABSTRACT

The rapid manufacturing of biocomposite scaffold made of saturated-Poly(ε-caprolactone) (PCL) and unsaturated Polyester (PE) blends with gelatin and modified gelatin (NCO-Gel) is demonstrated. Polyester blend-based scaffold are fabricated with and without applying potential in the melt electrowriting system. Notably, the applied potential induces phase separation between PCL and PE and drives the formation of PE rich spots at the interface of electrowritten fibers. The objective of the current study is to control the phase separation between saturated and unsaturated polyesters occurring in the melt electro-writing process and utilization of this phenomenon to improve efficiency of biofunctionalization at the interface of scaffold via Aza-Michael addition reaction. Electron-deficient triple bonds of PE spots on the fibers exhibit good potential for the biofunctionalization via the aza-Michael addition reaction. PE spots are found to be pronounced in which blend compositions are PCL-PE as 90:10 and 75:25 %. The biofunctionalization of scaffold is monitored through CN bond formation appeared at 400 eV via X-ray photoelectron spectroscopy (XPS) and XPS chemical mapping. The described biofunctionalization methodology suggest avoiding use of multi-step chemical modification on additive manufacturing products and thereby rapid prototyping of functional polymer blend based scaffolds with enhanced biocompatibility and preserved mechanical properties. Additionally one-step additive manufacturing method eliminates side effects of toxic solvents and long modification steps during scaffold fabrication.


Subject(s)
Polyesters , Polymers , Polyesters/chemistry , Tissue Scaffolds/chemistry , Gelatin/chemistry , Tissue Engineering/methods
17.
Bioeng Transl Med ; 9(1): e10614, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38193127

ABSTRACT

In articular cartilage (AC), the collagen arcades provide the tissue with its extraordinary mechanical properties. As these structures cannot be restored once damaged, functional restoration of AC defects remains a major challenge. We report that the use of a converged bioprinted, osteochondral implant, based on a gelatin methacryloyl cartilage phase, reinforced with precisely patterned melt electrowritten polycaprolactone micrometer-scale fibers in a zonal fashion, inspired by native collagen architecture, can provide long-term mechanically stable neo-tissue in an orthotopic large animal model. The design of this novel implant was achieved via state-of-the-art converging of extrusion-based ceramic printing, melt electrowriting, and extrusion-based bioprinting. Interestingly, the cell-free implants, used as a control in this study, showed abundant cell ingrowth and similar favorable results as the cell-containing implants. Our findings underscore the hypothesis that mechanical stability is more determining for the successful survival of the implant than the presence of cells and pre-cultured extracellular matrix. This observation is of great translational importance and highlights the aptness of advanced 3D (bio)fabrication technologies for functional tissue restoration in the harsh articular joint mechanical environment.

18.
Mater Today Bio ; 24: 100900, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38234463

ABSTRACT

Incorporating biomolecules as integral parts of computational systems represents a frontier challenge in bio- and nanotechnology. Using DNA to store digital data is an attractive alternative to conventional information technologies due to its high information density and long lifetime. However, developing an adequate DNA storage medium remains a significant challenge in permitting the safe archiving and retrieval of oligonucleotides. This work introduces composite nucleic acid-polymer fibers as matrix materials for digital information-bearing oligonucleotides. We devised a complete workflow for the stable storage of DNA in PEO, PVA, and PCL fibers by employing electrohydrodynamic processes to produce electrospun nanofibers with embedded oligonucleotides. The on-demand retrieval of messages is afforded by non-hazardous chemical treatment and subsequent PCR amplification and DNA sequencing. Finally, we develop a platform for melt-electrowriting of polymer-DNA composites to produce microfiber meshes of programmable patterns and geometries.

19.
Macromol Biosci ; 24(3): e2300397, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37902248

ABSTRACT

Polycaprolactone (PCL) is usually the material chosen for melt electrowriting (MEW) due to its biocompatibility, mechanical strength, and melt processability. This work first investigates the effect of different processing parameters to obtain optimum PCL-MEW scaffolds. Secondly, to increase PCL`s hydrophilicity and cell affinity, and to enable coating with superparamagnetic iron oxide nanoparticles (SPIONs) and silica-coated-SPIONs (Si-SPIONs), the scaffolds are modified with alkaline surface treatment. Finally, SPIONs and Si-SPIONs are successfully coated on MEW scaffolds. Results show that reproducible scaffolds are fabricated. Additionally, the alkaline treatment does not change the three-dimensional morphology of scaffolds while reducing fiber diameter. Furthermore, SEM images and ATR-FTIR results confirmed that SPIONs and Si-SPIONs-were coated on scaffolds. A cytocompatibility assay showed a non-toxic effect on MG-63 osteoblast-like cells in all scaffolds. Additionally, higher MC3T3-E1 pre-osteoblastic cell adhesion efficiency and proliferation are achieved for the alkaline-treated scaffolds and SPIONs/Si-SPIONs-coated scaffolds. All samples demonstrated the ability to generate heat, useful for hyperthermia-treatment, when subjected to an alternating magnetic field. Overall, the findings suggest that the strategy of coating PCL-MEW scaffolds with SPIONs/Si-SPIONs has the potential to improve scaffold performance for biomedical applications, especially for producing magnetically responsive MEW scaffolds.


Subject(s)
Osteoblasts , Tissue Scaffolds , Cell Adhesion , Magnetic Iron Oxide Nanoparticles
20.
Adv Healthc Mater ; 13(3): e2302057, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37933556

ABSTRACT

Emerging additive manufacturing (AM) strategies can enable the engineering of hierarchal scaffold structures for guiding tissue regeneration. Here, the advantages of two AM approaches, melt electrowriting (MEW) and fused deposition modelling (FDM), are leveraged and integrated to fabricate hybrid scaffolds for large bone defect healing. MEW is used to fabricate a microfibrous core to guide bone healing, while FDM is used to fabricate a stiff outer shell for mechanical support, with constructs being coated with pro-osteogenic calcium phosphate (CaP) nano-needles. Compared to MEW scaffolds alone, hybrid scaffolds prevent soft tissue collapse into the defect region and support increased vascularization and higher levels of new bone formation 12 weeks post-implantation. In an additional group, hybrid scaffolds are also functionalized with BMP2 via binding to the CaP coating, which further accelerates healing and facilitates the complete bridging of defects after 12 weeks. Histological analyses demonstrate that such scaffolds support the formation of well-defined annular bone, with an open medullary cavity, smooth periosteal surface, and no evidence of abnormal ectopic bone formation. These results demonstrate the potential of integrating different AM approaches for the development of regenerative biomaterials, and in particular, demonstrate the enhanced bone healing outcomes possible with hybrid MEW-FDM constructs.


Subject(s)
Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Biocompatible Materials/chemistry , Bone and Bones , Wound Healing , Bone Regeneration
SELECTION OF CITATIONS
SEARCH DETAIL