Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Biomater Adv ; 160: 213847, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657288

ABSTRACT

Three-dimensional (3D) organoid models have been instrumental in understanding molecular mechanisms responsible for many cellular processes and diseases. However, established organic biomaterial scaffolds used for 3D hydrogel cultures, such as Matrigel, are biochemically complex and display significant batch variability, limiting reproducibility in experiments. Recently, there has been significant progress in the development of synthetic hydrogels for in vitro cell culture that are reproducible, mechanically tuneable, and biocompatible. Self-assembling peptide hydrogels (SAPHs) are synthetic biomaterials that can be engineered to be compatible with 3D cell culture. Here we investigate the ability of PeptiGel® SAPHs to model the mammary epithelial cell (MEC) microenvironment in vitro. The positively charged PeptiGel®Alpha4 supported MEC viability, but did not promote formation of polarised acini. Modifying the stiffness of PeptiGel® Alpha4 stimulated changes in MEC viability and changes in protein expression associated with altered MEC function, but did not fully recapitulate the morphologies of MECs grown in Matrigel. To supply the appropriate biochemical signals for MEC organoids, we supplemented PeptiGels® with laminin. Laminin was found to require negatively charged PeptiGel® Alpha7 for functionality, but was then able to provide appropriate signals for correct MEC polarisation and expression of characteristic proteins. Thus, optimisation of SAPH composition and mechanics allows tuning to support tissue-specific organoids.


Subject(s)
Cell Culture Techniques, Three Dimensional , Collagen , Drug Combinations , Epithelial Cells , Hydrogels , Laminin , Peptides , Proteoglycans , Laminin/pharmacology , Laminin/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Proteoglycans/pharmacology , Proteoglycans/chemistry , Collagen/chemistry , Collagen/pharmacology , Peptides/pharmacology , Peptides/chemistry , Epithelial Cells/drug effects , Epithelial Cells/cytology , Humans , Female , Cell Culture Techniques, Three Dimensional/methods , Cell Survival/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Mammary Glands, Human/cytology , Organoids/drug effects , Organoids/cytology , Cell Culture Techniques/methods
2.
Sci Rep ; 14(1): 2696, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38302538

ABSTRACT

Osteoarthritis is the most common degenerative joint condition, leading to articular cartilage (AC) degradation, chronic pain and immobility. The lack of appropriate therapies that provide tissue restoration combined with the limited lifespan of joint-replacement implants indicate the need for alternative AC regeneration strategies. Differentiation of human pluripotent stem cells (hPSCs) into AC progenitors may provide a long-term regenerative solution but is still limited due to the continued reliance upon growth factors to recapitulate developmental signalling processes. Recently, TTNPB, a small molecule activator of retinoic acid receptors (RARs), has been shown to be sufficient to guide mesodermal specification and early chondrogenesis of hPSCs. Here, we modified our previous differentiation protocol, by supplementing cells with TTNPB and administering BMP2 at specific times to enhance early development (referred to as the RAPID-E protocol). Transcriptomic analyses indicated that activation of RAR signalling significantly upregulated genes related to limb and embryonic skeletal development in the early stages of the protocol and upregulated genes related to AC development in later stages. Chondroprogenitors obtained from RAPID-E could generate cartilaginous pellets that expressed AC-related matrix proteins such as Lubricin, Aggrecan, and Collagen II, but additionally expressed Collagen X, indicative of hypertrophy. This protocol could lay the foundations for cell therapy strategies for osteoarthritis and improve the understanding of AC development in humans.


Subject(s)
Benzoates , Cartilage, Articular , Osteoarthritis , Pluripotent Stem Cells , Retinoids , Humans , Chondrocytes/metabolism , Tretinoin/pharmacology , Chondrogenesis/genetics , Cell Differentiation , Cartilage, Articular/metabolism , Collagen/metabolism , Osteoarthritis/metabolism
3.
Nanoscale ; 16(11): 5653-5664, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38414413

ABSTRACT

Graphene Oxide (GO) has been shown to increase the expression of key cartilage genes and matrix components within 3D scaffolds. Understanding the mechanisms behind the chondroinductive ability of GO is critical for developing articular cartilage regeneration therapies but remains poorly understood. The objectives of this work were to elucidate the effects of GO on the key chondrogenic signalling pathway - TGFß and identify the mechanism through which signal activation is achieved in human chondrocytes. Activation of canonical signalling was validated through GO-induced SMAD-2 phosphorylation and upregulation of known TGFß response genes, while the use of a TGFß signalling reporter assay allowed us to identify the onset of GO-induced signal activation which has not been previously reported. Importantly, we investigate the cell-material interactions and molecular mechanisms behind these effects, establishing a novel link between GO, the plasma membrane and intracellular signalling. By leveraging fluorescent lifetime imaging (FLIM) and a membrane tension probe, we reveal GO-mediated increases in plasma membrane tension, in real-time for the first time. Furthermore, we report the activation of mechanosensory pathways which are known to be regulated by changes in plasma membrane tension and reveal the activation of endogenous latent TGFß in the presence of GO, providing a mechanism for signal activation. The data presented here are critical to understanding the chondroinductive properties of GO and are important for the implementation of GO in regenerative medicine.


Subject(s)
Cartilage, Articular , Chondrocytes , Graphite , Humans , Transforming Growth Factor beta/metabolism , Cell Line , Cell Membrane/metabolism
4.
Cell Rep ; 42(12): 113502, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38032796

ABSTRACT

Optogenetics is a rapidly advancing technology combining photochemical, optical, and synthetic biology to control cellular behavior. Together, sensitive light-responsive optogenetic tools and human pluripotent stem cell differentiation models have the potential to fine-tune differentiation and unpick the processes by which cell specification and tissue patterning are controlled by morphogens. We used an optogenetic bone morphogenetic protein (BMP) signaling system (optoBMP) to drive chondrogenic differentiation of human embryonic stem cells (hESCs). We engineered light-sensitive hESCs through CRISPR-Cas9-mediated integration of the optoBMP system into the AAVS1 locus. The activation of optoBMP with blue light, in lieu of BMP growth factors, resulted in the activation of BMP signaling mechanisms and upregulation of a chondrogenic phenotype, with significant transcriptional differences compared to cells in the dark. Furthermore, cells differentiated with light could form chondrogenic pellets consisting of a hyaline-like cartilaginous matrix. Our findings indicate the applicability of optogenetics for understanding human development and tissue engineering.


Subject(s)
Optogenetics , Pluripotent Stem Cells , Humans , Chondrocytes , Cell Differentiation/genetics , Cartilage/metabolism , Chondrogenesis/genetics , Bone Morphogenetic Protein 2/metabolism , Cells, Cultured
5.
Pharmaceutics ; 15(10)2023 Oct 07.
Article in English | MEDLINE | ID: mdl-37896192

ABSTRACT

Naturally derived polysaccharide-based hydrogels, such as alginate, are frequently used in the design of bioinks for 3D bioprinting. Traditionally, the formulation of such bioinks requires the use of pre-reticulated materials with low viscosities, which favour cell viability but can negatively influence the resolution and shape fidelity of the printed constructs. In this work, we propose the use of cellulose nanocrystals (CNCs) as a rheological modifier to improve the printability of alginate-based bioinks whilst ensuring a high viability of encapsulated cells. Through rheological analysis, we demonstrate that the addition of CNCs (1% and 2% (w/v)) to alginate hydrogels (1% (w/v)) improves shear-thinning behaviour and mechanical stability, resulting in the high-fidelity printing of constructs with superior resolution. Importantly, LIVE/DEAD results confirm that the presence of CNCs does not seem to affect the health of immortalised chondrocytes (TC28a2) that remain viable over a period of seven days post-encapsulation. Taken together, our results indicate a favourable effect of the CNCs on the rheological and biocompatibility properties of alginate hydrogels, opening up new perspectives for the application of CNCs in the formulation of bioinks for extrusion-based bioprinting.

6.
Adv Healthc Mater ; 12(23): e2300443, 2023 09.
Article in English | MEDLINE | ID: mdl-37353904

ABSTRACT

3D bioprinting has developed tremendously in the last couple of years and enables the fabrication of simple, as well as complex, tissue models. The international space agencies have recognized the unique opportunities of these technologies for manufacturing cell and tissue models for basic research in space, in particular for investigating the effects of microgravity and cosmic radiation on different types of human tissues. In addition, bioprinting is capable of producing clinically applicable tissue grafts, and its implementation in space therefore can support the autonomous medical treatment options for astronauts in future long term and far-distant space missions. The article discusses opportunities but also challenges of operating different types of bioprinters under space conditions, mainly in microgravity. While some process steps, most of which involving the handling of liquids, are challenging under microgravity, this environment can help overcome problems such as cell sedimentation in low viscous bioinks. Hopefully, this publication will motivate more researchers to engage in the topic, with publicly available bioprinting opportunities becoming available at the International Space Station (ISS) in the imminent future.


Subject(s)
Bioprinting , Cosmic Radiation , Space Flight , Weightlessness , Humans , Printing, Three-Dimensional
7.
Mar Drugs ; 21(2)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36827101

ABSTRACT

Collagen is the most ubiquitous biomacromolecule found in the animal kingdom and is commonly used as a biomaterial in regenerative medicine therapies and biomedical research. The collagens used in these applications are typically derived from mammalian sources which poses sociological issues due to widespread religious constraints, rising ethical concern over animal rights and the continuous risk of zoonotic disease transmission. These issues have led to increasing research into alternative collagen sources, of which marine collagens, in particular from jellyfish, have emerged as a promising resource. This study provides a characterization of the biophysical properties and cell adhesion interactions of collagen derived from the jellyfish Rhizostoma pulmo (JCol). Circular dichroism spectroscopy and atomic force microscopy were used to observe the triple-helical conformation and fibrillar morphology of JCol. Heparin-affinity chromatography was also used to demonstrate the ability of JCol to bind to immobilized heparin. Cell adhesion assays using integrin blocking antibodies and HT-1080 human fibrosarcoma cells revealed that adhesion to JCol is primarily performed via ß1 integrins, with the exception of α2ß1 integrin. It was also shown that heparan sulfate binding plays a much greater role in fibroblast and mesenchymal stromal cell adhesion to JCol than for type I mammalian collagen (rat tail collagen). Overall, this study highlights the similarities and differences between collagens from mammalian and jellyfish origins, which should be considered when utilizing alternative collagen sources for biomedical research.


Subject(s)
Cnidaria , Collagen , Scyphozoa , Animals , Humans , Rats , Cell Adhesion , Cnidaria/metabolism , Collagen/chemistry , Integrins/metabolism , Scyphozoa/chemistry
8.
Int J Mol Sci ; 23(13)2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35805921

ABSTRACT

Back pain is one of the leading causes of disability worldwide and is frequently caused by degeneration of the intervertebral discs. The discs' development, homeostasis, and degeneration are driven by a complex series of biochemical and physical extracellular matrix cues produced by and transmitted to native cells. Thus, understanding the roles of different cues is essential for designing effective cellular and regenerative therapies. Omics technologies have helped identify many new matrix cues; however, comparatively few matrix molecules have thus far been incorporated into tissue engineered models. These include collagen type I and type II, laminins, glycosaminoglycans, and their biomimetic analogues. Modern biofabrication techniques, such as 3D bioprinting, are also enabling the spatial patterning of matrix molecules and growth factors to direct regional effects. These techniques should now be applied to biochemically, physically, and structurally relevant disc models incorporating disc and stem cells to investigate the drivers of healthy cell phenotype and differentiation. Such research will inform the development of efficacious regenerative therapies and improved clinical outcomes.


Subject(s)
Intervertebral Disc Degeneration , Intervertebral Disc , Cell Differentiation , Cues , Extracellular Matrix/metabolism , Humans , Intervertebral Disc/metabolism , Intervertebral Disc Degeneration/metabolism
9.
Crit Rev Biotechnol ; 42(5): 774-793, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34488516

ABSTRACT

Here, we review the use of human pluripotent stem cells for skeletal tissue engineering. A number of approaches have been used for generating cartilage and bone from both human embryonic stem cells and induced pluripotent stem cells. These range from protocols relying on intrinsic cell interactions and signals from co-cultured cells to those attempting to recapitulate the series of steps occurring during mammalian skeletal development. The importance of generating authentic tissues rather than just differentiated cells is emphasized and enabling technologies for doing this are reported. We also review the different methods for characterization of skeletal cells and constructs at the tissue and single-cell level, and indicate newer resources not yet fully utilized in this field. There have been many challenges in this research area but the technologies to overcome these are beginning to appear, often adopted from related fields. This makes it more likely that cost-effective and efficacious human pluripotent stem cell-engineered constructs may become available for skeletal repair in the near future.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Animals , Cell Differentiation , Humans , Mammals , Tissue Engineering
10.
Polymers (Basel) ; 13(9)2021 May 09.
Article in English | MEDLINE | ID: mdl-34065114

ABSTRACT

Biofabrication strategies continue to gain considerable interest in the efforts to develop methods for better replicating in vitro models of human tissues [...].

11.
Polymers (Basel) ; 13(1)2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33401469

ABSTRACT

Additive manufacturing (AM) is changing our current approach to the clinical treatment of bone diseases, providing new opportunities to fabricate customized, complex 3D structures with bioactive materials. Among several AM techniques, the BioCell Printing is an advanced, integrated system for material manufacture, sterilization, direct cell seeding and growth, which allows for the production of high-resolution micro-architectures. This work proposes the use of the BioCell Printing to fabricate polymer-based scaffolds reinforced with ceramics and loaded with bisphosphonates for the treatment of osteoporotic bone fractures. In particular, biodegradable poly(ε-caprolactone) was blended with hydroxyapatite particles and clodronate, a bisphosphonate with known efficacy against several bone diseases. The scaffolds' morphology was investigated by means of Scanning Electron Microscopy (SEM) and micro-Computed Tomography (micro-CT) while Energy Dispersive X-ray Spectroscopy (EDX) and X-ray Photoelectron Spectroscopy (XPS) revealed the scaffolds' elemental composition. A thermal characterization of the composites was accomplished by Thermogravimetric analyses (TGA). The mechanical performance of printed scaffolds was investigated under static compression and compared against that of native human bone. The designed 3D scaffolds promoted the attachment and proliferation of human MSCs. In addition, the presence of clodronate supported cell differentiation, as demonstrated by the normalized alkaline phosphatase activity. The obtained results show that the BioCell Printing can easily be employed to generate 3D constructs with pre-defined internal/external shapes capable of acting as a temporary physical template for regeneration of cancellous bone tissues.

12.
Acta Biomater ; 119: 140-154, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33189954

ABSTRACT

The repair of tendon injuries is often compromised by post-operative peritendinous adhesions. Placing a physical barrier at the interface between the tendon and the surrounding tissue could potentially solve this problem by reducing adhesion formation. At present, no such system is available for routine use in clinical practice. Here, we propose the development of a bilayer membrane combining a nanofibrous poly(ε-caprolactone) (PCL) electrospun mesh with a layer of self-assembling peptide hydrogel (SAPH) laden with type-B synoviocytes. This bilayer membrane would act as an anti-adhesion system capable of restoring tendon lubrication, while assisting with synovial sheath regeneration. The PCL mesh showed adequate mechanical properties (Young's modulus=19±4 MPa, ultimate tensile stress=9.6±1.7 MPa, failure load=0.5±0.1 N), indicating that the membrane is easy to handle and capable to withstand the frictional forces generated on the tendon's surface during movement (~0.3 N). Morphological analysis confirmed the generation of a mesh with nanosized PCL fibres and small pores (< 3 µm), which prevented fibroblast infiltration to impede extrinsic healing but still allowing diffusion of nutrients and waste. Rheological tests showed that incorporation of SAPH layer allows good lubrication properties when the membrane is articulated against porcine tendon or hypodermis, suggesting that restoration of tendon gliding is possible upon implantation. Moreover, viability and metabolic activity tests indicated that the SAPH was conducive to rabbit synoviocyte growth and proliferation over 28 days of 3D culture, sustaining cell production of specific matrix components, particularly hyaluronic acid. Synoviocyte-laden peptide hydrogel promoted a sustained endogenous production of hyaluronic acid, providing an anti-friction layer that potentially restores the tendon gliding environment.


Subject(s)
Hydrogels , Tendon Injuries , Animals , Hyaluronic Acid , Polyesters , Rabbits , Swine , Tendon Injuries/pathology , Tendons/pathology , Tissue Adhesions/pathology , Tissue Engineering
13.
Chem Rev ; 120(19): 11128-11174, 2020 10 14.
Article in English | MEDLINE | ID: mdl-32937071

ABSTRACT

The lack of in vitro tissue and organ models capable of mimicking human physiology severely hinders the development and clinical translation of therapies and drugs with higher in vivo efficacy. Bioprinting allow us to fill this gap and generate 3D tissue analogues with complex functional and structural organization through the precise spatial positioning of multiple materials and cells. In this review, we report the latest developments in terms of bioprinting technologies for the manufacturing of cellular constructs with particular emphasis on material extrusion, jetting, and vat photopolymerization. We then describe the different base polymers employed in the formulation of bioinks for bioprinting and examine the strategies used to tailor their properties according to both processability and tissue maturation requirements. By relating function to organization in human development, we examine the potential of pluripotent stem cells in the context of bioprinting toward a new generation of tissue models for personalized medicine. We also highlight the most relevant attempts to engineer artificial models for the study of human organogenesis, disease, and drug screening. Finally, we discuss the most pressing challenges, opportunities, and future prospects in the field of bioprinting for tissue engineering (TE) and regenerative medicine (RM).


Subject(s)
Bioprinting , Polymers/chemistry , Precision Medicine , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds/chemistry , Humans
14.
Polymers (Basel) ; 12(7)2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32630145

ABSTRACT

Degeneration of articular cartilage (AC) is a common healthcare issue that can result in significantly impaired function and mobility for affected patients. The avascular nature of the tissue strongly burdens its regenerative capacity contributing to the development of more serious conditions such as osteoarthritis. Recent advances in bioprinting have prompted the development of alternative tissue engineering therapies for the generation of AC. Particular interest has been dedicated to scaffold-based strategies where 3D substrates are used to guide cellular function and tissue ingrowth. Despite its extensive use in bioprinting, the application of polycaprolactone (PCL) in AC is, however, restricted by properties that inhibit pro-chondrogenic cell phenotypes. This study proposes the use of a new bioprintable poly(ester urea) (PEU) material as an alternative to PCL for the generation of an in vitro model of early chondrogenesis. The polymer was successfully printed into 3D constructs displaying adequate substrate stiffness and increased hydrophilicity compared to PCL. Human chondrocytes cultured on the scaffolds exhibited higher cell viability and improved chondrogenic phenotype with upregulation of genes associated with type II collagen and aggrecan synthesis. Bioprinted PEU scaffolds could, therefore, provide a potential platform for the fabrication of bespoke, pro-chondrogenic tissue engineering constructs.

15.
ACS Biomater Sci Eng ; 6(1): 375-388, 2020 01 13.
Article in English | MEDLINE | ID: mdl-33463228

ABSTRACT

In the current study, Sr/Fe co-substituted hydroxyapatite (HAp) bioceramics were prepared by the sonication-assisted aqueous chemical precipitation method followed by sintering at 1100 °C for bone tissue regeneration applications. The sintered bioceramics were analyzed for various structural and chemical properties through X-ray diffraction, scanning electron microscopy, and Fourier transform infrared spectroscopy, which confirmed the phase purity of HAp and Sr/Fe co-substitution into its lattice. The Vickers hardness measurement, high blood compatibility (less than 5% hemolysis), and ability to support the adhesion, proliferation, and osteogenic differentiation of human mesenchymal stem cells suggest the suitability of Sr/Fe:HAp bioceramics for bone implant applications. The physicochemical analysis revealed that the developed Sr/Fe:HAp bioceramics exhibited a polyphasic nature (HAp and ßTCP) with almost identical structural morphology having a particle size less than 0.8 µm. The dielectric constant (ε') and dielectric loss (ε″) were potentially affected by the incorporated foreign ions together with the polyphasic nature of the material. The Sr/Fe co-substituted samples demonstrated extended drug (5-fluorouracil and amoxicillin) release profiles at the pH of physiological medium. The multifunctional properties of the developed HAp bioceramics enabled them to be an auspicious candidate for potential biomedical applications, including targeted drug-delivery applications, heating mediator in hyperthermia, and bone tissue repair implants.


Subject(s)
Durapatite , Tissue Engineering , Bone Regeneration , Bone and Bones , Humans , Osteogenesis
16.
Mater Sci Eng C Mater Biol Appl ; 104: 109904, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31499954

ABSTRACT

Brain extracellular matrix (ECM) is complex, heterogeneous and often poorly replicated in traditional 2D cell culture systems. The development of more physiologically relevant 3D cell models capable of emulating the native ECM is of paramount importance for the study of human induced pluripotent stem cell (iPSC)-derived neurons. Due to its structural similarity with hyaluronic acid, a primary component of brain ECM, alginate is a potential biomaterial for 3D cell culture systems. However, a lack of cell adhesion motifs within the chemical structure of alginate has limited its application in neural culture systems. This study presents a simple and accessible method of incorporating collagen fibrils into an alginate hydrogel by physical mixing and controlled gelation under physiological conditions and tests the hypothesis that such a substrate could influence the behaviour of human neurons in 3D culture. Regulation of the gelation process enabled the penetration of collagen fibrils throughout the hydrogel structure as demonstrated by transmission electron microscopy. Encapsulated human iPSC-derived neurons adhered to the blended hydrogel as evidenced by the increased expression of α1, α2 and ß1 integrins. Furthermore, immunofluorescence microscopy revealed that encapsulated neurons formed complex neural networks and matured into branched neurons expressing synaptophysin, a key protein involved in neurotransmission, along the neurites. Mechanical tuning of the hydrogel stiffness by modulation of the alginate ionic crosslinker concentration also influenced neuron-specific gene expression. In conclusion, we have shown that by tuning the physicochemical properties of the alginate/collagen blend it is possible to create different ECM-like microenvironments where complex mechanisms underpinning the growth and development of human neurons can be simulated and systematically investigated.


Subject(s)
Alginates/pharmacology , Cell Differentiation/drug effects , Collagen/pharmacology , Hydrogels/pharmacology , Neurogenesis/drug effects , Neurons/cytology , Cell Adhesion/drug effects , Cell Shape/drug effects , Cell-Matrix Junctions/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Neurons/drug effects , Phenotype , Rheology
17.
Adv Mater ; 31(37): e1902725, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31343084

ABSTRACT

Additive manufacturing (AM) technologies appear as a paradigm for scalable manufacture of electrochemical energy storage (EES) devices, where complex 3D architectures are typically required but are hard to achieve using conventional techniques. The combination of these technologies and innovative material formulations that maximize surface area accessibility and ion transport within electrodes while minimizing space are of growing interest. Herein, aqueous inks composed of atomically thin (1-3 nm) 2D Ti3 C2 Tx with large lateral size of about 8 µm possessing ideal viscoelastic properties are formulated for extrusion-based 3D printing of freestanding, high specific surface area architectures to determine the viability of manufacturing energy storage devices. The 3D-printed device achieves a high areal capacitance of 2.1 F cm-2 at 1.7 mA cm-2 and a gravimetric capacitance of 242.5 F g-1 at 0.2 A g-1 with a retention of above 90% capacitance for 10 000 cycles. It also exhibits a high energy density of 0.0244 mWh cm-2 and a power density of 0.64 mW cm-2 at 4.3 mA cm-2 . It is anticipated that the sustainable printing and design approach developed in this work can be applied to fabricate high-performance bespoke multiscale and multidimensional architectures of functional and structural materials for integrated devices in various applications.

18.
Trends Biotechnol ; 36(4): 457-472, 2018 04.
Article in English | MEDLINE | ID: mdl-29422410

ABSTRACT

Neurovascular dysfunction is a central process in the pathogenesis of stroke and most neurodegenerative diseases, including Alzheimer's disease. The multicellular neurovascular unit (NVU) combines the neural, vascular and extracellular matrix (ECM) components in an important interface whose correct functioning is critical to maintain brain health. Tissue engineering is now offering new tools and insights to advance our understanding of NVU function. Here, we review how the use of novel biomaterials to mimic the mechanical and functional cues of the ECM, coupled with precisely layered deposition of the different cells of the NVU through 3D bioprinting, is revolutionising the study of neurovascular function and dysfunction.


Subject(s)
Biocompatible Materials/chemistry , Bioprinting , Printing, Three-Dimensional , Tissue Engineering , Animals , Brain Diseases/therapy , Extracellular Matrix/chemistry , Humans , Hydrogels/chemistry , Models, Animal , Neuroglia/chemistry , Neurons/chemistry
19.
J Appl Biomater Funct Mater ; 15(3): e185-e195, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28623631

ABSTRACT

BACKGROUND: In recent years, the tissue engineering (TE) field has significantly benefited from advanced techniques such as additive manufacturing (AM), for the design of customized 3D scaffolds with the aim of guided tissue repair. Among the wide range of materials available to biomanufacture 3D scaffolds, poly(ε-caprolactone) (PCL) clearly arises as the synthetic polymer with the greatest potential, due to its unique properties - namely, biocompatibility, biodegradability, thermal and chemical stability and processability. This study aimed for the first time to investigate the effect of pore geometry on the in vitro enzymatic chain cleavage mechanism of PCL scaffolds manufactured by the AM extrusion process. METHODS: Methods: Morphological properties of 3D printed PCL scaffolds before and after degradation were evaluated using Scanning Electron Microscopy (SEM) and micro-computed tomography (µ-CT). Differential Scanning Calorimetry (DSC) was employed to determine possible variations in the crystallinity of the scaffolds during the degradation period. The molecular weight was assessed using Size Exclusion Chromatography (SEC) while the mechanical properties were investigated under static compression conditions. RESULTS: Morphological results suggested a uniform reduction of filament diameter, while increasing the scaffolds' porosity. DSC analysis revealed and increment in the crystallinity degree while the molecular weight, evaluated through SEC, remained almost constant during the incubation period (25 days). Mechanical analysis highlighted a decrease in the compressive modulus and maximum stress over time, probably related to the significant weight loss of the scaffolds. CONCLUSIONS: All of these results suggest that PCL scaffolds undergo enzymatic degradation through a surface erosion mechanism, which leads to significant variations in mechanical, physical and chemical properties, but which has little influence on pore geometry.


Subject(s)
Enzymes/chemistry , Polyesters/chemistry , Tissue Engineering , Tissue Scaffolds , Materials Testing , Microscopy, Electron, Scanning , X-Ray Microtomography
20.
Proc Inst Mech Eng H ; 231(6): 555-564, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28056713

ABSTRACT

Bone tissue engineering is strongly dependent on the use of three-dimensional scaffolds that can act as templates to accommodate cells and support tissue ingrowth. Despite its wide application in tissue engineering research, polycaprolactone presents a very limited ability to induce adhesion, proliferation and osteogenic cell differentiation. To overcome some of these limitations, different calcium phosphates, such as hydroxyapatite and tricalcium phosphate, have been employed with relative success. This work investigates the influence of nano-hydroxyapatite and micro-hydroxyapatite (nHA and mHA, respectively) particles on the in vitro biomechanical performance of polycaprolactone/hydroxyapatite scaffolds. Morphological analysis performed with scanning electron microscopy allowed us to confirm the production of polycaprolactone/hydroxyapatite constructs with square interconnected pores of approximately 350 µm and to assess the distribution of hydroxyapatite particles within the polymer matrix. Compression mechanical tests showed an increase in polycaprolactone compressive modulus ( E) from 105.5 ± 11.2 to 138.8 ± 12.9 MPa (PCL_nHA) and 217.2 ± 21.8 MPa (PCL_mHA). In comparison to PCL_mHA scaffolds, the addition of nano-hydroxyapatite enhanced the adhesion and viability of human mesenchymal stem cells as confirmed by Alamar Blue assay. In addition, after 14 days of incubation, PCL_nHA scaffolds showed higher levels of alkaline phosphatase activity compared to polycaprolactone or PCL_mHA structures.


Subject(s)
Cell Adhesion/drug effects , Cell Differentiation/drug effects , Durapatite/pharmacology , Mesenchymal Stem Cells/cytology , Nanostructures/chemistry , Printing, Three-Dimensional , Tissue Scaffolds/chemistry , Alkaline Phosphatase/metabolism , Cell Survival/drug effects , Durapatite/chemistry , Humans , Mechanical Phenomena , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Tissue Engineering
SELECTION OF CITATIONS
SEARCH DETAIL
...