Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Clin Med ; 8(10)2019 Oct 02.
Article in English | MEDLINE | ID: mdl-31581732

ABSTRACT

Bioprinting techniques use bioinks made of biocompatible non-living materials and cells to build 3D constructs in a controlled manner and with micrometric resolution. 3D bioprinted structures representative of several human tissues have been recently produced using cells derived by differentiation of induced pluripotent stem cells (iPSCs). Human iPSCs can be differentiated in a wide range of neurons and glia, providing an ideal tool for modeling the human nervous system. Here we report a neural construct generated by 3D bioprinting of cortical neurons and glial precursors derived from human iPSCs. We show that the extrusion-based printing process does not impair cell viability in the short and long term. Bioprinted cells can be further differentiated within the construct and properly express neuronal and astrocytic markers. Functional analysis of 3D bioprinted cells highlights an early stage of maturation and the establishment of early network activity behaviors. This work lays the basis for generating more complex and faithful 3D models of the human nervous systems by bioprinting neural cells derived from iPSCs.

2.
Biofabrication ; 11(4): 044101, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31151123

ABSTRACT

Osteochondral (OC) tissue is a biphasic material comprised of articular cartilage integrated atop subchondral bone. Damage to this tissue is highly problematic, owing to its intrinsic inability to regenerate functional tissue in response to trauma or disease. Further, the function of the tissue is largely conferred by its compartmentalized zonal microstructure and composition. Current clinical treatments fail to regenerate new tissue that recapitulates this zonal structure. Consequently, regenerated tissue often lacks long-term stability. To address this growing problem, we propose the development of tissue engineered biomaterials that mimic the zonal cartilage organization and extracellular matrix composition through the use of a microfluidic printing head bearing a mixing unit and incorporated into an extrusion-based bioprinter. The system is devised so that multiple bioinks can be delivered either individually or at the same time and rapidly mixed to the extrusion head, and finally deposited through a coaxial nozzle. This enables the deposition of either layers or continuous gradients of chemical, mechanical and biological cues and fabrication of scaffolds with very high shape fidelity and cell viability. Using such a system we bioprinted cell-laden hydrogel constructs recapitulating the layered structure of cartilage, namely, hyaline and calcified cartilage. The construct was assembled out of two bioinks specifically formulated to mimic the extracellular matrices present in the targeted tissues and to ensure the desired biological response of human bone marrow-derived mesenchymal stem cells and human articular chondrocytes. Homogeneous and gradient constructs were thoroughly characterized in vitro with respect to long-term cell viability and expression of hyaline and hypertrophic markers by means of real-time quantitative PCR and immunocytochemical staining. After 21 days of in vitro culture, we observed production of zone-specific matrix. The PCR analysis demonstrated upregulated expression of hypertrophic markers in the homogenous equivalent of calcified cartilage but not in the gradient heterogeneous construct. The regenerative potential was assessed in vivo in a rat model. The histological analysis of surgically damaged rat trochlea revealed beneficial effect of the bioprinted scaffolds on regeneration of OC defect when compared to untreated control.


Subject(s)
Bioprinting , Cartilage, Articular/pathology , Hydrogels/pharmacology , Microfluidics/instrumentation , Printing, Three-Dimensional , Regeneration , Animals , Cartilage, Articular/drug effects , Chondrogenesis/drug effects , Humans , Implants, Experimental , Ink , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Rats, Wistar , Regeneration/drug effects
3.
J Exp Clin Cancer Res ; 38(1): 117, 2019 Mar 22.
Article in English | MEDLINE | ID: mdl-30898166

ABSTRACT

Immune checkpoint inhibitor therapy has changed clinical practice for patients with different cancers, since these agents have demonstrated a significant improvement of overall survival and are effective in many patients. However, an intrinsic or acquired resistance frequently occur and biomarkers predictive of responsiveness should help in patient selection and in defining the adequate treatment options. A deep analysis of the complexity of the tumor microenvironment is likely to further advance the field and hopefully identify more effective combined immunotherapeutic strategies. Here we review the current knowledge on tumor microenvironment, focusing on T cells, cancer associated fibroblasts and extracellular matrix. The use of 3D cell culture models to resemble tumor microenvironment landscape and to screen immunomodulatory drugs is also reviewed.


Subject(s)
Models, Biological , Neoplasms/immunology , Spheroids, Cellular/cytology , Cancer-Associated Fibroblasts/immunology , Extracellular Matrix/immunology , Humans , Neoplasms/pathology , Printing, Three-Dimensional , Spheroids, Cellular/pathology , T-Lymphocytes/immunology , Tissue Scaffolds , Tumor Microenvironment
4.
Sci Rep ; 8(1): 17178, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30464219

ABSTRACT

An ideal illumination for light sheet fluorescence microscopy entails both a localized and a propagation invariant optical field. Bessel beams and Airy beams satisfy these conditions, but their non-diffracting feature comes at the cost of the presence of high-energy side lobes that notably degrade the imaging contrast and induce photobleaching. Here, we demonstrate the use of a light droplet illumination whose side lobes are suppressed by interfering Bessel beams of specific k-vectors. Our droplet illumination readily achieves more than 50% extinction of the light distributed across the Bessel side lobes, providing a more efficient energy localization without loss in transverse resolution. In a standard light sheet fluorescence microscope, we demonstrate a two-fold contrast enhancement imaging micron-scale fluorescent beads. Results pave the way to new opportunities for rapid and deep in vivo observations of large-scale biological systems.

5.
Biofabrication ; 11(1): 012001, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30284540

ABSTRACT

Nowadays, 3D bioprinting technologies are rapidly emerging in the field of tissue engineering and regenerative medicine as effective tools enabling the fabrication of advanced tissue constructs that can recapitulate in vitro organ/tissue functions. Selecting the best strategy for bioink deposition is often challenging and time consuming process, as bioink properties-in the first instance, rheological and gelation-strongly influence the suitable paradigms for its deposition. In this short review, we critically discuss one of the available approaches used for bioprinting-namely co-axial wet-spinning extrusion. Such a deposition system, in fact, demonstrated to be promising in terms of printing resolution, shape fidelity and versatility when compared to other methods. An overview of the performances of co-axial technology in the deposition of cellularized hydrogel fibres is discussed, highlighting its main features. Furthermore, we show how this approach allows (i) to decouple the printing accuracy from bioink rheological behaviour-thus notably simplifying the development of new bioinks-and (ii) to build heterogeneous multi-materials and/or multicellular constructs that can better mimic the native tissues when combined with microfluidic systems. Finally, the ongoing challenges and the future perspectives for the ultimate fabrication of functional constructs for advanced research studies are highlighted.


Subject(s)
Bioprinting/trends , Microfluidics/trends , Printing, Three-Dimensional/trends , Animals , Bioprinting/instrumentation , Bioprinting/methods , Humans , Microfluidics/instrumentation , Microfluidics/methods , Printing, Three-Dimensional/instrumentation , Tissue Engineering
6.
Methods Mol Biol ; 1612: 369-380, 2017.
Article in English | MEDLINE | ID: mdl-28634956

ABSTRACT

3D bioprinting is an emerging field that can be described as a robotic additive biofabrication technology that has the potential to build tissues or organs. In general, bioprinting uses a computer-controlled printing device to accurately deposit cells and biomaterials into precise architectures with the goal of creating on demand organized multicellular tissue structures and eventually intra-organ vascular networks. The latter, in turn, will promote the host integration of the engineered tissue/organ in situ once implanted. Existing biofabrication techniques still lay behind this goal. Here, we describe a novel microfluidic printing head-integrated within a custom 3D bioprinter-that allows for the deposition of multimaterial and/or multicellular within a single scaffold by extruding simultaneously different bioinks or by rapidly switching between one bioink and another. The designed bioprinting method effectively moves toward the direction of creating viable tissues and organs for implantation in clinic and research in lab environments.


Subject(s)
Bioprinting/instrumentation , Microfluidics/instrumentation , Printing, Three-Dimensional/instrumentation , Cell Survival , Computer-Aided Design , Human Umbilical Vein Endothelial Cells , Humans , Tissue Engineering/instrumentation , Tissue Scaffolds
7.
Biomaterials ; 110: 45-59, 2016 12.
Article in English | MEDLINE | ID: mdl-27710832

ABSTRACT

Engineering cardiac tissues and organ models remains a great challenge due to the hierarchical structure of the native myocardium. The need of integrating blood vessels brings additional complexity, limiting the available approaches that are suitable to produce integrated cardiovascular organoids. In this work we propose a novel hybrid strategy based on 3D bioprinting, to fabricate endothelialized myocardium. Enabled by the use of our composite bioink, endothelial cells directly bioprinted within microfibrous hydrogel scaffolds gradually migrated towards the peripheries of the microfibers to form a layer of confluent endothelium. Together with controlled anisotropy, this 3D endothelial bed was then seeded with cardiomyocytes to generate aligned myocardium capable of spontaneous and synchronous contraction. We further embedded the organoids into a specially designed microfluidic perfusion bioreactor to complete the endothelialized-myocardium-on-a-chip platform for cardiovascular toxicity evaluation. Finally, we demonstrated that such a technique could be translated to human cardiomyocytes derived from induced pluripotent stem cells to construct endothelialized human myocardium. We believe that our method for generation of endothelialized organoids fabricated through an innovative 3D bioprinting technology may find widespread applications in regenerative medicine, drug screening, and potentially disease modeling.


Subject(s)
Bioprinting/methods , Endothelial Cells , Myocardium , Organoids/growth & development , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Drug Evaluation, Preclinical , Endothelial Cells/chemistry , Endothelial Cells/cytology , Humans , Hydrogels/chemistry , Microfibrils/chemistry , Myocytes, Cardiac/chemistry , Myocytes, Cardiac/metabolism , Organoids/chemistry , Organoids/metabolism , Regenerative Medicine
8.
Mater Sci Eng C Mater Biol Appl ; 62: 668-77, 2016 May.
Article in English | MEDLINE | ID: mdl-26952471

ABSTRACT

In the design of scaffolds for tissue engineering applications, morphological parameters such as pore size, shape, and interconnectivity, as well as transport properties, should always be tailored in view of their clinical application. In this work, we demonstrate that a regular and ordered porous texture is fundamental to achieve an even cell distribution within the scaffold under perfusion seeding. To prove our hypothesis, two sets of alginate scaffolds were fabricated using two different technological approaches of the same method: gas-in-liquid foam templating. In the first one, foam was obtained by insufflating argon in a solution of alginate and a surfactant under stirring. In the second one, foam was generated inside a flow-focusing microfluidic device under highly controlled and reproducible conditions. As a result, in the former case the derived scaffold (GF) was characterized by polydispersed pores and interconnects, while in the latter (µFL), the porous structure was highly regular both with respect to the spatial arrangement of pores and interconnects and their monodispersity. Cell seeding within perfusion bioreactors of the two scaffolds revealed that cell population inside µFL scaffolds was quantitatively higher than in GF. Furthermore, seeding efficiency data for µFL samples were characterized by a lower standard deviation, indicating higher reproducibility among replicates. Finally, these results were validated by simulation of local flow velocity (CFD) inside the scaffolds proving that µFL was around one order of magnitude more permeable than GF.


Subject(s)
Gases/chemistry , Microfluidic Analytical Techniques/methods , Tissue Scaffolds/chemistry , Alginates/chemistry , Bioreactors , Cell Line , Elastic Modulus , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Microfluidic Analytical Techniques/instrumentation , Microscopy, Electron, Scanning , Porosity , Spectroscopy, Fourier Transform Infrared , Surface-Active Agents/chemistry , X-Ray Microtomography
9.
Adv Mater ; 28(4): 677-84, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26606883

ABSTRACT

A novel bioink and a dispensing technique for 3D tissue-engineering applications are presented. The technique incorporates a coaxial extrusion needle using a low-viscosity cell-laden bioink to produce highly defined 3D biostructures. The extrusion system is then coupled to a microfluidic device to control the bioink arrangement deposition, demonstrating the versatility of the bioprinting technique. This low-viscosity cell-responsive bioink promotes cell migration and alignment within each fiber organizing the encapsulated cells.


Subject(s)
Bioprinting/methods , Microfluidic Analytical Techniques/methods , Tissue Scaffolds , Alginates/chemistry , Bioprinting/instrumentation , Cell Survival/radiation effects , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Gelatin/chemistry , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Microfluidic Analytical Techniques/instrumentation , Microscopy, Confocal , Printing, Three-Dimensional , Tissue Engineering , Ultraviolet Rays , Viscosity
10.
ACS Appl Mater Interfaces ; 7(42): 23660-71, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26436204

ABSTRACT

Ordered porous polymeric materials can be engineered to present highly ordered pore arrays and uniform and tunable pore size. These features prompted a number of applications in tissue engineering, generation of meta materials, and separation and purification of biomolecules and cells. Designing new and efficient vistas for the generation of ordered porous materials is an active area of research. Here we investigate the potential of microfluidic foaming within a flow-focusing (FF) geometry in producing 3D regular sponge-like polymeric matrices with tailored morphological and permeability properties. The challenge in using microfluidic systems for the generation of polymeric foams is in the high viscosity of the continuous phase. We demonstrate that as the viscosity of the aqueous solution increases, the accessible range of foam bubble fraction (Φb) and bubble diameter (Db) inside the microfluidic chip tend to narrow progressively. This effect limits the accessible range of geometric properties of the resulting materials. We further show that this problem can be rationally tackled by appropriate choice of the concentration of the polymer. We demonstrate that via such optimization, the microfluidic assisted synthesis of porous materials becomes a facile and versatile tool for generation of porous materials with a wide range of pore size and pore volume. Moreover, we demonstrate that the size of interconnects among pores-for a given value of the gas fraction-can be tailored through the variation of surfactant concentration. This, in turn, affects the permeability of the materials, a factor of key importance in flow-through applications and in tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Lab-On-A-Chip Devices , Surface-Active Agents/chemistry , Tissue Engineering , Permeability/drug effects , Polymers/chemistry , Tissue Scaffolds , Viscosity/drug effects
11.
J Mater Chem B ; 2(16): 2290-2300, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-32261717

ABSTRACT

We demonstrate how to generate highly ordered porous matrices from dextran-methacrylate (DEX-MA) using microfluidics. We use a flow focusing device to inject an aqueous solution of DEX-MA and surfactant to break the flow of an organic solvent (cyclohexane) into monodisperse droplets at a high volume fraction (above 74% v/v) to form an ordered high internal phase emulsion (HIPE). We collect the crystalline HIPE structure and freeze it by gelling. The resulting polyHIPEs are characterized by an interconnected and ordered morphology. The size of pores and interconnects ranges between hundreds and tens of micrometers, respectively. The technique that we describe allows for precise tuning of all the structural parameters of the matrices, including their porosity, the size of the pores and the lumen of interconnects between the pores. The resulting ordered and precisely tailored HIPE gels represent a new class of scaffolds for applications in tissue engineering.

12.
J Mater Chem B ; 2(39): 6779-6791, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-32261874

ABSTRACT

Three dimensional, periodic scaffolds of chitosan-coated alginate are fabricated in a layer-by-layer fashion by rapid prototyping. A novel dispensing system based on two coaxial needles delivers simultaneously alginate and calcium chloride solutions permitting the direct deposition of alginate fibers according to any designed pattern. Coating of the alginate fiber with chitosan and subsequent cross-linking with EDC and genipin assured the endurance of the scaffold in the culture environment for a prolonged period of time. The cross-linking protocol adopted imparted to the scaffold a hierarchical chemical structure as evidenced by Confocal Laser Microscopy and FTIR spectroscopy. The core of the fibers making up the scaffold is represented by alginate chains cross-linked by ester bonds only, the periphery of the fiber is constituted by an inter-polyelectrolyte complex of alginate and chitosan cross-linked in all pair combinations. Fibers belonging to adjacent layers are glued together by the chitosan coating. Mechanical behavior of the scaffolds characterized by different layouts of deposition was determined revealing anisotropic properties. The biocompatibility and capability of the scaffolds to sustain hepatocyte (HepaRG) cultures were demonstrated. Typical hepatic functions such as albumin and urea secretion and induction of CYP3A4 enzyme activity following drug administration were excellent, thus proving the potential of these constructs in monitoring the liver specific function.

13.
Langmuir ; 29(1): 82-91, 2013 Jan 08.
Article in English | MEDLINE | ID: mdl-23214919

ABSTRACT

In this article, we have exploited a microfluidic foaming technique for the generation of highly monodisperse gas-in-liquid bubbles as a templating system for scaffolds characterized by an ordered and homogeneous porous texture. An aqueous poly(vinyl alcohol) (PVA) solution (containing a surfactant) and a gas (argon) are injected simultaneously at constant flow rates in a flow-focusing device (FFD), in which the gas thread breaks up to form monodisperse bubbles. Immediately after its formation, the foam is collected and frozen in liquid nitrogen, freeze-dried, and cross-linked with glutaraldehyde. In order to highlight the superior morphological quality of the obtained porous material, a comparison between this scaffold and another one, also constituted of PVA but obtained with a traditional gas foaming technique, was carried out. Such a comparison has been conducted by analyzing electron microscopy and X-ray microtomographic images of the two samples. It turned out that the microfluidic produced scaffold was characterized by much more uniform porous texture than the gas-foaming one as witnessed by narrower pore size, interconnection, and wall thickness distributions. On the other side, scarce pore interconnectivity, relatively low pore volume, and limited production rate represent, by now, the principal disadvantages of microfluidic foaming as scaffold fabrication method, emphasizing the kind of improvement that this technique needs to undergo.


Subject(s)
Microfluidics , Polyvinyl Alcohol/chemistry , Tissue Scaffolds/chemistry , Gases , Surface-Active Agents/chemistry
14.
J Mater Chem B ; 1(24): 3083-3098, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-32261012

ABSTRACT

Many whole cell-based assays in use today rely on flat, two-dimensional (2D) glass or plastic substrates that may not produce results characteristic of in vivo conditions. In this study, a three-dimensional (3D) cell-based assay scaffold was fabricated using a gas-in-foam templating technique. The scaffold was made of poly(vinyl alcohol), a water-soluble synthetic polymer with excellent film-forming, emulsifying, and biocompatible properties widely used in the biomedical field. The preliminary rheological studies on the solution of PVA and surfactant permitted us to disclose the significant physical parameters that influence the morphology of the ensuing materials. The scaffolds obtained were subjected to detailed analysis by light microscopy, Scanning Electron Microscopy (SEM), computed X-ray microtomography (µCT), infrared spectroscopy, and mechanical testing. Morphological investigations showed that the produced scaffolds are characterised by average void and interconnect diameters lying in the range of 200-300 and 30-150 µm, respectively, suitable for cell infiltration. Two different cross-linking procedures were adopted in order to modulate the mechanical properties of the PVA scaffolds. One made use of a bi-epoxide (PEGDGE), the other was based on glutaraldehyde (GA). The efficiency in terms of cross-linking density of the two procedures resulted in very different mechanical properties. Furthermore, in this article it is demonstrated how PVA foams can be processed into uniform, porous films suitable to be integrated with multi-well 2D culture plates in order to create a 3D analogue. The PEGDGE cross-linked scaffold was tested on C3A cells, a human hepatocyte cell line, representing an appropriate model for liver toxicity studies. Proliferation and cytotoxicity assays indicated good cell viability throughout the culture time, which was also confirmed by SEM analysis. Typical hepatic functions such as albumin and urea production and induction of Cyp3A4 enzyme activity following drug administration were satisfactory, thus proving the efficiency of this construct in maintaining specific liver functions.

SELECTION OF CITATIONS
SEARCH DETAIL