Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Sci Rep ; 14(1): 11003, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38744985

ABSTRACT

The future of organ and tissue biofabrication strongly relies on 3D bioprinting technologies. However, maintaining sterility remains a critical issue regardless of the technology used. This challenge becomes even more pronounced when the volume of bioprinted objects approaches organ dimensions. Here, we introduce a novel device called the Flexible Unique Generator Unit (FUGU), which is a unique combination of flexible silicone membranes and solid components made of stainless steel. Alternatively, the solid components can also be made of 3D printed medical-grade polycarbonate. The FUGU is designed to support micro-extrusion needle insertion and removal, internal volume adjustment, and fluid management. The FUGU was assessed in various environments, ranging from custom-built basic cartesian to sophisticated 6-axis robotic arm bioprinters, demonstrating its compatibility, flexibility, and universality across different bioprinting platforms. Sterility assays conducted under various infection scenarios highlight the FUGU's ability to physically protect the internal volume against contaminations, thereby ensuring the integrity of the bioprinted constructs. The FUGU also enabled bioprinting and cultivation of a 14.5 cm3 human colorectal cancer tissue model within a completely confined and sterile environment, while allowing for the exchange of gases with the external environment. This FUGU system represents a significant advancement in 3D bioprinting and biofabrication, paving the path toward the sterile production of implantable tissues and organs.


Subject(s)
Bioprinting , Bioreactors , Printing, Three-Dimensional , Bioprinting/methods , Humans , Tissue Engineering/methods , Sterilization , Tissue Scaffolds
2.
Hand Surg Rehabil ; : 101709, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38685316

ABSTRACT

OBJECTIVES: Surgery for congenital malformation of the hand is complex and protocols are not available. Simulation could help optimize results. The objective of the present study was to design, produce and assess a 3D-printed anatomical support, to improve success in rare and complex surgeries of the hand. MATERIAL AND METHODS: We acquired MRI imaging of the right hand of a 30 year-old subject, then analyzed and split the various skin layers for segmentation. Thus we created the prototype of a healthy hand, using 3D multi-material and silicone printing devices, and drew up a printing protocol suitable for all patients. We printed a base comprising bones, muscles and tendons, with a multi-material 3D printer, then used a 3D silicone printer for skin and subcutaneous fatty cell tissues in a glove-like shape. To evaluate the characteristics of the prototype, we performed a series of dissections on the synthetic hand and on a cadaveric hand in the anatomy lab, comparing realism, ease of handling and the final result of the two supports, and evaluated their respective advantages in surgical and training contexts. A grading form was given to each surgeon to establish a global score. RESULTS: This evaluation highlighted the positive and negative features of the model. The model avoided intrinsic problems of cadavers, such as muscle rigidity or tissue fragility and atrophy, and enables the anatomy of a specific patient to be rigorously respected. On the other hand, vascular and nervous networks, with their potential anatomical variants, are lacking. This preliminary phase highlighted the advantages and inconveniences of the prototype, to optimize the design and printing of future models. It is an indispensable prerequisite before performing studies in eligible pediatric patients with congenital hand malformation. CONCLUSION: The validation of 3D-printed anatomical model of a human hand opens a large field of applications in the area of preoperative surgical planning. The postoperative esthetic and functional benefit of such pre-intervention supports in complex surgery needs assessing.

3.
Article in English | MEDLINE | ID: mdl-38411533

ABSTRACT

The repair of nasal septal cartilage is a key challenge in cosmetic and functional surgery of the nose, as it determines its shape and its respiratory function. Supporting the dorsum of the nose is essential for both the prevention of nasal obstruction and the restoration of the nose structure. Most surgical procedures to repair or modify the nasal septum focus on restoring the external aspect of the nose by placing a graft under the skin, without considering respiratory concerns. Tissue engineering offers a more satisfactory approach, in which both the structural and biological roles of the nose are restored. To achieve this goal, nasal cartilage engineering research has led to the development of scaffolds capable of accommodating cartilaginous extracellular matrix-producing cells, possessing mechanical properties close to those of the nasal septum, and retaining their structure after implantation in vivo. The combination of a non-resorbable core structure with suitable mechanical properties and a biocompatible hydrogel loaded with autologous chondrocytes or mesenchymal stem cells is a promising strategy. However, the stability and immunotolerance of these implants are crucial parameters to be monitored over the long term after in vivo implantation, to definitively assess the success of nasal cartilage tissue engineering. Here, we review the tissue engineering methods to repair nasal cartilage, focusing on the type and mechanical characteristics of the biomaterials; cell and implantation strategy; and the outcome with regard to cartilage repair.

4.
Tissue Eng Part A ; 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37885209

ABSTRACT

The bioextrusion of mesenchymal stromal cells (MSCs) directly seeded in a bioink enables the production of three-dimensional (3D) constructs, promoting their chondrogenic differentiation. Our study aimed to evaluate the effect of different type I collagen concentrations in the bioink on MSCs' chondrogenic differentiation. We printed 3D constructs using an alginate, gelatin, and fibrinogen-based bioink cellularized with MSCs, with four different quantities of type I collagen addition (0.0, 0.5, 1.0, and 5.0 mg per bioink syringe). We assessed the influence of the bioprinting process, the bioink composition, and the growth factor (TGF-ꞵ1) on the MSCs' survival rate. We confirmed the biocompatibility of the process and the bioinks' cytocompatibility. We evaluated the chondrogenic effects of TGF-ꞵ1 and collagen addition on the MSCs' chondrogenic properties through macroscopic observation, shrinking ratio, reverse transcription polymerase chain reaction, glycosaminoglycan synthesis, histology, and type II collagen immunohistochemistry. The bioink containing 0.5 mg of collagen produces the richest hyaline-like extracellular matrix, presenting itself as a promising tool to recreate the superficial layer of hyaline cartilage. The bioink containing 5.0 mg of collagen enhances the synthesis of a calcified matrix, making it a good candidate for mimicking the calcified cartilaginous layer. Type I collagen thus allows the dose-dependent design of specific hyaline cartilage layers.

5.
Biotechnol Adv ; 68: 108211, 2023 11.
Article in English | MEDLINE | ID: mdl-37463610

ABSTRACT

Various research fields use the transfection of mammalian cells with genetic material to induce the expression of a target transgene or gene silencing. It is a tool widely used in biological research, bioproduction, and therapy. Current transfection protocols are usually performed on 2D adherent cells or suspension cultures. The important rise of new gene therapies and regenerative medicine in the last decade raises the need for new tools to empower the in situ transfection of tissues and 3D cell cultures. This review will present novel in situ transfection methods based on a chemical or physical non-viral transfection of cells in tissues and 3D cultures, discuss the advantages and remaining gaps, and propose future developments and applications.


Subject(s)
Genetic Therapy , Tissue Engineering , Animals , Tissue Engineering/methods , Transfection , Transgenes , Cell Culture Techniques, Three Dimensional , Mammals
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.
J Mech Behav Biomed Mater ; 134: 105365, 2022 10.
Article in English | MEDLINE | ID: mdl-35863297

ABSTRACT

Contraction assay based on surface measurement have been widely used to evaluate cell contractility in 3D models. This method is straightforward and requires no specific equipment, but it does not provide quantitative data about contraction forces generated by cells. We expanded this method with a new biomechanical model, based on the work-energy theorem, to provide non-destructive longitudinal monitoring of contraction forces generated by cells in 3D. We applied this method on hydrogels seeded with either fibroblasts or osteoblasts. Hydrogel mechanical characteristics were modulated to enhance (condition HCAHigh: hydrogel contraction assay high contraction) or limit (condition HCALow: hydrogel contraction assay low contraction) cell contractile behaviors. Macroscopic measures were further correlated with cell contractile behavior and descriptive analysis of their physiology in response to different mechanical environments. Fibroblasts and osteoblasts contracted their matrix up to 47% and 77% respectively. Contraction stress peaked at day 5 with 1.1 10-14 Pa for fibroblasts and 3.5 10-14 Pa for osteoblasts, which correlated with cell attachment and spreading. Negligible contraction was seen in HCALow. Both fibroblasts and osteoblasts expressed α-SMA contractile fibers in HCAHigh and HCALow. Failure to contract HCALow was attributed to increased cross-linking and resistance to proteolytic degradation of the hydrogel.


Subject(s)
Hydrogels , Mechanical Phenomena , Fibroblasts , Muscle Contraction
8.
Mol Ecol Resour ; 22(6): 2158-2170, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35218316

ABSTRACT

The study of environmental DNA (eDNA) released by aquatic organisms in their habitat offers a fast, noninvasive and sensitive approach to monitor their presence. Common eDNA sampling methods such as water filtration and DNA precipitation are time-consuming, require difficult-to-handle equipment and partially integrate eDNA signals. To overcome these limitations, we created the first proof of concept of a passive, 3D-printed and easy-to-use eDNA sampler. We designed the samplers from hydroxyapatite (HAp samplers), a natural mineral with a high DNA adsorption capacity. The porous structure and shape of the samplers were designed to optimize DNA adsorption and facilitate their handling in the laboratory and in the field. Here we show that HAp samplers can efficiently collect genomic DNA in controlled set-ups, but can also collect animal eDNA under controlled and natural conditions with yields similar to conventional methods. However, we also observed large variations in the amount of DNA collected even under controlled conditions. A better understanding of the DNA-hydroxyapatite interactions on the surface of the samplers is now necessary to optimize eDNA adsorption and to allow the development of a reliable, easy-to-use and reusable eDNA sampling tool.


Subject(s)
DNA, Environmental , Animals , DNA/genetics , Durapatite , Environmental Monitoring/methods , Printing, Three-Dimensional
9.
Plast Reconstr Surg Glob Open ; 10(1): e4056, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35186622

ABSTRACT

Intraoperative three-dimensional fabrication of living tissues could be the next biomedical revolution in patient treatment. APPROACH: We developed a surgery-ready robotic three-dimensional bioprinter and demonstrated that a bioprinting procedure using medical grade hydrogel could be performed using a 6-axis robotic arm in vivo for treating burn injuries. RESULTS: We conducted a pilot swine animal study on a deep third-degree severe burn model. We observed that the use of cell-laden bioink as treatment substantially affects skin regeneration, producing in situ fibroblast growth factor and vascular endothelial growth factor, necessary for tissue regeneration and re-epidermalization of the wound. CONCLUSIONS: We described an animal study of intraoperative three-dimensional bioprinting living tissue. This emerging technology brings the first proof of in vivo skin printing feasibility using a surgery-ready robotic arm-based bioprinter. Our positive outcome in skin regeneration, joined with this procedure's feasibility, allow us to envision the possibility of using this innovative approach in a human clinical trial in the near future.

10.
Cell Tissue Bank ; 23(3): 417-440, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35000046

ABSTRACT

The application of 3D printing technologies fields for biological tissues, organs, and cells in the context of medical and biotechnology applications requires a significant amount of innovation in a narrow printability range. 3D bioprinting is one such way of addressing critical design challenges in tissue engineering. In a more general sense, 3D printing has become essential in customized implant designing, faithful reproduction of microenvironmental niches, sustainable development of implants, in the capacity to address issues of effective cellular integration, and long-term stability of the cellular constructs in tissue engineering. This review covers various aspects of 3D bioprinting, describes the current state-of-the-art solutions for all aforementioned critical issues, and includes various illustrative representations of technologies supporting the development of phases of 3D bioprinting. It also demonstrates several bio-inks and their properties crucial for being used for 3D printing applications. The review focus on bringing together different examples and current trends in tissue engineering applications, including bone, cartilage, muscles, neuron, skin, esophagus, trachea, tympanic membrane, cornea, blood vessel, immune system, and tumor models utilizing 3D printing technology and to provide an outlook of the future potentials and barriers.


Subject(s)
Bioprinting , Bone and Bones , Ink , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds
11.
Front Bioeng Biotechnol ; 9: 721843, 2021.
Article in English | MEDLINE | ID: mdl-34671597

ABSTRACT

Since the emergence of regenerative medicine and tissue engineering more than half a century ago, one obstacle has persisted: the in vitro creation of large-scale vascular tissue (>1 cm3) to meet the clinical needs of viable tissue grafts but also for biological research applications. Considerable advancements in biofabrication have been made since Weinberg and Bell, in 1986, created the first blood vessel from collagen, endothelial cells, smooth muscle cells and fibroblasts. The synergistic combination of advances in fabrication methods, availability of cell source, biomaterials formulation and vascular tissue development, promises new strategies for the creation of autologous blood vessels, recapitulating biological functions, structural functions, but also the mechanical functions of a native blood vessel. In this review, the main technological advancements in bio-fabrication are discussed with a particular highlights on 3D bioprinting technologies. The choice of the main biomaterials and cell sources, the use of dynamic maturation systems such as bioreactors and the associated clinical trials will be detailed. The remaining challenges in this complex engineering field will finally be discussed.

12.
J Mech Behav Biomed Mater ; 122: 104649, 2021 10.
Article in English | MEDLINE | ID: mdl-34218017

ABSTRACT

Modern 3D printing of implantable devices provides an important opportunity for the development of personalized implants with good anatomical fit. Nevertheless, 3D printing of silicone has been challenging and the recent advances in technology are provided by the systems which can print medical grade silicone via extrusion. However, the potential impacts of the 3D printing process of silicone on its biomechanical properties has not been studied in sufficient detail. Therefore, the present study compares 3D printed and moulded silicone structures for their cytotoxicity, surface roughness, biomechanical properties, and in vivo tissue reaction. The 3D printing process creates increased nanoscale roughness and noticeably changes microscale topography. Neither the presence of these features nor the differences in processes were found to result in an increase in cytotoxicity or tissue reaction for 3D printed structures, exhibiting limited inflammatory reaction and cell viability above the threshold values. On the contrary, the biomechanical properties have demonstrated significant differences in static and dynamic conditions, and in thermal expansion. Our results demonstrate that 3D printing can be used for establishing a better biomechanical microenvironment for the surrounding tissue of the implant particularly for fragile soft tissue like epithelial mucosa without having any negative effect on the cytotoxicity or in vivo reaction to silicone. For engineering of the implants, however, one must consider the differences in mechanical properties to result in correct and personalized geometry and proper physical interaction with tissues.


Subject(s)
Printing, Three-Dimensional , Silicones , Prostheses and Implants
13.
Langmuir ; 37(14): 4154-4162, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33787263

ABSTRACT

Freeform reversible embedding of suspended hydrogel (FRESH) is an additive manufacturing technique enabling the 3D printing of soft materials with low or no yield stress. The printed material is embedded during the process until its solidification. From the literature, FRESH abilities are self-healing, reusability, suspending, thermal stability, and high-precision printing. This study proposes a new support hydrogel bath formulation for FRESH 3D printing. To do so, a poloxamer micellar thermoreversible hydrogel is tuned through the addition of poly(ethylene glycol) (PEG) to adapt rheological properties. PEG macromolecules interact with poly(ethylene oxide) blocks of poloxamer and favor micelle dehydration, and then decreasing the gelation temperature, the yield stress, and the viscosity. Parameters such as the Oldroyd number and the Rayleigh-Plateau instability, both dependent on yield stress, were studied to determine their impact on the FRESH 3D printing resolution and accuracy. It was found that print accuracy of embedded parts increases with increasing yield stress but then the self-healing property gets limited, leading to crevasse formation. The usefulness of this approach is distinctly demonstrated through a six-axis printing of a highly complex silicone anatomical model. Printing fidelity of 96.0 ± 3.58% (5-40 mm printed parts) is thus achieved using the newly formulated FRESH material, while only 56.0 ± 0.76% fidelity is obtained using the standard formulation. The present study thus showed that complex FRESH 3D printing of soft materials is possible in this tunable hydrogel and that parts can be manufactured on an industrial scale, thanks to the reusability of the support bath.

14.
Talanta ; 222: 121578, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33167265

ABSTRACT

This work reports the development of an electrical and optical biosensing for label-free detection of Aflatoxin B1 (AFB1) using gold (Au) nanobipyramids (NBPs). AuNBPs were synthesized through a two-step seed-mediated growth process followed by an exchange of capping agent from surfactant to lipoic acid. Pure and monodispersed AuNBPs of 70 nm base length were obtained and deposited on indium tin oxide (ITO)-coated glass substrate modified with self-assembled (3-Aminopropyl) triethoxysilane (APTES) film. The characterization of the obtained surfaces using spectroscopy, microscopy and diffractometry confirms the formation of AuNBPs, the conjugation to ITO electrode substrate and the immobilization of anti-AFB1 antibodies. AuNBPs modified ITO substrates were used for both electrochemical and Surface Plasmon Resonance biosensing studies. Localized Surface Plasmon Resonance (LSPR) local field enhancement was demonstrated. SPR based AFB1 detection was found to be linear in the 0.1-500 nM range with a limit of detection of 0.4 nM, whereas, impedimetric AFB1 detection was shown to be linear in the 0.1-25 nM range with a limit of detection of 0.1 nM. The practical utility of the impedimetric sensor was tested in spiked maize samples and 95-100% recovery percentage was found together with low relative standard deviation, proof of the robustness of this AFB1 sensor.


Subject(s)
Biosensing Techniques , Gold , Aflatoxin B1/analysis , Electrodes , Limit of Detection , Surface Plasmon Resonance
15.
J Tissue Eng Regen Med ; 15(1): 37-48, 2021 01.
Article in English | MEDLINE | ID: mdl-33170542

ABSTRACT

Clinical grade cultured epithelial autograft (CEA) are routinely used to treat burns covering more than 60% of the total body surface area. However, although the epidermis may be efficiently repaired by CEA, the dermal layer, which is not spared in deep burns, requires additional treatment strategies. Our aim is to develop an innovative method of skin regeneration based on in situ 3D bioprinting of freshly isolated autologous skin cells. We describe herein bioink formulation and cell preparation steps together with experimental data validating a straightforward enzyme-free protocol of skin cell extraction. This procedure complies with both the specific needs of 3D bioprinting process and the stringent rules of good manufacturing practices. This mechanical extraction protocol, starting from human skin biopsies, allows harvesting a sufficient amount of both viable and growing keratinocytes and fibroblasts. We demonstrated that a dermis may be reconstituted in vitro starting from a medical grade bioink and mechanically extracted skin cells. In these experiments, proliferation of the extracted cells can be observed over the first 21 days period after 3D bioprinting and the analysis of type I collagen exhibited a de novo production of extracellular matrix proteins. Finally, in vivo experiments in a murine model of severe burn provided evidences that a topical application of our medical grade bioink was feasible and well-tolerated. Overall, these results represent a valuable groundwork for the design of future 3D bioprinting tissue engineering strategies aimed at treating, in a single intraoperative step, patients suffering from extended severe burns.


Subject(s)
Bioprinting , Burns , Cells, Immobilized , Fibroblasts , Keratinocytes , Printing, Three-Dimensional , Tissue Scaffolds/chemistry , Animals , Burns/metabolism , Burns/pathology , Burns/therapy , Cells, Immobilized/metabolism , Cells, Immobilized/pathology , Cells, Immobilized/transplantation , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/transplantation , Heterografts , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/transplantation , Mice , Mice, Inbred BALB C , Mice, Nude
16.
Anal Bioanal Chem ; 412(25): 7029-7041, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32797305

ABSTRACT

Decoration of graphene quantum dots (GQDs) on molybdenum disulfide (MoS2) nanosheets serves as an active electrode material which enhances the electrochemical performance of the analyte detection system. Herein, ionic surfactant cetyltrimethylammonium bromide (CTAB)-exfoliated MoS2 nanosheets decorated with GQD material are used to construct an electrochemical biosensor for aflatoxin B1 (AFB1) detection. An antibody of AFB1 (aAFB1) was immobilized on the electrophoretically deposited MoS2@GQDs film on the indium tin oxide (ITO)-coated glass surface using a crosslinker for the fabrication of the biosensor. The immunosensing study investigated by the electrochemical method revealed a signal response in the range of 0.1 to 3.0 ng/mL AFB1 concentration with a detection limit of 0.09 ng/mL. Also, electrochemical parameters such as diffusion coefficient and heterogeneous electron transfer (HET) were calculated and found to be 1.67 × 10-5 cm2/s and 2 × 10-5 cm/s, respectively. The effective conjugation of MoS2@GQDs that provides abundant exposed edge sites, large surface area, improved electrical conductivity, and electrocatalytic activity has led to an excellent biosensing performance with enhanced electrochemical parameters. Validation of the fabricated immunosensor was performed in a spiked maize sample, and a good percentage of recoveries within an acceptable range were obtained (80.2 to 98.3%).Graphical abstract.


Subject(s)
Aflatoxin B1/analysis , Graphite/chemistry , Nanostructures/chemistry , Quantum Dots/chemistry , Aflatoxin B1/immunology , Biosensing Techniques , Electrochemical Techniques/methods , Food Contamination/analysis , Limit of Detection , Reproducibility of Results
17.
Stem Cells Int ; 2020: 2487072, 2020.
Article in English | MEDLINE | ID: mdl-32399041

ABSTRACT

3D bioprinting offers interesting opportunities for 3D tissue printing by providing living cells with appropriate scaffolds with a dedicated structure. Biological advances in bioinks are currently promising for cell encapsulation, particularly that of mesenchymal stem cells (MSCs). We present herein the development of cartilage implants by 3D bioprinting that deliver MSCs encapsulated in an original bioink at low concentration. 3D-bioprinted constructs (10 × 10 × 4 mm) were printed using alginate/gelatin/fibrinogen bioink mixed with human bone marrow MSCs. The influence of the bioprinting process and chondrogenic differentiation on MSC metabolism, gene profiles, and extracellular matrix (ECM) production at two different MSC concentrations (1 million or 2 million cells/mL) was assessed on day 28 (D28) by using MTT tests, real-time RT-PCR, and histology and immunohistochemistry, respectively. Then, the effect of the environment (growth factors such as TGF-ß1/3 and/or BMP2 and oxygen tension) on chondrogenicity was evaluated at a 1 M cell/mL concentration on D28 and D56 by measuring mitochondrial activity, chondrogenic gene expression, and the quality of cartilaginous matrix synthesis. We confirmed the safety of bioextrusion and gelation at concentrations of 1 million and 2 million MSC/mL in terms of cellular metabolism. The chondrogenic effect of TGF-ß1 was verified within the substitute on D28 by measuring chondrogenic gene expression and ECM synthesis (glycosaminoglycans and type II collagen) on D28. The 1 M concentration represented the best compromise. We then evaluated the influence of various environmental factors on the substitutes on D28 (differentiation) and D56 (synthesis). Chondrogenic gene expression was maximal on D28 under the influence of TGF-ß1 or TGF-ß3 either alone or in combination with BMP-2. Hypoxia suppressed the expression of hypertrophic and osteogenic genes. ECM synthesis was maximal on D56 for both glycosaminoglycans and type II collagen, particularly in the presence of a combination of TGF-ß1 and BMP-2. Continuous hypoxia did not influence matrix synthesis but significantly reduced the appearance of microcalcifications within the extracellular matrix. The described strategy is very promising for 3D bioprinting by the bioextrusion of an original bioink containing a low concentration of MSCs followed by the culture of the substitutes in hypoxic conditions under the combined influence of TGF-ß1 and BMP-2.

18.
Food Chem ; 307: 125530, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31639579

ABSTRACT

The Surface Plasmon resonance (SPR) based label-free detection of small targeted molecules is a great challenge and require substantial signal amplification for the accurate and precise quantification. The incorporation of noble metal nanoparticles (NPs) like gold (Au) NPs for the fabrication of SPR biosensor has shown remarkable impact both for anchoring the signal amplification and generate plasmonic resonant coupling between NPs and chip surface. In this work, we present comparative studies related to the fabrication of self-assembled monolayer (SAM) and the influence of AuNPs on Au chip for Aflatoxin B1 (AFB1) detection using SPRi apparatus. The SAM Au chip was sequentially modified by EDC-NHS crosslinkers, grafting of protein-A and finally interaction with anti-AFB1 antibodies. Similar multilayer chip surface was prepared using functionalized lipoic acid AuNPs deposited on SAM Au chips followed by in situ activation of functional groups using EDC-NHS crosslinkers, grafting of protein-A and immobilization of anti-AFB1 antibodies. This multilayer functionalized AuNPs modified Au chip was successfully utilized for AFB1 detection ranging from 0.01 to 50 nM with a limit of detection of 0.003 nM. When compared to bare self-assembled Au chip which was shown to exhibit a limit of detection of 0.19 nM and a linear detection ranging from 1 to 50 nM, the AuNPs modified Au chip was proven to clearly be a better analytical tool. Finally, validation of the proposed biosensor was evaluated by spiked wheat samples and average recoveries (93 and 90.1%) were found to be acceptable.


Subject(s)
Aflatoxin B1/analysis , Biosensing Techniques/instrumentation , Metal Nanoparticles/chemistry , Microfluidics , Surface Plasmon Resonance/instrumentation , Antibodies , Gold , Limit of Detection
19.
Biofabrication ; 12(2): 025006, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31578006

ABSTRACT

The mammalian retina contains multiple cellular layers, each carrying out a specific task. Such a controlled organization should be considered as a crucial factor for designing retinal therapies. The maintenance of retinal layered complexity through the use of scaffold-free techniques has recently emerged as a promising approach for clinical ocular tissue engineering. In an attempt to fabricate such layered retinal model, we are proposing herein a unique inkjet bioprinting system applied to the deposition of a photoreceptor cells (PRs) layer on top of a bioprinted retinal pigment epithelium (RPE), in a precise arrangement and without any carrier material. The results showed that, after bioprinting, both RPE and PRs were well positioned in a layered structure and expressed their structural markers, which was further demonstrated by ZO1, MITF, rhodopsin, opsin B, opsin R/G and PNA immunostaining, three days after bioprinting. We also showed that considerable amounts of human vascular endothelial growth factors (hVEGF) were released from the RPE printed layer, which confirmed the formation of a functional RPE monolayer after bioprinting. Microstructures of bioprinted cells as well as phagocytosis of photoreceptor outer segments by apical RPE microvilli were finally established through transmission electron microscopy (TEM) imaging. In summary, using this carrier-free bioprinting method, it was possible to develop a reasonable in vitro retina model for studying some sight-threatening diseases, such as age-related macular degeneration (AMD) and retinitis pigmentosa (RP).


Subject(s)
Bioprinting/methods , Retina/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Bioprinting/instrumentation , Cell Proliferation , Humans , Photoreceptor Cells/cytology , Photoreceptor Cells/metabolism , Printing, Three-Dimensional/instrumentation , Retina/metabolism , Retinal Pigment Epithelium/cytology , Rhodopsin/metabolism , Swine , Tissue Engineering/instrumentation , Vascular Endothelial Growth Factor A/metabolism
20.
Mater Sci Eng C Mater Biol Appl ; 104: 109898, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31499960

ABSTRACT

Surface of the implantable devices is the root cause of several complications such as infections, implant loosening and chronic inflammation. There is an urgent need for multifunctional coatings that can address these shortcomings simultaneously in a manner similar to the structures of extracellular matrix. Herein, we developed a coating system composed of ECM components and a naturally derived polypeptide. The interactions between the coating components create an environment that enables incorporation of an antimicrobial/angiogenic polypeptide. The film composition is based gelatin and hyaluronic acid modified with aldehyde groups (HA-Ald) that can react with poly (arginine) (PAR) through transient interactions. Nanoplasmon measurements demonstrated a significantly higher loading of PAR in films containing HA-Ald with longer retention of PAR in the structure. The presence of PAR not only provides to the film surface antimicrobial (contact-killing) properties but also increased endothelial cell-cell contacts (PECAM) and VEGFA gene expression and secretion by human vascular endothelial cells. This multifunctional coating can be easily applied to surface of implants where it can enact on several problems simultaneously.


Subject(s)
Coated Materials, Biocompatible/pharmacology , Gelatin/pharmacology , Hyaluronic Acid/pharmacology , Peptides/pharmacology , Polymers/pharmacology , Prostheses and Implants , Animals , Anti-Bacterial Agents/pharmacology , Cattle , Extracellular Matrix/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...