Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 185
Filter
1.
Artif Cells Nanomed Biotechnol ; 52(1): 345-354, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38829715

ABSTRACT

Cell encapsulation into spherical microparticles is a promising bioengineering tool in many fields, including 3D cancer modelling and pre-clinical drug discovery. Cancer microencapsulation models can more accurately reflect the complex solid tumour microenvironment than 2D cell culture and therefore would improve drug discovery efforts. However, these microcapsules, typically in the range of 1 - 5000 µm in diameter, must be carefully designed and amenable to high-throughput production. This review therefore aims to outline important considerations in the design of cancer cell microencapsulation models for drug discovery applications and examine current techniques to produce these. Extrusion (dripping) droplet generation and emulsion-based techniques are highlighted and their suitability to high-throughput drug screening in terms of tumour physiology and ease of scale up is evaluated.


3D microencapsulation models of cancer offer a customisable platform to mimic key aspects of solid tumour physiology in vitro. However, many 3D models do not recapitulate the hypoxic conditions and altered tissue stiffness established in many tumour types and stages. Furthermore, microparticles for cancer cell encapsulation are commonly produced using methods that are not necessarily suitable for scale up to high-throughput manufacturing. This review aims to evaluate current technologies for cancer cell-laden microparticle production with a focus on physiological relevance and scalability. Emerging techniques will then be touched on, for production of uniform microparticles suitable for high-throughput drug discovery applications.


Subject(s)
Drug Discovery , Neoplasms , Humans , Neoplasms/pathology , Neoplasms/drug therapy , Neoplasms/metabolism , Drug Discovery/methods , Cell Encapsulation/methods , Models, Biological , Capsules , Animals , Drug Compounding/methods , Tumor Microenvironment/drug effects
2.
Front Immunol ; 15: 1385022, 2024.
Article in English | MEDLINE | ID: mdl-38694507

ABSTRACT

Liver failure represents a critical medical condition with a traditionally grim prognosis, where treatment options have been notably limited. Historically, liver transplantation has stood as the sole definitive cure, yet the stark disparity between the limited availability of liver donations and the high demand for such organs has significantly hampered its feasibility. This discrepancy has necessitated the exploration of hepatocyte transplantation as a temporary, supportive intervention. In light of this, our review delves into the burgeoning field of hepatocyte transplantation, with a focus on the latest advancements in maintaining hepatocyte function, co-microencapsulation techniques, xenogeneic hepatocyte transplantation, and the selection of materials for microencapsulation. Our examination of hepatocyte microencapsulation research highlights that, to date, most studies have been conducted in vitro or using liver failure mouse models, with a notable paucity of experiments on larger mammals. The functionality of microencapsulated hepatocytes is primarily inferred through indirect measures such as urea and albumin production and the rate of ammonia clearance. Furthermore, research on the mechanisms underlying hepatocyte co-microencapsulation remains limited, and the practicality of xenogeneic hepatocyte transplantation requires further validation. The potential of hepatocyte microencapsulation extends beyond the current scope of application, suggesting a promising horizon for liver failure treatment modalities. Innovations in encapsulation materials and techniques aim to enhance cell viability and function, indicating a need for comprehensive studies that bridge the gap between small-scale laboratory success and clinical applicability. Moreover, the integration of bioengineering and regenerative medicine offers novel pathways to refine hepatocyte transplantation, potentially overcoming the challenges of immune rejection and ensuring the long-term functionality of transplanted cells. In conclusion, while hepatocyte microencapsulation and transplantation herald a new era in liver failure therapy, significant strides must be made to translate these experimental approaches into viable clinical solutions. Future research should aim to expand the experimental models to include larger mammals, thereby providing a clearer understanding of the clinical potential of these therapies. Additionally, a deeper exploration into the mechanisms of cell survival and function within microcapsules, alongside the development of innovative encapsulation materials, will be critical in advancing the field and offering new hope to patients with liver failure.


Subject(s)
Cell Encapsulation , Cell Survival , Hepatocytes , Animals , Humans , Cell Encapsulation/methods , Hepatocytes/transplantation , Hepatocytes/cytology , Liver Failure/therapy , Transplantation, Heterologous
3.
Lab Chip ; 24(11): 2958-2967, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38722067

ABSTRACT

Droplet-based microfluidic technologies for encapsulating single cells have rapidly evolved into powerful tools for single-cell analysis. In conventional passive single-cell encapsulation techniques, because cells arrive randomly at the droplet generation section, to encapsulate only a single cell with high precision, the average number of cells per droplet has to be decreased by reducing the average frequency at which cells arrive relative to the droplet generation rate. Therefore, the encapsulation efficiency for a given droplet generation rate is very low. Additionally, cell sorting operations are required prior to the encapsulation of target cells for specific cell type analysis. To address these challenges, we developed a cell encapsulation technology with a cell sorting function using a microfluidic chip. The microfluidic chip is equipped with an optical detection section to detect the optical information of cells and a sorting section to encapsulate cells into droplets by controlling a piezo element, enabling active encapsulation of only the single target cells. For a particle population including both targeted and non-targeted particles arriving at an average frequency of up to 6000 particles per s, with an average number of particles per droplet of 0.45, our device maintained a high purity above 97.9% for the single-target-particle droplets and achieved an outstanding throughput, encapsulating up to 2900 single target particles per s. The proposed encapsulation technology surpasses the encapsulation efficiency of conventional techniques, provides high efficiency and flexibility for single-cell research, and shows excellent potential for various applications in single-cell analysis.


Subject(s)
Lab-On-A-Chip Devices , Single-Cell Analysis , Single-Cell Analysis/instrumentation , Humans , Microfluidic Analytical Techniques/instrumentation , Equipment Design , High-Throughput Screening Assays/instrumentation , Animals , Cell Encapsulation/methods , Cell Encapsulation/instrumentation
4.
Carbohydr Polym ; 338: 122204, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38763712

ABSTRACT

This study presents the development and characterization of a novel double-network self-healing hydrogel based on N-carboxyethyl chitosan (CEC) and oxidized dextran (OD) with the incorporation of crosslinked collagen (CEC-OD/COL-GP) to enhance its biological and physicochemical properties. The hydrogel formed via dynamic imine bond formation exhibited efficient self-healing within 30 min, and a compressive modulus recovery of 92 % within 2 h. In addition to its self-healing ability, CEC-OD/COL-GP possesses unique physicochemical characteristics including transparency, injectability, and adhesiveness to various substrates and tissues. Cell encapsulation studies confirmed the biocompatibility and suitability of the hydrogel as a cell-culture scaffold, with the presence of a collagen network that enhances cell adhesion, spreading, long-term cell viability, and proliferation. Leveraging their unique properties, we engineered assemblies of self-healing hydrogel modules for controlled spatiotemporal drug delivery and constructed co-culture models that simulate angiogenesis in tumor microenvironments. Overall, the CEC-OD/COL-GP hydrogel is a versatile and promising material for biomedical applications, offering a bottom-up approach for constructing complex structures with self-healing capabilities, controlled drug release, and support for diverse cell types in 3D environments. This hydrogel platform has considerable potential for advancements in tissue engineering and therapeutic interventions.


Subject(s)
Cell Adhesion , Chitosan , Dextrans , Hydrogels , Hydrogels/chemistry , Hydrogels/pharmacology , Chitosan/chemistry , Dextrans/chemistry , Humans , Cell Adhesion/drug effects , Cell Survival/drug effects , Collagen/chemistry , Animals , Drug Liberation , Cell Proliferation/drug effects , Cell Encapsulation/methods , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Mice , Biomimetics/methods , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Tissue Scaffolds/chemistry
5.
Compr Rev Food Sci Food Saf ; 23(3): e13322, 2024 05.
Article in English | MEDLINE | ID: mdl-38597567

ABSTRACT

Adequate intake of live probiotics is beneficial to human health and wellbeing because they can help treat or prevent a variety of health conditions. However, the viability of probiotics is reduced by the harsh environments they experience during passage through the human gastrointestinal tract (GIT). Consequently, the oral delivery of viable probiotics is a significant challenge. Probiotic encapsulation provides a potential solution to this problem. However, the production methods used to create conventional encapsulation technologies often damage probiotics. Moreover, the delivery systems produced often do not have the required physicochemical attributes or robustness for food applications. Single-cell encapsulation is based on forming a protective coating around a single probiotic cell. These coatings may be biofilms or biopolymer layers designed to protect the probiotic from the harsh gastrointestinal environment, enhance their colonization, and introduce additional beneficial functions. This article reviews the factors affecting the oral delivery of probiotics, analyses the shortcomings of existing encapsulation technologies, and highlights the potential advantages of single-cell encapsulation. It also reviews the various approaches available for single-cell encapsulation of probiotics, including their implementation and the characteristics of the delivery systems they produce. In addition, the mechanisms by which single-cell encapsulation can improve the oral bioavailability and health benefits of probiotics are described. Moreover, the benefits, limitations, and safety issues of probiotic single-cell encapsulation technology for applications in food and beverages are analyzed. Finally, future directions and potential challenges to the widespread adoption of single-cell encapsulation of probiotics are highlighted.


Subject(s)
Cell Encapsulation , Probiotics , Humans , Gastrointestinal Tract , Biofilms
6.
J Mater Sci Mater Med ; 35(1): 19, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38526655

ABSTRACT

The efficacy of stem-cell therapy depends on the ability of the transplanted cells to escape early immunological reactions and to be retained at the site of transplantation. The use of tissue engineering scaffolds or injectable biomaterials as carriers has been proposed, but they still present limitations linked to a reliable manufacturing process, surgical practice and clinical outcomes. Alginate microbeads are potential candidates for the encapsulation of mesenchymal stromal cells with the aim of providing a delivery carrier suitable for minimally-invasive and scaffold-free transplantation, tissue-adhesive properties and protection from the immune response. However, the formation of stable microbeads relies on the cross-linking of alginate with divalent calcium ions at concentrations that are toxic for the cells, making control over the beads' size and a single-cell encapsulation unreliable. The present work demonstrates the efficiency of an innovative, high throughput, and reproducible microfluidic system to produce single-cell, calcium-free alginate coatings of human mesenchymal stromal cells. Among the various conditions tested, visible light and confocal microscopy following staining of the cell nuclei by DAPI showed that the microfluidic system yielded an optimal single-cell encapsulation of 2000 cells/min in 2% w/v alginate microcapsules of reproducible morphology and an average size of 28.2 ± 3.7 µm. The adhesive properties of the alginate microcapsules, the viability of the encapsulated cells and their ability to escape the alginate microcapsule were demonstrated by the relatively rapid adherence of the beads onto tissue culture plastic and the cells' ability to gradually disrupt the microcapsule shell after 24 h and proliferate. To mimic the early inflammatory response upon transplantation, the encapsulated cells were exposed to proliferating macrophages at different cell seeding densities for up to 2 days and the protection effect of the microcapsule on the cells assessed by time-lapse microscopy showing a shielding effect for up to 48 h. This work underscores the potential of microfluidic systems to precisely encapsulate cells by good manufacturing practice standards while favouring cell retention on substrates, viability and proliferation upon transplantation.


Subject(s)
Mesenchymal Stem Cells , Microfluidics , Humans , Cell Encapsulation , Capsules , Bone Marrow , Alginates/chemistry , Hexuronic Acids/chemistry , Cell Survival , Glucuronic Acid/chemistry
7.
Pharmacol Res ; 203: 107159, 2024 May.
Article in English | MEDLINE | ID: mdl-38554790

ABSTRACT

Encapsulated cell technology (ECT) is a targeted delivery method that uses the genetically engineered cells in semipermeable polymer capsules to deliver cytokines. Thus far, ECT has been extensively utilized in pharmacologic research, and shows enormous potentials in the treatment of posterior segment diseases. Due to the biological barriers within the eyeball, it is difficult to attain effective therapeutic concentration in the posterior segment through topical administration of drug molecules. Encouragingly, therapeutic cytokines provided by ECT can cross these biological barriers and achieve sustained release at the desired location. The encapsulation system uses permeable materials that allow growth factors and cytokines to diffuse efficiently into retinal tissue. Moreover, the ECT based treatment can be terminated timely when we need to retrieve the implant, which makes the therapy reversible and provides a safer alternative for intraocular gene therapy. Meanwhile, we also place special emphasis on optimizing encapsulation materials and enhancing preservation techniques to achieve the stable release of growth factors and cytokines in the eyeball. This technology holds great promise for the treatment of patients with dry AMD, RP, glaucoma and MacTel. These findings would enrich our understandings of ECT and promote its future applications in treatment of degenerative retinopathy. This review comprises articles evaluating the exactness of artificial intelligence-based formulas published from 2000 to March 2024. The papers were identified by a literature search of various databases (PubMed/MEDLINE, Google Scholar, Cochrane Library and Web of Science).


Subject(s)
Cytokines , Eye Diseases , Humans , Cytokines/metabolism , Animals , Eye Diseases/therapy , Eye Diseases/drug therapy , Drug Delivery Systems/methods , Cell Encapsulation/methods
8.
Adv Sci (Weinh) ; 11(16): e2304861, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38355304

ABSTRACT

An ideal hydrogel for stem cell therapy would be injectable and efficiently promote stem cell proliferation and differentiation in body. Herein, an injectable, single-component hydrogel with hyaluronic acid (HA) modified with phenylboronic acid (PBA) and spermidine (SM) is introduced. The resulting HAps (HA-PBA-SM) hydrogel is based on the reversible crosslinking between the diol and the ionized PBA, which is stabilized by the SM. It has a shear-thinning property, enabling its injection through a syringe to form a stable hydrogel inside the body. In addition, HAps hydrogel undergoes a post-injection "self-curing," which stiffens the hydrogel over time. This property allows the HAps hydrogel to meet the physical requirements for stem cell therapy in rigid tissues, such as bone, while maintaining injectability. The hydrogel enabled favorable proliferation of human mesenchymal stem cells (hMSCs) and promoted their differentiation and mineralization. After the injection of hMSCs-containing HAps into a rat femoral defect model, efficient osteogenic differentiation of hMSCs and bone regeneration is observed. The study demonstrates that simple cationic modification of PBA-based hydrogel enabled efficient gelation with shear-thinning and self-curing properties, and it would be highly useful for stem cell therapy and in vivo bone regeneration.


Subject(s)
Bone Regeneration , Boronic Acids , Cell Differentiation , Hydrogels , Mesenchymal Stem Cells , Animals , Bone Regeneration/physiology , Rats , Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Humans , Hyaluronic Acid/chemistry , Rats, Sprague-Dawley , Cell Encapsulation/methods , Cell Proliferation , Osteogenesis/physiology , Disease Models, Animal , Spermidine/pharmacology , Spermidine/chemistry
9.
Adv Healthc Mater ; 13(14): e2304386, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38373601

ABSTRACT

Tissue engineering at single-cell resolution has enhanced therapeutic efficacy. Droplet microfluidics offers a powerful platform that allows deterministic single-cell encapsulation into aqueous droplets, yet the direct encapsulation of cells into microgels remains challenging. Here, the design of a microfluidic device that is capable of single-cell encapsulation within polyethylene glycol norbornene (PEGNB) hydrogels on-chip is reported. Cells are first ordered in media within a straight microchannel via inertial focusing, followed by the introduction of PEGNB solution from two separate, converging channels. Droplets are thoroughly mixed by passage through a serpentine channel, and microgels are formed by photo-photopolymerization. This platform uniquely enables both single-cell encapsulation and excellent cell viability post-photo-polymerization. More than 90% of singly encapsulated mesenchymal stromal cells (MSCs) remain alive for 7 days. Notably, singly encapsulated MSCs have elevated expression levels in genes that code anti-inflammatory cytokines, for example, IL-10 and TGF-ß, thus enhancing the secretion of proteins of interest. Following injection into a mouse model with induced inflammation, singly encapsulated MSCs show a strong retention rate in vivo, reduce overall inflammation, and mitigate liver damage. These translational results indicate that deterministic single-cell encapsulation could find use in a broad spectrum of tissue engineering applications.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Norbornanes , Polyethylene Glycols , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Polyethylene Glycols/chemistry , Mice , Mesenchymal Stem Cell Transplantation/methods , Norbornanes/chemistry , Microgels/chemistry , Cell Encapsulation/methods , Hydrogels/chemistry , Hydrogels/pharmacology , Cell Survival/drug effects , Humans
10.
Ann Biomed Eng ; 52(5): 1222-1239, 2024 May.
Article in English | MEDLINE | ID: mdl-38353908

ABSTRACT

Surgical treatment of tracheal diseases, trauma, and congenital stenosis has shown success through tracheal reconstruction coupled with palliative care. However, challenges in surgical-based tracheal repairs have prompted the exploration of alternative approaches for tracheal replacement. Tissue-based treatments, involving the cultivation of patient cells on a network of extracellular matrix (ECM) from donor tissue, hold promise for restoring tracheal structure and function without eliciting an immune reaction. In this study, we utilized decellularized canine tracheas as tissue models to develop two types of cell carriers: a decellularized scaffold and a hydrogel. Our hypothesis posits that both carriers, containing essential biochemical niches provided by ECM components, facilitate cell attachment without inducing cytotoxicity. Canine tracheas underwent vacuum-assisted decellularization (VAD), and the ECM-rich hydrogel was prepared through peptic digestion of the decellularized trachea. The decellularized canine trachea exhibited a significant reduction in DNA content and major histocompatibility complex class II, while preserving crucial ECM components such as collagen, glycosaminoglycan, laminin, and fibronectin. Scanning electron microscope and fluorescent microscope images revealed a fibrous ECM network on the luminal side of the cell-free trachea, supporting epithelial cell attachment. Moreover, the ECM-rich hydrogel exhibited excellent viability for human mesenchymal stem cells encapsulated for 3 days, indicating the potential of cell-laden hydrogel in promoting the development of cartilage rings of the trachea. This study underscores the versatility of the trachea in producing two distinct cell carriers-decellularized scaffold and hydrogel-both containing the native biochemical niche essential for tracheal tissue engineering applications.


Subject(s)
Cell Encapsulation , Tissue Scaffolds , Humans , Animals , Dogs , Tissue Engineering/methods , Trachea , Extracellular Matrix/metabolism , Hydrogels
11.
Curr Protoc ; 3(11): e917, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37929691

ABSTRACT

Three-dimensional cell encapsulation has rendered itself a staple in the tissue engineering field. Using recombinantly engineered, biopolymer-based hydrogels to encapsulate cells is especially promising due to the enhanced control and tunability it affords. Here, we describe in detail the synthesis of our hyaluronan (i.e., hyaluronic acid) and elastin-like protein (HELP) hydrogel system. In addition to validating the efficacy of our synthetic process, we also demonstrate the modularity of the HELP system. Finally, we show that cells can be encapsulated within HELP gels over a range of stiffnesses, exhibit strong viability, and respond to stiffness cues. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Elastin-like protein modification with hydrazine Basic Protocol 2: Nuclear magnetic resonance quantification of elastin-like protein modification with hydrazine Basic Protocol 3: Hyaluronic acid-benzaldehyde synthesis Basic Protocol 4: Nuclear magnetic resonance quantification of hyaluronic acid-benzaldehyde Basic Protocol 5: 3D cell encapsulation in hyaluronan elastin-like protein gels.


Subject(s)
Hyaluronic Acid , Hydrogels , Elastin , Cell Encapsulation , Benzaldehydes , Hydrazines
12.
J Biosci Bioeng ; 136(6): 471-476, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37798227

ABSTRACT

Albumin is an attractive component for the development of biomaterials applied as biomedical implants, including drug carriers and tissue engineering scaffolds, because of its high biocompatibility and low immunogenicity. Additionally, albumin-based gelators facilitate cross-linking reactions under mild conditions, which maintains the high viability of encapsulated living cells. In this study, we synthesized albumin derivatives to undergo gelation under physiological conditions via the peroxidase-catalyzed formation of cross-links. Albumin was modified with phenolic hydroxyl groups (Alb-Ph-OH) using carbodiimide chemistry, and the effect of degree of substitution on gelation was investigated. Various properties of the Alb-Ph-OH hydrogels, namely the gelation time, swelling ratio, pore size, storage modulus, and enzymatic degradability, were easily controlled by adjusting the degree of substitution and the polymer concentration. Moreover, the viability of cells encapsulated within the Alb-Ph-OH hydrogel was high. These results demonstrate the potential applicability of Alb-Ph-OH hydrogels as cell-encapsulating materials for biomedical applications, including tissue engineering.


Subject(s)
Cell Encapsulation , Hydrogels , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Albumins
13.
ACS Biomater Sci Eng ; 9(11): 6024-6033, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37788301

ABSTRACT

Microgels are microscale particles of hydrogel that can be laden with cells and used to create macroporous tissue constructs. Their ability to support cell-ECM and cell-cell interactions, along with the high levels of nutrient and metabolite exchange facilitated by their high surface area-to-volume ratio, means that they are attracting increasing attention for a variety of tissue regeneration applications. Here, we present methods for fabricating and modifying the structure of microfluidic devices using commonly available laboratory consumables including pipet tips and PTFE and silicon tubing to produce microgels. Different microfluidic devices realized the controlled generation of a wide size range (130-800 µm) of microgels for cell encapsulation. Subsequently, we describe the process of encapsulating mesenchymal stromal cells in microgels formed by photo-cross-linking of gelatin-norbornene and PEG dithiol. The introduced pipet-based chip offers simplicity, tunability, and versatility, making it easily assembled in most laboratories to effectively produce cell-laden microgels for various applications in tissue engineering.


Subject(s)
Microgels , Cell Encapsulation , Gelatin/chemistry , Tissue Engineering/methods , Hydrogels/chemistry
14.
Prog Mol Biol Transl Sci ; 199: 109-127, 2023.
Article in English | MEDLINE | ID: mdl-37678968

ABSTRACT

Conventional two-dimensional (2-D) cultivation are easy to utilize for human pluripotent stem (hPS) cell cultivation in standard techniques and are important for analysis or development of the signal pathways to keep pluripotent state of hPS cells cultivated on 2-D cell culture materials. However, the most efficient protocol to prepare hPS cells is the cell culture in a three dimensional (3-D) cultivation unit because huge numbers of hPS cells should be utilized in clinical treatment. Some 3-D cultivation strategies for hPS cells are considered: (a) microencapsulated cell cultivation in suspended hydrogels, (b) cell cultivation on microcarriers (MCs), (c) cell cultivation on self-aggregated spheroid [cell aggregates; embryoid bodies (EBs) and organoids], (d) cell cultivation on microfibers or nanofibers, and (e) cell cultivation in macroporous scaffolds. These cultivation ways are described in this chapter.


Subject(s)
Cell Culture Techniques, Three Dimensional , Cell Differentiation , Stem Cells , Humans , Stem Cells/cytology , Cell Culture Techniques , Cell Culture Techniques, Three Dimensional/methods , Hepatocytes/cytology , Hydrogels , Cell Encapsulation , Tissue Scaffolds , Tissue Engineering
15.
Adv Drug Deliv Rev ; 201: 115010, 2023 10.
Article in English | MEDLINE | ID: mdl-37454931

ABSTRACT

The primary impetus of therapeutic cell encapsulation in the past several decades has been to broaden the options for donor cell sources by countering against immune-mediated rejection. However, another significant advantage of encapsulation is to provide donor cells with physiologically relevant cues that become compromised in disease. The advances in biomaterial design have led to the fundamental insight that cells sense and respond to various signals encoded in materials, ranging from biochemical to mechanical cues. The biomaterial design for cell encapsulation is becoming more sophisticated in controlling specific aspects of cellular phenotypes and more precise down to the single cell level. This recent progress offers a paradigm shift by designing single cell-encapsulating materials with predefined cues to precisely control donor cells after transplantation.


Subject(s)
Biocompatible Materials , Cell Encapsulation , Humans , Biology
16.
Appl Microbiol Biotechnol ; 107(18): 5715-5726, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37490127

ABSTRACT

Immobilized yeast cells are used industrially in winemaking processes such as sparkling wine and Sherry wine production. Here, a novel approach has been explored for the infusion and immobilization of yeast cells into filamentous fungal pellets, which serve as a porous natural material. This was accomplished through vacuum application to force the yeast cells towards the core of the fungal pellets followed by culture in YPD medium to promote their growth from the interior. This method represents an improved variation of a previous approach for the assembly of "yeast biocapsules," which entailed the co-culture of both fungal and yeast cells in the same medium. A comparison was made between both techniques in terms of biocapsule productivity, cell retention capacity, and cell biological activity through an alcoholic fermentation of a grape must. The results indicated a substantial increase in biocapsule productivity (37.40-fold), higher cell retention within the biocapsules (threefold), and reduction in cell leakage during fermentation (twofold). Although the majority of the chemical and sensory variables measured in the produced wine did not exhibit notable differences from those produced utilizing suspended yeast cells (conventional method), some differences (such as herbaceous and toasted smells, acidity, bitterness, and persistence) were perceived and wines positively evaluated by the sensory panel. As the immobilized cells remain functional and the encapsulation technique can be expanded to other microorganisms, it creates potential for additional industrial uses like biofuel, health applications, microbe encapsulation and delivery, bioremediation, and pharmacy. KEY POINTS: • New approach improves biocapsule productivity and cell retention. • Immobilized yeast remains functional in fermentation. • Wine made with immobilized yeast had positive sensory differences.


Subject(s)
Saccharomyces cerevisiae , Wine , Saccharomyces cerevisiae/chemistry , Cell Encapsulation , Vacuum , Fermentation , Wine/microbiology
17.
Biomater Sci ; 11(14): 4972-4984, 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37334482

ABSTRACT

Microfluidic on-chip production of polymeric hydrogel microspheres (MPs) can be designed for the loading of different biologically active cargos and living cells. Among different gelation strategies, ionically crosslinked microspheres generally show limited mechanical properties, meanwhile covalently crosslinked microspheres often require the use of crosslinking agents or initiators with limited biocompatibility. Inverse electron demand Diels Alder (iEDDA) click chemistry is a promising covalent crosslinking method with fast kinetics, high chemoselectivity, high efficiency and no cross-reactivity. Herein, in situ gellable iEDDA-crosslinked polymeric hydrogel microspheres are developed via water-in-oil emulsification (W/O) glass microfluidics. The microspheres are composed of two polyethylene glycol precursors modified with either tetrazine or norbornene as functional moieties. Using a single co-flow glass microfluidic platform, homogenous MPs of sizes 200-600 µm are developed and crosslinked within 2 minutes. The rheological properties of iEDDA crosslinked bulk hydrogels are maintained with a low swelling degree and a slow degradation behaviour under physiological conditions. Moreover, a high-protein loading capacity can be achieved, and the encapsulation of mammalian cells is possible. Overall, this work provides the possibility of developing microfluidics-produced iEDDA-crosslinked MPs as a potential drug vehicle and cell encapsulation system in the biomedical field.


Subject(s)
Heterocyclic Compounds , Hydrogels , Animals , Hydrogels/chemistry , Microfluidics , Cell Encapsulation , Click Chemistry , Electrons , Microspheres , Norbornanes/chemistry , Mammals
18.
ACS Appl Bio Mater ; 6(7): 2875-2885, 2023 07 17.
Article in English | MEDLINE | ID: mdl-37339251

ABSTRACT

Catechol (cat) is a highly adhesive diphenol that can be chemically grafted to polymers such as chitosan (CH) to make them adhesive as well. However, catechol-containing materials experimentally show a large variability of toxicity, especially in vitro. While it is unclear how this toxicity emerges, most concerns are directed toward the oxidation of catechol into quinone that releases reactive oxygen species (ROS) which can, in turn, cause cell apoptosis through oxidative stress. To better understand the mechanisms at play, we examined the leaching profiles, hydrogen peroxide (H2O2) production, and in vitro cytotoxicity of several cat-chitosan (cat-CH) hydrogels that were prepared with different oxidation levels and cross-linking methods. To create cat-CH with different propensities toward oxidation, we grafted either hydrocaffeic acid (HCA, more prone to oxidation) or dihydrobenzoic acid (DHBA, less prone to oxidation) to the backbone of CH. Hydrogels were cross-linked either covalently, using sodium periodate (NaIO4) to trigger oxidative cross-linking, or physically, using sodium bicarbonate (SHC). While using NaIO4 as a cross-linker increased the oxidation levels of the hydrogels, it also significantly reduced in vitro cytotoxicity, H2O2 production, and catechol and quinone leaching in the media. For all gels tested, cytotoxicity could be directly related to the release of quinones rather than H2O2 production or catechol release, showing that oxidative stress may not be the main reason for catechol cytotoxicity, as other pathways of quinone toxicity come into play. Results also suggest that the indirect cytotoxicity of cat-CH hydrogels fabricated through carbodiimide chemistry can be reduced if (i) catechol groups are chemically bound to the polymer backbone to prevent leaching or (ii) the chosen cat-bearing molecule has a high resistance to oxidation. Coupled with the use of other cross-linking chemistries or more efficient purification methods, these strategies can be adopted to synthesize various types of cytocompatible cat-containing scaffolds.


Subject(s)
Chitosan , Chitosan/chemistry , Hydrogels/chemistry , Hydrogen Peroxide , Cell Encapsulation , Polymers/chemistry , Catechols/chemistry
19.
ACS Appl Bio Mater ; 6(6): 2496-2504, 2023 06 19.
Article in English | MEDLINE | ID: mdl-37289861

ABSTRACT

Gelatin-based microgels are intriguing for various biomedical applications, which are conventionally prepared through photopolymerization of gelatin methacrylamide (GelMA). Here, we report on the modification of gelatin through acrylamidation to form gelatin acrylamide (GelA) with different substitution degrees, which was found to exhibit fast photopolymerization kinetics, better gelation, steady viscosity at elevated temperatures, and satisfying biocompatibility when compared to GelMA. By the online photopolymerization strategy with a home-made microfluidic setting, microgels of uniform sizes from GelA by blue light were obtained and their swollen properties were investigated. Compared to the microgels from GelMA, they showed an enhanced cross-linking degree and have better shape stability when swollen in water. Cell toxicities of the hydrogels from GelA and cell encapsulation from the corresponding microgels were investigated, which were found to exhibit superior properties than those from GelMA. We therefore believe that GelA has potential for constructing scaffolds for bioapplications and can be an excellent substitute for GelMA.


Subject(s)
Gelatin , Microgels , Microfluidics , Biocompatible Materials , Cell Encapsulation , Tissue Engineering , Acrylamide , Light , Acrylamides
20.
Adv Healthc Mater ; 12(26): e2300942, 2023 10.
Article in English | MEDLINE | ID: mdl-37235850

ABSTRACT

Thiol-norbornene photoclickable poly (ethylene glycol) (PEG)-based (PEG-NB) hydrogels are attractive biomaterials for cell encapsulation, drug delivery, and regenerative medicine applications. Although many crosslinking strategies and chemistries have been developed for PEG-NB bulk hydrogels, fabrication approaches of PEG-NB microgels have not been extensively explored. Here, a fabrication strategy for 4-arm amide-linked PEG-NB (PEG-4aNB) microgels using flow-focusing microfluidics for human mesenchymal stem/stromal cell (hMSCs) encapsulation is presented. PEG-4aNB photochemistry allows high-throughput, ultrafast generation, and cost-effective synthesis of monodispersed microgels (diameter 340 ± 18, 380 ± 24, and 420 ± 15 µm, for 6, 8, and 10 wt% of PEG-4aNB, respectively) using an in situ crosslinking methodology in a microfluidic device. PEG-4aNB microgels show in vitro degradability due to the incorporation of a protease-degradable peptide during photocrosslinking and encapsulated cells show excellent viability and metabolic activity for at least 13 days of culture. Furthermore, the secretory profile (i.e., MMP-13, ICAM-1, PD-L1, CXCL9, CCL3/MIP-1, IL-6, IL-12, IL-17E, TNF-α, CCL2/MCP-1) of encapsulated hMSCs shows increased expression in response to IFN-γ stimulation. Collectively, this work shows a versatile and facile approach for the fabrication of protease-degradable PEG-4aNB microgels for cell encapsulation.


Subject(s)
Microgels , Polyethylene Glycols , Humans , Cell Encapsulation , Peptide Hydrolases , Hydrogels , Biocompatible Materials , Norbornanes
SELECTION OF CITATIONS
SEARCH DETAIL
...