Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters










Publication year range
1.
Mater Today Bio ; 22: 100768, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37600348

ABSTRACT

Visualizing cells, tissues, and their components specifically without interference with cellular functions, such as biochemical reactions, and cellular viability remains important for biomedical researchers worldwide. For an improved understanding of disease progression, tissue formation during development, and tissue regeneration, labeling extracellular matrix (ECM) components secreted by cells persists is required. Bioorthogonal chemistry approaches offer solutions to visualizing and labeling ECM constituents without interfering with other chemical or biological events. Although biorthogonal chemistry has been studied extensively for several applications, this review summarizes the recent advancements in using biorthogonal chemistry specifically for metabolic labeling and visualization of ECM proteins and glycosaminoglycans that are secreted by cells and living tissues. Challenges, limitations, and future directions surrounding biorthogonal chemistry involved in the labeling of ECM components are discussed. Finally, potential solutions for improvements to biorthogonal chemical approaches are suggested. This would provide theoretical guidance for labeling and visualization of de novo proteins and polysaccharides present in ECM that are cell-secreted for example during tissue remodeling or in vitro differentiation of stem cells.

2.
Sci Rep ; 13(1): 12829, 2023 08 07.
Article in English | MEDLINE | ID: mdl-37550357

ABSTRACT

Hydrogels are used extensively as cell-culture scaffolds for both 2D and 3D cell cultures due to their biocompatibility and the ease in which their mechanical and biological properties can be tailored to mimic natural tissue. The challenge when working with hydrogel-based scaffolds is in their handling, as hydrogels that mimic e.g. brain tissue, are both fragile and brittle when prepared as thin (sub-mm) membranes. Here, we describe a method for facile handling of thin hydrogel cell culture scaffolds by molding them onto a polycaprolactone (PCL) mesh support attached to a commonly used Transwell set-up in which the original membrane has been removed. In addition to demonstrating the assembly of this set-up, we also show some applications for this type of biological membrane. A polyethylene glycol (PEG)-gelatin hydrogel supports cell adhesion, and the structures can be used for biological barrier models comprising either one or multiple hydrogel layers. Here, we demonstrate the formation of a tight layer of an epithelial cell model comprising MDCK cells cultured over 9 days by following the build-up of the transepithelial electrical resistances. Second, by integrating a pure PEG hydrogel into the PCL mesh, significant swelling is induced, which leads to the formation of a non-adherent biological scaffold with a large curvature that is useful for spheroid formation. In conclusion, we demonstrate the development of a handling platform for hydrogel cell culture scaffolds for easy integration with conventional measurement techniques and miniaturized organs-on-chip systems.


Subject(s)
Biocompatible Materials , Hydrogels , Hydrogels/chemistry , Biocompatible Materials/chemistry , Cell Culture Techniques , Cell Culture Techniques, Three Dimensional , Technology , Tissue Scaffolds/chemistry , Tissue Engineering/methods
3.
ACS Appl Mater Interfaces ; 15(29): 34407-34418, 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37435912

ABSTRACT

Injectable hydrogels show great promise in developing novel regenerative medicine solutions and present advantages for minimally invasive applications. Hydrogels based on extracellular matrix components, such as collagen, have the benefits of cell adhesiveness, biocompatibility, and degradability by enzymes. However, to date, reported collagen hydrogels possess severe shortcomings, such as nonbiocompatible cross-linking chemistry, significant swelling, limited range of mechanical properties, or gelation kinetics unsuitable for in vivo injection. To solve these issues, we report the design and characterization of an injectable collagen hydrogel based on covalently modified acetyl thiol collagen cross-linked using thiol-maleimide click chemistry. The hydrogel is injectable for up to 72 h after preparation, shows no noticeable swelling, is transparent, can be molded in situ, and retains its shape in solution for at least one year. Notably, the hydrogel mechanical properties can be fine-tuned by simply adjusting the reactant stoichiometries, which to date was only reported for synthetic polymer hydrogels. The biocompatibility of the hydrogel is demonstrated in vitro using human corneal epithelial cells, which maintain viability and proliferation on the hydrogels for at least seven days. Furthermore, the developed hydrogel showed an adhesion strength on soft tissues similar to fibrin glue. Additionally, the developed hydrogel can be used as a sealant for repairing corneal perforations and can potentially alleviate the off-label use of cyanoacrylate tissue adhesive for repairing corneal perforations. Taken together, these characteristics show the potential of the thiol collagen hydrogel for future use as a prefabricated implant, injectable filler, or as sealant for corneal repair and regeneration.


Subject(s)
Corneal Perforation , Hydrogels , Humans , Hydrogels/pharmacology , Hydrogels/chemistry , Click Chemistry , Sulfhydryl Compounds/chemistry , Collagen/pharmacology , Collagen/chemistry , Maleimides/pharmacology
4.
Biomater Adv ; 147: 213331, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36773382

ABSTRACT

Mesenchymal stem cells (MSCs) therapy is a promising approach for treating inflammatory diseases due to their immunosuppressive and tissue repair characteristics. However, allogenic transplantation of MSCs induces thrombotic complications in some patients which limits its potential for clinical translation. To address this challenge, we have exploited the bioactivity of heparin, a well-known anticoagulant and immunosuppressive polysaccharide that is widely used in clinics. We have developed a smart layer-by-layer (LbL) coating strategy using gelatin and heparin polymers exploiting their overall positive and negative charges that enabled efficient complexation with the MSCs' glycocalyx. The stable coating of MSCs suppressed complement attack and mitigated thrombotic activation as demonstrated in human whole blood. Gratifyingly, the MSC coating retained its immunosuppressive properties and differentiation potential when exposed to inflammatory conditions and differentiation factors. We believe the simple coating procedure of MSCs will increase allogenic tolerance and circumvent the major challenge of MSCs transplantation.


Subject(s)
Biomimetics , Mesenchymal Stem Cells , Humans , Polyelectrolytes , Heparin , Cell Differentiation , Immunosuppressive Agents
5.
Biomater Sci ; 10(22): 6399-6412, 2022 Nov 08.
Article in English | MEDLINE | ID: mdl-36214100

ABSTRACT

Hyaluronic acid (HA), one of the main components of the extracellular matrix (ECM), is extensively used in the design of hydrogels and nanoparticles for different biomedical applications due to its critical role in vivo, degradability by endogenous enzymes, and absence of immunogenicity. HA-based hydrogels and nanoparticles have been developed by utilizing different crosslinking chemistries. The development of such crosslinking chemistries indicates that even subtle differences in the structure of reactive groups or the procedure of crosslinking may have a profound impact on the intended mechanical, physical and biological outcomes. There are widespread examples of modified HA polymers that can form either covalently or physically crosslinked biomaterials. More recently, studies have been focused on dynamic covalent crosslinked HA-based biomaterials since these types of crosslinking allow the preparation of dynamic structures with the ability to form in situ, be injectable, and have self-healing properties. In this review, HA-based hydrogels and nanomaterials that are crosslinked by dynamic-covalent coupling (DCC) chemistry have been critically assessed.


Subject(s)
Hydrogels , Nanostructures , Hydrogels/chemistry , Hyaluronic Acid/chemistry , Biocompatible Materials/chemistry , Extracellular Matrix
6.
Int J Mol Sci ; 23(20)2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36293558

ABSTRACT

In severe malformations with a lack of native tissues, treatment options are limited. We aimed at expanding tissue in vivo using the body as a bioreactor and developing a sustainable single-staged procedure for autologous tissue reconstruction in malformation surgery. Autologous micro-epithelium from skin was integrated with plastically compressed collagen and a degradable knitted fabric mesh. Sixty-three scaffolds were implanted in nine rats for histological and mechanical analyses, up to 4 weeks after transplantation. Tissue integration, cell expansion, proliferation, inflammation, strength, and elasticity were evaluated over time in vivo and validated in vitro in a bladder wound healing model. After 5 days in vivo, we observed keratinocyte proliferation on top of the transplant, remodeling of the collagen, and neovascularization within the transplant. At 4 weeks, all transplants were fully integrated with the surrounding tissue. Tensile strength and elasticity were retained during the whole study period. In the in vitro models, a multilayered epithelium covered the defect after 4 weeks. Autologous micro-epithelial transplants allowed for cell expansion and reorganization in vivo without conventional pre-operative in vitro cell propagation. The method was easy to perform and did not require handling outside the operating theater.


Subject(s)
Rodentia , Tissue Engineering , Rats , Animals , Tissue Engineering/methods , Collagen , Tensile Strength , Transplantation, Autologous , Tissue Scaffolds
7.
Biomater Sci ; 9(11): 3939-3944, 2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34002185

ABSTRACT

There is an unmet need to develop strategies that allow site-specific delivery of short interfering RNA (siRNA) without any associated toxicity. To address this challenge, we have developed a novel siRNA delivery platform using chemically modified pluronic F108 as an amphiphilic polymer with a releasable bioactive disulfide functionality. The micelles exhibited thermoresponsive properties and showed a hydrodynamic size of ∼291 nm in DLS and ∼200-250 nm in SEM at 37 °C. The grafting of free disulfide pyridyl groups enhanced the transfection efficiency and was successfully demonstrated in human colon carcinoma (HCT116; 88%) and glioma cell lines (U87; 90%), non-cancerous human dermal fibroblast (HDF; 90%) cells as well as in mouse embryonic stem (mES; 54%) cells. To demonstrate the versatility of our modular nanocarrier design, we conjugated the MDGI receptor targeting COOP peptide on the particle surface that allowed the targeted delivery of the cargo molecules to human patent-derived primary BT-13 gliospheres. Transfection experiments with this design resulted in ∼65% silencing of STAT3 mRNA in BT-13 gliospheres, while only ∼20% of gene silencing was observed in the absence of the peptide. We believe that our delivery method solves current problems related to the targeted delivery of RNAi drugs for potential in vivo applications.


Subject(s)
Micelles , Poloxamer , Animals , Cell Line, Tumor , Mice , Oxidation-Reduction , RNA, Small Interfering/metabolism , Transfection
8.
Biomacromolecules ; 22(5): 1980-1989, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33813822

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) evoke great excitement for treating different human diseases due to their ability to home inflamed tissues, suppress inflammation, and promote tissue regeneration. Despite great promises, clinical trial results are disappointing as allotransplantation of MSCs trigger thrombotic activity and are damaged by the complement system, compromising their survival and function. To overcome this, a new strategy is presented by the silencing of tissue factor (TF), a transmembrane protein that mediates procoagulant activity. Novel Pluronic-based micelles are designed with the pendant pyridyl disulfide group, which are used to conjugate TF-targeting siRNA by the thiol-exchange reaction. This nanocarrier design effectively delivered the payload to MSCs resulting in ∼72% TF knockdown (KD) without significant cytotoxicity. Hematological evaluation of MSCs and TF-KD MSCs in an ex vivo human whole blood model revealed a significant reduction in an instant-blood-mediated-inflammatory reaction as evidenced by reduced platelet aggregation (93% of free platelets in the TF-KD group, compared to 22% in untreated bone marrow-derived MSCs) and thrombin-antithrombin complex formation. Effective TF silencing induced higher MSC differentiation in osteogenic and adipogenic media and showed stronger paracrine suppression of proinflammatory cytokines in macrophages and higher stimulation in the presence of endotoxins. Thus, TF silencing can produce functional cells with higher fidelity, efficacy, and functions.


Subject(s)
Mesenchymal Stem Cells , Cell Differentiation , Cells, Cultured , Humans , Micelles , Paracrine Communication , Poloxamer , Thromboplastin/genetics
9.
J Funct Biomater ; 12(1)2021 Jan 17.
Article in English | MEDLINE | ID: mdl-33477310

ABSTRACT

Acrylic bone cements modified with linoleic acid are a promising low-modulus alternative to traditional high-modulus bone cements. However, several key properties remain unexplored, including the effect of autoclave sterilization and the potential use of low-modulus cements in other applications than vertebral augmentation. In this work, we evaluate the effect of sterilization on the structure and stability of linoleic acid, as well as in the handling properties, glass transition temperature, mechanical properties, and screw augmentation potential of low-modulus cement containing the fatty acid. Neither 1H NMR nor SFC-MS/MS analysis showed any detectable differences in autoclaved linoleic acid compared to fresh one. The peak polymerization temperature of the low-modulus cement was much lower (28-30 °C) than that of the high-modulus cement (67 °C), whereas the setting time remained comparable (20-25 min). The Tg of the low-modulus cement was lower (75-78 °C) than that of the high-stiffness cement (103 °C). It was shown that sterilization of linoleic acid by autoclaving did not significantly affect the functional properties of low-modulus PMMA bone cement, making the component suitable for sterile production. Ultimately, the low-modulus cement exhibited handling and mechanical properties that more closely match those of osteoporotic vertebral bone with a screw holding capacity of under 2000 N, making it a promising alternative for use in combination with orthopedic hardware in applications where high-stiffness augmentation materials can result in undesired effects.

10.
Carbohydr Polym ; 254: 117291, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33357860

ABSTRACT

Anti-inflammatory drugs such as dexamethasone (DEX) are commonly administered to cancer patients along with anticancer drugs, however, the effect of DEX on human cancers is poorly understood. In this article, we have tailored self-assembled nanoparticles derived from hyaluronic acid (HA) wherein, anti-inflammatory DEX was used as a hydrophobic moiety for inducing amphiphilicity. The HA-DEX micelles were subsequently loaded with chemotherapeutic agent, doxorubicin (DOX) (HA-DEX-DOX) and was utilized to deliver drug cargo to human cancer cells expressing different levels of CD44 receptors. We found that DEX suppressed the cytotoxicity of DOX in HCT116, while it synergistically enhanced cytotoxicity in MCF-7 cells. When we tested DOX and HA-DEX-DOX in an ex-vivo human whole blood, we found activation of complement and the coagulation cascade in one group of donors. Encapsulation of DOX within the nanoparticle core eliminated such deleterious side-effects. The HA-DEX-DOX also polarized bone-marrow-derived anti-inflammatory M2 macrophages, to pro-inflammatory M1 phenotype with the upregulation of the cytokines TNF-α, iNOS and IL-1ß.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antibiotics, Antineoplastic/administration & dosage , Cell Polarity/drug effects , Dexamethasone/administration & dosage , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Macrophages/drug effects , Macrophages/immunology , Nanoparticles/chemistry , Animals , Cell Survival/drug effects , Cytokines/metabolism , Drug Combinations , Drug Liberation , HCT116 Cells , Humans , Hyaluronan Receptors/antagonists & inhibitors , Hyaluronic Acid/pharmacology , Inflammation/drug therapy , MCF-7 Cells , Mice , Mice, Inbred C57BL , Micelles , Phenotype , Platelet Aggregation/drug effects , Up-Regulation/drug effects
11.
Nat Mater ; 20(2): 250-259, 2021 02.
Article in English | MEDLINE | ID: mdl-32895507

ABSTRACT

Organoids can shed light on the dynamic interplay between complex tissues and rare cell types within a controlled microenvironment. Here, we develop gut organoid cocultures with type-1 innate lymphoid cells (ILC1) to dissect the impact of their accumulation in inflamed intestines. We demonstrate that murine and human ILC1 secrete transforming growth factor ß1, driving expansion of CD44v6+ epithelial crypts. ILC1 additionally express MMP9 and drive gene signatures indicative of extracellular matrix remodelling. We therefore encapsulated human epithelial-mesenchymal intestinal organoids in MMP-sensitive, synthetic hydrogels designed to form efficient networks at low polymer concentrations. Harnessing this defined system, we demonstrate that ILC1 drive matrix softening and stiffening, which we suggest occurs through balanced matrix degradation and deposition. Our platform enabled us to elucidate previously undescribed interactions between ILC1 and their microenvironment, which suggest that they may exacerbate fibrosis and tumour growth when enriched in inflamed patient tissues.


Subject(s)
Extracellular Matrix/metabolism , Intestinal Mucosa/metabolism , Lymphocytes/metabolism , Organoids/metabolism , Animals , Female , Humans , Intestinal Mucosa/cytology , Lymphocytes/cytology , Matrix Metalloproteinase 9/metabolism , Mice , Organoids/cytology , Transforming Growth Factor beta1/metabolism
12.
Biomed Mater ; 16(2): 022003, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33049725

ABSTRACT

Click chemistry is not a single specific reaction, but describes ways of generating products which emulate examples in nature. Click reactions occur in one pot, are not disturbed by water, generate minimal and inoffensive byproducts, and are characterized by a high thermodynamic driving force, driving the reaction quickly and irreversibly towards a high yield of a single reaction product. As a result, over the past 15 years it has become a very useful bio-orthogonal method for the preparation of chemical cross-linked biopolymer-based hydrogel, in the presence of e.g. growth factors and live cells, or in-vivo. Biopolymers are renewable and non-toxic, providing a myriad of potential backbone toolboxes for hydrogel design. The goal of this review is to summarize recent advances in the development of click chemistry-based biopolymeric hydrogels, and their applications in regenerative medicine. In particular, various click chemistry approaches, including copper-catalyzed azide-alkyne cycloaddition reactions, copper-free click reactions (e.g. the Diels-Alder reactions, the strain-promoted azide-alkyne cycloaddition reactions, the radical mediated thiol-ene reactions, and the oxime-forming reactions), and pseudo-click reactions (e.g. the thiol-Michael addition reactions and the Schiff base reactions) are highlighted in the first section. In addition, numerous biopolymers, including proteins (e.g. collagen, gelatin, silk, and mucin), polysaccharides (e.g. hyaluronic acid, alginate, dextran, and chitosan) and polynucleotides (e.g. deoxyribonucleic acid), are discussed. Finally, we discuss biopolymeric hydrogels, cross-linked by click chemistry, intended for the regeneration of skin, bone, spinal cord, cartilage, and cornea. This article provides new insights for readers in terms of the design of regenerative medicine, and the use of biopolymeric hydrogels based on click chemistry reactions.


Subject(s)
Biocompatible Materials/chemistry , Biopolymers/chemistry , Click Chemistry/methods , Copper/chemistry , Cross-Linking Reagents/chemistry , Hydrogels/chemistry , Polymers/chemistry , Regenerative Medicine/methods , Tissue Engineering/methods , Alginates , Animals , Cartilage , Collagen/chemistry , Cycloaddition Reaction , Drug Delivery Systems/methods , Gelatin , Humans , Hyaluronic Acid , Mice , Proteins/chemistry , Rats , Stress, Mechanical , Sulfhydryl Compounds/chemistry , Wound Healing
13.
Nat Commun ; 11(1): 1365, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32170076

ABSTRACT

Nanoclays have generated interest in biomaterial design for their ability to enhance the mechanics of polymeric materials and impart biological function. As well as their utility as physical cross-linkers, clays have been explored for sustained localization of biomolecules to promote in vivo tissue regeneration. To date, both biomolecule-clay and polymer-clay nanocomposite strategies have utilised the negatively charged clay particle surface. As such, biomolecule-clay and polymer-clay interactions are set in competition, potentially limiting the functional enhancements achieved. Here, we apply specific bisphosphonate interactions with the positively charged clay particle edge to develop self-assembling hydrogels and functionalized clay nanoparticles with preserved surface exchange capacity. Low concentrations of nanoclay are applied to cross-link hyaluronic acid polymers derivatised with a pendant bisphosphonate to generate hydrogels with enhanced mechanical properties and preserved protein binding able to sustain, for over six weeks in vivo, the localized activity of the clinically licensed growth factor BMP-2.


Subject(s)
Diphosphonates/metabolism , Hydrogels/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Nanocomposites/chemistry , Nanoparticles/chemistry , Animals , Bone Morphogenetic Protein 2/metabolism , Clay , Drug Delivery Systems , Female , Materials Testing , Mice , Polymers/chemistry , Protein Binding , Silicates
15.
Biomater Sci ; 8(1): 302-312, 2019 Dec 17.
Article in English | MEDLINE | ID: mdl-31701967

ABSTRACT

Statins are currently the most prescribed hypercholesterolemia-lowering drugs worldwide, with estimated usage approaching one-sixth of the population. However, statins are known to cause pleiotropic skeletal myopathies in 1.5% to 10% of patients and the mechanisms by which statins induce this response, are not fully understood. In this study, a 3D collagen-based tissue-engineered skeletal muscle construct is utilised as a screening platform to test the efficacy and toxicity of a new delivery system. A hyaluronic acid derived nanoparticle loaded with simvastatin (HA-SIM-NPs) is designed and the effect of free simvastatin and HA-SIM-NPs on cellular, molecular and tissue response is investigated. Morphological ablation of myotubes and lack of de novo myotube formation (regeneration) was evident at the highest concentrations (333.33 µM), independent of delivery vehicle (SIM or HA-SIM-NP). A dose-dependent disruption of the cytoskeleton, reductions in metabolic activity and tissue engineered (TE) construct tissue relaxation was evident in the free drug condition (SIM, 3.33 µM and 33.33 nM). However, most of these changes were ameliorated when SIM was delivered via HA-SIM-NPs. Significantly, homogeneous expressions of MMP2, MMP9, and myogenin in HA-SIM-NPs outlined enhanced regenerative responses compared to SIM. Together, these results outline statin delivery via HA-SIM-NP as an effective delivery mechanism to inhibit deleterious myotoxic side-effects.


Subject(s)
Hyaluronic Acid/chemistry , Muscle, Skeletal/cytology , Osteogenesis/drug effects , Simvastatin/adverse effects , Animals , Cell Differentiation/drug effects , Cell Line , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Mice , Muscle, Skeletal/chemistry , Muscle, Skeletal/drug effects , Myogenin/genetics , Myotoxicity , Nanoparticles , Simvastatin/chemistry , Simvastatin/pharmacology , Tissue Engineering , Tissue Scaffolds
16.
ACS Appl Mater Interfaces ; 11(41): 38232-38239, 2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31550878

ABSTRACT

Currently, there are limited approaches to tailor 3D scaffolds cross-linked with a stable covalent C-C bond that does not require any catalysts or initiators. We present here the first hydrogels employing aldol condensation chemistry that exhibit exceptional physicochemical properties. We investigated the aldol-cross-linking chemistry using two types of aldehyde-modified hyaluronic acid (HA) derivatives, namely, an enolizable HA-aldehyde (HA-Eal) and a non-enolizable HA-aldehyde (HA-Nal). Hydrogels formed using HA-Eal demonstrate inferior cross-linking efficiency (due to intramolecular loop formation), when compared with hydrogels formed by mixing HA-Eal and HA-NaI leading to a cross-aldol product. The change in mechanical properties as a result of cross-linking at different pH values is determined using rheological measurements and is interpreted in terms of molecular weight between cross-links (Mc). The novel HA cross-aldol hydrogel demonstrate excellent hydrolytic stability and favorable mechanical properties but allow hyaluronidase-mediated enzymatic degradation. Interestingly, residual aldehyde functionality within the aldol product rendered the tissue-adhesive properties by bonding two bone tissues. The aldehyde functionality also facilitated facile post-synthetic modifications with nucleophilic reagents. Finally, we demonstrate that the novel hydrogel is biocompatible with encapsulated stem cells that show a linear rate of expansion in our 3-6 days of study.


Subject(s)
Cells, Immobilized/metabolism , Hyaluronic Acid , Hydrogels , Materials Testing , Mesenchymal Stem Cells/metabolism , Tissue Adhesives , Cells, Immobilized/cytology , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogen-Ion Concentration , Mesenchymal Stem Cells/cytology , Tissue Adhesives/chemistry , Tissue Adhesives/pharmacology
17.
Biomacromolecules ; 20(9): 3475-3484, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31408340

ABSTRACT

Injectable hydrogels based on extracellular matrix-derived polymers show much promise in the field of tissue engineering and regenerative medicine. However, the hydrogels reported to date have at least one characteristic that limits their potential for clinical use, such as excessive swelling, complicated and potentially toxic cross-linking process, or lack of shear thinning and self-healing properties. We hypothesized that a collagen hydrogel cross-linked using thiol-Michael addition click reaction would be able to overcome these limitations. To this end, collagen was modified to introduce thiol groups, and hydrogels were prepared by cross-linking with 8-arm polyethylene glycol-maleimide. Rheological measurements on the hydrogels revealed excellent shear-thinning and self-healing properties. Additionally, only minimal swelling (6%) was observed over a period of 1 month in an aqueous buffer solution. Finally, tests using mesenchymal stromal cells and endothelial cells showed that the hydrogels are cell-compatible and suitable for cell encapsulation and delivery. Thus, the reported thiolated-collagen hydrogel cross-linked using thiol-Michael addition click reaction overcomes most of the challenges in the injectable hydrogel design and is an excellent candidate for cell delivery in regenerative medicine and tissue engineering applications. The hydrogel reported here is the first example of a self-healing hydrogel containing covalent cross-links.


Subject(s)
Drug Delivery Systems , Extracellular Matrix/drug effects , Hydrogels/pharmacology , Tissue Engineering , Cell Encapsulation , Click Chemistry , Collagen/chemistry , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , Extracellular Matrix/chemistry , Humans , Hydrogels/chemical synthesis , Hydrogels/chemistry , Maleimides/chemistry , Mesenchymal Stem Cells/drug effects , Sulfhydryl Compounds/chemical synthesis , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology
18.
Biomater Sci ; 7(9): 3906-3917, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31322163

ABSTRACT

Cardiovascular diseases represent a major socio-economic burden. In recent years, considerable effort has been invested in optimizing cell delivery strategies to advance cell transplantation therapies to restore heart function for example after an infarct. A particular issue is that the implantation of cells using a non-electroconductive matrix potentially causes arrhythmia. Here, we demonstrate that our hydrazide-functionalized nanotubes-pericardial matrix-derived electroconductive biohybrid hydrogel provides a suitable environment for maturation of human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes. hiPSC-derived cardiomyocytes exhibited an improved contraction amplitude (>500%) on conductive hydrogels compared to cells cultured on Matrigel®. This was accompanied by increased cellular alignment, enhanced connexin 43 expression, and improved sarcomere organization suggesting maturation of the hiPSC-derived cardiomyocytes. Sarcomeric length of these cells increased from 1.3 to 1.7 µm. Moreover, 3D cell-laden engineered tissues exhibited enhanced calcium handling as well as positive response to external electrical and pharmaceutical stimulation. Collectively, our data indicate that our biohybrid hydrogels consisting of solubilized nanostructured pericardial matrix and electroconductive positively charged hydrazide-conjugated carbon nanotubes provide a promising material for stem cell-based cardiac tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Hydrogels/chemistry , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Nanotubes, Carbon/chemistry , Pericardium/chemistry , Tissue Scaffolds/chemistry , Biomarkers/metabolism , Calcium/metabolism , Cell Differentiation , Cell Proliferation , Cell Survival , Collagen/chemistry , Connexin 43/metabolism , Drug Combinations , Electric Conductivity , Humans , Laminin/chemistry , Mesenchymal Stem Cells/cytology , Particle Size , Proteoglycans/chemistry
19.
Biomacromolecules ; 20(3): 1412-1420, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30726668

ABSTRACT

The disulfide bond plays a crucial role in protein biology and has been exploited by scientists to develop antibody-drug conjugates, sensors, and for the immobilization other biomolecules to materials surfaces. In spite of its versatile use, the disulfide chemistry suffers from some inevitable limitations such as the need for basic conditions (pH > 8.5), strong oxidants, and long reaction times. We demonstrate here that thiol-substrates containing electron-withdrawing groups at the ß-position influence the deprotonation of the thiol group, which is the key reaction intermediate in the formation of disulfide bonds. Evaluation of reaction kinetics using small molecule substrate such as l-cysteine indicated disulfide formation at a 2.8-fold higher ( k1 = 5.04 × 10-4 min-1) reaction rate as compared to the conventional thiol substrate, namely 3-mercaptopropionic acid ( k1 = 1.80 × 10-4 min-1) at physiological pH (pH 7.4). Interestingly, the same effect could not be observed when N-acetyl-l-cysteine substrate ( k1 = 0.51 × 10-4 min-1) was used. We further grafted such thiol-containing molecules (cysteine, N-acetyl-cysteine, and 3-mercaptopropionic acid) to a biopolymer namely hyaluronic acid (HA) and determined the p Ka value of different thiol groups by spectrophotometric analysis. The electron-withdrawing group at the ß-position reduced the p Ka of the thiol group to 7.0 for HA-cysteine (HA-Cys); 7.4 for N-acetyl cysteine (HA-ActCys); and 8.1 for HA-thiol (HA-SH) derivatives, respectively. These experiments further confirmed that the concentration of thiolate (R-S-) ions could be increased with the presence of electron-withdrawing groups, which could facilitate disulfide cross-linked hydrogel formation at physiological pH. Indeed, HA grafted with cysteine or N-acetyl groups formed hydrogels within 3.5 min or 10 h, respectively, at pH 7.4. After completion of cross-linking reaction, both gels demonstrated a storage modulus G' ≈ 3300-3500 Pa, which indicated comparable levels of cross-linking. The HA-SH gel, on the other hand, did not form any gel at pH 7.4 even after 24 h. Finally, we demonstrated that the newly prepared hydrogels exhibited excellent hydrolytic stability but can be degraded by cell-directed processes (enzymatic and reductive degradation). We believe our study provides a valuable insight on the factors governing the disulfide formation and our results are useful to develop strategies that would facilitate generation of stable thiol functionalized biomolecules or promote fast thiol oxidation according to the biomedical needs.


Subject(s)
Cross-Linking Reagents/chemistry , Disulfides/chemistry , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Hydrogen-Ion Concentration , Oxidation-Reduction
20.
Macromol Rapid Commun ; 40(7): e1800837, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30672628

ABSTRACT

Self-healing hydrogels based on metal-ligand coordination chemistry provide new and exciting properties that improve injectability, rheological behaviors, and even biological functionalities. The inherent reversibility of coordination bonds improves on the covalent cross-linking employed previously, allowing for the preparation of completely self-healing hydrogels. In this article, recent advances in the development of this class of hydrogels are summarized and their applications in biology and medicine are discussed. Various chelating ligands such as bisphosphonate, catechol, histidine, thiolate, carboxylate, pyridines (including bipyridine and terpyridine), and iminodiacetate conjugated onto polymeric backbones, as well as the chelated metal ions and metal ions containing inorganic particles, which are used to form dynamic networks, are highlighted. This article provides general ideas and methods for the design of self-healing hydrogel biomaterials based on coordination chemistry.


Subject(s)
Biomedical Technology , Coordination Complexes/chemistry , Drug Design , Hydrogel, Polyethylene Glycol Dimethacrylate/chemical synthesis , Polymers/chemical synthesis , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Ligands , Polymers/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...