Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Acta Biomater ; 177: 50-61, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38331132

ABSTRACT

Cell therapies offer great promise in the treatment of diseases and tissue regeneration, but their clinical use has many challenges including survival, optimal performance in their intended function, or localization at sites where they are needed for effective outcomes. We report here on a method to coat a biodegradable matrix of biomimetic nanofibers on single cells that could have specific functions ranging from cell signaling to targeting and helping cells survive when used for therapies. The fibers are composed of peptide amphiphile (PA) molecules that self-assemble into supramolecular nanoscale filaments. The PA nanofibers were able to create a mesh-like coating for a wide range of cell lineages with nearly 100 % efficiency, without interrupting the natural cellular phenotype or functions. The targeting abilities of this system were assessed in vitro using human primary regulatory T (hTreg) cells coated with PAs displaying a vascular cell adhesion protein 1 (VCAM-1) targeting motif. This approach provides a biocompatible method for single-cell coating that does not negatively alter cellular phenotype, binding capacity, or immunosuppressive functionality, with potential utility across a broad spectrum of cell therapies. STATEMENT OF SIGNIFICANCE: Cell therapies hold great promise in the treatment of diseases and tissue regeneration, but their clinical use has been limited by cell survival, targeting, and function. We report here a method to coat single cells with a biodegradable matrix of biomimetic nanofibers composed of peptide amphiphile (PA) molecules. The nanofibers were able to coat cells, such as human primary regulatory T cells, with nearly 100 % efficiency, without interrupting the natural cellular phenotype or functions. The approach provides a biocompatible method for single-cell coating that does not negatively alter cellular phenotype, binding capacity, or immunosuppressive functionality, with potential utility across a broad spectrum of cell therapies.


Subject(s)
Nanofibers , Humans , Nanofibers/chemistry , Biomimetics , Extracellular Matrix , Peptides/pharmacology , Peptides/chemistry
2.
Proc Natl Acad Sci U S A ; 117(24): 13329-13338, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32461372

ABSTRACT

Two-dimensional (2D) molybdenum disulfide (MoS2) nanomaterials are an emerging class of biomaterials that are photoresponsive at near-infrared wavelengths (NIR). Here, we demonstrate the ability of 2D MoS2 to modulate cellular functions of human stem cells through photothermal mechanisms. The interaction of MoS2 and NIR stimulation of MoS2 with human stem cells is investigated using whole-transcriptome sequencing (RNA-seq). Global gene expression profile of stem cells reveals significant influence of MoS2 and NIR stimulation of MoS2 on integrins, cellular migration, and wound healing. The combination of MoS2 and NIR light may provide new approaches to regulate and direct these cellular functions for the purposes of regenerative medicine as well as cancer therapy.


Subject(s)
Disulfides/radiation effects , Mesenchymal Stem Cells/radiation effects , Molybdenum/radiation effects , Nanostructures/radiation effects , Cell Adhesion/radiation effects , Cell Movement/radiation effects , Cell Survival , Disulfides/chemistry , Disulfides/metabolism , Gene Expression Profiling , Humans , Infrared Rays , Integrins/genetics , Integrins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Molybdenum/chemistry , Molybdenum/metabolism , Nanostructures/chemistry , Photosensitizing Agents , Signal Transduction/radiation effects
3.
ACS Biomater Sci Eng ; 6(8): 4551-4560, 2020 08 10.
Article in English | MEDLINE | ID: mdl-33455189

ABSTRACT

Supramolecular biomaterials are promising systems to bind or deliver therapeutic growth factors given their great structural versatility and tunability of properties by simply mixing molecules. In this work, we have investigated this approach for the growth factor cytokine TGFß-1, which is potentially important in the regeneration of damaged cartilage or in the prevention of fibrinogenesis of organs and the progression of tumors. Our previous work identified a peptide sequence capable of binding TGFß-1 and supramolecular peptide amphiphile (PA) nanofiber hydrogels that displayed the sequence were found to enhance regeneration of cartilage in a rabbit model. In this work, we have synthesized novel PA molecules motivated by the tendency of the original bioactive peptide to undergo deamidation during purification procedures, thus interfering with synthesis of molecularly well-defined structures. We report here on novel PA nanofibers that can be purified without deamidation to establish if the chemical reaction affects chondrogenesis. Interestingly, we found that gels formed from nanofibers displaying a fully deamidated sequence by introducing an asparagine to aspartic acid mutation retain 25% more growth factor relative to those displaying the original bioactive peptide even though the individual peptides have similar affinity for the cytokine. We attribute this difference in growth factor retention to bundling of nanofibers displaying the original asparagine-containing sequence, thus masking the growth factor-binding structure. Improved retention of the growth factor resulted in chondrogenesis of cells encapsulated in the gels as indicated by a more than 50% increase in Sox 9 expressing cells at 3 days and a 100% increase in glycosaminoglycan production at 21 days. We have therefore been able to design a more effective bioactive supramolecular biomaterial to bind TGFß-1, and also demonstrated how bioactive peptide sequences in supramolecular biomaterials can have impact on their structure at larger length scales that change their biological functions.


Subject(s)
Hydrogels , Nanofibers , Animals , Peptides , Phosphorylation , Protein Binding , Rabbits
4.
Adv Mater ; 31(23): e1900332, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30941811

ABSTRACT

Clay nanomaterials are an emerging class of 2D biomaterials of interest due to their atomically thin layered structure, charged characteristics, and well-defined composition. Synthetic nanoclays are plate-like polyions composed of simple or complex salts of silicic acids with a heterogeneous charge distribution and patchy interactions. Due to their biocompatible characteristics, unique shape, high surface-to-volume ratio, and charge, nanoclays are investigated for various biomedical applications. Here, a critical overview of the physical, chemical, and physiological interactions of nanoclay with biological moieties, including cells, proteins, and polymers, is provided. The state-of-the-art biomedical applications of 2D nanoclay in regenerative medicine, therapeutic delivery, and additive manufacturing are reviewed. In addition, recent developments that are shaping this emerging field are discussed and promising new research directions for 2D nanoclay-based biomaterials are identified.


Subject(s)
Biocompatible Materials/chemistry , Clay/chemistry , Drug Delivery Systems/methods , Nanostructures/chemistry , Regenerative Medicine/methods , Animals , Humans , Polymers/chemistry , Printing, Three-Dimensional , Proteins/chemistry , Silicates/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry
5.
Regen Biomater ; 6(1): 29-37, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30740240

ABSTRACT

Additive manufacturing (AM) has shown promise in designing 3D scaffold for regenerative medicine. However, many synthetic biomaterials used for AM are bioinert. Here, we report synthesis of bioactive nanocomposites from a poly(ethylene oxide terephthalate) (PEOT)/poly(butylene terephthalate) (PBT) (PEOT/PBT) copolymer and 2D nanosilicates for fabricating 3D scaffolds for bone tissue engineering. PEOT/PBT have been shown to support calcification and bone bonding ability in vivo, while 2D nanosilicates induce osteogenic differentiation of human mesenchymal stem cells (hMSCs) in absence of osteoinductive agents. The effect of nanosilicates addition to PEOT/PBT on structural, mechanical and biological properties is investigated. Specifically, the addition of nanosilicate to PEOT/PBT improves the stability of nanocomposites in physiological conditions, as nanosilicate suppressed the degradation rate of copolymer. However, no significant increase in the mechanical stiffness of scaffold due to the addition of nanosilicates is observed. The addition of nanosilicates to PEOT/PBT improves the bioactive properties of AM nanocomposites as demonstrated in vitro. hMSCs readily proliferated on the scaffolds containing nanosilicates and resulted in significant upregulation of osteo-related proteins and production of mineralized matrix. The synergistic ability of nanosilicates and PEOT/PBT can be utilized for designing bioactive scaffolds for bone tissue engineering.

6.
ACS Appl Mater Interfaces ; 11(7): 6741-6750, 2019 Feb 20.
Article in English | MEDLINE | ID: mdl-30676016

ABSTRACT

We present a nanoengineered system for sustained and prolonged delivery of protein therapeutics, which has the potential to impact current orthopedic regeneration strategies. Specifically, we introduce two-dimensional nanosilicates with a high surface area and charged characteristics for delivery of active proteins for more than 30 days. The nanosilicates show high binding efficacy without altering the protein conformation and bioactivity. The released proteins are able to maintain high activity as demonstrated by enhanced differentiation of human mesenchymal stem cells at 10-fold lower concentration compared to the exogenous control. Utilizing the nanosilicates as a delivery vehicle could minimize the negative side effects observed because of the use of supraphysiological dosages of protein therapeutics for orthopedic regeneration strategies.


Subject(s)
Bone Morphogenetic Protein 2 , Cell Differentiation/drug effects , Mesenchymal Stem Cells/metabolism , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/pharmacokinetics , Bone Morphogenetic Protein 2/pharmacology , Cell Line , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Humans , Mesenchymal Stem Cells/cytology
7.
Proc Natl Acad Sci U S A ; 115(17): E3905-E3913, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29643075

ABSTRACT

Two-dimensional nanomaterials, an ultrathin class of materials such as graphene, nanoclays, transition metal dichalcogenides (TMDs), and transition metal oxides (TMOs), have emerged as a new generation of materials due to their unique properties relative to macroscale counterparts. However, little is known about the transcriptome dynamics following exposure to these nanomaterials. Here, we investigate the interactions of 2D nanosilicates, a layered clay, with human mesenchymal stem cells (hMSCs) at the whole-transcriptome level by high-throughput sequencing (RNA-seq). Analysis of cell-nanosilicate interactions by monitoring changes in transcriptome profile uncovered key biophysical and biochemical cellular pathways triggered by nanosilicates. A widespread alteration of genes was observed due to nanosilicate exposure as more than 4,000 genes were differentially expressed. The change in mRNA expression levels revealed clathrin-mediated endocytosis of nanosilicates. Nanosilicate attachment to the cell membrane and subsequent cellular internalization activated stress-responsive pathways such as mitogen-activated protein kinase (MAPK), which subsequently directed hMSC differentiation toward osteogenic and chondrogenic lineages. This study provides transcriptomic insight on the role of surface-mediated cellular signaling triggered by nanomaterials and enables development of nanomaterials-based therapeutics for regenerative medicine. This approach in understanding nanomaterial-cell interactions illustrates how change in transcriptomic profile can predict downstream effects following nanomaterial treatment.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cells/metabolism , Nanoparticles , Silicates/pharmacology , Transcriptome/drug effects , Clathrin/metabolism , Endocytosis/drug effects , High-Throughput Nucleotide Sequencing , Humans , Mesenchymal Stem Cells/cytology
8.
J Biomed Mater Res A ; 106(7): 2048-2058, 2018 07.
Article in English | MEDLINE | ID: mdl-29577576

ABSTRACT

Nanoparticle shape has emerged as a key regulator of nanoparticle transport across physiological barriers, intracellular uptake, and biodistribution. We report a facile approach to synthesize ellipsoidal nanoparticles through self-assembly of poly(glycerol sebacate)-co-poly(ethylene glycol) (PGS-co-PEG). The PGS-PEG nanoparticle system is highly tunable, and the semiaxis length of the nanoparticles can be modulated by changing PGS-PEG molar ratio and incorporating therapeutics. As both PGS and PEG are highly biocompatible, the PGS-co-PEG nanoparticles show high hemo-, immuno-, and cytocompatibility. Our data suggest that PGS-co-PEG nanoparticles have the potential for use in a wide range of biomedical applications including regenerative medicine, stem cell engineering, immune modulation, and cancer therapeutics. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2048-2058, 2018.


Subject(s)
Decanoates/chemistry , Drug Delivery Systems/methods , Glycerol/analogs & derivatives , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , Animals , Cell Line , Decanoates/chemical synthesis , Endocytosis , Glycerol/chemical synthesis , Glycerol/chemistry , Intracellular Space , Mice , Nanoparticles/ultrastructure , Polyethylene Glycols/chemical synthesis , Polymers/chemical synthesis
9.
Acta Biomater ; 70: 35-47, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29425720

ABSTRACT

We report injectable nanoengineered hemostats for enhanced wound healing and tissue regeneration. The nanoengineered system consists of the natural polysaccharide, κ-carrageenan (κCA), loaded with synthetic two-dimensional (2D) nanosilicates. Nanoengineered hydrogels showed shear-thinning characteristics and can be injected for minimally invasive approaches. The injectable gels can be physically crosslinked in presence of monovalent ions to form mechanically strong hydrogels. By controlling the ratio between κCA and nanosilicates, compressive stiffness of crosslinked hydrogels can be modulated between 20 and 200 kPa. Despite high mechanical stiffness, nanocomposite hydrogels are highly porous with an interconnected network. The addition of nanosilicates to κCA increases protein adsorption on nanocomposite hydrogels that results in enhance cell adhesion and spreading, increase platelets binding and reduce blood clotting time. Moreover, due to presence of nanosilicates, a range of therapeutic biomacromolecules can be deliver in a sustain manner. The addition of nanosilicates significantly suppresses the release of entrap vascular endothelial growth factor (VEGF) and facilitate in vitro tissue regeneration and wound healing. Thus, this multifunctional nanocomposite hydrogel can be used as an injectable hemostat and an efficient vehicle for therapeutic delivery to facilitate tissue regeneration. STATEMENT OF SIGNIFICANCE: Hemorrhage is a leading cause of death in battlefield wounds, anastomosis hemorrhage and percutaneous intervention. Thus, there is a need for the development of novel bioactive materials to reduce the likelihood of hemorrhagic shock stemming from internal wounds. Here, we introduce an injectable hemostat from kappa-carrageenan and two-dimensional (2D) nanosilicates. Nanosilicates mechanically reinforce the hydrogels, provide enhanced physiological stability and accelerate the clotting time by two-fold. The sustained release of entrapped therapeutics due to presence of nanosilicates promotes enhanced wound healing. The multifunctional nanocomposite hydrogels could be used as an injectable hemostat for penetrating injury and percutaneous intervention during surgery.


Subject(s)
Carrageenan , Hydrogels , Mesenchymal Stem Cells/metabolism , Nanocomposites/chemistry , Wound Healing/drug effects , Carrageenan/chemistry , Carrageenan/pharmacology , Cell Adhesion/drug effects , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Mesenchymal Stem Cells/pathology , Vascular Endothelial Growth Factor A/biosynthesis
10.
ACS Appl Mater Interfaces ; 10(12): 9957-9968, 2018 Mar 28.
Article in English | MEDLINE | ID: mdl-29461795

ABSTRACT

We introduce an enhanced nanoengineered ionic-covalent entanglement (NICE) bioink for the fabrication of mechanically stiff and elastomeric 3D biostructures. NICE bioink formulations combine nanocomposite and ionic-covalent entanglement (ICE) strengthening mechanisms to print customizable cell-laden constructs for tissue engineering with high structural fidelity and mechanical stiffness. Nanocomposite and ICE strengthening mechanisms complement each other through synergistic interactions, improving mechanical strength, elasticity, toughness, and flow properties beyond the sum of the effects of either reinforcement technique alone. Herschel-Bulkley flow behavior shields encapsulated cells from excessive shear stresses during extrusion. The encapsulated cells readily proliferate and maintain high cell viability over 120 days within the 3D-printed structure, which is vital for long-term tissue regeneration. A unique aspect of the NICE bioink is its ability to print much taller structures, with higher aspect ratios, than can be achieved with conventional bioinks without requiring secondary supports. We envision that NICE bioinks can be used to bioprint complex, large-scale, cell-laden constructs for tissue engineering with high structural fidelity and mechanical stiffness for applications in custom bioprinted scaffolds and tissue engineered implants.


Subject(s)
Printing, Three-Dimensional , Bioprinting , Cell Survival , Tissue Engineering , Tissue Scaffolds
11.
Adv Mater ; 29(36)2017 Sep.
Article in English | MEDLINE | ID: mdl-28940819

ABSTRACT

A new approach of vacancy-driven gelation to obtain chemically crosslinked hydrogels from defect-rich 2D molybdenum disulfide (MoS2 ) nanoassemblies and polymeric binder is reported. This approach utilizes the planar and edge atomic defects available on the surface of the 2D MoS2 nanoassemblies to form mechanically resilient and elastomeric nanocomposite hydrogels. The atomic defects present on the lattice plane of 2D MoS2 nanoassemblies are due to atomic vacancies and can act as an active center for vacancy-driven gelation with a thiol-activated terminal such as four-arm poly(ethylene glycol)-thiol (PEG-SH) via chemisorption. By modulating the number of vacancies on the 2D MoS2 nanoassemblies, the physical and chemical properties of the hydrogel network can be controlled. This vacancy-driven gelation process does not require external stimuli such as UV exposure, chemical initiator, or thermal agitation for crosslinking and thus provides a nontoxic and facile approach to encapsulate cells and proteins. 2D MoS2 nanoassemblies are cytocompatible, and encapsulated cells in the nanocomposite hydrogels show high viability. Overall, the nanoengineered hydrogel obtained from vacancy-driven gelation is mechanically resilient and can be used for a range of biomedical applications including tissue engineering, regenerative medicine, and cell and therapeutic delivery.


Subject(s)
Nanocomposites , Hydrogels , Polyethylene Glycols , Tissue Engineering
12.
Acta Biomater ; 59: 33-44, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28647624

ABSTRACT

The synthesis of thermoset shape memory polymer (SMP) polyurethanes from symmetric, aliphatic alcohols and diisocyanates has previously demonstrated excellent biocompatibility in short term in vitro and in vivo studies, although long term stability has not been investigated. Here we demonstrate that while rapid oxidation occurs in these thermoset SMPs, facilitated by the incorporation of multi-functional, branching amino groups, byproduct analysis does not indicate toxicological concern for these materials. Through complex multi-step chemical reactions, chain scission begins from the amines in the monomeric repeat units, and results, ultimately, in the formation of carboxylic acids, secondary and primary amines; the degradation rate and product concentrations were confirmed using liquid chromatography mass spectrometry, in model compound studies, yielding a previously unexamined degradation mechanism for these biomaterials. The rate of degradation is dependent on the hydrogen peroxide concentration, and comparison of explanted samples reveals a much slower rate in vivo compared to the widely accepted literature in vitro real-time equivalent of 3% H2O2. Cytotoxicity studies of the material surface, and examination of the degradation product accumulations, indicate that degradation has negligible impact on cytotoxicity of these materials. STATEMENT OF SIGNIFICANCE: This paper presents an in-depth analysis on the degradation of porous, shape memory polyurethanes (SMPs), including traditional surface characterization as well as model degradation compounds with absolute quantification. This combination of techniques allows for determination of rates of degradation as well as accumulation of individual degradation products. These behaviors are used for in vivo-in vitro comparisons for determination of real time degradation rates. Previous studies have primarily been limited to surface characterization without examination of degradation products and accumulation rates. To our knowledge, our work presents a unique example where a range of material scales (atomistic-scale model compounds along with macroscopic porous SMPs) are used in conjunction with ex planted samples for calculation of degradation rates and toxicological risk.


Subject(s)
Biocompatible Materials , Biodegradable Plastics , Materials Testing , Polyurethanes , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacokinetics , Biocompatible Materials/pharmacology , Biodegradable Plastics/chemistry , Biodegradable Plastics/pharmacokinetics , Biodegradable Plastics/pharmacology , Mice , NIH 3T3 Cells , Oxidation-Reduction , Polyurethanes/chemistry , Polyurethanes/pharmacokinetics , Polyurethanes/pharmacology
13.
Biomacromolecules ; 17(11): 3516-3523, 2016 11 14.
Article in English | MEDLINE | ID: mdl-27656910

ABSTRACT

Click chemistry is a versatile tool for the synthesis and functionalization of polymeric biomaterials. Here, we describe a versatile new strategy for producing bioactive, protein-functionalized poly(ethylene glycol) (PEG) hydrogel microparticles that is based on sequential thiol-ene and tetrazine click reactions. Briefly, tetra-functional PEG-norbornene macromer and dithiothreitol (SH) cross-linker were combined at a 0.75:1 [SH]:[norbornene] ratio, emulsified in a continuous Dextran phase, and then photopolymerized to form PEG hydrogel microparticles that varied from 8 to 30 µm in diameter, depending on the PEG concentration used. Subsequently, tetrazine-functionalized protein was conjugated to unreacted norbornene groups in the PEG microparticles. Tetrazine-mediated protein tethering to the microparticles was first demonstrated using fluorescein-labeled ovalbumin as a model protein. Subsequently, bioactive protein tethering was demonstrated using alkaline phosphatase (ALP) and glucose oxidase (GOx). Enzyme activity assays demonstrated that both ALP and GOx maintained their bioactivity and imparted tunable bioactivity to the microparticles that depended on the amount of enzyme added. ALP-functionalized microparticles were also observed to initiate calcium phosphate mineralization in vitro when incubated with calcium glycerophosphate. Collectively, these results show that protein-functionalized hydrogel microparticles with tunable bioactive properties can be easily synthesized using sequential click chemistry reactions. This approach has potential for future applications in tissue engineering, drug delivery, and biosensing.


Subject(s)
Alkaline Phosphatase/chemistry , Drug Delivery Systems , Glucose Oxidase/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Biocompatible Materials/therapeutic use , Click Chemistry , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemical synthesis , Hydrogel, Polyethylene Glycol Dimethacrylate/therapeutic use , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polymerization
14.
Macromol Rapid Commun ; 37(23): 1945-1951, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27568830

ABSTRACT

Polyurethane shape memory polymer (SMP) foams are proposed for use as thrombogenic scaffolds to improve the treatment of vascular defects, such as cerebral aneurysms. However, gas blown SMP foams inherently have membranes between pores, which can limit their performance as embolic tissue scaffolds. Reticulation, or the removal of membranes between adjacent foam pores, is advantageous for improving device performance by increasing blood permeability and cellular infiltration. This work characterizes the effects of cold gas plasma reticulation processes on bulk polyurethane SMP films and foams. Plasma-induced changes on material properties are characterized using scanning electron microscopy, uniaxial tensile testing, goniometry, and free strain recovery experiments. Device specific performance is characterized in terms of permeability, platelet attachment, and cell-material interactions. Overall, plasma reticulated SMP scaffolds show promise as embolic tissue scaffolds due to increased bulk permeability, retained thrombogenicity, and favorable cell-material interactions.


Subject(s)
Intracranial Aneurysm/pathology , Plasma Gases/chemistry , Polyurethanes/chemistry , Tissue Scaffolds/chemistry , Animals , Cells, Cultured , Mice , NIH 3T3 Cells , Particle Size , Plasma Gases/chemical synthesis , Polyurethanes/chemical synthesis , Surface Properties
15.
Nanoscale ; 8(24): 12362-72, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27270567

ABSTRACT

Injectable hydrogels are investigated for cell encapsulation and delivery as they can shield cells from high shear forces. One of the approaches to obtain injectable hydrogels is to reinforce polymeric networks with high aspect ratio nanoparticles such as two-dimensional (2D) nanomaterials. 2D nanomaterials are an emerging class of ultrathin materials with a high degree of anisotropy and they strongly interact with polymers resulting in the formation of shear-thinning hydrogels. Here, we present 2D nanosilicate reinforced kappa-carrageenan (κCA) hydrogels for cellular delivery. κCA is a natural polysaccharide that resembles native glycosaminoglycans and can form brittle hydrogels via ionic crosslinking. The chemical modification of κCA with photocrosslinkable methacrylate groups renders the formation of a covalently crosslinked network (MκCA). Reinforcing the MκCA with 2D nanosilicates results in shear-thinning characteristics, and enhanced mechanical stiffness, elastomeric properties, and physiological stability. The shear-thinning characteristics of nanocomposite hydrogels are investigated for human mesenchymal stem cell (hMSC) delivery. The hMSCs showed high cell viability after injection and encapsulated cells showed a circular morphology. The proposed shear-thinning nanoengineered hydrogels can be used for cell delivery for cartilage tissue regeneration and 3D bioprinting.


Subject(s)
Carrageenan/chemistry , Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Nanocomposites/chemistry , Cartilage , Cells, Cultured , Humans
16.
ACS Nano ; 10(1): 246-56, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26670176

ABSTRACT

Although hydrogels are able to mimic native tissue microenvironments, their utility for biomedical applications is severely hampered due to limited mechanical stiffness and low toughness. Despite recent progress in designing stiff and tough hydrogels, it is still challenging to achieve a cell-friendly, high modulus construct. Here, we report a highly efficient method to reinforce collagen-based hydrogels using extremely low concentrations of a nanoparticulate-reinforcing agent that acts as a cross-link epicenter. Extraordinarily, the addition of these nanoparticles at a 10 000-fold lower concentration relative to polymer resulted in a more than 10-fold increase in mechanical stiffness and a 20-fold increase in toughness. We attribute the high stiffness of the nanocomposite network to the chemical functionality of the nanoparticles, which enabled the cross-linking of multiple polymeric chains to the nanoparticle surface. The mechanical stiffness of the nanoengineered hydrogel can be tailored between 0.2 and 200 kPa simply by manipulating the size of the nanoparticles (4, 8, and 12 nm), as well as the concentrations of the nanoparticles and polymer. Moreover, cells can be easily encapsulated within the nanoparticulate-reinforced hydrogel network, showing high viability. In addition, encapsulated cells were able to sense and respond to matrix stiffness. Overall, these results demonstrate a facile approach to modulate the mechanical stiffness of collagen-based hydrogels and may have broad utility for various biomedical applications, including use as tissue-engineered scaffolds and cell/protein delivery vehicles.


Subject(s)
Hardness , Hydrogels/chemistry , Nanocomposites/chemistry , Nanoparticles/chemistry , Animals , Cells, Immobilized/cytology , Cells, Immobilized/physiology , Collagen/chemistry , Dopamine/chemistry , Ferrosoferric Oxide/chemistry , Gelatin/chemistry , Hardness Tests , Humans , Materials Testing , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Methacrylates/chemistry , Mice , NIH 3T3 Cells , Nanocomposites/ultrastructure , Nanoparticles/ultrastructure , Particle Size , Polyethylene Glycols/chemistry
17.
Adv Healthc Mater ; 4(11): 1600-27, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26010739

ABSTRACT

Recent progress in nanotechnology has stimulated the development of multifunctional biomaterials for tissue engineering applications. Synergistic interactions between nanomaterials and stem cell engineering offer numerous possibilities to address some of the daunting challenges in regenerative medicine, such as controlling trigger differentiation, immune reactions, limited supply of stem cells, and engineering complex tissue structures. Specifically, the interactions between stem cells and their microenvironment play key roles in controlling stem cell fate, which underlines therapeutic success. However, the interactions between nanomaterials and stem cells are not well understood, and the effects of the nanomaterials shape, surface morphology, and chemical functionality on cellular processes need critical evaluation. In this Review, focus is put on recent development in nanomaterial-stem cell interactions, with specific emphasis on their application in regenerative medicine. Further, the emerging technologies based on nanomaterials developed over the past decade for stem cell engineering are reviewed, as well as the potential applications of these nanomaterials in tissue regeneration, stem cell isolation, and drug/gene delivery. It is anticipated that the enhanced understanding of nanomaterial-stem cell interactions will facilitate improved biomaterial design for a range of biomedical and biotechnological applications.


Subject(s)
Nanostructures/chemistry , Stem Cells/metabolism , Drug Carriers/chemistry , Humans , Regenerative Medicine , Stem Cell Transplantation , Stem Cells/cytology , Tissue Engineering , Tissue Scaffolds
SELECTION OF CITATIONS
SEARCH DETAIL
...