Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Soft Matter ; 19(33): 6224-6233, 2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37493066

ABSTRACT

Here, we designed enzymatically degradable hydrogels with tunable mesh sizes and crosslinking points to evaluate the effectiveness of network structure estimations in predicting dynamic mechanical properties and cargo retention or release. Poly(ethylene glycol) (PEG) hydrogels were prepared through a thiol-ene click reaction between four- or eight-arm PEG functionalized with vinyl sulfone and cysteine residues of collagenase-degradable peptides to create well-defined, homogenous, and robust materials with a range of mesh sizes estimated from the elasticity theory or Flory-Rehner theory. Time-dependent changes in mechanical properties associated with hydrogel degradation, i.e., dynamics of storage modulus, which is determined by the relationship between the hydrogel mesh and enzyme sizes, were characterized. The shear modulus G' decreased by enzyme addition, and the degradation rate decreased with the initial crosslinking density of the hydrogel. The degradation rate could also be controlled with the reactivity of peptide sequences against collagenase. With these findings, the retention and release of FITC-dextran were successfully controlled by tuning the mesh size and degradability of the hydrogel. This report provides useful insights for designing hydrogels as cell scaffolds or functional molecular delivery matrices with tunable dynamic mechanical properties and the resulting release of loaded drugs or proteins.


Subject(s)
Hydrogels , Polyethylene Glycols , Hydrogels/chemistry , Polyethylene Glycols/chemistry , Peptides/chemistry , Elasticity
2.
Langmuir ; 38(17): 5307-5314, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35143208

ABSTRACT

There is growing evidence that cellular functions are regulated by the viscoelastic nature of surrounding matrices. This study aimed to investigate the impact of interfacial viscoelasticity on adhesion and epithelial-mesenchymal transition (EMT) behaviors of epithelial cells. The interfacial viscoelasticity was manipulated using spin-coated thin films composed of copolymers of ε-caprolactone and d,l-lactide photo-cross-linked with benzophenone, whose mechanical properties were characterized using atomic force microscopy and a rheometer. The critical range for the morphological transition of epithelial Madin-Darby canine kidney (MDCK) cells was of the order of 102 ms relaxation time, which was 1-2 orders of magnitude smaller than the relaxation times reported (10-102 s). An analysis of strain rate-dependent viscoelastic properties revealed that the difference was caused by the different strain rate/frequency used for the mechanical characterization of the interface and bulk. Furthermore, decoupling of the interfacial viscous and elastic terms demonstrated that E/N-cadherin expression levels were regulated differently by interfacial relaxation and elasticity. These results confirm the significance of precise manipulation and characterization of interfacial viscoelasticity in mechanobiology studies on EMT progression.


Subject(s)
Epithelial-Mesenchymal Transition , Animals , Dogs , Elasticity , Madin Darby Canine Kidney Cells , Microscopy, Atomic Force , Viscosity
3.
Int J Mol Sci ; 23(23)2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36498966

ABSTRACT

The reactivating of disseminated dormant breast cancer cells in a soft viscoelastic matrix is mostly correlated with metastasis. Metastasis occurs due to rapid stress relaxation owing to matrix remodeling. Here, we demonstrate the possibility of promoting the permanent cell cycle arrest of breast cancer cells on a viscoelastic liquid substrate. By controlling the molecular weight of the hydrophobic molten polymer, poly(ε-caprolactone-co-D,L-lactide) within 35-63 g/mol, this study highlights that MCF7 cells can sense a 1000 times narrower relaxation time range (80-290 ms) compared to other studies by using a crosslinked hydrogel system. We propose that the rapid bulk relaxation response of the substrate promotes more reactive oxygen species generation in the formed semi-3D multicellular aggregates of breast cancer cells. Our finding sheds light on the potential role of bulk stress relaxation in a viscous-dominant viscoelastic matrix in controlling the cell cycle arrest depth of breast cancer cells.


Subject(s)
Breast Neoplasms , Humans , Female , Hydrogels/chemistry , Viscosity , Cell Cycle Checkpoints , Oxidative Stress
4.
Sci Technol Adv Mater ; 22(1): 522-531, 2021 Jun 24.
Article in English | MEDLINE | ID: mdl-34220340

ABSTRACT

We reports a novel thermally enhanced drug release system synthesized via a dynamic Diels-Alder (DA) reaction to develop chemotherapy for pancreatic cancer. The anticancer prodrug was designed by tethering gemcitabine (GEM) to poly(furfuryl methacrylate) (PFMA) via N-(3-maleimidopropionyloxy)succinimide as a linker by DA reaction (PFMA-L-GEM). The conversion rate of the DA reaction was found to be approximately 60% at room temperature for 120 h. The reversible deconstruction of the DA covalent bond in retro Diels-Alder (rDA) reaction was confirmed by proton nuclear magnetic resonance, and the reaction was significantly accelerated at 90 °C. A PFMA-LGEM film containing magnetic nanoparticles (MNPs) was prepared for thermally enhanced release of the drug via the rDA reaction. Drug release was initiated by heating MNPs by alternating magnetic field. This enables local heating within the film above the rDA reaction temperature while maintaining a constant surrounding medium temperature. The MNPs/PFMA-L-GEM film decreased the viability of pancreatic cancer cells by 49% over 24 h. Our results suggest that DA/rDA-based thermally enhanced drug release systems can serve as a local drug release platform and deliver the target drug within locally heated tissue, thereby improving the therapeutic efficiency and overcoming the side effects of conventional drugs used to treat pancreatic cancer.

5.
Int J Mol Sci ; 22(5)2021 Mar 03.
Article in English | MEDLINE | ID: mdl-33802613

ABSTRACT

This study demonstrates the rational fabrication of a magnetic composite nanofiber mesh that can achieve mutual synergy of hyperthermia, chemotherapy, and thermo-molecularly targeted therapy for highly potent therapeutic effects. The nanofiber is composed of biodegradable poly(ε-caprolactone) with doxorubicin, magnetic nanoparticles, and 17-allylamino-17-demethoxygeldanamycin. The nanofiber exhibits distinct hyperthermia, owing to the presence of magnetic nanoparticles upon exposure of the mesh to an alternating magnetic field, which causes heat-induced cell killing as well as enhanced chemotherapeutic efficiency of doxorubicin. The effectiveness of hyperthermia is further enhanced through the inhibition of heat shock protein activity after hyperthermia by releasing the inhibitor 17-allylamino-17-demethoxygeldanamycin. These findings represent a smart nanofiber system for potent cancer therapy and may provide a new approach for the development of localized medication delivery.


Subject(s)
Benzoquinones/pharmacology , Delayed-Action Preparations/pharmacology , Doxorubicin/pharmacology , Hyperthermia/drug therapy , Lactams, Macrocyclic/pharmacology , Nanofibers/chemistry , Neoplasms/drug therapy , Benzoquinones/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Delayed-Action Preparations/chemistry , Doxorubicin/chemistry , Drug Liberation , Drug Synergism , Ferric Compounds/chemistry , Humans , Lactams, Macrocyclic/chemistry , MCF-7 Cells , Magnetics/methods , Magnetite Nanoparticles/chemistry
6.
Int J Mol Sci ; 21(5)2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32143443

ABSTRACT

BACKGROUND: We propose the potential studies on material fluidity to induce epithelial to mesenchymal transition (EMT) in MCF-7 cells. In this study, we examined for the first time the effect of material fluidity on EMT using poly(ε-caprolactone-co-D,L-lactide) (P(CL-co-DLLA)) with tunable elasticity and fluidity. METHODS: The fluidity was altered by chemically crosslinking the polymer networks. The crosslinked P(CL-co-DLLA) substrate showed a solid-like property with a stiffness of 261 kPa, while the non-crosslinked P(CL-co-DLLA) substrate of 100 units (high fluidity) and 500 units (low fluidity) existed in a quasi-liquid state with loss modulus of 33 kPa and 30.8 kPa, respectively, and storage modulus of 10.8 kPa and 20.1 kPa, respectively. RESULTS: We observed that MCF-7 cells on low fluidic substrates decreased the expression of E-cadherin, an epithelial marker, and increased expression of vimentin, a mesenchymal marker. This showed that the cells lose their epithelial phenotype and gain a mesenchymal property. On the other hand, MCF-7 cells on high fluidic substrates maintained their epithelial phenotype, suggesting that the cells did not undergo EMT. CONCLUSION: Considering these results as the fundamental information for material fluidity induced EMT, our system could be used to regulate the degree of EMT by turning the fluidity of the material.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Polyesters/metabolism , Antigens, CD/metabolism , Biocompatible Materials , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Humans , MCF-7 Cells , Mesenchymal Stem Cells/metabolism , Microscopy, Confocal , Molecular Weight , Pressure , Signal Transduction , Transforming Growth Factor beta/metabolism , Vimentin/metabolism
7.
Small ; 15(5): e1804640, 2019 02.
Article in English | MEDLINE | ID: mdl-30624030

ABSTRACT

Mechanical cues of cellular microenvironments can modulate cell functions including cell spreading and differentiation. Most studies of cellular functions are performed using a solid substrate, and it is thought that cells cannot spread on fluid substrates because of rapid relaxation, which cannot resist against actomyosin-based cell contractility. Here, the spreading and growth of anchorage-dependent cells such as human mesenchymal stem cells at the liquid interface between a perfluorocarbon fluid and the culture medium are observed. It is demonstrated that a monomolecular protein nanosheet self-assembled at a fluid interface is sufficiently rigid to support cell spreading without additional treatment. Fine tuning of the packing of these proteins at the liquid interface permits tailoring of the mechanics of the protein layer, ultimately allowing for the regulation of cell spreading. The greater stiffness of the protein nanosheets triggers cell spreading, adhesion growth, and yes-associated protein nuclear translocation. Cell behavior at the fluid interface is explained within the framework of the molecular clutch model. In addition, the freestanding ultrathin protein nanosheets are extremely flexible, easily deformed, and perceived by cells as being much softer. The findings are expected to provide a new perspective for insights into cell-material interactions.


Subject(s)
Mechanotransduction, Cellular , Mesenchymal Stem Cells/cytology , Proteins/metabolism , Cell Culture Techniques , Fluorocarbons/chemistry , Focal Adhesions , Humans , Mesenchymal Stem Cells/metabolism
8.
Sci Technol Adv Mater ; 20(1): 291-304, 2019.
Article in English | MEDLINE | ID: mdl-30956733

ABSTRACT

Combining immunotherapeutic and radiotherapeutic technique has recently attracted much attention for advancing cancer treatment. If boron-incorporated hemagglutinating virus of Japan-envelope (HVJ-E) having high membrane fusion ability can be used as a boron delivery agent in boron neutron capture therapy (BNCT), a radical synergistic improvement of boron accumulation efficiency into tumor cells and antitumor immunity may be induced. In this study, we aimed to develop novel boron-containing biocompatible polymers modified onto HVJ-E surfaces. The copolymer consisting of 2-methacryloyloxyethyl phosphorylcholine (MPC) and methacrylamide benzoxaborole (MAAmBO), poly[MPC-co-MAAmBO], was successfully synthesized by using a simple free radical polymerization. The molecular structures and molecular weight of the poly[MPC-co-MAAmBO] copolymer were characterized by nuclear magnetic resonance and matrix-assisted laser desorption ionization time-of-flight mass spectrometry, respectively. The poly[MPC-co-MAAmBO] was coated onto the HVJ-E surface via the chemical bonding between the MAAmBO moiety and the sugar moiety of HVJ-E. DLS, AFM, UV-Vis, and fluorescence measurements clarified that the size of the poly[MPC-co-MAAmBO]-coated HVJ-E, HVJ-E/p[MPC-MAAmBO], to be about 130 ~ 150 nm in diameter, and that the polymer having 9.82 × 106 ~ 7 boron atoms was steadily coated on a single HVJ-E particle. Moreover, cellular uptake of poly[MPC-co-MAAmBO] could be demonstrated without cytotoxicity, and the hemolysis could be successfully suppressed by 20%. These results indicate that the HVJ-E/p[MPC-MAAmBO] may be used as boron nanocarriers in a combination of immunotherapy with BNCT.

9.
Prog Polym Sci ; 65: 53-82, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28522885

ABSTRACT

Human tissues are sophisticated ensembles of many distinct cell types embedded in the complex, but well-defined, structures of the extracellular matrix (ECM). Dynamic biochemical, physicochemical, and mechano-structural changes in the ECM define and regulate tissue-specific cell behaviors. To recapitulate this complex environment in vitro, dynamic polymer-based biomaterials have emerged as powerful tools to probe and direct active changes in cell function. The rapid evolution of polymerization chemistries, structural modulation, and processing technologies, as well as the incorporation of stimuli-responsiveness, now permit synthetic microenvironments to capture much of the dynamic complexity of native tissue. These platforms are comprised not only of natural polymers chemically and molecularly similar to ECM, but those fully synthetic in origin. Here, we review recent in vitro efforts to mimic the dynamic microenvironment comprising native tissue ECM from the viewpoint of material design. We also discuss how these dynamic polymer-based biomaterials are being used in fundamental cell mechanobiology studies, as well as towards efforts in tissue engineering and regenerative medicine.

10.
J Nanosci Nanotechnol ; 15(10): 7971-5, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26726449

ABSTRACT

In this study, we present anti-cancer drug containing nanofiber-mediated gene delivery to treat liver cancer. Electro-spun nanofibers have big potential for local delivery and sustained release of therapeutic gene and drugs. We reported a temperature-responsive nanofibers mainly compounded by branched poly(ε-caprolactone) (PCL) macro-monomers and anti-cancer drug paclitaxel. The nanofiber could be administrated into liver tumors to dramatically hinder their growth and prevent their metastasis. As a result, paclitaxel encapsulated PCL (PTX/PCL) nanofibers with diameters of around several tens nanometers to 10 nm were successfully obtained by electro-spinning and observed in scanning electron microscopy (SEM). Nanoparticles composed of disulfide cross-linked branched PEI (ssPEI) and anti-cancer therapeutic gene miRNA-145 were complexed based on the electrostatic interaction and coated over the paclitaxel-loaded nanofiber. MicroRNA 145/ssPEI nanoparticles (MSNs) immobilized on the PTX/PCL nanofiber showed time-dependent sustained release of the microRNA for enhanced uptake in neighboring liver cancer cells without any noticeable cytotoxicity. From this study we are expecting a synergistic effect on the cancer cell suppression since we have combined the drug and gene delivery. This approach uses the nanofibers and nanoparticles together for the treatment of cancer and the detailed investigation in vitro and in vivo must be conducted for the practicality of this study. The polymer is biodegradable and the toxicity issues must be cleared by our approach.


Subject(s)
Drug Delivery Systems/methods , Gene Transfer Techniques , Genetic Therapy/methods , Liver Neoplasms , Nanofibers/chemistry , Polyesters , Humans , Liver Neoplasms/genetics , Liver Neoplasms/therapy , MicroRNAs/genetics , Paclitaxel/chemistry , Paclitaxel/pharmacology , Polyesters/chemistry , Polyesters/pharmacology
11.
J Nanosci Nanotechnol ; 14(4): 3193-201, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24734754

ABSTRACT

(Apo)ferritins are cage-shaped proteins which have recently received a great deal of attention because the inner cavity of the protein shell can be used as a size-restricted reaction field for the synthesis of nanomaterials. The biomineralization behavior and inorganic nanoparticle (NP) synthesis mechanism of (apo)ferritin in solution systems have been studied but the mineralization behavior of (apo)ferritin on the substrates has not yet been well studied. Here, we conducted quantitative and kinetic analyses of the mineralization behavior of immobilized (apo)ferritin on a polyelectrolyte multilayer (PEM) using quartz crystal microbalance (QCM), scanning electron microscopy (SEM), and X-ray photoelectron spectroscopy (XPS) techniques. We demonstrated that the (apo)ferritin immobilized on a substrate synthesizes a ferrihydrite core within the confines of the protein cage; similar to a solution dispersed system. In addition, we applied a ferritin/apoferritin blended monolayer to the study of iron mineralization and revealed that biomineralization in this system is spatially selective. It is important to understand the mineralization mechanisms for the synthesis of other functional NPs as this approach has potential for a broad range of magnetic, catalytic, and biomedical sensing applications.


Subject(s)
Ferritins/chemistry , Immobilized Proteins/chemistry , Iron/chemistry , Minerals/chemistry , Adsorption , Animals , Apoproteins/chemistry , Ferric Compounds/chemistry , Horses , Microscopy, Electron, Scanning , Photoelectron Spectroscopy , Quartz Crystal Microbalance Techniques
12.
Int J Mol Sci ; 15(1): 1511-24, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24451135

ABSTRACT

We developed a dynamic cell culture platform with dynamically tunable nano-roughness and elasticity. Temperature-responsive poly(ε-caprolactone) (PCL) films were successfully prepared by crosslinking linear and tetra-branched PCL macromonomers. By optimizing the mixing ratios, the crystal-amorphous transition temperature (Tm) of the crosslinked film was adjusted to the biological relevant temperature (~33 °C). While the crosslinked films are relatively stiff (50 MPa) below the Tm, they suddenly become soft (1 MPa) above the Tm. Correspondingly, roughness of the surface was decreased from 63.4-12.4 nm. It is noted that the surface wettability was independent of temperature. To investigate the role of dynamic surface roughness and elasticity on cell adhesion, cells were seeded on PCL films at 32 °C. Interestingly, spread myoblasts on the film became rounded when temperature was suddenly increased to 37 °C, while significant changes in cell morphology were not observed for fibroblasts. These results indicate that cells can sense dynamic changes in the surrounding environment but the sensitivity depends on cell types.


Subject(s)
Biocompatible Materials/chemistry , Elasticity , Myoblasts/drug effects , Polyesters/chemistry , Animals , Biocompatible Materials/chemical synthesis , Biocompatible Materials/pharmacology , Cell Growth Processes , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Hot Temperature , Mice , Myoblasts/cytology , Myoblasts/physiology , NIH 3T3 Cells , Nanostructures/chemistry , Phase Transition , Polyesters/chemical synthesis , Polyesters/pharmacology , Rats , Surface Properties , Wettability
13.
Biomaterials ; 305: 122451, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38169189

ABSTRACT

Injectable hydrogels are promising carriers for cell delivery in regenerative medicine. However, injectable hydrogels composed of crosslinked polymer networks are often non-microporous and prevent biological communication with host tissues through signals, nutrients, oxygen, and cells, thereby limiting graft survival and tissue integration. Here we report injectable hydrogels with liquid-liquid phase separation-induced microcapillary networks (µCN) as stem cell-delivering scaffolds. The molecular modification of gelatin with hydrogen bonding moieties induced liquid-liquid phase separation when mixed with unmodified gelatin to form µCN structures in the hydrogels. Through spatiotemporally controlled covalent crosslinking and dissolution processes, porous µCN structures were formed in the hydrogels, which can enhance mass transport and cellular activity. The encapsulation of cells with injectable µCN hydrogels improved cellular spreading, migration, and proliferation. Transplantation of mesenchymal stem cells with injectable µCN hydrogels enhanced graft survival and recovered hindlimb ischemia by enhancing material-tissue communication with biological signals and cells through µCN. This facile approach may serve as an advanced scaffold for improving stem cell transplantation therapies in regenerative medicine.


Subject(s)
Gelatin , Hydrogels , Animals , Gelatin/chemistry , Hydrogels/chemistry , Phase Separation , Stem Cell Transplantation
14.
Adv Mater ; 36(26): e2310105, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38234135

ABSTRACT

In sharp contrast to conventional solid/hydrogel platforms, water-immiscible liquids, such as perfluorocarbons and silicones, allow the adhesion of mammalian cells via protein nanolayers (PNLs) formed at the interface. However, fluorocarbons and silicones, which are typically used for liquid cell culture, possess only narrow ranges of physicochemical parameters and have not allowed for a wide variety of cell culturing environments. In this paper, it is proposed that water-immiscible ionic liquids (ILs) are a new family of liquid substrates with tunable physicochemical properties and high solvation capabilities. Tetraalkylphosphonium-based ILs are identified as non-cytotoxic ILs, whereon human mesenchymal stem cells are successfully cultured. By reducing the cation charge distribution, or ionicity, via alkyl chain elongation, the interface allows cell spreading with matured focal contacts. High-speed atomic force microscopy observations of the PNL formation process suggest that the cation charge distribution significantly altered the protein adsorption dynamics, which are associated with the degree of protein denaturation and the PNL mechanics. Moreover, by exploiting dissolution capability of ILs, an ion-gel cell scaffold is fabricated. This enables to further identify the significant contribution of bulk subphase mechanics to cellular mechanosensing in liquid-based culture scaffolds.


Subject(s)
Ionic Liquids , Mesenchymal Stem Cells , Tissue Scaffolds , Ionic Liquids/chemistry , Humans , Mesenchymal Stem Cells/cytology , Tissue Scaffolds/chemistry , Cell Adhesion/drug effects , Water/chemistry
15.
Langmuir ; 29(24): 7384-92, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23441859

ABSTRACT

In this study, we created a nanoscale layer of hyaluronic acid (HA) on the inactivated Hemagglutinating Virus of Japan envelope (HVJ-E) via a layer-by-layer (LbL) assembly technique for CD-44 targeted delivery. HVJ-E was selected as the template virus because it has shown a tumor-suppressing ability by eliciting inflammatory cytokine production in dendritic cells. Although it has been required to increase the tumor-targeting ability and reduce nonspecific binding because HVJ-E fuses with virtually all cells and induces hemagglutination in the bloodstream, complete modifications of single-envelope-type viruses with HA have been difficult. Therefore, we studied the surface ζ potential of HVJ-E at different pH values and carefully examined the deposition conditions for the first layer using three cationic polymers: poly-L-lysine (PLL), chitosan (CH), and glycol chitosan (GC). GC-coated HVJ-E particles showed the highest disperse ability under physiological pH and salt conditions without aggregation. An HA layer was then prepared via alternating deposition of HA and GC. The successive decoration of multilayers on HVJ-E has been confirmed by dynamic light scattering (DLS), ζ potentials, and transmission electron microscopy (TEM). An enzymatic degradation assay revealed that only the outermost HA layer was selectively degraded by hyaluronidase. However, entire layers were destabilized at lower pH. Therefore, the HA/GC-coated HVJ-E describe here can be thought of as a potential bomb for cancer immunotherapy because of the ability of targeting CD44 as well as the explosion of nanodecorated HA/GC layers at endosomal pH while preventing nonspecific binding at physiological pH and salt conditions such as in the bloodstream or normal tissues.


Subject(s)
Nanotechnology , Sendai virus/chemistry , Viral Envelope Proteins/chemistry , Hydrogen-Ion Concentration , Surface Properties
16.
Adv Mater ; 35(30): e2300486, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37102709

ABSTRACT

Cell-picking technology is essential for cell culturing. Although the recently developed tools enable single-cell-level picking, they rely on special skills or additional devices. In this work, a dry powder that encapsulates single to several cells with a >95% aqueous culture medium, thereby acting as a powerful cell-picking tool, is reported. The proposed "drycells" are formed by spraying a cell suspension onto a powder bed of hydrophobic fumed silica nanoparticles. The particles adsorb to the droplet surface and form a superhydrophobic shell, which prevents the drycells from coalescence. The number of encapsulated cells per drycell can be controlled by adjusting the drycell size and cell-suspension concentration. Moreover, it is possible to encapsulate a pair of normal or cancerous cells and create several cell colonies within a single drycell. A sieving process can be used to sort the drycells according to size. The size of the droplet can range from one to hundreds of micrometers. The drycells are sufficiently stiff to be collected using tweezers; however, centrifugation separates them into nanoparticles and cell-suspension layers, with the separated particles being recyclable. Various handling techniques, such as splitting coalescence and inner liquid replacement, can be used. It is believed that the application of the proposed drycells will greatly improve the accessibility and productivity of single-cell analysis.


Subject(s)
Nanoparticles , Powders , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Water/chemistry , Hydrophobic and Hydrophilic Interactions
17.
Nanoscale ; 15(47): 18980-18998, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37990550

ABSTRACT

Liquid marbles (LMs) are nonsticking droplets whose surfaces are covered with low-wettability particles. Owing to their high mobility, shape reconfigurability, and widely accessible liquid/particle possibilities, the research on LMs has flourished since 2001. Their physical properties, fabrication mechanisms, and functionalisation capabilities indicate their potential for various applications. This review summarises the fundamental properties of LMs, the recent advances (mainly works published in 2020-2023) in the concept of LMs, physical properties, formation methods, LM-templated material design, and biochemical applications. Finally, the potential development and variations of LMs are discussed.

18.
Int J Biol Macromol ; 253(Pt 3): 126880, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37709226

ABSTRACT

Highly efficient adsorbents are needed to remove uremic toxins and reduce the economic and societal burden of the current dialysis treatments in resource-limited environments. In this study, nanostructured porous carbon nanofibers with nitrogen-doped zeolites (NZ-PCNF) were prepared, by electrospinning zeolites with chitosan-poly(ethylene oxide) blends, followed by a one-step carbonization process, without further activation steps or aggressive chemical additives for N-doping. The results showed that N-zeolites were successfully integrated into an ultrafine carbon nanofiber network, with a uniform nanofiber diameter of approximately 25 nm, hierarchical porous structure (micro- and mesopores), and high specific surface area (639.29 m2/g), facilitating uremic toxin diffusion and adsorption. The self-N-doped structure in the NZ-PCNF removed more creatinine (∼1.8 times) than the porous carbon nanofibers when using the same weight of precursor materials. Cytotoxicity and hemolysis tests were performed to verify the safety of NZ-PCNF. This study provides a novel strategy for transforming chitosan-based materials into state-of-the-art porous carbon nanofiber/zeolite self-N-doped composites, affording an efficient bioderived adsorbent for the removal of uremic toxins in patients with chronic kidney disease.


Subject(s)
Chitosan , Nanofibers , Zeolites , Humans , Carbon/chemistry , Uremic Toxins , Nanofibers/chemistry , Porosity , Nitrogen/chemistry , Renal Dialysis
19.
Article in English | MEDLINE | ID: mdl-37930044

ABSTRACT

OBJECTIVES: The use of bone wax (BW) is controversial for sternal haemostasis because it increases the risk of wound infection and inhibits bone healing. We developed new waxy bone haemostatic agents made from biodegradable polymers containing peptides and evaluated them using rabbit models. METHODS: We designed 2 types of waxy bone haemostatic agents: peptide wax (PW) and non-peptide wax (NPW), which used poly(ε-caprolactone)-based biodegradable polymers with or without an osteogenesis-enhancing peptide, respectively. Rabbits were randomly divided into 4 groups based on treatment with BW, NPW, PW or no treatment. In a tibial defect model, the bleeding amount was measured and bone healing was evaluated by micro-computed tomography over 16 weeks. Bone healing in a median sternotomy model was assessed for 2 weeks using X-ray, micro-computed tomography, histological examination and flexural strength testing. RESULTS: The textures of PW and NPW (n = 12 each) were similar to that of BW and achieved a comparable degree of haemostasis. The crevice area of the sternal fracture line in the BW group was significantly larger than that in other groups (n = 10 each). The PW group demonstrated the strongest sternal flexural strength (n = 10), with complete tibial healing at 16 weeks. No groups exhibited wound infection, including osteomyelitis. CONCLUSIONS: Waxy biodegradable haemostatic agents showed satisfactory results in haemostasis and bone healing in rabbit models and may be an effective alternative to BW.

20.
Sci Technol Adv Mater ; 13(6): 064202, 2012 Dec.
Article in English | MEDLINE | ID: mdl-27877529

ABSTRACT

We demonstrate a timed explosive drug release from smart pH-responsive hydrogels by utilizing a phototriggered spatial pH-jump reaction. A photoinitiated proton-releasing reaction of o-nitrobenzaldehyde (o-NBA) was integrated into poly(N-isopropylacrylamide-co-2-carboxyisopropylacrylamide) (P(NIPAAm-co-CIPAAm)) hydrogels. o-NBA-hydrogels demonstrated the rapid release of protons upon UV irradiation, allowing the pH inside the gel to decrease to below the pKa value of P(NIPAAm-co-CIPAAm). The generated protons diffused gradually toward the non-illuminated area, and the diffusion kinetics could be controlled by adjusting the UV irradiation time and intensity. After irradiation, we observed the enhanced release of entrapped L-3,4-dihydroxyphenylalanine (DOPA) from the gels, which was driven by the dissociation of DOPA from CIPAAm. Local UV irradiation also triggered the release of DOPA from the non-illuminated area in the gel via the diffusion of protons. Conventional systems can activate only the illuminated region, and their response is discontinuous when the light is turned off. The ability of the proposed pH-jump system to permit gradual activation via proton diffusion may be beneficial for the design of predictive and programmable devices for drug delivery.

SELECTION OF CITATIONS
SEARCH DETAIL