Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
bioRxiv ; 2024 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-38766224

RESUMO

Studies on the dynamics of single cell phenotyping have been hampered by the lack of quantitative high-throughput metabolism assays. Extracellular acidification, a prominent phenotype, yields significant insights into cellular metabolism, including tumorigenicity. Here, we develop a versatile microfluidic system for single cell optical pH analysis (SCO-pH), which compartmentalizes single cells in 140-pL droplets and immobilizes approximately 40,000 droplets in a two-dimensional array for temporal extracellular pH analysis. SCO-pH distinguishes cells undergoing hyperglycolysis induced by oligomycin A from untreated cells by monitoring their extracellular acidification. To facilitate pH sensing in each droplet, we encapsulate a cell-impermeable pH probe whose fluorescence intensities are quantified. Using this approach, we can differentiate hyperglycolytic cells and concurrently observe single cell heterogeneity in extracellular acidification dynamics. This high-throughput system will be useful in applications that require dynamic phenotyping of single cells with significant heterogeneity.

3.
Chem Eng J ; 446: 137054, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-35601362

RESUMO

Viruses/bacteria outbreaks have motivated us to develop a fabric that will inhibit their transmission with high potency and long-term stability. By creating a metal-ion-rich surface onto polyester (PET) fabric, a method is found to inhibit hospital-acquired infections by immobilizing microorganisms on its surface. ZIF-8 and APTES are utilized to overcome the limitations associated with non-uniform distribution, weak biomolecule interaction, and ion leaching on surfaces. Modified surfaces employing APTES enhance ZIF-8 nucleation by generating a monolayer of self-assembled amine molecules. An in-situ growth approach is then used to produce evenly distributed ZIF-8 throughout it. In comparison with pristine fabric, this large amount of zinc obtained from the modification of the fabric has a higher affinity for interacting with membranes of microorganisms, leading to a 4.55-fold increase in coronavirus spike-glycoprotein immobilization. A series of binding ability stability tests on the surface demonstrate high efficiency of immobilization, >90%, of viruses and model proteins. The immobilization capacity of the modification fabric stayed unchanged after durability testing, demonstrating its durability and stability. It has also been found that this fabric surface modification approach has maintained air/vapor transmittance and air permeability levels comparable to pristine fabrics. These results strongly advocate this developed fabric has the potential for use as an outer layer of face masks or as a medical gown to prevent hospital-acquired infections.

4.
Adv Healthc Mater ; 11(6): e2102095, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34826360

RESUMO

The delivery of nitric oxide (NO)-an intrinsic cellular signaling molecule-is promising for disease treatment, in particular to vascular diseases, due to its endothelial-derived inherent nature. The limited diffusion distance of labile NO prompts researchers to develop various carriers and targeting methods for specific sites. In contrast to the apoptotic effect of NO, such as anticancer, delivering low NO concentration at the desired targeting area is still intricate in a physiological environment. In this study, the layer-by-layer assembled nanocoating is leveraged to develop a direct NO delivery platform to individual endothelial cells (ECs). NO can be localized to individual ECs via S-nitrosothiol-bound polyacrylic acid which is a polymer directly providing an endothelial-like constant level of NO. To increase angiogenic activation along with NO, VEGF is additionally applied to specific receptors on the cell surface. Notably, the survival and proliferation of ECs are significantly increased by a synergistic effect of NO and VEGF co-localized via nanocoating. Furthermore, the nanocoating remarkably promoted cell migration and tubule formation-prerequisites of angiogenesis. The proposed unique technology based on nanocoating demonstrates great potential for conferring desired angiogenic functions to individual ECs through efficient NO delivery.


Assuntos
Células Endoteliais/fisiologia , Neovascularização Fisiológica , Óxido Nítrico/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Movimento Celular , Células Endoteliais/citologia , Humanos , Óxido Nítrico/metabolismo
5.
Chem Eng J ; 426: 130763, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131388

RESUMO

Infectious pollutants bioaerosols can threaten human public health. In particular, the indoor environment provides a unique exposure situation to induce infection through airborne transmission like SARS-CoV-2. To prevent the infection from spreading, personal protective equipment or indoor air purification is necessary. However, it has been discovered that the conventional filter can become contaminated by pathogen-containing aerosols, meaning that advanced filtering and self-sterilization systems are required. Here, we fabricate a multilayered nanocoating around the fabric using laponite (LAP) with Cu2+ ions (LAP-Cu2+ nanocoating) two contradictory functions in one system: trapping proteinaceous pathogens and antibacterial effect. Due to the strong LAP-protein interaction, albumin and spike protein (S-protein) are trapped into the fabric when proteins are sprayed using a nebulizer. The protein-blocking performance of the nanocoated fabric is 9.55-fold higher than bare fabric. These trapping capacities are retained after rinsing and repeated adsorption cycles, showing reproducibility for air filtration. Even though the protein-binding occurred, the LAP-Cu2+ fabric indicates antibacterial effect. LAP-Cu2+ fabric has an equivalent air and water transmittance rate to that of bare fabric with a stability under physiological environment. Therefore, given its excellent "Spear-and-shield" functions, the proposed LAP-Cu2+ fabric shows great potential for use in filter and masks during the viral pandemic.

6.
J Hazard Mater ; 414: 125472, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-33640729

RESUMO

The potential health hazards of particulates, such as micro/nano-sized plastics and carbon materials have recently received extensive attention. However, their toxicological properties in association with stem cell differentiation is still relatively unexplored. In this study, we elucidated the cytotoxic effects of 2D graphene oxide (GO), in relation to differentiation of human induced pluripotent stem cells (hiPSCs). Supplementation of GO to hiPSCs demonstrated uptake of GO through the plasma membrane and intracellular accumulation was observed. Increasing the concentration of GO led to reduced viability and increased likelihood of hiPSC colony detachment. Moreover, treatment of GO resulted in significant loss in pluripotency markers, OCT-4 and NANOG. In particular, when hiPSCs were cultured with GO in cardiomyocyte induction medium, upregulation of cardiomyocyte marker, NKX2.5, along with observation of early triggering of differentiation were observed. Taken together, our results highlight the risk in the uptake and accumulation of GO on the stem cell development by unwanted loss in pluripotency and accelerated initiation of differentiation.


Assuntos
Grafite , Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Diferenciação Celular , Grafite/toxicidade , Humanos
7.
Biomacromolecules ; 22(3): 1312-1322, 2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33617240

RESUMO

Implant-derived bacterial infection is a prevalent cause of diseases, and no antibacterial coating currently exists that is biocompatible and that does not induce multidrug resistance. To this end, nitric oxide (NO) has been emerging as an effective antimicrobial agent that acts on a broad range of bacteria and elicits no known resistance. Here, a method for accelerating NO release from multilayered nanofilms has been developed for facilitating antibacterial activity. A previously reported multilayered nanofilm (nbi film) was fabricated by alternative deposition of branched polyethyleneimine (BPEI) and alginate via the layer-by-layer assembly method. N-Diazeniumdiolate, a chemical NO donor, was synthesized at the secondary amine moiety of BPEI within the film (nbi/NO film). Cu(II) ions can be incorporated into the film by forming chelating compounds with unreacted amines that have not been converted to NO donors. The increase of the amine protonation state in the chelate caused destabilization of the NO donor by reducing hydrogen bonding between the deprotonated amine and the NO donor. Thus, the Cu(II) ion-embedding film presented accelerated NO release and was further subjected to antibacterial testing to demonstrate the correlation between the NO release rate and the antibacterial activity. This study aimed to establish a novel paradigm for NO-releasing material design based on multilayered nanofilms by presenting the correlation between the NO release rate and the antibacterial effect.


Assuntos
Anti-Infecciosos , Óxido Nítrico , Aceleração , Antibacterianos/farmacologia , Íons
8.
J Glaucoma ; 30(2): 195-202, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33086260

RESUMO

PRECIS: Silica nanoparticles (SiNPs), which are potential drug carriers for glaucoma treatment, may induce mild dose-dependent cytotoxicity but not so severe as to compromise a mammalian target of rapamycin (mTOR) pathway in immortalized trabecular meshwork (TM) cells. PURPOSE: Nanoparticle-based ophthalmic drug delivery is a promising field of drug development. In this study, we evaluated the effect of nonporous SiNPs on human TM cells. METHODS: TM cells were exposed to different concentrations (0 to 100 µg/mL) of SiNPs (50, 100, and 150 nm) for up to 48 hours. Transmission electron microscopy confirmed the intracellular distribution of SiNPs. Cellular viability assay, reactive oxygen species generation, autophagy, and activation of the mTOR pathway were evaluated. Histologic analysis of the TM structure was performed after intracameral injection of SiNPs (0.05 mL of 200 µg/mL concentration) in rabbits. RESULTS: SiNPs were taken up by TM cells and localized in the cytoplasm. Neither nuclear entry nor mitochondrial damage was observed. SiNPs induced a mild but dose-dependent increase of lactate dehydrogenase. However, neither increase of intracellular reactive oxygen species levels nor apoptosis was observed after SiNPs exposure. Significant coactivation of autophagy and the mTOR pathway were observed with exposure to SiNPs. Aqueous plexus structure was well maintained without inflammation in rabbits after SiNPs exposure. CONCLUSIONS: SiNPs induce mild and dose-dependent cytotoxicity in TM cells. However, the toxicity level is not enough to compromise the mTOR pathway of TM cells and histologic structure of the aqueous plexus tissue.


Assuntos
Nanopartículas , Dióxido de Silício , Animais , Sobrevivência Celular , Humanos , Pressão Intraocular , Nanopartículas/toxicidade , Coelhos , Dióxido de Silício/toxicidade , Malha Trabecular
9.
Biomacromolecules ; 21(12): 4972-4979, 2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33147008

RESUMO

Nitric oxide (NO) plays a key role in several physiological functions such as inflammatory responses and immune regulation. However, despite its beneficial functions, the short half-life and diffusion radius limit NO availability in biomedical applications. Hence, controlled release is important to achieve the desired therapeutic effects with exogenous NO delivery. In this study, we fabricated a poly(lactic-co-glycolic acid) (PLGA)-based NO delivery system to release NO in a sustained manner under physiological conditions. To prevent an initial burst release, branched polyethylenimine diazeniumdiolate (BPEI/NONOate), a pH-responsive NO donor, was encapsulated into the hydrophilic core of PLGA nanoparticles. Furthermore, low concentrations of NO released at a consistent level via a stabilization effect obtained as amine groups of BPEI/NONOate interacted with the nearby NONOate. Using the controlled-release profiles, we successfully regulated the inflammatory response in lipopolysaccharide-stimulated peripheral blood mononuclear cells. This work demonstrates the potential of a NO delivery carrier in the regulation of inflammation.


Assuntos
Anti-Inflamatórios , Nanopartículas , Óxido Nítrico , Ácido Poliglicólico , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacologia , Glicóis , Ácido Láctico , Leucócitos Mononucleares , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
10.
Mol Pharm ; 17(2): 656-665, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31913044

RESUMO

Incidence ofglaucoma, a severe disease leading to irreversible loss of vision, is increasing with global aging populations. Lowering intraocular pressure (IOP) is the only proven treatment method for glaucoma. Nitric oxide (NO) is an emerging material targeting the conventional outflow pathway by relaxing the trabecular meshwork (TM). However, there is little understanding on the NO level effective in IOP lowering without toxicity. Here, we report a novel long-term NO-releasing polydiazeniumdiolate (NOP) that enables lowering IOP via the conventional outflow pathway. NOP is composed of carbon-bound polydiazeniumdiolate, a stable NO donor moiety. NO release was monitored with accurate parameters by real-time detection of gas and analysis of the accumulated release profile. Based on the NO release information, the selected safe level of NOP exhibited effective TM relaxation and a potential IOP lowering effect in vivo without side effects. This work provides new insights into nitric oxide release behavior that should be considered for glaucoma treatment.


Assuntos
Compostos Azo/uso terapêutico , Glaucoma de Ângulo Aberto/tratamento farmacológico , Pressão Intraocular/efeitos dos fármacos , Doadores de Óxido Nítrico/uso terapêutico , Óxido Nítrico/uso terapêutico , Animais , Compostos Azo/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Humanos , Masculino , Óxido Nítrico/farmacologia , Doadores de Óxido Nítrico/farmacologia , Projetos Piloto , Coelhos , Pele/citologia , Malha Trabecular/citologia , Malha Trabecular/efeitos dos fármacos
11.
Sci Rep ; 9(1): 8308, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31165751

RESUMO

To utilize potentials of nitric oxide (NO) gas in anti-bacterial, anticancer, wound healing applications, numerous studies have been conducted to develop a NO delivery system in the past few decades. Even though a coating method and film types are essential to apply in biomedical device coating from previous NO delivery systems, release control from the coating system is still challenging. In this study, we introduced a multilayered polymeric coating system to overcome the uncontrollable NO release kinetics of film systems. We used biocompatible gelatin and tannic acid to construct a rough, porous structured film based on the layer-by-layer self-assembly method. The multilayered polymeric structure facilitated the controlled amount of NO release from (Gel/TA)n film and showed burst release in early period owing to their large surface area from the rough, porous structure. We synthesized the proton-responsive NO donor, N-diazeniumdiolate (NONOates), into the (Gel/TA)n film through a chemical reaction under high pressure NO gas. NO release profile was analyzed by a real-time NO analysis machine (NOA 280i). Then, the NO-releasing (Gel/TA)n film was tested its toxicity against human dermal fibroblast cells and bactericidal effects against Staphylococcus aureus.

12.
ACS Biomater Sci Eng ; 5(3): 1378-1383, 2019 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-33405613

RESUMO

Nitric oxide (NO) participates in various physiological and pathophysiological processes, for example, as a cell messenger and as an antimicrobial agent of the cell-mediated immune response. The development of NO-releasing materials to carry and deliver NO for biomedical applications has gained immense attention. NO-releasing perfluorooctane (PFO) microemulsion (ME) has been prepared using a simple and time-saving method. Perfluorocarbon (PFC) liquids are halogen-substituted carbon nonpolar oils with enhanced NO gas dissolution capacity. The solubility of NO in PFC liquids is higher than that in water-based fluids. Liquid-gas solubility is governed by Henry's Law. The cytotoxicity of the NO-unloaded and NO-loaded PFO MEs toward human dermal fibroblast (HDF) was evaluated. The results showed that the NO-loaded PFO ME was highly biocompatible. On the other hand, at high concentrations the NO-releasing PFO ME accelerated the bacteria (Staphylococcus aureus) death unlike the NO-unloaded PFO ME. Hence, NO-releasing PFO MEs are suitable for biomedical applications such as wound healing and antibacterial agents.

13.
Invest Ophthalmol Vis Sci ; 59(8): 3239-3248, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29971441

RESUMO

Purpose: Acanthamoeba keratitis is a well-known intractable corneal infectious disease. We investigated the anti-Acanthamoeba effect of exogenous nitric oxide (NO). Methods: Acanthamoeba castellanii was axenically cultured and exposed to various concentrations of NO donors, such as sodium nitrite, sodium nitroprusside (SNP), and NO-releasing silica nanoparticles (coated in branched polyethylene imine, size:100 nm), for 1 to 7 days (sodium nitrite and SNP: 0, 0.1, 1, 10, 100, and 1000 µM; silica nanoparticles: 0, 6.25, 12.5, 25, 50, and 100 µg/mL). Human corneal epithelial cells (HCECs) were cultured and exposed to sodium nitrite, SNP (0, 0.1, 1, 10, 100, and 1000 µM), and silica nanoparticles for 1, 2, and 3 days. Results: Sodium nitrite and SNP showed a dose-dependent inhibitory effect on A. castellanii viability. A more prominent inhibitory effect was observed with SNP (less than 10% of organisms survived at 7-day culture with 1000 µM) compared with sodium nitrite. However, more cytotoxicity on HCEC was observed with SNP. NO-releasing silica nanoparticles were successfully internalized into the amoebic cytoplasm and accumulated in large vacuoles. Although blank silica nanoparticles had no inhibitory effect on A. castellanii viability, NO-releasing silica nanoparticles showed a dose-dependent amoebicidal effect. Furthermore, no cystic transformation of A. castellanii was observed under a phase contrast microscope or transmission electron microscope after exogenous NO treatment. Conclusions: Our results demonstrated the anti-Acanthamoeba effect of exogenous NO. This finding suggests that NO-releasing drug platforms, including nano-carriers, can be a promising therapeutic strategy for Acanthamoeba keratitis.


Assuntos
Acanthamoeba castellanii/efeitos dos fármacos , Antiprotozoários/farmacologia , Sequestradores de Radicais Livres/farmacologia , Óxido Nítrico/farmacologia , Acanthamoeba castellanii/ultraestrutura , Animais , Sobrevivência Celular , Células Cultivadas , Relação Dose-Resposta a Droga , Epitélio Corneano/efeitos dos fármacos , Epitélio Corneano/ultraestrutura , Humanos , Microscopia Eletrônica de Transmissão , Microscopia de Contraste de Fase , Doadores de Óxido Nítrico/farmacologia
14.
RSC Adv ; 8(68): 38792-38800, 2018 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-35558288

RESUMO

Nitric oxide (NO) gas delivery has attracted extensive interest due to its endogenous therapeutic functions and potential biomedical applications for the treatment of various diseases. The important thing about NO delivery is the emission control due to the fast diffusion rate of gas molecules. To develop NO delivery platforms using macromolecules and to comprehend the chemical NO donor generation and release mechanisms, we studied branched polyethyleneimine/alginate (BPEI/ALG) nanoblended coatings fabricated by giving structural heterogeneity to the structure through a self-assembly process for the controlled release of gas molecules. NO release could be remarkably expected via the well-organized coating structures and explained by quantification of the NO donors. Taking advantage of these polymeric coatings, this process could be applied to the treatment of various diseases based on the biocompatibility of materials and the fine control of NO release rate and its amount.

15.
Sci Rep ; 7(1): 14566, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29109483

RESUMO

Nonporous silica nanoparticles (SiNPs) are promising drug carrier platforms for intraocular drug delivery. In this study, we investigated the safety of three different sizes of SiNPs (50, 100, and 150 nm) in a human corneal endothelial cell (HCEC) line, B4G12. The HCECs were exposed to different concentrations (0, 25, 50, and 100 µg/ml) of three sizes of SiNPs for up to 48 h. Cellular viability, autophagy, lactate dehydrogenase (LDH) assay, and mammalian target of rapamycin (mTOR) pathway activation were evaluated. Intracellular distribution of the SiNPs was evaluated with transmission electron microscopy (TEM). TEM revealed that the SiNPs were up-taken by the HCECs inside cytoplasmic vacuoles. No mitochondrial structural damage was observed. Both cellular viability and LDH level remained unchanged with up to 100 µg/mL of SiNP treatment. Autophagy showed a significant dose-dependent activation with 50, 100, and 150 nm SiNPs. However, the mTOR activation remained unchanged. Human corneal tissue culture with 100 µg/ml concentrations of SiNPs for 72 h revealed no significant endothelial toxicity. In vivo corneal safety of the SiNPs (0.05 ml intracameral injection, 200 mg/ml concentration) was also verified in rabbit models. These findings suggested that 50, 100, and 150 nm SiNPs did not induce acute significant cytotoxicity in corneal endothelial cells at concentrations up to 100 µg/mL. However, long-term toxicity of SiNPs remains unknown.


Assuntos
Perda de Células Endoteliais da Córnea/induzido quimicamente , Células Endoteliais/efeitos dos fármacos , Endotélio Corneano/efeitos dos fármacos , Nanopartículas/efeitos adversos , Dióxido de Silício/efeitos adversos , Autofagia/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Endotélio Corneano/ultraestrutura , Humanos , L-Lactato Desidrogenase/metabolismo , Masculino , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Nanopartículas/administração & dosagem , Dióxido de Silício/administração & dosagem , Serina-Treonina Quinases TOR/metabolismo
16.
Sci Rep ; 7(1): 9481, 2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28842713

RESUMO

Biocompatible polymers have been extensively applied to molecular assembly techniques on a micro- and nanoscale to miniaturize functional devices for biomedical uses. However, cytotoxic assessments of developed devices are prone to partially focus on non-specific cells or cells associated with the specific applications. Thereby, since toxicity is dependent on the type of cells and protocols, we do not fully understand the relative toxicities of polymers. Additionally, we need to ensure the blood cell biocompatibility of developed devices prior to that of targeted cells because most of the devices contact the blood before reaching the targeted regions. Motivated by this issue, we focused on screening cytotoxicity of polymers widely used for the layer-by-layer assembly technique using human blood cells. Cytotoxicity at the early stage was investigated on twenty types of polymers (positively charged, negatively charged, or neutral) and ten combination forms via hemolysis, cell viability, and AnnexinV-FITC/PI staining assays. We determined their effects on the cell membrane depending on their surface chemistry by molecular dynamics simulations. Furthermore, the toxicity of LbL-assembled nanofilms was assessed by measuring cell viability. Based on this report, researchers can produce nanofilms that are better suited for drug delivery and biomedical applications by reducing the possible cytotoxicity.


Assuntos
Materiais Biocompatíveis , Células Sanguíneas/metabolismo , Sobrevivência Celular , Polímeros , Materiais Biocompatíveis/efeitos adversos , Materiais Biocompatíveis/química , Fenômenos Químicos , Humanos , Bicamadas Lipídicas , Teste de Materiais , Simulação de Dinâmica Molecular , Nanoestruturas/efeitos adversos , Nanoestruturas/química , Polímeros/efeitos adversos , Polímeros/química , Testes de Toxicidade
17.
Mol Pharm ; 14(10): 3322-3330, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28841795

RESUMO

The ability to control drug loading and release is the most important feature in the development of medical devices. In this research, we prepared a functional nanocoating technology to incorporate a drug-release layer onto a desired substrate. The multilayer films were prepared using chitosan (CHI) and carboxymethyl cellulose (CMC) polysaccharides by the layer-by-layer (LbL) method. By using chemical cross-linking to change the inner structure of the assembled multilayer, we could control the extent of drug loading and release. The cross-linked multilayer film had a porous structure and enhanced water wettability. Interestingly, more of the small-molecule drug was loaded into and released from the non-cross-linked multilayer film, whereas more of the macromolecular drug was loaded into and released from the cross-linked multilayer film. These results indicate that drug loading and release can be easily controlled according to the molecular weight of the desired drug by changing the structure of the film.


Assuntos
Carboximetilcelulose Sódica/química , Quitosana/química , Liberação Controlada de Fármacos , Nanoestruturas/química , Química Farmacêutica , Reagentes de Ligações Cruzadas/química , Porosidade , Propriedades de Superfície , Molhabilidade
18.
Sci Rep ; 7(1): 8238, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28811672

RESUMO

Nonporous silica nanoparticles (SiNPs) have potential as promising carriers for ophthalmic drugs. However, the in vivo safety of ocular topical SiNPs remains unclear. This study investigated the in vivo safety of oral and ocular topical applications of 100 nm-sized SiNPs in Sprague-Dawley rats. The rats were divided into the following four groups: low-dose oral administration (total 100 mg/kg of SiNPs mixed with food for one week), high-dose oral administration (total 1000 mg/kg of SiNPs mixed with food for one week), ocular topical administration (10 mg/ml concentration, one drop, applied to the right eyes four times a day for one month), or a negative control (no SiNP treatment). The rats were observed for 12 weeks to investigate any signs of general or ocular toxicity. During the observation period, no differences were observed in the body weights, food and water intakes, behaviors and abnormal symptoms of the four groups. No animal deaths occurred. After 12 weeks, hematologic, blood biochemical parameters and ophthalmic examinations revealed no abnormal findings in any of the animals. The lack of toxicity of the SiNPs was further verified in autopsy findings of brain, liver, lung, spleen, heart, kidneys, intestine, eyeballs, and ovaries or testes.


Assuntos
Nanopartículas , Dióxido de Silício , Administração Oral , Administração Tópica , Animais , Biomarcadores , Técnicas de Diagnóstico Oftalmológico , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Olho/efeitos dos fármacos , Olho/patologia , Feminino , Imuno-Histoquímica , Masculino , Nanopartículas/administração & dosagem , Nanopartículas/efeitos adversos , Nanopartículas/química , Tamanho do Órgão , Ratos , Dióxido de Silício/química
19.
Invest Ophthalmol Vis Sci ; 58(1): 362-371, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28118663

RESUMO

Purpose: Silica nanoparticles (SiNPs) are promising carriers for ophthalmic drug delivery. In this study, we investigated the effect of various sizes of nonporous SiNPs on cultured human keratocytes. Methods: Three different sizes of SiNPs (50, 100, and 150 nm) were manufactured. Primarily cultured human keratocytes were exposed to different concentrations (0, 25, 50, and 100 µg/mL) of three sizes of SiNPs for up to 72 hours. Intracellular reactive oxygen species (ROS) generation, cellular viability, lactate dehydrogenase (LDH) assay, autophagy, vimentin expression, and mammalian target of rapamycin (mTOR) pathway activation were evaluated. Intracellular distribution of SiNPs was evaluated with transmission electron microscopy. Results: Transmission electron microscopy revealed SiNPs were taken up by keratocytes inside cytoplasmic vacuoles. Neither nuclear entry of SiNPs nor mitochondrial structural damage was observed. Both intracellular ROS generation and LDH level remained unchanged with up to 100 µg/mL SiNP treatment. Cellular viability was not affected by SiNP treatment. Autophagy showed significant dose-dependent activation with 50- and 100-nm SiNPs. However, mTOR activation remained unchanged. Vimentin expression did not show any significant increase with SiNPs. Conclusions: Our findings suggested that 50-, 100-, and 150-nm SiNPs did not induce significant cytotoxicity in cultured human keratocytes at concentrations up to 100 µg/mL.


Assuntos
Ceratócitos da Córnea/efeitos dos fármacos , Nanopartículas , Dióxido de Silício/farmacologia , Animais , Autofagia , Western Blotting , Contagem de Células , Sobrevivência Celular , Células Cultivadas , Ceratócitos da Córnea/metabolismo , Ceratócitos da Córnea/ultraestrutura , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , L-Lactato Desidrogenase/metabolismo , Masculino , Microscopia Eletrônica de Transmissão , Tamanho da Partícula , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
20.
Sci Rep ; 6: 37762, 2016 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-27876873

RESUMO

Ocular drug delivery is an interesting field in current research. Silica nanoparticles (SiNPs) are promising drug carriers for ophthalmic drug delivery. However, little is known about the toxicity of SiNPs on ocular surface cells such as human corneal epithelial cells (HCECs). In this study, we evaluated the cytotoxicity induced by 50, 100 and 150 nm sizes of SiNPs on cultured HCECs for up to 48 hours. SiNPs were up-taken by HCECs inside cytoplasmic vacuoles. Cellular reactive oxygen species generation was mildly elevated, dose dependently, with SiNPs, but no significant decrease of cellular viability was observed up to concentrations of 100 µg/ml for three different sized SiNPs. Western blot assays revealed that both cellular autophagy and mammalian target of rapamycin (mTOR) pathways were activated with the addition of SiNPs. Our findings suggested that 50, 100 and 150 nm sized SiNPs did not induce significant cytotoxicity in cultured HCECs.


Assuntos
Células Epiteliais/citologia , Epitélio Corneano/citologia , Nanopartículas/toxicidade , Dióxido de Silício/toxicidade , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Glutationa/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Espaço Intracelular/metabolismo , L-Lactato Desidrogenase/metabolismo , Nanopartículas/ultraestrutura , Estresse Oxidativo/efeitos dos fármacos , Tamanho da Partícula , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Eletricidade Estática , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA