Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Small ; 16(38): e2002529, 2020 09.
Article in English | MEDLINE | ID: mdl-32776465

ABSTRACT

Elevated temperatures might have promoted the nucleation, growth, and replication of protocells on the early Earth. Recent reports have shown evidence that moderately high temperatures not only permit protocell assembly at the origin of life, but can have actively supported it. Here, the fast nucleation and growth of vesicular compartments from autonomously formed lipid networks on solid surfaces, induced by a moderate increase in temperature, are shown. Branches of the networks, initially consisting of self-assembled interconnected nanotubes, rapidly swell into microcompartments which can spontaneously encapsulate RNA fragments. The increase in temperature further causes fusion of adjacent network-connected compartments, resulting in the redistribution of the RNA. The experimental observations and the mathematical model indicate that the presence of nanotubular interconnections between protocells facilitates the fusion process.


Subject(s)
Artificial Cells , Nanotubes , Cell Membrane , RNA
2.
Langmuir ; 35(11): 3999-4010, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30789270

ABSTRACT

Poly(ethylene glycol) (PEG) polymers and PEG-conjugated lipids are widely used in bioengineering and drug transport applications. A PEG layer in a drug carrier increases hydrophilic repulsion, inhibits membrane fusion and serum opsonin interactions, and prolongs the storage and circulation time. It can also change the carrier shape and have an influence on many properties related to the content release of the carrier. In this paper, we focus on the physicochemical effects of PEGylation in the lipid bilayer. We introduce laurdanC as a fluorophore for shape recognition and phase transition detection. Together with laurdanC, cryogenic transmission electron microscopy, differential scanning calorimetry, molecular dynamics simulations, and small-angle X-ray scattering/wide-angle X-ray scattering, we acquire information of the particle/bilayer morphology and phase behavior in systems containing 1,2-dipalmitoyl- sn-glycero-3-phosphocholine:1,2-distearoyl- sn-glycero-3-phosphoethanolamine-PEG(2000) with different fractions. We find that PEGylation leads to two important and potentially usable features of the system. (1) Spherical vesicles present a window of elevated chain-melting temperatures and (2) lipid packing shape-controlled liposome-to-bicelle transition. The first finding is significant for targets requiring multiple release sequences and the second enables tuning the release by composition and the PEG polymer length. Besides drug delivery systems, the findings can be used in other smart soft materials with trigger-polymers as well.

3.
J Control Release ; 244(Pt B): 157-166, 2016 12 28.
Article in English | MEDLINE | ID: mdl-27565215

ABSTRACT

Ocular drug delivery, especially to the retina and choroid, is a major challenge in drug development. Liposome technology may be useful in ophthalmology in enabling new routes of delivery, prolongation of drug action and intracellular drug delivery, but drug release from the liposomes should be controlled. For that purpose, light activation may be an approach to release drug at specified time and site in the eye. Technical advances have been made in the field of light activated drug release, particularly indocyanine green loaded liposomes are a promising approach with safe materials and effective light triggered release of small and large molecules. This review discusses the liposomal drug delivery with light activated systems in the context of ophthalmic drug delivery challenges.


Subject(s)
Administration, Ophthalmic , Drug Delivery Systems , Light , Liposomes/radiation effects , Animals , Eye/metabolism , Humans
4.
Mol Pharm ; 13(6): 2095-107, 2016 06 06.
Article in English | MEDLINE | ID: mdl-27097108

ABSTRACT

Light-triggered drug delivery systems enable site-specific and time-controlled drug release. In previous work, we have achieved this with liposomes containing gold nanoparticles in the aqueous core. Gold nanoparticles absorb near-infrared light and release the energy as heat that increases the permeability of the liposomal bilayer, thus releasing the contents of the liposome. In this work, we replaced the gold nanoparticles with the clinically approved imaging agent indocyanine green (ICG). The ICG liposomes were stable at storage conditions (4-22 °C) and at body temperature, and fast near-infrared (IR) light-triggered drug release was achieved with optimized phospholipid composition and a 1:50 ICG-to-lipid molar ratio. Encapsulated small molecular calcein and FITC-dextran (up to 20 kDa) were completely released from the liposomes after light exposure for 15 s. Location of ICG in the PEG layer of the liposomes was simulated with molecular dynamics. ICG has important benefits as a light-triggering agent in liposomes: fast content release, improved stability, improved possibility of liposomal size control, regulatory approval to use in humans, and the possibility of imaging the in vivo location of the liposomes based on the fluorescence of ICG. Near-infrared light used as a triggering mechanism has good tissue penetration and safety. Thus, ICG liposomes are an attractive option for light-controlled and efficient delivery of small and large drug molecules.


Subject(s)
Drug Liberation/drug effects , Indocyanine Green/chemistry , Liposomes/chemistry , Drug Delivery Systems/methods , Fluorescence , Gold/administration & dosage , Humans , Infrared Rays , Metal Nanoparticles/chemistry , Polyethylene Glycols/chemistry
5.
Langmuir ; 32(18): 4554-63, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27089512

ABSTRACT

In light-activated liposomal drug delivery systems (DDSs), the light sensitivity can be obtained by a photothermal agent that converts light energy into heat. Excess heat increases the drug permeability of the lipid bilayer, and drug is released as a result. In this work, two near-IR responsive photothermal agents in a model drug delivery system are studied: either gold nanorods (GNRs) encapsulated inside the liposomes or indocyanine green (ICG) embedded into the lipid bilayer. The liposome system is exposed to light, and the heating effect is studied with fluorescent thermometers: laurdan and CdSe quantum dots (QDs). Both photothermal agents are shown to convert light into heat in an extent to cause a phase transition in the surrounding lipid bilayer. This phase transition is also proven with laurdan generalized polarization (GP). In addition to the heating results, we show that the model drug (calcein) is released from the liposomal cavity with both photothermal agents when the light power is sufficient to cause a phase transition in the lipid bilayer.


Subject(s)
Drug Liberation , Gold/chemistry , Indocyanine Green/chemistry , Light , Lipid Bilayers/chemistry , Nanotubes/chemistry , Phase Transition , Temperature , Capsules , Liposomes , Safety
6.
J Control Release ; 203: 85-98, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25701610

ABSTRACT

Externally triggered drug release at defined targets allows site- and time-controlled drug treatment regimens. We have developed liposomal drug carriers with encapsulated gold nanoparticles for triggered drug release. Light energy is converted to heat in the gold nanoparticles and released to the lipid bilayers. Localized temperature increase renders liposomal bilayers to be leaky and triggers drug release. The aim of this study was to develop a drug releasing system capable of releasing its cargo to cell cytosol upon triggering with visible and near infrared light signals. The liposomes were formulated using either heat-sensitive or heat- and pH-sensitive lipid compositions with star or rod shaped gold nanoparticles. Encapsulated fluorescent probe, calcein, was released from the liposomes after exposure to the light. In addition, the pH-sensitive formulations showed a faster drug release in acidic conditions than in neutral conditions. The liposomes were internalized into human retinal pigment epithelial cells (ARPE-19) and human umbilical vein endothelial cells (HUVECs) and did not show any cellular toxicity. The light induced cytosolic delivery of calcein from the gold nanoparticle containing liposomes was shown, whereas no cytosolic release was seen without light induction or without gold nanoparticles in the liposomes. The light activated liposome formulations showed a controlled content release to the cellular cytosol at a specific location and time. Triggering with visual and near infrared light allows good tissue penetration and safety, and the pH-sensitive liposomes may enable selective drug release in the intracellular acidic compartments (endosomes, lysosomes). Thus, light activated liposomes with gold nanoparticles are an attractive option for time- and site-specific drug delivery into the target cells.


Subject(s)
Delayed-Action Preparations/chemistry , Fluoresceins/administration & dosage , Fluorescent Dyes/administration & dosage , Gold/chemistry , Liposomes/chemistry , Metal Nanoparticles/chemistry , Cell Line , Delayed-Action Preparations/metabolism , Drug Liberation , Gold/metabolism , Hot Temperature , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen-Ion Concentration , Light , Liposomes/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...