Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cell Mol Life Sci ; 79(1): 23, 2022 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-34984553

RESUMO

Vapor nanobubble (VNB) photoporation is a physical method for intracellular delivery that has gained significant interest in the past decade. It has successfully been used to introduce molecular cargo of diverse nature into different cell types with high throughput and minimal cytotoxicity. For translational purposes, it is important to understand whether and how photoporation affects cell homeostasis. To obtain a comprehensive view on the transcriptional rewiring that takes place after VNB photoporation, we performed a longitudinal shotgun RNA-sequencing experiment. Six hours after photoporation, we found a marked upregulation of LMNA transcripts as well as their protein products, the A-type lamins. At the same time point, we observed a significant increase in several heterochromatin marks, suggesting a global stiffening of the nucleus. These molecular features vanished 24 h after photoporation. Since VNB-induced chromatin condensation was prolonged in LMNA knockout cells, A-type lamins may be required for restoring the nucleus to its original state. Selective depletion of A-type lamins reduced cell viability after VNB photoporation, while pharmacological stimulation of LMNA transcription increased the percentage of successfully transfected cells that survived after photoporation. Therefore, our results suggest that cells respond to VNB photoporation by temporary upregulation of A-type lamins to facilitate their recovery.


Assuntos
Permeabilidade da Membrana Celular , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Lamina Tipo A/metabolismo , Nanopartículas/química , Perfilação da Expressão Gênica , Células HeLa , Humanos , Luz , Microtúbulos/metabolismo , Polimerização , Biossíntese de Proteínas , Temperatura , Transcrição Gênica , Transcriptoma/genética , Regulação para Cima/genética , Volatilização
2.
ACS Nano ; 17(8): 7180-7193, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37058432

RESUMO

Targeted drug delivery depends on the ability of nanocarriers to reach the target site, which requires the penetration of different biological barriers. Penetration is usually low and slow because of passive diffusion and steric hindrance. Nanomotors (NMs) have been suggested as the next generation of nanocarriers in drug delivery due to their autonomous motion and associated mixing hydrodynamics, especially when acting collectively as a swarm. Here, we explore the concept of enzyme-powered NMs designed as such that they can exert disruptive mechanical forces upon laser irradiation. The urease-powered motion and swarm behavior improve translational movement compared to passive diffusion of state-of-the-art nanocarriers, while optically triggered vapor nanobubbles can destroy biological barriers and reduce steric hindrance. We show that these motors, named Swarm 1, collectively displace through a microchannel blocked with type 1 collagen protein fibers (barrier model), accumulate onto the fibers, and disrupt them completely upon laser irradiation. We evaluate the disruption of the microenvironment induced by these NMs (Swarm 1) by quantifying the efficiency by which a second type of fluorescent NMs (Swarm 2) can move through the cleared microchannel and be taken up by HeLa cells at the other side of the channel. Experiments showed that the delivery efficiency of Swarm 2 NMs in a clean path was increased 12-fold in the presence of urea as fuel compared to when no fuel was added. When the path was blocked with the collagen fibers, delivery efficiency dropped considerably and only depicted a 10-fold enhancement after pretreatment of the collagen-filled channel with Swarm 1 NMs and laser irradiation. The synergistic effect of active motion (chemically propelled) and mechanical disruption (light-triggered nanobubbles) of a biological barrier represents a clear advantage for the improvement of therapies which currently fail due to inadequate passage of drug delivery carriers through biological barriers.


Assuntos
Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Humanos , Células HeLa
3.
Pharmaceutics ; 14(8)2022 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-36015342

RESUMO

The inner limiting membrane (ILM) represents a major bottleneck hampering efficient drug delivery to the retina after intravitreal injection. To overcome this barrier, we intend to perforate the ILM by use of a light-based approach which relies on the creation of vapor nanobubbles (VNBs) when irradiating photosensitizers with high intensity laser pulses. Upon collapse of these VNBs, mechanical effects can disrupt biological structures. As a photosensitizer, we explore indocyanine green (ICG) loaded nanoparticles (NPs) specifically designed for our application. In light of this, ICG liposomes and PLGA ICG NPs were characterized in terms of physicochemical properties, ICG incorporation and VNB formation. ICG liposomes were found to encapsulate significantly higher amounts of ICG compared to PLGA ICG NPs which is reflected in their VNB creating capacity. Since only ICG liposomes were able to induce VNB generation, this class of NPs was further investigated on retinal explants. Here, application of ICG liposomes followed by laser treatment resulted in subtle disruption effects at the ILM where zones of fully ablated ILM were alternated by intact regions. As the interaction between the ICG liposomes and ILM might be insufficient, active targeting strategies or other NP designs might improve the concept to a further extent.

4.
Nanomicro Lett ; 12(1): 185, 2020 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-34138203

RESUMO

Efficient and safe cell engineering by transfection of nucleic acids remains one of the long-standing hurdles for fundamental biomedical research and many new therapeutic applications, such as CAR T cell-based therapies. mRNA has recently gained increasing attention as a more safe and versatile alternative tool over viral- or DNA transposon-based approaches for the generation of adoptive T cells. However, limitations associated with existing nonviral mRNA delivery approaches hamper progress on genetic engineering of these hard-to-transfect immune cells. In this study, we demonstrate that gold nanoparticle-mediated vapor nanobubble (VNB) photoporation is a promising upcoming physical transfection method capable of delivering mRNA in both adherent and suspension cells. Initial transfection experiments on HeLa cells showed the importance of transfection buffer and cargo concentration, while the technology was furthermore shown to be effective for mRNA delivery in Jurkat T cells with transfection efficiencies up to 45%. Importantly, compared to electroporation, which is the reference technology for nonviral transfection of T cells, a fivefold increase in the number of transfected viable Jurkat T cells was observed. Altogether, our results point toward the use of VNB photoporation as a more gentle and efficient technology for intracellular mRNA delivery in adherent and suspension cells, with promising potential for the future engineering of cells in therapeutic and fundamental research applications.

5.
Int J Antimicrob Agents ; 56(6): 106194, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33039591

RESUMO

INTRODUCTION: Actively dispersed Pseudomonas aeruginosa biofilm cells differ from planktonic cells, as they have a lower intracellular cyclic di-guanosine monophosphate (c-di-GMP) concentration and show increased virulence. In addition, the nature of the dispersion trigger has been shown to influence the antibiotic susceptibility of dispersed cells. However, properties of passively-dispersed cells, in which the dispersion trigger directly releases cells from the biofilm, have not been described. The present study determined c-di-GMP concentration, virulence in Galleria mellonella and antibiotic susceptibility of P. aeruginosa cells dispersed from biofilm using various triggers. MATERIALS AND METHODS: P. aeruginosa biofilms grown in flow-cells were dispersed actively [exposure to the nitric oxide (NO)-donor sodium nitroprusside (SNP) or to glutamate] or passively [by stopping and restarting the flow or exposure to laser-induced vapor nanobubbles (VNB)], and properties of these dispersed cells were compared to those of spontaneously-dispersed cells. RESULTS: The passively dispersed P. aeruginosa biofilm cells had significantly lower intracellular c-di-GMP levels than actively-dispersed cells. However, this did not result in differences in virulence in Galleria mellonella, nor in tobramycin and ciprofloxacin susceptibility. Passively-dispersed cells were more susceptible to colistin than actively- and spontaneously-dispersed cells. In cells dispersed by interrupting the flow, increased susceptibility to colistin was immediate, whereas this was delayed for VNB-dispersed cells. CONCLUSION: Passively-dispersed P. aeruginosa biofilm cells have a decreased intracellular c-di-GMP concentration and an increased colistin susceptibility compared to actively-dispersed cells. No differences in virulence or susceptibility to tobramycin or colistin were observed.


Assuntos
Antibacterianos/farmacologia , Colistina/farmacologia , GMP Cíclico/metabolismo , Farmacorresistência Bacteriana/fisiologia , Pseudomonas aeruginosa/efeitos dos fármacos , Tobramicina/farmacologia , Animais , Carga Bacteriana , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Humanos , Mariposas/microbiologia , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade
6.
J Control Release ; 319: 262-275, 2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-31904400

RESUMO

Strategies for controlled delivery of therapeutic siRNA into living cells are in high demand as endosomal escape remains the most prominent bottleneck at the intracellular level. Photothermal properties of gold nanoparticles (AuNP) can be used to overcome the endosomal membrane barrier upon laser irradiation by two mechanisms: endosomal rupture by mechanical energy from water vapor nanobubbles (VNBs), or permeabilization of the endosomal membrane by heat diffusion. Here we evaluated how both mechanisms influence cargo release, transfection efficiency, acute cytotoxicity and cell homeostasis. Using a siRNA/AuNP drug delivery system we found that the in vitro release of siRNA from the AuNP carrier occurs equally efficiently by VNB formation or heat generation. Heat-mediated endosomal escape happened more efficiently in cells that had more particles per endosome, resulting in variable siRNA-induced downregulation (20-50%). VNB-mediated endosomal escape did not dependent on the number of AuNP per endosome, yielding high downregulations (50-60%) independent of the cell type. Effects on cell homeostasis by whole transcriptome analysis, showed a quick recover after 24 h or 48 h for either of both photothermal mechanisms. We conclude that VNBs are more consistent to induce efficient endosomal escape and gene silencing independent of the cell type without long lasting effects on cell homeostasis.


Assuntos
Ouro , Nanopartículas Metálicas , Endossomos , Homeostase , RNA Interferente Pequeno
7.
Biofilm ; 1: 100004, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33447791

RESUMO

Being responsible for delayed wound healing, the presence of biofilms in infected wounds leads to chronic, and difficult to treat infections. One of the reasons why antimicrobial treatment often fails to cure biofilm infections is the reduced penetration rate of antibiotics through dense biofilms. Strategies that have the ability to somehow interfere with the integrity of biofilms and allowing a better penetration of drugs are highly sought after. A promising new approach is the use of laser-induced vapor nanobubbles (VNB), of which it was recently demonstrated that it can substantially enhance the penetration of antibiotics into biofilms, resulting in a marked improvement of the killing efficiency. In this study, we examined if treatment of biofilms with laser-induced vapor nanobubbles (VNB) can enhance the potency of antimicrobials which are commonly used to treat wound infections, including povidone-iodine, chlorhexidine, benzalkonium chloride, cetrimonium bromide and mupirocin. Our investigations were performed on Pseudomonas aeruginosa and Staphylococcus aureus biofilms, which are often implicated in chronic wound infections. Pre-treatment of biofilms with laser-induced VNB did enhance the killing efficiency of those antimicrobials which experience a diffusion barrier in the biofilms, while this was not the case for those compounds for which there is no diffusion barrier. The magnitude of the enhanced potency was in most cases similar to the enhancement that was obtained when the biofilms were completely disrupted by vortexing and sonication. These results show that laser-induced VNB are indeed a very efficient way to enhance drug penetration deep into biofilms, and pave the way towards clinical translation of this novel approach for treatment of wound infections.

8.
Pharmaceutics ; 11(5)2019 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-31052369

RESUMO

Impaired penetration of antibiotics through bacterial biofilms is one of the reasons for failure of antimicrobial therapy. Hindered drug diffusion is caused on the one hand by interactions with the sticky biofilm matrix and on the other hand by the fact that bacterial cells are organized in densely packed clusters of cells. Binding interactions with the biofilm matrix can be avoided by encapsulating the antibiotics into nanocarriers, while interfering with the integrity of the dense cell clusters can enhance drug transport deep into the biofilm. Vapor nanobubbles (VNB), generated from laser irradiated nanoparticles, are a recently reported effective way to loosen up the biofilm structure in order to enhance drug transport and efficacy. In the present study, we explored if the disruptive force of VNB can be used simultaneously to interfere with the biofilm structure and trigger antibiotic release from light-responsive nanocarriers. The antibiotic tobramycin was incorporated in two types of light-responsive nanocarriers-liposomes functionalized with gold nanoparticles (Lip-AuNP) and graphene quantum dots (GQD)-and their efficacy was evaluated on Pseudomonas aeruginosa biofilms. Even though the anti-biofilm efficacy of tobramycin was improved by liposomal encapsulation, electrostatic functionalization with 70 nm AuNP unfortunately resulted in premature leakage of tobramycin in a matter of hours. Laser-irradiation consequently did not further improve P. aeruginosa biofilm eradication. Adsorption of tobramycin to GQD, on the other hand, did result in a stable formulation with high encapsulation efficiency, without burst release of tobramycin from the nanocarriers. However, even though laser-induced VNB formation from GQD resulted in biofilm disruption, an enhanced anti-biofilm effect was not achieved due to tobramycin not being efficiently released from GQD. Even though this study was unsuccessful in designing suitable nanocarriers for simultaneous biofilm disruption and light-triggered release of tobramycin, it provides insights into the difficulties and challenges that need to be considered for future developments in this regard.

9.
ACS Nano ; 12(8): 7791-7802, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30001106

RESUMO

The nuclear envelope (NE) has long been considered to dismantle only during mitosis. However, recent observations in cancer cells and laminopathy patient cells have revealed that the NE can also transiently rupture during interphase, thereby perturbing cellular homeostasis. Although NE ruptures are promoted by mechanical force and the loss of lamins, their stochastic nature and variable frequency preclude the study of their direct downstream consequences. We have developed a method based on vapor nanobubble-mediated photoporation that allows for deliberately inducing NE ruptures in a spatiotemporally controlled manner. Our method relies on wide-field laser illumination of perinuclear gold nanoparticles, resulting in the formation of short-lived vapor nanobubbles that inflict minute mechanical damage to the NE, thus creating small pores. We demonstrate that perinuclear localization of gold nanoparticles can be achieved after endocytic uptake or electroporation-facilitated delivery and that both strategies result in NE rupture upon laser irradiation. Furthermore, we prove that photoporation-induced nuclear ruptures are transient and recapitulate hallmarks of spontaneous NE ruptures that occur in A-type lamin-depleted cells. Finally, we show that the same approach can be used to promote influx of macromolecules that are too large to passively migrate through the NE. Thus, by providing unprecedented control over nuclear compartmentalization, nuclear photoporation offers a powerful tool for both fundamental cell biology research and drug delivery applications.


Assuntos
Núcleo Celular/metabolismo , Nanopartículas/metabolismo , Membrana Nuclear/metabolismo , Núcleo Celular/química , Sobrevivência Celular , Células HeLa , Humanos , Nanopartículas/química , Membrana Nuclear/química , Imagem Óptica , Processos Fotoquímicos , Células Tumorais Cultivadas , Volatilização
10.
Methods Mol Biol ; 1530: 165-192, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28150203

RESUMO

Laser pulse-induced vapor nanobubbles are nonstationary nanoevents that offer a broad range of applications, especially in the biomedical field. Plasmonic (usually gold) nanoparticles have the highest energy efficacy of the generation of vapor nanobubbles and such nanobubbles were historically named as plasmonic nanobubbles. Below we review methods (protocols) for generating and detecting plasmonic nanobubbles in liquids. The biomedical applications of plasmonic nanobubbles include in vivo and in vitro detection and imaging, gene transfer, micro-surgery, drug delivery, and other diagnostic, therapeutic, and theranostic applications.


Assuntos
Ouro , Terapia a Laser , Nanopartículas Metálicas , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Receptores ErbB/metabolismo , Ouro/química , Humanos , Nanopartículas Metálicas/química , Neoplasias/metabolismo
11.
J Control Release ; 267: 154-162, 2017 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-28778478

RESUMO

The success of cancer immunotherapy through the adoptive transfer of cytotoxic T lymphocytes (CTLs) is highly dependent on the potency of the elicited anti-tumor responses generated by the transferred cells, which can be hindered by a variety of upregulated immunosuppressive pathways. Downregulation of these pathways in the T cells via RNA interference (RNAi) could significantly boost their capacity to infiltrate tumors, proliferate, persist, and eradicate tumor cells, thus leading to a durable anti-tumor response. Unfortunately, it is well known that primary T cells are hard-to-transfect and conventional non-viral transfection agents are generally ineffective. Viral transduction and electroporation are more efficient but their use is restricted by high cost, safety issues, and cytotoxicity. Photoporation has recently gained interest as a more gentle alternative physical approach to deliver membrane-impermeable macromolecules into cells. By attaching gold nanoparticles (AuNPs) to the cell surface followed by pulsed laser illumination, transient membrane pores can be generated that allow the diffusion of macromolecules directly into the cell cytosol. Here, we evaluated this technique for the non-toxic and effective delivery of small interfering RNA (siRNA) and subsequent silencing of target genes in activated CTLs. We compared photoporation with nucleofection, the current standard physical technique for T cell transfection, and demonstrated a significantly reduced cytotoxicity and higher average dose per cell for the photoporation technique.


Assuntos
Ouro/administração & dosagem , Antígenos Comuns de Leucócito/genética , Nanopartículas Metálicas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Linfócitos T Citotóxicos/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Eletroporação/métodos , Feminino , Lasers , Camundongos Endogâmicos C57BL , Interferência de RNA , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA