Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Appl Mater Interfaces ; 16(9): 12042-12051, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38382003

RESUMO

Rapid detection of pathogens and analytes at the point of care offers an opportunity for prompt patient management and public health control. This paper reports an open microfluidic platform coupled with active whispering gallery mode (WGM) microsphere resonators for the rapid detection of influenza viruses. The WGM microsphere resonators, precoated with influenza A polyclonal antibodies, are mechanically trapped in the open micropillar array, where the evaporation-driven flow continuously transports a small volume (∼µL) of sample to the resonators without auxiliaries. Selective chemical modification of the pillar array changes surface wettability and flow pattern, which enhances the detection sensitivity of the WGM resonator-based virus sensor. The optofluidic sensing platform is able to specifically detect influenza A viruses within 15 min using a few microliters of sample and displays a linear response to different virus concentrations.


Assuntos
Técnicas Biossensoriais , Humanos , Microesferas
2.
J Nanobiotechnology ; 21(1): 273, 2023 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-37592297

RESUMO

BACKGROUND: Nanoinjection-the process of intracellular delivery using vertically configured nanostructures-is a physical route that efficiently negotiates the plasma membrane, with minimal perturbation and toxicity to the cells. Nanoinjection, as a physical membrane-disruption-mediated approach, overcomes challenges associated with conventional carrier-mediated approaches such as safety issues (with viral carriers), genotoxicity, limited packaging capacity, low levels of endosomal escape, and poor versatility for cell and cargo types. Yet, despite the implementation of nanoinjection tools and their assisted analogues in diverse cellular manipulations, there are still substantial challenges in harnessing these platforms to gain access into cell interiors with much greater precision without damaging the cell's intricate structure. Here, we propose a non-viral, low-voltage, and reusable electroactive nanoinjection (ENI) platform based on vertically configured conductive nanotubes (NTs) that allows for rapid influx of targeted biomolecular cargos into the intracellular environment, and for successful gene silencing. The localization of electric fields at the tight interface between conductive NTs and the cell membrane drastically lowers the voltage required for cargo delivery into the cells, from kilovolts (for bulk electroporation) to only ≤ 10 V; this enhances the fine control over membrane disruption and mitigates the problem of high cell mortality experienced by conventional electroporation. RESULTS: Through both theoretical simulations and experiments, we demonstrate the capability of the ENI platform to locally perforate GPE-86 mouse fibroblast cells and efficiently inject a diverse range of membrane-impermeable biomolecules with efficacy of 62.5% (antibody), 55.5% (mRNA), and 51.8% (plasmid DNA), with minimal impact on cells' viability post nanoscale-EP (> 90%). We also show gene silencing through the delivery of siRNA that targets TRIOBP, yielding gene knockdown efficiency of 41.3%. CONCLUSIONS: We anticipate that our non-viral and low-voltage ENI platform is set to offer a new safe path to intracellular delivery with broader selection of cargo and cell types, and will open opportunities for advanced ex vivo cell engineering and gene silencing.


Assuntos
Anticorpos , Dano ao DNA , Animais , Camundongos , Membrana Celular , Sobrevivência Celular , Inativação Gênica
3.
Adv Mater ; 35(44): e2304122, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37434421

RESUMO

Chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising cell-based immunotherapy approach for treating blood disorders and cancers, but genetically engineering CAR-T cells is challenging due to primary T cells' sensitivity to conventional gene delivery approaches. The current viral-based method can typically involve significant operating costs and biosafety hurdles, while bulk electroporation (BEP) can lead to poor cell viability and functionality. Here, a non-viral electroactive nanoinjection (ENI) platform is developed to efficiently negotiate the plasma membrane of primary human T cells via vertically configured electroactive nanotubes, enabling efficient delivery (68.7%) and expression (43.3%) of CAR genes in the T cells, with minimal cellular perturbation (>90% cell viability). Compared to conventional BEP, the ENI platform achieves an almost threefold higher CAR transfection efficiency, indicated by the significantly higher reporter GFP expression (43.3% compared to 16.3%). By co-culturing with target lymphoma Raji cells, the ENI-transfected CAR-T cells' ability to effectively suppress lymphoma cell growth (86.9% cytotoxicity) is proved. Taken together, the results demonstrate the platform's remarkable capacity to generate functional and effective anti-lymphoma CAR-T cells. Given the growing potential of cell-based immunotherapies, such a platform holds great promise for ex vivo cell engineering, especially in CAR-T cell therapy.


Assuntos
Linfoma , Receptores de Antígenos de Linfócitos T , Humanos , Linfócitos T , Transfecção , Eletroporação , Linfoma/metabolismo
4.
J Nanobiotechnology ; 20(1): 406, 2022 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-36076230

RESUMO

Nanofabrication technologies have been recently applied to the development of engineered nano-bio interfaces for manipulating complex cellular processes. In particular, vertically configurated nanostructures such as nanoneedles (NNs) have been adopted for a variety of biological applications such as mechanotransduction, biosensing, and intracellular delivery. Despite their success in delivering a diverse range of biomolecules into cells, the mechanisms for NN-mediated cargo transport remain to be elucidated. Recent studies have suggested that cytoskeletal elements are involved in generating a tight and functional cell-NN interface that can influence cargo delivery. In this study, by inhibiting actin dynamics using two drugs-cytochalasin D (Cyto D) and jasplakinolide (Jas), we demonstrate that the actin cytoskeleton plays an important role in mRNA delivery mediated by silicon nanotubes (SiNTs). Specifically, actin inhibition 12 h before SiNT-cellular interfacing (pre-interface treatment) significantly dampens mRNA delivery (with efficiencies dropping to 17.2% for Cyto D and 33.1% for Jas) into mouse fibroblast GPE86 cells, compared to that of untreated controls (86.9%). However, actin inhibition initiated 2 h after the establishment of GPE86 cell-SiNT interface (post-interface treatment), has negligible impact on mRNA transfection, maintaining > 80% efficiency for both Cyto D and Jas treatment groups. The results contribute to understanding potential mechanisms involved in NN-mediated intracellular delivery, providing insights into strategic design of cell-nano interfacing under temporal control for improved effectiveness.


Assuntos
Actinas , Nanotubos , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Citocalasina D/farmacologia , Mecanotransdução Celular , Camundongos , RNA Mensageiro , Silício/química
5.
Anal Chem ; 92(24): 16043-16050, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33269908

RESUMO

Microfluidic flow in lab-on-a-chip devices is typically very sensitive to the variable physical properties of complex samples, e.g., biological fluids. Here, evaporation-driven fluid transport (transpiration) is achieved in a configuration that is insensitive to interfacial tension, salinity, and viscosity over a wide range. Micropillar arrays ("pillar cuvettes") were preloaded by wicking a known volatile fluid (water) and then adding a microliter sample of salt, surfactant, sugar, or saliva solution to the loading zone. As the preloaded fluid evaporates, the sample is reliably drawn from a reservoir through the pillar array at a rate defined by the evaporation of the preloaded fluid (typically nL/s). Including a reagent in the preloaded fluid allows photometric reactions to take place at the boundary between the two fluids. In this configuration, a photometric signal enhancement is observed and chemical analysis is independent of both humidity and temperature. The ability to reliably transport and sense an analyte in microliter volumes without concern over salt, surfactant, viscosity (in part), humidity, and temperature is a remarkable advantage for analytical purposes.

6.
J Am Chem Soc ; 142(37): 15649-15653, 2020 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-32869983

RESUMO

Engineered nano-bio interfaces driven by tunable vertically configured nanostructures have recently emerged as a powerful tool for cellular manipulations and interrogations. Yet the interplay between substrate topography and cellular behavior is highly complex and not fully understood. A new experimental design is proposed that enables generation of ultrathin sections (lamellae) of cell-nanostructure imprints with minimal artifacts. We demonstrate the potential of such lamellae for efficient transmission electron microscopy (TEM) characterization of interfacial interactions between adherent cells and vertically aligned Si nanostructures. This approach will advance understanding of cellular responses to extracellular biophysical and biochemical cues-which is likely to facilitate the design of improved cellular manipulation technologies.

7.
Adv Mater ; 32(24): e2000036, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32378244

RESUMO

Engineered nano-bio cellular interfaces driven by vertical nanostructured materials are set to spur transformative progress in modulating cellular processes and interrogations. In particular, the intracellular delivery-a core concept in fundamental and translational biomedical research-holds great promise for developing novel cell therapies based on gene modification. This study demonstrates the development of a mechanotransfection platform comprising vertically aligned silicon nanotube (VA-SiNT) arrays for ex vivo gene editing. The internal hollow structure of SiNTs allows effective loading of various biomolecule cargoes; and SiNTs mediate delivery of those cargoes into GPE86 mouse embryonic fibroblasts without compromising their viability. Focused ion beam scanning electron microscopy (FIB-SEM) and confocal microscopy results demonstrate localized membrane invaginations and accumulation of caveolin-1 at the cell-NT interface, suggesting the presence of endocytic pits. Small-molecule inhibition of endocytosis suggests that active endocytic process plays a role in the intracellular delivery of cargo from SiNTs. SiNT-mediated siRNA intracellular delivery shows the capacity to reduce expression levels of F-actin binding protein (Triobp) and alter the cellular morphology of GPE86. Finally, the successful delivery of Cas9 ribonucleoprotein (RNP) to specifically target mouse Hprt gene is achieved. This NT-enhanced molecular delivery platform has strong potential to support gene editing technologies.


Assuntos
Edição de Genes/instrumentação , Espaço Intracelular/metabolismo , Nanotecnologia/instrumentação , Nanotubos/química , Silício/química , Animais , Caveolina 1/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Camundongos , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...