Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
J Nanobiotechnology ; 22(1): 131, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38532389

ABSTRACT

Effective intracellular DNA transfection is imperative for cell-based therapy and gene therapy. Conventional gene transfection methods, including biochemical carriers, physical electroporation and microinjection, face challenges such as cell type dependency, low efficiency, safety concerns, and technical complexity. Nanoneedle arrays have emerged as a promising avenue for improving cellular nucleic acid delivery through direct penetration of the cell membrane, bypassing endocytosis and endosome escape processes. Nanostraws (NS), characterized by their hollow tubular structure, offer the advantage of flexible solution delivery compared to solid nanoneedles. However, NS struggle to stably self-penetrate the cell membrane, resulting in limited delivery efficiency. Coupling with extra physiochemical perforation strategies is a viable approach to improve their performance. This study systematically compared the efficiency of NS coupled with polyethylenimine (PEI) chemical modification, mechanical force, photothermal effect, and electric field on cell membrane perforation and DNA transfection. The results indicate that coupling NS with PEI modification, mechanical force, photothermal effects provide limited enhancement effects. In contrast, NS-electric field coupling significantly improves intracellular DNA transfection efficiency. This work demonstrates that NS serve as a versatile platform capable of integrating various physicochemical strategies, while electric field coupling stands out as a form worthy of primary consideration for efficient DNA transfection.


Subject(s)
DNA , Electroporation , Transfection , Cell Membrane , Genetic Therapy , Polyethyleneimine/chemistry
2.
Small Methods ; 8(3): e2300915, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37994267

ABSTRACT

In vitro, drug assessment holds tremendous potential to success in novel drug development and precision medicine. Traditional techniques for drug assessment, however, face remarkable challenges to achieve high speed, as limited by incubation-based drug delivery (>several hours) and cell viability measurements (>1 d), which significantly compromise the efficacy in clinical trials. In this work, a nano-electroporation-DNA tensioner platform is reported that shortens the time of drug delivery to less than 3 s, and that of cellular mechanical force analysis to 30 min. The platform adopts a nanochannel structure to localize a safe electric field for cell perforation, while enhancing delivery speed by 103 times for intracellular delivery, as compared to molecular diffusion in coculture methods. The platform is further equipped with a DNA tensioner to detect cellular mechanical force for quantifying cell viability after drug treatment. Systematic head-to-head comparison, by analyzing FDA (food and drug administration)-approved drugs (paclitaxel, doxorubicin), demonstrated the platform with high speed, efficiency, and safety, showing a simple yet powerful tool for clinical drug screening and development.


Subject(s)
Drug Delivery Systems , Electroporation , United States , Electroporation/methods , Electroporation Therapies , Diffusion , DNA
3.
Adv Sci (Weinh) ; 10(33): e2302622, 2023 11.
Article in English | MEDLINE | ID: mdl-37847907

ABSTRACT

Regenerative medicine in tissue engineering often relies on stem cells and specific growth factors at a supraphysiological dose. These approaches are costly and may cause severe side effects. Herein, therapeutic small extracellular vesicles (t-sEVs) endogenously loaded with a cocktail of human vascular endothelial growth factor A (VEGF-A) and human bone morphogenetic protein 2 (BMP-2) mRNAs within a customized injectable PEGylated poly (glycerol sebacate) acrylate (PEGS-A) hydrogel for bone regeneration in rats with challenging femur critical-size defects are introduced. Abundant t-sEVs are produced by a facile cellular nanoelectroporation system based on a commercially available track-etched membrane (TM-nanoEP) to deliver plasmid DNAs to human adipose-derived mesenchymal stem cells (hAdMSCs). Upregulated microRNAs associated with the therapeutic mRNAs are enriched in t-sEVs for enhanced angiogenic-osteogenic regeneration. Localized and controlled release of t-sEVs within the PEGS-A hydrogel leads to the retention of therapeutics in the defect site for highly efficient bone regeneration with minimal low accumulation in other organs.


Subject(s)
Osteogenesis , Vascular Endothelial Growth Factor A , Rats , Humans , Animals , RNA, Messenger/genetics , Bone Regeneration/genetics , Hydrogels/pharmacology
4.
J Nanobiotechnology ; 21(1): 273, 2023 Aug 17.
Article in English | MEDLINE | ID: mdl-37592297

ABSTRACT

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.


Subject(s)
Antibodies , DNA Damage , Animals , Mice , Cell Membrane , Cell Survival , Gene Silencing
5.
Adv Mater ; 35(44): e2304122, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37434421

ABSTRACT

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.


Subject(s)
Lymphoma , Receptors, Antigen, T-Cell , Humans , T-Lymphocytes , Transfection , Electroporation , Lymphoma/metabolism
6.
ACS Sens ; 7(12): 3671-3681, 2022 12 23.
Article in English | MEDLINE | ID: mdl-36410738

ABSTRACT

Enhancers involved in the upregulation of multiple oncogenes play a fundamental role in tumorigenesis and immortalization. Exploring the activity of enhancers in living cells has emerged as a critical path to a deep understanding of cancer properties, further providing important clues to targeted therapy. However, identifying enhancer activity in living cells is challenging due to the double biological barriers of a cell cytoplasmic membrane and a nuclear membrane, limiting the sensitivity and responsiveness of conventional probing methods. In this work, we developed a nanoelectroporation-probing (NP) platform, which enables intranuclear probe delivery for sensitive interrogation of enhancer activity in living cells. The nanoelectroporation biochip achieved highly focused perforation of the cell cytoplasmic membrane and brought about additional driving force to expedite the delivery of probes into the nucleus. The probes targeting enhancer activity (named "PH probe") are programmed with a cyclic amplification strategy and enable an increase in the fluorescence signals over 100-fold within 1 h. The platform was leveraged to detect the activity of CCAT1 enhancers (CCAT1, colon cancer-associated transcript-1, a long noncoding RNA that functions in tumor invasion and metastasis) in cell samples from clinical lung cancer patients, as well as reveal the heterogeneity of enhancers among different patients. The observations may extend the linkages between enhancers and cancer cells while validating the robustness and reliability of the platform for the assay of enhancer activity. This platform will be a promising toolbox with wide applicable potential for the intranuclear study of living cells.


Subject(s)
Lung Neoplasms , Humans , Reproducibility of Results
7.
Biosensors (Basel) ; 12(7)2022 Jul 13.
Article in English | MEDLINE | ID: mdl-35884325

ABSTRACT

Cell perforation is a critical step for intracellular drug delivery and real-time biosensing of intracellular signals. In recent years, the nanostraws system has been developed to achieve intracellular drug delivery with minimal invasiveness to the cells. Repeated cell perforation via nano-system could allow delivery of multiple drugs into cells for cell editing, but the biosafety is rarely explored. In this work, a nanostraw-mediated nano-electroporation system was developed, which allowed repeated perforation of the same set of cells in a minimally invasive manner, while the biosafety aspect of this system was investigated. Highly controllable fabrication of Al2O3 nanostraw arrays based on a porous polyethylene terephthalate (PET) membrane was integrated with a microfluidic device to construct the nanostraw-electroporation system. The pulse conditions and intervals of nano-electroporation were systematically optimized to achieve efficient cells perforation and maintain the viability of the cells. The cells proliferation, the early apoptosis activities after nanostraw-electroporation and the changes of gene functions and gene pathways of cells after repeated nano-electroporation were comprehensively analyzed. These results revealed that the repeated nanostraw-electroporation did not induce obvious negative effects on the cells. This work demonstrates the feasibility of repeated nano-electroporation on cells and provides a promising strategy for future biomedical applications.


Subject(s)
Nanostructures , Containment of Biohazards , Electroporation/methods , Lab-On-A-Chip Devices , Pharmaceutical Preparations
8.
Biosens Bioelectron ; 210: 114281, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35487136

ABSTRACT

Probing nuclear protein expression while correlating cellular behavior is crucial for deciphering underlying causes of cellular disorders, such as tumor metastasis. Despite efforts to access nuclear proteins by trafficking the double barriers of cell membrane and nuclear membrane, they mostly fall short of the capacity for analyzing various proteins in different cells. Herein, we introduce a Companion-Probe & Race (CPR) platform that enables interrogating nuclear proteins in living cells, while guiding and tracking cellular behaviors (e.g., migration) in real time. The Companion-Probe consists of two polypeptide complexes that were structured with nuclear localization signal (NLS) for entering nucleus, recognition polypeptide for targeting different sites of nuclear proteins, and fragments of green fluorescent protein (GFP) that can recover a whole fluorescent GFP once the two polypeptide complexes combine with a same target protein. The two polypeptide complexes were expressed by two plasmids (named "probe plasmids") that were uniformly and efficiently delivered into cells by nano-electroporation (NEP), a high-performance delivery method for cell focal-poration and accelerated intracellular delivery. To track cell migration, multiple radial microchannels were designed with micro-landmarks on the platform to serve as addressable runways for cells. The proof-of-concept of CPR platform was validated with clinical primary cells that indicated the positive-correlation between nuclear protein murine double minute 2 (MDM2) expression level and cell migration velocity. This platform shows great promises to interrogate nuclear proteins in live cells, and to decode their roles in determining cellular behaviors on a chip.


Subject(s)
Biosensing Techniques , Nuclear Proteins , Animals , Cell Nucleus , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Nuclear Proteins/metabolism
9.
Methods Mol Biol ; 2050: 29-41, 2020.
Article in English | MEDLINE | ID: mdl-31468477

ABSTRACT

Electroporation has been one of the most commonly used physical methods for gene/drug delivery. Compared to other nonviral counterparts, electroporation enables optimization of delivery efficiency by tuning the electric field applied on cells. Commercial electroporation, however, results in stochastic transfection and significant cellular damage mostly due to its "bulk" environment. In this chapter, we introduce nanoelectroporation (NEP) which has demonstrated living cell transfection in a highly controllable manner. In NEP, the electric field can be precisely focused on a single cell positioned on nanochannels. Safe single-cell electroporation as well as "electrophoretic" molecular delivery can be achieved on the same device. This system achieves significantly higher transfection efficiency and cellular viability than commercial systems. This device is unique in that it can efficiently deliver genetic molecules (e.g., DNAs, RNAs) that exceed 10 kbp in size. The NEP device based on a 3D nanochannel array prototype was fabricated using cleanroom techniques. For achieving precise cell to nanochannel pairing, three on-chip high-throughput manipulation technologies were developed, that is, magnetic tweezers (MT), dielectrophoresis (DEP), and thin-film microfluidics.


Subject(s)
Electroporation/instrumentation , Microfluidic Analytical Techniques/instrumentation , Animals , Cell Line , Cell Survival , Equipment Design , High-Throughput Screening Assays/instrumentation , Humans , Lab-On-A-Chip Devices , Rats
10.
Methods Mol Biol ; 2050: 69-77, 2020.
Article in English | MEDLINE | ID: mdl-31468480

ABSTRACT

Delivery of macromolecular nucleotides into the living cells holds a great promise for the development of new therapeutics. However, its abilities for adoptive immunotherapy, cell reprogramming, and primary cell transfection have been long-term hindered by the lack of a system that can locally deliver engineered therapeutic nucleotides (e.g., plasmids, siRNAs, miRNAs) without causing any side effects. In this chapter, the performance of a novel 3D nanoelectroporation system (3D NEP) is highlighted in three scenarios-adoptive immunotherapy, cell reprogramming, and adult mouse primary cardiomyocyte transfection. Detailed protocols were given to introduce the 3D NEP system assembly, as well as their applications in (1) natural killer (NK) cells transfection by delivery of chimeric antigen receptor (CAR) plasmids; (2) mouse embryonic fibroblasts transfection with OSKM factors; and (3) miR-29b molecular beacon (BMs) delivery into primary cardiomyocytes for interrogating the side effect of miR-29b-assisted treatment.


Subject(s)
Electroporation/instrumentation , Fibroblasts/cytology , Killer Cells, Natural/cytology , Myocytes, Cardiac/cytology , Nucleotides/administration & dosage , Animals , Cells, Cultured , Cellular Reprogramming Techniques/instrumentation , Cellular Reprogramming Techniques/methods , Fibroblasts/chemistry , Humans , Immunotherapy, Adoptive/instrumentation , Immunotherapy, Adoptive/methods , Killer Cells, Natural/chemistry , Mice , Myocytes, Cardiac/chemistry , Nanotechnology , Transfection/instrumentation , Transfection/methods
11.
ACS Appl Mater Interfaces ; 11(47): 43936-43948, 2019 Nov 27.
Article in English | MEDLINE | ID: mdl-31696695

ABSTRACT

One-dimensional nanoneedle-like arrays have emerged as an attractive tool for penetrating the cell membrane to achieve intracellular applications including drug delivery, electrical recording, and biochemical detection. Hollow nanoneedles, also called nanostraws (NSs), combined with nanoelectroporation have been demonstrated as a powerful platform for intracellular drug delivery and extraction of intracellular contents. However, the fabrication technique of nanostraws still requires complicated and expensive atomic layer deposition and etching processes and fails to produce conductive nanostraws. Herein, we developed a commonly accessible and versatile electrodeposition approach to controllably fabricate conductive nanostraw arrays based on various types of metal or conductive polymer materials. Representatively, Pt nanostraws (Pt NSs) with 400 nm diameter were further integrated with a low-voltage nanoelectroporation system to achieve cell detection, intracellular drug delivery, and sensing of intracellular enzymes. Both theoretical simulations and experimental results revealed that the conductive nanostraws in direct contact with cells could induce high-efficiency cell electroporation at relatively low voltage (∼5 V). Efficient delivery of reagents into live cells with spatial control and repeated extraction of intracellular enzymes (e.g., caspase-3) for temporal monitoring from the same set of cells were demonstrated. This work not only pioneers a new avenue for universal production of conductive nanostraws on a large scale but also presents great potential for developing nanodevices to achieve a variety of biomedical applications including cell re-engineering, cell-based therapy, and signaling pathway monitoring.


Subject(s)
Drug Delivery Systems/instrumentation , Electroplating/methods , Equipment Design , Nanostructures/chemistry , Biosensing Techniques/instrumentation , Cell Survival , Cells/chemistry , Cells/enzymology , Electric Conductivity , Enzymes/analysis , HeLa Cells , Humans , Nanotechnology , Platinum/chemistry
12.
Nano Lett ; 19(10): 7201-7209, 2019 10 09.
Article in English | MEDLINE | ID: mdl-31557044

ABSTRACT

Downstream analysis of circulating tumor cells (CTCs) has provided new insights into cancer research. In particular, the detection of CTCs, followed by the regulation and monitoring of their intracellular activities, can provide valuable information for comprehensively understanding cancer pathogenesis and progression. However, current CTC detection techniques are rarely capable of in situ regulation and monitoring of the intracellular microenvironments of cancer cells over time. Here, we developed a multifunctional branched nanostraw (BNS)-electroporation platform that could effectively capture CTCs and allow for downstream regulation and monitoring of their intracellular activities in a real-time and in situ manner. The BNSs possessed numerous nanobranches on the outer sidewall of hollow nanotubes, which could be conjugated with specific antibodies to facilitate the effective capture of CTCs. Nanoelectroporation could be applied through the BNSs to nondestructively porate the membranes of the captured cells at a low voltage, allowing the delivery of exogenous biomolecules into the cytosol and the extraction of cytosolic contents through the BNSs without affecting cell viability. The efficient delivery of biomolecules (e.g., small molecule dyes and DNA plasmids) into cancer cells with spatial and temporal control and, conversely, the repeated extraction of intracellular enzymes (e.g., caspase-3) for real-time monitoring were both demonstrated. This technology can provide new opportunities for the comprehensive understanding of cancer cell functions that will facilitate cancer diagnosis and treatment.


Subject(s)
Electroporation/instrumentation , Microfluidic Analytical Techniques/instrumentation , Nanostructures , Neoplastic Cells, Circulating/pathology , Cell Line, Tumor , Epithelial Cell Adhesion Molecule/analysis , Equipment Design , Humans , Nanostructures/chemistry , Nanotechnology/instrumentation , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism
13.
Adv Ther (Weinh) ; 2(12)2019 Dec.
Article in English | MEDLINE | ID: mdl-37448511

ABSTRACT

Transfection is a critical step for gene editing and cell-based therapies. Nanoscale technologies have shown great promise to provide higher transfection efficiency and lower cell perturbation than conventional viral, biochemical and electroporation techniques due to their small size and localized effect. Although this has significant implications for using cells post-transfection, it has not been thoroughly studied. Here, we developed the nano-electro-injection (NEI) platform which makes use of localized electric fields to transiently open pores on cell membrane followed by electrophoretic delivery of DNA into cells. NEI provided two-folds higher net transfection efficiency than biochemicals and electroporation in Jurkat cells. Analysis of cell doubling time, intracellular calcium levels and mRNA expression changes after these gene delivery methods revealed that viruses and electroporation adversely affected cell behavior. Cell doubling times increased by more than 40% using virus and electroporation methods indicative of higher levels of cell stress, unlike NEI which only minimally affected cell division. Finally, electroporation, but not NEI, greatly altered the expression of immune-associated genes related to immune cell activation and trafficking. These results highlight that nanoscale delivery tools can have significant advantages from a cell health perspective for cell-based research and therapeutic applications.

14.
Small ; 12(43): 5971-5980, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27648733

ABSTRACT

While electroporation has been widely used as a physical method for gene transfection in vitro and in vivo, its application in gene therapy of cardiovascular cells remains challenging. Due to the high concentration of ion-transport proteins in the sarcolemma, conventional electroporation of primary cardiomyocytes tends to cause ion-channel activation and abnormal ion flux, resulting in low transfection efficiency and high mortality. In this work, a high-throughput nanoelectroporation technique based on a nanochannel array platform is reported, which enables massively parallel delivery of genetic cargo (microRNA, plasmids) into mouse primary cardiomyocytes in a controllable, highly efficient, and benign manner. A simple "dipping-trap" approach was implemented to precisely position a large number of cells on the nanoelectroporation platform. With dosage control, our device precisely titrates the level of miR-29, a potential therapeutic agent for cardiac fibrosis, and determines the minimum concentration of miR-29 causing side effects in mouse primary cardiomyocytes. Moreover, the dose-dependent effect of miR-29 on mitochondrial potential and homeostasis is monitored. Altogether, our nanochannel array platform provides efficient trapping and transfection of primary mouse cardiomyocyte, which can improve the quality control for future microRNA therapy in heart diseases.


Subject(s)
Myocytes, Cardiac/metabolism , Nanoparticles/chemistry , Nanotechnology/methods , Transfection/methods , Animals , Cells, Cultured , Computer Simulation , Electroporation , Mice , MicroRNAs/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL