Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Lab Chip ; 24(5): 1088-1120, 2024 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-38174732

RESUMO

Chimeric antigen receptor (CAR)-T cell therapies have revolutionized cancer treatment, particularly in hematological malignancies. However, their application to solid tumors is limited, and they face challenges in safety, scalability, and cost. To enhance current CAR-T cell therapies, the integration of microfluidic technologies, harnessing their inherent advantages, such as reduced sample consumption, simplicity in operation, cost-effectiveness, automation, and high scalability, has emerged as a powerful solution. This review provides a comprehensive overview of the step-by-step manufacturing process of CAR-T cells, identifies existing difficulties at each production stage, and discusses the successful implementation of microfluidics and related technologies in addressing these challenges. Furthermore, this review investigates the potential of microfluidics-based methodologies in advancing cell-based therapy across various applications, including solid tumors, next-generation CAR constructs, T-cell receptors, and the development of allogeneic "off-the-shelf" CAR products.


Assuntos
Microfluídica , Neoplasias , Humanos , Linfócitos T , Receptores de Antígenos de Linfócitos T , Imunoterapia/métodos , Imunoterapia Adotiva/métodos , Neoplasias/patologia
2.
Nano Lett ; 23(16): 7341-7349, 2023 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-37506062

RESUMO

Effective tumor regression has been observed with chimeric antigen receptor (CAR) T cells; however, the development of an affordable, safe, and effective CAR-T cell treatment remains a challenge. One of the major obstacles is that the suboptimal genetic modification of T cells reduces their yield and antitumor activity, necessitating the development of a next-generation T cell engineering approach. In this study, we developed a nonviral T cell nanoengineering system that allows highly efficient delivery of diverse functional nanomaterials into primary human T cells in a genetically stable and scalable manner. Our platform leverages the unique cell deformation and restoration process induced by the intrinsic inertial flow in a microchannel to create nanopores in the cellular membrane for macromolecule internalization, leading to effective transfection with high scalability and viability. The proposed approach demonstrates considerable potential as a practical alternative technique for improving the current CAR-T cell manufacturing process.


Assuntos
Imunoterapia Adotiva , Linfócitos T , Humanos , Imunoterapia Adotiva/métodos , Transfecção , Receptores de Antígenos de Linfócitos T/genética
3.
Lab Chip ; 23(7): 1758-1767, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36727443

RESUMO

In the past few years, messenger RNA (mRNA) has emerged as a promising therapeutic agent for the treatment and prevention of various diseases. Clinically, mRNA-based drugs have been used for cancer immunotherapy, infectious diseases, and genomic disorders. To maximize the therapeutic efficacy of mRNA, the exact amount of mRNAs must be delivered to the target locations without degradation; however, traditional delivery modalities, such as lipid carriers and electroporation, are suboptimal because of their high cost, cell-type sensitivity, low scalability, transfection/delivery inconsistency, and/or loss of cell functionality. Therefore, new effective and stable delivery methods are required. Accordingly, we present a novel nonlinear microfluidic cell stretching (µ-cell stretcher) platform that leverages viscoelastic fluids, i.e., methylcellulose (MC) solutions, and cell mechanoporation for highly efficient and robust intracellular mRNA delivery. In the proposed platform, cells suspended in MC solutions with mRNAs were injected into a microchannel where they rapidly passed through a single constriction. Owing to the use of viscoelastic MC solutions, a high shear force was applied to the cells, effectively creating transient nanopores. This feature allows mRNAs to be effectively internalized through generated membrane discontinuities. Using this platform, high delivery efficiency (∼97%), high throughput (∼3.5 × 105 cells per min), cell-type-/cargo-size-insensitive delivery, simple operation (single-step), low analyte consumption, low-cost operation (<$1), and nearly clogging-free operation were demonstrated, demonstrating the high potential of the proposed platform for application in mRNA-based cellular engineering research.


Assuntos
Eletroporação , Microfluídica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transfecção , Engenharia Celular
4.
Sci Rep ; 11(1): 17804, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34493759

RESUMO

For metastasis to occur, cancer cells must traverse a range of tissue environments. In part, this is accomplished by cells adjusting their migration mode to one that is best suited to the environment. Melanoma cells have been shown to be particularly plastic, frequently using both mesenchymal and amoeboid (bleb-based) modes of migration. It has been demonstrated that 2D confinement will promote the transition from mesenchymal to bleb-based migration. However, if melanoma cells similarly transition to bleb-based migration in response to 3D confinement, such as within narrow channels, is unknown. Here, using micro-fabricated channels, we demonstrate that metastatic, A375-M2, melanoma cells adopt features of both mesenchymal and bleb-based migration. In narrow (8 µm; height and width) channels coated with fibronectin, ~ 50% of melanoma cells were found to use either mesenchymal or bleb-based migration modes. In contrast, the inhibition of Src family kinases or coating channels with BSA, completely eliminated any features of mesenchymal migration. Detailed comparisons of migration parameters revealed that blebbing cells, particularly in the absence of adhesions, were faster than mesenchymal cells. In contrast to what has been previously shown under conditions of 2D confinement, pharmacologically inhibiting Arp2/3 promoted a fast filopodial-based mode of migration. Accordingly, we report that melanoma cells adopt a unique range of phenotypes under conditions of 3D confinement.


Assuntos
Técnicas de Cultura de Células/instrumentação , Melanoma/patologia , Metástase Neoplásica/patologia , Complexo 2-3 de Proteínas Relacionadas à Actina/antagonistas & inibidores , Movimento Celular , Forma Celular , Materiais Revestidos Biocompatíveis , Desenho de Equipamento , Fibronectinas , Adesões Focais , Humanos , Indóis/farmacologia , Mesoderma , Fenótipo , Pseudópodes/fisiologia , Estresse Mecânico
5.
Adv Sci (Weinh) ; 8(15): e2004595, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34096197

RESUMO

Innate cell function can be artificially engineered and reprogrammed by introducing biomolecules, such as DNAs, RNAs, plasmid DNAs, proteins, or nanomaterials, into the cytosol or nucleus. This process of delivering exogenous cargos into living cells is referred to as intracellular delivery. For instance, clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 gene editing begins with internalizing Cas9 protein and guide RNA into cells, and chimeric antigen receptor-T (CAR-T) cells are prepared by delivering CAR genes into T lymphocytes for cancer immunotherapies. To deliver external biomolecules into cells, tools, including viral vectors, and electroporation have been traditionally used; however, they are suboptimal for achieving high levels of intracellular delivery while preserving cell viability, phenotype, and function. Notably, as emerging solutions, microfluidic and nanofluidic approaches have shown remarkable potential for addressing this open challenge. This review provides an overview of recent advances in microfluidic and nanofluidic intracellular delivery strategies and discusses new opportunities and challenges for clinical applications. Furthermore, key considerations for future efforts to develop microfluidics- and nanofluidics-enabled next-generation intracellular delivery platforms are outlined.


Assuntos
Edição de Genes/métodos , Técnicas de Transferência de Genes , Terapia Genética/métodos , Microfluídica/métodos , Nanotecnologia/métodos , Humanos
6.
ACS Nano ; 14(3): 3048-3058, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32069037

RESUMO

In recent nanobiotechnology developments, a wide variety of functional nanomaterials and engineered biomolecules have been created, and these have numerous applications in cell biology. For these nanomaterials to fulfill their promises completely, they must be able to reach their biological targets at the subcellular level and with a high level of specificity. Traditionally, either nanocarrier- or membrane disruption-based method has been used to deliver nanomaterials inside cells; however, these methods are suboptimal due to their toxicity, inconsistent delivery, and low throughput, and they are also labor intensive and time-consuming, highlighting the need for development of a next-generation, intracellular delivery system. This study reports on the development of an intracellular nanomaterial delivery platform, based on unexpected cell-deformation phenomena via spiral vortex and vortex breakdown exerted in the cross- and T-junctions at moderate Reynolds numbers. These vortex-induced cell deformation and sequential restoration processes open cell membranes transiently, allowing effective and robust intracellular delivery of nanomaterials in a single step without the aid of carriers or external apparatus. By using the platform described here (termed spiral hydroporator), we demonstrate the delivery of different nanomaterials, including gold nanoparticles (200 nm diameter), functional mesoporous silica nanoparticles (150 nm diameter), dextran (hydrodynamic diameters between 2-55 nm), and mRNA, into different cell types. We demonstrate here that the system is highly efficient (up to 96.5%) with high throughput (up to 1 × 106 cells/min) and rapid delivery (∼1 min) while maintaining high levels of cell viability (up to 94%).


Assuntos
Dextranos/farmacologia , Sistemas de Liberação de Medicamentos , Nanoestruturas/química , Linhagem Celular Tumoral , Sobrevivência Celular , Dextranos/química , Humanos , Células K562 , Dispositivos Lab-On-A-Chip
7.
Small ; 13(28)2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28544415

RESUMO

Mechanical biomarkers associated with cytoskeletal structures have been reported as powerful label-free cell state identifiers. In order to measure cell mechanical properties, traditional biophysical (e.g., atomic force microscopy, micropipette aspiration, optical stretchers) and microfluidic approaches were mainly employed; however, they critically suffer from low-throughput, low-sensitivity, and/or time-consuming and labor-intensive processes, not allowing techniques to be practically used for cell biology research applications. Here, a novel inertial microfluidic cell stretcher (iMCS) capable of characterizing large populations of single-cell deformability near real-time is presented. The platform inertially controls cell positions in microchannels and deforms cells upon collision at a T-junction with large strain. The cell elongation motions are recorded, and thousands of cell deformability information is visualized near real-time similar to traditional flow cytometry. With a full automation, the entire cell mechanotyping process runs without any human intervention, realizing a user friendly and robust operation. Through iMCS, distinct cell stiffness changes in breast cancer progression and epithelial mesenchymal transition are reported, and the use of the platform for rapid cancer drug discovery is shown as well. The platform returns large populations of single-cell quantitative mechanical properties (e.g., shear modulus) on-the-fly with high statistical significances, enabling actual usages in clinical and biophysical studies.


Assuntos
Microfluídica/métodos , Animais , Citometria de Fluxo/métodos , Humanos , Técnicas Analíticas Microfluídicas
9.
Curr Opin Biotechnol ; 25: 114-23, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24484889

RESUMO

Micro-scale biological tools that have allowed probing of individual cells--from the genetic, to proteomic, to phenotypic level--have revealed important contributions of single cells to direct normal and diseased body processes. In analyzing single cells, sample heterogeneity between and within specific cell types drives the need for high-throughput and quantitative measurement of cellular parameters. In recent years, high-throughput single-cell analysis platforms have revealed rare genetic subpopulations in growing tumors, begun to uncover the mechanisms of antibiotic resistance in bacteria, and described the cell-to-cell variations in stem cell differentiation and immune cell response to activation by pathogens. This review surveys these recent technologies, presenting their strengths and contributions to the field, and identifies needs still unmet toward the development of high-throughput single-cell analysis tools to benefit life science research and clinical diagnostics.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Microtecnologia/métodos , Análise de Célula Única/métodos , Animais , Ensaios de Triagem em Larga Escala/instrumentação , Humanos , Microtecnologia/instrumentação , Neoplasias/química , Proteômica , Análise de Célula Única/instrumentação , Células-Tronco/química
10.
Lab Chip ; 9(5): 669-76, 2009 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-19224016

RESUMO

Small-scale insect inspired aircraft represent a promising approach to downscaling traditional aircraft designs. Despite advancements in microfabrication, however, it has proven difficult to fully replicate the mechanical complexities that enable these natural systems. As an alternative, recent efforts have used implanted electrical, optical or acoustic microsystems to exert direct control over insect flight. Here we demonstrate, for the first time, a method of directly and reversibly engineering insect flight metabolics using immature stage implanted microfluidics. We present our technique and device for on-command modulation of the internal levels of l-glutamic and l-aspartate acids and quantify the resulting changes in metabolic activity by monitoring respiratory CO(2) output. Microfluidic devices implanted 1 to 2 days prior to insects' emergence achieved survivability and flight-capable rates of 96% and 36%, respectively. Behavior ranging from retarded motion to complete, reversible paralysis, over timescales ranging from minutes to hours is demonstrated.


Assuntos
Voo Animal/fisiologia , Insetos/metabolismo , Insetos/fisiologia , Microfluídica/métodos , Atividade Motora/efeitos dos fármacos , Animais , Dióxido de Carbono/metabolismo , Cinética , Manduca , Metabolismo/fisiologia , Microcomputadores , Microinjeções , Microcirurgia , Nanotecnologia , Sistema Nervoso/efeitos dos fármacos , Paralisia/induzido quimicamente , Paralisia/fisiopatologia , Mecânica Respiratória/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA