Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Anal Chem ; 96(16): 6321-6328, 2024 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-38595097

RESUMO

Small extracellular vesicles (sEVs) are heterogeneous biological nanoparticles (NPs) with wide biomedicine applications. Tracking individual nanoscale sEVs can reveal information that conventional microscopic methods may lack, especially in cellular microenvironments. This usually requires biolabeling to identify single sEVs. Here, we developed a light scattering imaging method based on dark-field technology for label-free nanoparticle diffusion analysis (NDA). Compared with nanoparticle tracking analysis (NTA), our method was shown to determine the diffusion probabilities of a single NP. It was demonstrated that accurate size determination of NPs of 41 and 120 nm in diameter is achieved by purified Brownian motion (pBM), without or within the cell microenvironments. Our pBM method was also shown to obtain a consistent size estimation of the normal and cancerous plasma-derived sEVs without and within cell microenvironments, while cancerous plasma-derived sEVs are statistically smaller than normal ones. Moreover, we showed that the velocity and diffusion coefficient are key parameters for determining the diffusion types of the NPs and sEVs in a cancerous cell microenvironment. Our light scattering-based NDA and pBM methods can be used for size determination of NPs, even in cell microenvironments, and also provide a tool that may be used to analyze sEVs for many biomedical applications.


Assuntos
Vesículas Extracelulares , Vesículas Extracelulares/química , Humanos , Luz , Nanopartículas/química , Espalhamento de Radiação , Microambiente Celular , Tamanho da Partícula , Difusão , Microambiente Tumoral , Linhagem Celular Tumoral , Movimento (Física)
2.
Cytometry A ; 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-39101554

RESUMO

Imaging flow cytometry, which combines the advantages of flow cytometry and microscopy, has emerged as a powerful tool for cell analysis in various biomedical fields such as cancer detection. In this study, we develop multiplex imaging flow cytometry (mIFC) by employing a spatial wavelength division multiplexing technique. Our mIFC can simultaneously obtain brightfield and multi-color fluorescence images of individual cells in flow, which are excited by a metal halide lamp and measured by a single detector. Statistical analysis results of multiplex imaging experiments with resolution test lens, magnification test lens, and fluorescent microspheres validate the operation of the mIFC with good imaging channel consistency and micron-scale differentiation capabilities. A deep learning method is designed for multiplex image processing that consists of three deep learning networks (U-net, very deep super resolution, and visual geometry group 19). It is demonstrated that the cluster of differentiation 24 (CD24) imaging channel is more sensitive than the brightfield, nucleus, or cancer antigen 125 (CA125) imaging channel in classifying the three types of ovarian cell lines (IOSE80 normal cell, A2780, and OVCAR3 cancer cells). An average accuracy rate of 97.1% is achieved for the classification of these three types of cells by deep learning analysis when all four imaging channels are considered. Our single-detector mIFC is promising for the development of future imaging flow cytometers and for the automatic single-cell analysis with deep learning in various biomedical fields.

3.
Biomed Opt Express ; 15(4): 2063-2077, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38633087

RESUMO

Automatic and label-free screening methods may help to reduce cervical cancer mortality rates, especially in developing regions. The latest advances of deep learning in the biomedical optics field provide a more automatic approach to solving clinical dilemmas. However, existing deep learning methods face challenges, such as the requirement of manually annotated training sets for clinical sample analysis. Here, we develop Siamese deep learning video flow cytometry for the analysis of clinical cervical cancer cell samples in a smear-free manner. High-content light scattering images of label-free single cells are obtained via the video flow cytometer. Siamese deep learning, a self-supervised method, is built to introduce cell lineage cells into an analysis of clinical cells, which utilizes generated similarity metrics as label annotations for clinical cells. Compared with other deep learning methods, Siamese deep learning achieves a higher accuracy of up to 87.11%, with about 5.62% improvement for label-free clinical cervical cancer cell classification. The Siamese deep learning video flow cytometry demonstrated here is promising for automatic, label-free analysis of many types of cells from clinical samples without cell smears.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA