Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Sci (Weinh) ; 10(31): e2304108, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37702128

ABSTRACT

In this study, the potential of complex emulsions is investigated as transducers in sensing applications. Complex emulsions are stabilized without external detergents by developing a novel α-cyanostilbene substituted with PEG and semi-perfluoroalkyl chain (CNFCPEG). CNFCPEG exhibits unique variable emission properties depending on its aggregation state, allowing dual blue and green emissions in complex emulsions with hydrocarbon-in-fluorocarbon-in-water (H/F/W) morphology. The green excimer emissions result from the self-assembly of CNFCPEG at the fluorocarbon/water interface, while the blue emission observed is due to aggregation in the organic phase. A novel flow-injection method is developed by incorporating complex emulsions with CNFCPEG into multiple-well flow chips (MWFC). Iodine is successfully detected in a mobile aqueous solution by monitoring morphology changes. The findings demonstrate that self-stabilized complex emulsions with MWFC hold great promise for real-time sensing without costly instruments.

2.
Lab Chip ; 23(7): 1726-1751, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36811978

ABSTRACT

Single-cell proteomics (SCP) reveals phenotypic heterogeneity by profiling individual cells, their biological states and functional outcomes upon signaling activation that can hardly be probed via other omics characterizations. This has become appealing to researchers as it enables an overall more holistic view of biological details underlying cellular processes, disease onset and progression, as well as facilitates unique biomarker identification from individual cells. Microfluidic-based strategies have become methods of choice for single-cell analysis because they allow facile assay integrations, such as cell sorting, manipulation, and content analysis. Notably, they have been serving as an enabling technology to improve the sensitivity, robustness, and reproducibility of recently developed SCP methods. Critical roles of microfluidics technologies are expected to further expand rapidly in advancing the next phase of SCP analysis to reveal more biological and clinical insights. In this review, we will capture the excitement of the recent achievements of microfluidics methods for both targeted and global SCP, including efforts to enhance the proteomic coverage, minimize sample loss, and increase multiplexity and throughput. Furthermore, we will discuss the advantages, challenges, applications, and future prospects of SCP.


Subject(s)
Microfluidics , Proteomics , Microfluidics/methods , Proteomics/methods , Reproducibility of Results , Cell Separation , Single-Cell Analysis/methods
3.
Nat Commun ; 13(1): 37, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013269

ABSTRACT

Single-cell proteomics can reveal cellular phenotypic heterogeneity and cell-specific functional networks underlying biological processes. Here, we present a streamlined workflow combining microfluidic chips for all-in-one proteomic sample preparation and data-independent acquisition (DIA) mass spectrometry (MS) for proteomic analysis down to the single-cell level. The proteomics chips enable multiplexed and automated cell isolation/counting/imaging and sample processing in a single device. Combining chip-based sample handling with DIA-MS using project-specific mass spectral libraries, we profile on average ~1,500 protein groups across 20 single mammalian cells. Applying the chip-DIA workflow to profile the proteomes of adherent and non-adherent malignant cells, we cover a dynamic range of 5 orders of magnitude with good reproducibility and <16% missing values between runs. Taken together, the chip-DIA workflow offers all-in-one cell characterization, analytical sensitivity and robustness, and the option to add additional functionalities in the future, thus providing a basis for advanced single-cell proteomics applications.


Subject(s)
Lab-On-A-Chip Devices , Mass Spectrometry/methods , Microfluidics/methods , Proteomics/methods , Animals , Cell Line, Tumor , Cell Separation , Humans , Lung Neoplasms , Proteome , Reproducibility of Results , Workflow
SELECTION OF CITATIONS
SEARCH DETAIL
...