Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
J Colloid Interface Sci ; 678(Pt B): 925-937, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39270392

RÉSUMÉ

The effectiveness of chemokinetic therapy nanozymes is severely constrained because of the low H2O2 levels in the tumor microenvironment. Unlike other self-produced H2O2 nanozymes, the N-CNTs-encapsulated CoNi alloy (CoNiCoNC) with glucose oxidase and lactate oxidase activities has two ways to produce H2O2. It can facilitate the transformation of glucose and lactic acid into H2O2 simultaneously. First, the H2O2 generation pathway is favorable for aggravating energy metabolism. Second, some produced H2O2 can be decomposed by CoNiCoNC to H2O and O2 with the 4e- pathway to alleviate the TME hypoxia. Third, H2O2 can be catalyzed to form OH to enhance reactive oxygen species (ROS) content. Through proteomic analysis, nanozymes substantially impact the metabolic pathways of cancer cells because of their aggravating energy metabolism. The high levels of ROS can cause mitochondrial lipid peroxidation and cellular ferroptosis. Consequently, the two-way H2O2-selective nanoenzymatic platform realizes the synergistic effect of starvation therapy and chemokinetics.

2.
J Biophotonics ; 17(9): e202400192, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38938144

RÉSUMÉ

Non-invasive screening for bladder cancer is crucial for treatment and postoperative follow-up. This study combines digital microfluidics (DMF) technology with fluorescence lifetime imaging microscopy (FLIM) for urine analysis and introduces a novel non-invasive bladder cancer screening technique. Initially, the DMF was utilized to perform preliminary screening and enrichment of urine exfoliated cells from 54 participants, followed by cell staining and FLIM analysis to assess the viscosity of the intracellular microenvironment. Subsequently, a deep learning residual convolutional neural network was employed to automatically classify FLIM images, achieving a three-class prediction of high-risk (malignant), low-risk (benign), and minimal risk (normal) categories. The results demonstrated a high consistency with pathological diagnosis, with an accuracy of 91% and a precision of 93%. Notably, the method is sensitive for both high-grade and low-grade bladder cancer cases. This highly accurate non-invasive screening method presents a promising approach for bladder cancer screening with significant clinical application potential.


Sujet(s)
Apprentissage profond , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/imagerie diagnostique , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/diagnostic , Humains , Microscopie de fluorescence , Dépistage précoce du cancer/méthodes , Mâle , Traitement d'image par ordinateur/méthodes , Femelle , Microfluidique , Adulte d'âge moyen , Sujet âgé
3.
Cells ; 12(11)2023 05 31.
Article de Anglais | MEDLINE | ID: mdl-37296645

RÉSUMÉ

Mesenchymal stem cells (MSCs) play a crucial role in tissue engineering, as their differentiation status directly affects the quality of the final cultured tissue, which is critical to the success of transplantation therapy. Furthermore, the precise control of MSC differentiation is essential for stem cell therapy in clinical settings, as low-purity stem cells can lead to tumorigenic problems. Therefore, to address the heterogeneity of MSCs during their differentiation into adipogenic or osteogenic lineages, numerous label-free microscopic images were acquired using fluorescence lifetime imaging microscopy (FLIM) and stimulated Raman scattering (SRS), and an automated evaluation model for the differentiation status of MSCs was built based on the K-means machine learning algorithm. The model is capable of highly sensitive analysis of individual cell differentiation status, so it has great potential for stem cell differentiation research.


Sujet(s)
Adipogenèse , Cellules souches mésenchymateuses , Différenciation cellulaire , Cellules souches , Microscopie de fluorescence
4.
ACS Sens ; 5(1): 110-117, 2020 01 24.
Article de Anglais | MEDLINE | ID: mdl-31829015

RÉSUMÉ

Optofluidic lasers are currently of high interest for sensitive intracavity biochemical analysis. In comparison with conventional methods such as fluorescence and colorimetric detection, optofluidic lasers provide a method for amplifying small concentration differences in the gain medium, thus achieving high sensitivity. Here, we report the development of an on-chip ELISA (enzyme-linked immunosorbent assay) laser platform that is able to complete an assay in a short amount of time with small sample/reagent volumes, large dynamic range, and high sensitivity. The arrayed microscale reaction wells in the ELISA lasers can be microfabricated directly on dielectric mirrors, thus significantly improving the quality of the reaction wells and detection reproducibility. The details of the fabrication and characterization of those reaction wells on the mirror are described and the ELISA laser assay protocols are developed. Finally, we applied the ELISA laser to detecting IL-6, showing that a detection limit of about 0.1 pg/mL can be achieved in 1.5 h with 15 µL of sample/reagents per well. This work pushes the ELISA laser a step closer to solving problems in real-world biochemical analysis.


Sujet(s)
Techniques de biocapteur/méthodes , Test ELISA/méthodes , Humains , Lasers
5.
Biosens Bioelectron ; 131: 60-66, 2019 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-30826651

RÉSUMÉ

Turbidimetric inhibition immunoassay (TIIA) is a classic immunodiagnostic method that has been extensively used for biomarker detection. However, the low sensitivity of this technique hinders its applications in the early diagnosis of diseases. Here, a new concept, optofluidic laser TIIA (OFL-TIIA), is proposed and demonstrated for sensitive protein detection. In contrast to the immunoreaction in traditional TIIA, in which the single-pass laser loss is detected, the immunoreaction in the OFL-TIIA method takes place in a laser cavity, which considerably increases the loss induced by antigen-antibody complexes (AACs) via the amplification effect of the laser. A commercial IgG TIIA kit was selected as a demonstrative model to characterize the performance of OFL-TIIA. A wide dynamic range of five orders of magnitude with an exceptional limit of detection (LOD) (1.8 × 10-10 g/L) was achieved. OFL-TIIA is a fast, sensitive, and low-cost immunoassay with a simple homogeneous and wash-free process and low-volume sample consumption, thus providing a new detection platform for disease diagnostics.


Sujet(s)
Complexe antigène-anticorps/isolement et purification , Marqueurs biologiques/composition chimique , Techniques de biocapteur , Dosage immunologique , Complexe antigène-anticorps/immunologie , Humains , Lasers
6.
Biomed Opt Express ; 10(2): 838-854, 2019 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-30800518

RÉSUMÉ

We developed and applied rapid scanning laser-emission microscopy (LEM) to detect abnormal changes in cell nuclei for early diagnosis of cancer and cancer precursors. Regulation of chromatins is essential for genetic development and normal cell functions, while abnormal nuclear changes may lead to many diseases, in particular, cancer. The capability to detect abnormal changes in "apparently normal" tissues at a stage earlier than tumor development is critical for cancer prevention. Here we report using LEM to analyze colonic tissues from mice at-risk for colon cancer (induced by a high-fat diet) by detecting pre-polyp nuclear abnormality. By imaging the lasing emissions from chromatins, we discovered that, despite the absence of observable lesions, polyps, or tumors under stereoscope, high-fat mice exhibited significantly lower lasing thresholds than low-fat mice. The low lasing threshold is, in fact, very similar to that of adenomas and is caused by abnormal cell proliferation and chromatin deregulation that can potentially lead to cancer. Our findings suggest that conventional detection methods, such as colonoscopy followed by histopathology, by itself, may be insufficient to reveal hidden or early tumors under development. We envision that this innovative work will provide new insights into LEM and support existing tools for early tumor detection in clinical diagnosis, and fundamental biological and biomedical research of chromatin changes at the biomolecular level of cancer development.

7.
Lab Chip ; 18(18): 2741-2748, 2018 09 11.
Article de Anglais | MEDLINE | ID: mdl-30094434

RÉSUMÉ

Optofluidic lasers (OFLs) are an emerging technological platform for biochemical sensing, and their good performance especially high sensitivity has been demonstrated. However, high-throughput detection with an OFL remains a major challenge due to the lack of reproducible optical microcavities. Here, we introduce the concept of a distributed fibre optofluidic laser (DFOFL) and demonstrate its potential for high-throughput sensing applications. Due to the precise fibre geometry control via fibre drawing, a series of identical optical microcavities uniformly distributed along a hollow optical fibre (HOF) can be achieved to obtain a one-dimensional (1D) DFOFL. An enzymatic reaction catalysed by horseradish peroxidase (HRP) can be monitored over time, and the HRP concentration is detected by DFOFL-based arrayed colorimetric detection. Experimentally, five-channel detection in parallel with imaging has been demonstrated. Theoretically, spatial multiplexing of hundreds of channels is achievable with DFOFL-based detection. The DFOFL wavelength is tuned over hundreds of nanometers by optimizing the dye concentration or reconfiguring the liquid gain materials. Extending this concept to a two-dimensional (2D) chip through wavelength multiplexing can further enhance its multi-functionality, including multi-sample detection and spectral analysis. This work opens the door to high-throughput biochemical sensing.


Sujet(s)
Techniques de biocapteur/instrumentation , Lasers , Fibres optiques , Horseradish peroxidase/métabolisme
8.
Nanoscale ; 10(20): 9729-9735, 2018 May 24.
Article de Anglais | MEDLINE | ID: mdl-29762623

RÉSUMÉ

We investigate a cadmium sulfide (CdS) nanowire (NW) laser that is spontaneously internalized into a single cell to serve as a stand-alone intracellular probe. By pumping with nano-joule light pulses, green laser emission (500-520 nm) can be observed inside cells with a peak linewidth as narrow as 0.5 nm. Due to the sub-micron diameter (∼200 nm), the NW has an appreciable fraction of the evanescent field outside, facilitating a sensitive detection of cellular environmental changes. By monitoring the lasing peak wavelength shift in response to the intracellular refractive index change, our NW laser probe shows a sensitivity of 55 nm per RIU (refractive index units) and a figure of merit of approximately 98.

9.
Lab Chip ; 18(7): 1057-1065, 2018 03 27.
Article de Anglais | MEDLINE | ID: mdl-29511754

RÉSUMÉ

Laser emission-based detection and imaging technology has attracted significant interest in biomedical research due to its high sensitivity, narrow linewidth, and superior spectral and spatial resolution. Recent advances have further revealed the potential to use laser emission to investigate chromatin dynamics, as well as to diagnose cancer tissues based on nuclear biomarkers. To move the laser emission based detection technology a step further towards practical use, in this work, we developed a highly robust tissue laser platform by microfabricating an SU8 spacer with a fixed height on the top mirror of the Fabry-Pérot (FP) cavity, which allows generation of reproducible and stable lasing results regardless of tissue thickness. Then we applied this platform to achieve lasing emission from formalin-fixed, paraffin-embedded (FFPE) lung tissues, which account for an overwhelming fraction of tissues collected for research and clinical use worldwide. We further showed that the cancer and normal FFPE lung tissues can be distinguished by their respective lasing thresholds. Two different tissue thicknesses (10 µm and 5 µm) commonly used in pathological labs were explored. Finally, we tested three additional types of tissues (colon, stomach, and breast) that were prepared independently by lab technicians in a pathology lab in China and shipped to the US in order to validate the general applicability and practicality of the laser emission-based technology as well as the corresponding sample preparation protocol and the tissue laser platform. Our work will not only vastly broaden the applications of laser emission-based detection/imaging technology but also help translate it from the laboratory to an automated system for clinical practice that may eventually benefit biomedicine and biological research.


Sujet(s)
Formaldéhyde/composition chimique , Lasers , Inclusion en paraffine , Recherche biomédicale , Biopsie , Humains
10.
Nat Biomed Eng ; 1: 724-735, 2017.
Article de Anglais | MEDLINE | ID: mdl-29204310

RÉSUMÉ

Detection of nuclear biomarkers such as nucleic acids and nuclear proteins is critical for early-stage cancer diagnosis and prognosis. Conventional methods relying on morphological assessment of cell nuclei in histopathology slides may be subjective, whereas colorimetric immunohistochemical and fluorescence-based imaging are limited by strong light absorption, broad-emission bands and low contrast. Here, we describe the development and use of a scanning laser-emission-based microscope that maps lasing emissions from nuclear biomarkers in human tissues. 41 tissue samples from 35 patients labelled with site-specific and biomarker-specific antibody-conjugated dyes were sandwiched in a Fabry-Pérot microcavity while an excitation laser beam built a laser-emission image. We observed multiple sub-cellular lasing emissions from cancer cell nuclei, with a threshold of tens of µJ/mm2, sub-micron resolution (<700 nm), and a lasing band in the few-nanometre range. Different lasing thresholds of nuclei in cancer and normal tissues enabled the identification and multiplexed detection of nuclear proteomic biomarkers, with a high sensitivity for early-stage cancer diagnosis. Laser-emission-based cancer screening and immunodiagnosis might find use in precision medicine and facilitate research in cell biology.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE