Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
ACS Nano ; 17(9): 8108-8122, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37129374

ABSTRACT

Tumor derived exosomes (TEXs) have emerged as promising biomarkers for cancer liquid biopsy. Conventional methods (such as ELISA and qRT-PCR) and emerging biosensing technologies mainly detect a single type of exosomal biomarker due to the distinct properties of different biomolecules. Sensitive detection of two different types of TEX biomarkers, i.e., protein and microRNA combined biomarkers, may greatly improve cancer diagnostic accuracy. We developed an exosome protein microRNA one-stop (Exo-PROS) biosensor that not only selectively captured TEXs but also enabled in situ, simultaneous detection of TEX protein-microRNA pairs via a surface plasmon resonance mechanism. Exo-PROS assay is a fast, reliable, low sample consumption, and user-friendly test. With a total of 175 cancer patients and normal controls, we demonstrated that TEX protein-microRNA pairs measured by Exo-PROS assay detected lung cancer and breast cancer with 99% and 96% accuracy, respectively. Exo-PROS assay also showed superior diagnostic performance to conventional ELISA and qRT-PCR methods. Our results demonstrated that Exo-PROS assay is a potent liquid biopsy assay for cancer diagnosis.


Subject(s)
Biosensing Techniques , Exosomes , Lung Neoplasms , MicroRNAs , Humans , MicroRNAs/genetics , Exosomes/metabolism , Biomarkers, Tumor/analysis , Neoplasm Proteins/metabolism , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Biosensing Techniques/methods
2.
ACS Appl Nano Mater ; 4(3): 2806-2819, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-34849458

ABSTRACT

Exosomes are cell-derived, nanosized extracellular vesicles for intercellular communication. Exosomal RNAs have been shown as one type of promising cancer liquid biopsy biomarkers. Conventional methods to characterize exosomal RNAs such as quantitative reverse transcription polymerase chain reaction (qRT-PCR) are limited by low sensitivity, large sample consumption, time-consuming process, and high cost. Many technologies have been developed to overcome these challenges; however, many hours are still required to complete the assays, especially when exosome lysis and RNA extraction are required. We have developed a microfluidic cationic lipoplex nanoparticles (mCLN) assay that utilizes a micromixer biochip to allow for the effective capture of exosomes by cationic lipoplex nanoparticles and thus enables ultrafast and sensitive exosomal RNA detection for cancer diagnosis. The sensing performance and diagnostic performance of the mCLN assay were investigated using non-small cell lung cancer (NSCLC) as the disease model and exosomal microRNA-21 and TTF-1 mRNA as the biomarkers. The limits of detection of the mCLN assay were 2.06 × 109 and 3.71 × 109 exosomes/mL for microRNA-21 and TTF-1 mRNA, respectively, indicating that the mCLN assay may require as low as 1 µL of serum for exosomal RNA detection. The mCLN assay successfully distinguished NSCLC from normal controls by detecting significantly higher microRNA-21 and TTF-1 mRNA levels in exosomes from both NSCLC patient serum samples and A549 NSCLC cells than those from normal controls and BEAS-2B normal bronchial epithelial cells. Compared with conventional qRT-PCR assay, the mCLN assay showed a higher diagnostic accuracy in lung cancer, required less sample volume (30 vs 100 µL), and consumed much less time (10 min vs 4 h).

3.
Front Genet ; 11: 258, 2020.
Article in English | MEDLINE | ID: mdl-32265989

ABSTRACT

Circulating microRNAs carried by exosomes have emerged as promising diagnostic biomarkers for cancer because of their abundant amount and remarkable stability in body fluids. Exosomal microRNAs in blood are typically quantified using the RNA isolation-qRT-PCR workflow, which cannot distinguish circulating microRNAs secreted by cancer cells from those released by non-tumor cells, making it potentially less sensitive in detecting cancer-specific microRNA biomarkers. We have developed a sensitive and simple tethered cationic lipoplex nanoparticles (tCLN) biochip to detect exosomal microRNAs in human sera. The tCLN biochip allows the discrimination of tumor-derived exosomes from their non-tumor counterparts, and thus achieves higher detection sensitivity and specificity than qRT-PCR. We have demonstrated the clinical utility of the tCLN biochip in lung cancer diagnosis using sera from normal controls, therapy-naive early stage and late stage non-small cell lung cancer (NSCLC) patients. Total five microRNAs (miR-21, miR-25, miR-155, miR-210, and miR-486) were selected as the biomarkers. Each microRNA biomarker measured by tCLN assay showed higher sensitivity and specificity in lung cancer detection than that measured by qRT-PCR. When all five microRNAs were combined, the tCLN assay distinguished normal controls from all NSCLC patients with sensitivity of 0.969, specificity of 0.933 and AUC of 0.970, and provided much better diagnostic accuracy than qRT-PCR (sensitivity = 0.469, specificity = 1.000, AUC = 0.791). Remarkably, the tCLN assay achieved absolute sensitivity and specificity in discriminating early stage NSCLC patients from normal controls, demonstrating its great potential as a liquid biopsy assay for lung cancer early detection.

4.
J Extracell Vesicles ; 9(1): 1692401, 2020.
Article in English | MEDLINE | ID: mdl-31807236

ABSTRACT

Both exosomes and soluble factors have been implicated in the generation of an immunosuppressive tumour microenvironment. Determining the contribution of each requires stringent control of purity of the isolated analytes. The present study compares several conventional exosome isolation methods for the presence of co-enriched soluble factors while isolating exosomes from human melanoma-derived cell lines. The resultant preparations were analysed by multiplex bead array analysis for cytokine profiles, and by electron microscopy and nanotracking analysis for exosome size distribution and concentration. It is demonstrated that the amount and repertoire of soluble factors in exosome preparations is dependent upon the isolation method used. A combination of ultrafiltration and size exclusion chromatography yielded up to 58-fold more exosomes than ultracentrifugation, up to 836-fold lower concentrations of co-purified soluble factors when adjusted for exosome yield, and a greater than two-fold increase in PD-L1 expressing exosomes. Mechanistically, in context of the immunomodulatory effects of exosomes, the exosome isolation method should be carefully considered in order to limit any effects due instead to co-eluted soluble factors.

5.
Article in English | MEDLINE | ID: mdl-30983848

ABSTRACT

We report a plasmonic interferometer array (PIA) sensor and demonstrate its ability to detect circulating exosomal proteins in real-time with high sensitivity and low cost to enable the early detection of cancer. Specifically, a surface plasmon wave launched by the nano-groove rings interferes with the free-space light at the output of central nano-aperture and results in an intensity interference pattern. Under the single-wavelength illumination, when the target exosomal proteins are captured by antibodies bound on the surface, the biomediated change in the refractive index between the central aperture and groove rings causes the intensity change in transmitted light. By recording the intensity changes in real-time, one can effectively screen biomolecular binding events and analyze the binding kinetics. By integrating signals from multiple sensor pairs to enhance the signal-to-noise ratio, superior sensing resolutions of 1.63×10-6 refractive index unit (RIU) in refractive index change and 3.86×108 exosomes/mL in exosome detection were realized, respectively. Importantly, this PIA sensor can be imaged by a miniaturized microscope system coupled with a smart phone to realize a portable and highly sensitive healthcare device. The sensing resolution of 9.72×109 exosomes/mL in exosome detection was realized using the portable sensing system building upon a commercial smartphone.

6.
Sci Rep ; 9(1): 4959, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30874569

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

7.
ACS Appl Mater Interfaces ; 10(50): 43375-43386, 2018 Dec 19.
Article in English | MEDLINE | ID: mdl-30451486

ABSTRACT

Tumor-derived exosomes (TEXs) play instrumental roles in tumor growth, angiogenesis, immune modulation, metastasis, and drug resistance. TEX RNAs are a new class of noninvasive biomarkers for cancer. Neither current techniques, such as quantitative reverse transcription polymerase chain reaction (qRT-PCR) and next-generation sequencing, nor new ones, such as electrochemical or surface plasmon resonance-based biosensors, are able to selectively capture and separate TEXs from normal cell-derived exosomes, making TEX RNAs potentially less sensitive biomarkers. We developed an immuno-biochip that selectively captures TEXs using antibodies against tumor-associated proteins and quantifies in situ TEX RNAs using cationic lipoplexes containing molecular beacons. We used the immuno-biochip to measure the expression of miR-21 microRNA and TTF-1 mRNA in EGFR- or PD-L1-bearing exosomes from human sera and achieved absolute sensitivity and specificity in distinguishing normal controls from non-small cell lung cancer patients. Our results demonstrated that the effective separation of TEXs from other exosomes greatly improved the detection sensitivity and specificity. Compared with the traditional immunomagnetic separation-RNA isolation-qRT-PCR workflow, the immuno-biochip showed superior lung cancer diagnostic performance, consumed less samples (∼30 µL), and shortened assay time from ∼24 to 4 h.


Subject(s)
Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung , Cell-Free Nucleic Acids , Exosomes , Lung Neoplasms , Microarray Analysis , RNA, Neoplasm , Real-Time Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , A549 Cells , Antibodies, Neoplasm/chemistry , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/genetics , Exosomes/genetics , Exosomes/metabolism , Female , Humans , Lung Neoplasms/blood , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Male , Neoplasm Proteins/blood , Neoplasm Proteins/chemistry , RNA, Neoplasm/blood , RNA, Neoplasm/genetics
8.
Sci Rep ; 8(1): 12905, 2018 08 27.
Article in English | MEDLINE | ID: mdl-30150674

ABSTRACT

Local acidification of stroma is proposed to favour pre-metastatic niche formation but the mechanism of initiation is unclear. We investigated whether Human Melanoma-derived exosomes (HMEX) could reprogram human adult dermal fibroblasts (HADF) and cause extracellular acidification. HMEX were isolated from supernatants of six melanoma cell lines (3 BRAF V600E mutant cell lines and 3 BRAF wild-type cell lines) using ultracentrifugation or Size Exclusion Chromatography (SEC). Rapid uptake of exosomes by HADF was demonstrated following 18 hours co-incubation. Exposure of HDAF to HMEX leads to an increase in aerobic glycolysis and decrease in oxidative phosphorylation (OXPHOS) in HADF, consequently increasing extracellular acidification. Using a novel immuno-biochip, exosomal miR-155 and miR-210 were detected in HMEX. These miRNAs were present in HMEX from all six melanoma cell lines and were instrumental in promoting glycolysis and inhibiting OXPHOS in tumour cells. Inhibition of miR-155 and miR-210 activity by transfection of miRNA inhibitors into HMEX reversed the exosome-induced metabolic reprogramming of HADF. The data indicate that melanoma-derived exosomes modulate stromal cell metabolism and may contribute to the creation of a pre-metastatic niche that promotes the development of metastasis.


Subject(s)
Cellular Reprogramming/physiology , Exosomes/metabolism , Melanoma/metabolism , MicroRNAs/metabolism , Aerobiosis/genetics , Aerobiosis/physiology , Cell Line, Tumor , Cellular Reprogramming/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Glycolysis/genetics , Glycolysis/physiology , Humans , Melanoma/genetics , MicroRNAs/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology
9.
ACS Sens ; 3(8): 1471-1479, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30019892

ABSTRACT

Exosomes are small extracellular vesicles released by cells for cell-cell communication. They play important roles in cancer development, metastasis, and drug resistance. Exosomal proteins have been demonstrated by many studies as promising biomarkers for cancer screening, diagnosis, and monitoring. Among many detection techniques, surface plasmon resonance (SPR) is a highly sensitive, label-free, and real-time optical detection method. Commercial prism-based wavelength/angular-modulated SPR sensors afford high sensitivity and resolution, but their large footprint and high cost limit their adaptability for clinical settings. Recently, a nanoplasmonic exosome (nPLEX) assay was developed to detect exosomal proteins for ovarian cancer diagnosis. However, comparing with conventional SPR biosensors, the broad applications of nanoplasmonic biosensors are limited by the difficult and expensive fabrication of nanostructures. We have developed an intensity-modulated, compact SPR biosensor (25 cm × 10 cm × 25 cm) which uses a conventional SPR sensing mechanism and does not require nanostructure fabrication. Calibration from glycerol showed that the compact SPR biosensor offered sensitivity of 9.258 × 103%/RIU and resolution of 8.311 × 10-6 RIU. We have demonstrated the feasibility of the compact SPR biosensor in lung cancer diagnosis using exosomal epidermal growth factor receptor (EGFR) and programmed death-ligand 1 (PD-L1) as biomarkers. It detected a higher level of exosomal EGFR from A549 nonsmall cell lung cancer (NSCLC) cells than BEAS-2B normal cells. With human serum samples, the compact SPR biosensor detected similar levels of exosomal EGFR in NSCLC patients and normal controls, and higher expression of exosomal PD-L1 in NSCLC patients than normal controls. The compact SPR biosensor showed higher detection sensitivity than ELISA and similar sensing accuracy as ELISA. It is a simple and user-friendly sensing platform, which may serve as an in vitro diagnostic test for cancer.


Subject(s)
Biosensing Techniques/methods , Carcinoma, Non-Small-Cell Lung/diagnosis , ErbB Receptors/analysis , Exosomes/metabolism , Lung Neoplasms/diagnosis , Surface Plasmon Resonance , Aged , B7-H1 Antigen/analysis , Biomarkers, Tumor/analysis , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Protein Array Analysis , Surface Properties
10.
AAPS J ; 20(2): 41, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29520676

ABSTRACT

Current cancer diagnostic methods are challenged by low sensitivity, high false positive rate, limited tumor information, uncomfortable or invasive procedures, and high cost. Liquid biopsy that analyzes circulating biomarkers in body fluids represents a promising solution to these challenges. Exosomes are one of the promising cancer biomarkers for liquid biopsy because they are cell-secreted, nano-sized, extracellular vesicles that stably exist in all types of body fluids. Exosomes transfer DNAs, RNAs, proteins, and lipids from parent cells to recipient cells for intercellular communication and play important roles in cancer initiation, progression, and metastasis. Many liquid biopsy biosensors have been developed to offer non- or minimally-invasive, highly sensitive, simple, rapid, and cost-effective cancer diagnostics. This review summarized recent advances of liquid biopsy biosensors with a focus on the detection of exosomal proteins as biomarkers for cancer screening, diagnosis, and prognosis. We reviewed six major types of liquid biopsy biosensors including immunofluorescence biosensor, colorimetric biosensor, surface plasmon resonance (SPR) biosensor, surface-enhanced Raman scattering (SERS) biosensor, electrochemical biosensor, and nuclear magnetic resonance (NMR) biosensor. We shared our perspectives on future improvement of exosome-based liquid biopsy biosensors to accelerate their clinical translation.


Subject(s)
Biomarkers, Tumor/analysis , Biosensing Techniques/methods , Early Detection of Cancer/methods , Exosomes/metabolism , Neoplasms/diagnosis , Disease Progression , Humans , Liquid Biopsy/methods , Neoplasms/mortality , Neoplasms/pathology , Prognosis , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...