Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cancer Sci ; 115(5): 1602-1610, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38480462

RESUMEN

Anti-programmed death-ligand 1 (PD-L1) Ab-based therapies have demonstrated potential for treating metastatic urothelial cancer with high PD-L1 expression. Urinary exosomes are promising biomarkers for liquid biopsy, but urine's high variability requires normalization for accurate analysis. This study proposes using the PD-L1/Alix ratio to normalize exosomal PD-L1 signal intensity with Alix, an internal exosomal protein less susceptible to heterogeneity concerns than surface protein markers. Extracellular vesicles were isolated using ExoDisc and characterized using various methods, including ExoView to analyze tetraspanins, PD-L1, and Alix on individual exosomes. On-disc ELISA was used to evaluate PD-L1 and Alix-normalized PD-L1 in 15 urothelial cancer patients during the initial treatment cycle with Tecentriq. Results showed that Alix signal range was relatively uniform, whereas tetraspanin marker intensity varied for individual exosome particles. On-disc ELISA was more reliable for detecting exosomal PD-L1 expression than standard plate ELISA-based measurement. Using exosomal Alix expression for normalization is a more reliable approach than conventional methods for monitoring patient status. Overall, the study provides a practical and reliable method for detecting exosomal PD-L1 in urine samples from patients with urothelial cancer.


Asunto(s)
Antígeno B7-H1 , Biomarcadores de Tumor , Exosomas , Humanos , Exosomas/metabolismo , Antígeno B7-H1/orina , Biomarcadores de Tumor/orina , Proteínas de Ciclo Celular/orina , Ensayo de Inmunoadsorción Enzimática/métodos , Masculino , Neoplasias de la Vejiga Urinaria/orina , Neoplasias de la Vejiga Urinaria/patología , Femenino , Anciano , Persona de Mediana Edad , Neoplasias Urológicas/orina , Neoplasias Urológicas/patología , Biopsia Líquida/métodos
2.
Anal Chem ; 92(8): 6010-6018, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32207920

RESUMEN

Tumor-derived extracellular vesicles (EVs) have emerged as a promising source of circulating biomarkers for liquid biopsies. However, understanding the heterogeneous physical and biochemical properties of EVs originating from multiple complex biogenesis pathways remains a major challenge. Here, we introduce EV-Ident for preparation of subpopulations of EVs in three different size fractions: large EVs (EV200 nm; 200-1 000 nm), medium EVs (EV100 nm; 100-200 nm), and small EVs (EV20 nm; 20-100 nm). Furthermore, this technology enables the in situ labeling of fluorescence markers for the protein profiling of individual EVs. As a proof-of-concept, we analyzed the presence of human epidermal growth factor receptor 2 (HER2) and prostate-specific membrane antigen (PSMA) in breast cancer and prostate cancer cell-derived EVs, respectively, using three different size fractions at the single-EV level. By reducing the complexity of EV heterogeneity in each size fraction, we found that HER2-positive breast cancer cells showed the greatest expression of HER2 in EV20 nm, whereas PSMA expression was the highest in EV200 nm derived from PSMA-expressing prostate cancer cells. This increase in HER2 expression in EV20 nm and PSMA expression in EV200 nm was further confirmed in plasma-derived nanoparticles (PNPs) obtained from breast and prostate cancer patients, respectively. Our study demonstrates that single-EV analysis using EV-Ident provides a practical way to understand EV heterogeneity and to successfully identify potent subpopulation of EVs for breast and prostate cancer, which has promising translational implications for cancer theranostics. Furthermore, these findings have the potential to address fundamental questions surrounding the biology and clinical applications of EVs.


Asunto(s)
Antígenos de Superficie/sangre , Biomarcadores de Tumor/sangre , Neoplasias de la Mama/sangre , Vesículas Extracelulares/química , Glutamato Carboxipeptidasa II/sangre , Neoplasias de la Próstata/sangre , Receptor ErbB-2/sangre , Neoplasias de la Mama/diagnóstico , Femenino , Humanos , Masculino , Tamaño de la Partícula , Neoplasias de la Próstata/diagnóstico , Propiedades de Superficie
3.
Analyst ; 141(2): 371-81, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26535415

RESUMEN

Extracellular vesicles (EVs) are cell-derived nanovesicles, present in almost all types of body fluids, which play an important role in intercellular communication and are involved in the transport of biological signals for regulating diverse cellular functions. Due to the increasing clinical interest in the role of EVs in tumor promotion, various techniques for their isolation, detection, and characterization are being developed. In this review, we present an overview of the current EV isolation and characterization methods in addition to their applications and limitations. Furthermore, EVs as the potential emerging biomarkers in cancer management and their clinical implementation are briefly discussed.


Asunto(s)
Fraccionamiento Celular/métodos , Vesículas Extracelulares/patología , Neoplasias/diagnóstico , Neoplasias/patología , Vesículas Extracelulares/metabolismo , Humanos , Pronóstico
4.
Theranostics ; 12(5): 1988-1998, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35265194

RESUMEN

Extracellular vesicles (EVs) carry information inherited from parental cells, having significant potential for disease diagnosis. In blood, however, EVs are outnumbered >104-fold by low density lipoproteins (LDLs), yet similar in size and density. These fundamental disadvantages often cause LDL spillover into EV isolates, thus confounding assay results. We hypothesized that EVs can be further separated from LDLs based on electric charge: EVs and LDLs have different lipid composition, which can lead to differential surface charge densities. To test this hypothesis, we modeled and quantified the surface charge of EVs and LDLs, and used the information to optimally separate EVs from LDLs via ion-exchange chromatography. Methods: We built an enhanced dual-mode chromatography (eDMC) device which performed i) size-exclusion to remove particles smaller than EVs and LDLs and ii) cation-exchange in an acidic elution to retain LDLs longer than EVs. The performance of the eDMC, in comparison to size-exclusion only, was evaluated by analyzing the yield and purity of the isolated EVs. Results: By measuring and modeling zeta potentials at different buffer pH, we estimated surface charge densities of EVs (-6.2 mC/m2) and LDLs (-3.6 mC/m2), revealing that EVs are more negatively charged than LDLs. Furthermore, the charge difference between EVs and LDLs was maximal at a weak acidic condition (pH = 6.4). By applying these findings, we optimized eDMC operation to enrich EVs directly from plasma, depleting >99.8% of LPPs within 30 min. Minimizing LDL contamination improved analytical signals in EV molecular assays, including single vesicle imaging, bulk protein measurements, and mRNA detection. Conclusions: These developments will promote the translational value of the dual-mode separation - a fast, equipment-free, and non-biased way for EV isolation from plasma samples.


Asunto(s)
Vesículas Extracelulares , Vesículas Extracelulares/metabolismo , Lipoproteínas LDL/metabolismo , Plasma/metabolismo , Proteómica , ARN Mensajero/metabolismo
5.
Adv Mater ; 34(24): e2200981, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35429065

RESUMEN

Electrochemical biosensors have shown great potential for simple, fast, and cost-effective point-of-care diagnostic tools. However, direct analysis of complex biological fluids such as plasma has been limited by the loss of sensitivity caused by biofouling. By increasing the surface area, the nanostructured electrode can improve detection sensitivity. However, like a double-edged sword, a large surface area increases the nonspecific adsorption of contaminating proteins. The use of nanoporous structures may prevent fouling proteins. However, there is no straightforward approach for creating nanostructured and nanoporous surfaces compatible with microfabricated thin-film electrodes. Herein, the preferential etching of chloride and surfactant-assisted anisotropic gold reduction to create homogeneous, nanostructured, and nanoporous gold electrodes is demonstrated, yielding a 190 ± 20 times larger surface area within a minute without using templates. This process, "surfactant-based electrochemical etch-deposit interplay for nanostructure/nanopore growth" (SEEDING), on electrodes enhances the sensitivity and antibiofouling capabilities of amperometric biosensors, enabling direct analysis of tumor-derived extracellular vesicles (tEVs) in complex biofluids with a limit of detection of 300 tEVs µL-1  from undiluted plasma and good discrimination between patients with prostate cancer from healthy ones with an area under the curve of 0.91 in urine and 0.90 in plasma samples.


Asunto(s)
Técnicas Biosensibles , Nanoporos , Biomarcadores , Técnicas Electroquímicas , Electrodos , Oro/química , Humanos , Proteínas , Tensoactivos
6.
Sci Adv ; 8(38): eabq4022, 2022 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-36129990

RESUMEN

Electrochemiluminescence (ECL) has an inherently low background and enables precise chemical reactions through electrical control. Here, we report an advanced ECL system, termed ECLipse (ECL in paired signal electrode). We physically separated ECL generation from target detection: These two processes were carried out in isolated chambers and coupled through an electrode. The strategy allowed us to minimize cross-chemical reactions, design electrodes for high ECL signals, and integrate multiple sensors in a chip. As a proof of concept, we implemented an eight-plex ECLipse and applied it to detect host factors in human plasma. ECLipse achieved higher signal-to-noise ratio than conventional ECL assays and was >7000-fold more sensitive than enzyme-linked immunosorbent assay. In a pilot clinical study, we could detect septic conditions by measuring host factors [i.e., interleukin-3 (IL-3), IL-6, and procalcitonin (PCT)]. ECLipse assay further revealed distinct IL-3 and IL-6 patterns in patients with severe acute respiratory syndrome coronavirus 2 infection.

7.
Front Bioeng Biotechnol ; 9: 669537, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34164385

RESUMEN

Extracellular vesicles (EVs) have been lauded as next-generation medicines, but very few EV-based therapeutics have progressed to clinical use. Limited clinical translation is largely due to technical barriers that hamper our ability to mass produce EVs, i.e., to isolate, purify, and characterize them effectively. Technical limitations in comprehensive characterization of EVs lead to unpredicted biological effects of EVs. Here, using a range of optical and non-optical techniques, we showed that the differences in molecular composition of EVs isolated using two isolation methods correlated with the differences in their biological function. Our results demonstrated that the isolation method determines the composition of isolated EVs at single and sub-population levels. Besides the composition, we measured for the first time the dry mass and predicted sedimentation of EVs. These parameters were likely to contribute to the biological and functional effects of EVs on single cell and cell cultures. We anticipate that our new multiscale characterization approach, which goes beyond traditional experimental methodology, will support fundamental understanding of EVs as well as elucidate the functional effects of EVs in in vitro and in vivo studies. Our findings and methodology will be pivotal for developing optimal isolation methods and establishing EVs as mainstream therapeutics and diagnostics. This innovative approach is applicable to a wide range of sectors including biopharma and biotechnology as well as to regulatory agencies.

8.
Theranostics ; 9(7): 1851-1863, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31037143

RESUMEN

Extracellular vesicles (EVs) that circulate in body fluids possess significant potential for disease diagnosis. Their use in clinical settings, however, has been limited owing to lack of simple and robust isolation methods. To rectify this problem, a centrifugal device for automatic, fast, and efficient isolation of EVs from whole-blood, called Exodisc-B is presented in this paper. Methods: The device comprises a built-in chamber to facilitate plasma separation and two nanoporous filters-one for removing larger particles and the other for enriching EVs. The performance of the device in comparison to ultracentrifugation (UC) was evaluated by analyzing the yield, purity, protein and RNA content of the isolated EVs. Additionally, the EV protein marker expressions were measured by ELISA and statistically analyzed to differentiate prostate cancer patients from healthy donors. Results: Compared with the UC technique, the proposed device is capable of isolating at least an order of magnitude higher number of EVs with about 30-fold higher mRNA count within 40 min. Sandwich ELISA of EV-specific membrane proteins-CD9-CD81-confirmed that Exodisc-B can isolate EVs from a volume of whole blood as low as 30 µL with a capture efficiency exceeding 75%. The device also facilitates temporal monitoring of tumor progression within live mouse xenograft models over a period of 13 weeks while using minimal volumes of weekly collected blood samples. Further, in ELISA analyses of multiple cancer-related proteins, such as prostate-specific antigen (PSA), prostate-specific membrane antigen (PSMA), epithelial cell adhesion molecule (EpCAM), epidermal growth factor receptor 1 (EGFR1), and heat shock protein 90 (HSP90), extracted from EVs isolated from human plasma, 43 patients were differentiated from 30 healthy donors. Conclusion: The results demonstrated the ability of Exodisc-B to provide a rapid, sensitive, and point-of-care-type method for extracting intact EVs from small volumes of clinical blood samples for disease diagnosis and monitoring.


Asunto(s)
Automatización de Laboratorios/métodos , Vesículas Extracelulares/fisiología , Neoplasias de la Próstata/diagnóstico , Animales , Biomarcadores de Tumor/metabolismo , Donantes de Sangre , Ensayo de Inmunoadsorción Enzimática/métodos , Vesículas Extracelulares/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/metabolismo , ARN Mensajero/metabolismo , Ultracentrifugación/métodos
9.
Lab Chip ; 19(1): 87-97, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30500003

RESUMEN

Androgen-receptor splice variant 7 (AR-V7) is associated with castration-resistant prostate cancer (CRPC) and resistance to anti-androgen therapy. Despite its clinical importance, the lack of efficient methods for AR-V7 analysis remains a challenge for broader use of this biomarker in routine clinical practice. Herein, we suggest a practical and non-invasive liquid biopsy method for analysis of AR-V7 in the RNA of urine-derived extracellular vesicles (EVs) without the need for blood withdrawal. Urine-derived EVs were isolated by a lab-on-a-disc integrated with six independent nanofiltration units (Exo-Hexa) allowing simultaneous processing of six individual samples. Rapid enrichment of EVs (<30 min) from each 4 mL urine sample was followed by mRNA extraction, and AR-V7 and androgen receptor full-length (AR-FL) mRNA levels in the urinary EVs were quantified by droplet digital polymerase chain reaction (ddPCR) as absolute concentrations (copies per mL). Higher AR-V7 and lower AR-FL expressions were detected in urine-derived EVs from 14 patients with CRPC than in those from 22 patients with hormone-sensitive prostate cancer. Additionally, we found that AR-V7 transcript levels and the AR-V7/AR-FL ratio in urinary EVs were higher in patients with advanced prostate cancer. This study is the first to report that RNA of urine-derived EVs is a reliable source for AR-V7 expression analysis. The proposed method for quantifying AR-V7 in urinary EVs prepared by a lab-on-a-disc is therefore a simple and promising approach to liquid biopsy with great potential for therapeutic impact on prostate cancer.


Asunto(s)
Vesículas Extracelulares , Biopsia Líquida/métodos , Neoplasias de la Próstata Resistentes a la Castración/orina , Receptores Androgénicos/análisis , Humanos , Límite de Detección , Modelos Lineales , Masculino , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mensajero/orina , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Reproducibilidad de los Resultados
10.
ACS Nano ; 11(2): 1360-1370, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28068467

RESUMEN

Extracellular vesicles (EVs) are cell-derived, nanoscale vesicles that carry nucleic acids and proteins from their cells of origin and show great potential as biomarkers for many diseases, including cancer. Efficient isolation and detection methods are prerequisites for exploiting their use in clinical settings and understanding their physiological functions. Here, we presented a rapid, label-free, and highly sensitive method for EV isolation and quantification using a lab-on-a-disc integrated with two nanofilters (Exodisc). Starting from raw biological samples, such as cell-culture supernatant (CCS) or cancer-patient urine, fully automated enrichment of EVs in the size range of 20-600 nm was achieved within 30 min using a tabletop-sized centrifugal microfluidic system. Quantitative tests using nanoparticle-tracking analysis confirmed that the Exodisc enabled >95% recovery of EVs from CCS. Additionally, analysis of mRNA retrieved from EVs revealed that the Exodisc provided >100-fold higher concentration of mRNA as compared with the gold-standard ultracentrifugation method. Furthermore, on-disc enzyme-linked immunosorbent assay using urinary EVs isolated from bladder cancer patients showed high levels of CD9 and CD81 expression, suggesting that this method may be potentially useful in clinical settings to test urinary EV-based biomarkers for cancer diagnostics.


Asunto(s)
Biomarcadores de Tumor/análisis , Vesículas Extracelulares/química , Nanofibras/química , ARN Mensajero/análisis , Neoplasias de la Vejiga Urinaria/orina , Diseño Asistido por Computadora , Ensayo de Inmunoadsorción Enzimática , Oro/química , Humanos , Tamaño de la Partícula , Propiedades de Superficie , Células Tumorales Cultivadas , Ultracentrifugación , Neoplasias de la Vejiga Urinaria/diagnóstico
11.
Lab Chip ; 16(19): 3741-9, 2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27534824

RESUMEN

A lab-on-a-disc is a unique microfluidic platform that utilizes centrifugal force to pump liquids. This offers many benefits for point-of-care devices because it eliminates the need for connections to multiple pumps and complex tubing connections. A wide range of applications including clinical chemistry, immunoassay, cell analysis, and nucleic acid tests could be demonstrated on a spinning disc. To enable the performance of assays in a fully integrated and automated manner, the robust actuation of integrated valves is a prerequisite. However, conventional passive-type valves incur a critical drawback in that their operation is dependent on the rotational frequency, which is easily influenced by the channel geometry and chemistry, in addition to the physical properties of the liquids to be transferred. Even though a few active-type valving techniques permit the individual actuation of valves, independent of the rotational frequency, complex procedures for the fabrication as well as actuation mechanisms have prevented their broader acceptance in general applications. Here, we report on a lab-on-a-disc incorporating individually addressable diaphragm valves (ID valves) that enable the reversible and thermally stable actuation of multiple valves with unprecedented ease and robustness. These ID valves are configured from an elastic epoxy diaphragm embedded on a 3D printed push-and-twist valve, which can be easily actuated by a simple automatic driver unit. As a proof of concept experiment, an enzyme-linked immunosorbent assay (ELISA) and a polymerase chain reaction (PCR) were performed on a disc in a fully automated manner to demonstrate the robust, reversible, leak-free, and thermally stable actuation of the valves.


Asunto(s)
Dispositivos Laboratorio en un Chip , Membranas Artificiales , Temperatura , Elastómeros/química , Ensayo de Inmunoadsorción Enzimática , Compuestos Epoxi/química , Humanos , Reacción en Cadena de la Polimerasa , Antígeno Prostático Específico/sangre , Staphylococcus aureus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA