Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
ACS Sens ; 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836608

ABSTRACT

Immune checkpoint inhibitors (ICIs) targeting programmed cell death ligand 1 (PD-L1), or its receptor, PD-1 have improved survival in patients with non-small-cell lung cancer (NSCLC). Assessment of PD-L1 expression requires tissue biopsy or fine needle aspiration that are currently used to identify patients most likely to respond to single agent anti-PD-1/PD-L1 therapy. However, obtaining sufficient tissue to generate a PD-L1 tissue proportion score (TPS) ≥ 50% using immunohistochemistry remains a challenge that potentially may be overcome by liquid biopsies. This study utilized a mesoporous gold sensor (MGS) assay to examine the phosphorylation status of PD-L1 in plasma extracellular vesicles (EV pPD-L1) and PD-L1 levels in plasma from NSCLC patient samples and their association with tumor PD-L1 TPS. The 3-dimensional mesoporous network of the electrodes provides a large surface area, high signal-to-noise ratio, and a superior electro-conductive framework, thereby significantly improving the detection sensitivity of PD-L1 nanosensing. Test (n = 20) (Pearson's r = 0.99) and validation (n = 45) (Pearson's r = 0.99) cohorts show that EV pPD-L1 status correlates linearly with the tumor PD-L1 TPS assessed by immunohistochemistry irrespective of the tumor stage, with 64% of patients overall showing detectable EV pPD-L1 levels in plasma. In contrast to the EV pPD-L1 results, plasma PD-L1 levels did not correlate with the tumor PD-L1 TPS score or EV pPD-L1 levels. These data demonstrate that EV pPD-L1 levels may be used to select patients for appropriate PD-1 and PD-L1 ICI therapy regimens in early, locally advanced, and advanced NSCLC and should be tested further in randomized controlled trials. Most importantly, the assay used has a less than 24h turnaround time, facilitating adoption of the test into the routine diagnostic evaluation of patients prior to therapy.

2.
Anal Chem ; 96(19): 7651-7660, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38690989

ABSTRACT

Development of molecular diagnostics for lung cancer stratification and monitoring is crucial for the rational planning and timely adjustment of treatments to improve clinical outcomes. In this regard, we propose a nanocavity architecture to sensitively profile the protein signature on small extracellular vesicles (sEVs) to enable accurate, noninvasive staging and treatment monitoring of lung cancer. The nanocavity architecture is formed by molecular recognition through the binding of sEVs with the nanobox-based core-shell surface-enhanced Raman scattering (SERS) barcodes and mirrorlike, asymmetric gold microelectrodes. By imposing an alternating current on the gold microelectrodes, a nanofluidic shear force was stimulated that supported the binding of sEVs and the efficient assembly of the nanoboxes. The binding of sEVs further induced a nanocavity between the nanobox and the gold microelectrode that significantly amplified the electromagnetic field to enable the simultaneous enhancement of Raman signals from four SERS barcodes and generate patient-specific molecular sEV signatures. Importantly, evaluated on a cohort of clinical samples (n = 76) on the nanocavity architecture, the acquired patient-specific sEV molecular signatures achieved accurate identification, stratification, and treatment monitoring of lung cancer patients, highlighting its potential for transition to clinical utility.


Subject(s)
Extracellular Vesicles , Gold , Lung Neoplasms , Spectrum Analysis, Raman , Extracellular Vesicles/chemistry , Extracellular Vesicles/metabolism , Lung Neoplasms/metabolism , Humans , Gold/chemistry , Microelectrodes
3.
Anal Chem ; 96(11): 4495-4504, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38445954

ABSTRACT

The molecular detection of multiple respiratory viruses provides evidence for the rational use of drugs and effective health management. Herein, we developed and tested the clinical performance of an electrohydrodynamic-driven nanobox-on-mirror platform (E-NoM) for the parallel, accurate, and sensitive detection of four respiratory viral antigens. The E-NoM platform uses gold-silver alloy nanoboxes as the core material with the deposition of a silver layer as a shell on the core surfaces to amplify and enable a reproducible Raman signal readout that facilitates accurate detection. Additionally, the E-NoM platform employs gold microelectrode arrays as the mirror with electrohydrodynamics to manipulate the fluid flow and enhance molecular interactions for an improved biosensing response. The presence of viral antigens binds the nanobox-based core-shell nanostructure on the gold microelectrode and creates the nanocavity with extremely strong "hot spots" to benefit sensitive analysis. Significantly, in a large clinical cohort with 227 patients, the designed E-NoM platform demonstrates the capability of screening respiratory infection with achieved clinical specificity, sensitivity, and accuracy of 100.0, 96.48, and 96.91%, respectively. It is anticipated that the E-NoM platform can find a position in clinical usage for respiratory disease diagnosis.


Subject(s)
Biosensing Techniques , Metal Nanoparticles , Viruses , Humans , Metal Nanoparticles/chemistry , Silver/chemistry , Gold/chemistry , Antigens, Viral , Spectrum Analysis, Raman
4.
Proteomics ; 24(11): e2300094, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38343172

ABSTRACT

Microglia are a specialized population of innate immune cells located in the central nervous system. In response to physiological and pathological changes in their microenvironment, microglia can polarize into pro-inflammatory or anti-inflammatory phenotypes. A dysregulation in the pro-/anti-inflammatory balance is associated with many pathophysiological changes in the brain and nervous system. Therefore, the balance between microglia pro-/anti-inflammatory polarization can be a potential biomarker for the various brain pathologies. A non-invasive method of detecting microglia polarization in patients would have promising clinical applications. Here, we perform proteomic analysis of small extracellular vesicles (sEVs) derived from microglia cells to identify sEVs biomarkers indicative of pro-inflammatory and anti-inflammatory phenotypic changes. sEVs were isolated from microglia cell lines under different inflammatory conditions and analyzed by proteomics by liquid chromatography with mass spectrometry. Our findings provide the potential roles of sEVs that could be related to the pathogenesis of various brain diseases.


Subject(s)
Extracellular Vesicles , Microglia , Proteomics , Microglia/metabolism , Humans , Extracellular Vesicles/metabolism , Proteomics/methods , Cell Line , Proteome/analysis , Proteome/metabolism , Biomarkers/metabolism , Biomarkers/analysis , Inflammation/metabolism
5.
Methods ; 223: 146-147, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38354836
6.
Nanoscale Horiz ; 8(9): 1288-1289, 2023 Aug 21.
Article in English | MEDLINE | ID: mdl-37449356

ABSTRACT

Correction for 'Plasma extracellular vesicle phenotyping for the differentiation of early-stage lung cancer and benign lung diseases' by Liwen Yuan et al., Nanoscale Horiz., 2023, 8, 746-758, https://doi.org/10.1039/d2nh00570k.

7.
Methods Mol Biol ; 2679: 241-253, 2023.
Article in English | MEDLINE | ID: mdl-37300621

ABSTRACT

Small extracellular vesicles (sEVs) are nanoscopic bioparticles that transport biomolecular cargoes between cells. sEVs have been implicated in many pathological processes such as cancer, rendering them as promising targets for therapeutics and diagnostics. Characterizing phenotypic differences in sEV biomolecular cargos could support understanding their roles in cancer. However, this is difficult due to similar physical properties of sEVs and requirement for highly sensitive analysis. Our method describes the preparation and operation of a microfluidic immunoassay with surface-enhanced Raman scattering (SERS) readouts, termed sEV subpopulation characterization platform (ESCP). ESCP applies an alternating current induced electrohydrodynamic flow to enhance collisions of sEVs with the antibody-functionalized sensor surface. Captured sEVs are labeled with plasmonic nanoparticles to facilitate multiplexed and highly sensitive phenotypic characterization of sEVs by SERS. ESCP is demonstrated for characterizing the expression of three tetraspanins (CD9, CD63, CD81) and four cancer-associated biomarkers (MCSP, MCAM, ErbB3, LNGFR) in sEVs derived from cancer cell lines and plasma samples.


Subject(s)
Extracellular Vesicles , Neoplasms , Humans , Microfluidics , Neoplasms/genetics , Neoplasms/metabolism , Extracellular Vesicles/metabolism , Biomarkers, Tumor/metabolism , Electricity
8.
Nat Nanotechnol ; 18(10): 1222-1229, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37291255

ABSTRACT

Accurate and early detection of biomarkers provides the molecular evidence for disease management, allowing prompt actions and timely treatments to save lives. Multivalent biomolecular interactions between the probe and biomarker as well as controlled probe orientation on material surfaces are keys for highly sensitive detection. Here we report the bioengineering of programmable and multifunctional nanoprobes, which can provide rapid, specific and highly sensitive detection of emerging diseases in a range of widely used diagnostic systems. These nanoprobes composed of nanosized cell wall fragments, termed as synthetic bionanofragments (SynBioNFs), are generated by the fragmentation of genetically programmed yeast cells. SynBioNFs display multiple copies of biomolecules for high-affinity target binding and molecular handles for the precisely orientated attachment on surfaces used in diagnostic platforms. SynBioNFs are demonstrated for the capture and detection of SARS-CoV-2 virions using multiple diagnostic platforms, including surface-enhanced Raman scattering, fluorescence, electrochemical and colorimetric-based lateral flow systems with sensitivity comparable with the gold-standard reverse-transcription quantitative polymerase chain reaction.


Subject(s)
SARS-CoV-2 , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Indicators and Reagents , SARS-CoV-2/genetics
9.
Anal Chem ; 95(22): 8522-8532, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37224231

ABSTRACT

Phosphorylation is a post-translational modification in proteins that changes protein conformation and activity for regulating signal transduction pathways. This mechanism is frequently impaired in lung cancer, resulting in permanently active constitutive phosphorylation to initiate tumor growth and/or reactivate pathways in response to therapy. We developed a multiplexed phosphoprotein analyzer chip (MPAC) that enables rapid (detection time: 5 min) and sensitive (LOD: 2 pg/µL) detection of protein phosphorylation and presents phosphoproteomic profiling of major phosphorylation pathways in lung cancer. We monitored phosphorylated receptors and downstream proteins involved in mitogen-activated protein kinase (MAPK) and PI3K/AKT/mTOR pathways in lung cancer cell line models and patient-derived extracellular vesicles (EV). Using kinase inhibitor drugs in cell line models, we found that the drug can inhibit the phosphorylation and/or activation of the kinase pathway. We then generated a phosphorylation heatmap by EV phosphoproteomic profiling of plasma samples isolated from 36 lung cancer patients and 8 noncancer individuals. The heatmap showed a clear difference between the noncancer and cancer samples and identify the specific proteins that are activated in the cancer samples. Our data also showed that MPAC could monitor immunotherapy responses by assessment of the phosphorylation states of the proteins, particularly for PD-L1. Finally, with a longitudinal study, we found that the phosphorylation levels of the proteins were indicative of a positive response to therapy. We believe that this study will lead to personalized treatment by providing a better understanding of the active and resistant pathways and will provide a tool for selecting combined and targeted therapies for precision medicine.


Subject(s)
Lung Neoplasms , Phosphatidylinositol 3-Kinases , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/therapeutic use , Longitudinal Studies , Signal Transduction , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Cell Line, Tumor
10.
ACS Sens ; 8(4): 1648-1657, 2023 04 28.
Article in English | MEDLINE | ID: mdl-37026968

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 variants play an important role in predicting patient outcome during postinfection, and with growing fears of COVID-19 reservoirs in domestic and wild animals, it is necessary to adapt detection systems for variant detection. However, variant-specific detection remains challenging. Surface-enhanced Raman scattering is a sensitive and multiplexing technique that allows the simultaneous detection of multiple targets for accurate identification. Here we propose the development of a multiplex SERS microassay to detect both the spike and nucleocapsid structural proteins of SARS-CoV-2. The designed SERS microassay integrates gold-silver hollow nanobox barcodes and electrohydrodynamically induced nanomixing which in combination enables highly specific and sensitive detection of SARS-CoV-2 and the S-protein epitopes to delineate between ancestral prevariant strains with the newer variants of concern, Delta and Omicron. The microassay allows detection from as low as 20 virus/µL and 50 pg/mL RBD protein and can clearly identify the virus among infected versus healthy nasopharyngeal swabs, with the potential to identify between variants. The detection of both S- and N-proteins of SARS-CoV-2 and the differentiation of variants on the SERS microassay can aid the early detection of COVID-19 to reduce transmission rates and lead into adequate treatments for those severely affected by the virus.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , COVID-19/diagnosis , Epitopes , Gold , Nucleocapsid Proteins
11.
Nanoscale Horiz ; 8(6): 746-758, 2023 05 30.
Article in English | MEDLINE | ID: mdl-36974989

ABSTRACT

The development of a minimally invasive technique for early-stage lung cancer detection is crucial to reducing mortality. Phenotyping of tumor-associated extracellular vesicles (EVs) has the potential for early-stage lung cancer detection, yet remains challenging due to the lack of sensitive, integrated techniques that can accurately detect rare tumor-associated EV populations in blood. Here, we integrated gold core-silver shell nanoparticles and nanoscopic mixing in a microfluidic assay for sensitive phenotypic analysis of EVs directly in plasma without EV pre-isolation. The assay enabled multiplex detection of lung cancer-associated markers PTX3 and THBS1 and canonical EV marker CD63 by surface-enhanced Raman spectroscopy, providing a squared correlation coefficient of 0.97 in the range of 103-107 EVs mL-1 and a limit of detection of 19 EVs mL-1. Significantly, our machine learning-based nanostrategy provided 92.3% sensitivity and 100% specificity in differentiating early-stage lung cancer from benign lung diseases, superior to the CT scan-based lung cancer diagnosis (92.3% sensitivity and 71.4% specificity). Overall, our integrated nanostrategy achieved an AUC value of 0.978 in differentiating between early-stage lung cancer patients (n = 28) and controls consisting of patients with benign lung diseases (n = 23) and healthy controls (n = 26), which showed remarkable diagnostic performance and great clinical potential for detecting the early occurrence of lung cancer.


Subject(s)
Extracellular Vesicles , Lung Neoplasms , Humans , Biomarkers, Tumor , Lung Neoplasms/diagnosis , Extracellular Vesicles/chemistry , Plasma , Early Detection of Cancer/methods
12.
Semin Cancer Biol ; 88: 157-171, 2023 01.
Article in English | MEDLINE | ID: mdl-36581020

ABSTRACT

Extracellular vesicles (EVs) are nano-sized particles that hold tremendous potential in the clinical space, as their biomolecular profiles hold a key to non-invasive liquid biopsy for cancer diagnosis and prognosis. EVs are present in most bodily fluids, hence are easily obtainable from patients, advantageous to that of traditional, invasive tissue biopsies and imaging techniques. However, there are certain constraints that hinder clinical use of EVs. The translation of EV biomarkers from "bench-to-bedside" is encumbered by the methods of EV isolation and subsequent biomarker detection currently implemented in laboratories. Although current isolation and detection methods are effective, they lack practicality, with their requirement for high bodily fluid volumes, low equipment availability, slow turnaround times and high costs. The high demand for techniques that overcome these limitations has resulted in significant advancements in nanotechnological devices. These devices are designed to integrate EV isolation and biomarker detection into a one-step method of direct EV detection from bodily fluids. This provides promise for the acceleration of EVs into current clinical standards. This review highlights the importance of EVs as cancer biomarkers, the methodological obstacles currently faced in clinical studies and how novel nanodevices could advance clinical translation.


Subject(s)
Extracellular Vesicles , Humans , Biomarkers, Tumor , Liquid Biopsy/methods , Nanotechnology
13.
Adv Sci (Weinh) ; : e2204207, 2022 Nov 17.
Article in English | MEDLINE | ID: mdl-36394090

ABSTRACT

Accurate identification of malignant lung lesions is a prerequisite for rational clinical management to reduce morbidity and mortality of lung cancer. However, classification of lung nodules into malignant and benign cases is difficult as they show similar features in computer tomography and sometimes positron emission tomography imaging, making invasive tissue biopsies necessary. To address the challenges in evaluating indeterminate nodules, the authors investigate the molecular profiles of small extracellular vesicles (sEVs) in differentiating malignant and benign lung nodules via a liquid biopsy-based approach. Aiming to characterize phenotypes between malignant and benign groups, they develop a single-molecule-resolution-digital-sEV-counting-detection (DECODE) chip that interrogates three lung-cancer-associated sEV biomarkers and a generic sEV biomarker to create sEV molecular profiles. DECODE capturessEVs on a nanostructured pillar chip, confines individual sEVs, and profiles sEV biomarker expression through surface-enhanced Raman scattering barcodes. The author utilize DECODE to generate a digitally acquired sEV molecular profiles in a cohort of 33 people, including patients with malignant and benign lung nodules, and healthy individuals. Significantly, DECODE reveals sEV-specific molecular profiles that allow the separation of malignant from benign (area under the curve, AUC = 0.85), which is promising for non-invasive characterisation of lung nodules found in lung cancer screening and warrants further clinincal validaiton with larger cohorts.

14.
Anal Chem ; 94(42): 14573-14582, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36222247

ABSTRACT

Immune checkpoint blockade (ICB) therapy has achieved remarkable success in many cancers including melanoma. However, ICB therapy benefits only a small proportion of patients and produces severe side effects for some patients. Thus, there is an urgent need to identify patients who are more likely to respond to ICB therapy to improve outcomes and minimize side effects. To predict ICB therapy responses, we design a surface-enhanced Raman scattering (SERS) assay for multiplex profiling of circulating tumor cells (CTCs) under basal and interferon-γ (IFN-γ) stimulation. Through simultaneous ensemble and single-cell measurements of CTCs, the SERS assay can reveal tumor heterogeneity and offer a comprehensive CTC phenotype for decision-making. Anisotropic gold-silver alloy nanoboxes are utilized as SERS plasmonic substrates for improved signal readouts of CTC surface biomarkers. By generating a unique CTC signature with four surface biomarkers, the developed assay enables the differentiation of CTCs from three different patient-derived melanoma cell lines. Significantly, in a cohort of 14 melanoma patients who received programmed cell death-1 blockade therapy, the changes of CTC signature induced by IFN-γ stimulation to CTCs show the potential to predict responders. We expect that the SERS assay can help select patients for receiving ICB therapy in other cancers.


Subject(s)
Melanoma , Neoplastic Cells, Circulating , Humans , Immune Checkpoint Inhibitors , Silver , Interferon-gamma , Melanoma/drug therapy , Melanoma/pathology , Gold , Biomarkers , Alloys
15.
Anal Chem ; 94(41): 14177-14184, 2022 Oct 18.
Article in English | MEDLINE | ID: mdl-36194728

ABSTRACT

Dengue disease is an emerging global threat triggered by dengue virus (DENV) transmission, primarily by the mosquito Aedes aegypti. The accurate surveillance and sensitive detection of DENV in mosquito populations are critical for the protection of human populations worldwide that are in the habitat of these mosquito species. There are four DENV serotypes with DENV2 reported to cause the most severe complications. There are limited ultrasensitive methods to early detect DENV2 mosquito infection and prevent human infection. Herein, we report an innovative nanobased immunoassay platform for early, specific, and ultrasensitive detection of DENV2-secreted nonstructural 1 (NS1) protein biomarker in single infected mosquitoes with the limit of detection of 500 fg of recombinant DENV2 NS1. The high sensitivity and DENV2 serotype specificity of the platform are the result of using nanomixing, plasmonic SERS nanoboxes, and yeast affinity bionanofragments displaying single-chain variable fragments (nanoyeast scFvs). Nanoyeast scFvs used for high affinity capture of DENV2 NS1 provided an innovative and cost-efficient alternative to monoclonal antibodies and differentiated DENV2 NS1 from other DENV serotypes and Zika virus NS1. The platform used electrohydrodynamically driven nanomixing to enhance NS1 capture by the nanoyeast scFvs while reducing nonspecific interactions. High sensitivity detection of captured DENV2 NS1 was achieved using NS1-specific surface-enhanced Raman scattering (SERS) nanotags. These nanotechnologies provide a significant innovation for early DENV2 detection in single infected mosquitoes, improving the accurate surveillance of mosquito habitats and preventing infection and severe disease arising from DENV2 transmission.


Subject(s)
Aedes , Dengue Virus , Dengue , Single-Chain Antibodies , Zika Virus Infection , Zika Virus , Animals , Antibodies, Monoclonal , Dengue/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Humans , Saccharomyces cerevisiae , Viral Nonstructural Proteins
16.
Front Immunol ; 13: 842023, 2022.
Article in English | MEDLINE | ID: mdl-35345676

ABSTRACT

The early complement components have emerged as mediators of pro-oncogenic inflammation, classically inferred to cause terminal complement activation, but there are limited data on the activity of terminal complement in cancer. We previously reported elevated serum and tissue C9, the terminal complement component, in esophageal adenocarcinoma (EAC) compared to the precursor condition Barrett's Esophagus (BE) and healthy controls. Here, we investigate the level and cellular fates of the terminal complement complex C5b-9, also known as the membrane attack complex. Punctate C5b-9 staining and diffuse C9 staining was detected in BE and EAC by multiplex immunohistofluorescence without corresponding increase of C9 mRNA transcript. Increased C9 and C5b-9 staining were observed in the sequence normal squamous epithelium, BE, low- and high-grade dysplasia, EAC. C5b-9 positive esophageal cells were morphologically intact, indicative of sublytic or complement-evasion mechanisms. To investigate this at a cellular level, we exposed non-dysplastic BE (BAR-T and CP-A), high-grade dysplastic BE (CP-B and CP-D) and EAC (FLO-1 and OE-33) cell lines to the same sublytic dose of immunopurified human C9 (3 µg/ml) in the presence of C9-depleted human serum. Cellular C5b-9 was visualized by immunofluorescence confocal microscopy. Shed C5b-9 in the form of extracellular vesicles (EV) was measured in collected conditioned medium using recently described microfluidic immunoassay with capture by a mixture of three tetraspanin antibodies (CD9/CD63/CD81) and detection by surface-enhanced Raman scattering (SERS) after EV labelling with C5b-9 or C9 antibody conjugated SERS nanotags. Following C9 exposure, all examined cell lines formed C5b-9, internalized C5b-9, and shed C5b-9+ and C9+ EVs, albeit at varying levels despite receiving the same C9 dose. In conclusion, these results confirm increased esophageal C5b-9 formation during EAC development and demonstrate capability and heterogeneity in C5b-9 formation and shedding in BE and EAC cell lines following sublytic C9 exposure. Future work may explore the molecular mechanisms and pathogenic implications of the shed C5b-9+ EV.


Subject(s)
Adenocarcinoma , Barrett Esophagus , Extracellular Vesicles , Complement Activation , Complement C9/metabolism , Complement Membrane Attack Complex , Complement System Proteins/metabolism , Esophageal Neoplasms , Extracellular Vesicles/metabolism , Humans
17.
Anal Chem ; 93(50): 16787-16795, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34889595

ABSTRACT

Epithelial to mesenchymal transition (EMT) results in the genesis of circulating tumor cells (CTCs) from tumor sites and promotes the metastatic capability of CTCs in circulation. In this study, we develop a multiplex surface-enhanced Raman scattering nanotechnology for comprehensive characterization of EMT-associated phenotypes in CTCs, to monitor cancer metastasis. We observe the downregulation of the CTC marker (EpCAM) and the epithelial marker (E-cadherin), as well as the upregulation of a mesenchymal marker (N-cadherin) and a stem cell marker (ABCB5) during the transforming growth factor-ß-induced EMT process in breast cancer cell line models. Additionally, we also find changes in the heterogeneity levels of these selected markers in cells. With this method, we successfully detect the presence of disease in samples from breast cancer patients and characterize EMT-associated phenotypes in their CTCs. Overall, this approach and findings provide a new means for monitoring the EMT process in cancer, insights into the detailed mechanistic progress of the diseases, and have potential for detecting the early occurrence of cancer metastasis.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms , Humans
18.
ACS Biomater Sci Eng ; 7(12): 5850-5860, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34738789

ABSTRACT

Bioengineered yeast bio-nanomaterials termed nanoyeasts displaying antibody single-chain variable fragments (scFvs) against diagnostic targets are a promising alternative to monoclonal antibodies (mAbs). A potential limitation for translating nanoyeasts into diagnostic tools is batch-to-batch variability. Herein, we demonstrate a systematic approach for cost-efficient production of highly specific nanoyeasts that enabled accurate dengue virus (DENV) detection by immunoassay (2.5% CV). Yeasts bioengineered to surface express DENV-specific scFvs (up to 66% of the total cell population) were fragmented into nanoyeast fractions trialing sonication, bead beating, and high-pressure disruption methods. Nanoyeast fractions from sonication had optimal target binding, uniform particle size (±89 nm), were stable, and retained diagnostic activity for 7 days at 37 °C compared to traditional mAbs that lost activity after 1 day at 37 °C. We engineered a panel of nanoyeast scFvs targeting DENV nonstructural protein 1 (NS1): (i) specific for serotyping DENV 1-4 and (ii) cross-reactive anti-DENV scFvs that are suitable for "yes/no" diagnostic applications. We demonstrate highly specific nanoyeast scFvs for serotyping DENV. We show that nanoyeast scFvs specifically detect NS1 in simulated patient plasma with a limit of detection of 250 ng/mL, the concentration found in infected patients.


Subject(s)
Dengue Virus , Dengue , Single-Chain Antibodies , Antibodies, Viral , Biocompatible Materials , Dengue/diagnosis , Dengue Virus/genetics , Humans , Single-Chain Antibodies/genetics , Viral Nonstructural Proteins
19.
J Chromatogr A ; 1657: 462553, 2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34564029

ABSTRACT

We describe the chromatographic and electrochromatographic separation of small neutral and charged analytes using a fused silica capillary with a stationary pseudophase semi-permanent coating of didodecyldimethyl ammonium bromide (DDAB) aggregates. The coating was prepared by flushing the capillary with a DDAB solution that was rinsed out with the mobile phase. Our studies (i.e., electroosmotic flow measurements by capillary electrophoresis, chromatographic retention of a neutral probe and atomic force microscopy) suggested the formation of DDAB patchy admicelle, complete admicelle, or larger aggregates at the solid surface - liquid interface inside the capillary, depending on the concentration of DDAB used in coating the capillary. The analytical figures of merit for open tubular liquid chromatography (OT-LC, pressure driven) and open tubular capillary electrochromatography (OT-CEC, voltage driven) using a capillary coated with 0.5 mM DDAB and mobile phase/background solution of 25 mM borate buffer at pH 9.5 with 10% MeOH were the following: LOD = 3.0-5.0 µg/mL (OT-LC) and 2.5-5.0 µg/mL (OT-CEC); linearity R2 > 0.99 (peak area (OT-LC) and corrected peak area (OT-CEC)), intraday and interday repeatability%RSD < 5% (n = 12) for retention/migration time, peak area (OT-LC) and corrected peak area (OT-CEC). The reversed-phase and anion-exchange property of the stationary pseudophase was studied by the addition of organic solvents and sodium chloride to the mobile phase, respectively. We also demonstrate the increase in the ks of the tested analytes by implementing successive multiple ionic layer (SMIL) coating strategies with DDAB in combination with a cationic and/or anionic polyelectrolyte. The use of a stationary pseudophase coating is potentially an easy alternative way to conduct open-tubular liquid chromatography and electrochromatography.


Subject(s)
Capillary Electrochromatography , Chromatography, Liquid , Electroosmosis , Silicon Dioxide
20.
ACS Sens ; 6(9): 3182-3194, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34264628

ABSTRACT

Identifying small extracellular vesicle (sEV) subpopulations based on their different molecular signatures could potentially reveal the functional roles in physiology and pathology. However, it is a challenge to achieve this aim due to the nano-sized dimensions of sEVs, low quantities of biological cargo each sEV carries, and our incomplete knowledge of identifying features capable of separating heterogeneous sEV subpopulations. Here, a sensitive, multiplexed, and nano-mixing-enhanced sEV subpopulation characterization platform (ESCP) is proposed to precisely determine the sEV phenotypic heterogeneity and understand the role of sEV heterogeneity in cancer progression and metastasis. The ESCP utilizes spatially patterned anti-tetraspanin-functionalized micro-arrays for sEV subpopulation sorting and nanobarcode-based surface-enhanced Raman spectroscopy for multiplexed read-outs. An ESCP has been used for investigating sEV phenotypic heterogeneity in terms of canonical sEV tetraspanin molecules and cancer-associated protein biomarkers in both cancer cell line models and cancer patient samples. Our data explicitly demonstrate the selective enrichment of tetraspanins and cancer-associated protein biomarkers, in particular sEV subpopulations. Therefore, it is believed that the ESCP could enable the evaluation and broader application of sEV subpopulations as potential diagnostic disease biomarkers.


Subject(s)
Extracellular Vesicles , Neoplasms , Humans , Neoplasms/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...