Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
ACS Sens ; 8(3): 1085-1100, 2023 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-36853001

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a deadly malignancy that is often detected at an advanced stage. Earlier diagnosis of PDAC is key to reducing mortality. Circulating biomarkers such as microRNAs are gaining interest, but existing technologies require large sample volumes, amplification steps, extensive biofluid processing, lack sensitivity, and are low-throughput. Here, we present an advanced nanoplasmonic sensor for the highly sensitive, amplification-free detection and quantification of microRNAs (microRNA-10b, microRNA-let7a) from unprocessed plasma microsamples. The sensor construct utilizes uniquely designed -ssDNA receptors attached to gold triangular nanoprisms, which display unique localized surface plasmon resonance (LSPR) properties, in a multiwell plate format. The formation of -ssDNA/microRNA duplex controls the nanostructure-biomolecule interfacial electronic interactions to promote the charge transfer/exciton delocalization processes and enhance the LSPR responses to achieve attomolar (10-18 M) limit of detection (LOD) in human plasma. This improve LOD allows the fabrication of a high-throughput assay in a 384-well plate format. The performance of nanoplasmonic sensors for microRNA detection was further assessed by comparing with the qRT-PCR assay of 15 PDAC patient plasma samples that shows a positive correlation between these two assays with the Pearson correlation coefficient value >0.86. Evaluation of >170 clinical samples reveals that oncogenic microRNA-10b and tumor suppressor microRNA-let7a levels can individually differentiate PDAC from chronic pancreatitis and normal controls with >94% sensitivity and >94% specificity at a 95% confidence interval (CI). Furthermore, combining both oncogenic and tumor suppressor microRNA levels significantly improves differentiation of PDAC stages I and II versus III and IV with >91% and 87% sensitivity and specificity, respectively, in comparison to the sensitivity and specificity values for individual microRNAs. Moreover, we show that the level of microRNAs varies substantially in pre- and post-surgery PDAC patients (n = 75). Taken together, this ultrasensitive nanoplasmonic sensor with excellent sensitivity and specificity is capable of assaying multiple biomarkers simultaneously and may facilitate early detection of PDAC to improve patient care.


Asunto(s)
MicroARN Circulante , MicroARNs , Neoplasias Pancreáticas , Humanos , MicroARN Circulante/genética , Biomarcadores de Tumor/genética , MicroARNs/genética , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas
2.
Biomolecules ; 12(12)2022 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-36551284

RESUMEN

Our understanding of how osteocytes, the principal mechanosensors within bone, sense and perceive force remains unclear. Previous work identified "tethering elements" (TEs) spanning the pericellular space of osteocytes and transmitting mechanical information into biochemical signals. While we identified the heparan sulfate proteoglycan perlecan (PLN) as a component of these TEs, PLN must attach to the cell surface to induce biochemical responses. As voltage-sensitive calcium channels (VSCCs) are critical for bone mechanotransduction, we hypothesized that PLN binds the extracellular α2δ1 subunit of VSCCs to couple the bone matrix to the osteocyte membrane. Here, we showed co-localization of PLN and α2δ1 along osteocyte dendritic processes. Additionally, we quantified the molecular interactions between α2δ1 and PLN domains and demonstrated for the first time that α2δ1 strongly associates with PLN via its domain III. Furthermore, α2δ1 is the binding site for the commonly used pain drug, gabapentin (GBP), which is associated with adverse skeletal effects when used chronically. We found that GBP disrupts PLN::α2δ1 binding in vitro, and GBP treatment in vivo results in impaired bone mechanosensation. Our work identified a novel mechanosensory complex within osteocytes composed of PLN and α2δ1, necessary for bone force transmission and sensitive to the drug GBP.


Asunto(s)
Proteoglicanos de Heparán Sulfato , Mecanotransducción Celular , Proteoglicanos de Heparán Sulfato/metabolismo , Gabapentina/farmacología , Proteínas de la Matriz Extracelular/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo
3.
ACS Appl Mater Interfaces ; 14(23): 26517-26527, 2022 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-35639080

RESUMEN

Monitoring the human immune response by assaying (detection and quantification) the antibody level against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is important in conducting epidemiological surveillance and immunization studies at a population level. Herein, we present the design and fabrication of a solid-state nanoplasmonic biosensing platform that is capable of quantifying SARS-CoV-2 neutralizing antibody IgG with a limit of detection as low as 30.0 attomolar (aM) and a wide dynamic range spanning seven orders of magnitude. Based on IgG binding constant determination for different biological motifs, we show that the covalent attachment of highly specific SARS-CoV-2 linear epitopes with an appropriate ratio, in contrast to using SARS-CoV-2 spike protein subunits as receptor molecules, to gold triangular nanoprisms (Au TNPs) results in a construction of a highly selective and more sensitive, label-free IgG biosensor. The biosensing platform displays specificity against other human antibodies and no cross reactivity against MERS-CoV antibodies. Furthermore, the nanoplasmonic biosensing platform can be assembled in a multi-well plate format to translate to a high-throughput assay that allowed us to conduct SARS-CoV-2 IgG assays of COVID-19 positive patient (n = 121) and healthy individual (n = 65) plasma samples. Most importantly, performing a blind test in an additional cohort of 30 patient plasma samples, our nanoplasmonic biosensing platform successfully identified COVID-19 positive samples with 90% specificity and 100% sensitivity. Very recent studies show that our selected epitopes are conserved in the highly mutated SARS-CoV-2 variant "Omicron"; therefore, the demonstrated high-throughput nanoplasmonic biosensing platform holds great promise for a highly specific serological assay for conducting large-scale COVID-19 testing and epidemiological studies and monitoring the immune response and durability of immunity as part of the global immunization programs.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/diagnóstico , Prueba de COVID-19 , Anticuerpos Antivirales , Inmunoglobulina G
4.
ACS Nano ; 15(12): 19600-19612, 2021 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-34786933

RESUMEN

As interest continues to grow in Ti3C2Tx and other related MXenes, advancement in methods of manipulation of their surface functional groups beyond synthesis-based surface terminations (Tx: -F, -OH, and ═O) can provide mechanisms to enhance solution processability as well as produce improved solid-state device architectures and coatings. Here, we report a chemically important surface modification approach in which "solvent-like" polymers, polyethylene glycol carboxylic acid (PEG6-COOH), are covalently attached onto MXenes via esterification chemistry. Surface modification of Ti3C2Tx with PEG6-COOH with large ligand loading (up to 14% by mass) greatly enhances dispersibility in a wide range of nonpolar organic solvents (e.g., 2.88 mg/mL in chloroform) without oxidation of Ti3C2Tx two-dimensional flakes or changes in the structure ordering. Furthermore, cooperative interactions between polymer chains improve the nanoscale assembly of uniform microstructures of stacked MXene-PEG6 flakes into ordered thin films with excellent electrical conductivity (∼16,200 S·cm-1). Most importantly, our covalent surface modification approach with ω-functionalized PEG6 ligands (ω-PEG6-COOH, where ω: -NH2, -N3, -CH═CH2) allows for control over the degree of functionalization (incorporation of valency) of MXene. We believe that installing valency onto MXenes through short, ion conducting PEG ligands without compromising MXenes' features such as solution processability, structural stability, and electrical conductivity further enhance MXenes surface chemistry tunability and performance and widens their applications.

5.
Anal Chem ; 93(41): 13935-13944, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34606247

RESUMEN

Modulating optoelectronic properties of inorganic nanostructures tethered with light-responsive molecular switches by their conformational change in the solid state is fundamentally important for advanced nanoscale-device fabrication, specifically in biosensing applications. Herein, we present an entirely new solid-state design approach employing the light-induced reversible conformational change of spiropyran (SP)-merocyanine (MC) covalently attached to gold triangular nanoprisms (Au TNPs) via alkylthiolate self-assembled monolayers to produce a large localized surface plasmon resonance response (∼24 nm). This shift is consistent with the increase in thickness of the local dielectric shell-surrounded TNPs and perhaps short-range dipole-dipole (permanent and induced) interactions between TNPs and the zwitterionic MC form. Water contact angle measurement and Raman spectroscopy characterization unequivocally prove the formation of a stable TNP-MC structural motif. Utilizing this form, we fabricated the first adaptable nanoplasmonic biosensor, which uses an identical structural motif for ultrasensitive, highly specific, and programmable detection of microRNAs and proteins at attomolar concentrations in standard human plasma and urine samples, and at femtomolar concentrations from bladder cancer patient plasma (n = 10) and urine (n = 10), respectively. Most importantly, the TNP-MC structural motif displays a strong binding affinity with receptor molecules (i.e., single-stranded DNA and antibody) producing a highly stable biosensor. Taken together, the TNP-MC structural motif represents a multifunctional super biosensor with the potential to expand clinical diagnostics through simplifying biosensor design and providing highly accurate disease diagnosis.


Asunto(s)
Técnicas Biosensibles , Nanopartículas del Metal , MicroARNs , Oro , Humanos , Resonancia por Plasmón de Superficie
6.
Anal Chem ; 93(25): 8754-8763, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34125535

RESUMEN

To tackle the COVID-19 outbreak, which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there is an unmet need for highly accurate diagnostic tests at all stages of infection with rapid results and high specificity. Here, we present a label-free nanoplasmonic biosensor-based, multiplex screening test for COVID-19 that can quantitatively detect 10 different biomarkers (6 viral nucleic acid genes, 2 spike protein subunits, and 2 antibodies) with a limit of detection in the aM range, all within one biosensor platform. Our newly developed nanoplasmonic biosensors demonstrate high specificity, which is of the upmost importance to avoid false responses. As a proof of concept, we show that our detection approach has the potential to quantify both IgG and IgM antibodies directly from COVID-19-positive patient plasma samples in a single instrument run, demonstrating the high-throughput capability of our detection approach. Most importantly, our assay provides receiving operating characteristics, areas under the curve of 0.997 and 0.999 for IgG and IgM, respectively. The calculated p-value determined through the Mann-Whitney nonparametric test is <0.0001 for both antibodies when the test of COVID-19-positive patients (n = 80) is compared with that of healthy individuals (n = 72). Additionally, the screening test provides a calculated sensitivity (true positive rate) of 100% (80/80), a specificity (true negative rate) >96% (77/80), a positive predictive value of 98% at 5% prevalence, and a negative predictive value of 100% at 5% prevalence. We believe that our very sensitive, multiplex, high-throughput testing approach has potential applications in COVID-19 diagnostics, particularly in determining virus progression and infection severity for clinicians for an appropriate treatment, and will also prove to be a very effective diagnostic test when applied to diseases beyond the COVID-19 pandemic.


Asunto(s)
Técnicas Biosensibles , COVID-19 , Anticuerpos Antivirales , Humanos , Inmunoglobulina G , Inmunoglobulina M , Pandemias , ARN , SARS-CoV-2 , Sensibilidad y Especificidad , Glicoproteína de la Espiga del Coronavirus
7.
Anal Chem ; 93(4): 2578-2588, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33432809

RESUMEN

Surface-enhanced Raman scattering (SERS) is an ultrasensitive analytical technique, which is capable of providing high specificity; thus, it can be used for toxicological drug assay (detection and quantification). However, SERS-based drug analysis directly in human biofluids requires mitigation of fouling and nonspecificity effects that commonly appeared from unwanted adsorption of endogenous biomolecules present in biofluids (e.g., blood plasma and serum) onto the SERS substrate. Here, we report a bottom-up fabrication strategy to prepare ultrasensitive SERS substrates, first, by functionalizing chemically synthesized gold triangular nanoprisms (Au TNPs) with poly(ethylene glycol)-thiolate in the solid state to avoid protein fouling and second, by generating flexible plasmonic patches to enhance SERS sensitivity via the formation of high-intensity electromagnetic hot spots. Poly(ethylene glycol)-thiolate-functionalized Au TNPs in the form of flexible plasmonic patches show a twofold-improved signal-to-noise ratio in comparison to triethylamine (TEA)-passivated Au TNPs. Furthermore, the plasmonic patch displays a SERS enhancement factor of 4.5 ×107. Utilizing the Langmuir adsorption model, we determine the adsorption constant of drugs for two different surface ligands and observe that the drug molecules display stronger affinity for poly(ethylene glycol) ligands than TEA. Our density functional theory calculations unequivocally support the interaction between drug molecules and poly(ethylene glycol) moieties. Furthermore, the universality of the plasmonic patch for SERS-based drug detection is demonstrated for cocaine, JWH-018, and opioids (fentanyl, despropionyl fentanyl, and heroin) and binary mixture (trace amount of fentanyl in heroin) analyses. We demonstrate the applicability of flexible plasmonic patches for the selective assay of fentanyl at picogram/milliliter concentration levels from drug-of-abuse patients' blood plasma. The fentanyl concentration calculated in the patients' blood plasma from SERS analysis is in excellent agreement with the values determined using the paper spray ionization mass spectrometry technique. We believe that the flexible plasmonic patch fabrication strategy would be widely applicable to any plasmonic nanostructure for SERS-based chemical sensing for clinical toxicology and therapeutic drug monitoring.


Asunto(s)
Espectrometría Raman/métodos , Cocaína/química , Fentanilo/química , Toxicología Forense/métodos , Oro/química , Heroína/química , Humanos , Indoles/química , Límite de Detección , Espectrometría de Masas , Nanopartículas del Metal/química , Naftalenos/química , Preparaciones Farmacéuticas , Plasma
8.
Analyst ; 145(23): 7662-7672, 2020 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-32969415

RESUMEN

Herein we report the programmable preparation of ultrasensitive surface-enhanced Raman scattering (SERS)-based nanoplasmonic superlattice substrates to assay fentanyl and cocaine (detection and quantification) from 10 µL aliquots of emergency department patient plasma without the need for purification steps. Highly homogeneous three-dimensional (3D) nanoplasmonic superlattices are generated through the droplet evaporation-based self-assembly process of chemically-synthesized, polyethylene glycol thiolate-coated gold triangular nanoprisms (Au TNPs). Close-packed, solid-state 3D superlattice substrates produce electromagnetic hot spots due to near-field plasmonic coupling of Au TNPs, which display unique localized surface plasmonic resonance properties. These uniquely prepared superlattice substrates enable strong SERS enhancement to achieve a parts-per-quadrillion limit of detection using the label-free SERS-based technique. Our reported limit of detection is at least 100-fold better than any known SERS substrates for the drug assay. Importantly, our density functional theory calculations show that a specific electronic interaction between the drug molecule and novel nanoplasmonic superlattice substrates plays a critical role that may trigger achieving this unprecedentedly high sensitivity. Additionally, we show high selectivity of the superlattice substrate in the SERS-based detection of analytes from different patient samples, which do and do not contain target analytes (i.e., fentanyl and/or cocaine). The demonstrated sensitivity and selectivity of 3D superlattice substrates for SERS-based drug analysis in real toxicological samples are expected to advance the field of measurement science, and forensic and clinical toxicology by obviating the need for complicated sample processing steps, long assay times, and the low sensitivity of existing "gold standard" analytical techniques including gas chromatography/mass spectrometry, liquid chromatography/mass spectrometry and enzyme-linked immunosorbent assays. Taken together, we believe that this entirely new and reproducible superlattice substrate for the SERS analysis will aid scientific, forensic, and healthcare communities to battle the drug overdose epidemic in the United States.


Asunto(s)
Nanopartículas del Metal , Preparaciones Farmacéuticas , Fenómenos Electromagnéticos , Servicio de Urgencia en Hospital , Humanos , Límite de Detección , Espectrometría Raman
9.
Analyst ; 145(12): 4173-4180, 2020 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-32490854

RESUMEN

Studies have shown that microRNAs, which are small noncoding RNAs, hold tremendous promise as next-generation circulating biomarkers for early cancer detection via liquid biopsies. A novel, solid-state nanoplasmonic sensor capable of assaying circulating microRNAs through a combined surface-enhanced Raman scattering (SERS) and plasmon-enhanced fluorescence (PEF) approach has been developed. Here, the unique localized surface plasmon resonance properties of chemically-synthesized gold triangular nanoprisms (Au TNPs) are utilized to create large SERS and PEF enhancements. With careful modification to the surface of Au TNPs, this sensing approach is capable of quantifying circulating microRNAs at femtogram/microliter concentrations. Uniquely, the multimodal analytical methods mitigate both false positive and false negative responses and demonstrate the high stability of our sensors within bodily fluids. As a proof of concept, microRNA-10b and microRNA-96 were directly assayed from the plasma of six bladder cancer patients. Results show potential for a highly specific liquid biopsy method that could be used in point-of-care clinical diagnostics to increase early cancer detection or any other diseases including SARS-CoV-2 in which RNAs can be used as biomarkers.


Asunto(s)
MicroARN Circulante/sangre , Colorantes Fluorescentes/química , Espectrometría Raman , Neoplasias de la Vejiga Urinaria/diagnóstico , Betacoronavirus/aislamiento & purificación , Biomarcadores de Tumor/sangre , COVID-19 , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Oro/química , Humanos , Límite de Detección , Microscopía Confocal , Nanoestructuras/química , Pandemias , Neumonía Viral/diagnóstico , Neumonía Viral/patología , Neumonía Viral/virología , Sistemas de Atención de Punto , SARS-CoV-2 , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología
10.
Anal Chem ; 92(13): 9295-9304, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32469524

RESUMEN

There is an unmet need in clinical point-of-care (POC) cancer diagnostics for early state disease detection, which would greatly increase patient survival rates. Currently available analytical techniques for early stage cancer diagnosis do not meet the requirements for POC of a clinical setting. They are unable to provide the high demand of multiplexing, high-throughput, and ultrasensitive detection of biomarkers directly from low volume patient samples ("liquid biopsy"). To overcome these current technological bottle-necks, herein we present, for the first time, a bottom-up fabrication strategy to develop plasmonic nanoantenna-based sensors that utilize the unique localized surface plasmon resonance (LSPR) properties of chemically synthesized gold nanostructures, gold triangular nanoprisms (Au TNPs), gold nanorods (Au NRs), and gold spherical nanoparticles (Au SNPs). Our Au TNPs, NRs, and SNPs display refractive index unit (RIU) sensitivities of 318, 225, and 135 nm/RIU respectively. Based on the RIU results, we developed plasmonic nanoantenna-based multiplexing and high-throughput biosensors for the ultrasensitive assay of microRNAs. MicroRNAs are directly linked with cancer development, progression, and metastasis, thus they hold promise as next generation biomarkers for cancer diagnosis and prognosis. The developed biosensors are capable of assaying five different types of microRNAs at an attomolar detection limit. These sets of microRNAs include both oncogenic and tumor suppressor microRNAs. To demonstrate the efficiency as a POC cancer diagnostic tool, we analyzed the plasma of 20-bladder cancer patients without any sample processing steps. Importantly, our liquid biopsy-based biosensing approach is capable of differentiating healthy from early ("non-metastatic") and late ("metastatic") stage cancer with a p value <0.0001. Further, receiver operating characteristic analysis shows that our biosensing approach is highly specific, with an area under the curve of 1.0. Additionally, our plasmonic nanoantenna-based biosensors are regenerative, allowing multiple measurements using the same biosensors, which is essential in low- and middle-income countries. Taken together, our multiplexing and high-throughput biosensors have the unmatched potential to advance POC diagnostics and meet global needs for early stage detection of cancer and other diseases (e.g., infectious, autoimmune, and neurogenerative diseases).


Asunto(s)
Técnicas Biosensibles/métodos , MicroARNs/sangre , Nanoestructuras/química , Neoplasias de la Vejiga Urinaria/diagnóstico , Oro/química , Humanos , Nanopartículas del Metal/química , Nanotubos/química , Sistemas de Atención de Punto , Pronóstico , Resonancia por Plasmón de Superficie
11.
Anal Chem ; 91(3): 1894-1903, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30608133

RESUMEN

It is becoming understood that microRNAs hold great promise for noninvasive liquid biopsies for screening for different types of cancer, but current state-of-the-art RT-PCR and microarray techniques have sensitivity limitations that currently restrict their use. Herein, we report a new transduction mechanism involving delocalization of photoexcited conduction electrons wave function of gold triangular nanoprism (Au TNP) in the presence of -ssDNA/microRNA duplexes. This plasmoelectronic effect increases the electronic dimension of Au TNPs and substantially affects their localized surface plasmon resonance (LSPR) properties that together allow us to achieve a sensitivity for microRNA assay as low as 140 zeptomolar concentrations for our nanoplasmonic sensors. We show that the position of a single base-pair mismatch in the -ssDNA/microRNA duplex dramatically alters the LSPR properties and detection sensitivity. The unprecedentedly high sensitivity of nanoplasmonic sensors has allowed us to assay four different microRNAs (microRNA-10b, -182, -143, and -145) from bladder cancer patient plasma (50 µL/sample). For the first time, we demonstrate the utility of a label-free, nanoplasmonic sensor in quantification of tumor suppressor microRNAs, the level of tumor suppressor microRNAs goes down in a cancer patient as compared to normal healthy individuals, in metastatic and nonmetastatic bladder cancer patient plasma. Our statistical analysis of patient samples unequivocally suggests that the tumor suppressor microRNAs are more specific biomarkers ( p-value of <0.0001) than oncogenic microRNAs for differentiation between metastatic and nonmetastatic bladder cancer, and nonmetastatic cancer from healthy individuals. This work demonstrating the electron wave functions delocalization dependent ultrasensitive LSPR properties of noble metal nanoparticles has a great potential for fabrication of miniaturized and extremely powerful sensors to investigate microRNA properties in other cancers (for example breast, lung, and pancreatic) through liquid biopsy.


Asunto(s)
Biomarcadores de Tumor/sangre , Técnicas Biosensibles , Neoplasias de la Mama/diagnóstico , Técnicas Electroquímicas , Neoplasias Pulmonares/diagnóstico , MicroARNs/sangre , Neoplasias Pancreáticas/diagnóstico , Neoplasias de la Mama/sangre , ADN de Cadena Simple/sangre , ADN de Cadena Simple/química , Oro/química , Humanos , Neoplasias Pulmonares/sangre , Nanopartículas del Metal/química , MicroARNs/química , Neoplasias Pancreáticas/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA