Standard

AuNP Aptasensor for Hodgkin Lymphoma Monitoring. / Slyusarenko, Maria; Shalaev, Sergey; Valitova, Alina; Zabegina, Lidia; Nikiforova, Nadezhda; Nazarova, Inga; Rudakovskaya, Polina; Vorobiev, Maxim; Lezov, Alexey; Filatova, Larisa; Yevlampieva, Natalia; Gorin, Dmitry; Krzhivitsky, Pavel; Malek, Anastasia.

в: Biosensors, Том 12, № 1, 23, 04.01.2022.

Результаты исследований: Научные публикации в периодических изданияхстатьяРецензирование

Harvard

Slyusarenko, M, Shalaev, S, Valitova, A, Zabegina, L, Nikiforova, N, Nazarova, I, Rudakovskaya, P, Vorobiev, M, Lezov, A, Filatova, L, Yevlampieva, N, Gorin, D, Krzhivitsky, P & Malek, A 2022, 'AuNP Aptasensor for Hodgkin Lymphoma Monitoring', Biosensors, Том. 12, № 1, 23. https://doi.org/10.3390/bios12010023, https://doi.org/10.3390/bios12010023

APA

Slyusarenko, M., Shalaev, S., Valitova, A., Zabegina, L., Nikiforova, N., Nazarova, I., Rudakovskaya, P., Vorobiev, M., Lezov, A., Filatova, L., Yevlampieva, N., Gorin, D., Krzhivitsky, P., & Malek, A. (2022). AuNP Aptasensor for Hodgkin Lymphoma Monitoring. Biosensors, 12(1), [23]. https://doi.org/10.3390/bios12010023, https://doi.org/10.3390/bios12010023

Vancouver

Slyusarenko M, Shalaev S, Valitova A, Zabegina L, Nikiforova N, Nazarova I и пр. AuNP Aptasensor for Hodgkin Lymphoma Monitoring. Biosensors. 2022 Янв. 4;12(1). 23. https://doi.org/10.3390/bios12010023, https://doi.org/10.3390/bios12010023

Author

Slyusarenko, Maria ; Shalaev, Sergey ; Valitova, Alina ; Zabegina, Lidia ; Nikiforova, Nadezhda ; Nazarova, Inga ; Rudakovskaya, Polina ; Vorobiev, Maxim ; Lezov, Alexey ; Filatova, Larisa ; Yevlampieva, Natalia ; Gorin, Dmitry ; Krzhivitsky, Pavel ; Malek, Anastasia. / AuNP Aptasensor for Hodgkin Lymphoma Monitoring. в: Biosensors. 2022 ; Том 12, № 1.

BibTeX

@article{18051d46aa3244679dcf6b280b47c5e2,
title = "AuNP Aptasensor for Hodgkin Lymphoma Monitoring",
abstract = "A liquid biopsy based on circulating small extracellular vesicles (SEVs) has not yet been used in routine clinical practice due to the lack of reliable analytic technologies. Recent studies have demonstrated the great diagnostic potential of nanozyme-based systems for the detection of SEV markers. Here, we hypothesize that CD30-positive Hodgkin and Reed–Sternberg (HRS) cells secrete CD30 + SEVs; therefore, the relative amount of circulating CD30 + SEVs might reflect classical forms of Hodgkin lymphoma (cHL) activity and can be measured by using a nanozyme-based technique. A AuNP aptasensor analytics system was created using aurum nanoparticles (AuNPs) with peroxidase activity. Sensing was mediated by competing properties of DNA aptamers to attach onto surface of AuNPs inhibiting their enzymatic activity and to bind specific markers on SEVs surface. An enzymatic activity of AuNPs was evaluated through the color reaction. The study included characterization of the components of the analytic system and its functionality using transmission and scanning electron microscopy, nanoparticle tracking analysis (NTA), dynamic light scattering (DLS), and spectrophotometry. AuNP aptasensor analytics were optimized to quantify plasma CD30 + SEVs. The developed method allowed us to differentiate healthy donors and cHL patients. The results of the CD30 + SEV quantification in the plasma of cHL patients were compared with the results of disease activity assessment by positron emission tomography/computed tomography (PET-CT) scanning, revealing a strong positive correlation. Moreover, two cycles of chemotherapy resulted in a statistically significant decrease in CD30 + SEVs in the plasma of cHL patients. The proposed AuNP aptasensor system presents a promising new approach for monitoring cHL patients and can be modified for the diagnostic testing of other diseases.",
keywords = "Aptamer, Diagnostic, Enzyme-mimetic nanoparticles, Gold nanoparticles, Hodgkin lymphoma, Liquid biopsy, Monitoring, Small extracellular vesicles, gold nanoparticles, monitoring, enzyme-mimetic nanoparticles, ELECTROCHEMICAL DETECTION, EXTRACELLULAR VESICLES, aptamer, small extracellular vesicles, diagnostic, liquid biopsy, PEROXIDASE-LIKE ACTIVITY",
author = "Maria Slyusarenko and Sergey Shalaev and Alina Valitova and Lidia Zabegina and Nadezhda Nikiforova and Inga Nazarova and Polina Rudakovskaya and Maxim Vorobiev and Alexey Lezov and Larisa Filatova and Natalia Yevlampieva and Dmitry Gorin and Pavel Krzhivitsky and Anastasia Malek",
note = "Funding Information: Funding: This research was supported by the Ministry of Health of the Russian Federation (project no. AAAA-A18-118032890186-5). Publisher Copyright: {\textcopyright} 2022 by the authors. Licensee MDPI, Basel, Switzerland.",
year = "2022",
month = jan,
day = "4",
doi = "10.3390/bios12010023",
language = "English",
volume = "12",
journal = "Biosensors",
issn = "2079-6374",
publisher = "MDPI AG",
number = "1",

}

RIS

TY - JOUR

T1 - AuNP Aptasensor for Hodgkin Lymphoma Monitoring

AU - Slyusarenko, Maria

AU - Shalaev, Sergey

AU - Valitova, Alina

AU - Zabegina, Lidia

AU - Nikiforova, Nadezhda

AU - Nazarova, Inga

AU - Rudakovskaya, Polina

AU - Vorobiev, Maxim

AU - Lezov, Alexey

AU - Filatova, Larisa

AU - Yevlampieva, Natalia

AU - Gorin, Dmitry

AU - Krzhivitsky, Pavel

AU - Malek, Anastasia

N1 - Funding Information: Funding: This research was supported by the Ministry of Health of the Russian Federation (project no. AAAA-A18-118032890186-5). Publisher Copyright: © 2022 by the authors. Licensee MDPI, Basel, Switzerland.

PY - 2022/1/4

Y1 - 2022/1/4

N2 - A liquid biopsy based on circulating small extracellular vesicles (SEVs) has not yet been used in routine clinical practice due to the lack of reliable analytic technologies. Recent studies have demonstrated the great diagnostic potential of nanozyme-based systems for the detection of SEV markers. Here, we hypothesize that CD30-positive Hodgkin and Reed–Sternberg (HRS) cells secrete CD30 + SEVs; therefore, the relative amount of circulating CD30 + SEVs might reflect classical forms of Hodgkin lymphoma (cHL) activity and can be measured by using a nanozyme-based technique. A AuNP aptasensor analytics system was created using aurum nanoparticles (AuNPs) with peroxidase activity. Sensing was mediated by competing properties of DNA aptamers to attach onto surface of AuNPs inhibiting their enzymatic activity and to bind specific markers on SEVs surface. An enzymatic activity of AuNPs was evaluated through the color reaction. The study included characterization of the components of the analytic system and its functionality using transmission and scanning electron microscopy, nanoparticle tracking analysis (NTA), dynamic light scattering (DLS), and spectrophotometry. AuNP aptasensor analytics were optimized to quantify plasma CD30 + SEVs. The developed method allowed us to differentiate healthy donors and cHL patients. The results of the CD30 + SEV quantification in the plasma of cHL patients were compared with the results of disease activity assessment by positron emission tomography/computed tomography (PET-CT) scanning, revealing a strong positive correlation. Moreover, two cycles of chemotherapy resulted in a statistically significant decrease in CD30 + SEVs in the plasma of cHL patients. The proposed AuNP aptasensor system presents a promising new approach for monitoring cHL patients and can be modified for the diagnostic testing of other diseases.

AB - A liquid biopsy based on circulating small extracellular vesicles (SEVs) has not yet been used in routine clinical practice due to the lack of reliable analytic technologies. Recent studies have demonstrated the great diagnostic potential of nanozyme-based systems for the detection of SEV markers. Here, we hypothesize that CD30-positive Hodgkin and Reed–Sternberg (HRS) cells secrete CD30 + SEVs; therefore, the relative amount of circulating CD30 + SEVs might reflect classical forms of Hodgkin lymphoma (cHL) activity and can be measured by using a nanozyme-based technique. A AuNP aptasensor analytics system was created using aurum nanoparticles (AuNPs) with peroxidase activity. Sensing was mediated by competing properties of DNA aptamers to attach onto surface of AuNPs inhibiting their enzymatic activity and to bind specific markers on SEVs surface. An enzymatic activity of AuNPs was evaluated through the color reaction. The study included characterization of the components of the analytic system and its functionality using transmission and scanning electron microscopy, nanoparticle tracking analysis (NTA), dynamic light scattering (DLS), and spectrophotometry. AuNP aptasensor analytics were optimized to quantify plasma CD30 + SEVs. The developed method allowed us to differentiate healthy donors and cHL patients. The results of the CD30 + SEV quantification in the plasma of cHL patients were compared with the results of disease activity assessment by positron emission tomography/computed tomography (PET-CT) scanning, revealing a strong positive correlation. Moreover, two cycles of chemotherapy resulted in a statistically significant decrease in CD30 + SEVs in the plasma of cHL patients. The proposed AuNP aptasensor system presents a promising new approach for monitoring cHL patients and can be modified for the diagnostic testing of other diseases.

KW - Aptamer

KW - Diagnostic

KW - Enzyme-mimetic nanoparticles

KW - Gold nanoparticles

KW - Hodgkin lymphoma

KW - Liquid biopsy

KW - Monitoring

KW - Small extracellular vesicles

KW - gold nanoparticles

KW - monitoring

KW - enzyme-mimetic nanoparticles

KW - ELECTROCHEMICAL DETECTION

KW - EXTRACELLULAR VESICLES

KW - aptamer

KW - small extracellular vesicles

KW - diagnostic

KW - liquid biopsy

KW - PEROXIDASE-LIKE ACTIVITY

UR - http://www.scopus.com/inward/record.url?scp=85123101423&partnerID=8YFLogxK

UR - https://www.mendeley.com/catalogue/64817dfc-6523-303f-ac14-7ee0638df1f1/

U2 - 10.3390/bios12010023

DO - 10.3390/bios12010023

M3 - Article

C2 - 35049651

AN - SCOPUS:85123101423

VL - 12

JO - Biosensors

JF - Biosensors

SN - 2079-6374

IS - 1

M1 - 23

ER -

ID: 93143134