Cargando…

Characterization of a highly specific NQO1-activated near-infrared fluorescent probe and its application for in vivo tumor imaging

The Near-infrared Fluorescence (NIRF) molecular imaging of cancer is known to be superior in sensitivity, deeper penetration, and low phototoxicity compared to other imaging modalities. In view of an increased need for efficient and targeted imaging agents, we synthesized a NAD(P)H quinone oxidoredu...

Descripción completa

Detalles Bibliográficos
Autores principales: Punganuru, Surendra Reddy, Madala, Hanumantha Rao, Arutla, Viswanath, Zhang, Ruiwen, Srivenugopal, Kalkunte S.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6562040/
https://www.ncbi.nlm.nih.gov/pubmed/31189950
http://dx.doi.org/10.1038/s41598-019-44111-8
_version_ 1783426222140686336
author Punganuru, Surendra Reddy
Madala, Hanumantha Rao
Arutla, Viswanath
Zhang, Ruiwen
Srivenugopal, Kalkunte S.
author_facet Punganuru, Surendra Reddy
Madala, Hanumantha Rao
Arutla, Viswanath
Zhang, Ruiwen
Srivenugopal, Kalkunte S.
author_sort Punganuru, Surendra Reddy
collection PubMed
description The Near-infrared Fluorescence (NIRF) molecular imaging of cancer is known to be superior in sensitivity, deeper penetration, and low phototoxicity compared to other imaging modalities. In view of an increased need for efficient and targeted imaging agents, we synthesized a NAD(P)H quinone oxidoreductase 1 (NQO1)-activatable NIR fluorescent probe (NIR-ASM) by conjugating dicyanoisophorone (ASM) fluorophore with the NQO1 substrate quinone propionic acid (QPA). The probe remained non-fluorescent until activation by NQO1, whose expression is largely limited to malignant tissues. With a large Stokes shift (186 nm) and a prominent near-infrared emission (646 nm) in response to NQO1, NIR-ASM was capable of monitoring NQO1 activity in vitro and in vivo with high specificity and selectivity. We successfully employed the NIR-ASM to differentiate cancer cells from normal cells based on NQO1 activity using fluorescence microscopy and flow cytometry. Chemical and genetic approaches involving the use of ES936, a specific inhibitor of NQO1 and siRNA and gene transfection procedures unambiguously demonstrated NQO1 to be the sole target activating the NIR-ASM in cell cultures. NIR-ASM was successfully used to detect and image the endogenous NQO1 in three live tumor-bearing mouse models (A549 lung cancer, Lewis lung carcinoma, and MDMAMB 231 xenografts) with a high signal-to-low noise ratiometric NIR fluorescence response. When the NQO1-proficient A549 tumors and NQO1-deficient MDA-MB-231 tumors were developed in the same animal, only the A549 malignancies activated the NIR-ASM probe with a strong signal. Because of its high sensitivity, rapid activation, tumor selectivity, and nontoxic properties, the NIR-ASM appears to be a promising agent with clinical applications.
format Online
Article
Text
id pubmed-6562040
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-65620402019-06-20 Characterization of a highly specific NQO1-activated near-infrared fluorescent probe and its application for in vivo tumor imaging Punganuru, Surendra Reddy Madala, Hanumantha Rao Arutla, Viswanath Zhang, Ruiwen Srivenugopal, Kalkunte S. Sci Rep Article The Near-infrared Fluorescence (NIRF) molecular imaging of cancer is known to be superior in sensitivity, deeper penetration, and low phototoxicity compared to other imaging modalities. In view of an increased need for efficient and targeted imaging agents, we synthesized a NAD(P)H quinone oxidoreductase 1 (NQO1)-activatable NIR fluorescent probe (NIR-ASM) by conjugating dicyanoisophorone (ASM) fluorophore with the NQO1 substrate quinone propionic acid (QPA). The probe remained non-fluorescent until activation by NQO1, whose expression is largely limited to malignant tissues. With a large Stokes shift (186 nm) and a prominent near-infrared emission (646 nm) in response to NQO1, NIR-ASM was capable of monitoring NQO1 activity in vitro and in vivo with high specificity and selectivity. We successfully employed the NIR-ASM to differentiate cancer cells from normal cells based on NQO1 activity using fluorescence microscopy and flow cytometry. Chemical and genetic approaches involving the use of ES936, a specific inhibitor of NQO1 and siRNA and gene transfection procedures unambiguously demonstrated NQO1 to be the sole target activating the NIR-ASM in cell cultures. NIR-ASM was successfully used to detect and image the endogenous NQO1 in three live tumor-bearing mouse models (A549 lung cancer, Lewis lung carcinoma, and MDMAMB 231 xenografts) with a high signal-to-low noise ratiometric NIR fluorescence response. When the NQO1-proficient A549 tumors and NQO1-deficient MDA-MB-231 tumors were developed in the same animal, only the A549 malignancies activated the NIR-ASM probe with a strong signal. Because of its high sensitivity, rapid activation, tumor selectivity, and nontoxic properties, the NIR-ASM appears to be a promising agent with clinical applications. Nature Publishing Group UK 2019-06-12 /pmc/articles/PMC6562040/ /pubmed/31189950 http://dx.doi.org/10.1038/s41598-019-44111-8 Text en © The Author(s) 2019 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
spellingShingle Article
Punganuru, Surendra Reddy
Madala, Hanumantha Rao
Arutla, Viswanath
Zhang, Ruiwen
Srivenugopal, Kalkunte S.
Characterization of a highly specific NQO1-activated near-infrared fluorescent probe and its application for in vivo tumor imaging
title Characterization of a highly specific NQO1-activated near-infrared fluorescent probe and its application for in vivo tumor imaging
title_full Characterization of a highly specific NQO1-activated near-infrared fluorescent probe and its application for in vivo tumor imaging
title_fullStr Characterization of a highly specific NQO1-activated near-infrared fluorescent probe and its application for in vivo tumor imaging
title_full_unstemmed Characterization of a highly specific NQO1-activated near-infrared fluorescent probe and its application for in vivo tumor imaging
title_short Characterization of a highly specific NQO1-activated near-infrared fluorescent probe and its application for in vivo tumor imaging
title_sort characterization of a highly specific nqo1-activated near-infrared fluorescent probe and its application for in vivo tumor imaging
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6562040/
https://www.ncbi.nlm.nih.gov/pubmed/31189950
http://dx.doi.org/10.1038/s41598-019-44111-8
work_keys_str_mv AT punganurusurendrareddy characterizationofahighlyspecificnqo1activatednearinfraredfluorescentprobeanditsapplicationforinvivotumorimaging
AT madalahanumantharao characterizationofahighlyspecificnqo1activatednearinfraredfluorescentprobeanditsapplicationforinvivotumorimaging
AT arutlaviswanath characterizationofahighlyspecificnqo1activatednearinfraredfluorescentprobeanditsapplicationforinvivotumorimaging
AT zhangruiwen characterizationofahighlyspecificnqo1activatednearinfraredfluorescentprobeanditsapplicationforinvivotumorimaging
AT srivenugopalkalkuntes characterizationofahighlyspecificnqo1activatednearinfraredfluorescentprobeanditsapplicationforinvivotumorimaging