reporter gene imaging
Recently Published Documents


TOTAL DOCUMENTS

93
(FIVE YEARS 14)

H-INDEX

19
(FIVE YEARS 2)

2022 ◽  
Author(s):  
Ying-Ling Chen ◽  
Kuei-Yuan Hou ◽  
Min-Ying Lin ◽  
Yu-Chuan Lin ◽  
Hui-Yen Chuang ◽  
...  

Abstract The piggyBac transposon system is known to non-viral integrate exogenous genes to chromosomes of mammalian cells. For reporter gene imaging, this transposon system is believed to efficiently establish xenograft tumor model with low immunogenicity. Because tumor cells usually exhibit genomic instability, it is important to investigate if piggyBac mediated transduction of reporter genes would change tumor characteristics. In this study, reporter gene imaging mediated by the piggyBac transposon system was exploited to track the growth and dissemination of 4T1 triple-negative murine breast cancer cells in vivo, followed by ex vivo analysis of the metastatic cells expressing reporter genes. We demonstrated that several cell properties, including proliferation rate, invasion and migration rate, and mammosphere formation ability of 4T1 cells were not influenced by piggyBac transposon system. Further, we isolated the liver metastatic cells, named 4T1-3R_L cells for further analysis. Compared to parental 4T1 cells, 4T1-3R_L cells exhibited several cancer stem cells (CSC) related characteristics, including significant mammosphere formation ability, resistance to doxorubicin, high tumorigenicity potential in Balb/C mice and expression of CD44 CSC marker. We also found that 4T1-3R_L cells exhibited stronger migrated and invasive abilities, by wound healing assay and in vitro invasion assay, respectively. The cell adhesive ability of 4T1-3R_L cells was also lower than that of 4T1 cells. The microarray assay showed that several epithelial-mesenchymal transition (EMT) promoting markers, including vimentin, N-cadherin, Twist1, and Snail were up-regulated, and anti-EMT marker E-cadherin was down-regulated in 4T1-3R_L cells. Current data suggest that the piggyBac transposon system is a reliable and biocompatible tool to engineer cancer cells for tacking and characterizing tumor development in vivo and in vitro.


2021 ◽  
Vol 14 (9) ◽  
pp. 897
Author(s):  
Sophie Stotz ◽  
Gregory D. Bowden ◽  
Jonathan M. Cotton ◽  
Bernd J. Pichler ◽  
Andreas Maurer

There is a need for versatile in vivo nuclear imaging reporter systems to foster preclinical and clinical research. We explore the applicability of the SNAPTag and novel radiolabeled small-molecule ligands as a versatile reporter gene system for in vivo nuclear imaging. SNAPTag is a high-affinity protein tag used in a variety of biochemical research areas and based on the suicide DNA repair enzyme O6-methylguanine methyl transferase (MGMT). Its ligands are well suited for reporter gene imaging as the benzyl guanine core scaffold can be derivatized with fluorescent or radiolabeled moieties for various applications. Three guanine-based SNAPTag ligands ([18F]FBBG, [18F]pFBG and [18F]mFBG) were synthesized in high yields and were (radio)chemically characterized. HEK293 cells were engineered to express the SNAPTag on the cell surface and served as cell model to assess target affinity by radiotracer uptake assays, Western blotting and SDS-PAGE autoradiography. A subcutaneous HEK293-SNAPTag xenograft model in immunodeficient mice was used for in vivo evaluation of [18F]FBBG and [18F]pFBG while the biodistribution of [18F]mFBG was characterized in naïve animals. The results were validated by ex vivo biodistribution studies and immunofluorescence staining of the xenografts. All three radiotracers were produced in high radiochemical purity, molar activity and good yields. Western blot analysis revealed successful SNAPTag expression by the transfected HEK293 cells. In vitro testing revealed high target affinity of all three tracers with an up to 191-fold higher signal in the HEK293-SNAPTag cells compared to untransfected cells. This was further supported by a prominent radioactive protein band at the expected size in the SDS-PAGE autoradiograph of cells incubated with [18F]FBBG or [18F]pFBG. The in vivo studies demonstrated high uptake in HEK293-SNAP xenografts compared to HEK293 xenografts with excellent tumor-to-muscle ratios (7.5 ± 4.2 for [18F]FBBG and 10.6 ± 6.2 for [18F]pFBG). In contrast to [18F]pFBG and its chemical analogue [18F]mFBG, [18F]FBBG showed no signs of unspecific bone uptake and defluorination in vivo. Radiolabeled SNAPTag ligands bear great potential for clinical applications such as in vivo tracking of cell populations, antibody fragments and targeted radiotherapy. With excellent target affinity, good stability, and low non-specific binding, [18F]FBBG is a highly promising candidate for further preclinical evaluation.


Author(s):  
Sophie Stotz ◽  
Gregory D. Bowden ◽  
Jonathan M. Cotton ◽  
Bernd J. Pichler ◽  
Andreas Maurer

There is a need for versatile in vivo nuclear imaging reporter systems to foster preclinical and clinical research. We explore the applicability of the SNAPTag and novel radiolabeled small-molecule ligands as a versatile reporter gene system for in vivo nuclear imaging. SNAPTag is a high-affinity protein tag used in a variety of biochemical research areas and based on the suicide DNA repair enzyme O6-methylguanine methyl transferase (MGMT). Its ligands are well suited for reporter gene imaging as the benzyl guanine core scaffold can be derivatized with fluorescent or radiolabeled moieties for various applications. Three guanine-based SNAPTag ligands ([18F]FBBG, [18F]pFBG and [18F]mFBG) were synthesized in high yields and were (radio)chemically characterized. HEK293 cells were engineered to express the SNAPTag on the cell surface and served as cell model to assess target affinity by radiotracer uptake assays, Western blotting and SDS-PAGE autoradiography. A subcutaneous HEK293-SNAPTag xenograft model in immunodeficient mice was used for in vivo evaluation of [18F]FBBG amd [18F]pFBG while the biodistribution of [18F]mFBG was characterized in naïve animals. The results were validated by ex vivo biodistribution studies and immunofluorescence staining of the xenografts. All three radiotracers were produced in high radiochemical purity, molar activity and good yields. Western blot analysis revealed successful SNAPTag expression by the transfected HEK293 cells. In vitro testing revealed high target affinity of all three tracers with an up to 191-fold higher signal in the HEK293-SNAPTag cells compared to untransfected cells. This was further supported by a prominent radioactive protein band at the expected size in the SDS-PAGE autoradiograph of cells incubated with [18F]FBBG or [18F]pFBG. The in vivo studies demonstrated high uptake in HEK293-SNAP xenografts compared to HEK293 xenografts with excellent tumor-to-muscle ratios (7.5 ± 4.2 for [18F]FBBG and 10.6 ± 6.2 for [18F]pFBG). In contrast to [18F]pFBG and its chemical analogue [18F]mFBG, [18F]FBBG showed no signs of unspecific bone uptake and defluorination in vivo. Radiolabeled SNAPTag ligands bear great potential for clinical applications such as in vivo tracking of cell populations, antibody fragments and targeted radiotherapy. With excellent target affinity, good stability, and low non-specific binding, [18F]FBBG is a highly promising candidate for further preclinical evaluation.


2021 ◽  
Vol 21 ◽  
pp. 98-109
Author(s):  
Susanna C. Concilio ◽  
Stephen J. Russell ◽  
Kah-Whye Peng

2020 ◽  
Vol Volume 13 ◽  
pp. 11659-11668
Author(s):  
Xiaofeng Li ◽  
Guotao Yin ◽  
Wei Ji ◽  
Jianjing Liu ◽  
Yufan Zhang ◽  
...  

2020 ◽  
Vol 80 (21) ◽  
pp. 4731-4740 ◽  
Author(s):  
Surya Murty ◽  
Louai Labanieh ◽  
Tara Murty ◽  
Gayatri Gowrishankar ◽  
Tom Haywood ◽  
...  

2020 ◽  
Vol 28 (6) ◽  
pp. 1392-1416 ◽  
Author(s):  
Candice Ashmore-Harris ◽  
Madeleine Iafrate ◽  
Adeel Saleem ◽  
Gilbert O. Fruhwirth

Sign in / Sign up

Export Citation Format

Share Document