Faculty Opinions recommendation of Highly sensitive in vivo imaging of Trypanosoma brucei expressing "red-shifted" luciferase.

Author(s):  
Christine Clayton
2013 ◽  
Vol 7 (11) ◽  
pp. e2571 ◽  
Author(s):  
Alex P. McLatchie ◽  
Hollie Burrell-Saward ◽  
Elmarie Myburgh ◽  
Michael D. Lewis ◽  
Theresa H. Ward ◽  
...  

Nano Research ◽  
2010 ◽  
Vol 3 (10) ◽  
pp. 722-732 ◽  
Author(s):  
Liang Cheng ◽  
Kai Yang ◽  
Shuai Zhang ◽  
Mingwang Shao ◽  
Shuittong Lee ◽  
...  

2017 ◽  
Vol 247 ◽  
pp. 461-468 ◽  
Author(s):  
Xiaodong Jin ◽  
Shifan Wang ◽  
Wenzhu Yin ◽  
Tongxiang Xu ◽  
Yang Jiang ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2602-2602
Author(s):  
Nadia Terziyska ◽  
Catarina Castro Alves ◽  
Manfred Ogris ◽  
Ernst Wagner ◽  
Renier J Brentjens ◽  
...  

Abstract Abstract 2602 Novel therapeutic approaches require preclinical in vivo testing before first use in men. An important preclinical model to study acute lymphoblastic leukemia (ALL) was first described more than 2 decades ago (Dick et al., Science 1989). In this model, primary human tumor cells from patients with ALL are transplanted into severely immuno-compromised mice. The model is hampered by the lack of a sensitive readout for disease progression and treatment effects. As ALL is a systemic disease per se, tumor mass is difficult to measure; bone marrow aspirations remain exhausting for mice; secretion of tumor cells into the blood stream remains a late and only partially reliable surrogate parameter. Here, we optimized the preclinical individualized mouse model of ALL by introducing a highly sensitive and reliable readout parameter for disease progression and treatment efficacy. We introduced the lentiviral transduction of patient-derived ALL cells to express transgenes such as luciferase and fluochromes. Transduction efficiency ranged between 0,5 and 70 %, depending on the sample used. Expression of fluochromes was used for enrichment of transgenic cells by FACs sorting. Expression of luciferase was used to perform bioluminescence in vivo imaging in mice. Bioluminescence in vivo imaging was easy to perform and well tolerated by the mice, even as often as 3 times per week. Imaging enabled highly reliable measurement of disease progression over time in single mice and revealed a strictly logarithmic growth of leukemia in mice. Imaging results correlated perfectly well with post mortem determination of leukemic burden using immunohistochemistry and FACs analysis. In contrast to the later, imaging was highly sensitive and detected leukemia at disease stages representing minimal residual disease. Depending on the expression level of the transgenes, imaging was able to detect as few as 1 human ALL cells in >> 10.000 normal mouse bone marrow cells. By following up groups of mice over time, imaging revealed especially helpful for readout of limiting dilution transplantation assays to determine frequencies of leukemia initiating cells, e.g., with and without prior in vitro treatment. In preclinical treatment trials on ALL cells from individual patients, imaging quantified therapy effects precisely and with low variances discriminating treatment failure from partial and complete response as early as 4 days after treatment. Due to follow up of mice over time, imaging visualized disease regrowth after successful therapy and allowed determining classical clinical readout parameter such as time to tumor regrowth. Taken together, the most demanding challenges in anti-cancer treatment are modeled precisely and individually in mice using bioluminescence in vivo imaging. In vivo imaging upgrades the individualized xenograft mouse model of ALL by highly sensitive and reliable disease monitoring. Imaging enables performing preclinical trials at defined disease stages and using classical clinical readout parameters. Imaging-based individualized preclinical trials of ALL will facilitate translation of novel therapeutic approaches from bench to bedside. Disclosures: No relevant conflicts of interest to declare.


Author(s):  
Sang Bong Lee ◽  
Hyun Dong Ji ◽  
In-Kyu Lee ◽  
Kil Soo Kim ◽  
Sang-Woo Lee ◽  
...  

In vivo imaging of platelets will provide a better understanding of their critical roles in arterial cardiovascular disease, hemostasis, inflammation, and cancer. Here, we demonstrate the feasibility of using radioiodine...


2020 ◽  
Vol 21 (21) ◽  
pp. 7896
Author(s):  
Jun Nakayama ◽  
Ryohei Saito ◽  
Yusuke Hayashi ◽  
Nobuo Kitada ◽  
Shota Tamaki ◽  
...  

Bioluminescence imaging (BLI) is useful to monitor cell movement and gene expression in live animals. However, D-luciferin has a short wavelength (560 nm) which is absorbed by tissues and the use of near-infrared (NIR) luciferin analogues enable high sensitivity in vivo BLI. The AkaLumine-AkaLuc BLI system (Aka-BLI) can detect resolution at the single-cell level; however, it has a clear hepatic background signal. Here, to enable the highly sensitive detection of bioluminescence from the surrounding liver tissues, we focused on seMpai (C15H16N3O2S) which has been synthesized as a luciferin analogue and has high luminescent abilities as same as AkaLumine. We demonstrated that seMpai BLI could detect micro-signals near the liver without any background signal. The solution of seMpai was neutral; therefore, seMpai imaging did not cause any adverse effect in mice. seMpai enabled a highly sensitive in vivo BLI as compared to previous techniques. Our findings suggest that the development of a novel mutated luciferase against seMpai may enable a highly sensitive BLI at the single-cell level without any background signal. Novel seMpai BLI system can be used for in vivo imaging in the fields of life sciences and medicine.


RSC Advances ◽  
2014 ◽  
Vol 4 (19) ◽  
pp. 9656-9659 ◽  
Author(s):  
Sanju Das ◽  
Surajit Biswas ◽  
Santanu Mukherjee ◽  
Jaya Bandyopadhyay ◽  
Subhodip Samanta ◽  
...  

A solid non-toxic highly sensitive cyanide fluorosensor exhibits better biocompatibility over allied in situ complexes under physiological conditions.


2014 ◽  
Vol 20 (1) ◽  
pp. 36-43 ◽  
Author(s):  
Michael D. Lewis ◽  
Amanda Fortes Francisco ◽  
Martin C. Taylor ◽  
John M. Kelly

The protozoan Trypanosoma cruzi is the causative agent of Chagas disease, one of the world’s major neglected infections. Although development of improved antiparasitic drugs is considered a priority, there have been no significant treatment advances in the past 40 years. Factors that have limited progress include an incomplete understanding of pathogenesis, tissue tropism, and disease progression. In addition, in vivo models, which allow parasite burdens to be tracked throughout the chronic stage of infection, have been lacking. To address these issues, we have developed a highly sensitive in vivo imaging system based on bioluminescent T. cruzi, which express a red-shifted luciferase that emits light in the tissue-penetrating orange-red region of the spectrum. The exquisite sensitivity of this noninvasive murine model has been exploited to monitor parasite burden in real time throughout the chronic stage, has allowed the identification of the gastrointestinal tract as the major niche of long-term infection, and has demonstrated that chagasic heart disease can develop in the absence of locally persistent parasites. Here, we review the parameters of the imaging system and describe how this experimental model can be incorporated into drug development programs as a valuable tool for assessing efficacy against both acute and chronic T. cruzi infections.


2013 ◽  
Vol 87 (24) ◽  
pp. 13321-13329 ◽  
Author(s):  
V. Tran ◽  
L. A. Moser ◽  
D. S. Poole ◽  
A. Mehle

Sign in / Sign up

Export Citation Format

Share Document