Summary

生物发光成像检测到非洲锥虫病的晚期感染

Published: May 18, 2016
doi:

Summary

This manuscript describes the use of a bioluminescent strain of African trypanosomes to enable the tracking of late stage infection and demonstrates how in vivo live imaging can be used to visualize infections within the central nervous system in real-time.

Abstract

Human African trypanosomiasis (HAT) is a multi-stage disease that manifests in two stages; an early blood stage and a late stage when the parasite invades the central nervous system (CNS). In vivo study of the late stage has been limited as traditional methodologies require the removal of the brain to determine the presence of the parasites.

Bioluminescence imaging is a non-invasive, highly sensitive form of optical imaging that enables the visualization of a luciferase-transfected pathogen in real-time. By using a transfected trypanosome strain that has the ability to produce late stage disease in mice we are able to study the kinetics of a CNS infection in a single animal throughout the course of infection, as well as observe the movement and dissemination of a systemic infection.

Here we describe a robust protocol to study CNS infections using a bioluminescence model of African trypanosomiasis, providing real time non-invasive observations which can be further analyzed with optional downstream approaches.

Introduction

Human African trypanosomiasis (HAT), or sleeping sickness, is caused by the vector-borne protozoan parasites of the Trypanosoma brucei spp1. Estimated numbers of current cases is fewer than 7 thousand every year with almost 70 million people exposed to the risk of the parasite infection within the African continent. The disease, which is most often lethal if left untreated, comprises an early hemolymphatic stage where parasites are present in the blood, progressing to the late stage when parasites invade the central nervous system (CNS) and are no longer susceptible to treatment by early stage trypanosomal drugs2. The current drug therapies for late-stage HAT have both complex, prolonged, treatment regimens and severe adverse effects as well as reported resistance, therefore research into new drug therapies is imperative3,4.

The study of late-stage human African trypanosomiasis (HAT) within traditional mouse models is lengthy and complex, with the removal of brain tissue being required to monitor parasitic burden5. The animal infective strain T. b. brucei is used as the study model of trypanosomiasis with the late stage appearing 21 days post infection (dpi). To monitor the wild type nonbioluminescent parasite infection in the mouse model, peripheral blood films or quantitative PCR are the only methods to determine parasite burden. For parasite burden in the brain, the mouse needs to be culled, brain excised and qPCR carried out on tissues, making it impossible to track parasites through multiple time points in the late stage infection. This results in the inability to follow real-time infections within the central nervous system (CNS).

In vivo bioluminescence imaging (BLI) can provide highly sensitive, non-invasive detection of parasite dissemination and disease progression in a mouse model that can be followed in a single animal for the entirety of the experiment6. BLI is based on the emission of light in the visible spectrum produced by a luciferase-catalyzed reaction. The emitted photons are then detected by a charge coupled device (CCD) camera7. For this purpose, the pathogen is genetically modified to express a luciferase protein and the substrate, luciferin, is introduced at time points of interest by injection. The main advantage of this method is the ability to carry out longitudinal studies, in which the same animal can be imaged several times with minimal adverse effects. The acquired bioluminescence signal can be quantified, thus indicating the pathogen burden.

The optimization and validation of a red-shifted bioluminescent T. b. brucei has enabled the investigation of the late stage infection through non-invasive procedures, detecting parasites earlier than blood film microscopy and greatly reducing the time, cost and numbers of animals needed to study CNS infection and drug screening in late-stage trypanosomiasis8,9. In this protocol we demonstrate infection of mice with bioluminescent trypanosomes and how to then visualize the parasites in vivo for quantification of disease progression and CNS penetration.

Protocol

伦理 所有工作被英国内政部动物的批准(科学程序)1986年法令和卫生与热带医学院动物福利伦理审查委员会的伦敦经济学院下进行。 ARRIVE准则得到遵守本报告。 1.在生物发光布氏锥虫布氏的体内通道删除T的冷冻股票(称为stabilate) 湾布氏应变GVR35-VSL-2(可从约翰·凯利教授在卫生和热带医学伦敦学校通过MTA)从液氮9…

Representative Results

此协议演示了如何遵循以下小鼠感染T.疾病进展湾布氏 ,为非洲人类锥虫病的模型。 图1显示了实验方案的时间表,这表明对于治疗和成像的步骤的时间表; 图2演示了用于定量外围原虫固定吉姆萨染色的血涂片的视图的典型字段,用锥虫和红血细胞存在。后面可以通过测量生物发光如在图3,其显示了与在每个时间点拍摄?…

Discussion

生物发光T的发展湾布氏 GVR35应变允许锥虫感染从早期到晚期的可视化。以前感染模型是无法检测到后期,当寄生虫在大脑中,在从血液膜显微镜实时,和所需的大脑的剔除和除去从被感染的小鼠,以确定寄生虫负荷12。生物发光降低小鼠间变异性为单个鼠标可以在整个感染的全部,并且可以在任何阶段被观察感染的快速定性评估进行跟踪。生物发光成像的高度敏感的方法使?…

Divulgations

The authors have nothing to disclose.

Acknowledgements

我们感谢约翰·凯利和马丁·泰勒(伦敦卫生与热带医学院)提供T.湾布氏 GVR35-VSL-2和Andrea Zelmer博士(LSHTM),用于体内成像的建议。这项工作是由比尔和梅林达·盖茨基金会全球健康项目(批准号OPPGH5337)的支持。

Materials

PBS Sigma, UK P4417 tablets pH 7.4
Glucose Sigma, UK G8270 99.5% (molecular) grade
Ammonium chloride Sigma, UK A9434 99.5% (molecular) grade
Heparin (lithium salt) Sigma, UK H0878
Hi-FCS Gibco, Life Technologies, UK 10500-064 500 ml
DPBS Sigma, UK D4031 Sterile filtered
Mr. Frosty Nalgene, UK
Giemsa Sigma, UK G5637
D-Luciferin Perkin Elmer, UK
Sigma, UK 115144-35-9
Diminazene aceturate Sigma, UK D7770 Analytical grade
IVIS Lumina II Perkin Elmer, UK other bioimagers available e.g. from Bruker, Kodak
Living Image v. 4.2 Perkin Elmer, UK proprietary software for Perkin Elmer IVIS instruments; other instruments may have their own
1 ml syringe Fisher Scientific, UK 10142104
20 ml syringe Fisher Scientific, UK 10743785
25G Needles Greiner Bio-one N2516
21G Needles Greiner Bio-one N2138
Twin-frosted microscope slide VWR, UK 631-0117
1.5 ml microcentrifuge tube StarLab, UK I1415-1000
7 ml Bijou tube StarLab, UK E1412-0710
Mouse restrainer Sigma, UK Z756903 our restrainer was made in-house, this is a similar model

References

  1. Brun, R., Blum, J., Chappuis, F., Burri, C. Human African trypanosomiasis. Lancet. 375 (9709), 148-159 (2010).
  2. Rodgers, J. Trypanosomiasis and the brain. Parasitology. 137 (14), 1995-2006 (2010).
  3. Barrett, M. P., Boykin, D. W., Brun, R., Tidwell, R. R. Human African trypanosomiasis: pharmacological re-engagement with a neglected disease. Br. J. Pharmacol. 152 (8), 1155-1171 (2007).
  4. Barrett, M. P., Vincent, I. M., Burchmore, R. J. S., Kazibwe, A. J. N., Matovu, E. Drug resistance in human African trypanosomiasis. Future Microbiol. 6 (9), 1037-1047 (2011).
  5. Jennings, F. W., Whitelaw, D. D., Urquhart, G. M. The relationship between duration of infection with Trypanosoma brucei in mice and the efficacy of chemotherapy. Parasitology. 75 (2), 143-153 (1977).
  6. Andreu, N., Zelmer, A., Wiles, S. Noninvasive biophotonic imaging for studies of infectious disease. FEMS Microbiol. Rev. 35 (2), 360-394 (2011).
  7. Sadikot, R. T., Blackwell, T. S. Bioluminescence imaging. Proc. Am. Thorac. Soc. 2 (6), 537-540 (2005).
  8. Burrell-Saward, H., Rodgers, J., Bradley, B., Croft, S. L., Ward, T. H. A sensitive and reproducible in vivo imaging mouse model for evaluation of drugs against late-stage human African trypanosomiasis. J. Antimicrob. Chemother. 70 (2), 510-517 (2015).
  9. McLatchie, A. P., et al. Highly sensitive in vivo imaging of Trypanosoma brucei expressing ‘red-shifted’ luciferase. PLoS Negl. Trop. Dis. 7 (11), e2571 (2013).
  10. Jennings, F. W., et al. Human African trypanosomiasis: potential therapeutic benefits of an alternative suramin and melarsoprol regimen. Parasitol. Int. 51 (4), 381-388 (2002).
  11. Wiles, S., Robertson, B. D., Frankel, G., Kerton, A. Bioluminescent monitoring of in vivo colonization and clearance dynamics by light-emitting bacteria. Methods Mol. Biol. 574, 137-153 (2009).
  12. Rodgers, J., Bradley, B., Kennedy, P. G. E. Combination chemotherapy with a substance P receptor antagonist (aprepitant) and melarsoprol in a mouse model of human African trypanosomiasis. Parasitol. Int. 56 (4), 321-324 (2007).
  13. Kamerkar, S., Davis, P. H. Toxoplasma on the brain: understanding host-pathogen interactions in chronic CNS infection. J. Parasitol. Res. 2012, 589295 (2012).
  14. Franke-Fayard, B., et al. Murine malaria parasite sequestration: CD36 is the major receptor, but cerebral pathology is unlinked to sequestration. Proc. Natl. Acad. Sci. U.S.A. 102 (32), 11468-11473 (2005).
  15. Hitziger, N., Dellacasa, I., Albiger, B., Barragan, A. Dissemination of Toxoplasma gondii to immunoprivileged organs and role of Toll/interleukin-1 receptor signalling for host resistance assessed by in vivo bioluminescence imaging. Cell. Microbiol. 7 (6), 837-848 (2005).
  16. Lewis, M. D., et al. Bioluminescence imaging of chronic Trypanosoma cruzi infections reveals tissue-specific parasite dynamics and heart disease in the absence of locally persistent infection. Cell. Microbiol. 16 (9), 1285-1300 (2014).
check_url/fr/54032?article_type=t

Play Video

Citer Cet Article
Burrell-Saward, H., Ward, T. H. Bioluminescence Imaging to Detect Late Stage Infection of African Trypanosomiasis. J. Vis. Exp. (111), e54032, doi:10.3791/54032 (2016).

View Video