Abstract

Natural killer (NK) cells eliminate infected and tumorigenic cells through delivery of granzymes via perforin pores or by activation of caspases via death receptors. In order to understand how NK cells combine different cell death mechanisms, it is important to quantify target cell responses on a single cell level. However, currently existing reporters do not allow the measurement of several protease activities inside the same cell. Here, we present a strategy for the comparison of two different proteases at a time inside individual target cells upon engagement by NK cells. We developed single-fluorescent protein reporters containing the RIEAD or the VGPD cleavage site for the measurement of granzyme B activity. We show that these two granzyme B reporters can be applied in combination with caspase-8 or caspase-3 reporters. While we did not find that caspase-8 was activated by granzyme B, our method revealed that caspase-3 activity follows granzyme B activity with a delay of about 6 min. Finally, we illustrate the comparison of several different reporters for granzyme A, M, K, and H. The approach presented here is a valuable means for the investigation of the temporal evolution of cell death mediated by cytotoxic lymphocytes.

Highlights

  • As part of the innate immune system, natural killer (NK) cells can eliminate infected and tumorigenic cells [1]

  • We demonstrate our approach by measuring granzyme B, caspase-8, and caspase-3 activity in target cells exposed to Natural killer (NK) cells

  • We designed fluorescent reporters following the strategy that we previously established to measure caspase activity upon CD95 activation [34]. These reporters consist of one fluorescent protein fused to a localization domain through a linker that can be cleaved by proteases

Read more

Summary

Introduction

As part of the innate immune system, natural killer (NK) cells can eliminate infected and tumorigenic cells [1]. To do so, they adhere to a target cell and establish an immunological synapse [2]. The following NK cell receptor signaling can trigger the release of cytotoxic granules from the NK cell into the cleft of this synapse [3]. Granzymes are released from cytotoxic granules and enter the target cell via perforin pores and induce cell death. CD95L or TRAIL are presented at the surface of NK cells and induce extrinsic apoptosis in target cells through activation of the death receptors CD95 or TRAIL-R1/-R2 [4, 5]

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call