Abstract

Blocking lactate export in the parasitic protozoan Plasmodium falciparum is a novel strategy to combat malaria. We discovered small drug-like molecules that inhibit the sole plasmodial lactate transporter, PfFNT, and kill parasites in culture. The pentafluoro-3-hydroxy-pent-2-en-1-one BH296 blocks PfFNT with nanomolar efficiency but an in vitro selected PfFNT G107S mutation confers resistance against the drug. We circumvented the mutation by introducing a nitrogen atom as a hydrogen bond acceptor site into the aromatic ring of the inhibitor yielding BH267.meta. The current PfFNT inhibitor efficiency values were derived from yeast-based lactate transport assays, yet direct affinity and binding kinetics data are missing. Here, we expressed PfFNT fused with a green fluorescent protein in human embryonic kidney cells and generated fluorescent derivatives of the inhibitors, BH296 and BH267.meta. Using confocal imaging, we confirmed the location of the proposed binding site at the cytosolic transporter entry site. We then carried out fluorescence cross-correlation spectroscopy measurements to assign true Ki-values, as well as kon and koff rate constants for inhibitor binding to PfFNT wildtype and the G107S mutant. BH296 and BH267.meta gave similar rate constants for binding to PfFNT wildtype. BH296 was inactive on PfFNT G107S, whereas BH267.meta bound the mutant protein albeit with weaker affinity than to PfFNT wildtype. Eventually, using a set of PfFNT inhibitor compounds, we found a robust correlation of the results from the biophysical FCCS binding assay to inhibition data of the functional transport assay.

Highlights

  • Asexual plasmodia draw vital energy from glycolysis [1]

  • While we found that the biophysical affinity data generally correlated well with the IC50 values obtained from functional assays in yeast, the fluorescence cross-correlation spectroscopy (FCCS) approach revealed exceptions in which a compound exhibited high binding affinity yet showed low inhibitory efficacy in the yeast, most likely due to limited transmembrane diffusion

  • As the cytoplasmic domain of the transporter is inaccessible for the compound, we explored possibilities to permeabilize the plasma membrane and to retain cell viability for the time of incubation

Read more

Summary

Introduction

The pathway relies on the swift release of the metabolic end product lactate and its accompanying proton. For this purpose, in the blood stage, the parasites express a single lactate/H+ co-transporter, PfFNT, of the exclusively microbial formate-nitrite transporter family [2]. Inhibition of PfFNT by drug-like small molecules from the MMV Malaria Box [3] (hit compounds MMV007839 and MMV000972) led to the cessation of the energy metabolism and acidification of the cytosol, killing the parasite [4,5]. Despite its channel-like, rigid structure, PfFNT acts as a secondary-active transporter in the presence of a transmembrane proton

Methods
Results
Conclusion
Full Text
Published version (Free)

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