Abstract

Severe malaria caused by Plasmodium falciparum poses a major global health problem with high morbidity and mortality. P. falciparum harbors a family of pore-forming proteins (PFPs), known as perforin like proteins (PLPs), which are structurally equivalent to prokaryotic PFPs. These PLPs are secreted from the parasites and, they contribute to disease pathogenesis by interacting with host cells. The severe malaria pathogenesis is associated with the dysfunction of various barrier cells, including endothelial cells (EC). Several factors, including PLPs secreted by parasites, contribute to the host cell dysfunction. Herein, we have tested the hypothesis that PLPs mediate dysfunction of barrier cells and might have a role in disease pathogenesis. We analyzed various dysfunctions in barrier cells following rPLP2 exposure and demonstrate that it causes an increase in intracellular Ca2+ levels. Additionally, rPLP2 exposed barrier cells displayed features of cell death, including Annexin/PI positivity, depolarized the mitochondrial membrane potential, and ROS generation. We have further performed the time-lapse video microscopy of barrier cells and found that the treatment of rPLP2 triggers their membrane blebbing. The cytoplasmic localization of HMGB1, a marker of necrosis, further confirmed the necrotic type of cell death. This study highlights the role of parasite factor PLP in endothelial dysfunction and provides a rationale for the design of adjunct therapies against severe malaria.

Highlights

  • Malaria is probably one of the oldest life-threatening diseases caused by the protozoan parasites of the genus Plasmodium

  • Our data indicated significant conservation of the membrane attack complex/perforin (MACPF) domain between Plu-MACPF and PLP2, with two transmembrane helices present on either side of the β-sheet (Figure 1A). Since both the eukaryotic and bacterial pore-forming proteins (PFPs) share a common method of pore formation, we studied and compared the activity of PLP2 with respect to epsilon toxin

  • The major functions of the blood–brain barrier (BBB) are to mainly provide a controlled entry and exit of various molecules that infiltrate the brain through the endothelial cells (EC) tight junctions and its membrane proteins [40]

Read more

Summary

Introduction

Co-localization intensity plot analysis for DAPI and HMGB1 suggested that HMGB1 was present in the nucleus in untreated cells, while, in the case of treatment, co-localization was lacking, indicating the migration of HMGB1 from the nucleus to the cytoplasm (Figure 5D) Both rEtx (positive control) and rPLP2 exhibited simila9roaf c1t6ivity and led to the necrosis of their target cells in vitro. Both rEtx (positive control) and rPLP2 exhibited similar activity and led to the necrosis of their target cells in vitro

Discussion
Cell Culture
Recombinant Protein Purification
Endotoxin Contamination Assay in Recombinant Protein
RBC Lysis Assay
Live-Cell Video Microscopy
Erythrocyte Calcium Assay
Mitochondrial Membrane Potential and ROS Detection Assays
Findings
4.10. HMGB1 Assays
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