Cysteine-protease activity elicited by Ca2+ stimulus in Plasmodium

https://doi.org/10.1016/j.molbiopara.2005.01.015Get rights and content

Abstract

Bloodstage malaria parasites require proteolytic activity for key processes as invasion, hemoglobin degradation and merozoite escape from red blood cells (RBCs). We investigated by confocal microscopy the presence of cysteine-protease activity elicited by calcium stimulus in Plasmodium chabaudi and Plasmodium falciparum in free trophozoites or for the later parasite within RBC using fluorescence resonance energy transfer (FRET) peptides. Peptide probes access, to either free or intraerythrocytic parasites, was also tested by selecting a range of fluorescent peptides (653–3146 Da molecular mass) labeled with Abz or FITC. In the present work we show that Ca2+ stimulus elicited by treatment with either melatonin, thapsigargin, ionomicin or nigericin, promotes an increase of substrate hydrolysis, which was blocked by the specific cysteine-protease inhibitor E-64 and the intracellular Ca2+ chelator, BAPTA. When parasites were treated with cytoplasmic Ca2+ releasing compounds, a cysteine-protease was labeled in the parasite cytoplasm by the fluorescent specific irreversible inhibitor, Ethyl-Eps-Leu-Tyr-Cap-Lys(Abz)-NH2, where Ethyl-Eps is Ethyl-(2S,3S)-oxirane-2,3-dicarboxylate. In summary, we demonstrate that P. chabaudi and P. falciparum have a cytoplasmic dependent cysteine-protease activity elicited by Ca2+.

Introduction

Malaria is one of the most important infectious diseases in the world, being responsible for more than one million deaths each year, most of them children under five (WHO, www.rbm.who.int). The cell cycle of Plasmodium within red blood cells (RBCs) is marked by its 24 h multiplicity [1], [2] according to the host circadian rhythm. The host melatonin hormone peak during the night period [3], [4] has been shown to modulate the Plasmodium cell cycle through a cytoplasmic Ca2+ increase resulting in parasite synchronization [5], [6]. Calcium homeostatic control permits a series of related processes to be readily turned on and off in response to ion mobilization from the major intracellular calcium reservoirs, the endoplasmic reticulum, mitochondria and acidic compartments [7], [8], [9]. Molecular and cellular studies on Ca2+ signaling in Plasmodium parasites [10], [11] have revealed the Ca2+ storage organelles to include the endoplasmic reticulum based in its sensitivity to Ca2+-ATPase inhibitors such as thapsigargin and ciclopiazonic acid [12], [13], [14], [15], [16], [17], [18], [19] and acidic Ca2+ pool [15], [16], [17], [20]. In addition, mitochondria are thought to play a role in Ca2+ homeostatic control in these parasites [21]. Exposure to a high extracellular Ca2+ concentration might create appropriate conditions for the use of Ca2+ as a second messenger in the modulation of parasite intracellular signaling events [22].

Downstream mechanisms related to melatonin and Ca2+ signaling might involve molecular changes such as protein activation or switches in gene expression in the parasites. Such events usually involve remodeling proteolysis [23], [24], [25], [26], a key-driving element able to activate or inactivate intermediate regulating factors, as it occurs with cyclins in the progression of cell cycle in mammals [27], [28].

Cysteine-proteases are involved in the progression of the intraerythrocytic life cycle, with roles in degradation of hemoglobin [26], [29], [30], [31] and erythrocyte cytoskeletal proteins, such as ankyrin and band 4.1 [32], and erythrocyte rupture [33]. We report in this work the presence of a cysteine-protease activity elicited by Ca2+-in malaria parasites, which was detected by hydrolysis of FRET peptides and inhibited by an irreversible fluorescent-labeled cysteine-protease inhibitor. The FRET peptides and the inhibitor Ethyl-Eps-Leu-Tyr-Cap-Lys(Abz)-NH2 (Abz-peptidyl-epoxide) were incorporated by the parasites and the hydrolytic activity or enzyme labeling by inhibitor were observed at the same time as the cytoplasmic Ca2+ release. The loading of peptides into free parasites and parasites inside RBC have been described previously [34]. Here we further investigate the permeation of peptides into the Plasmodium developmental stages (ring, trophozoite, and schizont) using fluorescent Abz- and FITC-peptides of molecular weight ranging from 653 to 3146 Da.

Section snippets

Reagents

Ionomycin, nigericin, thapsigargin (Thg), melatonin, PMSF, pepstatin A, E-64, saponin, probenecid, l-polylysine, and MOPS were purchased from Sigma-Aldrich (St. Louis, MO). BAPTA and Fluo-4 AM were from Molecular Probes, Inc. (Eugene, OR). All other reagents were analytical grade.

Plasmodium falciparum parasites

P. falciparum parasites (Palo Alto strain) were maintained in continuous in vitro culture in adult RBC [35] and synchronization was achieved by sorbitol treatment [36]. Parasitemias were determined from microscopic

Confocal microscopy measurements

The FRET peptides Abz-AIKFFARQ-EDDnp or Abz-KLRSSKQ-EDDnp with donor/receptor pair Abz/EDDnp have low fluorescence emission at 420 nm after excitation at 320 nm. The hydrolysis at any peptide bond within the spacer peptide results in an increase of fluorescence. The peptide sequence AIKFFAR was chosen to reveal aspartyl, cysteine or metaloprotease activities that preferentially cleave at Phe–Phe or at Phe–Ala bonds [39], [40], and the sequence KLRSSK [41] to reveal cysteine and serine proteases,

Discussion

This work provides functional evidence for the existence in Plasmodium of a cysteine-protease activity triggered by an increase of Ca2+ in the cytoplasm of the parasites. The peptide sequences AIKFFAR and KLRSSK used as FRET substrates are also susceptible to hydrolysis by serine-, metalo-, and aspartyl-proteases, which can participate in the continuous background hydrolysis, observed in confocal microscopy and spectrofluorimetric measurements within free cells of P. chabaudi. However, only

Acknowledgements

This work was funded by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) to C.R.S.G. (02/06194-7) and L.J. (03/09994-7). S.L.F. received fellowship from CAPES and M.L.G. from FAPESP. C.R.S.G. and L.J. are research fellows of Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq). C.R.S.G. is a John Simon Memorial Guggenheim Fellow.

References (53)

  • M.E. Wickham et al.

    Selective inhibition of a two-step egress of malaria parasites from the host erythrocyte

    J Biol Chem

    (2003)
  • M. Bogyo et al.

    Selective targeting of lysosomal cysteine proteases with radiolabeled electrophilic substrate analogs

    Chem Biol

    (2000)
  • D.C. Pimenta et al.

    Substrate specificity of human cathepsin D using internally quenched fluorescent peptides derived from reactive site loop of kallistatin

    Biochim Biophys Acta

    (2001)
  • K. Becker et al.

    Of malaria, metabolism and membrane transport

    Trends Parasitol

    (2004)
  • K. Kirk

    Channels and transporters as drug targets in the Plasmodium-infected erythrocyte

    Acta Trop

    (2004)
  • J.V. Cohn et al.

    Extracellular lysines on the plasmodial surface anion channel involved in Na+ exclusion

    Mol Biochem Parasitol

    (2003)
  • C. Braun-Breton et al.

    Activation of a Plasmodium falciparum protease correlated with merozoite maturation and erythrocyte invasion

    Biol Cell

    (1988)
  • F. Hawking et al.

    The asexual and sexual circadian rhythms of Plasmodium vinckei chabaudi, of P. berghei and of P. gallinaceum

    Parasitology

    (1972)
  • C.R. Garcia et al.

    Tertian and quartan fevers: temporal regulation in malarial infection

    J Biol Rhythms

    (2001)
  • P. Pevet

    Melatonin: from seasonal to circadian signal

    J Neuroendocrinol

    (2003)
  • C. Hotta et al.

    Calcium dependent modulation by melatonin of the circadian rhythm in malarial parasites

    Nat Cell Biol

    (2000)
  • C.T. Hotta et al.

    Melatonin and N-acetyl-serotonin cross the red blood cell membrane and evoke calcium mobilization in malarial parasites

    Braz J Med Biol Res

    (2003)
  • T. Pozzan et al.

    Molecular and cellular physiology of intracellular calcium stores

    Physiol Rev

    (1994)
  • H.T. Ma et al.

    Science

    (2000)
  • M.J. Berridge et al.

    Calcium signalling: dynamics, homeostasis and remodelling

    Nat Rev Mol Cell Biol

    (2003)
  • K. Murakami et al.

    Structure of a Plasmodium yoelii gene-encoded protein homologous to the Ca2+-ATPase of rabbit skeletal muscle sarcoplasmic reticulum

    J Cell Sci

    (1990)
  • Cited by (39)

    • Identification and structure–activity relationship (SAR) studies of carvacrol derivatives as potential anti-malarial against Plasmodium falciparum falcipain-2 protease

      2020, Bioorganic Chemistry
      Citation Excerpt :

      Among the probable targets for drugs leads falcipain-2 (PfFP-2), known cysteine proteases hydrolyze Hb to provide amino acids for parasite protein synthesis. It is important to find inhibitors of falcipain-2 with suitable properties for novel antimalarial drugs [57–60]. Recombinant PfFP-2 was prepared as described earlier [36].

    • Inhibition of malaria parasite Plasmodium falciparum development by crotamine, a cell penetrating peptide from the snake venom

      2016, Peptides
      Citation Excerpt :

      The acidic compartments, in addition to the endoplasmic reticulum and mitochondria, present in Plasmodium cells, possess an important role in the intracellular ionic homeostasis (i.e. Ca2+ and H+) [9,16,17]. They are also the key elements to provide, during the Plasmodium development, the necessary environment for the functions of vital enzymes [18]. For instance, the hemoglobin degradation, which is dependent of the action of different proteases as falcipains and plasmepsinas, occurs in acidic compartments, and therefore, the ion homeostasis is undoubtedly crucial [8,19,20].

    View all citing articles on Scopus
    View full text