Elsevier

Cell Calcium

Volume 94, March 2021, 102337
Cell Calcium

The role of potassium and host calcium signaling in Toxoplasma gondii egress

https://doi.org/10.1016/j.ceca.2020.102337Get rights and content

Highlights

  • ā€¢

    Toxoplasma gondii replicates inside host cells and takes up Ca2+ from the host cytosol.

  • ā€¢

    T. gondii uses extracellular Ca2+ which contributes to reach a threshold needed for egress.

  • ā€¢

    Two peaks of Ca2+ precede parasite egress. Intracellular and extracellular stores contribute.

  • ā€¢

    It is possible to patch infected cells to deliver defined Ca2+ concentrations to trigger egress.

  • ā€¢

    Reduction of the surrounding potassium concentration modulates the rate of egress.

Abstract

Toxoplasma gondii is an obligate intracellular parasite and replicates inside a parasitophorous vacuole (PV) within the host cell. The membrane of the PV (PVM) contains pores that permits for equilibration of ions and small molecules between the host cytosol and the PV lumen. Ca2+ signaling is universal and both T. gondii and its mammalian host cell utilize Ca2+ signals to stimulate diverse cellular functions. Egress of T. gondii from host cells is an essential step for the infection cycle of T. gondii, and a cytosolic Ca2+ increase initiates a Ca2+ signaling cascade that culminates in the stimulation of motility and egress. In this work we demonstrate that intracellular T. gondii tachyzoites are able to take up Ca2+ from the host cytoplasm during host cell signaling events. Both intracellular and extracellular Ca2+ sources are important in reaching a threshold of parasite cytosolic Ca2+ needed for successful egress. Two peaks of Ca2+ were observed in egressing single parasites with the second peak resulting from Ca2+ entry. We patched infected host cells to allow the delivery of precise concentrations of Ca2+ for the stimulation of motility and egress. Using this approach of patching infected host cells, allowed us to determine that increasing the host cytosolic Ca2+ to a specific concentration can trigger egress, which is further accelerated by diminishing the concentration of potassium (K+).

Introduction

Toxoplasma gondii is an obligate intracellular parasite that infects approximately one third of the world's population. T. gondii replicates inside cells and causes disease by engaging in multiple rounds of a lytic cycle, which consists of invasion of host cells, replication inside a parasitophorous vacuole (PV), egress resulting in lysis of the host cell, and invasion of a new host cell [1,2]. Several key steps of the lytic cycle of T. gondii: motility, attachment, invasion, and egress, are regulated by fluctuations in its cytosolic Ca2+ concentration ([Ca2+]c) [3,4].

Ca2+ signaling is universal and plays important roles in the regulation of a large number of cellular functions [5]. The [Ca2+]c is highly regulated, because prolonged high cytosolic Ca2+ levels are toxic and may result in cell death. A variety of Ca2+ pumps, channels, and transporters, located at the plasma membrane (PM) and intracellular organelles (endoplasmic reticulum (ER), acidic stores, and mitochondria) are involved in regulating cytosolic Ca2+ [6].

In T. gondii, the controlled influx of extracellular and intracellular Ca2+ into the parasite cytosol initiates a cascade of signaling pathways that promotes progression through the biological steps of the parasite lytic cycle. Motile parasites loaded with fluorescent Ca2+ indicators, as well as expressing Genetically Encoded Calcium Indicators (GECIs) exhibit Ca2+ oscillations [7,8]. Previous studies have shown that a rise in the cytosolic Ca2+ activates the motility machinery leading to egress. Blocking these cytosolic Ca2+ fluxes with BAPTA-AM (membrane permeable cytosolic Ca2+ chelator), blocks motility, conoid extrusion (apical tip of the parasite necessary for attachment), invasion, and host cell egress [9].

Measurements of T. gondii [Ca2+]c (using Fura2-AM) showed influx of Ca2+ from the extracellular environment, which did not operate as store-operated calcium entry (SOCE) as shown with experiments testing surrogate ions like Mn [10]. This result was supported by the lack of components of the SOCE pathway, STIM and ORAI from the T. gondii genome [11]. We showed that influx of Ca2+, as well as invasion-linked traits [10] and egress [8] were inhibited by nifedipine a Voltage Operated Calcium Chanel blocker (VOCC), supporting the function of this type of channel in Ca2+ influx.

Active egress of T. gondii from host cells requires rupture of the parasitophorous vacuole membrane (PVM) and the host cell membrane [12]. Egress is essential for the dissemination of the infection, and it has been known for several years that Ca2+ ionophores can trigger egress [13]. The final and conclusive evidence of a cytosolic Ca2+ increase preceding egress was obtained by expressing GECIs in the cytosol of T. gondii tachyzoites [8]. Secretion of the Perforin-Like Protein 1 (TgPLP1) from micronemes (specialized secretory organelles involved in egress, motility, and invasion by tachyzoites), assists in the permeabilization of the PVM and host cell membrane [14]. Both secretion of the microneme protein TgPLP1 and initiation of motility during egress are stimulated by an increase in cytosolic Ca2+. It has been proposed that the trigger for this cytosolic Ca2+ increase is the rupture of the host plasma membrane and the ensuing reduction in the concentration of the surrounding potassium [K+]. It was proposed that low [K+] would activate a phospholipase C activity in T. gondii that, in turn, would cause an increase in [Ca2+]c in the parasite via Ca2+ release from intracellular stores [15].

As an obligate intracellular parasite, T. gondii resides and replicates within the PV that functions as a molecular sieve and passively permits the exchange of small molecules; thus, the surrounding milieu of intracellular parasites is likely in equilibrium with the host cell cytoplasm [16]. Therefore, intracellular parasites would be exposed to the same fluctuations of the host cytosolic ionic composition. The host cytosolic Ca2+ is highly regulated and the resting [Ca2+]c is maintained at āˆ¼70āˆ’100 nM, which is similar to the [Ca2+]c of replicating parasites. Ca2+ efflux from intracellular stores of the parasite must be the first step of the Ca2+ signaling pathway leading to activation of egress, so maintaining these stores replenished with Ca2+ is fundamental for continuation of the lytic cycle.

In this work, we investigated how T. gondii manages to replenish its intracellular Ca2+ stores during its intracellular replication and how the decrease of the surrounding K+ concentration impacts parasite egress. Using a variety of pharmacological tools, fluorescence microscopy, and a new approach using patched infected host cells, we show that Ca2+ signaling of the host cell influences parasite cytosolic Ca2+ and contributes to parasite egress. Rupture of the host cell during egress facilitates Ca2+ influx. The ensuing decrease in the K+ concentration modulates but does not trigger egress directly.

Section snippets

Calcium influx in intracellular parasites

We previously characterized a Ca2+ influx pathway at the plasma membrane of extracellular T. gondii. Following on this finding we wanted to determine if Ca2+ influx was also operational in intracellular replicating parasites. For this, we measured cytosolic Ca2+ responses of intracellular tachyzoites and used specific natural host receptor agonists to stimulate Ca2+ signaling in the host. We expressed Genetically Encoded Ca2+ Indicators (GECIs) [17,18] in the cytosol of HeLa cells (jRGECO1a or

Discussion

In this work we showed that intracellular replicating Toxoplasma gondii take up Ca2+ through their plasma membrane. The host cytosolic Ca2+ is likely tightly regulated and kept low although physiological Ca2+ signaling events will result in Ca2+ increase. In our experimental set-up we stimulated signaling in host cells with specific agonists that had no direct effect on the parasite Ca2+. Host cytosol Ca2+ did respond to these agonists and was followed by a simultaneous increase in the PV Ca2+

Cell culture

T. gondii tachyzoites (RH strain) were maintained in hTERT human fibroblasts (BD Biosciences) using Dulbecoā€™s modified essential media (DMEM) with 1% fetal bovine serum (FBS), as described previously [46]. GCaMP6f expressing tachyzoites were maintained under similar conditions, in the presence of 20 ā€ÆĪ¼M chloramphenicol. The selection-less strain of GCaMP6f was grown under the same conditions as the RH strain. hTERT cells were maintained in high glucose DMEM with 10 % calf serum. HeLa cells

Funding

This work was supported by an NIH grant R01AI128356 to SNJM. SAV and EP were partially supported by fellowships (pre-doc and post-doc respectively) through a Training Grant in Tropical and Emerging Global Diseases (T32AI060546).

CRediT authorship contribution statement

Stephen A. Vella: Conceptualization, Methodology, Writing - original draft. Christina A. Moore: Methodology, Data curation. Zhu-Hong Li: Visualization, Investigation. Miryam A. Hortua Triana: Methodology, Investigation. Evgeniy Potapenko: Methodology, Investigation. Silvia N.J. Moreno: Supervision, Conceptualization, Writing - review & editing, Resources.

Declaration of Competing Interest

The authors report no declarations of interest.

Acknowledgements

We would like to thank Alex W. Chan and Dr. Sebastian Lourido for the protocol of natural egress using Compound 1; Dr. Vern Carruthers for the Ī”PLP1 mutant parasites; Dr. Diego Huet for reading the manuscript; Daniel Williamson for assisting on the egress of the Ī”PLP1 mutant; Beejan Asady for technical assistance; Julie Nelson for the FACā€™s sorting assistance; Dr. Kandasamy for technical assistance on the use of the microscopes at the Biomedical Microscopy Core.

References (53)

  • M.C. Berman

    Characterisation of thapsigargin-releasable Ca(2+) from the Ca(2+)-ATPase of sarcoplasmic reticulum at limiting [Ca(2+)]

    Biochim. Biophys. Acta

    (2000)
  • S.M. Sidik et al.

    Using a genetically encoded sensor to identify inhibitors of Toxoplasma gondii Ca2+ signaling

    J. Biol. Chem.

    (2016)
  • K.M. Brown et al.

    Serum albumin stimulates protein kinase G-dependent microneme secretion in Toxoplasma gondii

    J. Biol. Chem.

    (2016)
  • A.M. Gurnett et al.

    Purification and molecular characterization of cGMP-dependent protein kinase from Apicomplexan parasites. A novel chemotherapeutic target

    J. Biol. Chem.

    (2002)
  • N. Fertig et al.

    Whole cell patch clamp recording performed on a planar glass chip

    Biophys. J.

    (2002)
  • H.E. Bullen et al.

    Phosphatidic acid-mediated signaling regulates microneme secretion in toxoplasma

    Cell Host Microbe

    (2016)
  • J. Chemin et al.

    Overexpression of T-type calcium channels in HEK-293 cells increases intracellular calcium without affecting cellular proliferation

    FEBS Lett.

    (2000)
  • M.W. Black et al.

    Lytic cycle of Toxoplasma gondii

    Microbiol. Mol. Biol. Rev.

    (2000)
  • I.J. Blader et al.

    Lytic cycle of toxoplasma gondii: 15 years later

    Annu. Rev. Microbiol.

    (2015)
  • M.D. Bootman et al.

    Fundamentals of cellular calcium signaling: a primer

    Cold Spring Harb. Perspect. Biol.

    (2020)
  • J.L. Lovett et al.

    Intracellular calcium stores in Toxoplasma gondii govern invasion of host cells

    J. Cell. Sci.

    (2003)
  • D.L. Prole et al.

    Identification of intracellular and plasma membrane calcium channel homologues in pathogenic parasites

    PLoS One

    (2011)
  • B.F. Kafsack et al.

    Rapid membrane disruption by a perforin-like protein facilitates parasite exit from host cells

    Science

    (2009)
  • J.C. Schwab et al.

    The parasitophorous vacuole membrane surrounding intracellular Toxoplasma gondii functions as a molecular sieve

    Proc. Natl. Acad. Sci. U. S. A.

    (1994)
  • H. Dana et al.

    Sensitive red protein calcium indicators for imaging neural activity

    Elife

    (2016)
  • Y. Zhao et al.

    An expanded palette of genetically encoded Ca(2)(+) indicators

    Science

    (2011)
  • Cited by (18)

    • The immunomodulatory effects of roflumilast on tachyzoite-bradyzoite transition in a murine model of Toxoplasma gondii

      2022, International Immunopharmacology
      Citation Excerpt :

      Roflumilast increases intracellular cAMP, crucial in the transition of T. gondii tachyzoites to bradyzoites [9,34], which may lead to a decrease in intracellular calcium (Ca 2+) [35] that is fundamental to cellular and growth changes of T. gondii [36]. Parasites must be able to uptake Ca 2+ derived from host Ca 2+ signaling while replicating to keep their intracellular stores filled to be used during egress [37]. PDE4 inhibitors can interfere with the interconversion of tachyzoiteā€“bradyzoite by inhibiting cytokines, TNF-Ī±, IFN-Ī³, and IL-12, which have critical roles in this interconversion [10,38].

    • Pan-phylum genome-wide identification of sodium calcium exchangers reveal heterogeneous expansions and possible roles in nematode parasitism

      2022, Gene
      Citation Excerpt :

      In the free-living nematode, Caenorhabditis elegans, calcium has been shown to influence dauer stage regulation (Neal et al., 2015), which is the life stage in C. elegans that is analogous to the infective stage of parasitic nematodes. Calcium signaling is also required in diverse parasites for infection, including the apicomplexan Toxoplasma gondii where calcium regulates host cell egress (Vella et al., 2021), and also in the trypanosomatid species Trypanosoma cruzi, calcium is necessary for cell invasion (Moreno et al., 1994). In fact, targeting calcium signaling represents a strategy to target T. cruzi in order to combat Chagas disease (Benaim et al., 2020) as well as the parasite Plasmodium falciparum to tackle malaria (Scheibel et al., 1987).

    • Calcium signaling in intracellular protist parasites

      2021, Current Opinion in Microbiology
      Citation Excerpt :

      The PV of some strains of T. gondii are surrounded by host cell mitochondria and it has been suggested that these mitochondria buffer these Ca2+ increases in the cytosol of the host [19]. Another protection in T. gondii is the need of a threshold for Ca2+ increase to stimulate egress [20ā€¢]. Trypanosomatids possess an IP3R with structural similarity to the animal IP3Rs.

    View all citing articles on Scopus
    1

    Present addresses: Department of Orthopaedic Surgery, Duke University, 308 Research Drive, LSRC Room B323A, Durham, NC 27710.

    2

    Present addresses: School of Medicine and Public Health, Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, C4141 Veterans Administration Hosp.

    View full text