Calcium Signaling and The Lytic Cycle of - 2018 - Biochimica Et Biophysica Acta

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

BBA - Molecular Cell Research 1865 (2018) 1846–1856

Contents lists available at ScienceDirect

BBA - Molecular Cell Research


journal homepage: www.elsevier.com/locate/bbamcr

Calcium signaling and the lytic cycle of the Apicomplexan parasite T


Toxoplasma gondii☆
Miryam Andrea Hortua Trianaa,1, Karla M. Márquez-Noguerasa,1, Stephen A. Vellaa,1,

Silvia N.J. Morenoa,b,
a
Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
b
Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA

A R T I C LE I N FO A B S T R A C T

Keywords: Toxoplasma gondii has a complex life cycle involving different hosts and is dependent on fast responses, as the
Calcium signaling parasite reacts to changing environmental conditions. T. gondii causes disease by lysing the host cells that it
Toxoplasma gondii infects and it does this by reiterating its lytic cycle, which consists of host cell invasion, replication inside the
Lytic cycle host cell, and egress causing host cell lysis. Calcium ion (Ca2+) signaling triggers activation of molecules in-
Pathogenicity
volved in the stimulation and enhancement of each step of the parasite lytic cycle. Ca2+ signaling is essential for
Calcium dependent protein kinases
the cellular and developmental changes that support T. gondii parasitism.
Calcium binding proteins
The characterization of the molecular players and pathways directly activated by Ca2+ signaling in
Toxoplasma is sketchy and incomplete. The evolutionary distance between Toxoplasma and other eukaryotic
model systems makes the comparison sometimes not informative. The advent of new genomic information and
new genetic tools applicable for studying Toxoplasma biology is rapidly changing this scenario. The Toxoplasma
genome reveals the presence of many genes potentially involved in Ca2+ signaling, even though the role of most
of them is not known. The use of Genetically Encoded Calcium Indicators (GECIs) has allowed studies on the role
of novel calcium-related proteins on egress, an essential step for the virulence and dissemination of Toxoplasma.
In addition, the discovery of new Ca2+ players is generating novel targets for drugs, vaccines, and diagnostic
tools and a better understanding of the biology of these parasites.

1. Calcium and the Toxoplasma lytic cycle with the feces of infected cats, the definitive host [3,5]. Intermediate
hosts, such as animals and humans, can be infected via mature oocysts
Toxoplasma gondii belongs to the phylum Apicomplexa, which in- containing sporozoites or via contaminated meat with tissue cysts
cludes a number of unicellular eukaryotes that infect humans and an- containing bradyzoites. Both sporozoites and bradyzoites transform
imals and cause diseases, such as malaria (Plasmodium spp.), babesiosis into tachyzoites, the fast replicating form, which change back into slow
(Babesia spp.), toxoplasmosis (Toxoplasma gondii), cryptosporidiosis replicating bradyzoites within tissue cysts. These are characteristic of
(Cryptosporidium parvum), and cyclosporiasis (Cyclospora cayetanensis), the chronic infection and are usually found in the brain, eye, and stri-
among others. Apicomplexan are highly divergent from mammals and ated muscle tissues [6]. Disseminated toxoplasmosis can cause severe
more closely related to ciliates and dinoflagellates [1]. The name of the complications in immunocompromised patients including HIV-infected
phylum Apicomplexa denotes the presence of a unique set of secretory individuals, fetuses, and organ transplant recipients [7,8]. Some of the
organelles at the apical complex of zoites. These specialized organelles most devastating effects of toxoplasmosis are the result of the parasite
secrete unique effectors, in a highly regulated manner, critical for en- lytic cycle (Fig. 1), which starts when the tachyzoite attaches and in-
tering into their host cell [2]. vades host cells, replicates inside a parasitophorous vacuole and
Toxoplasma gondii is an obligate intracellular parasite that infects egresses to find another host cell to invade and re-start the cycle [9,10].
approximately one third of the world population [3,4]. Toxoplasma can Invasion and egress are dynamic processes that are highly regulated
be transmitted through contaminated food or water via oocyts released and essential for the propagation of the infection. Toxoplasma and other


This article is part of a Special Issue entitled: Calcium signaling in health, disease and therapy edited by Geert Bultynck and Jan Parys.

Corresponding author at: Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA.
E-mail address: smoreno@uga.edu (S.N.J. Moreno).
1
Equal Contribution, Authors' names are alphabetical.

https://doi.org/10.1016/j.bbamcr.2018.08.004
Received 15 April 2018; Received in revised form 6 August 2018; Accepted 7 August 2018
Available online 10 August 2018
0167-4889/ © 2018 Elsevier B.V. All rights reserved.
M.A. Hortua Triana et al. BBA - Molecular Cell Research 1865 (2018) 1846–1856

Fig. 1. The lytic cycle of Toxoplasma gondii. Toxoplasma tachyzoites attach and invade host cells and form a parasitophorous vacuole (PV) where they reside and
replicate by endodyogeny. The PV is surrounded by a PV membrane (PVM) that allows small molecules to sieve through. After several rounds of replication, they
rupture the PVM and the host cell membrane and egress followed by a short extracellular phase and invasion of another host cell. Christina Moore designed the Lytic
Cycle.

Apicomplexan parasites like Plasmodium rely on gliding motility for recent studies have provided evidence of their function in the lytic cycle
host-cell invasion, egress and dissemination in the infected host. of Toxoplasma.
Gliding motility, vital to a successful lytic cycle, is a peculiar mode of
substrate dependent motility, unique to Apicomplexan parasites, that
facilitates entry into host cells and is powered by a macromolecular 2. The Toxoplasma calcium signaling toolkit
complex termed the glideosome [11].
During invasion, the parasite secretes proteins from specific apical The information on the elements that form part of the calcium
organelles [12], and their contents mediate attachment to host cells and signaling toolkit of Toxoplasma and other Apicomplexan parasites is
formation of the specialized parasitophorous vacuole (PV) occupied by fragmented. The main challenge is the evolutionary distance of the
the parasite [13]. Each step during the lytic cycle has to be precise, fast Apicomplexa from mammalian model systems for which most of the
and effective and the parasite possesses specific molecules that become information is available, and making direct comparisons is not always
activated at the right instant. A strong body of evidence supports that informative. However, the characterization of the molecular pathways
intracellular calcium oscillations in the parasite precede the activation involved in calcium signaling in Toxoplasma and other pathogens is
or stimulation of the specific steps of the lytic cycle [14]. Both extra- highly relevant for several reasons. First, it is known that calcium
cellular and intracellular Ca2+ pools contribute to cytosolic Ca2+ in- fluctuations control vital cellular processes essential for parasitic life.
creases resulting in downstream signaling pathways that are decoded Second, it is very likely that new molecules with unique characteristics
into critical biological functions of the parasite lytic cycle. may result in the discovery of druggable targets. Lastly, as an early
Genomic analysis suggests the presence of a variety of calcium branching eukaryote, Toxoplasma occupies a unique phylogenetic po-
channels, which could potentially play a role in Ca2+ entry or release sition providing insights into the early origin of complex signaling
from intracellular stores [15]. There is evidence for Ca2+ release sti- pathways.
mulated by IP3 and cADP ribose [16,17] but no receptors for these Toxoplasma possesses characteristic calcium compartments such as
second messengers have been identified for any Apicomplexan parasite the endoplasmic reticulum (ER), Golgi apparatus and mitochondrium,
[18]. A number of Ca2+-binding proteins including calmodulins, cal- and additional compartments that could contribute to calcium signaling
modulin-like proteins, and an array of Ca2+-dependent protein kinases like acidocalcisomes [21–23] and endosome-like compartments like the
are predicted to be present in these parasites [19,20]. A large number of plant-like vacuole [24].
The concentration of Ca2+ in the cytosol of Toxoplasma tachyzoites

1847
M.A. Hortua Triana et al. BBA - Molecular Cell Research 1865 (2018) 1846–1856

is around 50–100 nM [25], similar to the cytosolic concentration of channel, termed transmembrane and coiled-coil domains 1 (TMCO1) or
other eukaryotic cells. Ca2+ signaling starts when the cytoplasmic le- Ca2+ load-activated Ca2+ (CLAC) channel has been shown to assemble
vels of Ca2+ are raised due to Ca2+ entry or Ca2+ release from in- as a homotetramer upon ER Ca2+ overload [38]. The main function of
tracellular stores. Ca2+ entry has an important role in refilling in- the channel is to prevent ER Ca2+ overload. A small portion of TMCO1
tracellular organelles that participate in signaling pathways that exists as homotetramer even under normal levels of ER Ca2+, which
respond to elevated cytosolic Ca2+ [26]. T. gondii does not appear to could also cause passive leak of Ca2+ from the ER. The genome of
express the molecular elements of Store Operated Calcium Entry Toxoplasma shows evidence for the presence of a TMCO1 ortholog
(SOCE). There is no genomic evidence for the presence of store-oper- (TGME49_310870), predicted to have a signal peptide at its N-terminus.
ated channels (ORAI), or for the ER sensor protein stromal interaction This Toxoplasma gene has not been characterized.
molecule (STIM). It appears that ligand-operated channels [26], or Acidocalcisomes are acidic organelles that store large amounts of
second messenger-operated channels are also missing in T. gondii. Gene Ca2+, which is mostly bound to polymers of phosphate [39]. Acid-
sequences with similarity to voltage dependent Ca2+ channels are ocalcisomes of T. gondii possess a plasma membrane-type Ca2+-ATPase
present in its genome [15,19]. The role of the encoded proteins as for Ca2+ uptake named TgA1 [40]. Purified acidocalcisome fractions
functional Ca2+ channels has not been demonstrated although evidence from T. gondii tachyzoites show vanadate-sensitive Ca2+ uptake sup-
has been presented for the presence of a nifedipine-sensitive Ca2+ entry porting the presence of this enzyme [23].
pathway that supports the function of a voltage-gated Ca2+ channel A vacuolar compartment with lysosomal characteristics (Plant-Like
[27]. Sequences with similarity to transient receptor potential (TRP) Vacuole or PLV) that expresses several orthologs of pumps and trans-
channels were also found in T. gondii [15] but have not been char- porters usually found in the plant vacuole has been characterized in
acterized. TRP channels are a superfamily of channels with diverse Toxoplasma and shown to store Ca2+ [24]. An increase in cytosolic
cellular functions [28] that can localize at the plasma membrane and Ca2+ in response to glycyl-L-phenylalanine-naphthylamide (GPN) in-
allow Ca2+ influx or to intracellular stores and allow Ca2+ release into dicated the presence of Ca2+ in the PLV where lytic activities are also
the cell cytosol. present. GPN has been used to study lysosomal Ca2+ because it causes
In vertebrate cells binding of a ligand to a surface receptor like a G- swelling of these compartments leading to leakage of Ca2+ into the
protein linked receptor (GPCR) may result in the stimulation of the cytosol. In Toxoplasma, GPN-dependent Ca2+ release was independent
phosphoinositide-signaling pathway via the activation of a phosphati- from release from other Ca2+ stores, such as the ER [24].
dylinositol phospholipase C (PI-PLC), which hydrolyzes phosphatidyli- The mitochondrion of T. gondii maintains a membrane potential and
nositol 4,5-bisphosphate (PIP2) to form inositol 1,4,5-trisphosphate carries out energy-linked functions like respiration coupled to oxidative
(IP3) and diacylglycerol (DAG). Binding of IP3 to an IP3 receptor usually phosphorylation [41]. However, it was not possible to demonstrate
present in the membrane of the ER, will result in its opening allowing Ca2+-uptake reliant on its membrane potential (unpublished results). In
Ca2+ to be released into the cytosol [29]. A diversity of cell signaling addition, there is no genomic evidence for the presence of a mi-
pathways can be generated through this mechanism [29]. Cyclic ADP tochondrial Ca2+ uniporter (MCU) in the inner membrane of any api-
ribose (cADPR) is also able to release intracellular Ca2+ acting through complexan parasite [42]. A Ca2+/H+ antiporter (CAX) was localized to
ryanodine receptors usually located in the ER [30]. Some of the ele- the mitochondria of P. falciparum [43] but the Toxoplasma ortholog did
ments of this signaling pathway have been found and characterized in not localize to the mitochondrion [44]. The potential role of the Tox-
Toxoplasma but a large number of them remains to be identified. The T. oplasma mitochondria in cytoplasmic Ca2+ homeostasis or even the role
gondii phosphoinositide phospholipase C (TgPIPLC) has been studied of mitochondrial Ca2+ on the regulation of mitochondrial enzymes is
[31] and characterization of conditional mutants of the Tgpiplc gene unknown [45].
highlighted its importance for parasite survival and its participation in Downstream to Ca2+, the signaling pathways that control essential
a vast and currently incompletely understood signaling cascade [32]. It and specific biological functions of Toxoplasma are not clearly defined.
is not clear yet, what is the target of the IP3 generated by TgPIPLC However, the information available is growing and several important
because there is no genomic support for the presence of IP3 receptors in players are emerging (Fig. 2). A role for a cyclic GMP (cGMP)-activated
any Apicomplexan parasite. However, IP3 stimulates Ca2+ release from Protein Kinase G (PKG) in the activation of Toxoplasma egress, motility
isolated microsomes of T. gondii [17] and cyclic ADP ribose does as and differentiation, has been reported [46,47]. Plant like Ca2+ de-
well, presumably by stimulating a ryanodine-type receptor [17]. Ca2+ pendent proteins kinases (CDPKs) are expanded across Apicomplexa
release by IP3 is inhibited by the IP3 receptor inhibitor xestospongin C and play pivotal roles in Ca2+ signaling throughout the lytic cycle [20].
[17], which also inhibited secretion of micronemes, secretory proteins CDPKs have been implicated in a range of processes in Toxoplasma, like
involved in host invasion [16]. T. gondii possesses ADP ribosyl cyclase invasion [48], egress [49,50,51] and microneme secretion [20,48]. A
and hydrolase [17] but there is no molecular evidence of a ryanodine recent study linked the central regulating role of PKG in the Ca2+
receptor. signals that precede egress to the activity of Protein kinase A. It was
The ER plays critical roles in calcium signaling in eukaryotic cells. proposed that PKA down-regulates PKG-dependent signaling leading to
Ca2+ influx into the ER is driven by a SERCA-type Ca2+ ATPases, which egress by potentially activating a phosphodiesterase that would hy-
has been characterized in T. gondii [33]. Inhibition of the T. gondii drolyze cGMP [52]. In a second study on Toxoplasma PKA the authors
SERCA-type Ca2+-ATPase by thapsigargin [34] results in increase of proposed that PKA is in the pathway that leads to suppression of cy-
cytosolic Ca2+ in T. gondii [25], due to Ca2+ leakage from the ER tosolic Ca2+ right after invasion promoting a decrease in motility and
through an unknown pathway. Ca2+ leakage is the passive calcium the beginning of intracellular replication [53]. It is clear that Ca2+, PKG
efflux from the ER that is thought to prevent ER calcium overload and and PKA signaling pathways crosstalk but we still do not know all the
thus allow cytosolic Ca2+ signaling [35]. In mammalian cells it has elements involved and the order in which they interact (Fig. 2). How-
been proposed that leak channels in the membrane of the ER play a role ever, the expanse of information during the last three years about sig-
in the steady state concentration of the ER luminal Ca2+ [36]. Several naling pathways involved in the regulation of Toxoplasma parasitism is
types of membrane proteins have been proposed to be involved in the encouraging and it is very likely that we will soon know how those
ER calcium leak pathway, Bcl-2, pannexin 1, presenilins and TRPC1. elements interact.
The Toxoplasma genome contains ortholog genes for a presenilin
(TGME49_204040) and two TRP channels (TGME49_247370 and 3. Calcium binding proteins in T. gondii
TGME49_310560) [15]. These Toxoplasma molecules have not been
characterized except for a recent study using high-affinity tags, which Intracellular Ca2+ increase activates a variety of cellular processes,
localized TGME49_247370 to the ER [37]. An active ER Ca2+ release including the mechanisms that return cytosolic Ca2+ concentration to

1848
M.A. Hortua Triana et al. BBA - Molecular Cell Research 1865 (2018) 1846–1856

Fig. 2. Ca2+ signaling in T. gondii tachyzoites. Ca2+ enters the parasite likely through a Ca2+ channel that is sensitive to nifedipine. Inside the cell, Ca2+ is pumped
either outside by a plasma membrane type Ca2+ ATPase or into organelles like the SERCA-Ca2+-ATPases localized to the endoplasmic reticulum (ER). The plasma
membrane type Ca2+-ATPase in Toxoplasma also localizes to acidic stores (AS) like acidocalcisome or PLV. A signaling cascade that starts by an increase in cGMP
level would lead to activation of a cGMP-protein kinase G that would result in cytosolic Ca2+ fluctuations that would activate biological responses like microneme
(MIC) secretion. In this hypothetical scenario PKA could phosphorylate an unidentified phosphodiesterase (PDE) resulting in its inhibition and leading to an increase
in cGMP levels. Downstream to PKG the pathway is hypothetical because the evidence is incomplete but CDPK3 could be part of it together with PI-PLC. The
hydrolysis of phosphatidylinositol 4,5 bisphosphate (PIP2) by PI-PLC forms two second messengers inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3
would act on an unknown release channel in the ER allowing Ca2+ to be released into the cytosol, which could activate CDPK1 stimulating microneme secretion.
Changes in DAG levels would activate DAGK1 (DAG kinase 1), which converts DAG into phosphatidic acid (PA), which is sensed by the micronemal protein APH
(acylated pleckstrin homology domain-containing protein) stimulating secretion. A Ca2+/H+ exchanger that was characterized in the PLV could use the proton
gradient to take-up Ca2+ into the acidic store [140]. How Ca2+ is released into the cytoplasm is not known. The bar at the upper right represents a Ca2+
concentration scale by blue color gradient (light to dark). Known pathways are shown by continuous lines, while hypothetical ones are represented by dashed lines.

its steady-state level, such as the membrane transporters that sequester Toxoplasma possesses a large number of CML proteins with low se-
Ca2+ into intracellular organelles or actively pump it out of the cell. quence identity and with degenerate EF-hand motifs [60]. CML proteins
Most signaling responses, however, involve Ca2+-binding proteins can have diverse subcellular localizations broadening the potential
(CaBPs). One of the most common motifs that binds Ca2+ and trans- substrates for their interaction [61]. Three of the CaM-like proteins,
duces these signals is the helix-loop-helix motif known as EF-hand. EF- named CaM1, CaM2, and CaM3, are important for motility and invasion
hands frequently occur in pairs (also called EF-hand domains) that and localize to the conoid where they overlap with the motor protein
allow for the cooperative binding of two Ca2+ ions per domain. CaBPs myosin H (MyoH). These proteins were shown to interact with MyoH
with single or odd number of EF hands sometimes function via dimer- and its light chain MLC7 [62,63]. CaM1 has two conserved EF hands,
ization mechanisms. Analysis of the T. gondii genome has revealed ap- while CaM2 has one conserved and one degenerate EF hand. Analysis of
proximately 68 EF-hand domain-containing proteins encoded by its mutants unable to bind Ca2+ was consistent with Ca2+ binding to the
genome (ToxoDB, http://toxodb.org/toxo/). The majority of these conserved EF hands for the regulation of function of CaM1 and CaM2
genes have not been characterized. [62]. CaM3 is essential for growth and it was proposed that it may
EF-hand containing proteins can be grouped into the calmodulin replace CaM1 and CaM2 in the activation of MyoH [64]. In agreement
(CaM) family, the calcineurin (CN) family, and the Ca2+-dependent with the function of CaMs in invasion, the CaM inhibitors calmidazo-
protein kinase (CDPK) family. Both CaM and CN family members lack lium and trifluoperazine were shown to inhibit T. gondii host cell entry
effector domains and are thought to act by regulating other proteins. [65].
The CaM group includes classical CaMs (with > 75% sequence identity Centrins are Ca2+ binding proteins usually present with centro-
to human CaM), calmodulin-like (CML) proteins, which include myosin somes. Apicomplexan centrins have been shown to associate with var-
light chains (sequence identity with human CaM < 75%), and centrins ious cytoskeletal components including the centriole and conoid
or calcactrins. Calmodulins comprise two globular domains (each with [66,67], but none have been demonstrated to be Ca2+ regulated. In-
a pair of EF hands) linked by a flexible helical region [54,55]. The T. terestingly, a recent finding showed that TgCentrin2 co-localized with
gondii genome encodes a single prototypical CaM and a variable number the TgPIPLC [37]. This localization was secondary to the plasma
of CML proteins. The T. gondii prototypical CaM binds Ca2+ in vitro membrane localization of the TgPIPLC and it was only possible to ob-
[56] and was localized to the apical domain [57], and more recently to serve with high-affinity tags. The significance of this finding is intri-
the conoid where it may interact with calcineurin [58]. No studies have guing and deserves further investigation.
addressed directly the role of CaM in the specific steps of the parasite Myosin light chains (MLCs) are calmodulin (CaM)-related proteins
lytic cycle. containing four EF-hand motifs and are important for the stability of the
Toxoplasma contains plant-like features because of its early myosin motor complex [68–70]. It is unlikely that these CaM-like
branching prior to the animal-plant separation [1] and also because of proteins will bind Ca2+ given that most of their EF-hands are degen-
the acquisition of an endosymbiont derived from an algal cell [59]. erate. Two myosin regulatory light chain proteins, essential light chains
Because of this, signaling pathways and molecules present in Tox- 1 and 2 (ELC1, ELC2), which form part of the glideosome, have been
oplasma will likely be very unique. For example, as in plants, studied and ELC1 has been shown to bind Ca2+ as estimated by

1849
M.A. Hortua Triana et al. BBA - Molecular Cell Research 1865 (2018) 1846–1856

modeling studies and thermal shift assays [71,72]. The affinity for Ca2+ microneme secretion, motility, host-cell invasion and egress [48]. These
of this protein is low (37 ± 9 μM) suggesting that for this interaction to studies highlighted the role of CDPKs in transducing Ca2+ signals.
be physiologically relevant it would have to be in a high Ca2+ con- A remarkable characteristic of TgCDPK1 is the presence of a small
centration microdomain. It is interesting that four CML proteins localize glycine gatekeeper residue in the ATP binding pocket making it sensi-
to the conoid. Considering what occurs with plant CML proteins and tive to ATP-competitive inhibitors with bulky substituents. Because of
CaMs, which can activate a variety of target proteins that regulate this, it is possible to inhibit TgCDPK1 with bulky ATP-analogues de-
metabolism, transcription and ion transport, it is very likely that the signed for kinases with extended ATP-binding pockets [48,79,81], as
Toxoplasma CML proteins that co-localize could have more than one well as with similar compounds designed for their improved anti-
target. The redundancy of these proteins, suggests that they play more parasitic properties [82–84].
than one role in Ca2+ signaling. TgCDPK3 has been shown to have a more specific function in
The CN family members have been identified only in some protist parasite egress, which was shown by characterizing a direct knockout of
genomes. Calcineurin is a Ca2+ regulated-phosphatase that consists of a the gene as well as with a chemical-genetic approach [49–51]. CDPKs
catalytic subunit (CnA) and a Ca2+-binding regulatory subunit (CnB). represent attractive drug targets because of their absence from the
Calcineurin phosphatase activity is usually regulated by changes in mammalian genome [20]. Several recent screens have targeted CDPKs
intracellular Ca2+ [73]. The role of calcineurin in T. gondii was studied for the development of new antiparasitic compounds [85–87].
by analyzing conditional CnA mutants and found to be important for T. gondii also expresses a number of noncanonical CDPKs with dif-
host cell attachment, invasion and stage-specific development of the ferent numbers of EF-hands, N-terminal EF hands, or with additional
parasite [58]. The next challenge will be to determine which are the domains. Much less is known about the function of these noncanonical
substrates for T. gondii calcineurin. CDPKs with the exception of TgCDPK7, which contains the kinase do-
A less prominent Ca2+-binding motif is the C2 domain, which was main preceded by a pleckstrin homology (pH) domain and two or more
originally identified as the domain responsible for the Ca2+-dependent incomplete EF-hands. Conditional knockout of TgCDPK7 showed a
phospholipid binding of protein kinase C (PKC). Although no PKC can defect in cell division [88]. Another study developed seven individual
be found in apicomplexan genomes, various C2-domain containing mutants of noncanonical CDPKs, plus double and triple mutants [89].
proteins have been identified. Among them, DOC2 has been shown to Only one of these mutants, ΔCDPK6, showed a mild growth phenotype.
be required for the Ca2+-regulated secretion of micronemes in T. gondii There is not a clear link between Ca2+ signaling and the potential
[74]. Apicomplexan PIPLC, which is thought to mediate intracellular function of these noncanonical CDPKs.
Ca2+ store release through the production of IP3, is also known to The challenge has been to characterize the biological targets of
contain a C2 domain, in addition to an EF-hand, although it is unknown CDPKs. Several studies have addressed this question by searching for
how Ca2+ impacts its regulation [31]. widespread changes in the phosphorylation state of diverse proteins
In plants there is a unique group of Ca2+ sensors and regulators following an increase in intracellular Ca2+ [70,90]. These studies have
known as Calcium Dependent Protein Kinases (CDPKs). Canonically, uncovered interesting molecules but it is still not clear what are the
CDPKs are composed of a kinase domain followed by a CaM-like do- specific effectors that are controlled by Ca2+. A comparison of the
main, also known as the CDPK activation domain (CAD) [75,76]. The phosphoproteomes of wild type and TgCDPK3-defficient parasites un-
activity of these kinases is directly regulated by Ca2+ binding to the covered hundreds of changes in diverse cellular pathways including
CAD. The important role of CDPKs in apicomplexans is reflected by proteins associated with gliding motility [90]. Several components of
their overrepresentation in the parasite genome. T. gondii CDPKs reg- the motor complex are heavily phosphorylated and this has been
ulate a variety of events in the parasites like egress, replication, motility documented [91]. Mutation of the identified phosphorylation sites was
and protein secretion. undertaken to identify which one was relevant for motor function [91].
The central player of the glideosome, a multisubunit motor complex, is
4. Ca2+-dependent protein kinases T. gondii myosin A (TgMyoA), an unconventional myosin that is es-
sential for motility, invasion and egress [69,92]. An increase in cyto-
The classical calmodulin-dependent kinases (CaMK) typical of an- plasmic Ca2+ resulted in increased MyoA phosphorylation and muta-
imal cells [77] are absent in Apicomplexan. Instead, they contain cal- tion of the major sites for TgMyoA phosphorylation altered the parasite
cium-dependent protein kinases (CDPKs), a family of serine/threonine response to cytoplasmic Ca2+ elevation by ionophores and other drugs
kinases that is present in plants and protists, including ciliates [20,78]. [93]. The expression of MyoA bearing phosphoro-mimetic mutations
In plants, CDPKs regulate a diversity of pathways like cell cycle pro- rescued the response to ionophores [93]. A more recent study using the
gression, stress response, stomatal closure, and root-nodule formation proximity-based protein interaction trap BioID identified 13 targets of
[75]. In Apicomplexan parasites CDPKs represent the best characterized CDPK3, among them TgMyoA [94]. In this study, a peptide array ap-
link between Ca2+ signaling and parasite biological functions like in- proach identified serines 21 and 743 as the targets for CDPK3 phos-
vasion, motility, egress and differentiation [20]. phorylation. CDPK3 phosphorylation of MyoA was important for in-
The typical structure of CDPKs comprises an N-terminal serine/ itiation of motility and egress providing a mechanistic link between
threonine kinase domain (KD) connected by a junction region to a CDPK3 regulation and the lytic cycle [94]. A study on egress and the
calcium-binding regulatory domain with four EF-hands forming a cal- role of CDPKs on Ca2+ signaling showed that CDPK1 and CDPK3 play a
modulin-like domain. The regulatory domain is termed the CDPK-ac- role in the Ca2+ signal recovery prior to egress [95]. A forward-genetic
tivation domain (CAD) [76]. The structures of T. gondii CDPK1 in the screen to isolate gain-of-function CDPK3 mutants [96] followed this
presence and absence of Ca2+ revealed the molecular details of their study, which uncovered a new component that was characterized as
activation [76,79]. In the absence of Ca2+, the junction region lodges in suppressor of Ca2+-dependent cell egress [96].
the substrate-binding region, blocking the catalytic pocket of the en- It is very likely that the essential phenotypes attributed to CDPKs
zyme keeping it inactive. Upon binding of Ca2+ to the EF-hands a are the result of their important regulatory functions but it is clear that
dramatic structural rearrangement leads to a segmentation of the au- more work is needed to clarify the signaling network that is triggered
toinhibitory helix around the calmodulin like-domain altering the in- upon cytoplasmic Ca2+ increase and its downstream stimulation of
teraction and relieving the inhibition [76]. A study with CDPK1 showed kinases, phosphatases or other targets. Deciphering the specific targets
that upon binding calcium the CAD domain rotates with respect to the of CDPKs will be an essential task to understand the signaling pathways
kinase domain and in addition to relieving autoinhibition stabilizes the that lead to the biological functions that are essential for Toxoplasma
active form of the enzyme [80]. The Toxoplasma CDPK1 is essential and parasitism.
conditional knockdown mutants of TgCDPK1 are defective in

1850
M.A. Hortua Triana et al. BBA - Molecular Cell Research 1865 (2018) 1846–1856

5. Microneme secretion, motility, host cell invasion and egress homologue of the human redox chaperone protein, T. gondii DJ-1, with
specific function in parasite motility and microneme secretion [112],
Toxoplasma is an obligate intracellular parasite that multiplies in- even when stimulating secretion with ionophore or ethanol. This sug-
side host cells. The process of invasion is actively driven by the parasite gested that the role of TgDJ-1 is downstream of Ca2+ signaling [112] as
and is fast, precise, and involves gliding motility, conoid extrusion, and was also shown for TgCDPK1. It was proposed that the regulation of
microneme secretion [97–99]. An increase in cytosolic Ca2+ artificially microneme secretion by CDPK1 was in part due to its interaction with
elevated with ionophores, like ionomycin or A23187, stimulated conoid TgDJ-1 in a Ca2+ and H2O2-dependent manner [113].
extrusion of T. gondii tachyzoites, an effect that was prevented if the The role of the cGMP-dependent protein kinase (PKG) in microneme
parasites were pre-loaded with BAPTA-AM [98]. Conoid extrusion also exocytosis was first demonstrated by its inhibition with compound 1
responded to other cytosolic Ca2+ triggers like ethanol [100] and ex- (Cpd1), a trisubstituted pyrrole derivative with in vivo activity against
tracellular Ca2+ enhanced and extended the extrusion time [27]. Toxoplasma [114]. The validation of PKG as target of Cpd1 was done
BAPTA-AM also prevented gliding motility [97] and host cell invasion with transgenic parasites complemented with mutated versions of
[101] by T. gondii, supporting a role for Ca2+ in these essential parasite TgPKG (T761Q), which are insensitive to the inhibition of microneme
functions. Conditional downregulation of the expression of a conoid secretion by Cpd1 [46]. Increase of cytosolic Ca2+ with ionophores did
protein hub 1 (CPH1) caused the conoid to collapse and become shorter not reverse the inhibition of microneme secretion by Cpd1 suggesting a
[64]. In this work, the stimulation of conoid extrusion or microneme downstream role for PKG in this process [46]. Toxoplasma possesses a
secretion by ionophore was not affected even though CPH1 was es- single gene for PKG, which is essential, but expresses two isoforms with
sential for invasion and conoid structure [64]. The specifics about how identical regulatory and catalytic domains, PKGI is membrane bound
Ca2+ increase leads to extension of the conoid are not clearly defined. and PKGII is cytosolic [115]. Recent work on PKG differentiated the
Several Ca2+ binding proteins have been localized to the structure [62]. contribution of each PKG isoform to Toxoplasma biology using the
A recently described conoid ring protein, RNG2, was shown to change auxin-inducible degron (AID) tagging system for conditional depletion
its orientation in response to stimulation of conoid extrusion with Ca2+ of PKG protein followed by complementation with mutated versions of
ionophores [102]. Based on this result, a role for RNG2 in secretion was PKG [116]. PKGI, the membrane associated isoform is essential and
proposed. sufficient for microneme secretion and parasite survival. PKGII, the
Toxoplasma gondii contains micronemes, specialized secretory or- cytosolic version is dispensable but its function became relevant when
ganelles important for gliding motility and host cell invasion [103,104]. the gene was modified so the protein localized to the plasma membrane
Micronemes are small apical organelles with an elongated shape and [116]. These results support a role for PKG downstream or in-
electron-dense structure [104]. Micronemes store adhesins that upon dependently of the events that lead to intracellular Ca2+ increase
secretion participate in the interactions with the host-cell surface and [46,116]. However, studies with parasites loaded with the Ca2+ in-
participate in the early stages of host-cell invasion. Secretion of mi- dicator Fura2-AM showed that accumulation of cGMP by inhibiting its
cronemes can be triggered by elevating cytosolic Ca2+ with ionophores phosphodiesterases (PDE) with zaprinast, resulted in an increase in
and can be blocked by chelating intracellular Ca2+ with BAPTA-AM cytosolic Ca2+, which is dependent on PKG activity because this
[103,105]. A number of studies have shown that multiple external Ca2+increase was almost 80–90% inhibited by Cpd1 [117].
stimuli can trigger microneme secretion such as a drop in extracellular It is interesting that serum albumin acts synergistically with ethanol
K+ [106], an increase in H+ [107], serum albumin [108] and extra- to increase cytosolic Ca2+ and microneme secretion [108]. The sti-
cellular Ca2+ [27]. The precise signaling pathway that leads to mi- mulation of microneme secretion was proposed to occur through a
croneme exocytosis, and especially how Ca2+ stimulates it, is not well Ca2+-independent pathway on the basis of Ca2+ measurements using
defined, but some participating molecules have been identified. One the genetic indicator GCaMP6f. However, it remains to be determined if
study on the characterization of Ca2+-ATPase (TgA1) [40] knockout the Ca2+ increase was below the GCaMP6 sensitivity as this indicator
mutants, which showed altered levels of intracellular Ca2+, examined exhibits different kinetics and affinities for Ca2+ than Fura2 [118].
the role of Ca2+ in the lytic cycle of Toxoplasma. These mutant parasites Early studies evaluating microneme secretion of Toxoplasma were per-
are deficient in microneme secretion, invasion, and showed reduced formed in the presence of bovine serum, which has been shown to
virulence in vivo [109]. stimulate microneme secretion in related apicomplexan
A study looking at the mechanism of action of the antimalarial drug [16,99,105,108]. More recently it was determined that serum albumin,
artemisinin, showed that thapsigargin and artemisinin triggered Ca2+- the major component of serum, was sufficient to induce microneme
dependent secretion of micronemes. Artemisinin treatment altered secretion, while γ-globulins, the second major component of serum had
Toxoplasma intracellular Ca2+ and increased the periodicity of Ca2+ no effect [108]. It is well characterized that Ca2+ ionophores such as
oscillations as imaged in parasites loaded with the Ca2+ fluorescent ionomycin or A23187 induce microneme secretion, however in a study
indicator Fluo4 [33]. Previous reports had shown in Plasmodium, that characterizing the role of extracellular Ca2+, microneme secretion was
the SERCA orthologue PfATPase6 could be the target of artemisinin shown to correlate with the concentration of extracellular Ca2+ sup-
[110] and considering that artemisinin altered Ca2+ signaling in Tox- porting an enhancing role for Ca2+ influx [27]. This study did not use
oplasma [111] it was proposed that the drug could inhibit TgSERCA, ionophores or other triggers, or serum and parasites were kept in low
which was further supported by the sensitivity of yeast expressing Ca2+ buffer until the microneme assay was performed. It is possible
TgSERCA to artemisinin. Presently, the mechanism (s) of action of ar- that Ca2+ influx leads to an increase of Ca2+ at specific peripheral
temisinin, still remains unclear because Toxoplasma artemisinin-re- domains of the parasite and bypasses the intracellular signaling trig-
sistant mutants did not show apparent mutations or alterations in the gered by PKG. Additional studies are needed to clarify the link between
expression levels of TgSERCA or other Ca2+-ATPase genes [111]. Ca2+ and PKG signaling in T. gondii.
CDPK1 conditional knockdowns highlighted the role of the Ca2+- Most evidence linking Ca2+ signaling to parasite replication is in-
stimulated kinase in attachment and secretion of micronemes [48]. The direct. Conditional knock-downs of CDPK7, a potentially Ca2+ regu-
authors characterized the inhibition of TgCDPK1 by 3-methylbenzyl- lated kinase, results in parasites with replication phenotypes [88].
PP1 (3-MB-PP1), an ATP analog that inhibited the enzyme due to the Knockdown of CDPK7 alters centrosome duplication and positioning.
presence of the amino acid glycine in the gate-keeper position allowing CDPK7 appears to play a role in the positioning of secretory organelles
the inhibitor to block the ATP binding site. The PP1 inhibitor blocked like rhoptries and micronemes. The miss-positioning of these organelles
microneme secretion and related functions. Mutation of glycine to a in the mutants implicated CDPK7 in the trafficking of these organelles
methionine (G for M) made the enzyme insensitive to the inhibitor [48]. to daughter cells [88]. During intracellular growth, GRA1, a dense
A chemical genetic study with the inhibitor WRR-086, identified an granule protein secreted within the PV was shown to be important for

1851
M.A. Hortua Triana et al. BBA - Molecular Cell Research 1865 (2018) 1846–1856

the formation of the tubulovesicular network [119,120]. GRA1 pos- gliding motility and microneme secretion [134], which has been used
sesses two EF-hand domains, which were shown to bind Ca2+ and it to deduce our current understanding of this form of parasite movement.
was proposed that it would act as a Ca2+ buffer within the PV [119]. For example, increasing the frequency and lowering the amplitude of
The role of host Ca2+, specifically Ca2+ entry, was shown with the Ca2+ oscillations with calmidazolium enhanced microneme secretion
inhibitor L-651,582, which apparently blocks Ca2+ entry in the host and gliding motility [97]. Calmidazolium stimulated gliding and this
cell affecting parasite replication [121]. L-651,582 inhibited Tox- was proposed to depend on MIC2 secretion. The increase of cytosolic
oplasma and Eimeria growth in a variety of mammalian host cells [121]. Ca2+ levels was shown to occur through release from intracellular
This result suggests that during its intracellular replication phase the stores and entry from the extracellular milieu. The calmidazolium study
parasite might need to take up Ca2+ from the host to keep its stores highlighted the importance of Ca2+ oscillations vs. constant Ca2+ in-
replenished. It is also possible that a number of Ca2+ regulated proteins crease for efficient gliding of T. gondii [97]. It is possible that Ca2+
are involved in the replication of the parasite. There is evidence for the oscillations enable the parasite to traverse longer distances over a
presence of a higher concentration of Ca2+ within the PV [122] and longer period of time, via coordinating the release of micronemes only
host Ca2+ oscillation do cause oscillations in the parasite [123]. It is when necessary instead of constitutively [95,123]. Whether Ca2+ reg-
still not clear how host cytosolic Ca2+ impacts the biological functions ulates aspects of gliding motility beyond secretion of adhesins remains
of the parasite. Parasites mutants on a dense granule protein secreted to to be determined.
the PV, GRA41, showed alteration in their cytosolic Ca2+ levels and Release of intracellular Ca2+ stores was proposed to be sufficient to
egress [124]. initiate gliding motility and host-cell invasion by use of ionophores or
Active egress of Toxoplasma from host cells requires rupture of the intracellular Ca2+ chelators like BAPTA [16]. However, extracellular
parasitophorous vacuole membrane (PVM) and the host cell membrane Ca2+ influx impacted and enhanced invasion-linked traits like motility,
[125]. Egress is essential for the dissemination of the infection and it conoid extrusion, microneme secretion and invasion [27]. T. gondii
has been known for several years that Ca2+ ionophores can trigger replicates through endodyogeny, and its lytic cycle is complex and
egress [126]. However, it was the use of Genetically Encoded Ca2+ parasites commonly invade and exit with little or no replication
Indicators (GECIs) that provided the final and conclusive evidence that [135,136]. This complex behavior, all of which depends on Ca2+, likely
there was a cytosolic Ca2+ increase right before egress [123]. The use requires replenishing intracellular Ca2+ stores with extracellular Ca2+.
of these indicators has vastly impacted the studies of Ca2+ in in- It would be advantageous for the parasite to utilize the high con-
tracellular parasites [95,117,123,127]. Secretion from the micronemes centration of extracellular Ca2+ to enhance invasion-related processes
of the perforin-like protein TgPLP1, assists in the permeabilization of and to resupply intracellular stores for use during subsequent rounds of
the PVM and host cell membrane [128]. Initiation of parasite motility invasion and egress. Experimental evidence supports a general me-
results in mechanical pressure and final rupture of the host cell allowing chanism of Ca2+ entry, which may not be sensitive to the filling state of
the release of the parasites. Both, secretion of the micronemal protein the ER, as is the case in mammalian cells, but which could work in
TgPLP1 and initiation of motility during egress are stimulated by an parallel with intracellular store release to elevate intracellular Ca2+,
increase in cytosolic Ca2+. A role for a change in extracellular po- and activate downstream events like invasion and motility [27].
tassium concentration was proposed to stimulate Ca2+ signaling The glideosome is the molecular machinery that generates the me-
leading to egress [106]. The presence of extracellular Ca2+ also affected chanical force needed for completion of the lytic cycle [137]. Inhibition
the rate of egress, which was blocked with the Ca2+-channel blocker (using pharmacological agents) or disruption (using knockouts or con-
nifedipine [123]. A plant-like pathway involving the phytohormone ditional depletion of core components) of the glideosome has been
abscisic acid (ABA) was proposed to be the pathway leading to the known to adversely affect the lytic cycle events of egress, extracellular
production of the natural signal for Toxoplasma egress [129]. The initial motility, and invasion thus highlighting its significance across the lytic
trigger that starts the Ca2+ signaling leading to egress is still a mystery. cycle [10,125]. Egress results from the initiation of the glideosome and
The isolation of parasite mutants resistant to ionophore-induced parasites will continue to be actively motile until invasion of new host
egress (or delayed response) identified mutants defective in Ca2+ sig- cells suggesting that extracellular motility serves as the bridge con-
naling. A mutant defective in a Na+/H+ exchanger (TgNHE1), was necting egress and invasion [10]. As an intracellular parasite, T. gondii
isolated in this way and found to have elevated intracellular Ca2+ levels must replicate inside host cells; and as such, glideosome activation must
and reduced pathogenicity [130]. A similar screen identified TgCDPK3 be tightly regulated across time and space [91]. Several studies have
as a mediator of ionophore-induced egress [50]. TgCDPK3 likely con- demonstrated that glideosome activation occurs in a Ca2+-dependent
trols rapid exit from the host by phosphorylating proteins involved in manner, through multiple, interconnected and cross-talking mechanism
the activation of motility. Another CDPK3 substrate that plays a role in (s) [51,94,125] (Fig. 4A). Furthermore, a recent study identified that
egress is the Suppressor of Ca2+ Egress 1 or SCE1, and its phosphor- the force and direction of gliding motility occurs in a Ca2+-dependent
ylation relieves the suppression of Ca2+-dependent host cell egress manner. At basal levels the net mechanical force of the parasite is
[96]. random and disorganized. Post increases in cytosolic Ca2+, the net
Extracellular tachyzoites exhibit three types of motility (Fig. 3): mechanical force of the parasite becomes polarized and oriented across
circular (counter-clockwise circular motion), helical (forward cork- the long axis of the parasite thus sufficient for productive motility
screw motion), and twirling (attachment to posterior end and spinning [138].
vertically) [131], though within a 3D matrix, motility is one continuous The glideosome is a multi-subunit actomyosin motor complex that
irregular corkscrew motion [132]. Helical gliding is considered the resides at the periphery of the parasite; confined to the space below the
most effective and essential for invasion [131]. Ca2+ oscillations were plasma membrane (PM) and above an underlying structure termed the
first observed in actively motile parasites loaded with Fluo-4 that pre- inner membrane complex (IMC) (Fig. 4A) [11,125]. The complex
ceded periods of microneme secretion and bursts of speed [97,133]. (Fig. 4B) is anchored to the IMC via the scaffolding complex of gli-
Cytosolic Ca2+ fluctuations were further characterized with parasites deosome-associated proteins (GAP's), and members of the GAPM family
expressing GECIs and a direct correlation between distance traveled [11]. TgGAP45 serves as a molecular tether that links the IMC to the PM
and amplitude of the oscillations was exposed [95,123]. Cytosolic Ca2+ through lipid modification links [11,70]. TgMyoA, an important com-
levels are reduced at the moment of invasion and stay low right after ponent of the glideosome [70,92], is an unconventional myosin of the
invasion. The oscillation pattern of cytosolic Ca2+ in motile parasites class XIVa that interacts with its associated light chain, TgMLC1 and
suggests that Ca2+ influx channels and reuptake mechanisms are highly either the essential light chain 1 or 2 (ELC1 or ELC2) [70,72]. Though
active in gliding parasites and necessary for effective lytic cycle events MyoA serves as the primary source for the generation of force needed
[95,123,133]. Similar perturbations of Ca2+ signaling also affect for motility, it is the interaction between the MyoA neck with ELC1/2

1852
M.A. Hortua Triana et al. BBA - Molecular Cell Research 1865 (2018) 1846–1856

Fig. 3. T. gondii Motility Types. (From left to right)


Representative stills of Circular (counter clockwise
circular motion), Helical (forward corkscrew mo-
tion), and Twirling (attachment on posterior end with
windmill-like motion) motility post extracellular
Ca2+ addition. T. gondii tachyzoites are expressing
cytosolic GCaMP6f [123] and exposure to extra-
cellular Ca2+ stimulates motility and fluorescence of
the indicator. Size bar: 5 μM.

that supports force transduction throughout the motor complex and coordinate the dynamic cycles of phosphorylation/dephosphorylation
more broadly throughout the body of the parasite as GAP proteins are of MyoA, needed for effective gliding motility/invasion [51,93,94].
anchored to the cytoskeleton [11,72,139]. Within the glideosome An in vitro assay utilized to monitor actin displacement of purified
MLC1, ELC1 and ELC2, interact either directly with Ca2+ or via Ca2+ MyoA, MLC1, and ELC1 identified that when ELC1 was added to
dependent phosphorylation to stimulate motility and microneme se- complexed MyoA-MLC1, MyoA moved actin filaments at more than
cretion [14,51,70] (Fig. 4B). twice the speed (and more in line with motor complexes isolate from
Though several Ca2+-dependent phosphorylation sites of the gli- parasites) as compared to MyoA-MLC1 alone [139]. Later it was de-
deosome are dispensable, mutations of S20, S21, and S29 of MyoA have termined that in addition to enhancing the thermostability of ELC1 in
been determined to be important for MyoA activity, whereas S21 ap- the presence of Ca2+, ELC1 had an enhanced affinity for MyoA and is
pears to be the primary site for phosphorylation [70,91,93]. A recent recruited by MLC1 [70,72]. Given the low affinity (μM) of ELC1 (and
study determined that disruption of the phosphorylation state(s) of presumably ELC2) for Ca2+, it is reasonable to hypothesize that ELC1(/
MyoA by expressing mutated versions of MyoA serine residues, altered 2) may have evolved to optimally bind Ca2+ at high concentrations
the motility behavior of gliding parasites as compared to wild type cells and/or rapidly release Ca2+ in continuous succession [72]. Perhaps
[93]. These results suggest that the summation of Ca2+ oscillations may through rapidly binding and releasing Ca2+, during the Ca2+

Fig. 4. A) Signaling and activation of Motility. Activation of Ca2+ signaling will translate into the activation of CDPK3 which will phosphorylate proteins from the
molecular machinery and activating motility. The glideosome is the main molecular motility machinery and is anchored to the underlying cytoskeleton termed the
inner membrane complex (IMC). Adapted from [14], with permission. (B) The glideosome of T. gondii. MLC1 and/or ELC1 or ELC2 interact with Ca2+ directly or via
Ca2+ dependent phosphorylation events to stimulate motility. The glideosome was adapted from [71].

1853
M.A. Hortua Triana et al. BBA - Molecular Cell Research 1865 (2018) 1846–1856

oscillations that govern parasite motility, ELC1(/2) may serve as a [4] G. Pappas, N. Roussos, M.E. Falagas, Toxoplasmosis snapshots: global status of
“molecular throttle” to enhance MyoA activity, thus highlighting its Toxoplasma gondii seroprevalence and implications for pregnancy and congenital
toxoplasmosis, Int. J. Parasitol. 39 (2009) 1385–1394.
potential role in decoding the Ca2+ signals that govern parasite moti- [5] J.P. Dubey, Advances in the life cycle of Toxoplasma gondii, Int. J. Parasitol. 28
lity. (1998) 1019–1024.
[6] J.P. Dubey, D.S. Lindsay, C.A. Speer, Structures of Toxoplasma gondii tachyzoites,
bradyzoites, and sporozoites and biology and development of tissue cysts, Clin.
Outlook Microbiol. Rev. 11 (1998) 267–299.
[7] L.M. Weiss, J.P. Dubey, Toxoplasmosis: a history of clinical observations, Int. J.
In the last 3 years since our last review of the Ca2+ research lit- Parasitol. 39 (2009) 895–901.
[8] B.J. Luft, J.S. Remington, Toxoplasmic encephalitis in AIDS, Clin. Infect. Dis. 15
erature [14] we have witnessed how new genetic tools have advanced (1992) 211–222.
our knowledge on Ca2+ signaling in Toxoplasma. Some of these ad- [9] M.W. Black, J.C. Boothroyd, Lytic cycle of Toxoplasma gondii, Microbiol. Mol.
vances include the use of CRISPR-Cas9 for genome editing increasing Biol. Rev. 64 (2000) 607–623.
[10] I.J. Blader, B.I. Coleman, C.T. Chen, M.J. Gubbels, Lytic cycle of toxoplasma
the efficiency for the characterization of Toxoplasma genes, the ex-
gondii: 15 years later, Annu. Rev. Microbiol. 69 (2015) 463–485.
pression of genetically encoded Ca2+ indicators in Toxoplasma tachy- [11] A. Keeley, D. Soldati, The glideosome: a molecular machine powering motility and
zoites, the use of a biotin ligase fusion protein to identify proximal and host-cell invasion by Apicomplexa, Trends Cell Biol. 14 (2004) 528–532.
interacting proteins, and the adaptation of tools to down-regulate [12] V.B. Carruthers, L.D. Sibley, Sequential protein secretion from three distinct or-
ganelles of Toxoplasma gondii accompanies invasion of human fibroblasts, Eur. J.
protein levels allowing studies of more immediate phenotypic differ- Cell Biol. 73 (1997) 114–123.
ences without previous adaptation. The field is moving leap wise and it [13] V.B. Carruthers, Host cell invasion by the opportunistic pathogen Toxoplasma
is expected that we will see more and more discoveries of new elements gondii, Acta Trop. 81 (2002) 111–122.
[14] S. Lourido, S.N. Moreno, The calcium signaling toolkit of the Apicomplexan
and pathways components. parasites Toxoplasma gondii and Plasmodium spp, Cell Calcium 57 (2015)
During these last three years we have finally been able to see Ca2+ 186–193.
oscillations prior to exit of the parasite from its host cells. This was [15] D.L. Prole, C.W. Taylor, Identification of intracellular and plasma membrane
calcium channel homologues in pathogenic parasites, PLoS One 6 (2011) e26218.
years after Endo [126] showed that ionophores trigger egress and as- [16] J.L. Lovett, N. Marchesini, S.N. Moreno, L.D. Sibley, Toxoplasma gondii micro-
sumed that it was because of a Ca2+ response. These studies were neme secretion involves intracellular ca(2+) release from inositol 1,4,5-tripho-
possible thanks to the use of GECIs that allowed to specifically detect sphate (IP(3))/ryanodine-sensitive stores, J. Biol. Chem. 277 (2002)
25870–25876.
Ca2+ changes in live intracellular parasites. We still need to know how [17] E.N. Chini, K. Nagamune, D.M. Wetzel, L.D. Sibley, Evidence that the cADPR
intracellular vs extracellular Ca2+ crosstalk to deliver the threshold signalling pathway controls calcium-mediated microneme secretion in
needed for increase in motility and egress. Toxoplasma gondii, Biochem. J. 389 (2005) 269–277.
[18] C.R.S. Garcia, E. Alves, P.H.S. Pereira, P.J. Bartlett, A.P. Thomas, K. Mikoshiba,
We have also seen evidence of crosstalk between second messengers
H. Plattner, L.D. Sibley, InsP3 signaling in apicomplexan parasites, Curr. Top.
like Ca2+, cGMP and cAMP. The picture is still not very clear, especially Med. Chem. 17 (2017) 2158–2165.
about which one is upstream of which, but there are no doubts that [19] K. Nagamune, L.D. Sibley, Comparative genomic and phylogenetic analyses of
information is flowing and we will see this mystery uncovered in the calcium ATPases and calcium-regulated proteins in the apicomplexa, Mol. Biol.
Evol. 23 (2006) 1613–1627.
near future. [20] O. Billker, S. Lourido, L.D. Sibley, Calcium-dependent signaling and kinases in
There is no doubt about the significance of Ca2+ for gliding moti- apicomplexan parasites, Cell Host Microbe 5 (2009) 612–622.
lity, microneme secretion, host cell invasion and egress of T. gondii. [21] C.O. Rodrigues, F.A. Ruiz, P. Rohloff, D.A. Scott, S.N. Moreno, Characterization of
isolated acidocalcisomes from Toxoplasma gondii tachyzoites reveals a novel pool
How Ca2+ regulates aspects of gliding motility beyond secretion of of hydrolyzable polyphosphate, J. Biol. Chem. 277 (2002) 48650–48656.
adhesins remains to be determined. The same argument can be ex- [22] K. Miranda, W. de Souza, H. Plattner, J. Hentschel, U. Kawazoe, J. Fang,
tended to our understanding of parasite invasion and egress from host S.N. Moreno, Acidocalcisomes in Apicomplexan parasites, Exp. Parasitol. 118
(2008) 2–9.
cells, although certain cellular components may affect the different [23] P. Rohloff, K. Miranda, J.C. Rodrigues, J. Fang, M. Galizzi, H. Plattner,
phenotypes to different extents. Ca2+ signaling is controlled by its J. Hentschel, S.N. Moreno, Calcium uptake and proton transport by acidocalci-
uptake and release from different cellular compartments and there are somes of Toxoplasma gondii, PLoS One 6 (2011) e18390.
[24] K. Miranda, D.A. Pace, R. Cintron, J.C. Rodrigues, J. Fang, A. Smith, P. Rohloff,
important differences with the processes that control Ca2+ homeostasis
E. Coelho, F. de Haas, W. de Souza, I. Coppens, L.D. Sibley, S.N. Moreno,
in other eukaryotic cells, providing opportunities for finding novel drug Characterization of a novel organelle in Toxoplasma gondii with similar compo-
targets. It is also apparent that the more we learn about Ca2+ signaling sition and function to the plant vacuole, Mol. Microbiol. 76 (2010) 1358–1375.
[25] S.N. Moreno, L. Zhong, Acidocalcisomes in Toxoplasma gondii tachyzoites,
in Toxoplasma the more we know about the basic mechanisms that
Biochem. J. 313 (Pt 2) (1996) 655–659.
regulate how Toxoplasma, and other related pathogens, cause disease. [26] A.B. Parekh, J.W. Putney Jr., Store-operated calcium channels, Physiol. Rev. 85
(2005) 757–810.
Transparency document [27] D.A. Pace, C.A. McKnight, J. Liu, V. Jimenez, S.N. Moreno, Calcium entry in
Toxoplasma gondii and its enhancing effect of invasion-linked traits, J. Biol.
Chem. 289 (2014) 19637–19647.
The Transparency document associated with this article can be [28] K. Venkatachalam, C. Montell, TRP channels, Annu. Rev. Biochem. 76 (2007)
found, in online version. 387–417.
[29] M.J. Berridge, Inositol trisphosphate and calcium signalling mechanisms, Biochim.
Biophys. Acta 1793 (2009) 933–940.
Acknowledgments [30] H.C. Lee, Cyclic ADP-ribose and NAADP: fraternal twin messengers for calcium
signaling, Sci. China Life Sci. 54 (2011) 699–711.
[31] J. Fang, N. Marchesini, S.N. Moreno, A Toxoplasma gondii phosphoinositide
S.N.J.M. is supported by grants from the U.S. National Institutes of phospholipase C (TgPI-PLC) with high affinity for phosphatidylinositol, Biochem.
Health (AI-096836, and AI128356). KMN was supported in part by a J. 394 (2006) 417–425.
fellowship from the T32 training grant to the Center for Tropical and [32] H.E. Bullen, Y. Jia, Y. Yamaryo-Botte, H. Bisio, O. Zhang, N.K. Jemelin, J.B. Marq,
V. Carruthers, C.Y. Botte, D. Soldati-Favre, Phosphatidic acid-mediated signaling
Emerging Global Diseases (AI060546). SAV was partially supported by
regulates Microneme secretion in Toxoplasma, Cell Host Microbe 19 (2016)
a fellowship from the UGA Office of the Vice-President for Research 349–360.
(OVPR). We thank Christina Moore for help with the drawings. [33] K. Nagamune, W.L. Beatty, L.D. Sibley, Artemisinin induces calcium-dependent
protein secretion in the protozoan parasite Toxoplasma gondii, Eukaryot. Cell 6
(2007) 2147–2156.
References [34] O. Thastrup, P.J. Cullen, B.K. Drobak, M.R. Hanley, A.P. Dawson, Thapsigargin, a
tumor promoter, discharges intracellular Ca2+ stores by specific inhibition of the
[1] S.L. Baldauf, The deep roots of eukaryotes, Science 300 (2003) 1703–1706. endoplasmic reticulum Ca2(+)-ATPase, Proc. Natl. Acad. Sci. U. S. A. 87 (1990)
[2] L.D. Sibley, Intracellular parasite invasion strategies, Science 304 (2004) 248–253. 2466–2470.
[3] D. Hill, J.P. Dubey, Toxoplasma gondii: transmission, diagnosis and prevention, [35] D.E. Clapham, Calcium signaling, Cell 131 (2007) 1047–1058.
Clin. Microbiol. Infect. 8 (2002) 634–640. [36] A. Guerrero-Hernandez, A. Dagnino-Acosta, A. Verkhratsky, An intelligent sarco-
endoplasmic reticulum Ca2+ store: release and leak channels have differential

1854
M.A. Hortua Triana et al. BBA - Molecular Cell Research 1865 (2018) 1846–1856

access to a concealed Ca2+ pool, Cell Calcium 48 (2010) 143–149. replication of Toxoplasma gondii, PLoS Pathog. 4 (2008) e10.
[37] M.A. Hortua Triana, K. Marquez-Nogueras, L. Chang, A.J. Stasic, C. Li, K. Spiegel, [67] J. Hartmann, K. Hu, C.Y. He, L. Pelletier, D.S. Roos, G. Warren, Golgi and cen-
A. Sharma, Z.H. Li, S.N.J. Moreno, Tagging of weakly expressed toxoplasma gondii trosome cycles in Toxoplasma gondii, Mol. Biochem. Parasitol. 145 (2006)
calcium-related genes with high-affinity tags, J. Eukaryot. Microbiol. (2018) (In 125–127.
press). [68] V. Polonais, B. Javier Foth, K. Chinthalapudi, J.B. Marq, D.J. Manstein, D. Soldati-
[38] Q.C. Wang, Q. Zheng, H. Tan, B. Zhang, X. Li, Y. Yang, J. Yu, Y. Liu, H. Chai, Favre, K. Frenal, Unusual anchor of a motor complex (MyoD-MLC2) to the plasma
X. Wang, Z. Sun, J.Q. Wang, S. Zhu, F. Wang, M. Yang, C. Guo, H. Wang, Q. Zheng, membrane of Toxoplasma gondii, Traffic 12 (2011) 287–300.
Y. Li, Q. Chen, A. Zhou, T.S. Tang, TMCO1 is an ER Ca(2+) load-activated Ca(2+) [69] S. Egarter, N. Andenmatten, A.J. Jackson, J.A. Whitelaw, G. Pall, J.A. Black,
channel, Cell 165 (2016) 1454–1466. D.J. Ferguson, I. Tardieux, A. Mogilner, M. Meissner, The toxoplasma Acto-MyoA
[39] R. Docampo, W. de Souza, K. Miranda, P. Rohloff, S.N. Moreno, Acidocalcisomes - motor complex is important but not essential for gliding motility and host cell
conserved from bacteria to man, Nat. Rev. Microbiol. 3 (2005) 251–261. invasion, PLoS One 9 (2014) e91819.
[40] S. Luo, M. Vieira, J. Graves, L. Zhong, S.N. Moreno, A plasma membrane-type Ca [70] T. Nebl, J.H. Prieto, E. Kapp, B.J. Smith, M.J. Williams, J.R. Yates 3rd,
(2+)-ATPase co-localizes with a vacuolar H(+)-pyrophosphatase to acidocalci- A.F. Cowman, C.J. Tonkin, Quantitative in vivo analyses reveal calcium-depen-
somes of toxoplasma gondii, EMBO J. 20 (2001) 55–64. dent phosphorylation sites and identifies a novel component of the toxoplasma
[41] A.E. Vercesi, C.O. Rodrigues, S.A. Uyemura, L. Zhong, S.N. Moreno, Respiration invasion motor complex, PLoS Pathog. 7 (2011) e1002222.
and oxidative phosphorylation in the apicomplexan parasite Toxoplasma gondii, J. [71] M.J. Williams, H. Alonso, M. Enciso, S. Egarter, L. Sheiner, M. Meissner,
Biol. Chem. 273 (1998) 31040–31047. B. Striepen, B.J. Smith, C.J. Tonkin, Two essential light chains regulate the MyoA
[42] A.G. Bick, S.E. Calvo, V.K. Mootha, Evolutionary diversity of the mitochondrial lever arm to promote toxoplasma gliding motility, MBio 6 (2015) e00845.
calcium uniporter, Science 336 (2012) 886. [72] C.J. Powell, M.L. Jenkins, M.L. Parker, R. Ramaswamy, A. Kelsen, D.M. Warshaw,
[43] A. Rotmann, C. Sanchez, A. Guiguemde, P. Rohrbach, A. Dave, N. Bakouh, G.E. Ward, J.E. Burke, M.J. Boulanger, Dissecting the molecular assembly of the
G. Planelles, M. Lanzer, PfCHA is a mitochondrial divalent cation/H+ antiporter Toxoplasma gondii MyoA motility complex, J. Biol. Chem. 292 (2017)
in Plasmodium falciparum, Mol. Microbiol. 76 (2010) 1591–1606. 19469–19477.
[44] D.S. Guttery, J.K. Pittman, K. Frenal, B. Poulin, L.R. McFarlane, K. Slavic, [73] F. Rusnak, P. Mertz, Calcineurin: form and function, Physiol. Rev. 80 (2000)
S.P. Wheatley, D. Soldati-Favre, S. Krishna, R. Tewari, H.M. Staines, The 1483–1521.
Plasmodium berghei Ca(2+)/H(+) exchanger, PbCAX, is essential for tolerance to [74] A. Farrell, S. Thirugnanam, A. Lorestani, J.D. Dvorin, K.P. Eidell, D.J. Ferguson,
environmental Ca(2+) during sexual development, PLoS Pathog. 9 (2013) B.R. Anderson-White, M.T. Duraisingh, G.T. Marth, M.J. Gubbels, A DOC2 protein
e1003191. identified by mutational profiling is essential for apicomplexan parasite exocy-
[45] R. Docampo, J. Lukes, Trypanosomes and the solution to a 50-year mitochondrial tosis, Science 335 (2012) 218–221.
calcium mystery, Trends Parasitol. 28 (2012) 31–37. [75] J.F. Harper, A. Harmon, Plants, symbiosis and parasites: a calcium signalling
[46] H.I. Wiersma, S.E. Galuska, F.M. Tomley, L.D. Sibley, P.A. Liberator, R.G. Donald, connection, Nat. Rev. Mol. Cell Biol. 6 (2005) 555–566.
A role for coccidian cGMP-dependent protein kinase in motility and invasion, Int. [76] A.K. Wernimont, J.D. Artz, P. Finerty Jr., Y.H. Lin, M. Amani, A. Allali-Hassani,
J. Parasitol. 34 (2004) 369–380. G. Senisterra, M. Vedadi, W. Tempel, F. Mackenzie, I. Chau, S. Lourido,
[47] M.S. Eaton, L.M. Weiss, K. Kim, Cyclic nucleotide kinases and tachyzoite-brady- L.D. Sibley, R. Hui, Structures of apicomplexan calcium-dependent protein kinases
zoite transition in Toxoplasma gondii, Int. J. Parasitol. 36 (2006) 107–114. reveal mechanism of activation by calcium, Nat. Struct. Mol. Biol. 17 (2010)
[48] S. Lourido, J. Shuman, C. Zhang, K.M. Shokat, R. Hui, L.D. Sibley, Calcium-de- 596–601.
pendent protein kinase 1 is an essential regulator of exocytosis in Toxoplasma, [77] S.S. Hook, A.R. Means, Ca(2+)/CaM-dependent kinases: from activation to
Nature 465 (2010) 359–362. function, Annu. Rev. Pharmacol. Toxicol. 41 (2001) 471–505.
[49] J.M. McCoy, L. Whitehead, G.G. van Dooren, C.J. Tonkin, TgCDPK3 regulates [78] A.C. Harmon, M. Gribskov, J.F. Harper, CDPKs - a kinase for every Ca2+ signal?
calcium-dependent egress of Toxoplasma gondii from host cells, PLoS Pathog. 8 Trends Plant Sci. 5 (2000) 154–159.
(2012) e1003066. [79] K.K. Ojo, E.T. Larson, K.R. Keyloun, L.J. Castaneda, A.E. Derocher, K.K. Inampudi,
[50] E. Garrison, M. Treeck, E. Ehret, H. Butz, T. Garbuz, B.P. Oswald, M. Settles, J.E. Kim, T.L. Arakaki, R.C. Murphy, L. Zhang, A.J. Napuli, D.J. Maly,
J. Boothroyd, G. Arrizabalaga, A forward genetic screen reveals that calcium-de- C.L. Verlinde, F.S. Buckner, M. Parsons, W.G. Hol, E.A. Merritt, W.C. van Voorhis,
pendent protein kinase 3 regulates egress in Toxoplasma, PLoS Pathog. 8 (2012) Toxoplasma gondii calcium-dependent protein kinase 1 is a target for selective
e1003049. kinase inhibitors, Nat. Struct. Mol. Biol. 17 (2010) 602–607.
[51] S. Lourido, K. Tang, L.D. Sibley, Distinct signalling pathways control Toxoplasma [80] J.R. Ingram, K.E. Knockenhauer, B.M. Markus, J. Mandelbaum, A. Ramek, Y. Shan,
egress and host-cell invasion, EMBO J. 31 (2012) 4524–4534. D.E. Shaw, T.U. Schwartz, H.L. Ploegh, S. Lourido, Allosteric activation of api-
[52] Y. Jia, J.B. Marq, H. Bisio, D. Jacot, C. Mueller, L. Yu, J. Choudhary, M. Brochet, complexan calcium-dependent protein kinases, Proc. Natl. Acad. Sci. U. S. A. 112
D. Soldati-Favre, Crosstalk between PKA and PKG controls pH-dependent host cell (2015) E4975–E4984.
egress of Toxoplasma gondii, EMBO J. 36 (2017) 3250–3267. [81] T. Sugi, K. Kato, K. Kobayashi, S. Watanabe, H. Kurokawa, H. Gong, K. Pandey,
[53] A.D. Uboldi, M.L. Wilde, E.A. McRae, R.J. Stewart, L.F. Dagley, L. Yang, H. Takemae, H. Akashi, Use of the kinase inhibitor analog 1NM-PP1 reveals a role
N.J. Katris, S.V. Hapuarachchi, M.J. Coffey, A.M. Lehane, C.Y. Botte, R.F. Waller, for Toxoplasma gondii CDPK1 in the invasion step, Eukaryot. Cell 9 (2010)
A.I. Webb, C.J. Tonkin, Protein kinase A negatively regulates Ca2+ signaling 1 in 667–670.
Toxoplasma gondii, bioRxiv (2018), https://doi.org/10.1101/265371. [82] S. Lourido, C. Zhang, M.S. Lopez, K. Tang, J. Barks, Q. Wang, S.A. Wildman,
[54] D. Chin, A.R. Means, Calmodulin: a prototypical calcium sensor, Trends Cell Biol. K.M. Shokat, L.D. Sibley, Optimizing small molecule inhibitors of calcium-de-
10 (2000) 322–328. pendent protein kinase 1 to prevent infection by Toxoplasma gondii, J. Med.
[55] A.P. Yamniuk, M. Rainaldi, H.J. Vogel, Calmodulin has the potential to function as Chem. 56 (2013) 3068–3077.
a Ca-dependent adaptor protein, Plant Signal. Behav. 2 (2007) 354–357. [83] R.C. Murphy, K.K. Ojo, E.T. Larson, A. Castellanos-Gonzalez, B.G. Perera,
[56] F. Seeber, B. Beuerle, H.H. Schmidt, Cloning and functional expression of the K.R. Keyloun, J.E. Kim, J.G. Bhandari, N.R. Muller, C.L. Verlinde, A.C. White Jr.,
calmodulin gene from Toxoplasma gondii, Mol. Biochem. Parasitol. 99 (1999) E.A. Merritt, W.C. van Voorhis, D.J. Maly, Discovery of potent and selective in-
295–299. hibitors of calcium-dependent protein kinase 1 (CDPK1) from C. Parvum and T.
[57] N. Pezzella-D'Alessandro, H. Le Moal, A. Bonhomme, A. Valere, C. Klein, J. Gomez- Gondii, ACS Med. Chem. Lett. 1 (2010) 331–335.
Marin, J.M. Pinon, Calmodulin distribution and the actomyosin cytoskeleton in [84] S.M. Johnson, R.C. Murphy, J.A. Geiger, A.E. DeRocher, Z. Zhang, K.K. Ojo,
toxoplasma gondii, J. Histochem. Cytochem. 49 (2001) 445–454. E.T. Larson, B.G. Perera, E.J. Dale, P. He, M.C. Reid, A.M. Fox, N.R. Mueller,
[58] A.S. Paul, S. Saha, K. Engelberg, R.H. Jiang, B.I. Coleman, A.L. Kosber, C.T. Chen, E.A. Merritt, E. Fan, M. Parsons, W.C. van Voorhis, D.J. Maly, Development of
M. Ganter, N. Espy, T.W. Gilberger, M.J. Gubbels, M.T. Duraisingh, Parasite Toxoplasma gondii calcium-dependent protein kinase 1 (TgCDPK1) inhibitors
Calcineurin regulates host cell recognition and attachment by apicomplexans, Cell with potent anti-toxoplasma activity, J. Med. Chem. 55 (2012) 2416–2426.
Host Microbe 18 (2015) 49–60. [85] E. Moine, I. Dimier-Poisson, C. Enguehard-Gueiffier, C. Loge, M. Penichon,
[59] R.F. Waller, G.I. McFadden, The apicoplast: a review of the derived plastid of N. Moire, C. Delehouze, B. Foll-Josselin, S. Ruchaud, S. Bach, A. Gueiffier,
apicomplexan parasites, Curr. Issues Mol. Biol. 7 (2005) 57–79. F. Debierre-Grockiego, C. Denevault-Sabourin, Development of new highly potent
[60] A. Perochon, D. Aldon, J.P. Galaud, B. Ranty, Calmodulin and calmodulin-like imidazo[1,2-b]pyridazines targeting Toxoplasma gondii calcium-dependent pro-
proteins in plant calcium signaling, Biochimie 93 (2011) 2048–2053. tein kinase 1, Eur. J. Med. Chem. 105 (2015) 80–105.
[61] N. Bouche, A. Yellin, W.A. Snedden, H. Fromm, Plant-specific calmodulin-binding [86] F.U. Rutaganira, J. Barks, M.S. Dhason, Q. Wang, M.S. Lopez, S. Long, J.B. Radke,
proteins, Annu. Rev. Plant Biol. 56 (2005) 435–466. N.G. Jones, A.R. Maddirala, J.W. Janetka, M. El Bakkouri, R. Hui, K.M. Shokat,
[62] S. Long, K.M. Brown, L.L. Drewry, B. Anthony, I.Q.H. Phan, L.D. Sibley, L.D. Sibley, Inhibition of calcium dependent protein kinase 1 (CDPK1) by
Calmodulin-like proteins localized to the conoid regulate motility and cell inva- Pyrazolopyrimidine analogs decreases establishment and reoccurrence of central
sion by Toxoplasma gondii, PLoS Pathog. 13 (2017) e1006379. nervous system disease by Toxoplasma gondii, J. Med. Chem. 60 (2017)
[63] A. Graindorge, K. Frénal, D. Jacot, J. Salamun, J.B. Marq, D. Soldati-Favre, The 9976–9989.
conoid associated motor MyoH is indispensable for Toxoplasma gondii entry and [87] E.M. Cardew, C. Verlinde, E. Pohl, The calcium-dependent protein kinase 1 from
exit from host cells, PLoS Pathog. 12 (2016) e1005388. Toxoplasma gondii as target for structure-based drug design, Parasitology 145
[64] S. Long, B. Anthony, L.L. Drewry, L.D. Sibley, A conserved ankyrin repeat-con- (2018) 210–218.
taining protein regulates conoid stability, motility and cell invasion in Toxoplasma [88] J. Morlon-Guyot, L. Berry, C.T. Chen, M.J. Gubbels, M. Lebrun, W. Daher, The
gondii, Nat. Commun. 8 (2017) 2236. Toxoplasma gondii calcium-dependent protein kinase 7 is involved in early steps
[65] N. Pezzella, A. Bouchot, A. Bonhomme, L. Pingret, C. Klein, H. Burlet, G. Balossier, of parasite division and is crucial for parasite survival, Cell. Microbiol. 16 (2014)
P. Bonhomme, J.M. Pinon, Involvement of calcium and calmodulin in Toxoplasma 95–114.
gondii tachyzoite invasion, Eur. J. Cell Biol. 74 (1997) 92–101. [89] S. Long, Q. Wang, L.D. Sibley, Analysis of noncanonical calcium-dependent pro-
[66] K. Hu, Organizational changes of the daughter basal complex during the parasite tein kinases in Toxoplasma gondii by targeted gene deletion using CRISPR/Cas9,

1855
M.A. Hortua Triana et al. BBA - Molecular Cell Research 1865 (2018) 1846–1856

Infect. Immun. 84 (2016) 1262–1273. cGMP dependent protein kinase, Mol. Biochem. Parasitol. 120 (2002) 165–175.
[90] M. Treeck, J.L. Sanders, R.Y. Gaji, K.A. LaFavers, M.A. Child, G. Arrizabalaga, [115] R.G. Donald, J. Allocco, S.B. Singh, B. Nare, S.P. Salowe, J. Wiltsie, P.A. Liberator,
J.E. Elias, J.C. Boothroyd, The calcium-dependent protein kinase 3 of Toxoplasma Toxoplasma gondii cyclic GMP-dependent kinase: chemotherapeutic targeting of
influences basal calcium levels and functions beyond egress as revealed by an essential parasite protein kinase, Eukaryot. Cell 1 (2002) 317–328.
quantitative phosphoproteome analysis, PLoS Pathog. 10 (2014) e1004197. [116] K.M. Brown, S. Long, L.D. Sibley, Plasma membrane association by N-acylation
[91] D. Jacot, K. Frenal, J.B. Marq, P. Sharma, D. Soldati-Favre, Assessment of phos- governs PKG function in toxoplasma gondii, MBio 8 (2017).
phorylation in Toxoplasma glideosome assembly and function, Cell. Microbiol. 16 [117] S.M. Sidik, M.A. Hortua Triana, A.S. Paul, M. El Bakkouri, C.G. Hackett, F. Tran,
(2014) 1518–1532. N.J. Westwood, R. Hui, W.J. Zuercher, M.T. Duraisingh, S.N. Moreno, S. Lourido,
[92] M. Meissner, D. Schlüter, D. Soldati, Role of Toxoplasma gondii myosin A in Using a genetically encoded sensor to identify inhibitors of toxoplasma gondii
powering parasite gliding and host cell invasion, Science (New York, N.Y.) 298 Ca2+ signaling, J. Biol. Chem. 291 (2016) 9566–9580.
(2002) 837–840. [118] J.T. Lock, I. Parker, I.F. Smith, A comparison of fluorescent Ca2+ indicators for
[93] Q. Tang, N. Andenmatten, M.A. Hortua Triana, B. Deng, M. Meissner, S.N. Moreno, imaging local Ca2+ signals in cultured cells, Cell Calcium 58 (2015) 638–648.
B.A. Ballif, G.E. Ward, Calcium-dependent phosphorylation alters class XIVa [119] M.F. Cesbron-Delauw, B. Guy, G. Torpier, R.J. Pierce, G. Lenzen, J.Y. Cesbron,
myosin function in the protozoan parasite Toxoplasma gondii, Mol. Biol. Cell 25 H. Charif, P. Lepage, F. Darcy, J.P. Lecocq, et al., Molecular characterization of a
(2014) 2579–2591. 23-kilodalton major antigen secreted by Toxoplasma gondii, Proc. Natl. Acad. Sci.
[94] R.Y. Gaji, D.E. Johnson, M. Treeck, M. Wang, A. Hudmon, G. Arrizabalaga, U. S. A. 86 (1989) 7537–7541.
Phosphorylation of a myosin motor by TgCDPK3 facilitates rapid initiation of [120] L.D. Sibley, I.R. Niesman, S.F. Parmley, M.F. Cesbron-Delauw, Regulated secretion
motility during toxoplasma gondii egress, PLoS Pathog. 11 (2015). of multi-lamellar vesicles leads to formation of a tubulo-vesicular network in host-
[95] R.J. Stewart, L. Whitehead, B. Nijagal, B.E. Sleebs, G. Lessene, M.J. McConville, cell vacuoles occupied by Toxoplasma gondii, J. Cell Sci. 108 (Pt 4) (1995)
K.L. Rogers, C.J. Tonkin, Analysis of Ca(2)(+) mediated signaling regulating 1669–1677.
Toxoplasma infectivity reveals complex relationships between key molecules, Cell. [121] D.J. Hupe, E.R. Pfefferkorn, N.D. Behrens, K. Peters, L-651,582 inhibition of in-
Microbiol. 19 (2017). tracellular parasitic protozoal growth correlates with host-cell directed effects, J.
[96] J.M. McCoy, R.J. Stewart, A.D. Uboldi, D. Li, J. Schroder, N.E. Scott, Pharmacol. Exp. Ther. 256 (1991) 462–467.
A.T. Papenfuss, A.M. Lehane, L.J. Foster, C.J. Tonkin, A forward genetic screen [122] A. Bonhomme, L. Pingret, P. Bonhomme, J. Michel, G. Balossier, M. Lhotel,
identifies a negative regulator of rapid Ca(2+)-dependent cell egress (MS1) in the M. Pluot, J.M. Pinon, Subcellular calcium localization in toxoplasma gondii by
intracellular parasite Toxoplasma gondii, J. Biol. Chem. 292 (2017) 7662–7674. electron microscopy and by X-ray and electron energy loss spectroscopies,
[97] D.M. Wetzel, L.A. Chen, F.A. Ruiz, S.N. Moreno, L.D. Sibley, Calcium-mediated Microsc. Res. Tech. 25 (1993) 276–285.
protein secretion potentiates motility in Toxoplasma gondii, J. Cell Sci. 117 (2004) [123] L. Borges-Pereira, A. Budu, C.A. McKnight, C.A. Moore, S.A. Vella, M.A. Hortua
5739–5748. Triana, J. Liu, C.R. Garcia, D.A. Pace, S.N. Moreno, Calcium signaling throughout
[98] R. Mondragon, E. Frixione, Ca(2+)-dependence of conoid extrusion in the toxoplasma gondii lytic cycle: a study using genetically encoded calcium in-
Toxoplasma gondii tachyzoites, J. Eukaryot. Microbiol. 43 (1996) 120–127. dicators, J. Biol. Chem. 290 (2015) 26914–26926.
[99] V.B. Carruthers, L.D. Sibley, Mobilization of intracellular calcium stimulates mi- [124] K.A. LaFavers, K.M. Marquez-Nogueras, I. Coppens, S.N.J. Moreno,
croneme discharge in Toxoplasma gondii, Mol. Microbiol. 31 (1999) 421–428. G. Arrizabalaga, A novel dense granule protein, GRA41, regulates timing of egress
[100] M.G. Del Carmen, M. Mondragon, S. Gonzalez, R. Mondragon, Induction and and calcium sensitivity in Toxoplasma gondii, Cell Microbiol. 19 (2017).
regulation of conoid extrusion in Toxoplasma gondii, Cell. Microbiol. 11 (2009) [125] K. Frenal, J.F. Dubremetz, M. Lebrun, D. Soldati-Favre, Gliding motility powers
967–982. invasion and egress in Apicomplexa, Nat. Rev. Microbiol. 15 (2017) 645–660.
[101] M.C. Vieira, S.N. Moreno, Mobilization of intracellular calcium upon attachment [126] T. Endo, K.K. Sethi, G. Piekarski, Toxoplasma gondii: calcium ionophore A23187-
of Toxoplasma gondii tachyzoites to human fibroblasts is required for invasion, mediated exit of trophozoites from infected murine macrophages, Exp. Parasitol.
Mol. Biochem. Parasitol. 106 (2000) 157–162. 53 (1982) 179–188.
[102] N.J. Katris, G.G. van Dooren, P.J. McMillan, E. Hanssen, L. Tilley, R.F. Waller, The [127] A. Kuchipudi, R.D. Arroyo-Olarte, F. Hoffmann, V. Brinkmann, N. Gupta,
apical complex provides a regulated gateway for secretion of invasion factors in Optogenetic monitoring identifies phosphatidylthreonine-regulated calcium
Toxoplasma, PLoS Pathog. 10 (2014) e1004074. homeostasis in Toxoplasma gondii, Microb. Cell 3 (2016) 215–223.
[103] V.B. Carruthers, O.K. Giddings, L.D. Sibley, Secretion of micronemal proteins is [128] B.F. Kafsack, J.D. Pena, I. Coppens, S. Ravindran, J.C. Boothroyd, V.B. Carruthers,
associated with toxoplasma invasion of host cells, Cell. Microbiol. 1 (1999) Rapid membrane disruption by a perforin-like protein facilitates parasite exit from
225–235. host cells, Science 323 (2009) 530–533.
[104] V.B. Carruthers, F.M. Tomley, Microneme proteins in apicomplexans, Subcell. [129] K. Nagamune, L.M. Hicks, B. Fux, F. Brossier, E.N. Chini, L.D. Sibley, Abscisic acid
Biochem. 47 (2008) 33–45. controls calcium-dependent egress and development in Toxoplasma gondii, Nature
[105] V.B. Carruthers, S.N. Moreno, L.D. Sibley, Ethanol and acetaldehyde elevate in- 451 (2008) 207–210.
tracellular [Ca2+] and stimulate microneme discharge in Toxoplasma gondii, [130] G. Arrizabalaga, F. Ruiz, S. Moreno, J.C. Boothroyd, Ionophore-resistant mutant of
Biochem. J. 342 (Pt 2) (1999) 379–386. Toxoplasma gondii reveals involvement of a sodium/hydrogen exchanger in cal-
[106] R. Moudy, T.J. Manning, C.J. Beckers, The loss of cytoplasmic potassium upon cium regulation, J. Cell Biol. 165 (2004) 653–662.
host cell breakdown triggers egress of Toxoplasma gondii, J. Biol. Chem. 276 [131] S. Hakansson, H. Morisaki, J. Heuser, L.D. Sibley, Time-lapse video microscopy of
(2001) 41492–41501. gliding motility in Toxoplasma gondii reveals a novel, biphasic mechanism of cell
[107] M.S. Roiko, N. Svezhova, V.B. Carruthers, Acidification activates toxoplasma locomotion, Mol. Biol. Cell 10 (1999) 3539–3547.
gondii motility and egress by enhancing protein secretion and Cytolytic activity, [132] J.M. Leung, M.A. Rould, C. Konradt, C.A. Hunter, G.E. Ward, Disruption of
PLoS Pathog. 10 (2014) e1004488. TgPHIL1 alters specific parameters of Toxoplasma gondii motility measured in a
[108] K.M. Brown, S. Lourido, L.D. Sibley, Serum albumin stimulates protein kinase G- quantitative, three-dimensional live motility assay, PLoS One 9 (2014) e85763.
dependent Microneme secretion in toxoplasma gondii, J. Biol. Chem. 291 (2016) [133] J.L. Lovett, L.D. Sibley, Intracellular calcium stores in Toxoplasma gondii govern
9554–9565. invasion of host cells, J. Cell Sci. 116 (2003) 3009–3016.
[109] S. Luo, F.A. Ruiz, S.N. Moreno, The acidocalcisome Ca2+-ATPase (TgA1) of [134] J.M. Dobrowolski, V.B. Carruthers, L.D. Sibley, Participation of myosin in gliding
toxoplasma gondii is required for polyphosphate storage, intracellular calcium motility and host cell invasion by Toxoplasma gondii, Mol. Microbiol. 26 (1997)
homeostasis and virulence, Mol. Microbiol. 55 (2005) 1034–1045. 163–173.
[110] U. Eckstein-Ludwig, R.J. Webb, I.D. Van Goethem, J.M. East, A.G. Lee, M. Kimura, [135] T. Tomita, T. Yamada, L.M. Weiss, A. Orlofsky, Externally triggered egress is the
P.M. O'Neill, P.G. Bray, S.A. Ward, S. Krishna, Artemisinins target the SERCA of major fate of Toxoplasma gondii during acute infection, J. Immunol. 183 (2009)
Plasmodium falciparum, Nature 424 (2003) 957–961. 6667–6680.
[111] K. Nagamune, S.N. Moreno, L.D. Sibley, Artemisinin-resistant mutants of tox- [136] A.A. Koshy, H.K. Dietrich, D.A. Christian, J.H. Melehani, A.J. Shastri, C.A. Hunter,
oplasma gondii have altered calcium homeostasis, Antimicrob. Agents Chemother. J.C. Boothroyd, Toxoplasma co-opts host cells it does not invade, PLoS Pathog. 8
51 (2007) 3816–3823. (2012) e1002825.
[112] C.I. Hall, M.L. Reese, E. Weerapana, M.A. Child, P.W. Bowyer, V.E. Albrow, [137] M.B. Heintzelman, Gliding motility in apicomplexan parasites, Semin. Cell Dev.
J.D. Haraldsen, M.R. Phillips, E.D. Sandoval, G.E. Ward, B.F. Cravatt, Biol. 46 (2015) 135–142.
J.C. Boothroyd, M. Bogyo, Chemical genetic screen identifies toxoplasma DJ-1 as a [138] R.V. Stadler, L.A. White, K. Hu, B.P. Helmke, W.H. Guilford, Direct measurement
regulator of parasite secretion, attachment, and invasion, Proc. Natl. Acad. Sci. U. of cortical force generation and polarization in a living parasite, Mol. Biol. Cell 28
S. A. 108 (2011) 10568–10573. (2017) 1912–1923.
[113] M.A. Child, M. Garland, I. Foe, P. Madzelan, M. Treeck, W.A. van der Linden, [139] C.S. Bookwalter, A. Kelsen, J.M. Leung, G.E. Ward, K.M. Trybus, A Toxoplasma
K. Oresic Bender, E. Weerapana, M.A. Wilson, J.C. Boothroyd, M.L. Reese, gondii class XIV myosin, expressed in Sf9 cells with a parasite co-chaperone, re-
M. Bogyo, Toxoplasma DJ-1 regulates organelle secretion by a direct interaction quires two light chains for fast motility, J. Biol. Chem. 289 (2014) 30832–30841.
with calcium-dependent protein kinase 1, MBio 8 (2017). [140] M. Melchionda, J.K. Pittman, R. Mayor, S. Patel, Ca2+/H+ exchange by acidic
[114] R.G. Donald, P.A. Liberator, Molecular characterization of a coccidian parasite organelles regulates cell migration in vivo, J. Cell Biol. 212 (2016) 803–813.

1856

You might also like