You are on page 1of 21

Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

Contents lists available at ScienceDirect

Neuroscience and Biobehavioral Reviews


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

Review article

Toxoplasmosis: A pathway to neuropsychiatric disorders T


a,b,c,⁎ a,b c,d a,b
Shiraz Tyebji , Simona Seizova , Anthony J. Hannan , Christopher J. Tonkin
a
The Walter and Eliza Hall Institute of Medical Research, Melbourne, 3052, Australia
b
Department of Medical Biology, The University of Melbourne, Melbourne, 3052, Australia
c
Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, Victoria, Australia
d
Department of Anatomy and Neuroscience, University of Melbourne, Parkville, 3052, Victoria, Australia

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

Keywords: Toxoplasma gondii is an obligate intracellular parasite that resides, in a latent form, in the human central nervous
Toxoplasma gondii system. Infection with Toxoplasma drastically alters the behaviour of rodents and is associated with the incidence
Neuroimmune of specific neuropsychiatric conditions in humans. But the question remains: how does this pervasive human
Neuropathology pathogen alter behaviour of the mammalian host? This fundamental question is receiving increasing attention as
Transgenerational
it has far reaching public health implications for a parasite that is very common in human populations. Our
Behavioural manipulations
Central nervous system
current understanding centres on neuronal changes that are elicited directly by this intracellular parasite versus
Inflammation indirect changes that occur due to activation of the immune system within the CNS, or a combination of both. In
this review, we explore the interactions between Toxoplasma and its host, the proposed mechanisms and con-
sequences on neuronal function and mental health, and discuss Toxoplasma infection as a public health issue.

1. Introduction tendency to develop mental health issues in life, such as malnutrition,


exposure to heavy metals, urban living, socio-economic situation,
When the genetic architecture of complex human diseases was un- stress, drug abuse etc. (Uher and Zwicker, 2017). Apart from external
covered by the genome-wide association studies, scientists quickly factors, the effect of gut flora on brain function has also revealed a bi-
realized that the expected Mendelian mechanism of inheritance cannot directional gut-brain cross-talk that affects a person’s mood, motiva-
be applied to many of the complex human traits and disorders observed tion, cognitive abilities as well as decision making. Perhaps the most
in the population (Maher, 2008; Manolio et al., 2009). This was re- intriguing and poorly understood link in the aetiology of mental health
ferred to as the ‘missing heritability’, which may involve a wide range is the effect of infections on brain function.
of genetic, environmental and epigenetic factors (Eichler et al., 2010; In 1979, it was discovered that infection of rodents with a ubiqui-
Hannan, 2010; Yeshurun and Hannan, 2018). In addition to rapid tous human parasite, Toxoplasma gondii, had drastic effects on learning
progress in human genetics, various diseases, including mental health performance. Toxoplasma gondii is an obligate intracellular eukaryotic
disorders, are increasingly being associated with specific environmental parasite of the phylum Apicomplexa. Worldwide, 30%–80% of the
exposures and perturbations surrounding an individual. Researchers human population is known to carry the infection, which is acquired via
have now identified numerous factors that could impact a person’s the consumption of contaminated water and food, or via vertical

Abbreviations: AAH, aromatic amino acid hydrolase; AD, Alzheimer’s disease; ADHD, attention deficit hyperactivity disorder; AMPA, α-amino-3-hydroxy-5-methyl-
4-isoxazolepropionic acid; APP, amyloid precursor protein; Aß, beta-amyloid; BBB, blood brain barrier; BD, bipolar disorder; BLA, basolateral amygdala; CNS, central
nervous system; CORT, corticosterone; DC, dendritic cells; DG, dense granules; EAE, autoimmune encephalomyelitis; EEG, electroencephalography; EM, electron
microscopy; GABA, gamma-aminobutyric acid; GAD67, glutamic acid decarboxylase 67; GAS, interferon gamma activated sequence; GLT-1, glutamate transporter-1;
HD, Huntington’s disease; HPA, hypothalamic-pituitary-adrenal; IDO, indoleamine 2,3-dioxygenase; IFNγ, interferon-gamma; IST, inhibitor of STAT1 transcriptional
activity; KA, kynurenic acid; KO, knock-out; MAP2, microtubule associated protein-2; MAPK, mitogen-activated protein kinase; MD, major depression; mGluR,
metabotropic glutamate receptor; miR-132, microRNA-132; MRI, magnetic resonance imaging; MS, multiple sclerosis; NK, natural killer cells; NMDA, N-methyl-d-
aspartic acid; NMDARs, NMDA receptors; NO, nitric oxide; NOS, nitric oxide synthase; PD, Parkinson’s disease; PSD-95, post-synaptic density protein-95; PSEN,
presenilin protein -1; PV, parasitophorous vacuole; PVM, parasitophorous vacuole membrane; QA, quinolinic acid; STAT1, signal transducer and activator of
transcription 1; SZ, schizophrenia; TBI, traumatic brain injury; TE, toxoplasma encephalitis; TH, tyrosine hydroxylase; VGLUT1, vesicular glutamate transporter 1;
WT, wild-type

Corresponding author at: The Walter and Eliza Hall Institute of Medical Research, 1G, Royal Parade, Parkville, Melbourne, 3052, Australia.
E-mail addresses: tyebji.s@wehi.edu.au (S. Tyebji), seizova.s@wehi.edu.au (S. Seizova), anthony.hannan@florey.edu.au (A.J. Hannan),
tonkin@wehi.edu.au (C.J. Tonkin).

https://doi.org/10.1016/j.neubiorev.2018.11.012
Received 25 May 2018; Received in revised form 23 October 2018; Accepted 22 November 2018
Available online 23 November 2018
0149-7634/ © 2018 Elsevier Ltd. All rights reserved.
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

transmission during pregnancy (Montoya and Liesenfeld, 2004; Tenter meaning that it must invade host cells to replicate. Members of the
et al., 2000). Acute infection spreads throughout the body and presents Felidae family are definitive hosts for Toxoplasma, wherein it undergoes
as mild flu-like symptoms, which is largely controlled by a healthy the sexual stage of its lifecycle within the gut. Fecal shedding leads to
immune system. Upon infection, acute-stage tachyzoites differentiates environmental contamination of water and food sources, causing acute
into latent bradyzoite forms which reside in the muscle and CNS, and infection in warm-blooded animals, including humans, when consumed
are resistant to immune clearance thus leading to life-long chronic in- (Dubey, 2009). During the acute stage, tachyzoites, the fast-replicating
fection. Whilst initially thought to be clinically silent, latent Toxoplasma form of the parasite, go through cycles of invasion, replication and
is now known to cause changes in learning and memory, locomotion, egress, destroying the host cells in the process. Throughout its lifecycle,
anxiety, depression-like behaviours and novelty preference in rodents the parasite secrets multiple parasite proteins into the host cells that
(Table 1; Worth et al., 2014). However, the most fascinating effect of ensure its survival within the host (see Section 9). These secreted pro-
Toxoplasma on rodent behaviour is its ability to cause rodents to lose teins protect immune killing of Toxoplasma and allow it to persist and
their natural aversion to cat odours (Berdoy et al., 2000; Vyas et al., hijack monocytes causing them to become hypermigratory, enabling
2007). This is especially interesting as Toxoplasma is transmitted by dissemination from the site of infection (the gut) and spread throughout
cats, thus suggesting that this cunning intracellular parasite may ma- the body (Hammoudi et al., 2015; Kanatani et al., 2017; Lambert et al.,
nipulate its host for transmission to its definitive host. How Toxoplasma 2006; Weidner et al., 2016, 2013; Weidner and Barragan, 2014). Tox-
achieves this in a way that it undergoes minimal damage, avoiding oplasma then undergoes differentiation into a slow growing encysted
lethal outcome, is the focus of current research. form (bradyzoites) within cells of the muscle and CNS tissue (Dubey,
It has now been shown that Toxoplasma infection is associated with 2008).
several neuropsychiatric conditions thus posing interesting questions An area of active research is understanding how Toxoplasma is able
regarding how the biology of the parasite and its interactions with the to cross blood-brain barrier (BBB). Three models have been proposed:
brain and immune system could contribute to mental health disorders. paracellular entry, transcellular migration, and infiltration of infected
The mere association of seropositivity to Toxoplasma with the devel- immune cells (Fig. 1). The ability of Toxoplasma to cross the gut epi-
opment of mental disorders in humans does not answer critical ques- thelia via a paracellular pathway suggests that a similar means may be
tions such as: is Toxoplasma the cause of neurological and psychiatric applied to cross comparable barriers, such as the BBB (Barragan and
problems? Does Toxoplasma exacerbate the disease onset/symptoms in Sibley, 2002; Harker et al., 2014). Alternatively, it was demonstrated
susceptible individuals? What role does the parasite itself play in causing that tachyzoites are able to invade and replicate inside the endothelial
these symptoms? cells of the BBB, finally bursting the cells and crossing over into the
To date, studies evaluating the direct effects of Toxoplasma on brain parenchyma (Konradt et al., 2016). Lastly, a ‘Trojan horse’ me-
neurons and their associated neural functions have provided little chanism has also been suggested; wherein acutely infected monocytes
concrete evidence of cellular mechanisms that go awry in the presence cross the BBB, delivering the parasites into the brain parenchyma
of this parasite, and even less that prove it is a direct result of parasitic (Lachenmaier et al., 2011; Lambert et al., 2006; Ueno et al., 2014). In
activity. In an attempt to understand the effects of Toxoplasma on host reality, it’s likely that a combination of these strategies is used by the
behaviour, murine models of Toxoplasmosis have been developed parasite to gain entry into the CNS (Fig. 1).
which have provided us with a good snapshot of the plethora of cog-
nitive changes caused as a result of infection (Worth et al., 2014). Apart 2.1. Tropism in the CNS
from changes in behaviour, a wide range of deficits have been observed
in the brain at the structural and functional level (Parlog et al., 2015; In the CNS, Toxoplasma has a selective tropism for neuronal cells
Tedford and Mcconkey, 2017). Moreover, recent advancements in un- over resident glial cells and establishes a latent chronic infection by
derstanding the effects of immune activation on brain function have differentiating into encysted bradyzoites (Fig. 1). Early in vitro experi-
indicated that direct parasite invasion of the brain as well as recruit- ments used primary cells from murine and human foetal brains to study
ment of effector molecules from the peripheral immune system in re- the growth of Toxoplasma in astrocytes, microglia, and neurons
sponse to the CNS inflammation can lead to chronic neurological dys- (Contreras-Ochoa et al., 2013; Fischer et al., 1997; Halonen et al., 2001,
function that can persist long after pathogen clearance (Fung et al., 1998, 1996; Jones et al., 1986; Lüder et al., 1999; Peterson et al., 1995,
2017; Klein et al., 2017; Yarovinsky, 2014). This opens up an intriguing 1993). When a mixed culture of rat primary cortical cells were infected
possibility that neurological changes observed upon infection could be with Toxoplasma, intracellular parasites were observed in all cell types;
due to the host CNS altering itself as a defence mechanism against this but as infection progressed, astrocytes and microglia were able to ef-
infection. Interestingly, above studies indicate no single definitive cause ficiently inhibit the growth and replication of the parasites. Subse-
for mental health disorders, genetic or otherwise, suggesting that a quently, vacuoles containing bradyzoites were observed predominantly
combination of external and genetic factors could contribute to such in neurons (Lüder et al., 1999). In studies with co-cultures of only
outcomes. microglia and astrocytes, microglial cells could efficiently eliminate
Although humans are secondary, dead-end hosts of Toxoplasma, its parasites while cysts could be observed in astrocytes (Jones et al., 1986;
effects on human neuropathology are still noteworthy. This review will Peterson et al., 1993; Schlüter et al., 2001). Microglial cells were able to
further propound the idea that the major threat from Toxoplasma to clear parasites due to their intrinsic phagocytosis-associated anti-Tox-
apparently healthy but susceptible individuals is its role as an en- oplasma activity (Jones et al., 1986; Peterson et al., 1993), possibly via
vironmental modifier of disease onset and/or progression. Further, its nitric-oxide (NO) mediated mechanisms (Chao et al., 1993; Gazzinelli
ability to affect the offspring of infected individuals, and its prevalence et al., 1993); while NO production (Peterson et al., 1995) and interferon
in our society, begets an overhaul of our understanding of gamma (IFNγ) signalling (Fischer et al., 1997; Halonen et al., 2001;
Toxoplasmosis as a major public health issue. The indirect and un- Halonen and Weiss, 2000; Jones et al., 1986; Peterson et al., 1993;
detected implications might outweigh the presence of the actual para- Schlüter et al., 2001) inhibits intracellular parasite replication. The
site in its host. Here, we review our current understanding of changes in development of cysts in astrocytes was not dependent on the presence
brain functions upon infection, and link that to the changes observed in of IFNγ, but IFNγ did control the reactivation of tachyzoites that en-
known neuropsychiatric disorders. abled cysts to remain for prolonged periods without rupturing (Jones
et al., 1986). It was recently demonstrated that the ability of IFNγ to
2. Toxoplasma gondii – the organism limit parasite replication was dependent on signal transducer and ac-
tivator of transcription 1 (STAT1) signalling in astrocytes, and deletion
Toxoplasma is a single-celled obligate intracellular parasite, of STAT1 in astrocytes led to an uncontrolled parasite replication

73
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

Fig. 1. Toxoplasma invasion of the brain. To successfully infect the brain and form cysts therein, the first challenge faced by Toxoplasma is to cross the BBB. It could do
so by one or more of the three proposed mechanisms: 1. Invade the epithelial cells of the blood vessel, replicate within and burst out into the brain parenchyma
(transcellular entry); 2. Hijack an immune cell and ride it across the BBB; 3. Adapt a paracellular mode of entry. After entry into the brain, the presence of Toxoplasma
leads to activation of microglia and astrocytes, leading to a heavy immune response that aids parasite clearance (A and B). Such events are thought to be responsible
for the lack of cysts observed in these cell types in vivo. On the other hand, the process of invasion of neurons by Toxoplasma is known to create three neuronal
subpopulations: C, cells that have been injected by rhoptry proteins, but remain uninfected; D, cells that harbor Toxoplasma in their tachyzoite forms; and, E, cells
that harbor stable parasite cysts. It is known that in their tachyzoite stages, Toxoplasma export DG proteins (GRA) across the parasitophorous vacuole membrane into
the host cell, but it’s ability to do the same in the stable cysts stage still remains a mystery. Elucidating if such protein export events occur in the chronic stages of
infection would help us understand mechanisms that lead to changes in neuronal function and behavioural alterations.

74
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

Table 1
A summary of the documented behavioural changes upon infection with Toxoplasma gondii using rodent models. M, male; F, female; n/a, information not available.
Behavioural paradigm Effect Model Sex Toxoplasma strain Reference

Anxiety
Elevated plus maze Reduced Mouse F ME49 (Machado et al., 2016)
No change Rat M Pru (Vyas et al., 2007)
Reduced Rat n/a RH (Gonzalez et al., 2007)
Reduced Mouse F Pru & ME49 (Kannan et al., 2010)
Open field Reduced Mouse F ME49 (Afonso et al., 2012)
Increased Mouse M ME49 (Gatkowska et al., 2012)
Reduced Mouse M HIF (Skallová et al., 2006)
Increased Mouse F HIF (Skallová et al., 2006)
Increased Mouse n/a Rabbit A (Hay et al., 1984)
Recognition memory
Object recognition Reduced Mouse F ME49 (Machado et al., 2016)
Reduced Mouse F GT1 (Xiao et al., 2016b)
No change Mouse M ME49 (Gulinello et al., 2010)
Working Memory
Y-maze, No change Mouse F GT1 (Xiao et al., 2016b)
continuous alternations
No change Mouse n/a ME49 (Jung et al., 2012)
No change Mouse F Pru (Kannan et al., 2010)
Impaired Mouse F ME49 (Kannan et al., 2010)
Spatial Memory
Y-maze, No change Mice F GT1 (Xiao et al., 2016b)
time in novel arm
No change Mouse F Pru (Kannan et al., 2010)
No change Mouse F ME49 (Kannan et al., 2010)
Impaired Mouse n/a Rabbit A (Hay et al., 1984)
Impaired Mouse M N/a (Hutchison et al., 1980)
Morris water maze test No change Mouse n/a ME49 (Jung et al., 2012)
Impaired Rat M PTg(II) (Daniels et al., 2014)
No change Rat M Pru (Vyas et al., 2007)
8-arm radial maze Impaired Mouse F HIF (Hodkova et al., 2007)
Social food transmission No change Mouse F Pru (Xiao et al., 2012)
Reduced Mouse M Pru (Xiao et al., 2012)
Social interaction Increased Rat n/a RH (Gonzalez et al., 2007)
Associative emotional learning
Passive avoidance test No change Mouse n/a ME49 (Jung et al., 2012)
Fear Conditioning No change Rat M Pru (Vyas et al., 2007)
Impaired Mouse M PLK(II) (Ihara et al., 2016a)
Response to Novelty
Neophobia No change Mouse F Pru (Vyas et al., 2007)
(novel food avoidance)
Anhedonia Reduced Mouse F PLK(II) (Mahmoud et al., 2017, 2016)
Depressive-like behaviour
Forced swim test Increased Mouse F PLK(II) (Mahmoud et al., 2017, 2016)
Static rod test Impaired Mouse F HIF (Hodkova et al., 2007)
Motor control
Locomotor activity-open field No change Mouse M Pru (Xiao et al., 2012)
No change Mouse F ME49 (Kannan et al., 2010)
No change Rat M Pru (Vyas et al., 2007)
No change Rat n/a RH (Gonzalez et al., 2007)
Increased Mouse F Pru (Xiao et al., 2012)
Increased Mouse F Pru (Kannan et al., 2010)
Increased Mouse F HIF (Hodkova et al., 2007)
Increased Mouse F ME49 (Afonso et al., 2012)
Reduced Mouse F GT1 (Xiao et al., 2016b)
Reduced Mouse M ME49 (Gulinello et al., 2010)
Gait and Balance beam Impaired Mouse M ME49 (Gulinello et al., 2010)

(Hidano et al., 2016). These findings were transferable to in vivo mouse neuronal cytoskeleton and that intact cysts were separated from the
studies, where deletion of STAT1 in astrocytes lead to an increased cyst extracellular compartment by a layer of neurofibrillae enclosed within
count in astrocytes (Hidano et al., 2016), suggesting that astrocyte in- the host cell membrane (Sims et al., 1988). This was one of the earliest
vasion is possible but astrocytes are able to clear the infection or un- indications that cysts within the brain were physically protected from
dergo pyroptosis, and thus cysts are not observed in these cell types in the host’s immune response. Newer EM techniques revealed that cysts
vivo. were predominantly found in the grey matter, were intracellular, and
One key question involves the extent of cellular tropism of the majority of the host cells were neurons, as identified by the presence
Toxoplasma cysts in vivo, and the mechanisms mediating this phenom- of synapses (Ferguson et al., 1994; Ferguson and Hutchison, 1987).
enon. Early studies described the presence of cysts in the brains of in- More recently, a confocal study in chronically infected mice found that
fected mice using electron microscopy (EM) techniques (Sims et al., cysts were almost exclusively present in the neurons, as observed by
1988; Wanko et al., 1962), but failed to demonstrate their cell type encapsulation of cysts by neurofilaments (Melzer et al., 2010).
preference conclusively. Nevertheless, one of these studies did observe The tropism for neurons in vivo was elegantly shown by using a
that the wall of the tissue cysts contained components derived from the florescent protein expressing Toxoplasma strain (Koshy et al., 2010) and

75
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

Fig. 2. Structural and functional changes in the brain upon Toxoplasma infection. 1. At the macroscopic level, Toxoplasma infection causes lesions in the brain, alters
fibre density and grey matter volume and forms parasite cysts that remain in the brain throughout the lifetime of the host. 2. At the cellular level, the presence of
parasites in the brain parenchyma activates microglia, the immune cells of the brain, that generates the host’s immune response to counteract the invading pathogen.
Such a response is also mediated by the infiltrating T-cells. Infection also leads to a reduced dendritic spine count, causes axonal damage and causes an imbalance in
the proportions of kynurenic acid and quinolinic acid in the neurons. 3. Impairments in brain function can be attributed to changes that occur down at the synaptic
levels in the infected brain. Such changes can be structural, such as synaptic pruning by the classic complement system and altered microtubule and post-synaptic
density organization, or it can be functional, such as imbalance in neurotransmitter synthesis, release, clearance and metabolism, as well as perturbations in
downstream signalling at the post-synapse that lead to changes in gene expression. 5-HT; 5-hydroxytryptamine or Serotonin; BNDF, brain-derived neurotropic factor;
DAPRR-32, Dopamine and cAMP-regulated phosphoprotein, Mr 32 kD; DOP, Dopamine; GABA, Gamma-aminobutyric acid; GABA(A), Gamma-aminobutyric acid
receptor A; GAD67, Glutamic acid decarboxylase 67; GLT-1, Glutamate transporter-1; GLU, Glutamate; GluN1, NMDA receptor subunit N1; GluN2B, NMDA receptor
subunit N2B; IDO, Indoleamine 2,3-dioxygenase; IFNγ, Interferon-gamma; IL, Interleukin; iNOS, Induced nitric oxide synthase; KA, Kynurenic acid; MiR-132,
MicroRNA-132; PSD, Post-synaptic density; QA, Quinolinic acid; VLGUT1, Vesicular glutamate transporter 1.

76
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

exploiting the ability of the parasite to export effector proteins into the to actual onset of neuropsychiatric diseases, the obvious place to look is
host cells (Boothroyd and Dubremetz, 2008; Bradley and Sibley, 2007). the communication between brain cells, and understand the cascade of
These parasites injected Cre into the cells of Cre reporter mice, which events at the molecular level which are translated to global changes in
enables the visualisation of cells that the parasite has interacted with the brain and dictate behaviour (Fig. 2). Research into various neu-
allowing the authors to track GFP-positive cells in the brain. These ropsychiatric conditions aims to do exactly this, and understanding the
studies showed that neurons were the primary target cells for Tox- effects of Toxoplasma on brain has revealed that many changes observed
oplasma and that cysts reside in the cell’s processes rather than at the during latent infection recapitulate at least some of the effects known to
cell’s nucleus as seen in vitro (Cabral et al., 2016; Koshy et al., 2012; occur in specific brain disorders.
Koshy and Cabral, 2014). The most surprising finding using this model
was that Toxoplasma injects proteins into cells that it does not invade. 4.1. Changes in neuronal signalling
Overall, the observation that the number of cells affected by Tox-
oplasma (by protein injection) far exceeds cells that actually contain Neurotransmitters are chemical messengers in the brain that
cysts. The heterogeneity of infection sites observed in vivo indicates that transmit signals at synaptic junctions, and are classified into excitatory,
the parasite could ‘’explore’’ the brain parenchyma before settling on a inhibitory or modulatory, depending on their effects on signal trans-
host cell and forming a cyst. Deciphering why this occurs could address duction, and also have region-specific expressional patterns and func-
how and why Toxoplasma prefer neurons over other cell types and tional attributes. Dopamine, a slow-acting modulatory neuro-
suggests that this parasite may be able to bring about more distal transmitter, is essential for long-term regulation of brain functions
changes in the brain that could aid parasite survival and affect neuronal (Beaulieu and Gainetdinov, 2011; Girault and Greengard, 2004;
function behavioural output. Greengard, 2001). Dopaminergic neurons innervate regions of the brain
Apart from the cell type, tropism to specific brain region could also chiefly involved with motor function, motivation and working memory
help explain the behavioural consequences. Several studies have tried (Beaulieu and Gainetdinov, 2011), and are a central element of the
to elucidate this parameter, albeit with no conclusive results. An initial brain reward system and addiction (Di Chiara and Bassareo, 2007).
study by Vyas et al. found a mild tropism towards the amygdala, but Thus, it is not surprising that dopamine dysfunction has been im-
cysts were distributed throughout the mouse brain (Vyas et al., 2007). plicated in the aetiology of several human disorders such as Parkinson’s
One study exclusively observed the distribution of Toxoplasma cysts in disease (PD) (Matsumoto, 2015), Alzheimer’s disease (AD) (Martorana
the brain of mice with latent toxoplasmosis by hematoxylin-eosin and Koch, 2014), Huntington’s disease (HD) (Cyr et al., 2006; Tyebji
staining of brain sections and observed no particular tropism. Striking and Hannan, 2017), Schizophrenia (SZ) (Li et al., 2016), Tourette’s
interindividual differences in the total parasite load and cyst number syndrome (Mink, 2006) and attention deficit hyperactivity disorder
and distribution were also observed (Berenreiterová et al., 2011). Using (ADHD) (Gizer et al., 2009).
similar staining techniques, other studies found higher cyst densities in Dopamine dysregulation has perhaps generated the most interest in
the cerebral cortex, thalamus and cerebellum in infected rats (Dubey the study of Toxoplasma mediated neurochemical changes. Early report
et al., 2016) and mice (Hermes et al., 2008), while others found no by Stibbs showed higher dopamine levels in chronically infected mice.
tropism (Daniels et al., 2014; Gonzalez et al., 2007). Tracking of bio- This was not the case at acute stages, although the levels of homo-
luminescent Toxoplasma expressing luciferase under the control of a vanillic acid, a dopamine metabolite, was increased (Stibbs, 1985).
bradyzoite-specific gene promoter showed increased luminescence in Later in 2009, Gaskell et al. found two genes (aromatic amino acid
the cerebral cortex, colliculi, cerebellum and olfactory bulbs (Di hydrolase 1 and 2; AAH1 and AAH2) in Toxoplasma that encoded for
Cristina et al., 2008). The advent of the cre-injecting line in conjunction tyrosine hydroxylase (TH) that produces L-DOPA, the precursor of do-
with reporter mice should be able to provide a more definitive answer pamine (Gaskell et al., 2009). This was the first study to report the
to this question whilst also determining if there is any tropism for presence of aromatic amino acid hydroxylase in an apicomplexan
specific types of neurons using immunohistochemistry. parasite, and gave one of the first evidence of possible direct modula-
tion of neuronal function by the parasite. Reports from the same lab
3. Behavioural changes in rodents later showed that presence of Toxoplasma in mammalian dopaminergic
cells enhanced the levels of K+-induced release of dopamine several
The majority of the body of work that attempts to answer the basic fold, and also saw increased staining for dopamine and TH within cysts
questions on how Toxoplasma affects the host behaviour has involved present in the brain sections of infected mice (Prandovszky et al.,
observing infected rodent models (Table 1). Rodents are intermediate 2011). A later study by Wang et al. challenged this view by disrupting
hosts of Toxoplasma, and changes observed in them perhaps represent the AAH2 gene, whose expression increases at the dormant bradyzoite
the quintessential characteristics of the proposed ‘manipulation hy- stages (Webster et al., 2006), and showed that it had no effects on either
pothesis’, by which a parasite can manipulate its host’s behaviour in a the growth and differentiation of the parasite nor on the global dopa-
way that can increase its transmission into its definitive host. For ex- mine levels of the brain (Wang et al., 2015). They reported that the role
ample, Plasmodium infection makes humans more attractive to mos- of AAH genes was to increase the parasite’s capabilities of transmission
quitoes (Koella, 2005; Lacroix et al., 2005; Robinson et al., 2018), ra- in the cat (Wang et al., 2017). This discrepancy between the two la-
bies increases aggression which is thought to aid more biting (Sapolsky, boratories was proposed to be due to different methodological ap-
2003), and infection by parasitic worm Echinorhynchus truttae increases proaches taken (McConkey et al., 2015). More recently, it was shown
the respiratory rate of the fishes, forcing them into regions where they that disruption of the AAH2 gene did not alter the parasite induced
can be preyed upon (MacNeil et al., 2003). All of these promote behaviour abnormalities (Afonso et al., 2017; McFarland et al., 2018).
transmission of the infectious agent. Therefore, there is little doubt that Nevertheless, some studies have indeed reported lower dopamine levels
Toxoplasma is also able to affect its host behaviour in an insidious and higher dopamine turnover rates in the brains of mice chronically
manner to promote its own selfish means, while at the same time in- infected with Toxoplasma (Ihara et al., 2016b; Kannan et al., 2016;
ducing a plethora of related ‘by-product’ neuropsychiatric changes. Mahmoud et al., 2017), but its cause remains elusive.
Here, we provide an overview of the behavioural changes in rodents A mechanism by which Toxoplasma could disrupt dopamine sig-
observed upon infection with Toxoplasma. nalling is by altering microRNA-132 (miR-132) expression. MicroRNAs
are a class of small non-coding RNAs that can regulate gene expression
4. Functional effects on neurons (Lim et al., 2005), and miR-132 dysregulation is associated with several
neurological disorders such as SZ, AD, and PD (Miller et al., 2012;
When evaluating changes in the brain and linking pathogenic events Wanet et al., 2012). MiR-132 expression was found to be increased

77
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

during acute Toxoplasma infection (Li et al., 2015), while a chronic 2017), has essentially been proposed as a mechanistic link between
infection had an opposite effect (Xiao et al., 2014). This effect varied Toxoplasma infection and onset of SZ (Lucchese, 2017; Wang et al.,
amongst brain region, was sex-dependent and did not correlate with 2013). Two recent studies by Kannan et al. demonstrate that exposure
density of Toxoplasma in brain regions. Interestingly, when target pre- to Toxoplasma produces serum IgG antibodies to NMDARs that might
diction was performed followed by pathway enrichment analysis in the underlie glutamate receptor hypofunction and related impairment
transcriptome of Toxoplasma infected mice, dopamine signalling was found in SZ. In mice, autoantibodies to GluN2 were elevated when
the strongest associated pathway regulated by miR-132 expression. exposed to live Toxoplasma parasite, but not UV-inactivated parasites
Further, levels of dopamine D1-like receptors, the metabolising enzyme (Kannan et al., 2017, 2016), indicating that antibodies were not pro-
and intracellular proteins associated with the transduction of dopamine duced to the parasite itself, but involved an antigenic mimic of NMDAR
signalling (DARPP-32 phosphorylation at Thr34 and Ser97) were found that is generated during the dormant cyst phase. Interestingly, this in-
to be decreased as well (Xiao et al., 2014). Thus, although there is no crease was more pronounced in juvenile infected mice when compared
conclusive evidence of Toxoplasma infection directly increasing dopa- to adults (Kannan et al., 2016). Furthermore, evaluation of Toxoplasma-
mine levels in the host cells, sufficient evidence is present to indicate seropositive individuals with SZ indicated higher NMDA IgG titres
that dopaminergic pathways are indeed altered and further investiga- compared to seronegative SZ patients as well as seropositive non-SZ
tion in this area is required. controls (Kannan et al., 2017). Indeed, a sequence-matching analysis
Glutamate is another important neurotransmitter whose metabolism carried out between seven human NMDAR subunit amino acid se-
and signalling is altered upon Toxoplasma infection. Glutamate is an quences and the Toxoplasma proteome revealed vast epitopic peptide
excitatory neurotransmitter of the CNS and its receptors are localised on sharing between Toxoplasma and NMDAR subunits, indicating that anti-
neuronal as well as non-neuronal cells and regulate a broad range of Toxoplasma immune responses cross-react with NMDARs (Lucchese,
cognitive processes (Daikhin and Yudkoff, 2000; Dauvermann et al., 2017). This reaction to Toxoplasma proteins does in fact alter the levels
2017; Miladinovic et al., 2015). Under normal physiological conditions, of NMDA receptors in the cortex and the hippocampus in mice 8 weeks
glutamate is released at the synaptic cleft where it potentiates fast ex- post infection (Kannan et al., 2016; Torres et al., 2018).
citatory neurotransmission by activating its ionotropic receptors at the Another interesting neuron modulatory response generated by
post synapse, which include the α-amino-3-hydroxy-5-methyl-4-iso- Toxoplasma in the brain is its ability to alter GABAergic synapses and
xazolepropionic acid (AMPA), kainite and N-methyl-D-aspartic acid signalling in the CNS. Gamma-aminobutyric acid (GABA), an inhibitory
(NMDA) receptors and its metabotropic receptors (mGluRs) (Südhof, neurotransmitter, is primarily responsible for preventing the onset of
2013). At the matured glutamatergic synapses, a steep glutamate con- seizures by modulating the flow and timing of excitatory neuro-
centration gradient is maintained at the synaptic cleft by astrocytes at transmission (Treiman, 2001). Essentially, it acts as a tonic brake on the
the synapses that express the sodium-dependent glutamate transporters excitatory tone of glutamate, and as discussed above, perturbations in
and remove excess glutamate after the action potential from the ex- glutamate levels and signalling observed in presence of Toxoplasma
tracellular field (Daikhin and Yudkoff, 2000). This gradient provides could exacerbate any modifications of GABA signalling in the brain.
the driving force for rapid synaptic transmission to occur. Under pa- Epidemiological studies and meta-analysis have indicated that presence
thological conditions and dysregulation of the glutamate transporter of Toxoplasma in human population is associated with epilepsy (de
expression and/or function, excess extracellular glutamate becomes Bittencourt et al., 1996; Ngô et al., 2017; Ngoungou et al., 2015;
neurotoxic by over-activating the ionotropic glutamate receptors, Palmer, 2007), indicating possible dysregulation of inhibitory neuro-
especially at the extra-synaptic sites, triggering excitotoxic cell death in transmission in the presence of Toxoplasma. GABA is used as a carbon
surrounding postsynaptic neurons (Choi, 1994). source by Toxoplasma (MacRae et al., 2012) and infection leads to a
Altered extracellular glutamate levels upon Toxoplasma infection release of GABA from dendritic cells (Fuks et al., 2012). It was de-
has been shown in a recent study (David et al., 2016), while changes in monstrated that GABA activated GABA(A) receptors on Toxoplasma-
brain glutamate levels have been reported upon infection with a closely infected dendritic cells which led to a hypermigratory phenotype in
related parasite Neospora caninum (Ihara et al., 2016a). This report in- those cells. This process was postulated as a means of parasite dis-
dicates that glutamate homeostasis is disrupted upon infection with semination. Further, Brooks et al. recently showed that Toxoplasma
Toxoplasma. David et al. demonstrated that Toxoplasma infection in infection in mice leads to spontaneous seizures and re-distribution of
mice leads to a gradual reduction in the astrocytic glutamate trans- glutamic acid decarboxylase 67 (GAD67), a key enzyme that catalysis
porter, GLT-1, in the forebrain (David et al., 2016). In correlation, they GABA synthesis in the brain, from clustering at pre-synaptic termini, to
found a significant increase in extracellular glutamate during the a more diffused localization throughout the neuropil (Brooks et al.,
chronic stages of infection. Furthermore, these mice displayed impair- 2015). Heteromerisation of GAD67 with GAD65, a membrane an-
ments in electroencephalography (EEG) recordings from the frontal choring isoform of glutamic acid decarboxylase, is one of the proposed
cortex and reduction in anxiety-like behaviour. Interestingly, treatment mechanism by which GAD67 localises at the pre-synapse (Kanaani
with ceftriaxone, a β-lactam that is known to increase GLT-1 expression et al., 2010, 1999). Levels of GAD65 have not been studied in Tox-
(Rothstein et al., 2005), lowered extracellular glutamate and rescued oplasma infection and remain a plausible explanation for GAD67 re-
neuronal pathology, but failed to improve behavioural deficits (David distribution. Moreover, GABAergic dysfunction in the prefrontal cortex
et al., 2016). In contrast, and probably a hint towards a possible com- is a known signature of major depression (MD) (Croarkin et al., 2011;
pensatory mechanism, Kannan et al. showed that levels of hippocampal Karolewicz et al., 2010; Levinson et al., 2010), a phenotype present in
VGLUT1, a major vesicular glutamate transporter at the excitatory sy- murine models of Toxoplasma infection (Mahmoud et al., 2017, 2016).
napse, was reduced in infected mice (Kannan et al., 2016), although Thus, the association between loss of GABAergic inhibition and Tox-
such a change was not observed by another study (Brooks et al., 2015), oplasma-induced behavioural changes needs more investigation.
and demands more investigation. Serotonin is another neurotransmitter with a role in pathogenesis of
But perhaps more important in the glutamate dysregulation model depression (Oxenkrug, 2013) and whose levels are altered upon Tox-
of Toxoplasma is the role of its ionotropic receptors, specifically the oplasma infection (Ihara et al., 2016b; Kannan et al., 2016; Mahmoud
NMDA receptors (NMDARs). Toxoplasma infection has been strongly et al., 2017, 2016; Xiao et al., 2014). Chronic inflammation, as in the
associated with SZ, in light of numerous studies finding increased ser- case of toxoplasmosis, pushes tryptophan metabolism towards pro-
oprevalence of toxoplasmosis in individuals with SZ (Arias et al., 2012; duction of kynurenine and away from serotonin, leading to serotonin
Esshili et al., 2016; Monroe et al., 2015; Sutterland et al., 2015; Torrey deficiency. Indeed, reduced serotonin was found in models of acute
and Yolken, 2003). NMDAR, a key player in the pathophysiology of SZ infection or reactivated models of chronic toxoplasmosis, which was
(Javitt, 2015, 2010; Moghaddam and Javitt, 2012; Papouin et al., shown to be due to enhanced turnover to its metabolite 5-

78
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

hydroxyindolacetic acid (Mahmoud et al., 2017, 2016). There are in- HD individuals are susceptible to exacerbation of disease phenotype by
dications that such alterations in serotonin levels could be brain-region Toxoplasma. Nevertheless, its effects on psychiatric changes present in
specific, as one study found increased serotonin levels in the striatum HD are yet to be explored.
(Xiao et al., 2014), while an earlier study was unable to detect any The effect of Toxoplasma on AD is probably quite heterogeneous, in
serotonin changes in the brain (Stibbs, 1985). that the effects of infection depend upon the immune responses due to
Apart from the canonical neurotransmitter-mediated changes, other interaction between strain of parasite and the genetic mouse model of
pathways that could directly or indirectly alter brain function and lead AD used in the study. In mice overexpressing a mutant form of amyloid
to behavioural changes have also been explored in Toxoplasma infec- precursor protein (APP; Tg2576) (Jung et al., 2012) and in mice ex-
tion. One such pathway is the classical complement system that bridges pressing mutant APP and presenilin protein-1 (PSEN1; 5xFAD) (Möhle
innate and adaptive immunity and functions to clear antigen-antibody et al., 2016), Toxoplasma infection leads to a reduction in ß-amyloid
immune complexes from systemic circulation (Walport, 2001). This was (Aß) plaque burden, possibly due to enhanced production of anti-in-
first investigated as a likely candidate that could mediate elimination of flammatory cytokines TGF-B and IL-10, and an increase in monocytes
Toxoplasma tachyzoites from circulation. On the contrary, studies re- capable of phagocytosing Aß. Interestingly, such neuroprotective ef-
vealed that although the complement system was activated in presence fects led to an improvement in cognitive capacities of AD mice infected
of Toxoplasma, it failed to kill the parasites (Fuhrman and Joiner, 1989; with Toxoplasma (Jung et al., 2012). A later study using the human APP
Sacks and Sher, 2002). Interestingly, one study observed that this effect AD mouse model (J20) determined that only the Type II strain of
was specific in the presence of complement in cat sera (Kaneko et al., Toxoplasma was able to induce the above discussed changes, not Type I
2004). C1q, an immune protein of the classical complement system, is or III (Cabral et al., 2017). On the other hand, reports also suggest that
involved in synaptic pruning and elimination during development as Toxoplasma infection could exacerbate AD symptoms. One study using a
well as adult neurodegenerative processes (Stephan et al., 2012; triple transgenic AD mouse model (3xTg-AD) observed exacerbated
Stevens et al., 2007), and therefore is implicated in the aetiology of immune response in infected AD mice and severe morbidity (Montacute
several neuropathological conditions such as AD (Kolev et al., 2009), SZ et al., 2017). Another study reported an enhancement of cognitive
(Severance et al., 2014, 2012; Stevens et al., 2007) and multiple impairments in infected-BALB/c mice injected with subdoses of Aß
sclerosis (MS) (Michailidou et al., 2015). Recently, it has been reported 1–42 peptides (1 μl) in the hippocampus and considerable impairments
that levels of C1q mRNA and proteins are increased in the brains of in learning and memory functions, similar to AD models injected with
Toxoplasma infected mice, and can be found as punctate patterns 2 μl Aß peptides (Mahmoudvand et al., 2016a). More recently, Torres
around cysts, indicative of synapses along neuronal cell processes et al. showed that Toxoplasma infection induces two major hallmarks of
(Kannan et al., 2016; Xiao et al., 2016a). Thus, loss of synaptic com- AD such as Aß immunoreactivity and hyperphosphorylated Tau (Torres
plexity could be part of the collateral damage to neurons, due to et al., 2018). It is noteworthy that complement component C1q, acti-
complement system activated by the host as a response to pathogen vated upon Toxoplasma infection (Xiao et al., 2016a), can initiate mi-
invasion. croglia-mediated synaptic loss (Fonseca et al., 2004; Hong et al., 2016)
as well as phagocytosis of pre-amyloid aggregates providing neuro-
4.2. Toxoplasma – a disease modifier protection in AD (Pisalyaput and Tenner, 2008). This confounding role
of the complement system in AD pathology could explain the incon-
Emerging studies now posit the presence of Toxoplasma as an en- sistent results discussed above and therefore, more thorough and con-
vironmental factor that can either act as a trigger for disease onset, a clusive results are required to assess the susceptibility of AD patients to
factor that can exacerbate the disease condition, or sometimes even Toxoplasma infection.
ameliorate or halt disease progression. The strongest link is perhaps
between Toxoplasma and the susceptibility of individuals towards de- 5. Neuropathology of infection
velopment of SZ and bipolar disorder (BD), and recent reviews have
discussed this possible connection (Brown and Derkits, 2010; Del Changes in structure and morphology of neurons is a common
Grande et al., 2017; Elsheikha et al., 2016; Henriquez et al., 2009; phenomenon in neuropsychiatric diseases. Morphological integrity of
Kannan and Pletnikov, 2012; Sutterland et al., 2015; Yolken et al., neurons dictate its function by establishing synaptic partnerships, and
2009). SZ, affecting about 1% of the world population (Saha et al., thus it is no surprise that any change that compromise its structural
2005), has major socio-economic impact worldwide, and studies have rigour could have direct implications on how the cells respond to sti-
consistently shown that the prevalence of Toxoplasma infection is muli i.e. affect learning and memory. Behavioural changes in
unusually high in SZ patients (Torrey et al., 2007). Toxoplasma infection Toxoplasmosis have thus led to studies that analyse if neuronal integrity
is not only correlated with the appearance of SZ (Ebadi et al., 2014; is compromised, or the damage is also taken even further so as leading
Fuglewicz et al., 2017; Torrey and Yolken, 1995; Yolken et al., 2009), to cell death.
but the effect of latent toxoplasmosis on the risk of developing SZ is Toxoplasma infection causes significant neuronal pathology, and few
stronger than that of any SZ-associated gene variant identified in recent studies have demonstrated this in vivo as well as in vitro (Fig. 2).
genome-wide analyses (Purcell et al., 2009). Toxoplasma infection was More recently, in vivo non-invasive magnetic resonance imaging (MRI)
also shown to exacerbate open field activity in a genetic mouse model was performed at high magnetic fields on Toxoplasma infected mice
of SZ (Eells et al., 2015) and mimic the depression-like phenotype in a which identified and anatomically localised the preferential lesion sites
drug-induced model of SZ (Wang et al., 2013). of parasite-induced brain pathology (Parlog et al., 2014). High density
A recent study has evaluated the effects of Toxoplasma infection on of hypointense lesions were found in the somatosensory and motor
aetiology of HD, working on the hypothesis that alterations in the ky- cortices, hippocampus and the striatum of all the infected mice. Fur-
nurenine pathway, that occur during both Toxoplasma infection (Silva ther, a detailed qualitative and quantitative analysis of the brain con-
et al., 2002) as well as in HD (Beal et al., 1992; Thevandavakkam et al., nectional microstructure using in vivo diffusion-tensor MRI and high-
2010), could represent a point of synergism between these two condi- resolution fiber mapping revealed that the somatosensory cortical re-
tions (Donley et al., 2016). Indeed, they found that Toxoplasma-infected gions showed the highest density of lesions, and a loss in fiber co-
HD mice have elevated brain indoleamine 2,3-dioxygenase (IDO) ac- herence and density at the lesion sites (Parlog et al., 2014). Such fiber
tivity, and underwent a premature death. Interestingly, Toxoplasma density maps showed an overall fiber loss in the infected group. Inter-
infection in HD mice produced a blunted CD8+ T-cell IFNγ response as estingly, a voxel-based morphometry MRI in schizophrenic individuals
compared to Toxoplasma-infected wild-type (WT) mice, that lead to an positive for IgG against Toxoplasma revealed significantly reduced grey
increased parasite burden in the brain. This is the first indication that matter volume of several cortical regions compared to non-infected

79
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

patients (Horacek et al., 2012). Above studies indicate that apart from et al., 2008) and also because programmed cell death is an essential
local neuronal changes, alterations in intra-regional brain connectivity mechanism of the host to combat infectious diseases (Contreras-Ochoa
might also play a significant role in the observed behavioural outcomes. et al., 2013; Graumann et al., 2009). Possible excitotoxic mechanisms
When individual neurons were analysed, Parlog et al. observed associated with Toxoplasma infection is discussed in detail in Section 6.
significantly reduced dendritic complexity of the layer II/III pyramidal The general lack of cell death in vivo could be due to the strategies
neurons of the cortex, the CA1 pyramidal neurons as well as the granule evolved by Toxoplasma to directly inhibit host cell apoptosis in order to
cells of the dentate gyrus of the hippocampus (Parlog et al., 2014). hamper elimination (Contreras-Ochoa et al., 2013; Graumann et al.,
Total dendritic length was reduced only for the layer II/III and the 2009; Ni Nyoman and Lüder, 2013; Payne et al., 2003). These include
granule cells. Reduced dendritic complexity was also reported in a more upregulation of the anti-apoptotic mechanism in host cells such as re-
recent study where dendritic spines from the frontal cortex were duction or inhibition of caspase molecules, upregulation of anti-apop-
quantified using dil labelling in frozen sections from naïve and infected totic factors and downregulation of poly ADP-ribose polymerase ex-
mice (David et al., 2016). Dendritic spines are specialised membrane pression (Goebel et al., 2001; Kim and Denkers, 2006). This effect is not
protrusions from a neuronal dendrite that receive synaptic input from only induced in infected cells, but neighbouring cells as well (Orlofsky
the neighbouring neurons (Blanpied and Ehlers, 2004). Abnormalities et al., 2002; Vutova et al., 2007), possibly contributing to protecting the
in dendritic structure and number are a characteristic observed in brain from neurodegenerative events. Although, recently a study
several neuropsychiatric disorders (Glantz and Lewis, 2000; Qiao et al., showed increased cell death in the cortex and olfactory bulb of mice 60
2017; Tyebji and Hannan, 2017; van Spronsen and Hoogenraad, 2010). days after infection (Torres et al., 2018).
An initial study by Mitra et al. showed that the total dendritic length of Apart from changes in cell structure, Toxoplasma infection has also
the principal neurons of the basolateral amygdala (BLA) were reduced been reported to alter neuronal structure and function at the synaptic
in rats 8 weeks after infection (Mitra et al., 2013). While both the level. Synapses are the units of information processing in the brain, and
hemispheres were affected, the right hemisphere showed a greater they play a dynamic role in learning and memory processes by being
dendritic retraction. Interestingly, when such an analysis was done in modified in structure and function upon receiving external stimuli and
the neighbouring brain region, the pyriform cortex, dendritic retraction environmental cues. Post-synaptic density protein-95 (PSD-95) main-
was not observed. This precluded a generalised retraction across the tains the correct molecular organisation of the postsynaptic density
brain, and is indicative of brain region specific effects of toxoplasmosis. complex (Chen et al., 2011) and regulates synaptic plasticity
Weather such effects are a result of a differential response of particular (Montgomery and Madison, 2004). At the pre-synapse, synaptophysin
neurons to the infection remains to be discovered. modulates vesicle cycle and neurotransmitter release at the synaptic
Neuronal architecture is maintained by the cytoskeletal structure of cleft (Valtorta et al., 2004). Thus, predictably, dysregulation of these
the cells and Toxoplasma infection also causes disruption in neuronal important synaptic proteins have major implications for the beha-
cytoskeleton which have been mapped in some recent studies. In one vioural outcome of the animal (Deng et al., 2012; Irie et al., 1997;
model of murine toxoplasmosis, infection with ME49 type II strains led Newpher and Ehlers, 2008; Schmitt et al., 2009). Indeed, upon infection
to a significant reduction in the neuronal neurofilament marker SMI311 with Toxoplasma, a significant reduction in PSD-95 and synaptophysin
along with reduction in microtubule associated protein-2 (MAP2) in the was observed in the hippocampus and cortex of mice 8 weeks later
somatosensory cortex (Parlog et al., 2014). Staining for β-III tubulin, a (Kannan et al., 2016; Parlog et al., 2014). Whether such changes in
component of neuronal cytoskeleton, revealed a disruption in the synaptic protein precede abnormalities observed in the dendritic com-
neuronal structure, with mice infected with ME49 strains displaying a plexity during chronic toxoplasmosis remain to be studied.
reduced number of β-III tubulin positive cells in the layer II/III of the
cortex (David et al., 2016). In the same study, damage to neurons was 6. The brain – immune axis
also observed via a significant reduction in staining for the neuronal
nuclei marker NeuN. Interestingly, such a reduction in neuronal mar- The brain has classically been accepted as an immune privileged
kers was not due to a reduction in total cell count, as Nissl staining organ due to the lack of the classical lymphatic system draining CNS
revealed no significant changes in cell density. Further, Toxoplasma antigens and the tight capillary junctions of the BBB that limits the
infection in a mouse model of AD blocked neurodegeneration and re- entry of immune cells into the brain parenchyma, as well as astrocyte
duced amyloid plaque formation in the cortex and hippocampus com- endfeet processes. Although this notion still holds true, it has been in-
pared to their non-infected counterparts (Jung et al., 2012). Above ef- creasingly accepted now that immune cell infiltration of the CNS occurs
fects could be explained by studies that show that during latent not only during infection to control pathogenic invasion but also to
infections, Toxoplasma contributes to control the host immune system maintain brain homeostasis and function (Carson et al., 2006; Kipnis,
in a way that prevents neurodegeneration (Rozenfeld et al., 2005, 2003; 2016; Louveau et al., 2015). Such neuroimmune interactions can have
Wagner et al., 2009), possibly via the production of transforming both beneficial and detrimental effects on brain functions. Even before
growth factor ß-1 secreted by IFNγ-activated microglia (Rozenfeld reaching the brain, peripheral immune activation caused by Toxoplasma
et al., 2005). On the contrary, it is interesting to note that cell death was can produce a host of pro-inflammatory immune cells that can cause
observed in vitro in mouse neuronal cells co-cultured with Toxoplasma- neuroinflammation, leading to local or global disruption in neuronal
infected microglial cell as well as in vivo in mouse models of tox- physiology (Filiano et al., 2015). Interestingly, peripheral immune
oplasmosis encephalitis (TE) (Zhang et al., 2014). Such model of re- mediators have been implicated in the pathophysiology of SZ, HD, MD,
activated toxoplasmosis is particularly relevant to im- AD, stroke, autism as well as traumatic brain injury (TBI) (Carter, 2011;
munocompromised individuals or those on continuous Haroon et al., 2012; Onore et al., 2012; Zheng et al., 2015). Discussing
immunosuppressive medication due to its devastating effects on the every effector cell/molecule activated in the central and peripheral
brain (Dellacasa-Lindberg et al., 2011; Jones et al., 1996; Reiter-Owona immune system upon Toxoplasma infection is beyond the scope of this
et al., 2000). This effect was due to microglial activation and sub- review article, and has been discussed elsewhere (Aliberti, 2005; Klein
sequent release of pro-inflammatory cytokines, because an inhibitor of et al., 2017; Yarovinsky, 2014). Instead, here we shall discuss changes
microglial activation blocked the above described effects (Zhang et al., in immune and hormonal milieu after Toxoplasma infection which di-
2014). The effects on Toxoplasma in vitro are indeed different in terms of rectly or indirectly impinge on the brain function that might result in
survival and function. alterations in cognitive abilities of the host.
In toxoplasmosis, cell death can occur due to the fact that IFNγ production represents the first line of defence against
Toxoplasma infection leads to a robust induction of pathways associated Toxoplasma infection when innate immune cells such as dendritic cells
with excitotoxicity (Cervenka et al., 2017; Lucchese, 2017; Mizuno (DC) and neutrophils, and macrophages via IL-10 release, initiate

80
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

natural killer (NK) cell and T-cell mediated IFNγ production (Denkers implicated in the aetiology of AD (Gulaj et al., 2010), HD (Guidetti
et al., 2004; Hunter et al., 1996). In the brain, invasion by Toxoplasma et al., 2006, 2004), depression (Steiner et al., 2011), autism
leads to rapid expression of chemoattractant such as CCL5, CXCL9 and (Zimmerman et al., 2005), as well as in cases of increased suicidal at-
CXCL10 by astrocytes and microglia that recruit IFNγ producing CD8+ tempts (Erhardt et al., 2013). Interestingly, activation of the kynurenine
and CD4+ T-cells directed at Toxoplasma antigens, leading to parasite pathway is observed in the brains of Toxoplasma infected mice, as
clearance (Landrith et al., 2015). Also, Toxoplasma infected CNS shows measured by increased IDO production, kynurenine metabolism and QA
increased astrocytic expression of TNF, IL-1B and IL-6 (Mahmoudvand levels (Murakami et al., 2012; Notarangelo et al., 2014; Silva et al.,
et al., 2016b). It is noteworthy that each of these effector molecules has 2002). Many recent studies have shown that activation of IDO, likely
been implicated in altering neural correlates that are involved in via disruption in kynurenine metabolism pathway and subsequent QA
memory formation such as neurogenesis, synaptic plasticity and mod- production and NMDAR-mediated excitotoxicity, leads to depression-
ulation of long-term potentiation (del Rey et al., 2013; Filiano et al., like phenotype observed in Toxoplasma infected mice (Dinel et al.,
2016; Mandolesi et al., 2017; Riazi et al., 2015; Wu et al., 2013). IFNγ 2014; Mahmoud et al., 2017; Mazarei et al., 2013; Parrott et al., 2016).
has an important role in regulating social behaviour, anxiety- and de- The changes in NMDAR signalling, PSD organization as well as altera-
pressive-like behaviour as well as spatial recognition memory, and tions in receptor levels discussed before could be an outcome of Tox-
dysregulation of these has been associated with Toxoplasma infection oplasma-mediated changes in the kynurenine pathway. Further studies
(Gonzalez et al., 2007; Goodwin et al., 2012; Mahmoud et al., 2017, are required to ascertain the role of kynurenine pathway in Toxoplasma
2016). Interestingly, results show that increase in IFNγ signalling has infection and if such changes are associated with onset and/or ex-
positive and reinforcing roles on social behaviour (Filiano et al., 2016), acerbation of neuropsychiatric conditions already displayed by pa-
but caused affective dysfunction in mice (Mandolesi et al., 2017). Fi- tients.
liano et al. demonstrated that IFNγ knock-out mice (KO) displayed so- Another downstream effector of IFNγ signalling and an important
cial deficits, that could be rescued by a single injection of IFNγ (Filiano microbicidal mechanism for the control of Toxoplasma infection is NO
et al., 2016). Also, IFNγ increased tonic GABAergic inhibitory current, generated by the action of nitric oxide synthase (NOS). Depending on
increased latency to reach seizures, and reduced the maximum severity the site of expression, NOS has three isoforms: endothelial (eNOS, ex-
of seizures. Moreover, diazepam, a GABA activator, rescued social pressed in vascular endothelium and choroid plexus), neuronal (nNOS,
deficits in IFNγ KO mice. These results demonstrate that IFNγ mediates expressed in neuronal cell bodies) and inducible (iNOS, expressed in
GABAergic signalling, that regulates social behaviour in mice. Given macrophages and glial cells). Interestingly, all three isoforms are pro-
that Toxoplasma infection leads to redistribution and dysregulation of duced in the brain of mice in response to Toxoplasma infection (Dincel
GABA in the brain (Brooks et al., 2015), this could impair IFNγ medi- and Atmaca, 2015; Tonin et al., 2014). NO production upon Toxoplasma
ated social activity in infected mice. Another study found that IFNγ KO infection has a protective role by killing and eliminating the parasites
mice displayed increased anxiety- and depression-like phenotype, with (Khan et al., 1997), and thus it is no surprise that Toxoplasma activity
reduced neurogenesis in the hippocampus (Campos et al., 2014). It is inhibits NO production to better its odds of survival (Guillermo and
interesting to note that, although IFNγ KO mice displayed impaired DaMatta, 2004; Seabra et al., 2002). In fact, recent attempts at pro-
spatial recognition memory in basal state, under conditions of chronic ducing vaccines against the parasite exploit this protective NO me-
stress, IFNγ KO mice displayed enhanced memory performance chanism to reduce brain parasitism (Czarnewski et al., 2017). However,
(Litteljohn et al., 2014). This indicates that IFNγ signalling, under although NO production helps the host cells control parasite pro-
conditions of chronic stress such as in Toxoplasma infection, exacerbates liferation, it has a detrimental, sometimes lethal effect on the cells as
memory impairments. Indeed, recently it was shown that in models of well as the infected mice. iNOS KO mice had a higher brain cyst burden
experimental autoimmune encephalomyelitis (EAE), increase in IFNγ (Sa et al., 2017), but much lower brain inflammation than their par-
signalling in the striatum caused anxiety- and depressive-like beha- ental lines, with no change in IFNγ or TFNα levels (Khan et al., 1997).
viours in mice (Mandolesi et al., 2017). This occurred by dysregulation Despite increased proliferation of Toxoplasma in the brains of iNOS KO
of the cannabinoid CB1 receptor at the GABAergic synapses in the mice, these mice displayed increased survival post-infection (Khan
striatum. Thus, the direct role of IFNγ signalling in regulating memory et al., 1997). Thus, NO production plays an important role in regulating
performance in infected mice requires a thorough inspection. Never- parasitism in the host.
theless, downstream effectors of IFNγ signalling, generally studied as Apart from its microbicidal function, NO also signals as a neuro-
part of IFNγ mediated mechanism of parasite killing, are also involved transmitter in the brain, and has not only been implicated in processes
in memory regulation and their effects on regulating host behaviour that regulate learning and memory (Paul and Ekambaram, 2011;
seem more plausible under conditions such as Toxoplasma infection. Steinert et al., 2010) but also in brain homeostasis by regulating cere-
One mechanism by which IFNγ suppresses Toxoplasma growth is by bral blood flow at rest and under physiological conditions (Garry et al.,
inducing tryptophan degradation via IDO activation (Pfefferkorn, 1984; 2015). NO is synthesized in the brain on demand upon neuronal activity
for other mechanims see: Hunter and Sibley, 2012). Tryptophan is an under cognitive conditions, when an increase in intracellular Ca2+
essential amino acid and a precursor for the synthesis of serotonin. Also, leads to the activation of eNOS or nNOS leading to NO creation, while
IDO can cleave the indole ring of serotonin to form its breakdown Ca2+-independent induction of NO by iNOS mediates immune function
product formyl-5-hydroxykynurenamine (Steiner et al., 2011). Thus, (Steinert et al., 2010). Interestingly, NO derived from eNOS and nNOS
IFNγ induction could lead to changes in serotonin metabolism, and its is neuroprotective and facilitates synaptic plasticity, while NO derived
effects have been discussed earlier. Tryptophan metabolism also in- from iNOS may have neurotoxic effects. Under physiological condi-
duces the kynurenine pathway, whose end products are the two neu- tions, levels of iNOS in brain and vasculature is low (Ortiz and Garvin,
rotoxic agents, kynurenic acid (KA) and quinolinic acid (QA) (Ruddick 2003), but under conditions of inflammation, such as upon Toxoplasma
et al., 2006). Under basal conditions, most kynurenine is metabolized to infection, iNOS levels are upregulated (Fujigaki et al., 2003), which
KA (Hilmas et al., 2001), but under inflammatory conditions, kynur- might lead to nitrostative stress: this is the ability of NO and its deri-
enine metabolism shifts towards QA production (Saito et al., 1992). QA, vatives (reactive nitrogen species) to cause damage to lipids, proteins
an NMDA agonist, is heavily implicated in driving glutamate mediated and DNA, leading to neuronal toxicity (Beckman and Koppenol, 1996).
excitotoxicity as well as precipitating oxidative damage in neurons, Another detrimental consequence of increased NO is the inhibition of
which is now well documented as a precursor for development of the mitochondrial respiratory chain leading to oxidative stress
neuropsychiatric disorders (Lugo-Huitrón et al., 2013). Indeed, higher (Erusalimsky and Moncada, 2007; Heales et al., 1999). NO production
KA levels are a characteristic of SZ (Erhardt et al., 2001; Nilsson et al., due to inflammation and resulting oxidative stress has been implicated
2005; Schwarcz and Hunter, 2007), while QA upregulation has been in the aetiology of AD, PD, MS and HD, as well as brain damage

81
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

following ischemia and reperfusion (Bennett et al., 2009; Steinert et al., Kocazeybek et al., 2009; Yereli et al., 2006). Other documented changes
2010). Increased iNOS expression and axonal damage was reported in in human traits upon infection with Toxoplasma were poor impulse
experimental models of PD (Barthwal et al., 2001; Gatto et al., 2000; regulation in males and violent and risk-taking behaviour along with
Singh et al., 2005), as well as post-mortem brain tissues from patients self-directed violence in women (Cook et al., 2015; Pedersen et al.,
with MS (Bö et al., 1994), AD (Wallace et al., 1997) and PD (Hunot 2012). Also, some studies have found an increased association between
et al., 1996). Thus, the above discussed data strongly suggests that upon Toxoplasma antibody titer and suicide attempts (Arling et al., 2009;
infection with Toxoplasma, the interplay among different isoforms of Ling et al., 2011; Yagmur et al., 2010). Moreover, population com-
NOS might play a pivotal role in determining its effects in the brain, and parison studies have documented a correlation between Toxoplasma
a balance between anti-parasitic and pro-cognitive properties of NO antibody titers to increased homicide and suicide (Lester, 2012, 2010),
signalling and detrimental cytotoxic properties of NO and its deriva- in line with another study that reported higher seropositivity among
tives requires a further detailed investigation and understanding. prison inmates (Alvarado-Esquivel et al., 2014). In order to make this
Cytokines such as IL-1ß, IL-6, IL-1 and TNFα, all of which are data more definitive, longitudinal studies are required that evaluates
known to be produced in abundance upon Toxoplasma infection (Burke the mental health status of people before and after acquiring infection.
et al., 1994; Hunter et al., 1992a, 1992b), activate the hypothalamic- Moreover, monitoring the infection status should be made a routine for
pituitary-adrenal (HPA) axis (Tilders et al., 1994; Turnbull and Rivier, mental health patients, which could possibly uncover previously un-
1995), an essential adaptive mechanism to stressful environment known effects of Toxoplasma on human behaviour.
(López et al., 1999). HPA activation leads to secretion of corticosterone
(CORT) that mediates emotional (Pariante and Lightman, 2008), an- 8. Transgenerational influence of Toxoplasma
xiety (Mitra and Sapolsky, 2008) and depressive-like behaviour
(Johnson et al., 2006), possibly involving serotonin signalling in the The most prominent deleterious effect of acquiring Toxoplasma in-
amygdala (Herman et al., 2003; Mitra et al., 2009a;b). Interestingly, it fection during pregnancy is its effects on the offspring. The effects of
was demonstrated that Toxoplasma infection leads to reduction in basal maternal immune activation on offspring mental health is a widely
CORT levels as well as CORT in response to stressful stimuli such as studied field of research (Canetta and Brown, 2012; Knuesel et al.,
exposure to cat odour (Mitra et al., 2013). This could possibly explain 2014), and thus it is no surprise that Toxoplasma infection in utero is
the loss of fear to cat odours, but characterizing other behavioural associated with high risk of developing psychiatric diseases, by af-
changes in Toxoplasma-infected mice has indicated an increase in an- fecting fetal brain development and increasing the vulnerability to
xiety and depressive-like phenotypes (Gatkowska et al., 2012; develop SZ later in life (Brown et al., 2005; Elsheikha et al., 2016;
Mahmoud et al., 2017, 2016; Skallová et al., 2006), indicating me- Mortensen et al., 2007a, 2007b). Studies that document specific can-
chanisms separate from HPA-axis- mediated signalling. didate infections associated with the incidence of neuropsychiatric
Thus, activation of the central and peripheral immune system in disorders have shown high predilection between prenatal Toxoplasma
response to parasitic infection is a ‘slippery slope’ that allows the brain infection and SZ (Brown et al., 2009; Brown and Derkits, 2010). It is
to control parasitism but leads to unintended changes in neuronal noteworthy, however, that SZ was shown to relate to high maternal
function, manifesting as neuropsychiatric problems. Toxoplasma IgG levels and not the presence of parasite itself (Brown
et al., 2000; Mortensen et al., 2007a). However, no association was
7. Implications for human behaviour and brain disorders found between maternal Toxoplasma infection and risk of BD
(Freedman et al., 2016; Mortensen et al., 2011), suggesting that effects
Toxoplasma infection has major implications on our society and of Toxoplasma infection may be specific to development of SZ. Studies
global healthcare systems. Humans are secondary hosts in the trans- on mouse models of congenital Toxoplasma infection indicated that
mission cycle of Toxoplasma in the environment and its effect on human infection acquired during gestation or prior to mating resulted in off-
personality and behaviour are wide ranged and mostly subtle (Flegr, spring that displayed increased ambulatory activity and reduced pre-
2013; Sugden et al., 2016). In Toxoplasma-infected human subjects, ference for novelty (Hay et al., 1984) and learning and memory deficits
characterizing the personality profile has revealed several changes in (Wang et al., 2011). However, a later study failed to confirm these
their psychology, some based on their gender. Infected men were found findings, and moreover reported that maternal Toxoplasma infection
to be more suspicious and jealous, and had higher tendency to disregard does not result in neurotransmitter changes in offspring brain (Goodwin
rules (Flegr, 2007). Meanwhile, infected women were more easy-going et al., 2012). It is likely that changes observed upon maternal Tox-
and warm-hearted than their uninfected counterparts. Further, both oplasma infection could be due to exposure to inflammatory environ-
men and women showed reduced novelty-seeking behaviours (Flegr, ment during development, rather than a direct effect of parasite IgG or
2010; Flegr et al., 2003; Skallová et al., 2005), were more submissive in the parasite itself. Indeed, maternal inflammation leads to changes in
a conflict and had a blunted reflex behaviour under imminent danger, anxiety and performance in spatial learning tasks in the offspring
while reporting that diplomacy is not their strong point (Flegr, 2010). (Schaafsma et al., 2017), and also activates maternal complement C1q
In terms of behavioural changes, there have been some association system, a risk factor in the development of SZ and psychosis in offspring
studies linking Toxoplasma infection to depression (Gale et al., 2014; (Severance et al., 2014). Apart from the development of neu-
Pearce et al., 2012) and epilepsy (de Bittencourt et al., 1996; Ngoungou ropsychiatric disorders in offspring, maternal infection with Toxoplasma
et al., 2015; Palmer, 2007), while one case report also suggested that causes spontenous abortions (Giakoumelou et al., 2016; Jones et al.,
depressive-like symptoms could be alleviated upon successful Tox- 2001; Wong and Remington, 1994) and lower body weight at birth
oplasma treatment (Kar and Misra, 2004). Infection was also correlated (Wong and Remington, 1994) is associated with harm to the developing
with depression in pregnant women (Nourollahpour Shiadeh et al., fetus due to an increased risk of self-directed violence in pregnant fe-
2016). Toxoplasma infected subjects showed prolonged reaction times males (Pedersen et al., 2012).
measured by a simple reaction test (Havlícek et al., 2001). Performance Inheritance of Toxoplasma effects via the paternal line of transmis-
worsened proportional to the levels of anti-Toxoplasma antibodies and sion has recently gained attention. Rodent studies have shown that
tests suggested that toxoplasmosis impaired long-term concentration males infected with Toxoplasma display reduced sexual rigor, reduced
ability rather than maximum performance. Also, such an impaired sperm motility, sperm count and viability and increased sperm defor-
psychomotor performance, along with slower reaction times (Havlícek mities (Abdoli et al., 2012; Dalimi and Abdoli, 2013; Dvorakova-
et al., 2001; Příplatová et al., 2014) and poor attention (Beste et al., Hortova et al., 2014; Lim et al., 2013; Terpsidis et al., 2009). Mating
2014), could explain the increased incidence of Toxoplasma infected with infected males resulted in a significant drop in successful preg-
drivers to be involved in road traffic accidents (Flegr et al., 2009; nancies (Asgari et al., 2015). Interestingly, Toxoplasma parasites were

82
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

detected in the semen of infected human, rat, rabbit, dog, sheep, cattle, (Bougdour et al., 2013; Braun et al., 2013; Franco et al., 2016; Gay
goat, and swine (Dalimi and Abdoli, 2013; Flegr et al., 2014), while in et al., 2016; Nadipuram et al., 2016).
some species such as rabbit, sheep, and dogs, Toxoplasma can also be One of the first nuclear DG proteins to be characterized was GRA16,
transmitted via semen to infect the females during mating (Flegr et al., which alters the expression of proteins involved in the cell cycle
2014), but such a phenomenon has not yet been found in mice (Asgari (Bougdour et al., 2013). Once exported, GRA16 interacts with tran-
et al., 2015). Thus, Toxoplasma indeed affects the male reproductive scription factors and p53 regulators, thus preventing apoptosis and
parameters, but does it have any effects on the next generation? causing cell cycle arrest (Bougdour et al., 2013; Li et al., 2002). Further
There has been only one study conducted on human population that analysis identified GRA24, a protein that localizes to the host nucleus
reported that fathers of congenitally infected infants were also more where it forms a complex with p38α mitogen-activated protein kinase
likely to be seropositive for Toxoplasma infection. (Contopoulos- (MAPK), inducing its auto-phosphorylation independently of the MAPK
Ioannidis et al., 2015). However, what is more important in this context cascade (Braun et al., 2013). This resulted in significant upregulation of
is the effect of parasite infection on the epigenetic changes that can be pro-inflammatory cytokines such as IL-12, TNF, and IFNγ. Furthermore,
transmitted from father to offspring via sperm resulting in behavioural the prolonged nuclear localisation of the GRA24-p38α complex led to
changes. Indeed, Toxoplasma infection has been reported to induce al- an increased expression of immediate early gene products such as of
terations in DNA methylation status of genes in the brain (Hari Dass and Egr-1 and c-Fos, resulting in a wave of downstream transcriptional
Vyas, 2014) as well as promoters of spermatogenesis genes (Dvorakova- changes (Braun et al., 2013).
Hortova et al., 2014). It is noteworthy that multiple studies have re- The ability to respond to IFNγ and initiate STAT1-dependent tran-
ported that paternal exposure to stress results in epigenetic changes in scription is vital to TH1 defence against Toxoplasma. It has been known
sperm small non-coding microRNAs that can transmit experience-de- for some time that Toxoplasma suppresses STAT1 dependent tran-
pendent information from parent to offspring and results in detectable scription, despite normal STAT1 activation and nuclear localization in
changes in behavioural phenotype up to three generations (Bale, 2015; response to IFNγ (Kim et al., 2007; Rosowski et al., 2014; Rosowski and
Short et al., 2016; Yeshurun et al., 2017; Yeshurun and Hannan, 2018). Saeij, 2012). The DG protein IST (Inhibitor of STAT1 Transcriptional
Microinjection of specific sperm microRNAs, known to be modified activity) was identified and was found to localise at the host nucleus,
upon paternal stress experience, into the zygote recapitulated stress were it inhibits STAT1 transcription, not by preventing its DNA binding
dysregulation phenotype in the offspring (Rodgers et al., 2015). What is capacity, but rather increasing it (Gay et al., 2016; Olias et al., 2016;
yet unknown is whether Toxoplasma’s presence in the testis and semen Rosowski et al., 2014). The mechanism by which IST increases STAT1-
somehow reprograms or establishes epigenetic marks in the father’s DNA binding and mediates the repression of transcription is two-fold:
germ line which is inherited by the offspring. Such investigations would (1) by increasing the phosphorylation of STAT1 and therefore acti-
open up a whole new arena of congenital toxoplasmosis, with major vating and localising additional STAT1 to the nucleus, all in the absence
public health implications. of IFNγ, and (2) by recruiting the chromatin remodelling complex (Mi-
2/NuRD) to the GAS (interferon gamma activated sequence) site, re-
9. Host-parasite interactions sulting in histone acetylation modification and inactivation. In contrast
to the proteins described above, DG protein, GRA15, does not localise at
Alternatively, the changes discussed above could occur as a result of the host nucleus, but rather it is thought to be embedded into host-side
active manipulation by the parasite. Toxoplasma is known to manip- of the PVM where it activates the host’s NF-kB pathway by the nuclear
ulate the host response during an acute infection by actively exporting translocation of NF-kB subunit, p65 (Rosowski et al., 2011).
proteins (Coffey et al., 2016), to prevent clearance and thus enabling Translocation of all these factors across the vacuolar membrane and
the establishment of a chronic infection. Toxoplasma appears to have into the host cell requires machinery. A parasite Golgi-resident protease
two pathways which it can export proteins into the infected host cell or (ASP5) has been shown to be required for maturation of exported
at the host parasite interface, both of which are immunomodulatory. proteins (Coffey et al 2015, Hammoudi et al., 2015, Curt-Varesano et al
Invading tachyzoites inject proteins from unique secretory organelles 2015, Gay et al., 2016). Further, translocation complex comprising
shared by related parasites known as ‘rhoptries’ (Boothroyd and MYR1, 2 and 3 is essential for Toxoplasma proteins to cross the va-
Dubremetz, 2008; Bradley and Sibley, 2007). Rhoptries inject several cuolar membrane into the host cell (Franco et al., 2016; Marino et al.,
known proteins, including a fascinating group of kinases and pseudo 2018). Removal of any of this translocation machinery has drastic ef-
kinases, which are involved in initial immune evasion (Clough and fects on parasite-induced changes to host cell transcription (Naor et al.,
Frickel, 2017; Delorme-Walker et al., 2012; Peixoto et al., 2010; Saeij 2018).
et al., 2007). One of the most interesting findings regarding host cell To date, the majority of research into host-Toxoplasma interactions
manipulation by Toxoplasma was the discovery that, in the brain, the has focussed on the tachyzoite stage of the lifecycle. However, there is
parasite will also inject proteins into cells that it does not invade (Koshy no evidence to show that encysted bradyzoites export proteins and
et al., 2012). These ‘injected but uninfected’ neurons outnumbered in- whether these factors, if any, are the same or different to that known to
fected cells suggesting that parasites can move through the brain par- be exported by tachyzoites. Nevertheless, it seems unlikely that the
enchyma and modify surrounding host cells without directly invading parasites in bradyzoite stages can remain concealed from the immune
them. This potentially has an impact on avoiding immune clearance system without some kind of host manipulation. Furthermore, many of
and may also be important for eliciting changes in neurons, causing the pathways modulated by tachyzoites, such as the MAPK, IFNγ, and
brain dysfunction. NF-kB pathways, have been implicated in important neuronal functions
Once the parasites have invaded, concomitantly generating a va- and behaviours. To simply ignore the function of the bradyzoites, and
cuole (parasitophorous vacuole; PV) that separates it from the host their potential role in neuropsychiatric diseases and mental health,
cytoplasm, it induces the expression of a second round of effector would give us an incomplete picture of the successful lifecycle this
proteins. These proteins are exported via dense granules (DG) orga- parasite has evolved in order to survive and spread. Manipulating the
nelles into the host cell or become embedded in the PV membrane behaviour of the infected host to promote transmission is an observed
(PVM). The DG proteins (GRA) are associated with the maturation of characteristic in many parasitic infections (Koella, 2005; Lacroix et al.,
PV/PVM as well as the modulation of the host cell in order to manip- 2005; MacNeil et al., 2003; Sapolsky, 2003), but whether such ma-
ulate the immune signalling and upregulate host pro-survival pathways nipulation is a direct result of parasite molecules or due to an indirect
(Clough and Frickel, 2017; Mercier and Cesbron-Delauw, 2015). Four effect of the immune response against Toxoplasma in the brain remains
DG proteins identified to date manipulate host pathways, which result unclear. Distinguishing between events occurring in tachyzoite and
in an altered immune response and changes to cell survival pathways bradyzoite stages, and understanding host immune responses, is critical

83
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

to elucidate links between Toxoplasma infection and mental health. It that appear not to be able to persist (Fox et al., 2016; Uboldi et al.,
may also provide insights into how environmental factors (such as in- 2015). Using these strains, as well as others that are conditionally at-
fection), internal factors (the immune response) and genetics of the tenuated (Brown et al., 2017), in either growth or ability to export
host, interact to affect an individual’s health. proteins into the host neurons, may also help shed light on the role of
Toxoplasma in neuronal dysfunction.
10. Countermeasures for effects on the brain In this context, it is important to note that current research into the
role of Toxoplasma in mental health is mostly associative, and solid
Studies have tried to investigate the anti-Toxoplasma effect of anti- links between cause and effects have not been established. The next big
psychotic drugs, with an aim to counteract symptoms of SZ and/or BD, discoveries in this field would now focus on characterising specific
while at the same time targeting parasitism (Fond et al., 2015). Al- molecules that are responsible for the observed changes from both the
though anti-psychotics showed inhibitory activity of parasite replica- parasite and host side. In terms of developing a functional link between
tion in vitro (Fond et al., 2014; Goodwin et al., 2011; Jones-Brando Toxoplasma infection and human neuropsychiatric conditions then
et al., 2003), other studies failed to show any effects in vivo (Goodwin clinically available drugs would need to be developed and trials per-
et al., 2008; Saraei et al., 2015). A recent study by David et al. eval- formed to see if treatment leads to improvements in conditions. This
uated the efficacy of a ß-lactam antibiotic ceftriaxone, that rescues may not show anything as the damage by Toxoplasma (direct or in-
extracellular glutamate concentrations on neuronal pathology and directly through immune activation) may have already been enough to
function (David et al., 2016). Although ceftriaxone had a positive effect yield ongoing brain dysfunction. Nevertheless, developing an under-
on infection-induced glutamate dysregulation and was neuroprotective standing of the link between Toxoplasma and neuropsychiatric disease
in mice, it did not alter the immune response to the infection, neither is still important as it may provide impetus to develop vaccines to
did it affect mouse behaviour. More recently, efforts to develop vac- prevent infection and thus alleviating one environmental factor that
cines against infection has shown that Toxoplasma-expressed heat shock could be causing problems of brain function.
protein 70 induces massive iNOS expression and reduces brain para-
sitism, pointing towards a possible protective therapy against the Acknowledgements
parasite (Czarnewski et al., 2017). Nevertheless, effects of NO pro-
duction on behaviour remain to be determined, and further research is We would like to thank members of the Tonkin Laboratory for
required before any viable vaccine can be developed. Recently, a DNA helpful discussions that has contributed towards the preparation of this
vaccine based on a recombinant eukaryotic plasmid was shown to offer manuscript. The research in this laboratory is supported by The David
protective immunity in mice against the highly virulent RH strain (Chen Winston Turner Foundation, Melbourne. AJH is an NHMRC Principal
et al., 2017) Research Fellow.

11. Conclusions and future perspectives References

The current model of Toxoplasma-induced behavioural changes Abdoli, A., Dalimi, A., Movahedin, M., 2012. Impaired reproductive function of male rats
posits that a complex interplay between changes in the brain as well as infected with Toxoplasma gondii. Andrologia 44, 679–687. https://doi.org/10.1111/
j.1439-0272.2011.01249.x.
immune activation leads to an overall deficit in cognitive and affective Afonso, C., Paixão, V.B., Costa, R.M., 2012. Chronic toxoplasma infection modifies the
function, manifesting as neuropsychiatric dysfunction in the infected structure and the risk of host behavior. PLoS One 7. https://doi.org/10.1371/journal.
host. There may be changes in neuronal activity caused by the parasite pone.0032489.
Afonso, C., Paixão, V.B., Klaus, A., Lunghi, M., Piro, F., Emiliani, C., Di Cristina, M., Costa,
itself, an adaptation towards the inflammatory environment, and/or an R.M., 2017. Toxoplasma-induced changes in host risk behaviour are independent of
effect by the pro-inflammatory elements that cause collateral damage to parasite-derived AaaH2 tyrosine hydroxylase. Sci. Rep. 7, 13822. https://doi.org/10.
the neurons while the host actively fights the parasitism. Either way, 1038/s41598-017-13229-y.
Alday, H., Doggett, J., 2017. Drugs in development for toxoplasmosis: advances, chal-
there is no magic bullet that can relieve the host of the parasite infec- lenges, and current status. Drug Des. Devel. Ther. 11, 273–293. https://doi.org/10.
tion while at the same time preventing or reversing the neurological 2147/DDDT.S60973.
damage already caused. Research in the fairly new field of neu- Aliberti, J., 2005. Host persistence: exploitation of anti-inflammatory pathways by
Toxoplasma gondii. Nat. Rev. Immunol. 5, 162–170. https://doi.org/10.1038/
roimmune communication has already produced some exciting data on
nri1547.
how the peripheral immune system can communicate with the CNS Alvarado-Esquivel, C., Hernández-Tinoco, J., Sánchez-Anguiano, L.F., Ramos-Nevárez, A.,
using common molecular signalling cues, leading to dysregulation in Cerrillo-Soto, S.M., Sáenz-Soto, L., Liesenfeld, O., 2014. High seroprevalence of
brain functions. However, in terms of specific mechanisms related to Toxoplasma gondii infection in inmates: a case control study in Durango City.
Mexico. Eur. J. Microbiol. Immunol. 4, 76–82. https://doi.org/10.1556/EuJMI.4.
Toxoplasma infection, our understanding still remains naive. 2014.1.7.
An important aspect of Toxoplasma infection is its ability to export Arias, I., Sorlozano, A., Villegas, E., de Dios Luna, J., McKenney, K., Cervilla, J., Gutierrez,
effector proteins into the host cells (Coffey et al., 2016; Hakimi et al., B., Gutierrez, J., 2012. Infectious agents associated with schizophrenia: a meta-
analysis. Schizophr. Res. 136, 128–136. https://doi.org/10.1016/j.schres.2011.10.
2017). What is still unknown is which proteins, if any, Toxoplasma 026.
exports into the host neurons during the chronic stages and what are Arling, T.A., Yolken, R.H., Lapidus, M., Langenberg, P., Dickerson, F.B., Zimmerman, S.A.,
their effects on the function and survival of those neurons. Balis, T., Cabassa, J.A., Scrandis, D.A., Tonelli, L.H., Postolache, T.T., 2009.
Toxoplasma gondii antibody titers and history of suicide attempts in patients with
Another way in which a links could be established between recurrent mood disorders. J. Nerv. Ment. Dis. 197, 905–908. https://doi.org/10.
Toxoplasma and changes in the structure and function of the brain is by 1097/NMD.0b013e3181c29a23.
studying parasite clearance, or by not allowing chronic Toxoplasma Asgari, Q., Keshavarz Valian, H., Rezaeian, M., Shojaee, S., Mehrabani, D., 2015.
Toxoplasma gondii: sexual transmission in mice. J. Parasit. Dis. 39, 253–257. https://
infection to be established in the brain. A recent study suggests that
doi.org/10.1007/s12639-013-0330-5.
upon infection with an attenuated Toxoplasma strain, mice still dis- Bale, T.L., 2015. Epigenetic and transgenerational reprogramming of brain development.
played changes in behaviour, suggesting that an ongoing (chronic) in- Nat. Rev. Neurosci. 16, 332–344. https://doi.org/10.1038/nrn3818.
Barragan, A., Sibley, L.D., 2002. Transepithelial migration of Toxoplasma gondii is linked
fection may not be needed to alter the brain (Ingram et al., 2013).
to parasite motility and virulence. J. Exp. Med. 195, 1625–1633.
Unfortunately, complete clearance of Toxoplasma cysts by drugs is not Barthwal, M.K., Srivastava, N., Dikshit, M., 2001. Role of nitric oxide in a progressive
yet possible, but a few mouse studies have tested compounds that lead neurodegeneration model of Parkinson’s disease in the rat. Redox Rep. 6, 297–302.
to a reduction in brain cysts (Alday and Doggett, 2017). This is an https://doi.org/10.1179/135100001101536436.
Beal, M.F., Matson, W.R., Storey, E., Milbury, P., Ryan, E.A., Ogawa, T., Bird, E.D., 1992.
avenue of continued research in the hopes that clearing brain cysts Kynurenic acid concentrations are reduced in Huntington’s disease cerebral cortex.
might help alleviate mental illnesses. Furthermore, several parasite J.Neurol.Sci. 108, 80–87.
mutants have become available that have reduced chronic infection or Beaulieu, J.-M., Gainetdinov, R.R., 2011. The physiology, signaling, and pharmacology of

84
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

dopamine receptors. Pharmacol. Rev. 63. Chao, C.C., Anderson, W.R., Hu, S., Gekker, G., Martella, A., Peterson, P.K., 1993.
Beckman, J.S., Koppenol, W.H., 1996. Nitric oxide, superoxide, and peroxynitrite: the Activated microglia inhibit multiplication of Toxoplasma gondii via a nitric oxide
good, the bad, and ugly. Am. J. Physiol. 271, C1424–37. mechanism. Clin. Immunol. Immunopathol. 67, 178–183.
Bennett, S., Grant, M.M., Aldred, S., 2009. Oxidative stress in vascular dementia and Chen, K., Wang, J.-L., Huang, S.-Y., Yang, W.-B., Zhu, W.-N., Zhu, X.-Q., 2017. Immune
Alzheimer’s disease: a common pathology. J. Alzheimers Dis. 17, 245–257. https:// responses and protection after DNA vaccination against Toxoplasma gondii calcium-
doi.org/10.3233/JAD-2009-1041. dependent protein kinase 2 (TgCDPK2). Parasite 24, 41. https://doi.org/10.1051/
Berdoy, M., Webster, J.P., Macdonald, D.W., 2000. Fatal attraction in rats infected with parasite/2017045.
Toxoplasma gondii. Proc. R. Soc. B Biol. Sci. 267, 1591–1594. https://doi.org/10. Chen, X., Nelson, C.D., Li, X., Winters, C.A., Azzam, R., Sousa, A.A., Leapman, R.D.,
1098/rspb.2000.1182. Gainer, H., Sheng, M., Reese, T.S., 2011. PSD-95 is required to sustain the molecular
Berenreiterová, M., Flegr, J., Kuběna, A.A., Němec, P., Witter, M., 2011. The distribution organization of the postsynaptic density. J. Neurosci. 31, 6329–6338. https://doi.
of toxoplasma gondii cysts in the brain of a mouse with latent toxoplasmosis: im- org/10.1523/JNEUROSCI.5968-10.2011.
plications for the behavioral manipulation hypothesis. PLoS One 6https://doi.org/10. Choi, D.W., 1994. Glutamate receptors and the induction of excitotoxic neuronal death.
1371/journal.pone.0028925. e28925. Prog. Brain Res. 100, 47–51.
Beste, C., Getzmann, S., Gajewski, P.D., Golka, K., Falkenstein, M., 2014. Latent Clough, B., Frickel, E.-M., 2017. The toxoplasma parasitophorous vacuole: an evolving
Toxoplasma gondii infection leads to deficits in goal-directed behavior in healthy host-parasite frontier. Trends Parasitol. 33, 473–488. https://doi.org/10.1016/j.pt.
elderly. Neurobiol. Aging 35, 1037–1044. https://doi.org/10.1016/j.neurobiolaging. 2017.02.007.
2013.11.012. Coffey, M.J., Jennison, C., Tonkin, C.J., Boddey, J.A., 2016. Role of the ER and Golgi in
Blanpied, T.A., Ehlers, M.D., 2004. Microanatomy of dendritic spines: emerging principles protein export by Apicomplexa. Curr. Opin. Cell Biol. 41, 18–24. https://doi.org/10.
of synaptic pathology in psychiatric and neurological disease. Biol. Psychiatry 55, 1016/j.ceb.2016.03.007.
1121–1127. https://doi.org/10.1016/j.biopsych.2003.10.006. Contopoulos-Ioannidis, D., Wheeler, K.M., Ramirez, R., Press, C., Mui, E., Zhou, Y., Van
Bö, L., Dawson, T.M., Wesselingh, S., Mörk, S., Choi, S., Kong, P.A., Hanley, D., Trapp, Tubbergen, C., Prasad, S., Maldonado, Y., Withers, S., Boyer, K.M., Noble, A.G.,
B.D., 1994. Induction of nitric oxide synthase in demyelinating regions of multiple Rabiah, P., Swisher, C.N., Heydemann, P., Wroblewski, K., Karrison, T., Grigg, M.E.,
sclerosis brains. Ann. Neurol. 36, 778–786. https://doi.org/10.1002/ana. Montoya, J.G., McLeod, R., 2015. Clustering of toxoplasma gondii infections within
410360515. families of congenitally infected infants. Clin. Infect. Dis. 61, 1815–1824. https://doi.
Boothroyd, J.C., Dubremetz, J.-F., 2008. Kiss and spit: the dual roles of Toxoplasma org/10.1093/cid/civ721.
rhoptries. Nat. Rev. Microbiol. 6, 79–88. https://doi.org/10.1038/nrmicro1800. Contreras-Ochoa, C.O., Lagunas-Martínez, A., Belkind-Gerson, J., Díaz-Chávez, J., Correa,
Bougdour, A., Durandau, E., Brenier-Pinchart, M.-P., Ortet, P., Barakat, M., Kieffer, S., D., 2013. Toxoplasma gondii invasion and replication within neonate mouse astro-
Curt-Varesano, A., Curt-Bertini, R.-L., Bastien, O., Coute, Y., Pelloux, H., Hakimi, M.- cytes and changes in apoptosis related molecules. Exp. Parasitol. 134, 256–265.
A., 2013. Host cell subversion by Toxoplasma GRA16, an exported dense granule https://doi.org/10.1016/j.exppara.2013.03.010.
protein that targets the host cell nucleus and alters gene expression. Cell Host Cook, T.B., Brenner, L.A., Cloninger, C.R., Langenberg, P., Igbide, A., Giegling, I.,
Microbe 13, 489–500. https://doi.org/10.1016/j.chom.2013.03.002. Hartmann, A.M., Konte, B., Friedl, M., Brundin, L., Groer, M.W., Can, A., Rujescu, D.,
Bradley, P.J., Sibley, L.D., 2007. Rhoptries: an arsenal of secreted virulence factors. Curr. Postolache, T.T., 2015. “Latent” infection with Toxoplasma gondii: association with
Opin. Microbiol. 10, 582–587. https://doi.org/10.1016/j.mib.2007.09.013. trait aggression and impulsivity in healthy adults. J. Psychiatr. Res. 60, 87–94.
Braun, L., Brenier-Pinchart, M.-P., Yogavel, M., Curt-Varesano, A., Curt-Bertini, R.-L., https://doi.org/10.1016/j.jpsychires.2014.09.019.
Hussain, T., Kieffer-Jaquinod, S., Coute, Y., Pelloux, H., Tardieux, I., Sharma, A., Croarkin, P.E., Levinson, A.J., Daskalakis, Z.J., 2011. Evidence for GABAergic inhibitory
Belrhali, H., Bougdour, A., Hakimi, M.-A., 2013. A Toxoplasma dense granule protein, deficits in major depressive disorder. Neurosci. Biobehav. Rev. 35, 818–825. https://
GRA24, modulates the early immune response to infection by promoting a direct and doi.org/10.1016/j.neubiorev.2010.10.002.
sustained host p38 MAPK activation. J. Exp. Med. 210, 2071–2086. https://doi.org/ Cyr, M., Sotnikova, T.D., Gainetdinov, R.R., Caron, M.G., 2006. Dopamine enhances
10.1084/jem.20130103. motor and neuropathological consequences of polyglutamine expanded huntingtin.
Brooks, J.M., Carrillo, G.L., Su, J., Lindsay, D.S., Fox, M.A., Blader, I.J., 2015. Toxoplasma FASEB J. 20, 2541–2543. https://doi.org/10.1096/fj.06-6533fje.
gondii infections alter GABAergic synapses and signaling in the central nervous Czarnewski, P., Araújo, E.C.B., Oliveira, M.C., Mineo, T.W.P., Silva, N.M., 2017.
system. MBio 6, 1–9. https://doi.org/10.1128/mBio.01428-15. Recombinant TgHSP70 immunization protects against toxoplasma gondii brain cyst
Brown, A.S., Derkits, E.J., 2010. Prenatal infection and schizophrenia: a review of epi- formation by enhancing inducible nitric oxide expression. Front. Cell. Infect.
demiologic and translational studies. Am. J. Psychiatry 167, 261–280. https://doi. Microbiol. 7, 142. https://doi.org/10.3389/fcimb.2017.00142.
org/10.1176/appi.ajp.2009.09030361. Daikhin, Y., Yudkoff, M., 2000. Compartmentation of brain glutamate metabolism in
Brown, A.S., Schaefer, C.A., Quesenberry, C.P., Liu, L., Babulas, V.P., Susser, E.S., 2005. neurons and glia. J. Nutr. 130 1026S–31S.
Maternal exposure to toxoplasmosis and risk of Schizophrenia in adult offspring. Am. Dalimi, A., Abdoli, A., 2013. Toxoplasma gondii and male reproduction impairment: a
J. Psychiatry 162, 767–773. https://doi.org/10.1176/appi.ajp.162.4.767. new aspect of toxoplasmosis research. Jundishapur J. Microbiol. 6, 1–5. https://doi.
Brown, A.S., Schaefer, C.A., Wyatt, R.J., Goetz, R., Begg, M.D., Gorman, J.M., Susser, E.S., org/10.5812/jjm.7184.
2000. Maternal exposure to respiratory infections and adult schizophrenia spectrum Daniels, B.P., Sestito, S.R., Rouse, S.T., 2014. An expanded task battery in the Morris
disorders: a prospective birth cohort study. Schizophr. Bull. 26, 287–295. water maze reveals effects of Toxoplasma gondii infection on learning and memory in
Brown, A.S., Vinogradov, S., Kremen, W.S., Poole, J.H., Deicken, R.F., Penner, J.D., rats. Parasitol. Int. 64, 5–12. https://doi.org/10.1016/j.parint.2014.09.002.
McKeague, I.W., Kochetkova, A., Kern, D., Schaefer, C.A., 2009. Prenatal exposure to Dauvermann, M.R., Lee, G., Dawson, N., 2017. Glutamatergic regulation of cognition and
maternal infection and executive dysfunction in adult schizophrenia. Am. J. functional brain connectivity: insights from pharmacological, genetic and transla-
Psychiatry 166, 683–690. https://doi.org/10.1176/appi.ajp.2008.08010089. tional schizophrenia research. Br. J. Pharmacol. 174, 3136–3160. https://doi.org/10.
Brown, K.M., Long, S., Sibley, L.D., 2017. Plasma membrane association by N-acylation 1111/bph.13919.
governs PKG function inToxoplasma gondii. MBio 8, e00375–17. https://doi.org/10. David, C.N., Frias, E.S., Szu, J.I., Vieira, P.A., Hubbard, J.A., Lovelace, J., Michael, M.,
1128/mBio.00375-17. Worth, D., McGovern, K.E., Ethell, I.M., Stanley, B.G., Korzus, E., Fiacco, T.A., Binder,
Burke, J.M., Roberts, C.W., Hunter, C.A., Murray, M., Alexander, J., 1994. Temporal D.K., Wilson, E.H., 2016. GLT-1-dependent disruption of CNS glutamate homeostasis
differences in the expression of mRNA for IL-10 and IFN-gamma in the brains and and neuronal function by the protozoan parasite toxoplasma gondii. PLoS Pathog. 12,
spleens of C57BL/10 mice infected with Toxoplasma gondii. Parasite Immunol. 16, 1–29. https://doi.org/10.1371/journal.ppat.1005643.
305–314. de Bittencourt, P.R., Adamolekum, B., Bharucha, N., Carpio, A., Cossío, O.H., Danesi,
Cabral, C.M., McGovern, K.E., MacDonald, W.R., Franco, J., Koshy, A.A., 2017. Dissecting M.A., Dumas, M., Meinardi, H., Ordinario, A., Senanayake, N., Shakir, R., Sotelo, J.,
amyloid beta deposition using distinct strains of the neurotropic parasite Toxoplasma 1996. Epilepsy in the tropics: I. Epidemiology, socioeconomic risk factors, and
gondii as a novel tool. ASN Neuro 9, 175909141772491. https://doi.org/10.1177/ etiology. Epilepsia 37, 1121–1127.
1759091417724915. Del Grande, C., Galli, L., Schiavi, E., Dell’Osso, L., Bruschi, F., 2017. Is Toxoplasma gondii
Cabral, C.M., Tuladhar, S., Dietrich, H.K., Nguyen, E., MacDonald, W.R., Trivedi, T., a Trigger of Bipolar Disorder? Pathogens 6, 3. https://doi.org/10.3390/
Devineni, A., Koshy, A.A., 2016. Neurons are the primary target cell for the brain- pathogens6010003.
tropic intracellular parasite Toxoplasma gondii. PLoS Pathog. 12, 1–20. https://doi. del Rey, A., Balschun, D., Wetzel, W., Randolf, A., Besedovsky, H.O., 2013. A cytokine
org/10.1371/journal.ppat.1005447. network involving brain-borne IL-1β, IL-1ra, IL-18, IL-6, and TNFα operates during
Campos, A.C., Vaz, G.N., Saito, V.M., Teixeira, A.L., 2014. Further evidence for the role of long-term potentiation and learning. Brain Behav. Immun. 33, 15–23. https://doi.
interferon-gamma on anxiety- and depressive-like behaviors: involvement of hippo- org/10.1016/j.bbi.2013.05.011.
campal neurogenesis and NGF production. Neurosci. Lett. 578, 100–105. https://doi. Dellacasa-Lindberg, I., Fuks, J.M., Arrighi, R.B.G., Lambert, H., Wallin, R.P.A., Chambers,
org/10.1016/j.neulet.2014.06.039. B.J., Barragan, A., 2011. Migratory activation of primary cortical microglia upon
Canetta, S.E., Brown, A.S., 2012. Prenatal infection, maternal immune activation, and risk infection with toxoplasma gondii. Infect. Immun. 79, 3046–3052. https://doi.org/10.
for schizophrenia. Transl. Neurosci. 3, 320–327. https://doi.org/10.2478/s13380- 1128/IAI.01042-10.
012-0045-6. Delorme-Walker, V., Abrivard, M., Lagal, V., Anderson, K., Perazzi, A., Gonzalez, V., Page,
Carson, M.J., Doose, J.M., Melchior, B., Schmid, C.D., Ploix, C.C., 2006. CNS immune C., Chauvet, J., Ochoa, W., Volkmann, N., Hanein, D., Tardieux, I., 2012. Toxofilin
privilege: hiding in plain sight. Immunol. Rev. 213, 48–65. https://doi.org/10.1111/ upregulates the host cortical actin cytoskeleton dynamics, facilitating Toxoplasma
j.1600-065X.2006.00441.x. invasion. J. Cell. Sci. 125, 4333–4342. https://doi.org/10.1242/jcs.103648.
Carter, C.J., 2011. Schizophrenia: a pathogenetic autoimmune disease caused by viruses Deng, X.-H., Ai, W.-M., Lei, D.-L., Luo, X.-G., Yan, X.-X., Li, Z., 2012. Lipopolysaccharide
and pathogens and dependent on genes. J. Pathog. 2011, 128318. https://doi.org/10. induces paired immunoglobulin-like receptor B (PirB) expression, synaptic alteration,
4061/2011/128318. and learning-memory deficit in rats. Neuroscience 209, 161–170. https://doi.org/10.
Cervenka, I., Agudelo, L.Z., Ruas, J.L., 2017. Kynurenines: Tryptophan’s metabolites in 1016/j.neuroscience.2012.02.022.
exercise, inflammation, and mental health. Science 80, 357. https://doi.org/10. Denkers, E.Y., Butcher, B.A., Del Rio, L., Bennouna, S., 2004. Neutrophils, dendritic cells
1126/science.aaf9794. eaaf9794. and Toxoplasma. Int. J. Parasitol. 34, 411–421. https://doi.org/10.1016/j.ijpara.

85
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

2003.11.001. Flegr, J., Klapilova, K., Kankova, S., 2014. Toxoplasmosis can be a sexually transmitted
Di Chiara, G., Bassareo, V., 2007. Reward system and addiction: what dopamine does and infection with serious clinical consequences. Not all routes of infection are created
doesn’t do. Curr. Opin. Pharmacol. 7, 69–76. https://doi.org/10.1016/j.coph.2006. equal. Med. Hypotheses 83, 286–289. https://doi.org/10.1016/j.mehy.2014.05.019.
11.003. Flegr, J., Klose, J., Novotná, M., Berenreitterová, M., Havlíček, J., 2009. Increased in-
Di Cristina, M., Marocco, D., Galizi, R., Proietti, C., Spaccapelo, R., Crisanti, A., 2008. cidence of traffic accidents in Toxoplasma-infected military drivers and protective
Temporal and spatial distribution of toxoplasma gondii differentiation into effect RhD molecule revealed by a large-scale prospective cohort study. BMC Infect.
Bradyzoites and tissue cyst formation in vivo. Infect. Immun. 76, 3491–3501. https:// Dis. 9, 72. https://doi.org/10.1186/1471-2334-9-72.
doi.org/10.1128/IAI.00254-08. Flegr, J., Preiss, M., Klose, J., Havlícek, J., Vitáková, M., Kodym, P., 2003. Decreased level
Dincel, G.C., Atmaca, H.T., 2015. Nitric oxide production increases during Toxoplasma of psychobiological factor novelty seeking and lower intelligence in men latently
gondii encephalitis in mice. Exp. Parasitol. 156, 104–112. https://doi.org/10.1016/j. infected with the protozoan parasite Toxoplasma gondii Dopamine, a missing link
exppara.2015.06.009. between schizophrenia and toxoplasmosis? Biol. Psychol. 63, 253–268.
Dinel, A.L., André, C., Aubert, A., Ferreira, G., Layé, S., Castanon, N., 2014. Fond, G., Boyer, L., Gaman, A., Laouamri, H., Attiba, D., Richard, J.-R., Delavest, M.,
Lipopolysaccharide-induced brain activation of the indoleamine 2,3-dioxygenase and Houenou, J., Le Corvoisier, P., Charron, D., Krishnamoorthy, R., Oliveira, J.,
depressive-like behavior are impaired in a mouse model of metabolic syndrome. Tamouza, R., Yolken, R., Dickerson, F., Leboyer, M., Hamdani, N., 2015. Treatment
Psychoneuroendocrinology 40, 48–59. https://doi.org/10.1016/j.psyneuen.2013.10. with anti-toxoplasmic activity (TATA) for toxoplasma positive patients with bipolar
014. disorders or schizophrenia: a cross-sectional study. J. Psychiatr. Res. 63, 58–64.
Donley, D.W., Olson, A.R., Raisbeck, M.F., Fox, J.H., Gigley, J.P., 2016. Huntingtons https://doi.org/10.1016/j.jpsychires.2015.02.011.
disease mice infected with toxoplasma gondii demonstrate early kynurenine pathway Fond, G., Macgregor, A., Tamouza, R., Hamdani, N., Meary, A., Leboyer, M., Dubremetz,
activation, altered CD8+ T-Cell responses, and premature mortality. PLoS One 11, J.-F., 2014. Comparative analysis of anti-toxoplasmic activity of antipsychotic drugs
1–16. https://doi.org/10.1371/journal.pone.0162404. and valproate. Eur. Arch. Psychiatry Clin. Neurosci. 264, 179–183. https://doi.org/
Dubey, J., 2009. Toxoplasmosis of Animals and Humans, second edition. CRC 10.1007/s00406-013-0413-4.
Presshttps://doi.org/10.1201/9781420092370. Fonseca, M.I., Kawas, C.H., Troncoso, J.C., Tenner, A.J., 2004. Neuronal localization of
Dubey, J.P., 2008. The history of Toxoplasma gondii–the first 100 years. J. Eukaryot. C1q in preclinical Alzheimer’s disease. Neurobiol. Dis. 15, 40–46. https://doi.org/10.
Microbiol. 55, 467–475. https://doi.org/10.1111/j.1550-7408.2008.00345.x. 1016/j.nbd.2003.09.004.
Dubey, J.P., Ferreira, L.R., Alsaad, M., Verma, S.K., Alves, D.A., Holland, G.N., McConkey, Fox, B.A., Rommereim, L.M., Guevara, R.B., Falla, A., Hortua Triana, M.A., Sun, Y., Bzik,
G.A., 2016. Experimental toxoplasmosis in rats induced orally with eleven strains of D.J., 2016. The Toxoplasma gondii rhoptry kinome is essential for chronic infection.
toxoplasma gondii of seven genotypes: tissue tropism, tissue cyst size, neural lesions, MBio 7, 1–12. https://doi.org/10.1128/mBio.00193-16.
tissue cyst rupture without reactivation, and ocular lesions. PLoS One 11https://doi. Franco, M., Panas, M.W., Marino, N.D., Lee, M.-C.W., Buchholz, K.R., Kelly, F.D.,
org/10.1371/journal.pone.0156255. e0156255. Bednarski, J.J., Sleckman, B.P., Pourmand, N., Boothroyd, J.C., 2016. A novel se-
Dvorakova-Hortova, K., Sidlova, A., Ded, L., Hladovcova, D., Vieweg, M., Weidner, W., creted protein, MYR1, is central to toxoplasma’s manipulation of host cells. MBio
Steger, K., Stopka, P., Paradowska-Dogan, A., 2014. Toxoplasma gondii decreases the 7https://doi.org/10.1128/mBio.02231-15. e02231-15.
reproductive fitness in mice. PLoS One 9. https://doi.org/10.1371/journal.pone. Freedman, D., Bao, Y., Shen, L., Schaefer, C.A., Brown, A.S., 2016. Maternal T. gondii,
0096770. offspring bipolar disorder and neurocognition. Psychiatry Res. 243, 382–389.
Ebadi, M., Akhlaghi, H., Zamani, M.M., Beheshti, H., Abolhassani, H., Ayadi, A., Dezdar, https://doi.org/10.1016/j.psychres.2016.06.057.
J., Mortazzavi, S.H., Karami, G.R., Izadi, M., 2014. The correlation between tox- Fuglewicz, A., Piotrowski, P., Stodolak, A., 2017. Relationship between toxoplasmosis
oplama gondii infection and schizophrenia: a comparative study with family mem- and schizophrenia: a review. Adv. Clin. Exp. Med. 26, 1033–1038. https://doi.org/
bers (Control group). Scimetr 2, 1–5. https://doi.org/10.5812/scimetr.15386. 10.17219/acem/61435.
Eells, J.B., Varela-Stokes, A., Guo-Ross, S.X., Kummari, E., Smith, H.M., Cox, E., Lindsay, Fuhrman, S.A., Joiner, K.A., 1989. Toxoplasma gondii: mechanism of resistance to
D.S., 2015. Chronic toxoplasma gondii in nurr1-null heterozygous mice exacerbates complement-mediated killing. J. Immunol. 142, 940–947.
elevated open field activity. PLoS One 10, 1–26. https://doi.org/10.1371/journal. Fujigaki, S., Takemura, M., Hamakawa, H., Seishima, M., Saito, K., 2003. The mechanism
pone.0119280. of interferon-gamma induced anti Toxoplasma gondii by indoleamine 2,3-dioxy-
Eichler, E.E., Flint, J., Gibson, G., Kong, A., Leal, S.M., Moore, J.H., Nadeau, J.H., 2010. genase and/or inducible nitric oxide synthase vary among tissues. Adv. Exp. Med.
Missing heritability and strategies for finding the underlying causes of complex dis- Biol. 527, 97–103.
ease. Nat. Rev. Genet. 11, 446–450. https://doi.org/10.1038/nrg2809. Fuks, J.M., Arrighi, R.B.G., Weidner, J.M., Kumar Mendu, S., Jin, Z., Wallin, R.P.A., Rethi,
Elsheikha, H.M., Büsselberg, D., Zhu, X.Q., 2016. The known and missing links between B., Birnir, B., Barragan, A., 2012. GABAergic signaling is linked to a hypermigratory
Toxoplasma gondii and schizophrenia. Metab. Brain Dis. 31, 749–759. https://doi. phenotype in dendritic cells infected by toxoplasma gondii. PLoS Pathog. 8https://
org/10.1007/s11011-016-9822-1. doi.org/10.1371/journal.ppat.1003051. e1003051.
Erhardt, S., Blennow, K., Nordin, C., Skogh, E., Lindström, L.H., Engberg, G., 2001. Fung, T.C., Olson, C.A., Hsiao, E.Y., 2017. Interactions between the microbiota, immune
Kynurenic acid levels are elevated in the cerebrospinal fluid of patients with schi- and nervous systems in health and disease. Nat. Neurosci. 20, 145–155. https://doi.
zophrenia. Neurosci. Lett. 313, 96–98. org/10.1038/nn.4476.
Erhardt, S., Lim, C.K., Linderholm, K.R., Janelidze, S., Lindqvist, D., Samuelsson, M., Gale, S.D., Brown, B.L., Berrett, A., Erickson, L.D., Hedges, D.W., 2014. Association be-
Lundberg, K., Postolache, T.T., Träskman-Bendz, L., Guillemin, G.J., Brundin, L., tween latent toxoplasmosis and major depression, generalised anxiety disorder and
2013. Connecting inflammation with glutamate agonism in suicidality. panic disorder in human adults. Folia Parasitol. (Praha) 61, 285–292.
Neuropsychopharmacology 38, 743–752. https://doi.org/10.1038/npp.2012.248. Garry, P.S., Ezra, M., Rowland, M.J., Westbrook, J., Pattinson, K.T.S., 2015. The role of
Erusalimsky, J.D., Moncada, S., 2007. Nitric oxide and mitochondrial signaling: from the nitric oxide pathway in brain injury and its treatment - from bench to bedside.
physiology to pathophysiology. Arterioscler. Thromb. Vasc. Biol. 27, 2524–2531. Exp. Neurol. 263, 235–243. https://doi.org/10.1016/j.expneurol.2014.10.017.
https://doi.org/10.1161/ATVBAHA.107.151167. Gaskell, E.A., Smith, J.E., Pinney, J.W., Westhead, D.R., McConkey, G.A., 2009. A unique
Esshili, A., Thabet, S., Jemli, A., Trifa, F., Mechri, A., Zaafrane, F., Gaha, L., Juckel, G., dual activity amino acid hydroxylase in Toxoplasma gondii. PLoS One 4, 1–10.
Babba, H., Hadj, Bel, Jrad, B., 2016. Toxoplasma gondii infection in schizophrenia https://doi.org/10.1371/journal.pone.0004801.
and associated clinical features. Psychiatry Res. 245, 327–332. https://doi.org/10. Gatkowska, J., Wieczorek, M., Dziadek, B., Dzitko, K., Dlugonska, H., 2012. Behavioral
1016/j.psychres.2016.08.056. changes in mice caused by Toxoplasma gondii invasion of brain. Parasitol. Res. 111,
Ferguson, D.J., Huskinson-Mark, J., Araujo, F.G., Remington, J.S., 1994. A morphological 53–58. https://doi.org/10.1007/s00436-011-2800-y.
study of chronic cerebral toxoplasmosis in mice: comparison of four different strains Gatto, E.M., Riobó, N.A., Carreras, M.C., Cherñavsky, A., Rubio, A., Satz, M.L., Poderoso,
of Toxoplasma gondii. Parasitol. Res. 80, 493–501. J.J., 2000. Overexpression of neutrophil neuronal nitric oxide synthase in parkinson’s
Ferguson, D.J., Hutchison, W.M., 1987. An ultrastructural study of the early development disease. Nitric Oxide 4, 534–539. https://doi.org/10.1006/niox.2000.0288.
and tissue cyst formation of Toxoplasma gondii in the brains of mice. Parasitol. Res. Gay, G., Braun, L., Brenier-Pinchart, M.-P., Vollaire, J., Josserand, V., Bertini, R.-L.,
73, 483–491. Varesano, A., Touquet, B., De Bock, P.-J., Coute, Y., Tardieux, I., Bougdour, A.,
Filiano, A.J., Gadani, S.P., Kipnis, J., Scientist, M., Program, T., 2015. Interactions of Hakimi, M.-A., 2016. Toxoplasma gondii TgIST co-opts host chromatin repressors
innate and adaptive immunity in brain development and function. Brain Res. 1617, dampening STAT1-dependent gene regulation and IFN-γ-mediated host defenses. J.
18–27. https://doi.org/10.1016/j.brainres.2014.07.050.Interactions. Exp. Med. 213, 1779–1798. https://doi.org/10.1084/jem.20160340.
Filiano, A.J., Xu, Y., Tustison, N.J., Marsh, R.L., Baker, W., Smirnov, I., Overall, C.C., Gazzinelli, R.T., Eltoum, I., Wynn, T.A., Sher, A., 1993. Acute cerebral toxoplasmosis is
Gadani, S.P., Turner, S.D., Weng, Z., Peerzade, S.N., Chen, H., Lee, K.S., Scott, M.M., induced by in vivo neutralization of TNF-alpha and correlates with the down-regu-
Beenhakker, M.P., Litvak, V., Kipnis, J., 2016. Unexpected role of interferon-γ in lated expression of inducible nitric oxide synthase and other markers of macrophage
regulating neuronal connectivity and social behaviour. Nature 535, 425–429. activation. J. Immunol. 151, 3672–3681.
https://doi.org/10.1038/nature18626. Giakoumelou, S., Wheelhouse, N., Cuschieri, K., Entrican, G., Howie, S.E.M., Horne, A.W.,
Fischer, H.G., Nitzgen, B., Reichmann, G., Hadding, U., 1997. Cytokine responses induced 2016. The role of infection in miscarriage. Hum. Reprod. Update 22, 116–133.
by Toxoplasma gondii in astrocytes and microglial cells. Eur. J. Immunol. 27, https://doi.org/10.1093/humupd/dmv041.
1539–1548. https://doi.org/10.1002/eji.1830270633. Girault, J.-A., Greengard, P., 2004. The neurobiology of dopamine signaling. Arch.
Flegr, J., 2013. Influence of latent Toxoplasma infection on human personality, phy- Neurol. 61, 641. https://doi.org/10.1001/archneur.61.5.641.
siology and morphology: pros and cons of the Toxoplasma-human model in studying Gizer, I.R., Ficks, C., Waldman, I.D., 2009. Candidate gene studies of ADHD: a meta-
the manipulation hypothesis. J. Exp. Biol. 216, 127–133. https://doi.org/10.1242/ analytic review. Hum. Genet. 126, 51–90. https://doi.org/10.1007/s00439-009-
jeb.073635. 0694-x.
Flegr, J., 2010. Influence of latent toxoplasmosis on the phenotype of intermediate hosts. Glantz, L.A., Lewis, D.A., 2000. Decreased dendritic spine density on prefrontal cortical
Folia Parasitol. (Praha). 57, 81–87. pyramidal neurons in schizophrenia. Arch. Gen. Psychiatry 57, 65–73.
Flegr, J., 2007. Effects of toxoplasma on human behavior. Schizophr. Bull. 33, 757–760. Goebel, S., Gross, U., Lüder, C.G., 2001. Inhibition of host cell apoptosis by Toxoplasma
https://doi.org/10.1093/schbul/sbl074. gondii is accompanied by reduced activation of the caspase cascade and alterations of

86
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

poly(ADP-ribose) polymerase expression. J. Cell. Sci. 114, 3495–3505. R.P., Suzuki, Y., Reardon, C., Hickey, A.W., Chen, L., McLeod, R., 2008. Neurological
Gonzalez, L.E., Rojnik, B., Urrea, F., Urdaneta, H., Petrosino, P., Colasante, C., Pino, S., and behavioral abnormalities, ventricular dilatation, altered cellular functions, in-
Hernandez, L., 2007. Toxoplasma gondii infection lower anxiety as measured in the flammation, and neuronal injury in brains of mice due to common, persistent,
plus-maze and social interaction tests in rats. A behavioral analysis. Behav. Brain Res. parasitic infection. J. Neuroinflammation 5, 48. https://doi.org/10.1186/1742-2094-
177, 70–79. https://doi.org/10.1016/j.bbr.2006.11.012. 5-48.
Goodwin, D., Hrubec, T.C., Klein, B.G., Strobl, J.S., Werre, S.R., Han, Q., Zajac, A.M., Hidano, S., Randall, L.M., Dawson, L., Dietrich, H.K., Konradt, C., Klover, P.J., John, B.,
Lindsay, D.S., 2012. Congenital infection of mice with toxoplasma gondii induces Harris, T.H., Fang, Q., Turek, B., Kobayashi, T., Hennighausen, L., Beiting, D.P.,
minimal change in behavior and no change in neurotransmitter concentrations. J. Koshy, A.A., Hunter, C.A., 2016. STAT1 signaling in astrocytes is essential for control
Parasitol. 98, 706–712. https://doi.org/10.1645/GE-3068.1. of infection in the central nervous system. MBio 7, e01881–16. https://doi.org/10.
Goodwin, D.G., Strobl, J., Mitchell, S.M., Zajac, A.M., Lindsay, D.S., 2008. Evaluation of 1128/mBio.01881-16.
the mood-stabilizing agent valproic acid as a preventative for toxoplasmosis in mice Hilmas, C., Pereira, E.F., Alkondon, M., Rassoulpour, A., Schwarcz, R., Albuquerque, E.X.,
and activity against tissue cysts in mice. J. Parasitol. 94, 555–557. https://doi.org/ 2001. The brain metabolite kynurenic acid inhibits alpha7 nicotinic receptor activity
10.1645/GE-1331.1. and increases non-alpha7 nicotinic receptor expression: physiopathological implica-
Goodwin, D.G., Strobl, J.S., Lindsay, D.S., 2011. Evaluation of five antischizophrenic tions. J. Neurosci. 21, 7463–7473.
agents against Toxoplasma gondii in human cell cultures. J. Parasitol. 97, 148–151. Hodkova, H., Kodym, P., Flegr, J., 2007. Poorer results of mice with latent toxoplasmosis
https://doi.org/10.1645/GE-2536.1. in learning tests: impaired learning processes or the novelty discrimination me-
Graumann, K., Hippe, D., Gross, U., Lüder, C.G.K., 2009. Mammalian apoptotic signalling chanism? Parasitology 134, 1329–1337. https://doi.org/10.1017/
pathways: multiple targets of protozoan parasites to activate or deactivate host cell S0031182007002673.
death. Microbes Infect. 11, 1079–1087. https://doi.org/10.1016/j.micinf.2009.08. Hong, S., Beja-Glasser, V.F., Nfonoyim, B.M., Frouin, A., Li, S., Ramakrishnan, S., Merry,
011. K.M., Shi, Q., Rosenthal, A., Barres, B.A., Lemere, C.A., Selkoe, D.J., Stevens, B.,
Greengard, P., 2001. The neurobiology of dopamine signaling. Biosci. Rep. 21, 247–269. 2016. Complement and microglia mediate early synapse loss in Alzheimer mouse
Guidetti, P., Bates, G.P., Graham, R.K., Hayden, M.R., Leavitt, B.R., MacDonald, M.E., models. Science 352 (80-), 712–716. https://doi.org/10.1126/science.aad8373.
Slow, E.J., Wheeler, V.C., Woodman, B., Schwarcz, R., 2006. Elevated brain 3-hy- Horacek, J., Flegr, J., Tintera, J., Verebova, K., Spaniel, F., Novak, T., Brunovsky, M.,
droxykynurenine and quinolinate levels in Huntington disease mice. Neurobiol. Dis. Bubenikova-Valesova, V., Holub, D., Palenicek, T., Höschl, C., 2012. Latent tox-
23, 190–197. https://doi.org/10.1016/j.nbd.2006.02.011. oplasmosis reduces gray matter density in schizophrenia but not in controls: voxel-
Guidetti, P., Luthi-Carter, R.E., Augood, S.J., Schwarcz, R., 2004. Neostriatal and cortical based-morphometry (VBM) study. World J. Biol. Psychiatry 13, 501–509. https://doi.
quinolinate levels are increased in early grade Huntington’s disease. Neurobiol. Dis. org/10.3109/15622975.2011.573809.
17, 455–461. https://doi.org/10.1016/j.nbd.2004.07.006. Hunot, S., Boissière, F., Faucheux, B., Brugg, B., Mouatt-Prigent, A., Agid, Y., Hirsch, E.C.,
Guillermo, L.V.C., DaMatta, R.A., 2004. Nitric oxide inhibition after Toxoplasma gondii 1996. Nitric oxide synthase and neuronal vulnerability in Parkinson’s disease.
infection of chicken macrophage cell lines. Poult. Sci. 83, 776–782. Neuroscience 72, 355–363.
Gulaj, E., Pawlak, K., Bien, B., Pawlak, D., 2010. Kynurenine and its metabolites in Hunter, C.A., Roberts, C.W., Alexander, J., 1992a. Kinetics of cytokine mRNA production
Alzheimer’s disease patients. Adv. Med. Sci. 55, 204–211. https://doi.org/10.2478/ in the brains of mice with progressive toxoplasmic encephalitis. Eur. J. Immunol. 22,
v10039-010-0023-6. 2317–2322. https://doi.org/10.1002/eji.1830220921.
Gulinello, M., Acquarone, M., Kim, J.H., Spray, D.C., Barbosa, H.S., Sellers, R., Tanowitz, Hunter, C.A., Roberts, C.W., Murray, M., Alexander, J., 1992b. Detection of cytokine
H.B., Weiss, L.M., 2010. Acquired infection with Toxoplasma gondii in adult mice mRNA in the brains of mice with toxoplasmic encephalitis. Parasite Immunol. 14,
results in sensorimotor deficits but normal cognitive behavior despite widespread 405–413.
brain pathology. Microbes Infect. 12, 528–537. https://doi.org/10.1016/j.micinf. Hunter, C.A., Sibley, L.D., 2012. Modulation of innate immunity by Toxoplasma gondii
2010.03.009. virulence effectors. Nat. Rev. Microbiol. 10, 766–778. https://doi.org/10.1038/
Hakimi, M.-A., Olias, P., Sibley, L.D., 2017. Toxoplasma effectors targeting host signaling nrmicro2858.
and transcription. Clin. Microbiol. Rev. 30, 615–645. https://doi.org/10.1128/CMR. Hunter, C.A., Suzuki, Y., Subauste, C.S., Remington, J.S., 1996. Cells and cytokines in
00005-17. resistance to Toxoplasma gondii. Curr. Top. Microbiol. Immunol. 219, 113–125.
Halonen, S.K., Chiu, F., Weiss, L.M., 1998. Effect of cytokines on growth of Toxoplasma Hutchison, W.M., Aitken, P.P., Wells, B.W.P., 1980. Chronic toxoplasma infections and
gondii in murine astrocytes. Infect. Immun. 66, 4989–4993. familiarity-novelty discrimination in the mouse. Ann. Trop. Med. Parasitol. 74,
Halonen, S.K., Lyman, W.D., Chiu, F.C., 1996. Growth and development of Toxoplasma 145–150. https://doi.org/10.1080/00034983.1980.11687324.
gondii in human neurons and astrocytes. J. Neuropathol. Exp. Neurol. 55, Ihara, F., Nishimura, M., Muroi, Y., Furuoka, H., Yokoyama, N., Nishikawa, Y., 2016a.
1150–1156. Changes in neurotransmitter levels and expression of immediate early genes in brain
Halonen, S.K., Taylor, G.A., Weiss, L.M., 2001. Gamma interferon-induced inhibition of of mice infected with Neospora caninum. Sci. Rep. 6, 23052. https://doi.org/10.
Toxoplasma gondii in astrocytes is mediated by IGTP. Infect. Immun. 69, 5573–5576. 1038/srep23052.
Halonen, S.K., Weiss, L.M., 2000. Investigation into the mechanism of gamma interferon- Ihara, F., Nishimura, M., Muroi, Y., Mahmoud, M.E., Yokoyama, N., Nagamune, K.,
mediated inhibition of Toxoplasma gondii in murine astrocytes. Infect. Immun. 68, Nishikawa, Y., 2016b. Toxoplasma gondii infection in mice impairs long-term fear
3426–3430. memory consolidation through dysfunction of the cortex and amygdala. Infect.
Hammoudi, P.M., Jacot, D., Mueller, C., Di Cristina, M., Dogga, S.K., Marq, J.B., Romano, Immun. 84, 2861–2870. https://doi.org/10.1128/IAI.00217-16.
J., Tosetti, N., Dubrot, J., Emre, Y., Lunghi, M., Coppens, I., Yamamoto, M., Sojka, D., Ingram, W.M., Goodrich, L.M., Robey, E.A., Eisen, M.B., 2013. Mice infected with low-
Pino, P., Soldati-Favre, D., 2015. Fundamental roles of the golgi-associated tox- virulence strains of toxoplasma gondii lose their innate aversion to cat urine, even
oplasma aspartyl protease, ASP5, at the host-parasite interface. PLoS Pathog. 11, after extensive parasite clearance. PLoS One 8, 1–6. https://doi.org/10.1371/journal.
1–32. https://doi.org/10.1371/journal.ppat.1005211. pone.0075246.
Hannan, A.J., 2010. Tandem repeat polymorphisms: modulators of disease susceptibility Irie, M., Hata, Y., Takeuchi, M., Ichtchenko, K., Toyoda, A., Hirao, K., Takai, Y., Rosahl,
and candidates for “missing heritability”. Trends Genet. 26, 59–65. https://doi.org/ T.W., Südhof, T.C., 1997. Binding of neuroligins to PSD-95. Science 277, 1511–1515.
10.1016/j.tig.2009.11.008. https://doi.org/10.1126/science.277.5331.1511.
Hari Dass, S.A., Vyas, A., 2014. Toxoplasma gondii infection reduces predator aversion in Javitt, D.C., 2015. Current and emergent treatments for symptoms and neurocognitive
rats through epigenetic modulation in the host medial amygdala. Mol. Ecol. 23, impairment in schizophrenia. Curr. Treat. Options Psychiatry 1, 107–120. https://
6114–6122. https://doi.org/10.1111/mec.12888. doi.org/10.1007/s40501-014-0010-9.
Harker, K.S., Jivan, E., McWhorter, F.Y., Liu, W.F., Lodoen, M.B., 2014. Shear forces Javitt, D.C., 2010. Glutamatergic theories of schizophrenia. Isr. J. Psychiatry Relat. Sci.
enhance toxoplasma gondii tachyzoite motility on vascular endothelium. MBio 47, 4–16.
5https://doi.org/10.1128/mBio.01111-13. e01111-13-e01111-13. Johnson, S.A., Fournier, N.M., Kalynchuk, L.E., 2006. Effect of different doses of corti-
Haroon, E., Raison, C.L., Miller, A.H., 2012. Psychoneuroimmunology meets neu- costerone on depression-like behavior and HPA axis responses to a novel stressor.
ropsychopharmacology: translational implications of the impact of inflammation on Behav. Brain Res. 168, 280–288. https://doi.org/10.1016/j.bbr.2005.11.019.
behavior. Neuropsychopharmacology 37, 137–162. https://doi.org/10.1038/npp. Jones-Brando, L., Torrey, E.F., Yolken, R., 2003. Drugs used in the treatment of schizo-
2011.205. phrenia and bipolar disorder inhibit the replication of Toxoplasma gondii. Schizophr.
Havlícek, J., Gasová, Z.G., Smith, A.P., Zvára, K., Flegr, J., 2001. Decrease of psycho- Res. 62, 237–244.
motor performance in subjects with latent “asymptomatic” toxoplasmosis. Jones, J.L., Hanson, D.L., Chu, S.Y., Ciesielski, C.A., Kaplan, J.E., Ward, J.W., Navin, T.R.,
Parasitology 122, 515–520. 1996. Toxoplasmic encephalitis in HIV-infected persons: risk factors and trends. The
Hay, J., Aitken, P.P., Graham, D.I., 1984. Toxoplasma infection and response to novelty in Adult/Adolescent Spectrum of Disease Group. AIDS 10, 1393–1399.
mice. Z. 70, 575–588. Jones, J.L., Lopez, A., Wilson, M., Schulkin, J., Gibbs, R., 2001. Congenital toxoplasmosis:
Heales, S.J., Bolaños, J.P., Stewart, V.C., Brookes, P.S., Land, J.M., Clark, J.B., 1999. a review. Obstet. Gynecol. Surv. 56, 296–305.
Nitric oxide, mitochondria and neurological disease. Biochim. Biophys. Acta 1410, Jones, T.C., Bienz, K.A., Erb, P., 1986. In vitro cultivation of Toxoplasma gondii cysts in
215–228. astrocytes in the presence of gamma interferon. Infect. Immun. 51, 147–156.
Henriquez, S.A., Brett, R., Alexander, J., Pratt, J., Roberts, C.W., 2009. Neuropsychiatric Jung, B.-K., Pyo, K.-H., Shin, K.Y., Hwang, Y.S., Lim, H., Lee, S.J., Moon, J.-H., Lee, S.H.,
disease and Toxoplasma gondii infection. Neuroimmunomodulation 16, 122–133. Suh, Y.-H., Chai, J.-Y., Shin, E.-H., 2012. Toxoplasma gondii infection in the brain
https://doi.org/10.1159/000180267. inhibits neuronal degeneration and learning and memory impairments in a murine
Herman, J.P., Figueiredo, H., Mueller, N.K., Ulrich-Lai, Y., Ostrander, M.M., Choi, D.C., model of alzheimer’s disease. PLoS One 7https://doi.org/10.1371/journal.pone.
Cullinan, W.E., 2003. Central mechanisms of stress integration: hierarchical circuitry 0033312. e33312.
controlling hypothalamo-pituitary-adrenocortical responsiveness. Front. Kanaani, J., Kolibachuk, J., Martinez, H., Baekkeskov, S., 2010. Two distinct mechanisms
Neuroendocrinol. 24, 151–180. target GAD67 to vesicular pathways and presynaptic clusters. J. Cell Biol. 190,
Hermes, G., Ajioka, J.W., Kelly, K.A., Mui, E., Roberts, F., Kasza, K., Mayr, T., Kirisits, 911–925. https://doi.org/10.1083/jcb.200912101.
M.J., Wollman, R., Ferguson, D.J., Roberts, C.W., Hwang, J.-H., Trendler, T., Kennan, Kanaani, J., Lissin, D., Kash, S.F., Baekkeskov, S., 1999. The hydrophilic isoform of

87
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

glutamate decarboxylase, GAD67, is targeted to membranes and nerve terminals Landrith, T.A., Harris, T.H., Wilson, E.H., 2015. Characteristics and critical function of
independent of dimerization with the hydrophobic membrane-anchored isoform, CD8+ T cells in the Toxoplasma-infected brain. Semin. Immunopathol. 37, 261–270.
GAD65. J. Biol. Chem. 274, 37200–37209. https://doi.org/10.1007/s00281-015-0487-3.
Kanatani, S., Fuks, J.M., Olafsson, E.B., Westermark, L., Chambers, B., Varas-Godoy, M., Lester, D., 2012. Toxoplasma gondii and homicide. Psychol. Rep. 111, 196–197. https://
Uhlén, P., Barragan, A., 2017. Voltage-dependent calcium channel signaling mediates doi.org/10.2466/12.15.16.PR0.111.4.196-197.
GABAA receptor-induced migratory activation of dendritic cells infected by Lester, D., 2010. Brain parasites and suicide. Psychol. Rep. 107, 424. https://doi.org/10.
Toxoplasma gondii. PLoS Pathog. 13https://doi.org/10.1371/journal.ppat.1006739. 2466/12.13.PR0.107.5.424.
e1006739. Levinson, A.J., Fitzgerald, P.B., Favalli, G., Blumberger, D.M., Daigle, M., Daskalakis, Z.J.,
Kaneko, Y., Takashima, Y., Xuaun, X., Igarashi, I., Nagasawa, H., Mikami, T., Otsuka, H., 2010. Evidence of cortical inhibitory deficits in major depressive disorder. Biol.
2004. Natural IgM antibodies in sera from various animals but not the cat kill Psychiatry 67, 458–464. https://doi.org/10.1016/j.biopsych.2009.09.025.
Toxoplasma gondii by activating the classical complement pathway. Parasitology Li, M., Chen, D., Shiloh, A., Luo, J., Nikolaev, A.Y., Qin, J., Gu, W., 2002.
128, 123–129. Deubiquitination of p53 by HAUSP is an important pathway for p53 stabilization.
Kannan, G., Crawford, J.A., Yang, C.X., Gressitt, K.L., Ihenatu, C., Krasnova, I.N., Cadet, Nature 416, 648–653. https://doi.org/10.1038/nature737.
J.L., Yolken, R.H., Severance, E.G., Pletnikov, M.V., 2016. Anti-NMDA receptor au- Li, P., Snyder, G.L., Vanover, K.E., 2016. Dopamine targeting drugs for the treatment of
toantibodies and associated neurobehavioral pathology in mice are dependent on age schizophrenia: past, present and future. Curr. Top. Med. Chem. 16, 3385–3403.
of first exposure to Toxoplasma gondii. Neurobiol. Dis. 91, 307–314. https://doi.org/ Li, Y., Kannan, G., Pletnikov, M., Yolken, R., Xiao, J., 2015. Chronic infection of
10.1016/j.nbd.2016.03.005. Toxoplasma gondii downregulates miR-132 expression in multiple brain regions in a
Kannan, G., Gressitt, K.L., Yang, S., Stallings, C.R., Katsafanas, E., Schweinfurth, L.A., sex-dependent manner. Parasitology 142, 623–632. https://doi.org/10.1017/
Savage, C.L.G., Adamos, M.B., Sweeney, K.M., Origoni, A.E., Khushalani, S., Bahn, S., S003118201400167X.
Leweke, F.M., Dickerson, F.B., Yolken, R.H., Pletnikov, M.V., Severance, E.G., 2017. Lim, A., Kumar, V., Hari Dass, S.A., Vyas, A., 2013. Toxoplasma gondii infection enhances
Pathogen-mediated NMDA receptor autoimmunity and cellular barrier dysfunction in testicular steroidogenesis in rats. Mol. Ecol. 22, 102–110. https://doi.org/10.1111/
schizophrenia. Transl. Psychiatry 7, e1186. https://doi.org/10.1038/tp.2017.162. mec.12042.
Kannan, G., Moldovan, K., Xiao, J.C., Yolken, R.H., Brando, L.J., Pletnikov, M.V., 2010. Lim, L.P., Lau, N.C., Garrett-Engele, P., Grimson, A., Schelter, J.M., Castle, J., Bartel, D.P.,
Toxoplasma gondii strain dependent effects on mouse behaviour. Folia Parasitol Linsley, P.S., Johnson, J.M., 2005. Microarray analysis shows that some microRNAs
(Praha) 5683, 151–155. downregulate large numbers of target mRNAs. Nature 433, 769–773. https://doi.org/
Kannan, G., Pletnikov, M.V., 2012. Toxoplasma gondii and cognitive deficits in schizo- 10.1038/nature03315.
phrenia: an animal model perspective. Schizophr. Bull. 38, 1155–1161. https://doi. Ling, V.J., Lester, D., Mortensen, P.B., Langenberg, P.W., Postolache, T.T., 2011.
org/10.1093/schbul/sbs079. Toxoplasma gondii Seropositivity and suicide rates in women. J. Nerv. Ment. Dis.
Kar, N., Misra, B., 2004. Toxoplasma seropositivity and depression: a case report. BMC 199, 440–444. https://doi.org/10.1097/NMD.0b013e318221416e.
Psychiatry 4, 1. https://doi.org/10.1186/1471-244X-4-1. Litteljohn, D., Nelson, E., Hayley, S., 2014. IFN-γ differentially modulates memory-related
Karolewicz, B., Maciag, D., O’Dwyer, G., Stockmeier, C.A., Feyissa, A.M., Rajkowska, G., processes under basal and chronic stressor conditions. Front. Cell. Neurosci. 8, 391.
2010. Reduced level of glutamic acid decarboxylase-67 kDa in the prefrontal cortex https://doi.org/10.3389/fncel.2014.00391.
in major depression. Int. J. Neuropsychopharmacol. 13, 411. https://doi.org/10. López, J.F., Akil, H., Watson, S.J., 1999. Neural circuits mediating stress. Biol. Psychiatry
1017/S1461145709990587. 46, 1461–1471.
Khan, I.A., Schwartzman, J.D., Matsuura, T., Kasper, L.H., 1997. A dichotomous role for Louveau, A., Harris, T.H., Kipnis, J., 2015. Revisiting the mechanisms of CNS immune
nitric oxide during acute Toxoplasma gondii infection in mice. Med. Sci. 94, privilege. Trends Immunol. 36, 569–577. https://doi.org/10.1016/j.it.2015.08.006.
13955–13960. Lucchese, G., 2017. From toxoplasmosis to schizophrenia via NMDA dysfunction: peptide
Kim, L., Denkers, E.Y., 2006. Toxoplasma gondii triggers Gi-dependent PI 3-kinase sig- overlap between Toxoplasma Gondii and n-methyl-d-aspartate receptors as a poten-
naling required for inhibition of host cell apoptosis. J. Cell. Sci. 119, 2119–2126. tial mechanistic link. Front. Psychiatry 8, 1–8. https://doi.org/10.3389/fpsyt.2017.
https://doi.org/10.1242/jcs.02934. 00037.
Kim, S.-K., Fouts, A.E., Boothroyd, J.C., 2007. Toxoplasma gondii dysregulates IFN- Lüder, C.G.K., Giraldo-Velásquez, M., Sendtner, M., Gross, U., 1999. Toxoplasma gondii
gamma-inducible gene expression in human fibroblasts: insights from a genome-wide in primary rat CNS cells: differential contribution of neurons, astrocytes, and mi-
transcriptional profiling. J. Immunol. 178, 5154–5165 https://doi.org/178/8/5154 croglial cells for the intracerebral development and stage differentiation. Exp.
[pii]. Parasitol. 93, 23–32. https://doi.org/10.1006/expr.1999.4421.
Kipnis, J., 2016. Multifaceted interactions between adaptive immunity and the central Lugo-Huitrón, R., Ugalde Muñiz, P., Pineda, B., Pedraza-Chaverrí, J., Ríos, C., Pérez-de la
nervous system. Science 353, 766–771. https://doi.org/10.1126/science.aag2638. Cruz, V., 2013. Quinolinic acid: an endogenous neurotoxin with multiple targets.
Klein, R.S., Garber, C., Howard, N., 2017. Infectious immunity in the central nervous Oxid. Med. Cell. Longev. 2013, 104024. https://doi.org/10.1155/2013/104024.
system and brain function. Nat. Immunol. 18, 132–141. https://doi.org/10.1038/ni. Machado, V.S., Bottari, N.B., Baldissera, M.D., Rech, V.C., Ianiski, F.R., Signor, C., Rubin,
3656. M.A., Waczuk, E.P., Schwertz, C.I., Mendes, R.E., Camillo, G., Vogel, F.F., de la Rue,
Knuesel, I., Chicha, L., Britschgi, M., Schobel, S.A., Bodmer, M., Hellings, J.A., Toovey, S., M.L., Morsch, V.M., Schetinger, M.R.C., Fr??hauf, P.K.S., Da Silva, A.S., 2016.
Prinssen, E.P., 2014. Maternal immune activation and abnormal brain development Diphenyl diselenide supplementation in infected mice by Toxoplasma gondii: pro-
across CNS disorders. Nat. Rev. Neurol. 10, 643–660. https://doi.org/10.1038/ tective effect on behavior, neuromodulation and oxidative stress caused by disease.
nrneurol.2014.187. Exp. Parasitol. 169, 51–58. https://doi.org/10.1016/j.exppara.2016.07.006.
Kocazeybek, B., Oner, Y.A., Turksoy, R., Babur, C., Cakan, H., Sahip, N., Unal, A., MacNeil, C., Fielding, N.J., Hume, K.D., Dick, J.T.A., Elwood, R.W., Hatcher, M.J., Dunn,
Ozaslan, A., Kılıc, S., Saribas, S., Aslan, M., Taylan, A., Koc, S., Dirican, A., Uner, H.B., A.M., 2003. Parasite altered micro-distribution of Gammarus pulex (Crustacea: am-
Oz, V., Ertekin, C., Kucukbasmaci, O., Torun, M.M., 2009. Higher prevalence of phipoda). Int. J. Parasitol. 33, 57–64.
toxoplasmosis in victims of traffic accidents suggest increased risk of traffic accident MacRae, J.I., Sheiner, L., Nahid, A., Tonkin, C., Striepen, B., McConville, M.J., 2012.
in Toxoplasma-infected inhabitants of Istanbul and its suburbs. Forensic Sci. Int. 187, Mitochondrial metabolism of glucose and glutamine is required for intracellular
103–108. https://doi.org/10.1016/j.forsciint.2009.03.007. growth of Toxoplasma gondii. Cell Host Microbe 12, 682–692. https://doi.org/10.
Koella, J.C., 2005. Malaria as a manipulator. Behav. Processes 68, 271–273. https://doi. 1016/j.chom.2012.09.013.
org/10.1016/j.beproc.2004.10.004. Maher, B., 2008. Personal genomes: the case of the missing heritability. Nature 456,
Kolev, M.V., Ruseva, M.M., Harris, C.L., Morgan, B.P., Donev, R.M., 2009. Implication of 18–21. https://doi.org/10.1038/456018a.
complement system and its regulators in Alzheimer’s disease. Curr. Neuropharmacol. Mahmoud, M.E., Fereig, R., Nishikawa, Y., 2017. Involvement of host defense mechan-
7, 1–8. https://doi.org/10.2174/157015909787602805. isms against toxoplasma gondii infection in Anhedonic and despair-like behaviors in
Konradt, C., Ueno, N., Christian, D.A., Delong, J.H., Pritchard, G.H., Herz, J., Bzik, D.J., mice. Infect. Immun. 85, e00007–17. https://doi.org/10.1128/IAI.00007-17.
Koshy, A.A., McGavern, D.B., Lodoen, M.B., Hunter, C.A., 2016. Endothelial cells are Mahmoud, M.E., Ihara, F., Fereig, R.M., Nishimura, M., Nishikawa, Y., 2016. Induction of
a replicative niche for entry of Toxoplasma gondii to the central nervous system. Nat. depression-related behaviors by reactivation of chronic Toxoplasma gondii infection
Microbiol. 1, 16001. https://doi.org/10.1038/nmicrobiol.2016.1. in mice. Behav. Brain Res. 298, 125–133. https://doi.org/10.1016/j.bbr.2015.11.
Koshy, A.A., Cabral, C.M., 2014. 3-D imaging and analysis of neurons infected in vivo with 005.
Toxoplasma gondii. J. Vis. Exp. 1–7. https://doi.org/10.3791/52237. Mahmoudvand, H., Sheibani, V., Shojaee, S., Mirbadie, S.R., Keshavarz, H., Esmaeelpour,
Koshy, A.A., Dietrich, H.K., Christian, D.A., Melehani, J.H., Shastri, A.J., Hunter, C.A., K., Keyhani, A.R., Ziaali, N., 2016a. Toxoplasma gondii infection potentiates cognitive
Boothroyd, J.C., 2012. Toxoplasma co-opts host cells it does not invade. PLoS Pathog. impairments of alzheimer’s disease in the BALB/c mice. J. Parasitol. 102, 629–635.
8, 18. https://doi.org/10.1371/journal.ppat.1002825. https://doi.org/10.1645/16-28.
Koshy, A.A., Fouts, A.E., Lodoen, M.B., Alkan, O., Blau, H.M., Boothroyd, J.C., 2010. Mahmoudvand, H., Ziaali, N., Ghazvini, H., Shojaee, S., Keshavarz, H., Esmaeilpour, K.,
Toxoplasma secreting Cre recombinase for analysis of host-parasite interactions. Nat. Sheibani, V., 2016b. Toxoplasma gondii infection promotes neuroinflammation
Methods 7, 307–309. https://doi.org/10.1038/nmeth.1438. through cytokine networks and induced hyperalgesia in BALB/c mice. Inflammation
Lachenmaier, S.M., Deli, M.A., Meissner, M., Liesenfeld, O., 2011. Intracellular transport 39, 405–412. https://doi.org/10.1007/s10753-015-0262-6.
of Toxoplasma gondii through the blood–brain barrier. J. Neuroimmunol. 232, Mandolesi, G., Bullitta, S., Fresegna, D., Gentile, A., De Vito, F., Dolcetti, E., Rizzo, F.R.,
119–130. https://doi.org/10.1016/j.jneuroim.2010.10.029. Strimpakos, G., Centonze, D., Musella, A., 2017. Interferon-γ causes mood abnorm-
Lacroix, R., Mukabana, W.R., Gouagna, L.C., Koella, J.C., 2005. Malaria infection in- alities by altering cannabinoid CB1 receptor function in the mouse striatum.
creases attractiveness of humans to mosquitoes. PLoS Biol. 3, e298. https://doi.org/ Neurobiol. Dis. 108, 45–53. https://doi.org/10.1016/j.nbd.2017.07.019.
10.1371/journal.pbio.0030298. Manolio, T.A., Collins, F.S., Cox, N.J., Goldstein, D.B., Hindorff, L.A., Hunter, D.J.,
Lambert, H., Hitziger, N., Dellacasa, I., Svensson, M., Barragan, A., 2006. Induction of McCarthy, M.I., Ramos, E.M., Cardon, L.R., Chakravarti, A., Cho, J.H., Guttmacher,
dendritic cell migration upon Toxoplasma gondii infection potentiates parasite dis- A.E., Kong, A., Kruglyak, L., Mardis, E., Rotimi, C.N., Slatkin, M., Valle, D.,
semination. Cell. Microbiol. 8, 1611–1623. https://doi.org/10.1111/j.1462-5822. Whittemore, A.S., Boehnke, M., Clark, A.G., Eichler, E.E., Gibson, G., Haines, J.L.,
2006.00735.x. Mackay, T.F.C., McCarroll, S.A., Visscher, P.M., 2009. Finding the missing heritability

88
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

of complex diseases. Nature 461, 747–753. https://doi.org/10.1038/nature08494. development of schizophrenia. Schizophr. Bull. 33, 741–744. https://doi.org/10.
Marino, N.D., Panas, M.W., Franco, M., Theisen, T.C., Naor, A., Rastogi, S., Buchholz, 1093/schbul/sbm009.
K.R., Lorenzi, H.A., Boothroyd, J.C., 2018. Identification of a novel protein complex Mortensen, P.B., Pedersen, C.B., Mcgrath, J.J., Hougaard, D.M., Nørgaard-Petersen, B.,
essential for effector translocation across the parasitophorous vacuole membrane of Mors, O., Børglum, A.D., Yolken, R.H., 2011. Neonatal antibodies to infectious agents
Toxoplasma gondii. PLoS Pathog. 14https://doi.org/10.1371/journal.ppat.1006828. and risk of bipolar disorder: a population-based case-control study. Bipolar Disord.
e1006828. 13, 624–629. https://doi.org/10.1111/j.1399-5618.2011.00962.x.
Martorana, A., Koch, G., 2014. Is dopamine involved in Alzheimer’s disease? Front. Aging Murakami, Y., Hoshi, M., Hara, A., Takemura, M., Arioka, Y., Yamamoto, Y., Matsunami,
Neurosci. 6, 252. https://doi.org/10.3389/fnagi.2014.00252. H., Funato, T., Seishima, M., Saito, K., 2012. Inhibition of increased indoleamine 2,3-
Matsumoto, M., 2015. Dopamine signals and physiological origin of cognitive dysfunction dioxygenase activity attenuates Toxoplasma gondii replication in the lung during
in Parkinson’s disease. Mov. Disord. 30, 472–483. https://doi.org/10.1002/mds. acute infection. Cytokine 59, 245–251. https://doi.org/10.1016/j.cyto.2012.04.022.
26177. Nadipuram, S.M., Kim, E.W., Vashisht, A.A., Lin, A.H., Bell, H.N., Coppens, I.,
Mazarei, G., Budac, D.P., Lu, G., Lee, H., M??ller, T., Leavitt, B.R., 2013. The absence of Wohlschlegel, J.A., Bradley, P.J., 2016. In vivo biotinylation of the toxoplasma
indoleamine 2,3-dioxygenase expression protects against NMDA receptor-mediated parasitophorous vacuole reveals novel dense granule proteins important for parasite
excitotoxicity in mouse brain. Exp. Neurol. 249, 144–148. https://doi.org/10.1016/j. growth and pathogenesis. MBio 7https://doi.org/10.1128/mBio.00808-16.
expneurol.2013.08.007. e00808-16.
McConkey, G.A., Peers, C., Prandovszky, E., 2015. Reproducing increased dopamine with Naor, A., Panas, M.W., Marino, N., Coffey, M.J., Tonkin, C.J., Boothroyd, J.C., 2018.
infection to evaluate the role of parasite-encoded tyrosine hydroxylase activity. MYR1-dependent effectors are the major drivers of a host cell’s early response to
Infect. Immun. 83, 3334–3335. https://doi.org/10.1128/IAI.00605-15. toxoplasma, including counteracting MYR1-independent effects. MBio 9. https://doi.
McFarland, R., Wang, Z.T., Jouroukhin, Y., Li, Y., Mychko, O., Coppens, I., Xiao, J., Jones- org/10.1128/mBio.02401-17.
Brando, L., Yolken, R.H., Sibley, L.D., Pletnikov, M.V., 2018. AAH2 gene is not re- Newpher, T.M., Ehlers, M.D., 2008. Glutamate receptor dynamics in dendritic micro-
quired for dopamine-dependent neurochemical and behavioral abnormalities pro- domains. Neuron. https://doi.org/10.1016/j.neuron.2008.04.030.
duced by Toxoplasma infection in mouse. Behav. Brain Res. https://doi.org/10. Ngô, H.M., Zhou, Y., Lorenzi, H., Wang, K., Kim, T.-K., Zhou, Y., Bissati, K.El, Mui, E.,
1016/j.bbr.2018.03.023. Fraczek, L., Rajagopala, S.V., Roberts, C.W., Henriquez, F.L., Montpetit, A.,
Melzer, T.C., Cranston, H.J., Weiss, L.M., Halonen, S.K., 2010. Host cell preference of Blackwell, J.M., Jamieson, S.E., Wheeler, K., Begeman, I.J., Naranjo-Galvis, C.,
toxoplasma gondii cysts in murine brain: a confocal study. J. Neuroparasitology 1. Alliey-Rodriguez, N., Davis, R.G., Soroceanu, L., Cobbs, C., Steindler, D.A., Boyer, K.,
https://doi.org/10.4303/jnp/N100505. Noble, A.G., Swisher, C.N., Heydemann, P.T., Rabiah, P., Withers, S., Soteropoulos,
Mercier, C., Cesbron-Delauw, M.-F., 2015. Toxoplasma secretory granules: one population P., Hood, L., McLeod, R., 2017. Toxoplasma modulates signature pathways of human
or more? Trends Parasitol. 31, 60–71. https://doi.org/10.1016/j.pt.2014.12.002. epilepsy, neurodegeneration & Cancer. Sci. Rep. 7, 11496. https://doi.org/10.1038/
Michailidou, I., Willems, J.G.P., Kooi, E.-J., van Eden, C., Gold, S.M., Geurts, J.J.G., Baas, s41598-017-10675-6.
F., Huitinga, I., Ramaglia, V., 2015. Complement C1q-C3-associated synaptic changes Ngoungou, E.B., Bhalla, D., Nzoghe, A., Dardé, M.-L., Preux, P.-M., 2015. Toxoplasmosis
in multiple sclerosis hippocampus. Ann. Neurol. 77, 1007–1026. https://doi.org/10. and epilepsy — systematic review and Meta analysis. PLoS Negl. Trop. Dis. 9https://
1002/ana.24398. doi.org/10.1371/journal.pntd.0003525. e0003525.
Miladinovic, T., Nashed, M.G., Singh, G., 2015. Overview of glutamatergic dysregulation Ni Nyoman, A.D., Lüder, C.G.K., 2013. Apoptosis-like cell death pathways in the uni-
in central pathologies. Biomolecules 5, 3112–3141. https://doi.org/10.3390/ cellular parasite Toxoplasma gondii following treatment with apoptosis inducers and
biom5043112. chemotherapeutic agents: a proof-of-concept study. Apoptosis 18, 664–680. https://
Miller, B.H., Zeier, Z., Xi, L., Lanz, T.A., Deng, S., Strathmann, J., Willoughby, D., Kenny, doi.org/10.1007/s10495-013-0832-8.
P.J., Elsworth, J.D., Lawrence, M.S., Roth, R.H., Edbauer, D., Kleiman, R.J., Nilsson, L.K., Linderholm, K.R., Engberg, G., Paulson, L., Blennow, K., Lindström, L.H.,
Wahlestedt, C., 2012. MicroRNA-132 dysregulation in schizophrenia has implications Nordin, C., Karanti, A., Persson, P., Erhardt, S., 2005. Elevated levels of kynurenic
for both neurodevelopment and adult brain function. Proc. Natl. Acad. Sci. 109, acid in the cerebrospinal fluid of male patients with schizophrenia. Schizophr. Res.
3125–3130. https://doi.org/10.1073/pnas.1113793109. 80, 315–322. https://doi.org/10.1016/j.schres.2005.07.013.
Mink, J.W., 2006. Neurobiology of basal ganglia and Tourette syndrome: basal ganglia Notarangelo, F.M., Wilson, E.H., Horning, K.J., Thomas, M.A.R., Harris, T.H., Fang, Q.,
circuits and thalamocortical outputs. Adv. Neurol. 99, 89–98. Hunter, C.A., Schwarcz, R., 2014. Evaluation of kynurenine pathway metabolism in
Mitra, R., Ferguson, D., Sapolsky, R.M., 2009a. SK2 potassium channel overexpression in Toxoplasma gondii-infected mice: implications for schizophrenia. Schizophr. Res.
basolateral amygdala reduces anxiety, stress-induced corticosterone secretion and 152, 261–267. https://doi.org/10.1016/j.schres.2013.11.011.
dendritic arborization. Mol. Psychiatry 14 (847–55), 827. https://doi.org/10.1038/ Nourollahpour Shiadeh, M., Rostami, A., Pearce, B.D., Gholipourmalekabadi, M.,
mp.2009.9. Newport, D.J., Danesh, M., Mehravar, S., Seyyedtabaei, S.J., 2016. The correlation
Mitra, R., Ferguson, D., Sapolsky, R.M., 2009b. Mineralocorticoid receptor overexpression between Toxoplasma gondii infection and prenatal depression in pregnant women.
in basolateral amygdala reduces corticosterone secretion and anxiety. Biol. Eur. J. Clin. Microbiol. Infect. Dis. 35, 1829–1835. https://doi.org/10.1007/s10096-
Psychiatry 66, 686–690. https://doi.org/10.1016/j.biopsych.2009.04.016. 016-2734-5.
Mitra, R., Sapolsky, R.M., 2008. Acute corticosterone treatment is sufficient to induce Olias, P., Etheridge, R.D., Zhang, Y., Holtzman, M.J., Sibley, L.D., 2016. Toxoplasma
anxiety and amygdaloid dendritic hypertrophy. Proc. Natl. Acad. Sci. U. S. A. 105, effector recruits the Mi-2/NuRD complex to repress STAT1 transcription and block
5573–5578. https://doi.org/10.1073/pnas.0705615105. IFN-γ-dependent gene expression. Cell Host Microbe 20, 72–82. https://doi.org/10.
Mitra, R., Sapolsky, R.M., Vyas, A., 2013. Toxoplasma gondii infection induces dendritic 1016/j.chom.2016.06.006.
retraction in basolateral amygdala accompanied by reduced corticosterone secretion. Onore, C., Careaga, M., Ashwood, P., 2012. The role of immune dysfunction in the pa-
Dis. Model. Mech. 6, 516–520. https://doi.org/10.1242/dmm.009928. thophysiology of autism. Brain Behav. Immun. 26, 383–392. https://doi.org/10.
Mizuno, T., Zhang, G., Takeuchi, H., Kawanokuchi, J., Wang, J., Sonobe, Y., Jin, S., 1016/j.bbi.2011.08.007.
Takada, N., Komatsu, Y., Suzumura, A., 2008. Interferon-gamma directly induces Orlofsky, A., Weiss, L.M., Kawachi, N., Prystowsky, M.B., 2002. Deficiency in the anti-
neurotoxicity through a neuron specific, calcium-permeable complex of IFN-gamma apoptotic protein A1-a results in a diminished acute inflammatory response. J.
receptor and AMPA GluR1 receptor. FASEB J. 22, 1797–1806. https://doi.org/10. Immunol. 168, 1840–1846.
1096/fj.07-099499. Ortiz, P.A., Garvin, J.L., 2003. Cardiovascular and renal control in NOS-deficient mouse
Moghaddam, B., Javitt, D., 2012. From revolution to evolution: the glutamate hypothesis models. Am. J. Physiol. Regul. Integr. Comp. Physiol. 284, R628–38. https://doi.org/
of schizophrenia and its implication for treatment. Neuropsychopharmacology 37, 10.1152/ajpregu.00401.2002.
4–15. https://doi.org/10.1038/npp.2011.181. Oxenkrug, G., 2013. Serotonin – kynurenine hypothesis of depression: historical overview
Möhle, L., Israel, N., Paarmann, K., Krohn, M., Pietkiewicz, S., Müller, A., Lavrik, I.N., and recent developments. Curr. Drug Targets 14, 514–521. https://doi.org/10.2174/
Buguliskis, J.S., Schott, B.H., Schlüter, D., Gundelfinger, E.D., Montag, D., Seifert, U., 1389450111314050002.
Pahnke, J., Dunay, I.R., 2016. Chronic Toxoplasma gondii infection enhances β- Palmer, B.S., 2007. Meta-analysis of three case controlled studies and an ecological study
amyloid phagocytosis and clearance by recruited monocytes. Acta Neuropathol. into the link between cryptogenic epilepsy and chronic toxoplasmosis infection.
Commun. 4, 25. https://doi.org/10.1186/s40478-016-0293-8. Seizure 16, 657–663. https://doi.org/10.1016/j.seizure.2007.05.010.
Monroe, J.M., Buckley, P.F., Miller, B.J., 2015. Meta-analysis of anti-Toxoplasma gondii Papouin, T., Dunphy, J.M., Tolman, M., Dineley, K.T., Haydon, P.G., 2017. Septal cho-
IgM antibodies in acute psychosis. Schizophr. Bull. 41, 989–998. https://doi.org/10. linergic neuromodulation tunes the astrocyte-dependent gating of hippocampal
1093/schbul/sbu159. NMDA receptors to wakefulness. Neuron 94, 840–854. https://doi.org/10.1016/j.
Montacute, R., Foley, K., Forman, R., Else, K.J., Cruickshank, S.M., Allan, S.M., 2017. neuron.2017.04.021. e7.
Enhanced susceptibility of triple transgenic Alzheimer’s disease (3xTg-AD) mice to Pariante, C.M., Lightman, S.L., 2008. The HPA axis in major depression: classical theories
acute infection. J. Neuroinflammation 14, 50. https://doi.org/10.1186/s12974-017- and new developments. Trends Neurosci. 31, 464–468. https://doi.org/10.1016/j.
0826-5. tins.2008.06.006.
Montgomery, J.M., Madison, D.V., 2004. Discrete synaptic states define a major me- Parlog, A., Harsan, L.-A., Zagrebelsky, M., Weller, M., von Elverfeldt, D., Mawrin, C.,
chanism of synapse plasticity. Trends Neurosci. 27, 744–750. https://doi.org/10. Korte, M., Dunay, I.R., 2014. Chronic murine toxoplasmosis is defined by subtle
1016/j.tins.2004.10.006. changes in neuronal connectivity. Dis. Model. Mech. 7, 459–469. https://doi.org/10.
Montoya, J.G., Liesenfeld, O., 2004. Toxoplasmosis. Lancet (Lond. Engl.) 363, 1242/dmm.014183.
1965–1976. https://doi.org/10.1016/S0140-6736(04)16412-X. Parlog, A., Schlüter, D., Dunay, I.R., 2015. Toxoplasma gondii-induced neuronal altera-
Mortensen, P.B., Nørgaard-Pedersen, B., Waltoft, B.L., Sørensen, T.L., Hougaard, D., tions. Parasite Immunol. 37, 159–170. https://doi.org/10.1111/pim.12157.
Torrey, E.F., Yolken, R.H., 2007a. Toxoplasma gondii as a risk factor for early-onset Parrott, J.M., Redus, L., Santana-Coelho, D., Morales, J., Gao, X., O’Connor, J.C., 2016.
schizophrenia: analysis of filter paper blood samples obtained at birth. Biol. Neurotoxic kynurenine metabolism is increased in the dorsal hippocampus and drives
Psychiatry 61, 688–693. https://doi.org/10.1016/j.biopsych.2006.05.024. distinct depressive behaviors during inflammation. Transl. Psychiatry 6, e918.
Mortensen, P.B., Nørgaard-Pedersen, B., Waltoft, B.L., Sørensen, T.L., Hougaard, D., https://doi.org/10.1038/tp.2016.200.
Yolken, R.H., 2007b. Early infections of Toxoplasma gondii and the later Paul, V., Ekambaram, P., 2011. Involvement of nitric oxide in learning & memory

89
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

processes. Indian J. Med. Res. 133, 471–478. org/10.1371/journal.pone.0051448.


Payne, T.M., Molestina, R.E., Sinai, A.P., 2003. Inhibition of caspase activation and a Rothstein, J.D., Patel, S., Regan, M.R., Haenggeli, C., Huang, Y.H., Bergles, D.E., Jin, L.,
requirement for NF-kappaB function in the Toxoplasma gondii-mediated blockade of Hoberg, M.D., Vidensky, S., Chung, D.S., Toan, S.V., Bruijn, L.I., Su, Z., Gupta, P.,
host apoptosis. J. Cell. Sci. 116, 4345–4358. https://doi.org/10.1242/jcs.00756. Fisher, P.B., 2005. |[Beta]|-Lactam antibiotics offer neuroprotection by increasing
Pearce, B.D., Kruszon-Moran, D., Jones, J.L., 2012. The relationship between toxoplasma glutamate transporter expression. Nature 433, 73. https://doi.org/10.1038/
gondii infection and mood disorders in the third national health and nutrition survey. nature03180. Publ. online 06 January 2005.
Biol. Psychiatry 72, 290–295. https://doi.org/10.1016/j.biopsych.2012.01.003. Rozenfeld, C., Martinez, R., Figueiredo, R.T., Bozza, M.T., Lima, F.R.S., Pires, A.L., Silva,
Pedersen, M.G., Mortensen, P.B., Norgaard-Pedersen, B., Postolache, T.T., 2012. P.M., Bonomo, A., Lannes-Vieira, J., De Souza, W., Moura-Neto, V., 2003. Soluble
Toxoplasma gondii infection and self-directed violence in mothers. Arch. Gen. factors released by Toxoplasma gondii-infected astrocytes down-modulate nitric
Psychiatry 69, 1123–1130. https://doi.org/10.1001/archgenpsychiatry.2012.668. oxide production by gamma interferon-activated microglia and prevent neuronal
Peixoto, L., Chen, F., Harb, O.S., Davis, P.H., Beiting, D.P., Brownback, C.S., Ouloguem, degeneration. Infect. Immun. 71, 2047–2057.
D., Roos, D.S., 2010. Integrative genomic approaches highlight a family of parasite- Rozenfeld, C., Martinez, R., Seabra, S., Sant’anna, C., Gonçalves, J.G.R., Bozza, M.,
specific kinases that regulate host responses. Cell Host Microbe 8, 208–218. https:// Moura-Neto, V., De Souza, W., 2005. Toxoplasma gondii prevents neuron degen-
doi.org/10.1016/j.chom.2010.07.004. eration by interferon-gamma-activated microglia in a mechanism involving inhibi-
Peterson, P.K., Gekker, G., Hu, S., Chao, C.C., 1995. Human astrocytes inhibit in- tion of inducible nitric oxide synthase and transforming growth factor-beta1 pro-
tracellular multiplication of Toxoplasma gondii by a nitric oxide-mediated me- duction by infected microglia. Am. J. Pathol. 167, 1021–1031.
chanism. J. Infect. Dis. 171, 516–518. Ruddick, J.P., Evans, A.K., Nutt, D.J., Lightman, S.L., Rook, G.A.W., Lowry, C.A., 2006.
Peterson, P.K., Gekker, G., Hu, S., Chao, C.C., 1993. Intracellular survival and multi- Tryptophan metabolism in the central nervous system: medical implications. Expert
plication of Toxoplasma gondii in astrocytes. J. Infect. Dis. 168, 1472–1478. Rev. Mol. Med. 8, 1–27. https://doi.org/10.1017/S1462399406000068.
Pfefferkorn, E.R., 1984. Interferon gamma blocks the growth of Toxoplasma gondii in Sa, Q., Tiwari, A., Ochiai, E., Mullins, J., Suzuki, Y., 2017. Inducible nitric oxide synthase
human fibroblasts by inducing the host cells to degrade tryptophan. Proc. Natl. Acad. in innate immune cells is important for restricting cyst formation of Toxoplasma
Sci. U. S. A. 81, 908–912. https://doi.org/10.1073/pnas.81.3.908. gondii in the brain but not required for the protective immune process to remove the
Pisalyaput, K., Tenner, A.J., 2008. Complement component C1q inhibits beta-amyloid- cysts. Microbes Infect. https://doi.org/10.1016/j.micinf.2017.12.004.
and serum amyloid P-induced neurotoxicity via caspase- and calpain-independent Sacks, D., Sher, A., 2002. Evasion of innate immunity by parasitic protozoa. Nat.
mechanisms. J. Neurochem. 104, 696–707. https://doi.org/10.1111/j.1471-4159. Immunol. 3, 1041–1047. https://doi.org/10.1038/ni1102-1041.
2007.05012.x. Saeij, J.P.J., Coller, S., Boyle, J.P., Jerome, M.E., White, M.W., Boothroyd, J.C., 2007.
Prandovszky, E., Gaskell, E., Martin, H., Dubey, J.P., Webster, J.P., McConkey, G.A., Toxoplasma co-opts host gene expression by injection of a polymorphic kinase
2011. The neurotropic parasite toxoplasma gondii increases dopamine metabolism. homologue. Nature 445, 324–327. https://doi.org/10.1038/nature05395.
PLoS One 6. https://doi.org/10.1371/journal.pone.0023866. Saha, S., Chant, D., Welham, J., McGrath, J., 2005. A systematic review of the prevalence
Příplatová, L., Šebánková, B., Flegr, J., 2014. Contrasting effect of prepulse signals on of schizophrenia. PLoS Med. 2, e141. https://doi.org/10.1371/journal.pmed.
performance of toxoplasma-infected and toxoplasma-free subjects in an acoustic re- 0020141.
action times test. PLoS One 9, e112771. https://doi.org/10.1371/journal.pone. Saito, K., Markey, S.P., Heyes, M.P., 1992. Effects of immune activation on quinolinic acid
0112771. and neuroactive kynurenines in the mouse. Neuroscience 51, 25–39.
Purcell, S.M., Wray, N.R., Stone, J.L., Visscher, P.M., O’Donovan, M.C., Sullivan, P.F., Sapolsky, R., 2003. Bugs in the brain. Sci. Am. 288, 94–97.
Sklar, P., Purcell (Leader), S.M., Stone, J.L., Sullivan, P.F., Ruderfer, D.M., McQuillin, Saraei, M., Samadzadeh, N., Khoeini, J., Shahnazi, M., Asl, N., Jahanihashemi, H., 2015.
A., Morris, D.W., O’Dushlaine, C.T., Corvin, A., Holmans, P.A., O’Donovan, M.C., In vivo anti ‐ Toxoplasma activity of aripiprazole. Iran. J. Basic Med. Sci. 18,
Sklar, P., Wray, N.R., Macgregor, S., Sklar, P., Sullivan, P.F., O’Donovan, M.C., 938–941.
Visscher, P.M., Gurling, H., Blackwood, D.H.R., Corvin, A., Craddock, N.J., Gill, M., Schaafsma, W., Basterra, L.B., Jacobs, S., Brouwer, N., Meerlo, P., Schaafsma, A.,
Hultman, C.M., Kirov, G.K., Lichtenstein, P., McQuillin, A., Muir, W.J., O’Donovan, Boddeke, E.W.G.M., Eggen, B.J.L., 2017. Maternal inflammation induces immune
M.C., Owen, M.J., Pato, C.N., Purcell, S.M., Scolnick, E.M., St Clair, D., Stone, J.L., activation of fetal microglia and leads to disrupted microglia immune responses,
Sullivan, P.F., Sklar (Leader), P., O’Donovan, M.C., Kirov, G.K., Craddock, N.J., behavior, and learning performance in adulthood. Neurobiol. Dis. 106, 291–300.
Holmans, P.A., Williams, N.M., Georgieva, L., Nikolov, I., Norton, N., Williams, H., https://doi.org/10.1016/j.nbd.2017.07.017.
Toncheva, D., Milanova, V., Owen, M.J., Hultman, C.M., Lichtenstein, P., Thelander, Schlüter, D., Deckert, M., Hof, H., Frei, K., 2001. Toxoplasma gondii infection of neurons
E.F., Sullivan, P., Morris, D.W., O’Dushlaine, C.T., Kenny, E., Quinn, E.M., Gill, M., induces neuronal cytokine and chemokine production, but gamma interferon- and
Corvin, A., McQuillin, A., Choudhury, K., Datta, S., Pimm, J., Thirumalai, S., Puri, V., tumor necrosis factor-stimulated neurons fail to inhibit the invasion and growth of T.
Krasucki, R., Lawrence, J., Quested, D., Bass, N., Gurling, H., Crombie, C., Fraser, G., Gondii. Infect. Immun. 69, 7889–7893. https://doi.org/10.1128/IAI.69.12.7889-
Leh Kuan, S., Walker, N., St Clair, D., Blackwood, D.H.R., Muir, W.J., McGhee, K.A., 7893.2001.
Pickard, B., Malloy, P., Maclean, A.W., Van Beck, M., Wray, N.R., Macgregor, S., Schmitt, U., Tanimoto, N., Seeliger, M., Schaeffel, F., Leube, R.E., 2009. Detection of
Visscher, P.M., Pato, M.T., Medeiros, H., Middleton, F., Carvalho, C., Morley, C., behavioral alterations and learning deficits in mice lacking synaptophysin.
Fanous, A., Conti, D., Knowles, J.A., Paz Ferreira, C., Macedo, A., Helena Azevedo, Neuroscience 162, 234–243. https://doi.org/10.1016/j.neuroscience.2009.04.046.
M., Pato, C.N., Stone, J.L., Ruderfer, D.M., Kirby, A.N., Ferreira, M.A.R., Daly, M.J., Schwarcz, R., Hunter, C.A., 2007. Toxoplasma gondii and schizophrenia: linkage through
Purcell, S.M., Sklar, P., Purcell, S.M., Stone, J.L., Chambert, K., Ruderfer, D.M., astrocyte-derived kynurenic acid? Schizophr. Bull. 33, 652–653. https://doi.org/10.
Kuruvilla, F., Gabriel, S.B., Ardlie, K., Moran, J.L., Daly, M.J., Scolnick, E.M., Sklar, 1093/schbul/sbm030.
P., 2009. Common polygenic variation contributes to risk of schizophrenia and bi- Seabra, S.H., de Souza, W., DaMatta, R.A., 2002. Toxoplasma gondii partially inhibits
polar disorder. Nature 460, 748–752. https://doi.org/10.1038/nature08185. nitric oxide production of activated murine macrophages. Exp. Parasitol. 100, 62–70.
Qiao, H., An, S.-C., Xu, C., Ma, X.-M., 2017. Role of proBDNF and BDNF in dendritic spine https://doi.org/10.1006/expr.2001.4675.
plasticity and depressive-like behaviors induced by an animal model of depression. Severance, E.G., Gressitt, K.L., Buka, S.L., Cannon, T.D., Yolken, R.H., 2014. Maternal
Brain Res. 1663, 29–37. https://doi.org/10.1016/j.brainres.2017.02.020. complement C1q and increased odds for psychosis in adult offspring. Schizophr. Res.
Reiter-Owona, I., Seitz, H., Gross, U., Sahm, M., Rockstroh, J.K., Seitz, H.M., 2000. Is 159, 14–19. https://doi.org/10.1016/j.schres.2014.07.053.
stage conversion the initiating event for reactivation of Toxoplasma gondii in brain Severance, E.G., Gressitt, K.L., Halling, M., Stallings, C.R., Origoni, A.E., Vaughan, C.,
tissue of AIDS patients? J. Parasitol. 86, 531–536. https://doi.org/10.1645/0022- Khushalani, S., Alaedini, A., Dupont, D., Dickerson, F.B., Yolken, R.H., 2012.
3395(2000)086[0531:ISCTIE]2.0.CO;2. Complement C1q formation of immune complexes with milk caseins and wheat
Riazi, K., Galic, M.A., Kentner, A.C., Reid, A.Y., Sharkey, K.A., Pittman, Q.J., 2015. glutens in schizophrenia. Neurobiol. Dis. 48, 447–453. https://doi.org/10.1016/j.
Microglia-dependent alteration of glutamatergic synaptic transmission and plasticity nbd.2012.07.005.
in the hippocampus during peripheral inflammation. J. Neurosci. 35, 4942–4952. Short, a K., Fennell, Ka, Perreau, V.M., Fox, A., O’Bryan, M.K., Kim, J.H., Bredy, T.W.,
https://doi.org/10.1523/JNEUROSCI.4485-14.2015. Pang, T.Y., Hannan, a J., 2016. Elevated paternal glucocorticoid exposure alters the
Robinson, A., Busula, A.O., Voets, M.A., Beshir, K.B., Caulfield, J.C., Powers, S.J., small noncoding RNA profile in sperm and modifies anxiety and depressive pheno-
Verhulst, N.O., Winskill, P., Muwanguzi, J., Birkett, M.A., Smallegange, R.C., Masiga, types in the offspring. Transl. Psychiatry 6, e837. https://doi.org/10.1038/tp.2016.
D.K., Mukabana, W.R., Sauerwein, R.W., Sutherland, C.J., Bousema, T., Pickett, J.A., 109.
Takken, W., Logan, J.G., de Boer, J.G., 2018. Plasmodium -associated changes in Silva, N.M., Rodrigues, C.V., Santoro, M.M., Reis, L.F.L., Alvarez-leite, J.I., Ricardo, T.,
human odor attract mosquitoes. Proc. Natl. Acad. Sci. 115, 201721610. https://doi. Gazzinelli, R.T., 2002. Expression of Indoleamine and kynurenine formation during
org/10.1073/pnas.1721610115. in vivo infection with toxoplasma gondii : induction by endogenous gamma inter-
Rodgers, A.B., Morgan, C.P., Leu, N.A., Bale, T.L., 2015. Transgenerational epigenetic feron and requirement of interferon regulatory factor 1 expression of indoleamine 2,
programming via sperm microRNA recapitulates effects of paternal stress. Proc. Natl. 3-Dioxygenase, tryptophan degrad. Infect. Immun. 70, 859–868. https://doi.org/10.
Acad. Sci. 112, 13699–13704. https://doi.org/10.1073/pnas.1508347112. 1128/IAI.70.2.859.
Rosowski, E.E., Lu, D., Julien, L., Rodda, L., Gaiser, R.A., Jensen, K.D.C., Saeij, J.P.J., Sims, T.A., Hay, J., Talbot, I.C., 1988. Host—parasite relationship in the brains of mice
2011. Strain-specific activation of the NF-kappaB pathway by GRA15, a novel with congenital toxoplasmosis. J. Pathol. 156, 255–261. https://doi.org/10.1002/
Toxoplasma gondii dense granule protein. J. Exp. Med. 208, 195–212. https://doi. path.1711560311.
org/10.1084/jem.20100717. Singh, S., Das, T., Ravindran, A., Chaturvedi, R.K., Shukla, Y., Agarwal, A.K., Dikshit, M.,
Rosowski, E.E., Nguyen, Q.P., Camejo, A., Spooner, E., Saeij, J.P.J., Adams, J.H., 2014. 2005. Involvement of nitric oxide in neurodegeneration: a study on the experimental
Toxoplasma gondii inhibits gamma interferon (IFN-γ)- and IFN-β-induced host cell models of Parkinson’s disease. Redox Rep. 10, 103–109. https://doi.org/10.1179/
STAT1 transcriptional activity by increasing the association of STAT1 with DNA. 135100005X38842.
Infect. Immun. 82, 706–719. https://doi.org/10.1128/IAI.01291-13. Skallová, A., Kodym, P., Frynta, D., Flegr, J., 2006. The role of dopamine in Toxoplasma-
Rosowski, E.E., Saeij, J.P.J., 2012. Toxoplasma gondii clonal strains all inhibit STAT1 induced behavioural alterations in mice: an ethological and ethopharmacological
transcriptional activity but polymorphic effectors differentially modulate IFNγ in- study. Parasitology 133, 525–535. https://doi.org/10.1017/S0031182006000886.
duced gene expression and STAT1 phosphorylation. PLoS One 7, e51448. https://doi. Skallová, A., Novotná, M., Kolbeková, P., Gasová, Z., Veselý, V., Sechovská, M., Flegr, J.,

90
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

2005. Decreased level of novelty seeking in blood donors infected with Toxoplasma. Vutova, P., Wirth, M., Hippe, D., Gross, U., Schulze-Osthoff, K., Schmitz, I., Lüder, C.G.K.,
Neuro Endocrinol. Lett. 26, 480–486. 2007. Toxoplasma gondii inhibits Fas/CD95-triggered cell death by inducing aber-
Steiner, J., Walter, M., Gos, T., Guillemin, G.J., Bernstein, H.-G., Sarnyai, Z., Mawrin, C., rant processing and degradation of caspase 8. Cell. Microbiol. 9, 1556–1570. https://
Brisch, R., Bielau, H., zu Schwabedissen, L., Bogerts, B., Myint, A.-M., 2011. Severe doi.org/10.1111/j.1462-5822.2007.00893.x.
depression is associated with increased microglial quinolinic acid in subregions of the Vyas, A., Kim, S.-K., Giacomini, N., Boothroyd, J.C., Sapolsky, R.M., 2007. Behavioral
anterior cingulate gyrus: evidence for an immune-modulated glutamatergic neuro- changes induced by Toxoplasma infection of rodents are highly specific to aversion of
transmission? J. Neuroinflammation 8, 94. https://doi.org/10.1186/1742-2094- cat odors. Proc. Natl. Acad. Sci. U. S. A. 104, 6442–6447. https://doi.org/10.1073/
8-94. pnas.0608310104.
Steinert, J.R., Chernova, T., Forsythe, I.D., 2010. Nitric oxide signaling in brain function, Wagner, A., Förster-Waldl, E., Garner-Spitzer, E., Schabussova, I., Kundi, M., Pollak, A.,
dysfunction, and dementia. Neuroscientist 16, 435–452. https://doi.org/10.1177/ Scheiner, O., Joachim, A., Wiedermann, U., 2009. Immunoregulation by Toxoplasma
1073858410366481. gondii infection prevents allergic immune responses in mice. Int. J. Parasitol. 39,
Stephan, A.H., Barres, B.A., Stevens, B., 2012. The complement system: an unexpected 465–472. https://doi.org/10.1016/j.ijpara.2008.09.003.
role in synaptic pruning during development and disease. Annu. Rev. Neurosci. 35, Wallace, M.N., Geddes, J.G., Farquhar, D.A., Masson, M.R., 1997. Nitric oxide synthase in
369–389. https://doi.org/10.1146/annurev-neuro-061010-113810. reactive astrocytes adjacent to beta-amyloid plaques. Exp. Neurol. 144, 266–272.
Stevens, B., Allen, N.J., Vazquez, L.E., Howell, G.R., Christopherson, K.S., Nouri, N., https://doi.org/10.1006/exnr.1996.6373.
Micheva, K.D., Mehalow, A.K., Huberman, A.D., Stafford, B., Sher, A., Litke, A.M., Walport, M.J., 2001. Complement. N. Engl. J. Med. 344, 1058–1066. https://doi.org/10.
Lambris, J.D., Smith, S.J., John, S.W.M., Barres, B.A., 2007. The classical complement 1056/NEJM200104053441406.
cascade mediates CNS synapse elimination. Cell 131, 1164–1178. https://doi.org/10. Wanet, A., Tacheny, A., Arnould, T., Renard, P., 2012. miR-212/132 expression and
1016/j.cell.2007.10.036. functions: within and beyond the neuronal compartment. Nucleic Acids Res. 40,
Stibbs, H.H., 1985. Changes in brain concentrations of catecholamines and indoleamines 4742–4753. https://doi.org/10.1093/nar/gks151.
in Toxoplasma gondii infected mice. Ann. Trop. Med. Parasitol. 79, 153–157. Wang, T., Liu, M., Gao, X.J., Zhao, Z.J., Chen, X.G., Lun, Z.R., 2011. Toxoplasma gondii:
Südhof, T.C., 2013. Neurotransmitter release: the last millisecond in the life of a synaptic the effects of infection at different stages of pregnancy on the offspring of mice. Exp.
vesicle. Neuron 80, 675–690. https://doi.org/10.1016/j.neuron.2013.10.022. Parasitol. 127, 107–112. https://doi.org/10.1016/j.exppara.2010.07.003.
Sugden, K., Moffitt, T.E., Pinto, L., Poulton, R., Williams, B.S., Caspi, A., 2016. Is tox- Wang, T., Tang, Z., Li, J., Li, X., Wang, X., Zhao, Z., 2013. Experimental Parasitology A
oplasma gondii infection related to brain and behavior impairments in humans? potential association between Toxoplasma gondii infection and schizophrenia in
Evidence from a population-representative birth cohort. PLoS One 11, e0148435. mouse models. Exp. Parasitol. 135, 497–502. https://doi.org/10.1016/j.exppara.
https://doi.org/10.1371/journal.pone.0148435. 2013.08.012.
Sutterland, A.L., Fond, G., Kuin, A., Koeter, M.W.J., Lutter, R., van Gool, T., Yolken, R., Wang, Z.T., Harmon, S., O’Malley, K.L., Sibley, L.D., 2015. Reassessment of the role of
Szoke, A., Leboyer, M., de Haan, L., 2015. Beyond the association. Toxoplasma gondii aromatic amino acid hydroxylases and the effect of infection by Toxoplasma gondii
in schizophrenia, bipolar disorder, and addiction: systematic review and meta-ana- on host dopamine. Infect. Immun. 83, 1039–1047. https://doi.org/10.1128/IAI.
lysis. Acta Psychiatr. Scand. 132, 161–179. https://doi.org/10.1111/acps.12423. 02465-14.
Tedford, E., Mcconkey, G., 2017. Neurophysiological changes induced by chronic tox- Wang, Z.T., Verma, S.K., Dubey, J.P., Sibley, L.D., 2017. The aromatic amino acid hy-
oplasma gondii infection. Pathogens 6, 19. https://doi.org/10.3390/ droxylase genes AAH1 and AAH2 in Toxoplasma gondii contribute to transmission in
pathogens6020019. the cat. PLoS Pathog. 13, 1–19. https://doi.org/10.1371/journal.ppat.1006272.
Tenter, A.M., Heckeroth, A.R., Weiss, L.M., 2000. Toxoplasma gondii: from animals to Wanko, T., Jacobs, L., Gavin, M.A., 1962. Electron microscope study of Toxoplasma cysts
humans. Int. J. Parasitol. 30, 1217–1258. in mouse brain. J. Protozool. 9, 235–242.
Terpsidis, K.I., Papazahariadou, M.G., Taitzoglou, I.A., Papaioannou, N.G., Georgiadis, Webster, J., Lamberton, P.H., Donnelly, C., Torrey, E., 2006. Parasites as causative agents
M.P., Theodoridis, I.T., 2009. Toxoplasma gondii: reproductive parameters in ex- of human affective disorders? The impact of anti-psychotic, mood-stabilizer and anti-
perimentally infected male rats. Exp. Parasitol. 121, 238–241. https://doi.org/10. parasite medication on Toxoplasma gondii’s ability to alter host behaviour. Proc. R.
1016/j.exppara.2008.11.006. Soc. Lond. B Biol. Sci. 273.
Thevandavakkam, M.A., Schwarcz, R., Muchowski, P.J., Giorgini, F., 2010. Targeting Weidner, J.M., Barragan, A., 2014. Tightly regulated migratory subversion of immune
kynurenine 3-monooxygenase (KMO): implications for therapy in Huntington’s dis- cells promotes the dissemination of Toxoplasma gondii. Int. J. Parasitol. 44, 85–90.
ease. CNS Neurol. Disord. Drug Targets 9, 791–800. https://doi.org/10.1016/j.ijpara.2013.09.006.
Tilders, F.J., DeRijk, R.H., Van Dam, A.M., Vincent, V.A., Schotanus, K., Persoons, J.H., Weidner, J.M., Kanatani, S., Hernández-Castañeda, M.A., Fuks, J.M., Rethi, B., Wallin,
1994. Activation of the hypothalamus-pituitary-adrenal axis by bacterial endotoxins: R.P.A., Barragan, A., 2013. Rapid cytoskeleton remodelling in dendritic cells fol-
routes and intermediate signals. Psychoneuroendocrinology 19, 209–232. lowing invasion by Toxoplasma gondii coincides with the onset of a hypermigratory
Tonin, A.A., Da Silva, A.S., Thomé, G.R., Sangoi, M.B., Oliveira, L.S., Flores, M.M., phenotype. Cell. Microbiol. 15, 1735–1752. https://doi.org/10.1111/cmi.12145.
Schetinger, M.R.C., Fighera, R.A., Moresco, R.N., Camillo, G., Vogel, F.S.F., Lopes, Weidner, J.M., Kanatani, S., Uchtenhagen, H., Varas-Godoy, M., Schulte, T., Engelberg,
S.T.A., 2014. Influence of toxoplasmosis on acetylcholinesterase activity, nitric oxide K., Gubbels, M.-J., Sun, H.S., Harrison, R.E., Achour, A., Barragan, A., 2016.
levels and cellular lesion on the brain of mice. Pathol. Res. Pract. 210, 526–532. Migratory activation of parasitized dendritic cells by the protozoan Toxoplasma gondii
https://doi.org/10.1016/j.prp.2014.04.025. 14-3-3 protein. Cell. Microbiol. 18, 1537–1550. https://doi.org/10.1111/cmi.12595.
Torres, L., Robinson, S.-A., Kim, D.-G., Yan, A., Cleland, T.A., Bynoe, M.S., 2018. Wong, S.Y., Remington, J.S., 1994. Toxoplasmosis in pregnancy. Clin. Infect. Dis. 18,
Toxoplasma gondii alters NMDAR signaling and induces signs of Alzheimer’s disease 853–861 quiz 862.
in wild-type, C57BL/6 mice. J. Neuroinflammation 15, 57. https://doi.org/10.1186/ Worth, A.R., Andrew Thompson, R.C., Lymbery, A.J., 2014. Reevaluating the evidence for
s12974-018-1086-8. toxoplasma gondii-induced behavioural changes in rodents. Advances in
Torrey, E.F., Bartko, J.J., Lun, Z.-R., Yolken, R.H., 2007. Antibodies to toxoplasma gondii Parasitology, 1st ed. Elsevier Ltdhttps://doi.org/10.1016/B978-0-12-800182-0.
in patients with schizophrenia: a meta-analysis. Schizophr. Bull. 33, 729–736. 00003-9.
https://doi.org/10.1093/schbul/sbl050. Wu, M.D., Montgomery, S.L., Rivera-Escalera, F., Olschowka, J.A., O’Banion, M.K., 2013.
Torrey, E.F., Yolken, R.H., 2003. Toxoplasma gondii and schizophrenia. Emerg. Infect. Dis. Sustained IL-1β expression impairs adult hippocampal neurogenesis independent of
9, 1375–1380. https://doi.org/10.3201/eid0911.030143. IL-1 signaling in nestin+ neural precursor cells. Brain Behav. Immun. 32, 9–18.
Torrey, E.F., Yolken, R.H., 1995. Could schizophrenia be a viral zoonosis transmitted from https://doi.org/10.1016/j.bbi.2013.03.003.
house cats? Schizophr. Bull. 21, 167–171. Xiao, J., Kannan, G., Jones-Brando, L., Brannock, C., Krasnova, I.N., Cadet, J.L.,
Treiman, D.M., 2001. GABAergic mechanisms in epilepsy. Epilepsia 42 (Suppl 3), 8–12. Pletnikov, M., Yolken, R.H., 2012. Sex-specific changes in gene expression and be-
Turnbull, A.V., Rivier, C., 1995. Regulation of the HPA axis by cytokines. Brain Behav. havior induced by chronic Toxoplasma infection in mice. Neuroscience 206, 39–48.
Immun. 9, 253–275. https://doi.org/10.1006/brbi.1995.1026. https://doi.org/10.1016/j.neuroscience.2011.12.051.
Tyebji, S., Hannan, A.J., 2017. Synaptopathic mechanisms of neurodegeneration and Xiao, J., Li, Y., Gressitt, K.L., He, H., Kannan, G., Schultz, T.L., Svezhova, N., Carruthers,
dementia: insights from Huntington’s disease. Prog. Neurobiol. https://doi.org/10. V.B., Pletnikov, M.V., Yolken, R.H., Severance, E.G., 2016a. Cerebral complement
1016/j.pneurobio.2017.03.008. C1q activation in chronic Toxoplasma infection. Brain Behav. Immun. 58, 52–56.
Uboldi, A.D., McCoy, J.M., Blume, M., Gerlic, M., Ferguson, D.J.P., Dagley, L.F., Beahan, https://doi.org/10.1016/j.bbi.2016.04.009.
C.T., Stapleton, D.I., Gooley, P.R., Bacic, A., Masters, S.L., Webb, A.I., McConville, Xiao, J., Li, Y., Prandovszky, E., Kannan, G., Viscidi, R.P., Pletnikov, M.V., Yolken, R.H.,
M.J., Tonkin, C.J., 2015. Regulation of starch stores by a Ca2+-Dependent protein 2016b. Behavioral abnormalities in a mouse model of chronic toxoplasmosis are
kinase is essential for viable cyst development in toxoplasma gondii. Cell Host associated with MAG1 antibody levels and cyst burden. PLoS Negl. Trop. Dis. 10,
Microbe 18, 670–681. https://doi.org/10.1016/j.chom.2015.11.004. 1–19. https://doi.org/10.1371/journal.pntd.0004674.
Ueno, N., Harker, K.S., Clarke, E.V., McWhorter, F.Y., Liu, W.F., Tenner, A.J., Lodoen, Xiao, J., Li, Y., Prandovszky, E., Karuppagounder, S.S., Talbot, C.C., Dawson, V.L.,
M.B., 2014. Real-time imaging of Toxoplasma-infected human monocytes under Dawson, T.M., Yolken, R.H., 2014. MicroRNA-132 dysregulation in Toxoplasma
fluidic shear stress reveals rapid translocation of intracellular parasites across en- gondii infection has implications for dopamine signaling pathway. Neuroscience 268,
dothelial barriers. Cell. Microbiol. 16, 580–595. https://doi.org/10.1111/cmi.12239. 128–138. https://doi.org/10.1016/j.neuroscience.2014.03.015.
Uher, R., Zwicker, A., 2017. Etiology in psychiatry: embracing the reality of poly-gene- Yagmur, F., Yazar, S., Temel, H.O., Cavusoglu, M., 2010. May Toxoplasma gondii increase
environmental causation of mental illness. World Psychiatry 16, 121–129. https:// suicide attempt-preliminary results in Turkish subjects? Forensic Sci. Int. 199, 15–17.
doi.org/10.1002/wps.20436. https://doi.org/10.1016/j.forsciint.2010.02.020.
Valtorta, F., Pennuto, M., Bonanomi, D., Benfenati, F., 2004. Synaptophysin: leading actor Yarovinsky, F., 2014. Innate immunity to Toxoplasma gondii infection. Nat. Rev.
or walk-on role in synaptic vesicle exocytosis? Bioessays 26, 445–453. https://doi. Immunol. 14, 109–121. https://doi.org/10.1038/nri3598.
org/10.1002/bies.20012. Yereli, K., Balcioğlu, I.C., Özbilgin, A., 2006. Is Toxoplasma gondii a potential risk for
van Spronsen, M., Hoogenraad, C.C., 2010. Synapse pathology in psychiatric and neu- traffic accidents in Turkey? Forensic Sci. Int. 163, 34–37. https://doi.org/10.1016/j.
rologic disease. Curr. Neurol. Neurosci. Rep. 10, 207–214. https://doi.org/10.1007/ forsciint.2005.11.002.
s11910-010-0104-8. Yeshurun, S., Hannan, A.J., 2018. Transgenerational epigenetic influences of paternal

91
S. Tyebji et al. Neuroscience and Biobehavioral Reviews 96 (2019) 72–92

environmental exposures on brain function and predisposition to psychiatric dis- neuronal apoptosis in Toxoplasmic encephalitis. Parasites Vectors 7, 372. https://doi.
orders. Mol. Psychiatry 1. https://doi.org/10.1038/s41380-018-0039-z. org/10.1186/1756-3305-7-372.
Yeshurun, S., Rogers, J., Short, A.K., Renoir, T., Pang, T.Y., Hannan, A.J., 2017. Elevated Zheng, X., Zhang, X., Kang, A., Ran, C., Wang, G., Hao, H., 2015. Thinking outside the
paternal glucocorticoid exposure modifies memory retention in female offspring. brain for cognitive improvement: Is peripheral immunomodulation on the way?
Psychoneuroendocrinology 83, 9–18. https://doi.org/10.1016/j.psyneuen.2017.05. Neuropharmacology 96, 94–104. https://doi.org/10.1016/j.neuropharm.2014.06.
014. 020.
Yolken, R., Dickerson, F., Torrey, F., 2009. Toxoplasma and schizophrenia. Parasite Zimmerman, A.W., Jyonouchi, H., Comi, A.M., Connors, S.L., Milstien, S., Varsou, A.,
Immunol. 31, 706–715. https://doi.org/10.1111/j.1365-3024.2009.01131.x. Heyes, M.P., 2005. Cerebrospinal fluid and serum markers of inflammation in autism.
Zhang, Y., Chen, H., Chen, Y., Wang, L., Cai, Y., Li, M., Wen, H., Du, J., An, R., Luo, Q., Pediatr. Neurol. 33, 195–201. https://doi.org/10.1016/j.pediatrneurol.2005.03.014.
Wang, X., Lun, Z.-R., Xu, Y., Shen, J., 2014. Activated microglia contribute to

92

You might also like