You are on page 1of 18

Acta Tropica 231 (2022) 106432

Contents lists available at ScienceDirect

Acta Tropica
journal homepage: www.elsevier.com/locate/actatropica

Toxoplasmosis in Human and Animals Around the World. Diagnosis and


Perspectives in the One Health Approach
Rosangela Aparecida Müller de Barros a, b, 1, Ana Claudia Torrecilhas c, 2,
Maria Aparecida Moraes Marciano d, 3, Monica Leszkowicz Mazuz f, 4,
Vera Lucia Pereira-Chioccola e, 5, **, Blima Fux a, b, 6, *
a
Unidade de Medicina Tropical, Departamento de Patologia, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil.
b
Programa em Doenças Infecciosas, Centro de Doenças Infecciosas, Universidade Federal do Espirito Santo, Vitoria, ES, Brazil.
c
Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP),
Campus Diadema, Sao Paulo, SP, Brazil.
d
Núcleo de Morfologia e Microscopia, Instituto Adolfo Lutz, Sao Paulo, SP, Brazil.
e
Centro de Parasitologia e Micologia, Instituto Adolfo Lutz, Sao Paulo, SP, Brazil.
f
Parasitology Division, Kimron Veterinary Institute, Israeli Veterinary Service and Animal Health, Ministry of Agriculture and Rural Development Beit Dagan, 5025000,
Israel.

A R T I C L E I N F O A B S T R A C T

Key-words: Toxoplasmosis is a unique health disease that significantly affects the health of humans, domestic animals,
Toxoplasma gondii wildlife and is present in ecosystems, including water, soil and food. Toxoplasma gondii is one of the best-adapted
Toxoplasmosis parasites in the word. This parasite is able to persist for long periods in its hosts, in different geographic regions
Laboratorial diagnosis
of the word. This review summarizes the current literature of these themes, focusing on: (1) toxoplasmosis, a
One health approach
Extracellular vesicles
zoonotic infection; (2) One health approach and toxoplasmosis; (3) human toxoplasmosis; (4) animal toxo­
plasmosis; (5) toxoplasmosis diagnosis, as immunological, parasitological and molecular diagnosis; (6) T. gondii
outbreaks caused by infected meat, milk and dairy products, as well as, vegetables and water consume; (7)
studies in experimental models; (8) genetic characterization of T. gondii strains; (9) extracellular vesicles and
miRNA; and (10) future perspectives on T. gondii and toxoplasmosis. The vast prevalence of toxoplasmosis in
both humans and animals and the dispersion and resistence of T. gondii parasites in environment highlight the
importance of the one health approach in diagnostic and control of the disease. Here the different aspects of the
one health approach are presented and discussed.

1. Toxoplasmosis, a Zoonotic Infection South American felines with relative expansion through migratory birds.
Probably, the transatlantic slave trade promoted the migration of do­
Toxoplasma gondii is an obligate intracellular parasitic protozoan, mestic cats, rats and mice (Lehmann et al., 2006).
causative agent of toxoplasmosis that is widely distributed (Ybañez T. gondii is one of the best adapted parasites, being able to infect
et al., 2020). According to recent studies T. gondii was originated in innumerous species of animals and different types of cells. Thus, these

* Corresponding author at: Unidade de Medicina Tropical, Departamento de Patologia, Universidade Federal do Espirito Santo, Av. Marechal Campos, 1468,
Vitória, ES, CEP 29043-900, Brazil.
** Corresponding author at: Centro de Parasitologia e Micologia, Instituto Adolfo Lutz, Sao Paulo, SP, Brazil.
E-mail addresses: ramuller.es@gmail.com (R.A.M. de Barros), ana.torrecilhas@unifesp.br (A.C. Torrecilhas), maria.marciano@ial.sp.gov.br (M.A.M. Marciano),
monical@moag.gov.il (M.L. Mazuz), pchioccola@gmail.com (V.L. Pereira-Chioccola), blima.fux@ufes.br (B. Fux).
1
ORCID: 0000-0001-5924-8789
2
ORCID: 0000-0001-5724-2199
3
ORCID: 0000-0002-6720-736X
4
ORCID: 0000-0001-5357-0136
5
ORCID: 0000-0003-3317-195X
6
ORCID: 0000-0002-5038-3551

https://doi.org/10.1016/j.actatropica.2022.106432
Received 25 January 2022; Received in revised form 25 March 2022; Accepted 27 March 2022
Available online 4 April 2022
0001-706X/© 2022 Elsevier B.V. All rights reserved.
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

parasites are able to persist for long periods in their hosts, probably for a In recent years, toxoplasmosis has become the subject of numerous
lifetime. The course of the infection and pathogenicity depends on the studies. There is solid knowledge of T. gondii genome, life cycle, trans­
doses of inoculated parasites, parasite genetic background, host genetics mission routes and epidemiology. Furthermore, T. gondii strains are
and immunological status (Suzuki et al., 1996; Habegger de Sorrentino easily maintained and manipulated in the laboratory, making it a model
et al, 2005; Hunter and Sibley 2012). organism for studying many aspects of its biology (Boothroyd, 2009).
T. gondii is a tissue cyst forming coccidia thus having the ability to Studies also evaluated the high and less virulent strains, susceptibility
encyst within host tissue cells. In a failure of the host immune system, based on genetic characteristics (Carme et al., 2009; Ngô et al., 2019).
parasites are converted back to their proliferative stage. This is an The infection is subclinical in the vast majority of cases, and this char­
evaluative shift, circumventing the need of parasites to go through their acteristic makes difficult the effective control of toxoplasmosis.
sexual stage to be transmitted to new hosts. This property gives to Despite advances in the knowledge of T. gondii biology and the
T. gondii a second route of transmission and the ability to clonally spread development of new diagnostic methods, there is still no efficient
through intermediate hosts, especially in immunocompromised animals parasite control. The development of a vaccine for use in humans is still
or during pregnancy (Sullivan and Jeffers, 2012). a global challenge, as it is considered a neglected disease. Therefore, in
Felids are the definitive hosts of T. gondii and are responsible for the the absence of an effective vaccine the control of the human infection
production and shedding the environmentally resistant oocyst. Human should be based on the one health control approaches such as hygienic
and animal infections are mostly acquired by ingesting food or water measures, reduction of infection in animals, reduction of contamination
contaminated with sporulated oocysts or undercooked meat infected of the environment and water, and others. To date, there is only one
with latent cysts. Other ways of infection include congenital trans­ commercially available vaccine that is licensed for use in sheep against
mission (during pregnancy), blood transfusion and organ trans­ abortion (Innes et al., 2019).
plantation (Montoya and Liesenfeld, 2004). The zoonotic potential and pathogenicity of atypical strains of
The infection occurs in the different geographic regions. The prev­ T. gondii common in wildlife are often unknown (Thompson, 2013).
alence is higher in area with warm, humid climates and lower altitudes. Virulence varies between different genotypes (Dardé, 2008; Costa Vie­
The oocysts are resistant and survive better in humid environments gas de Lima et al, 2019). There is evidence of abnormally high levels of
(Weiss and Kim, 2020). human exposure to T. gondii in some regions of the Canadian Arctic.
Estimates of human infection in Central America, South America and Transmission of T. gondii is complex and enigmatic in the Arctic, where
Continental Europe range from 30 to 90% (Dubey and Jones, 2008; definitive feline hosts are scarce. Possible routes of transmission include
Jones and Dubey, 2010; Dubey, 2010a; Minbaeva et al., 2013; Wilking oocysts shed in feline feces driven by marine and freshwater routes from
et al., 2016). Epidemiological studies carried out in different countries subarctic regions and (Lindsay et al., 2003; Simon et al., 2013) trophic
revealed that the prevalence in Germany was around 55% (Wilking and vertical routes of transmission.
et al., 2016; Lassen et al., 2016); in China, from 3.4% to 17.5% (Liu A better understanding of the contamination of the environment by
et al., 2017, Pan et al., 2017); in the United States of America, around oocysts and the actual epidemiology of the cases are critical to deter­
11.4% (Jones et al., 2001; Jones et al., 2009; Jones and Dubey, 2014); in mine actions to control the disease. Studies indicate that large regions of
Brazil, the prevalence ranging from 52.6% to 72.3% (Costa et al., 2018). terrestrial, aquatic and marine environments may be contaminated (Du
Studies conducted in Israel showed a wide variation of seropositivity, et al., 2012; Vanwormer et al., 2013; Gao et al., 2016). Although
ranging from 9.8% to 54.9% in different ethnic populations (Markovich different studies, additional research is needed to elucidate environ­
et al., 2014). mental conditions responsible for oocyst survival and resistance, extent
of environmental contamination, and tissue cyst survival or infectivity
2. One Health Approach and Toxoplasmosis after host death VanWormer et al., 2013; VanWormer et al., 2013a).
The first strategy to reduce the burden of toxoplasmosis in humans
One Health recognizes the interconnectedness of human, animal, would be the knowledge of prevalence, in selected areas, of the human
plant and environmental health at the local and global levels. One toxoplasmosis Next, the intervention of animal reservoirs. These actions
Health still employs an interdisciplinary approach encouraging and would require close collaboration of different disciplines, such as med­
expanding collaborations in integrative research, capacity building, icine, public health, veterinary, food safety, biology, genetics, environ­
clinical practice, policy and communication across diverse fields mental and water science and water science (Patz and Hahn, 2013).
(Aguirre and Wilcox, 2008; Aguirre et al., 2019). This approach can Collaboration between professionals from different fields as well as,
cross bureaucratic boundaries and represents an opportunity for new organizations and institutions from various countries would aid the
partnerships focused on solutions to complex problems (Zinsstag, 2012). understanding of the ecological interactions. As summarized in Fig. 1,
Toxoplasmosis as a zoonotic infection, significantly affects the health such actions would facilitate a clearer understanding the impacts of this
of humans, domestic and wildlife animals. T. gondii is able to infect zoonotic disease on animal and human health, as well as, in
animals in all ecosystems, including water, soil and food (Crozier and environmental.
Schulte-Hostedde, 2014; Jenkins et al. 2015). Due to its ability to suc­
cessfully spread throughout the ecosystem and on diverse hosts, T. gondii 3. Human Toxoplasmosis
is one of the most successful parasites. Studies estimate that T. gondii
already infected more than a third of the global human population Toxoplasmosis prevalence in humans is highly variable since it is
(Montoya and Liesenfeld, 2004). Thus, toxoplasmosis is considerate as a influenced by socioeconomic and environmental conditions, as well as,
global health hazard since its global seroprevalence is high. However, cultural habits (Dubey et al., 2012; Mareze et al., 2019). The three main
this seroprevalence varies widely from one region to another due to routes of T. gondii transmission include vertical transmission from
differences in climate, diet, hygiene, habits and host susceptibility mother to fetus, ingestion of tissue cysts from infected animal tissues,
(Flegr et al., 2014; Nogareda et al., 2014). and ingestion of oocysts from contaminated water, soil or food (Tenter
As disease burden, toxoplasmosis is ranked among one of the highest et al., 2000; Montoya and Liesenfeld, 2004).
infections (Suijkerbuijk et al., 2018; Smith et al., 2021). The toxoplas­ The infection is normally asymptomatic or sub-clinical however
mosis burden can be identified as years of life lost and years lived in less primary infection during pregnancy can cause damage to fetus and even
health. The aggregate of both measures determines quantification of abortion. In addition, primary infection or recrudescence of parasitemia
years of healthy life lost due to a particular disease or infection. This in immunocompromised humans and animals lead to severe neurolog­
equation is known as "Disability Adjusted Life Years" (Mangen et al., ical clinical signs causing significant impacts on public health and ani­
2015). mal production (Dubey and Jones, 2008; Dubey et al., 2020).

2
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

Fig. 1. One Health an interdisciplinary approach for toxoplasmosis control. T. gondii lives in ecosystems, as water, soil and food. Due to it successfully ability to
spread throughout the ecosystem and on many hosts, T. gondii has become one of the most successful parasites on the planet. Successful public health interventions
require the cooperation of partners working with human and animal health, as well as, the environmental actions. The One Health concept proposes collaborative
multi-sectorial and interdisciplinary approaches with the goal of achieving optimal health outcomes. Efforts to better understand toxoplasmosis and its real
epidemiology are crucial for controlling of this infection. The collaboration between professionals from different areas, such human health professionals (physicians,
nurses, biologists, epidemiologists and others), animal health (veterinarians, pet owners, paraprofessionals, agricultural workers), environment (biologists, ecolo­
gists, educators, wildlife specialists), and other areas of expertise need to communicate, collaborate and coordinate activities to better understand the ecological
interactions and impacts of this zoonotic disease. Other relevant actors include policy makers, farmers and different communities.

In the beginning of infection, T. gondii can spread to mesenteric multiple clinical manifestations in chronic infection are widely
lymph nodes and then to distant organs by invasion of lymphatic vessels described as in inflammation, usually following necrosis (Holland Gary,
and blood and can multiply in virtually any cell in the body. Like many 2004; Bogitsh et al., 2018). Around 10-20% of the infected immuno­
obligate intracellular microbial pathogens, T. gondii infection induces a competent individuals, during the chronic infection are reactive by the
type 1 polarized immune response, engaging both innate and adaptive rupture of a tissue cyst in eyes, brain or muscles causing local necrosis
immune systems. It is clear that the major mechanism of host resistance accompanied by inflammation. Hypersensitivity plays a major role in
to toxoplasmosis is mediated by production of pro-inflammatory cyto­ such reactions. However, when infection collapses the eyes establishe
kines (Habegger de Sorrentino et al, 2005; Bogitsh et al., 2018). the tachyzoite multiplication (Montoya and Liesenfeld, 2004; Vidal and
The chronic infection appears after two-three weeks of infection, Pereira-Chioccola, 2021).
when the production of cytokines and antibodies against many T. gondii Ocular toxoplasmosis usually manifests as recurrent attacks of
proteins begin. Extracellular tachyzoites are cleared from host tissues necrotizing retinal inflammation with subsequent scarring (Bala­
and intracellular parasites differentiate into latent bradyzoite forms, sundaram et al., 2010; Kianersi et al., 2012). The clinical manifestations
which are surrounded by the parasitophorous vacuole that is enclosed in of ocular toxoplasmosis partly reflect the parasite infection and partly
a cyst wall. In this clinical phase, cysts preferentially reside in neural and the host immune response, which often involves exuberant inflamma­
muscular tissues and are periodically ruptured. However, the majority of tion, is recognized as a leading cause of blindness in many parts of the
them persist indefinitely in the brain and muscles, developing life-long world.
protective immunity against reinfection, since bradyzoites released are Ocular lesions may result from congenital or after birth-acquired
normally destroyed by the host immune response (Suzuki et al., 1989; infections. The lesion is often necrotic, destroying the architecture of
Gazzinelli e al, 1992; Schlüter et al., 1997; Schlüter et al., 2003; Dubey the neural retina and sometimes involving the choroid (retinochor­
et al., 2008; Pereira-Chioccola et al., 2009). The ability of bradyzoites to oiditis), the most common lesion caused by infection with this proto­
escape the host immune response and persist in a quiescent form within zoan. Typical findings of toxoplasmic retinochoroiditis include white
the host is another event in the T. gondii life cycle. focal lesions with an intense overlaying and vitreal inflammatory reac­
If parasite multiplication is not controlled by the immune system, it tion (Montoya and Remington, 1996; Klaren and Kijlstra, 2002; Vallochi
causes a generalized toxoplasmosis, which is always fatal (Vidal and et al., 2005; Arevalo et al., 2010). Alternatively, retinochoroiditis might
Pereira-Chioccola, 2021). Host T-cell-mediated immune responses play result from a hypersensitivity reaction triggered by unknown causes
an important role in suppression of tachyzoite replication and resistance (Commodaro et al., 2009), resulting in irreversible damage to the retina
to T. gondii, resulting in chronic infection or possibly clearance of the involved, whose consequences may be severe visual morbidity (Previato
parasites (Montoya and Liesenfeld, 2004; Melo et al., 2020). Although et al, 2015).
the infection is usually asymptomatic, in immunocompetent individuals, Toxoplasmic retinochoroiditis has been reported as more severe in

3
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

Brazil than in Europe. Brazilian mothers received later prenatal treat­ 2011; Oliveira et al., 2018).
ment. Consequently, seroconvertion determination is detected later in T. gondii cause acute and fatal disseminated toxoplasmosis in wild
pregnancy. European mothers received early prenatal treatment animals with macropodidae, monkeys and meerkats being highly sus­
reducing the risk of fetuses with ocular disease (Gilbert et al., 2008). ceptible for infection (Cunningham et al., 1992; Juan-Sallés et al., 1997;
Another point that favors the frequency and severity of ocular disease in Portas, 2010). Due to its great importance as a causative agent of a
Brazil compared with Europe is due to exposure to more virulent strains zoonosis, T. gondii has been studied more intensively among coccidian
of T.gondii in Brazil. Type 1 and atypical strains appear to be associated (Tenter et al., 2000).
with more severe ocular disease, compared with type II strains, which The environmental contamination occurs by oocysts that are shed by
predominate in Europe and North America (Vallochi et al., 2005; Kan domestic cats and other wild felids (Frenkel et al., 1970; Dubey, 2008;
et al., 2006). These data favor understanding differences in the risk of Dubey, 2009). The oocyst stage has a resistant wall that protects para­
retinochoroiditis in Brazil and Europe (Gilbert et al., 2008). sites from severe environmental conditions until their ingestion by the
Despite that, the diagnosis of ocular toxoplasmosis is typically clin­ host (Dumètre et al., 2013). Different T. gondii biological parameters are
ical; laboratory testing normally provides a definitive diagnosis. The not totally known, as the environmental conditions that cause the oocyst
presence of anti T. gondii IgG antibodies do not confirm a toxoplasmic long survival and resistance. (Dubey, 2010a). Despite the important role
etiology, but such antibodies can often persist at high titers for years of epidemiology, environmental transmission remains the least studied
after acute infection, and a negative result generally contradicts the (Shahet al, 2019). Even with epidemiological progress about T. gondii
diagnosis. T. gondii DNA has been identified in ocular tissue sections and infecting humans and animals, the environmental contamination and
vitreous fluid and blood (Grigg et al., 2001; Rothova et al., 2008, Mattos the factors that contribute to the survival of oocysts in the environment
et al., 2011; Ferreira et al., 2014). are not totally established (Maleki et al., 2021; Oliveira et al., 2021).
The reactivation of latent infection in asymptomatic individuals can Domestic and wild felids are the only known definitive hosts capable
be caused by some immunodeficiency. Thus, bradyzoites convert into of releasing environmentally resistant oocysts into their feces (Hutch­
active and rapidly replicating tachyzoites that may result in clinical ison et al., 1969; Jewell et al., 1972; Dubey et al., 2008; Miller et al.,
toxoplasmosis and even cause fatal tissue injury. As the cysts have a 2018). Young cats can release greater numbers of oocysts than adult
predilection for neural and muscle tissue as well as the eye, most cases of domestic cats. Interest in the evaluation of T. gondii infection has focused
reactivation disease presenting, more frequently, cerebral toxoplasmosis on domestic animals that cohabit or serve as food for humans, as these
that is a life-threatening condition (Pereira-Chioccola et al., 2009; animals can act as reservoirs for human infections (Sogorb et al., 1977;
Bogitsh et al., 2018). Cerebral toxoplasmosis is usually the most com­ Ruiz and Frenkel 1980; Dubey and Beattie, 1988).
mon cerebral opportunistic disease in both developed and developing T. gondii infects a variety of domestic animals, including dogs
countries. The disease reactivation caused by immunosuppression cau­ (Machado et al., 2019; Dubey et al., 2020a; Arruda et al., 2021); sheep
ses a serious neurological disease. Usually, immunosuppression is and goats (Bártová et al, 2009; Chikweto et al, 2011; Gharekhani et al.,
caused by the use of immunosuppressed drugs such as transplant 2018; Moskwa et al., 2018); pigs (Saavedra and Ortega, 2004; Guo
recipient patients, patients with lymph-proliferative diseases or HIV et al., 2015; Flores et al., 2021); chickens (Mahami-Oskouei et al., 2017;
infection (Vidal et al., 2003; Pereira-Chioccola et al., 2009; Vidal et al., Ferreira et al., 2018; Dubey et al., 2021a; Lachkhem et al., 2021); ducks
2011; Vidal and Oliveira, 2013; Vidal and Pereira-Chioccola, 2021). (Lv et al., 2021); cattle (Moore et al., 2008; Jokelainen et al., 2017;
Globally, T. gondii causes the most common focal brain lesion in Gomes et al., 2020; Shariatzadeh et al., 2021); horses (Li et al., 2020);
HIV-infected patients (Manzardo et al., 2005; Vidal et al., 2005; Vidal rabbits (Almería et al., 2021); wild boar (Rostami et al., 2017; Laforet
et al., 2008). et al., 2019); camels (Maspi et al., 2021); among others. In sheep and
In congenital infection, tachyzoites cross the placenta and reach the goats, congenital infection is a major cause of stillbirth. Those who
fetus. Women acutely infected for the first-time during pregnancy may survive generally grow normally, but still pose a public health risk if
cause severe damage to the fetus with high morbidity and mortality in their infected meat is consumed (Dubey, 2009; Oliveira et al., 2018).
congenitally infected infants. Infections acquired during the first Sheep and pigs are the most susceptible to toxoplasmosis. Cattle and
trimester are more severe for the fetus than those acquired in the second equines are easily infected, but resistant to acute disease, which makes
and third trimesters (Desmonts and Remington 1980; Remington et al., them important reservoirs for transmission to humans and other ani­
1995; Dubey et al., 2021). mals. Chickens are also resistant to T. gondii infection, but studies have
The clinical signs can be stillbirth, premature birth, intrauterine shown a high frequency of T. gondii in free-range chicken (Dubey et al.,
growth retardation, fever, pneumonia, hepatosplenomegaly, thrombo­ 2005; Dubey and Jones, 2014; Magalhães et al., 2016).
cytopenia involvement in eyes and brain (McAuley et al., 1994; Peyron Several wildlife species play an important role in the transmission
et al., 2016). However, a great part of the congenitally infected new­ and maintenance of T. gondii in the environment (Yai et al., 2009; Dubey
borns presents no clinical signs, but they are at risk of developing ocular et al, 2010; Pena et al., 2011; Cabral et al., 2013; Cañón-Franco et al.,
lesions in childhood or adolescence (Remington et al., 2001; Dubey 2013). Detection in apparently healthy and free-living wild animals
et al., 2021). suggests that asymptomatic or subclinical infections may occur. Wildlife
routes of infection include consumption of infected felines, predation or
4. Animal Toxoplasmosis elimination of infected intermediate hosts, direct ingestion of oocysts in
contaminated environments, or congenital transmission.
T. gondii is found in all habitats and regions, from the Arctic to the T. gondii infections were shown in a wide range of wild species,
tropics in terrestrial, aquatic and marine environments, affecting a wide including freshwater mammals (Santos et al., 2011; Ribas et al., 2018);
range of hosts (Sibley 2003; Dubey and Jones 2008; Dubey, 2010). marine mammals (Fayer et al., 2004; Jensen et al., 2012; Rengifo-­
Theoretically all warm-blooded animals can be infected and at least 350 Herrera et al., 2012; Miller et al., 2018); non-human primates
host species have been described to date (Zhao and Ewald, 2020). (Catão-Dias et al., 2013; Cano-Terriza et al., 2019; Paula et al., 2020);
Toxoplasmosis causes important clinical infection in small ruminants birds (Frenkel et al., 1995; Dubey, 2002; Dubey, 2010a; Darwich et al.,
being a major cause of abortion (Anderson et al., 1994; Oliveira et al., 2012; Barroso et al., 2020; Ammar et al., 2021; Dubey et al., 2021a);
2018; Lindsay and Dubey, 2020). In addition, T. gondii infection is ungulates (Dubey et al., 2014; Almería et al., 2018); polar, brown and
considered an important cause of reproductive disorders in goats (Cal­ black bears (Chomel et al., 1995; Oksanen et al., 2009); terrestrial
deira et al., 2011; Unzaga et al., 2014). Abortion cases in goats have herbivores (Ballash et al., 2015); among others.
been described in different regions of the world such as Spain, (Moreno Aquatic invertebrates can significantly influence the aquatic trans­
et al., 2012), Argentina, (Unzaga et al., 2014) and Brazil (Caldeira et al., port of T. gondii by ingestion, which can transmit the infectious stage to

4
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

susceptible hosts, including marine mammals and humans (Shapiro seroprevalence for T. gondii in the general population is very high. In
et al., 2014). Although cold-blooded animals may not serve true inter­ Brazil, most AIDS patients with any focal brain lesion also show positive
mediate hosts, T. gondii oocysts can be ingested and retained by fish serological diagnosis for toxoplasmosis However, the identification of a
(Massie et al., 2010); bivalves (Arkush et al., 2003; Lindsay et al., 2003) negative serological result presents a high negative predictive value
and marine snails (Krusor et al., 2015; Schott et al., 2016). Ingestion of (Vidal et al., 2008; Vidal and Pereira-Chioccola, 2021).
these invertebrates has been epidemiologically associated with an MAT is a sensitive serological method to detect T. gondii IgG anti­
increased risk of T. gondii infections in marine mammals or people who bodies in herds and wild animals. MAT primarily detects antibodies from
consume seafood (Jones et al., 2009). animal tissue fluid, serum or plasma (Al-Adhami, 2016). It is the most
When cats have free access to a farm premises and, principally, to widely used method for diagnosis, the most economical and the simplest
pastures, the other domestic or farm animals are very exposured to among many methods available to detect T. gondii infection, and there is
T. gondii infection (Guo et al., 2015). The exposure to contaminated soil no need for specific equipment (Hill et al., 2006, Dubey, 2010a). In most
with oocysts is the main transmission route to these animals that are laboratories, the development of MAT is performed with the aid of
intermediate hosts and will be food to humans (Gilot-Fromont et al., tachyzoites collected by traditional propagation performed in murine
2012). peritoneal cavity in vitro culture (Bachand et al, 2019). Among the
advantages of this method is that it can be used for birds and mammals
5. Diagnosis of Toxoplasmosis with easy interpretation of results, its high sensitivity and specificity, as
well as its ease of execution, with no need for specific equipment
The diagnosis is crucial for the surveillance, prevention and control (Desmonts and Remington, 1980). The antigen is stable at 4◦ C for many
of toxoplasmosis. Traditional approaches to the laboratorial diagnosis months and does not need species-specific reagents (Langoni et al.,
include etiological, immunological, molecular and histopathological/ 2007; Macrì et al, 2009).
immunohistochemical methodologies. Currently, two biological ap­ ELISA has been used as the most reliable, practical, economical and
proaches are used: (i) indirect detection through immunological assays widely used for detecting exposure to T. gondii in hosts (Titilincu et al.,
and (ii) direct detection as parasitological methods using microscopy to 2009; Tumurjav et al, 2010). Only a small sample volume is needed, and
parasite detection. The other is the molecular methodology that detects the assay can be semi-automatic, making it suitable for large-scale
the parasite DNA in suspected samples (Table 1). screening (Felin et al., 2017). Furthermore, different ELISAs can
differentiate between immunoglobulin classes. Therefore, it is useful for
5.1. Indirect Methodologies: Immunological and Imunohistochemical determining the infection stage (Lind et al., 1997). Different method­
Diagnosis ologies for ELISA were standardized using various antigens (native, re­
combinant, chimeric), secondary antibodies and antibody binding
A wide range of serological tests for T. gondii detection is available reagents (Ferra et al., 2015). However, the choice of components can
with various methodologies and antigens that can detect different significantly influence test performance. Commercial ELISA kits for the
immunoglobulin isotypes. Most are not Rapid Diagnostic Test (RDT) and detection of T. gondii antibodies in domesticated animals are available,
are aimed at laboratories with good infrastructure and qualified making large-scale and routine screening feasible.
personnel. Immunological techniques are relatively inexpensive, require IFAT is a simple test that detects IgG and IgM antibodies and has been
a small sample volume, and can be used on live animals (Elmore et al., widely used in the detection of antibodies against T. gondii in humans
2016; Cakir-Koc, 2016). Assays include the Sabin-Feldman (DT) dye test, and animals (Miller et al., 2002). Fluorescence-labeled antibodies to a
based on tachyzoite stains under microscopy; enzyme-linked immuno­ variety of species are commercially available. However, a fluorescence
sorbent assay (ELISA), immunosorbent agglutination assay (ISAGA), microscope is required for the test and the results are read with the
indirect hemagglutination assay (IHA), indirect fluorescent antibody naked eye, and individual variation may occur. Some species-specific
test (IFAT), modified agglutination test (MAT), Latex Agglutination Test conjugates can be difficult to find, and there is a risk of possible
(LAT) and Western blot (WB) (Liu et al., 2015; Sun et al., 2015; Dard cross-reactivity with rheumatoid factor and antinuclear antibodies
et al., 2016). (Filice et al., 1983).
Humans present a lifelong persistence of anti-T. gondii IgG antibodies Immunohistochemical diagnosis is frequently used for determination
and, in many geographical areas, toxoplasmosis prevalence is high. of fetal brain damage in early abortions of ovine toxoplasmosis
Thus, in many cases, the serological titers do not reflect a recent infec­ (Gutiérrez-Expósito et al, 2020), necropsies of wild and marine mam­
tion. In this way, it is often the lack of reliability in discriminating recent mals as cetaceans (Sanots et al., 2018; Costa-Silva et al., 2019), and
from old infection by detection of anti-T. gondii IgM, IgA, or IgE. This autopsies for undefined death investigation (Bastos da Silva et al, 2016;
situation is very common in congenital toxoplasmosis diagnosis (Per­ Hippólito et al., 2019).
eira-Chioccola et al. 2009). In the case of cerebral toxoplasmosis, the In addition bioassays in mice and cats can be used for T. gondii
identification of a positive serology is less useful in settings where the isolation, infection confirmation. This method is considered gold

Table 1
Strategies for T. gondii diagnostic in samples from humans, animals and environmental.
Methods for diagnostic of T. gondii infection/contamination Humans Animals *Environment

Indirected detection Serology Sabin –Feldman (DT) X


ISAGA/IHA X
ELISA/Avidity ELISA X X
MAT/LAT X X
IFAT/ WB X X
Biopsy/ Necropsy/authopsy Pathology (histology) X X
Immunohistochemical X X
Direct detection Microscopic examination X X X
Electron microscopy X X X
Molecular detection X X X
Isolation (bioassays and culture) X
*
Detection of T. gondii oocysts in environmental samples (soil, water, feed) is technically difficult. Normally samples are concentrated (filtrated) and prepared with
the combination of several techniques.

5
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

standard for detecting viable tissue cysts (Dubey et al., 2016). In tissues levels in different moments of infection, as during the course of therapy
of chronically infected animals the number of parasites is low therefore (Contini et al., 2005). Among them, two are more frequently used by the
bioassays can be of great importance. Bioassay in cats is considered a great sensitivity and specificity. One is the 529-bp sequence, which has
highly sensitive method of detecting T. gondii bradyzoites in tissues. Cats 200-300 copies in the genome of T. gondii. The other is the B1 gene that
experiment fed with as few as one bradyzoite may excrete large numbers has 35 copies in the genome and is conserved in different parasite strains
of oocysts in their feces (Dubey et al., 1995). (Costa and Bretagne 2012; Robert-Gangneux et al., 2010). The sensi­
tivity and accuracy of the targets 529-bp sequence, B1 gene and the
5.2. Direct T. gondii Detection: Parasitological and Molecular Diagnosis comparison of both were analyzed in conventional PCR and qPCR
(Holman et al., 2000; Belaz et al., 2015; Camilo et al., 2017). They have
T. gondii detection in feces, water, environmental and tissue samples shown markers to 529 bp repeat region that were more sensitive than
has been traditionally performed using microscopy. However, the those from B1 gene.
identification based on this method is less sensitive and unreliable (Liu
et al., 2015). 6. T. gondii Outbreaks
Oocysts in feces, water and environment can be previously stained,
which helps to distinguish the parasites from the host cells. Giemsa/ 6.1. Meat products
Hematoxylin/Eosin (HE) staining is simple and economical and
commonly used for this purpose (Silva et al., 2010). Electron microscopy One of the major public health problems in the world is the food-
is also used to detect tissue cysts in the brain of mice and oocysts in the borne diseases, since they cause several infections in the population,
small intestine of infected cats, but it is difficult to apply for routine use with significant economic losses. Protozoa as T. gondii, which is trans­
(Sims et al., 1989). mitted by water and food, cause to severe clinical involvements. The
According to molecular diagnosis, the methodology used to detect consumption of raw or undercooked meat and its by-products, mainly,
T. gondii DNA in clinical, veterinary and environment samples is poly­ those from pigs and sheep, is one of the main risk factors for acquiring
merase chain reaction (PCR). This methodology is considered highly toxoplasmosis (Kijlstra and Jongert, 2009; Belluco et al., 2018; Tenter
specific and highly sensitive (Belaz et al., 2015). The test sensitivity can et al., 2000; Smith et al., 2021). The cyst number found in meat is
be influenced by different factors such as variation in tissue predilection generally slight (around one cyst/100g of meat). The cyst localization in
by T. gondii in host, heterogeneous distribution of bradyzoite cysts musculature and/or viscera may vary from host to host. Cysts in tissues
within tissues, tissue selection and tissue quantity (Hill et al., 2006). PCR from swine, sheep and goats are found more frequently (Hill and Dubey,
can be used in situations where sera are not available or antibody 2002; Djokic et al., 2016; Sroka et al., 2020). On the other hand, cysts
preservation is compromised, for example, in wildlife studies when the are less frequent in tissues of chickens, rabbits, horses and, rarely found
eliminated carcasses are the most readily available set of samples. In in tissues of infected cattle and buffaloes (Tenter et al., 2000; Tenter,
addition, PCR-based techniques can be expanded to allow for 2009).
sequencing and typing of strains, which can be important in detailed Toxoplasmosis control by “Sanitary Inspection Services” is unfeasi­
epidemiological studies. ble, since T. gondii cysts are microscopic and cannot be visualized with
In humans, PCR have allowed the sensitive detection of T. gondii DNA the naked eye. Therefore, the sanitary control of T. gondii is a challenge
in clinical specimens such amniotic fluid, aqueous humor, cerebrospinal for regulatory surveillance services and science (Kijlstra and Jongert,
fluid (CSF), bone marrow, and blood (Colombo et al., 2005, Contini 2009). The strategies adopted include prevention measures, as hygienic
et al., 2005; Edvinsson et al., 2008; Mattos et al., 2011; Camilo et al., and sanitary guidelines on care in the correct meat cooking and its
2017). PCR is appropriate for AIDS patients, since this method is not products as well as, the proper handling of cooking utensils (Germano
affected by the immunological status and has shown to be rapid, sensi­ and Germano, 2011; CDC, 2021). One third of the human population
tive and specific, avoiding invasive and expensive brain biopsy pro­ already had contact with T. gondii by ingestion of contaminated water or
cedures. As an alternative approach, PCR in CSF and peripheral blood food, mainly in places where sanitary conditions are precarious (Kijlstra
samples has also been used with several reported sensitivities (Pelloux and Jongert 2008; Almeria and Dubey, 2021; CDC, 2021).
et al., 1997; Priya et al., 2002; Contini et al. 2005; Colombo et al., 2005). Strategies to reduce the occurrence risk of tissue cysts in meat
The development of the Real-time PCR (qPCR) has revolutionized products are considered preventive measures in T. gondii transmission
molecular diagnosis by adding reliability and speed, representing that can be by monitoring of fresh meat or cyst inactivation by pro­
considerable progress (Robert-Gangneux et al., 2010). Many reports cessing the contaminated meat (Berger-Schoch et al., 2011). The inac­
tend to generalize the idea that qPCR in general has an improved tivation of T. gondii cysts in meat products can be carried out by heating
sensitivity compared with conventional PCR (Lin et. al., 2000; Mesquita homogeneously, at temperature above 67◦ C or by freezing at -13◦ C
et al., 2010; Camilo et al., 2017). (Sommer et al., 1965; Dubey et al., 1990; Kotula et al., 1991; Kijlstra and
Subsequently, varieties of PCR sensitivity and specificity depend also Jongert, 2009). Cured process in some meat products causes unviable
on factors including the standardization of reagents and protocols for cysts. However, they depend on the salt concentration and storage
DNA extraction, the inadequate storage of the clinical sample and the temperature. Experimentally, cysts were inactivated in 6% NaCl solu­
time elapsing between the start of specific therapy and collection (blood tion at 4◦ C to 20◦ C, but they were resistant for weeks in 0.85 to 3.3%
or CSF) which often affects PCR reproducibility and makes comparison NaCl solutions at the same temperatures (Dubey, 1998). The salting of
of results difficult. The use of a second reaction using parallel amplifi­ fresh-type homemade pork sausages does not inactivate cysts. The
cation of a marker that amplifies a human sequence as the β-globulin inactivation occurs only at high NaCl concentrations. However, the
gene in case of human samples guarantees the quality of the DNA consumption is unfeasible, mainly due to changes in organoleptic
extraction and PCR inhibitors, avoiding false results (Camilo et al., characteristics (Jamra et al., 1991; Navarro et al., 1992). Cultural habits
2017). are relevant in the prevalence of T. gondii infection (Kijlstra and Jongert,
Several DNA targets for PCR were evaluated and have been used 2009). Studies report that most infections in humans in the US are likely
regularly in the different laboratories to determine the diagnosis of the result of ingesting cysts contained in undercooked meat. In France,
congenital, ocular, disseminated or cerebral toxoplasmosis. Targets such the habit of eating raw or undercooked meat products also increases the
as bradyzoite genes encoding for specific antigens, B1 gene; P30, ribo­ infection prevalence. Around 84% of pregnant women present anti­
somal DNA genes and 529-bp sequence have been frequently used bodies anti-T. gondii (Hill et al., 2005; Rodrigues et al., 2018).
(Chabbert et al., 2004; Contini et al., 2005; Mesquita et al., 2010; Case-control studies developed in Europe have shown that meat is the
Camilo et al., 2017), or to detect and quantify parasite DNA load and its main risk factor for infection in pregnant women. Small toxoplasmosis

6
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

outbreaks have also been associated with raw meat consumption in 6.3. Water Consume
Korea, USA, France, French Guiana, and New Zealand (Choi et al., 1997;
Ross et al., 2001; Carme et al., 2002; Lake et al., 2020). It is important to Studies reporting the detection of T. gondii oocysts in water samples
emphasize the management meat from hunting animals, since epide­ are scarce when compared to those from Cryptosporidium spp. and
miological studies and reports of outbreaks have pointed to the man­ Giardia spp. Vieira et al. (2015) studied the vulnerability of groundwater
agement and consumption of raw or undercooked meat as a source of associated with toxoplasmosis prevalence in humans and discussed the
toxoplasmosis (Cook et al., 2000; Ross et al., 2001; Carme et al., 2002; importance of oocysts in aquatic environments. However, waterborne
Dubey and Jones, 2008; England et al, 2019; Gaulin et al., 2020). outbreaks have been described in different regions of the world (Franco,
In recent years, various parasitological and molecular methods have 2007; Jones and Dubey, 2010). An epidemiological study associated the
been developed to detect T. gondii cysts in meat from different animal high toxoplasmosis prevalence to oocysts in contaminated food, irriga­
species. These methodologies aimed to improve toxoplasmosis control tion water, fresh fruits and vegetables. The authors highlighted water as
and prevention measures (Tenter et al., 2000; Hill and Dubey, 2002). an efficient carrier source of T. gondii due to of its great dispersal ca­
Alternative approaches were developed to employ meat exudate as pacity, its wide use and for being of fundamental consumption for the
biological material for antibody detection in rabbit meat, fresh and maintenance of life (Hussain et al., 2017).
cured bovine meat (Mecca et al., 2011; Marciano et al., 2018). This The largest epidemic outbreak of human toxoplasmosis reported in
alternative approach was, also, tested in exudates from cattle, swine, the literature occurred in Santa Isabel do Ivaí city, Paraná state, Brazil
sheep and poultry retailed in Scotland. The authors obtained a good due to water transmission. Oocyst dissemination occurred in a water
positivity in swine and sheep exudates (Plaza et al., 2020). Schares et al. treatment plant (Moura et al., 2006). In October/2018, 2,000 cases were
(2018) compared the ELISA results of serum and meat exudate from notified in Santa Maria city, Rio Grande do Sul state, Brazil (Minuzzi
different bovine organs and muscles. They concluded that samples from et al., 2020; Minuzzi et al., 2020a). The main suspicion was water
the heart had high immunoglobulin levels. These alternative approaches transmission. Other studies have also linked toxoplasmosis outbreaks to
are interesting for use in quality control and safety of meat products, oocyst contamination in drinking water in the United States and Canada
since they constitute a promising tool for Public Health Laboratories, (Bowie et al., 1997; Dubey, 2004). These studies suggested the envi­
allowing the monitoring and elucidation of T. gondii outbreaks. ronmental contamination occur via oocysts, which are more restricted in
places with high feline concentration (Afonso et al., 2008). Less than
0.9% of cats actively excrete oocysts in feces and this may still be an
6.2. Milk and Dairy Products overestimation as it is difficult to distinguish T. gondii oocysts from
Hammondia hammondi oocysts by microscopic analysis (Schares et al.,
Normally, milk and its products are industrialized and pasteurized, 2008).
therefore, the ingestion of milk or dairy products is considered low to the Investigations of T. gondii oocysts in water bring a series of scientific
risk of toxoplasmosis transmission. Occasional reports related the and technological challenges, since internationally recognized meth­
occurrence of small outbreaks (Sacks et al., 1982; Jones et al., 2009; odologies are of high complexity, combined with difficulties in detection
Dubey and Jones, 2014; Dubey et al., 2014; Bezerra et al., 2015). Thus, and possible presence of interfering in the matrices. Although different
dairy products cannot be excluded as a potential source of transmission, studies have been carried out to investigate the infectivity of parasites in
since they may represent a risk to human health, especially when they soil and water, the viability of T. gondii oocysts in water is poorly re­
are manipulated by informal producers that use unpasteurized milk, in ported. Therefore, parasite viability is an important tool for the effective
the production of dairy products intended for consumption by children, monitoring of drinking water quality (Dubey, 1998; Dubey, 2009).
elderly and immunocompromised individuals (Riemann et al., 1975; Recently, Marciano et al., (2020) evaluated the viability of T. gondii
Skinner et al., 1990; Bezerra et al., 2015). This evidence was tested by oocysts by real-time PCR in samples treated or not with phorbol 12-myr­
Hiramoto et al. (2001). A sample of bovine milk previously contami­ istate 13-acetate (PMA), a DNA intercalant. In intact oocysts, the
nated with T. gondii (ME-49 strain) was used to prepare fresh cheese. intercalant did not bind to the DNA. In contrast, in non-viable oocysts,
Next, this cheese was used in bioassay to infect two mice, which an intercalation by PMA occurred in the DNA, allowing the differenti­
developed an expressive infection after inoculation. ation between them. The implementation of specific methodologies for
Studies related the presence of tachyzoites and cysts in milk from detecting this pathogen allows preventing or elucidating epidemic out­
sheep, goats, cattle and mice (Remington et al., 2004, Ragozo et al., breaks, as well as monitoring the quality of water available to the
2009; Camossi et al., 2011; Tavassoli et al., 2013). However, most of population.
them were related with small outbreaks and indicated that transmissions
were caused by ingestion of unpasteurized and raw goat milk (Riemann 6.4. Vegetables
et al., 1975; Chiari and Neves, 1984; Skinner et al., 1990; Dubey et al.,
2014; Silva, 2015). Recently, T. gondii DNA was detected in milk of 42 In the daily diet, vegetables are essential to gain vitamins and min­
seropositive sheep in Santa Catarina State, Brazil. These DNA samples erals. However, the consumption of raw vegetables, without proper
were sequenced and results showed around 97% of identity with the hygiene, can represent a health risk if they are contaminated by any
membrane antigen of T. gondii P22 gene. Besides the risk of human pathogen. Vegetable contamination can occur during irrigation, storage,
infection by consumption of milk and/or dairy products, the lactogenic transport and inadequate handling (Soares, 2006; Fernandes et al.,
transmission to lambs is possible (Ossani et al., 2017). 2015; Gois et al., 2018).The investigation of T. gondii oocysts in vege­
The importance of adopting measures for sanitary management of tables is appropriate, since they are widely consumed by the population.
herds was shown in a study that IgG antibodies or DNA were determined However, the oocysts removal is hampered by intrinsic conditions of the
in 15.78% in samples of raw milk from goats and sheep analyzed in parasite including adherence to surfaces and resistance to environment
northern Italy (Gazzonis et al., 2019). Equality, a study in Poland (Bekele et al., 2017).
showed that milk was associated as a potential risk factor for T. gondii Different T. gondii outbreaks related to consumption of raw vegeta­
transmission in humans. In Sudan a study showed a high seropositivity bles were reported in epidemiological studies (Ekman et al., 2012;
(67%) for T. gondii in camels. These data represent public health sig­ Hohweyer et al., 2016; Lalonde and Gajadhar, 2016). However, oocyst
nificance since nomads consume raw milk from camels (Elamin et al., isolation and detection in these matrices are still a challenge (Lass et al.,
1992). These studies show the importance of preventive measures, 2012). A toxoplasmosis outbreak in 2016, in Para state, Brazil reported
health education and development of new research are essential to curb 73 cases. The contamination was correlated with the consumption of
the high toxoplasmosis incidence via food. acai juice, sold in three sale points of the product. The epidemiological

7
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

data pointed to food origin and the probable failure of good practices in Several animal models have been suggested as a model for toxo­
production of acai juice (Morais et al., 2016). For transmission reduction plasmosis, but they have never been widely used. Most studies were
is essential the adoption of good practices, such as correct vegetable conducted with rodents, due to the nature of their placenta and also for
sanitizing before consumption in order to remove the oocysts. practical reasons, due to the ease of maintenance and handling in the
laboratory (Araujo and Remington, 1974; Dubey and Shen, 1991; Fux
7. Studies in Experimental Models et al., 2000; Freyre et al., 2001; Oz and Tobin, 2012). However, studies
involving a detailed anatomical analysis and assessment of fetal infec­
Toxoplasma gondii is a protozoan intensively studied and used as a tion often require larger animals such as rabbits, pigs, sheep or primates
model organism for the study of other clinically relevant human and (Schoondermark-van de Ven et al, 1994; Jungersen et al., 2001; Nau
veterinary parasites due to its ease of in vitro culture and genetic et al., 2017).
manipulation. An ideal animal model for congenital toxoplasmosis, for drug studies,
Experimental models have largely contributed to a better under­ as well as for immunological and vaccine studies would be one that
standing of the pathogenesis of toxoplasmosis in different experimental mimics the conditions of infection in the human host (Weiss and Kim,
approaches and applications, such as the pathophysiology of infection, 2020). To date, no animal model has been able to mimic all clinical
drug efficacy, treatment and prevention. However, these models do not symptoms and signs developed by humans in response to T. gondii
always reflect the natural dynamics of the infection, so it is important to infection (Munoz et al., 2011).
choose the most appropriate model for each scientific question.
Basically, all warm-blooded animals can be infected with T. gondii. 8. Genetic Characterization of T. gondii Strains
However, different animal species differ markedly in their resistance to
infection (Zenner et al., 1998; Gao et al., 2014; Dubey et al., 2016). The Extensive research regarding the genotyping of T. gondii in humans
outcome of infection depends not only on the animal species, but also on and in different animals worldwide has been done in the last years (Fux
the parasite strain (McLeod et al., 1984; Brown et al., 1995). Genetically, et al 2003). Investigation of the genotypes of T. gondii infection in ani­
the T. gondii strains are all relatively similar, differing only by about mals aims to ascertain the correlation between the specific variant and
1-2% at the nucleotide level (Fujii et al., 1983; Zenner et al., 1998). its biological properties and to epidemiologically track the relation of
Despite their genetic similarity, they induce strong phenotypic differ­ the particular isolate with diseases outcome, outbreaks and pathoge­
ences in the experimental models (Sibley et al., 2009). nicity (Dardé 2008; Xiao and Yolken, 2015).
T. gondii is capable of infecting most laboratory animals, although, Genotyping of T. gondii isolates from different geographical places
rat is partially resistant. The use of these animals has been useful for showed several T. gondii subpopulations. Howe and Sibley (1995) sug­
understanding the mechanisms of resistance and immunity induction gest that the genetic structure of T. gondii are from three widespread
(Benedetto et al, 1993; Darcy and Zener, 1993). clonal lineages named type I, II and III isolates from both wild and do­
Mice, rats, rabbits, pigs and non-human primates are commonly used mestic animals in diverse regions of North America has revealed greater
to study the efficacy of drugs against T. gondii infection. Mice have been genetic diversity than previously described and a fourth clonal lineage
used as a model to assess therapeutic assessment (Fux et al., 2000; Oz have been proposed (Khan et al., 2011). Yet, some isolates from animals
and Tobin, 2012; Dunay et al., 2018), pathogenic mechanisms and, more and humans are different from the known clonal lineage (I, II, III) and
recently, for vaccine approaches (Ismael et al., 2006). Among the therefore classified as atypical or exotic genotypes (Ajzenberg et al.,
experimental models, the most economical and widely used is the mouse 2004).
(Buxton, 1998; Que et al., 2004). Although murine models have many For better understanding of the epidemiological population of
advantages, extrapolation of results to other species must be done with T. gondii worldwide an integrated approach using PCR-RFLP of 10
care (Dunay et al., 2018). different genetic markers has been proposed by Su and collaborates in
Animal models of congenital toxoplasmosis were effectively used to 2009 (Su et al, 2012). The use of this integrated approach revealed
evaluate efficacy of treatment and vaccines and to simulate fetal damage several new major genotypes (Su et al., 2012). In order to standardize
and vertical transmission of human according to different doses and the nomenclature of T. gondii genotypes, each identified genotype is
time of pregnancies in a different dose and time of pregnancy (Var­ designated as a specific genotype number at ToxoDB–PCR-RFLP geno­
gas-Villavicencio, et al., 2016). types (http://ToxoDB.org) database (Gajriaet al, 2008). Nowadays at
In recent years, the development of new technology of staining least, 278 variations have been described using PCR-RFLP (Su and
(signaling) and genetic engineering of parasites has provided an addi­ Dubey, 2020). Geographical analyses of 1457 samples revealed 189
tional alternative for experimental models of infection permitting real- ToxoDB genotypes by the PCR-RFLP method with few genotypes present
time analyses of infection with reduction of the number of animals in the northern hemisphere, whereas hundreds of genotypes coexist in
(Avci et al, 2018; Ribeiro-Andrade et al., 2019). The use of transfected the southern hemisphere (Su and Dubey, 2020).
T. gondii parasites make it possible to evaluate progression of infection In North America, Europe, Asia, Africa, most isolates belong to the
and immune response to the parasite including the maintenance of clonal type I, II, and III lineages, and there is no host boundary for
T. gondii tissue cysts and tachyzoites (Subauste, 2012; Ribeiro-Andrade different genotypes (Sibley and Boothroyd, 1992; Howe and Sibley,
et al., 2019). 1995; Lindstrom et al., 2006; Zakimi et al., 2006). The three lineages are
Other animal models can be used, such as sheep, rats, guinea pigs, highly similar at DNA sequence level. As the differences were less than
hamsters or (Santos et al., 2016; Freyre et al., 2012). Rats and sheep are 1% on average, it was suggested that I, II and III lineages were expanded
widely used in studies that address the efficacy of drugs for T. gondii globally in the past 10,000 years (Su et al., 2003; Sibley and Ajioka,
(Dunay et al., 2018). Rats have placental development and hemochorial 2008).
placentation identical to humans (Fonseca et al., 2012). In sheep, Human toxoplasmosis has been most often associated with strains of
congenital toxoplasmosis is very similar to that which occurs in humans. a type II genotype (Howe and Sibley, 1995; Hosseini et al., 2019).
Thus, sheep are an appropriate animal model for the study of congenital Concerning virulence type I lineage is highly virulent compared to types
toxoplasmosis, not only because they share important aspects of human II and III (Dardé 2008; Robert-Gangneux and Dardé., 2012; Hosseini
infection, but also because they directly contribute to the study of new et al., 2019).
disease control measures in livestock, also severely affected by T. gondii. Studies on T. gondii strains in South America have shown a high
There are well-established models of toxoplasmosis in pregnant ewes genetic variability with distinct genotypes not identified in North
that provide a starting point for the preparation and testing of new America, Europe, Asia and Africa (Fux et al 2003, Dubey et al., 2005;
vaccines (Innes et al., 2019). Lehmann et al., 2006; Pena et al., 2008; Dubey et al., 2008). These

8
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

analyses were performed in isolates collected from clinical samples nm. The “microvesicles” vary in size between 100 to 1,000 nm. Micro­
(pregnant women with congenital infection, patients with ocular and vesicles are produced by external budding from plasma membrane
cerebral forms) (Ferreira et al., 2011; Bastos da Silva et al, 2016; Cunha (Delabranch et al., 2012, Lagana et al., 2013; Akers et al., 2013; Théry
et al., 2016) and a great variety of isolates from domestic animals. The et al., 2018). These nanoparticles carry proteins, RNA, DNA, and act as
investigations revealed more than 50 genotypes and they were consid­ messengers between cells. Patients and experimental mice developing
ered to be the common clonal lineages in Brazil (Ferreira et al 2018, Yai symptomatic toxoplasmosis release EVs in blood when compared with
et al., 2009; Pena et al, 2011; Cabral et al, 2013; Cañón-Franco et al., those without toxoplasmosis infection (Cruz et al., 2020; Maia et al.,
2013). However, various isolates from different Brazilian regions had 2021; Maia et al., 2021a). The subclass “exosomes” are the smallest EVs
atypical genotypes (Blaizot al., 2019). This genotype diversity suggests (size 30 to 100 nm). Exosomes do not originate from plasma membrane.
the constant emergence of new or atypical genotypes. In addition, These particles are derived from internal budding of vesicles in the
T. gondii strains in South America have a high rate of transmission and lumen of early endosome (Cocucci et al., 2009; Akers et al., 2013;
out-crossing (Ajzenberg et al., 2004; Lehmann, et al., 2006; Pereir­ Fleming et al., 2014; Kowal et al., 2014; Coakley et al., 2015). These
a-Chioccola et al., 2009). The genetic exchange could occur when the intracellular multivesicular bodies fuse with the plasma membrane and
definitive host ingests different types of parasites from their interme­ are released to the extracellular medium (Théry et al., 2009). As the
diate hosts at nearly the same time, or the intermediate host has mixed microvesicles, they also contain bioactive proteins, mRNA, micro-RNA
infection of different strains. T. gondii isolates in South America seem to (miRNA) and other small non-coding RNA species (Delabranch et al.,
be different in physiological characteristics since in some cases, are 2012; Coakley et al., 2015; Schwab et al., 2015; Tkach and Théry, 2016;
highly pathogenic in immunocompetent individuals (Ferreira et al., Théry et al., 2018).
2011; Bastos da Silva et al, 2016). Atypical genotypes have also been Protozoan parasites also release the three EVs subclasses that can be
associated with human outbreak toxoplasmosis as in Santa Maria, Brazil generated by endocytic pathways or are directly released from the
with 809 confirmed cases (Minuzzi et al., 2020; Paraboni et al., 2020; plasma membrane (Trocoli Torrecilhas et al, 2009; Torrecilhas et al.,
Costa et al., 2020). In North America atypical genotypes have been 2012; Marcilla et al., 2014; Montaner et al., 2014; Twu and Johnson,
frequently identified and associated with severe ocular and systemic 2014; Campos et al., 2015; Torrecilhas et al., 2020). These bioactive
disease in humans (Pomares et al., 2018). molecules have functions as host-parasite interaction, modulation of
Genotyping studies on T. gondii in wildlife showed extended genetic host immune system and inflammatory response via TLR2 (Nogueira
diversity and evidence for recombination present in wildlife samples et al., 2015). EVs released by protozoan parasites provide a mechanism
thus it has been suggested that atypical strains are not insignificant for inserting the parasite cargo into host cells, as virulence factors
anomalies in the population structure, but significantly demonstrate (Torrecilhas et al., 2012; Mantel and Marti, 2014).
gene pool generated by the vast host range utilized by this parasite The effect of EVs excreted by T. gondii in immune host response and
(Wendte et al., 2011). physiological environment are not totally established. However, the
In ruminants (goat, sheep and cattle), type II strains and atypical host-T gondii interactions certainly occur, also, via EVs. The first in­
genotypes were predominant, with a high diversity of genotypes present vestigations of EVs and T. gondii were reported in 2004 (Aline et al.,
in sheep (Sharif et al., 2017). High prevalence of atypical genotypes was 2004). The authors described the participation of exosomes excreted by
observed in isolates from lambs, chickens, beef and pigs destined for dendritic cells infected with T. gondii in protective immune response
human consumption (Dubey et al., 2020a; Santos Silva, et al., 2020; against T. gondii infection. Later, Kim et al. (2016) confirmed these re­
Frazão-Teixeira et al., 2011 Berger-Schoch et al., 2011a). sults reporting that treatment of exosomes isolated from
A study conducted in the Midwestern Brazil (Mato Grosso do Sul) T. gondii-infected cells attenuated the cell proliferation and altered the
described a high genetic diversity of T. gondii among isolates from do­ host cell cycle. The capacity of exosomes excreted by infected host cells
mestic animals, wildlife, and humans. Among wildlife, an ocelot, two proceeded as an effective non-cellular vaccine with adjuvant properties
crab-eating foxes, one Azara’s agouti and one roadside hawk were for immunization. The experiments were performed in an experimental
infected with BrIII strain, as well as, the environment where they model with mice and, during the pregnancy (Beauvillain et al., 2007;
circulate. This Brazilian clonal lineage was the most frequent among Beauvillain et al., 2009). These studies also demonstrated that exosomes
wild animals in this region. Other reports indicated diverse genotypes in secreted from infected cells with T. gondii alter the cell proliferation
Brazilian wildlife in various regions of the country (Yai et al., 2009; mechanisms causing changes in neighboring cells (Kim et al., 2016).
Pena et al., 2011; Cañón-Franco et al., 2013, Witter et al., 2020). Subsequent studies were correlated with the specific characteriza­
tion of EVs (microvesicles and exosomes) secreted/excreted by T. gondii.
9. Extracellular Vesicles and miRNA Studies using scanning (SEM) and transmission electron microscopy
(TEM) demonstrated the morphology of these EVs inside of tachyzoites
The interaction parasite-host modulating the immune system and and at the moment to be released by plasma membrane (Silva et al.,
precise mechanisms of delivery and functions of cargo excreted by 2018; Ramírez-Flores et al., 2019; Quiarim et al., 2021). In addition,
T. gondii are a little known and must be answered for higher control these studies showed that T. gondii derived EVs carry virulent factors.
toxoplasmosis, principally in outbreaks. The release of different T. gondii Virulent strains, as RH secreted high concentration of EVs, when
antigens is essential to modulate the host immune system. Many articles compared as those non-virulent strains (VEG and ME49) (Quiarim et al.,
about T. gondii host interactions have been great advances in the past 2021).
years. T. gondii EVs carry a high concentration of proteins. In the proteomic
In the last decades, different studies have focused on extracellular profile of exosomes includes a wide range of proteins as the secretory
vesicles (EVs) released from prokaryotic or eukaryotic cells (Schwab proteins used by tachyzoites for invasion and posterior replication
et al., 2015; Théry et al., 2018). EVs participate of cell-cell communi­ within host cells (Pope and Lasser, 2013; Wowk et al., 2017; Silva et al.,
cation for transfer of proteins, lipids, nucleic acids, biomarkers of dis­ 2018; Quiarim et al., 2021). These EV-derived proteins inducing the
eases, bio reactive macromolecules and others (Cocucci et al., 2009; production of IL-10, IL-12, IFN-α, TNF-α and iNOS in host cells (Pope
Akers et al., 2013; Schwab et al., 2015; Tkach and Théry, 2016; Théry and Lasser, 2013; Silva et al., 2018; Li et al., 2018; Maia et al, 2021,
et al., 2018). EVs comprise a variety of membrane-limited vesicles and Maia et al., 2021a; Quiarim et al., 2021). EVs purified of T. gondii used as
are released by cells. EVs are classified into three subclasses of particles vaccine in experimental mice were able to stimulate the protective im­
and based on their origin, markers and size. The subclass “apoptotic mune response. Immunized mice developed high levels of IgG1, IFN-γ,
bodies”, released as blebs from undergoing programmed death cells. IL-10, IL-17 and TNF-α. When they were challenged with RH strain
They represent the largest EVs with size ranging from 1,000 to 5,000 (virulent), they presented reduced parasitemia and increased survival of

9
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

mice (Maia et al, 2020). and miR-29c-3p. The unbalance of miRNAs production in toxoplasmosis
Studies on investigations of host-produced EVs collected from human is correlated with the unbalance of immune response as elevation of
and murine sera showed that the concentration and not size was able to TNF-α, IL-6 and down-regulation of IFN-γ in cerebral and ocular toxo­
distinguish infected and non-infected hosts. Serum-derived EVs from plasmosis forms (Pereira et al., 2020; Meira-Strejevitch et al., 2020;
patients with symptomatic toxoplasmosis (cerebral and gestational) had Cruz et al.; 2020).
more EVs when compared with those purified from non-infected in­
dividuals. Equality infected and immunized mice had more concentra­ 10. T gondii and Toxoplasmosis Studies: Future Perspectives
tion of EVs in sera than non-infected ones (Cruz et al., 2020, Maia et al.,
2021a). Sera from chronically infected humans or mice recognized The overall knowledge of T. gondii biology in it different aspects
distinct electrophoretic patterns in immunoblotting of EV proteins involving humans, animals and environment are essential for the control
produced by RH strain. However, these sera are poorly reactive to of toxoplasmosis worldwide. In the last few decades, significant research
EV-derived proteins of ME-49 and VEG strains (Silva et al., 2018; Maia efforts have led to the development of several studies with promising
et al, 2021; Maia et al., 2021a; Quiarim et al., 2021). Fig. 2 shows a results. Despite such knowledge as parasite-host interactions, other
scheme of the interaction T. gondii/host cells by EVs contribution. cellular and molecular mechanisms underlying to such interactions are
T. gondii EVs carry mRNA and small-RNAs, including microRNAs not completely understood. Thus, a significant level of academic-public-
(miRNAs) and these molecules participate in the modulation of host private partnership is important to support future studies.
immune response against T gondii infection (Silva et al., 2018). The In this review, the epidemiology, diagnostics, and control strategies
numerous small-RNAs participate in biological processes such as cell are addressed, with the objective of facilitating awareness of toxoplas­
differentiation, proliferation, apoptosis and post-transcriptional gene mosis and promoting trans disciplinary collaborations, an opportunity
regulation in cell development and pathological processes. Among for new partnerships focused on solutions to the complex problems.
small-RNAs, miRNAs are regulatory elements that can modulate gene According to the topics covered in this review, future research will
expression at the post-transcriptional level. These small single-strand likely focus in aspects emphasizing studies from the perspective of
RNA molecules, with lengths of 18–25 nt do not encode proteins. They toxoplasmosis associated with the one health concept. The knowledge of
act as negative regulators of the host response by the miRNA-induced T. gondii interaction with its hosts is critical for the development of
silencing complex (Bartel, 2004; Schorey et al., 2015). vaccines, drugs and other approaches. In addition the presence of
T. gondii has complete machinery for generation of small-RNA and its T. gondii oocysts in environment is a continue challenge to the efficient
gene regulation. The exosomal miRNAs produced by T. gondii control the control of toxoplasmosis. For these reasons, clinical and basic research
regulation of target genes related to cell proliferation. Probably, EVs about this infection and environment contamination should be main­
released by T. gondii transport miRNA to the host cells (Braun et al., tained and stimulated, particularly in low-and middle-income countries,
2010; Pope and Lasser, 2013; Kowal et al., 2014; Kim et al., 2016; Silva where the epidemic causes its major social and economic cost. In addi­
et al., 2018; Faria Junior et al, 2021). However, there is a fine-tuning tion, these studies could contribute to improvement in the clinical care
mechanism of miRNA biogenesis in distinct strains of T. gondii, since of patients to get better health and that will require a closer collabora­
each strain has different miRNA species (Wang et al., 2012; Li et al., tion between physicians and scientists.
2019; Li et al., 2020). miRNAs can be imbalanced in mice and humans Finally, the collaboration between professionals from different fields,
during T. gondii infection. Different studies related that T. gondii infec­ as well as, organizations and institutions from various countries would
tion modifies the levels of miRNAs in host cells by modulating the innate aid the understanding of the ecological interactions cooperating for a
and adaptive immune responses. clearer understanding the impacts of this zoonotic disease on animal and
Human macrophages infected with T. gondii trigger a cluster of human health, as well as, in environment actions.
miRNAs (miRNA-30c-1, miRNA-125b-2, miRNA-23b-27b-24-1 and
miRNA-17~92) that bind to the STAT3 promoter of macrophages, Author contribution statement
which cause an increased apoptosis of these cells. Mice recently infected
expressed three miRNAs (mmu-miR-712-3p, mmu-miR-511-5p and RAMB; ACT, VLP-C and BF conceived and designed the review
mmu-miR-217-5p) that were up-expressed in RH and ME49 strains (Cai manuscript. RAMB, ACT, MAMM, MLM, VLP-C and BF wrote, contrib­
et al., 2013; Cai et al., 2014). Infected and immunized mice expressed uted, and revised the manuscript. All authors approved the final version
the same miRNAs (miR-155-5p, miR-125b-5p and miR-29c-3p) than submitted and published.
expressed by T. gondii strains (RH, ME-49 and VEG) (Quiarim et al.,
2021; Maia et al., 2021a). Disclosures
Analyses of miRNA species in plasma collected of patients with
toxoplasmosis showed an up-expression of the miR-21-5p and miR- The authors have no other relevant affiliations or financial involve­
146a-5p in patients with co-infection cerebral toxoplasmosis/AIDS. ment with any organization or entire with a financial interest in or
Patients with ocular toxoplasmosis had up-expression of the miR-155-5p financial conflict with the subject matter or materials discussed in the

Fig. 2. EVs release from T. gondii and interac­


tion with host cells. Image captured by scan­
ning electron microscopy showing a tachyzoite
(magnification: 130,000x), and a schematic
figure releasing extracellular vesicles (EVs).
These nanomolecules carry virulent factors,
high concentration of parasite proteins used for
invasion and posterior replication within host
cells (A). Host cells releasing EVs after parasite
stimulus. The EVs-release cause an intense im­
mune response, with production of cytokines,
antibodies and miRNAs (B).

10
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

manuscript apart from those disclosed. No writing assistance was uti­ Arruda, IF, Millar, PR, Barbosa, ADS, Abboud, LCS, Dos Reis, IC, Moreira, ASDC,
Guimarães, MPP, Amendoeira, MRR, 2021. 2021. T. gondii in domiciled dogs and
lized in the production of this manuscript or financial relationships that
cats in urban areas of Brazil: risk factors and spatial distribution. Parasite 28, 56.
could be construed as a potential conflict of interest. https://doi.org/10.1051/parasite/2021049.
Bachand, N, Ravel, A, Leighton, P, Stephen, C, Ndao, M, Avard, E, Jenkins, E, 2019.
Serological and molecular detection of Toxoplasma gondii in terrestrial and marine
Support wildlife harvested for food in Nunavik. Canada. Parasit Vectors. 12 (1), 155. https://
doi.org/10.1186/s13071-019-3408-9, 3.
This review article was supported by grants from FAPESP (Fundação Balasundaram, MB, Andavar, R, Palaniswamy, M, Venkatapathy, N, 2010. Outbreak of
acquired ocular toxoplasmosis involving 248 patients. Arch Ophthalmol 128 (1),
de Amparo à Pesquisa do Estado de Sao Paulo, Brazil) 2018/04708-8, 28–32. https://doi.org/10.1001/archophthalmol.2009.354.
2019/15909-0, 2020/07870-4; CNPq (Conselho Nacional de Desenvol­ Ballash, GA, Dubey, JP, Kwok, OC, Shoben, AB, Robison, TL, Kraft, TJ, Dennis, PM.,
vimento Científico e Tecnológico) 302327/2018-5. RAMB was sup­ 2015. Seroprevalence of Toxoplasma gondii in White-Tailed Deer (Odocoileus
virginianus) and Free-Roaming Cats (Felis catus) Across a Suburban to Urban
ported by scholarship from FAPES (Fundação de Amparo à Pesquisa e Gradient in Northeastern Ohio. Ecohealth 2, 359–367. https://doi.org/10.1007/
Inovação do Espírito Santo) 13/2019 and PROCAP 2020; VLP-C and s10393-014-0975-2.
ACT, from CNPq, 302327/2018-5 and 302104/2019-4, respectively. Barroso, P, García-Bocanegra, I, Acevedo, P, Palencia, P, Carro, F, Jiménez-Ruiz, S,
Almería, S, Dubey, JP, Cano-Terriza, D, Vicente, J., 2020. Long-Term Determinants
of the Seroprevalence of Toxoplasma gondii in a Wild Ungulate Community. Animals
(Basel) 10 (12), 2349. https://doi.org/10.3390/ani10122349.
Declaration of Competing Interest Bartel, DP., 2004. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 23
(2), 281–297. https://doi.org/10.1016/s0092-8674(04)00045-5, 116.
Bártová, E, Sedlák, K, Literák, I., 2009. Toxoplasma gondii and Neospora caninum
The authors have no other affiliations or financial involvement with antibodies in sheep in the Czech Republic. Vet Parasitol 6 (1-2), 131–132. https://
any organization or entire with a financial interest in or financial con­ doi.org/10.1016/j.vetpar.2008.12.022, 161.
flict with the subject matter or materials discussed in the manuscript Bastos da Silva, I, Batista, TP, Martines, RB, Kanamura, CT, Ferreira, IM, Vidal, JE,
Pereira-Chioccola, VL., 2016. Genotyping of Toxoplasma gondii: DNA extraction from
apart from those disclosed. No writing assistance was utilized in the formalin-fixed paraffin-embedded autopsy tissues from AIDS patients who died by
production of this manuscript or financial relationships that could be severe disseminated toxoplasmosis. Exp Parasitol 165, 16–21. https://doi.org/
construed as a potential conflict of interest. 10.1016/j.exppara.2016.03.004.
Beauvillain, C, Juste, MO, Dion, S, Pierre, J, Dimier-Poisson, I, 2009. Exosomes are an
effective vaccine against congenital toxoplasmosis in mice. Vaccine 10 (11),
References 1750–1757. https://doi.org/10.1016/j.vaccine.2009.01.022, 27.
Beauvillain, C, Ruiz, S, Guiton, R, Bout, D, Dimier-Poisson, I., 2007. A vaccine based on
exosomes secreted by a dendritic cell line confers protection against T. gondii
Afonso, E, Lemoine, M, Poulle, ML, et al., 2008. Spatial distribution of soil contamination
infection in syngeneic and allogeneic mice. Microbes Infect 9 (14-15), 1614–1622.
by Toxoplasma gondii in relation to cat defecation behavior in an urban area. Int J
https://doi.org/10.1016/j.micinf.2007.07.002.
Parasitol 38, 1017–1023. https://doi.org/10.1016/j.ijpara.2008.01.004.
Bekele, F, Tefera, T, Biresaw, G, Yohannes, T., 2017. Parasitic contamination of raw
Aguirre, A, Wilcox, BA., 2008 Sep. EcoHealth: Envisioning and creating a truly global
vegetables and fruits collected from selected local markets in Arba Minch town,
transdiscipline. Ecohealth 5 (3), 238–239. https://doi.org/10.1007/s10393-008-
Southern Ethiopia. Infect Dis Poverty 6 (1), 19.
0197-6.
Belaz, S, Gangneux, JP, Dupretz, P, Guiguen, C, Robert-Gangneux, F., 2015. A 10-year
Aguirre, AA, Longcore, T, Barbieri, M., et al., 2019. The One Health Approach to
retrospective comparison of two target sequences, REP-529 and B1, for Toxoplasma
Toxoplasmosis: Epidemiology, Control, and Prevention Strategies. EcoHealth 16 (2),
gondii detection by quantitative PCR. J Clin Microbiol 53 (4), 1294–1300. https://
378–390. https://doi.org/10.1007/s10393-019-01405-7.
doi.org/10.1128/JCM.02900-14.
Ajzenberg, D, Bañuls, AL, Su, C, Dumètre, A, Demar, M, Carme, B, Dardé, ML, 2004.
Belluco, S, Patuzzi, I, Ricci, A, 2018. Bovine meat versus pork in Toxoplasma gondii
Genetic diversity, clonality and sexuality in Toxoplasma gondii. Int J Parasitol 34
transmission in Italy: A quantitative risk assessment model. International Journal of
(10), 1185–1196. https://doi.org/10.1016/j.ijpara.2004.06.007.
Food Microbiology 269, 1–11.
Akers, JC, Gonda, D, Kim, R, Carter, BS, Chen, CC, 2013. Biogenesis of extracellular
Benedetto, A, Garbuglia, AR, Di Caro, A, Lo Presti, E, Alfani, E, Delfini, C, 1993. Virus-
vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies.
free survival and down-regulation of CD4 in C8166 cells infected with human
J Neurooncol 113 (1), 1–11. https://doi.org/10.1007/s11060-013-1084-8.
immunodeficiency virus type 1 at low density. J Gen Virol 74 (Pt 12), 2595–2601.
Al-Adhami, B.H., Simard, M., Hernández-Ortiz, A., Boireau, C., Gajadhar, A.A, 2016.
https://doi.org/10.1099/0022-1317-74-12-2595.
Development and evaluation of a modified agglutination test for diagnosis of
Berger-Schoch, AE, Bernet, D, Doherr, MG, Gottstein, B, Frey, CF., 2011. Toxoplasma
Toxoplasma infection using tachyzoites cultivated in cell culture. Food and
gondii in Switzerland: A Serosurvey Based on meat juice analysis of slaughtered pigs,
Waterborne Parasitology 2, 15–21. https://doi.org/10.1016/j.faw0par.2015.12.001.
wild boar, sheep and cattle. Zoonoses Public Health 58, 472–478. https://doi.org/
Aline, F, Bout, D, Amigorena, S, Roingeard, P, Dimier-Poisson, I, 2004. Toxoplasma gondii
10.1111/j.1863-2378.2011.01395.x.
antigen-pulsed-dendritic cell-derived exosomes induce a protective immune
Berger-Schoch, AE, Herrmann, DC, Schares, G, et al., 2011a. Prevalence and genotypes of
response against T. gondii infection. Infect Immun 72 (7), 4127–4137. https://doi.
Toxoplasma gondii in feline faeces (oocysts) and meat from sheep, cattle and pigs in
org/10.1128/IAI.72.7.4127-4137.2004.
Switzerland. Vet Parasitol 177 (3-4), 290–297. https://doi.org/10.1016/j.
Almería, S, Cabezón, O, Paniagua, J0, Cano-Terriza, D, Jiménez-Ruiz, S, Arenas-
vetpar.2010.11.046.
Montes, A, Dubey, JP, García-Bocanegra, I., 2018. Toxoplasma gondii in sympatric
Bezerra, M.J.G., KIM, P.C.P., Moraes, É.P.B.X., et al., 2015. Detection of Toxoplasma
domestic and wild ungulates in the Mediterranean ecosystem. Parasitol Res 117 (3),
gondii in the milk of naturally infected goats in the Northeast of Brazil. Transbound.
665–671. https://doi.org/10.1007/s00436-017-5705-6.
Emerg. Dis. 62, 421–424.
Almeria, S, Dubey, JP, 2021 Mar. Foodborne transmission of Toxoplasma gondii
Blaizot, R, Nabet, C, Blanchet, D, et al., 2019. Pediatric Amazonian Toxoplasmosis
infection in the last decade. An overview. Res Vet Sci 135, 371–385. https://doi.org/
Caused by Atypical Strains in French Guiana, 2002-2017. Pediatr Infect Dis J 2 (3),
10.1016/j.rvsc.2020.10.019. Epub 2020 Oct 24. PMID: 33148402.
e39–e42. https://doi.org/10.1097/INF.0000000000002130, 38.
Almería, S., Murata, F., Cerqueira-Cézar, C.K., Kwok, O., Shipley, A., Dubey, J.P, 2021.
Bogitsh, Burton J., Carter, Clint E, Oeltmann, Thomas N., 2018. Human parasitology.
Epidemiological and Public Health significance of Toxoplasma gondii infection in
Academic Press. https://doi.org/10.1016/C2010-0-65681-1.
wild rabbits and hares: 2010-2020. Microorganisms 9 (3), 597. https://doi.org/
Boothroyd, J.C., 2009. Toxoplasma gondii: 25 years and 25 major advances for the field.
10.3390/microorganisms9030597.
Int J Parasitol 39 (8), 935–946. https://doi.org/10.1016/j.ijpara.2009.02.003.
Ammar, S., Wood, L., Su, C., Spriggs, M., Brown, J., Van Why, K., Gerhold, R., 2021.
Bowie, WR, King, AS, Werker, DH, Isaac Renton, JL, Bell, A, Eng, SB, et al., 1997.
Toxoplasma gondii prevalence in carnivorous wild birds in the eastern United States.
Outbreak of toxoplasmosis associated with municipal drinking water. Lancet 350,
International journal for parasitology. Parasites and wildlife 15, 153–157. https://
173–177.
doi.org/10.1016/j.ijppaw.2021.04.010.
Braun, L, Cannella, D, Ortet, P, Barakat, M, Sautel, CF, Kieffer, S, Garin, J, Bastien, O,
Anderson, ML, Barr, BC, Conrad, PA., 1994. Protozoal causes of reproductive failure in
Voinnet, O, Hakimi, MA., 2010. A complex small RNA repertoire is generated by a
domestic ruminants. Vet Clin North Am Food Anim Pract 10 (3), 439–461. https://
plant/fungal-like machinery and effected by a metazoan-like Argonaute in the
doi.org/10.1016/s0749-0720(15)30531-4.
single-cell human parasite Toxoplasma gondii. PLoS Pathog 27 (5), e1000920.
Araujo, FG, Remington, JS., 1974. Effect of clindamycin on acute and chronic
https://doi.org/10.1371/journal.ppat.1000920, 6.
toxoplasmosis in mice. Antimicrob Agents Chemother 5 (6), 647–651. https://doi.
Brown, CR, Hunter, CA, Estes, RG, Beckmann, E, Forman, J, David, C, Remington, JS,
org/10.1128/AAC.5.6.647.
McLeod, R, 1995. Definitive identification of a gene that confers resistance against
Arevalo, JF, Belfort Jr, R, Muccioli, C, Espinoza, JV., 2010. Ocular toxoplasmosis in the
Toxoplasma cyst burden and encephalitis. Immunology 419–428, 85.
developing world. Int Ophthalmol Clin. Spring 50 (2), 57–69. https://doi.org/
Buxton, D., 1998. Protozoan infections Toxoplasma gondii, Neospora caninum and
10.1097/IIO.0b013e3181d26bf4.
Sarcocystis spp. in sheep and goats: recent advances. Vet Res 29 (3-4), 289–310.
Arkush, KD, Miller, MA, Leutenegger, CM, Gardner, IA, Packham, AE, Heckeroth, AR,
https://doi.org/10.1128/IAI.72.5.2915-2921.2004. PMID: 9689743.
Tenter, AM, Barr, BC, Conrad, PA., 2003. Molecular and bioassay-based detection of
Cabral, AD, Gama, AR, Sodré, MM, Savani, ES, Galvão-Dias, MA, Jordão, LR, 2013. First
Toxoplasma gondii oocyst uptake by mussels (Mytilus galloprovincialis). Int J
isolation and genotyping of Toxoplasma gondii from bats (Mammalia: Chiroptera).
Parasitol 15 (10), 1087–1097. https://doi.org/10.1016/s0020-7519(03)00181-4,
Vet Parasitol 100–104. https://doi.org/10.1016/j.vetpar.2012.11.015, 193.
33.

11
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

Cai, Y, Chen, H, Mo, X, Tang, Y, Xu, X, Zhang, A, et al., 2014. Toxoplasma gondii inhibits Toxoplasmosis. BMJ 15 (7254), 142–147. https://doi.org/10.1136/
apoptosis via a novel STAT3-miR-17-92-Bim pathway in macrophages. Cell Signal bmj.321.7254.142, 321.
26, 1204–1212. https://doi.org/10.1016/j.cellsig.2014.02.013. Costa, DF, Madeira, RP, Torrecilhas, AC, Nascimento, H, Ribeiro, KS, Paraboni, MLR,
Cai, Y, He, C, Lei, J, You, Y, Shen, J., 2013. STAT3-dependent transactivation of miRNA Rizzo, LV, Silveira, C, Burnier, MN, Lowen, MS, Garcia, JL, Gava, R, Pereira-
genes following Toxoplasma gondii infection in macrophage. Parasit Vectors 6, 356. Chioccola, VL, Commodaro, AG, Belfort, R, 2020. Ocular Disease in Mice Inoculated
https://doi.org/10.1186/1756-3305-6-356. with Pork Heart Samples Infected with Toxoplasma gondii. Ocul Immunol Inflamm
Cakir-Koc, R., 2016. Production of anti-SAG1 IgY antibody against Toxoplasma gondii 25, 1–7. https://doi.org/10.1080/09273948.2020.1807024.
parasites and evaluation of antibody activity by ELISA method. Parasitology research Costa, GB, De Oliveira, MC, Gadelha, SR, Albuquerque, GR, Teixeira, M, Raiol, MRDS,
115 (8), 2947–2952. https://doi.org/10.1007/s00436-016-5047-9. Sousa, SMB, Marin, LJ., 2018. Infectious diseases during pregnancy in Brazil:
Caldeira, FHB, Ubiali, DG, de Godoy, I, Dutra, V, de Aguiar, DM, Melo, ALT, Riet- seroprevalence and risk factors. J Infect Dev Ctries 31 (8), 657–665. https://doi.org/
Correa, F, Colodel, EM, Pescador, CA., 2011. Outbreak of caprine abortion by 10.3855/jidc.9492, 12.
Toxoplasma gondii in Midwest Brazil. Pesqui. Vet. Bras. 31, 933–937. https://doi. Costa, JM, Bretagne, S., 2012. Variation of B1 gene and AF146527 repeat element copy
org/10.1590/S0100-736X2011001100001. numbers according to Toxoplasma gondii strains assessed using real-time quantitative
Camilo, LM, Pereira-Chioccola, VL, Gava, R, Meira-Strejevitch, CDS, et al., 2017. PCR. J Clin Microbiol 50, 1452–1454.
Molecular diagnosis of symptomatic toxoplasmosis: a 9-year retrospective and Costa Viegas de Lima, D, de Melo, RPB, Campos de Almeida, J, Rodrigues Magalhães, FJ,
prospective study in a referral laboratory in São Paulo. Brazil. Braz J Infect Dis. 21 Ribeiro Andrade, M, de Morais Pedrosa, C, Vasconcelos de Oliveira, CA, Nascimento
(6), 638–647. https://doi.org/10.1016/j.bjid.2017.07.003. Porto, WJ, Su, C, 2019 Dec. Aparecido Mota R. Toxoplasma gondii in invasive animals
Camossi, LG, Greca-Júnior, H, Corrêa, AP, Richini-Pereira, VB, Silva, RC, Da Silva, AV, on the Island of Fernando de Noronha in Brazil: Molecular characterization and
Langoni, H, 2011. Detection of Toxoplasma gondii DNA in the milk of naturally mouse virulence studies of new genotypes. Comp Immunol Microbiol Infect Dis 67,
infected ewes. Vet Parasitol 11 (3-4), 256–261. https://doi.org/10.1016/j. 101347. https://doi.org/10.1016/j.cimid.2019.101347. Epub 2019 Sep 10. PMID:
vetpar.2010.12.007, 177. 31546127.
Campos, J.H., Soares, R.P., Ribeiro, K., Andrade, A.C., Batista, W.L., Torrecilhas, A.C, Costa-Silva, S, Sacristán, C, Gonzales-Viera, O, Díaz-Delgado, J, Sánchez-Sarmiento, AM,
2015. Extracellular Vesicles: Role in Inflammatory Responses and Potential Uses in Marigo, J, Groch, KR, Carvalho, VL, Ewbank, AC, Colosio, AC, Marcondes, MCC,
Vaccination in Cancer and Infectious Diseases. Journal of immunology research Meirelles, ACO, Bertozzi, CP, Lailson-Brito, J, Azevedo, AF, Ruoppolo, V, Oliveira, L,
832057. https://doi.org/10.1155/2015/832057. Ott, PH, Catão-Dias, JL., 2019 Aug 12. Toxoplasma gondii in cetaceans of Brazil: a
Cañón-Franco, WA, Araújo, FA, López-Orozco, N, et al., 2013. Toxoplasma gondii in free- histopathological and immunohistochemical survey. Rev Bras Parasitol Vet 28 (3),
ranging wild small felids from Brazil: molecular detection and genotypic 395–402. https://doi.org/10.1590/S1984-29612019051. PMID: 31411314.
characterization. Vet Parasitol 8 (3-4), 462–469. https://doi.org/10.1016/j. Crozier, G, Schulte-Hostedde, AI., 2014. The ethical dimensions of wildlife disease
vetpar.2013.07.019, 197. management in an evolutionary context. Evol Appl 7 (7), 788–798. https://doi.org/
Cano-Terriza D, Almería S, Caballero-Gómez, J., Díaz-Cao, J. M., Jiménez-Ruiz, S., 10.1111/eva.12171.
Dubey JP, García-Bocanegra I. 2019. Serological survey of Toxoplasma gondii in Cruz, AB, Maia, MM, Pereira, IS, Taniwaki, NN, Namiyama, GM, Telles, JPM, Vidal, JE,
captive nonhuman primates in zoos in Spain. Comparative immunology, Spegiorin, LCJF, Brandão de Mattos, CC, Mattos, LC, Meira-Strejevitch, CDS, Pereira-
microbiology and infectious diseases, 65, 54–57. 10.1016/j.cimid.2019.04.002. Chioccola, VL., 2020. Human extracellular vesicles and correlation with two clinical
Carme, B, Bissuel, F, Ajzenberg, D, Bouyne, R, Aznar, C, et al., 2002. Severe acquired forms of toxoplasmosis. PLoS One 3 (3), e0229602. https://doi.org/10.1371/
toxoplasmosis in immunocompetent adult patients in French Guiana. J Clin journal.pone, 15.
Microbiol 40 (11), 4037–4044. https://doi.org/10.1128/JCM.40.11.4037- Cunha, MM, Carneiro, AC, Costa, JG, Vitor, RW, 2016. Genotyping of Toxoplasma gondii
4044.2002. Directly from Human and Animal Biological Samples: From Partial Genotypes to a
Carme, B., Demar, M., Ajzenberg, D., Dardé, M.L., 2009. Severe acquired toxoplasmosis New Genotype. J Parasitol 102 (1), 157–160. https://doi.org/10.1645/15-813. Epub
caused by wild cycle of Toxoplasma gondii, French Guiana. Emerging infectious 2015 Sep 29. PMID: 26418172.
diseases 15 (4), 656–658. https://doi.org/10.3201/eid1504.081306. Cunningham, AA, Buxton, D, Thomson, KM., 1992. An epidemic of toxoplasmosis in a
Catão-Dias, JL, Epiphanio, S, Kierulff, MCM, 2013. Neotropical Primates and Their captive colony of squirrel monkeys (Saimiri sciureus). J Comp Pathol 107 (2),
Susceptibility to Toxoplasma gondii: New Insights for an Old Problem. In: 207–219. https://doi.org/10.1016/0021-9975(92)90037-u.
Brinkworth, J., Pechenkina, K. (Eds.), Primates, Pathogens, and Evolution. Darcy, F, Zenner, L, 1993. Experimental models of toxoplasmosis. Res Immunol 144 (1),
Developments in Prima0tology: Progress and Prospects. Springer, New York, NY. 16–23. https://doi.org/10.1016/s0923-2494(05)80091-4.
https://doi.org/10.1007/978-1-4614-7181-3_9. Dard, C, Fricker-Hidalgo, H, Brenier-Pinchart, MP, Pelloux, H, 2016. Relevance of and
CDC-Centers for Disease Control and Prevention USA, 2021. Human Services. Parasites- New Developments in Serology for Toxoplasmosis. Trends in parasitology 32 (6),
Toxoplasmosis (Toxoplasma infection). https://www.cdc.gov/parasites/ 492–506. https://doi.org/10.1016/j.pt.2016.04.001.
toxoplasmosis/index.html. and Parasitic Infections in the United States. https:// Dardé, ML., 2008. Toxoplasma gondii, "new" genotypes and virulence. Parasite (Paris,
www.cdc.gov/parasites/npi/index.html (accessed 12 December 2021. France) 15 (3), 366–371. https://doi.org/10.1051/parasite/2008153366.
Chabbert, E, Lachaud, L, Crobu, L, et al., 2004. Comparison of two widely used PCR Darwich, L., Cabezón, O., Echeverria, I., Pabón, M., Marco, I., Molina-López, R., Alarcia-
primer systems for detection of Toxoplasma in amniotic fluid, blood and tissues. Alejos, O., López-Gatius, F., Lavín, S., Almería, S., 2012. Presence of Toxoplasma
J. Clin. Microbiol. 42, 1719–1722. gondii and Neospora caninum DNA in the brain of wild birds. Veterinary Parasitology
Chiari CDA, Neves DP. 1984. Toxoplasmose humana adquirida através da ingestão de 183 (3-4), 377–381. https://doi.org/10.1016/j.vetpar.2011.07.024.
leite de cabra. Memórias do Instituto Oswaldo Cruz, 79, 337-340.Disponível em: < Delabranch, X, Berger, A, Boisramé-Helms, J, Meziani, F., 2012. Microparticles and
10.1590/S0074-02761984000300007 >. infectious diseases. Med Mal Infect 42 (8), 335–343. https://doi.org/10.1016/j.
Chikweto, A, Kumthekar, S, Tiwari, K, Nyack, B, Deokar, MS, Stratton, G, medmal.2012.05.011.
Macpherson, CN, Sharma, RN, Dubey, JP., 2011. Seroprevalence of Toxoplasma Desmonts, G, Remington, JS., 1980. Direct agglutination test for diagnosis of Toxoplasma
gondii in pigs, sheep, goats, and cattle from Grenada and Carriacou, West Indies. infection: method for increasing sensitivity and specificity. Journal of clinical
J Parasitol 97 (5), 950–951. https://doi.org/10.1645/GE-2811.1. microbiology 11 (6), 562–568. https://doi.org/10.1128/jcm.11.6.562-568.1980.
Choi, WY, Nam, HW, Kwak, NH, Huh, W, Kim, YR, Kang, MW, Cho, SY, Dubey, JP., 1997. Djokic, V, Fablet, C, Blaga, R, et al., 2016. Factors associated with Toxoplasma gondii
Foodborne outbreaks of human toxoplasmosis. J Infect Dis 175 (5), 1280–1282. infection in confined farrow-to-finish pig herds in western France: an exploratory
https://doi.org/10.1086/593702. study in 60 herds. Parasites Vectors 9 466. https://doi.org/10.1186/s13071-016-
Chomel, BB, Zarnke, RL, Kasten, RW, Kass, PH, Mendes, E., 1995. Serologic survey of 1753-5.
Toxoplasma gondii in grizzly bears (Ursus arctos) and black bears (Ursus americanus), Du, F, Zhang, Q, Yu, Q, Hu, M, Zhou, Y, Zhao, J., 2012. Soil contamination of Toxoplasma
from Alaska, 1988 to 1991. J Wildl Dis 31 (4), 472–479. https://doi.org/10.7589/ gondii oocysts in pig farms in central China. Vet Parasitol 8 (1-2), 53–56. https://doi.
0090-3558-31.4.472. org/10.1016/j.vetpar.2011.12.036, 187.
Coakley, G, Maizels, RM, Buck, AH., 2015. Exosomes and other extracellular vesicles: Dubey, JP, Beattie, CP., 1988. Toxoplasmosis of animals and man. CRC Press.
The new communicators in parasite infections. Trends Parasitol 31, 477–489. Dubey, JP, Ferreira, LR, Alsaad, M, Verma, SK, DA, Alves, Holland, GN, McConkey, GA.,
https://doi.org/10.1016/j.pt.2015.06.009. 2016. Experimental Toxoplasmosis in Rats Induced Orally with Eleven Strains of
Cocucci, E, Racchetti, G, Meldolesi, J., 2009. Shedding microvesicles: artefacts no more. Toxoplasma gondii of Seven Genotypes: Tissue Tropism, Tissue Cyst Size, Neural
Trends Cell Biol 19 (2), 43–51. https://doi.org/10.1016/j.tcb.2008.11.003. Lesions, Tissue Cyst Rupture without Reactivation, and Ocular Lesions. PLoS One 26
Colombo, FA, Vidal, JE, Penalva de Oliveira, AC, et al., 2005. Diagnosis of cerebral (5), e0156255. https://doi.org/10.1371/journal.pone.0156255, 11.
toxoplasmosis in AIDS patients in Brazil: importance of molecular and Dubey, JP, Gomez-Marin, JE, Bedoya, A, et al., 2005. Genetic and biologic characteristics
immunological methods using peripheral blood samples. J. Clin. Microbiol. 43, of Toxoplasma gondii isolates in chickens from Colombia. South America. Vet.
5044–5047. Parasitol. 134, 67–72.
Commodaro AG, Belfort RN, Rizzo LV, Muccioli C, Silveira C, Burnier Jr MN, Belfort Jr. Dubey, JP, Jones, JL., 2008. Toxoplasma gondii infection in humans and animals in the
2009. Ocular toxoplasmosis: an update and review of the literature. Memórias do United States. Int J Parasitol 38 (11), 1257–1278. https://doi.org/10.1016/j.
Instituto Oswaldo Cruz. 10.1590/S0074-02762009000200030 104:345-350. ijpara.2008.03.007.
Contini, C, Seraceni, S, Cultrera, R, et al., 2005. Evaluation of a Real-time PCR-based Dubey, JP, Jones, JL., 2014. Comments on “detection of Toxoplasma gondii in raw
assay using the light cycler system for detection of Toxoplasma gondii bradyzoite caprine, ovine, buffalo, bovine, and camel milk using cell cultivation, cat bioassay,
genes in blood specimens from patients with toxoplasmic retinochoroiditis. Int J capture ELISA, and PCR methods in Iran”. Foodborne pathogens and disease 11 (6),
Parasitol; 35, 275–283. 500–501.
Cook, AJ, Gilbert, RE, Buffolano, W, Zufferey, J, Petersen, E, Jenum, PA, Foulon, W, Dubey, JP, Kotula, AW, Sharar, A, Andrews, CD, Lindsay, DS., 1990. Effect of high
Semprini, AE, Dunn, DT., 2000. Sources of toxoplasma infection in pregnant women: temperature on infectivity of Toxoplasma gondii tissue cysts in pork. J Parasitol 76
European multicentre case-control study. European Research Network on Congenital (2), 201–204. PMID: 2319420.

12
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

Dubey, JP, Lago, EG, Gennari, SM, Su, C, Jones, JL, 2012. Toxoplasmosis in humans and from human patients in Sao Paulo State, Brazil identified distinct genotypes. Exp
animals in Brazil: high prevalence, high burden of disease, and epidemiology. Parasitol 129 (2), 190–195. https://doi.org/10.1016/j.exppara.2011.06.002.
Parasitology 139 (11), 1375–1424. https://doi.org/10.1017/S0031182012000765. Ferreira, TCR, Buery, JC, Moreira, NIB, Santos, CB, Costa, JGL, Pinto, LV,
Dubey, JP, Murata, FH, Cerqueira-Cézar, CK, Kwok, O, Villena, I., 2021. Congenital Baraviera, RCA, Vitor, RWA, Fux, B., 2018. Toxoplasma gondii: isolation, biological
toxoplasmosis in humans: an update of worldwide rate of congenital infections. and molecular characterization of samples from free-range Gallus gallus domesticus
Parasitology. https://doi.org/10.1017/S0031182021001013. from countryside Southeast Brazil. Rev Bras Parasitol Vet 27 (3), 384–389. https://
Dubey, JP, Murata, FH, Cerqueira-Cézar, CK, Kwok, OC, Yang, Y, Su, C., 2020. doi.org/10.1590/s1984-296120180028.
Toxoplasma gondii infections in dogs. 2009-2020. Veterinary Parasitology 109223. Filice, G., Meroni, V., Carnevale, G., Olliaro, P., Carosi, G., 1983. Comparison of ELISA
https://doi.org/10.1016/j.vetpar.2020a.109223. and indirect immunofluorescence in the detection of IgG and IgM antitoxoplasma
Dubey, JP, Murata, FHA, Cerqueira-Cézar, CK, Kwok, OCH, Su, C., 2020a. Economic and antibodies. Bollettino dell’Istituto sieroterapico milanese 62 (5), 445–450.
public health importance of Toxoplasma gondii infections in sheep: 2009-2020. Vet Flegr, J, Prandota, J, Sovičková, M., Israili, Z.H, 2014. Toxoplasmosis–a global threat.
Parasitol 286, 109195. https://doi.org/10.1016/j.vetpar.2020.109195. Correlation of latent toxoplasmosis with specific disease burden in a set of 88
Dubey, JP, Murata, F, Cerqueira-Cézar, CK, Kwok, O, Su, C., 2021. Epidemiologic countries. PloS One. https://doi.org/10.1371/journal.pone.0090203.
significance of Toxoplasma gondii infections in turkeys, ducks, ratites and other wild Fleming, A, Sampey, G, Chung, M., et al., 2014. The carrying pigeons of the cell:
birds: 2009-2020. Parasitology 148 (1), 1–30. https://doi.org/10.1017/ exosomes and their role in infectious diseases caused by human pathogens. Pathog
S0031182020001961. Dis. 71, 109–120. https://doi.org/10.1111/2049-632X.12135.
Dubey, JP, Shen, SK., 1991. Rat model of congenital toxoplasmosis. Infect Immun 59 (9), Flores, CA, Jimenez, J, Gomez-Puerta, LA, Palacios, C, O’Neal, SE, Muro, C,
3301–3302. https://doi.org/10.1128/iai.59.9.3301-3302.1991. Gonzalez, AE, Gilman, RH, Calderón, M, 2021. Seroprevalence of Toxoplasma gondii
Dubey, JP, Thulliez, P, Powell, EC, 1995. Toxoplasma gondii in Iowa sows: comparison of in free-range pigs in northern Peru. Vet Parasitol Reg Stud Reports 23, 100533.
antibody titers to isolation of T. gondii by bioassays in mice and cats. Journal of https://doi.org/10.1016/j.vprsr.2021.100533.
Parasitology 81, 48–53. Fonseca, AL, Silva, R.A., Fux, B., Madureira, A.P., Sousa, F.F.D., Margonari, C., 2012.
Dubey, JP, Velmurugan, GV, Chockalingan, A, et al., 2008. Genetic diversity of Epidemiologic aspects of toxoplasmosis and evaluation of its seroprevalence in
Toxoplasma gondii isolates from chickens from Brazil. Vet. Parasitol. 157, 299–305. pregnant women. Revista da Sociedade Brasileira de Medicina Tropical 45, 357–364.
Dubey, JP, Verma, SK, Ferreira, LR, Oliveira, S, Cassinelli, AB, Ying, Y, Jones, JL, 2014. https://doi.org/10.1590/S0037-86822012000300015.
Detection and survival of Toxoplasma gondii in milk and cheese from experimentally Franco, RM., 2007. Protozoários de veiculação hídrica: relevância em saúde Pública. Rev
infected goats. Journal of food protection 77 (10), 1747–1753. Panam Infectol 9, 36–43.
Dubey, JP., 2002. A review of toxoplasmosis in wild birds. Veterinary Parasitology 106 Frazão-Teixeira, E, Sundar, N, Dubey, JP, Grigg, ME, de Oliveira, FC., 2011. Multi-locus
(2), 121–153. https://doi.org/10.1016/s0304-4017(02)00034-1. DNA sequencing of Toxoplasma gondii isolated from Brazilian pigs identifies
Dubey, JP., 1998. Advances in the life cycle of Toxoplasma gondii. Int J Parasitol 28, genetically divergent strains. Vet Parasitol 175 (1-2), 33–39. https://doi.org/
1019–1024. 10.1016/j.vetpar.2010.09.030.
Dubey, JP., 2008. The History of Toxoplasma gondii-The First 100 Years. J. Eukaryot. Frenkel, JK, Dubey, JP, Miller, NL., 1970. Toxoplasma gondii in cats: fecal stages
Microbiol. 55 (6), 467–475. identified as coccidian oocysts. Science 6 (3919), 893–896. https://doi.org/
Dubey, JP., 2009. History of the discovery of the life cycle of Toxoplasma gondii. Int J 10.1126/science.167.3919.893, 167.
Parasitol 1 (8), 877–882. https://doi.org/10.1016/j.ijpara.2009.01.005, 39. Frenkel, JK, Hassanein, KM, Hassanein, RS, Brown, E, Thulliez, P, Quintero-Nunez, R.,
Dubey, JP., 2010. Toxoplasma gondii infections in chickens (Gallus domesticus): 1995. Transmission of Toxoplasma gondii in Panama City, Panama: a five-year
prevalence, clinical disease, diagnosis and public health significance. Zoonoses prospective cohort study of children, cats, rodents, birds, and soil. Am J Trop Med
Public Health 57, 60–73. https://doi.org/10.1111/j.1863-2378.2009.01274.x. Hyg 53 (5), 458–468. https://doi.org/10.4269/ajtmh.1995.53.458.
Dubey, JP., 2010. Toxoplasmosis of Animals and Humans, 2nd ed. CRC Press, Boca Freyre, A, Correa, O, Falcón, J, Mendez, J, González, M, Venzal, JM, 2001. Some factors
Raton, FL, pp. 1–313. https://doi.org/10.1186/1756-3305-3-112. influencing transmission of toxoplasma in pregnant rats fed cysts. Parasitol Res 87
Dubey, JP., 2004. Toxoplasmosis – a waterborne zoonosis. Vet Parasitol 126, 57–72. (11), 941–944. https://doi.org/10.1007/s004360100486.
Dumètre, A, Dubey, JP, Ferguson, DJ, Bongrand, P, Azas, N, Puech, PH, 2013. Mechanics Freyre, A., Araujo, F.A., Fialho, C.G., Bigatti, L.E., Falcón, J.D., 2012. Protection in a
of the Toxoplasma gondii oocyst wall. Proc Natl Acad Sci U S A 110 (28), hamster model of congenital toxoplasmosis. Veterinary parasitology 183 (3-4),
11535–11540. https://doi.org/10.1073/pnas.1308425110. 359–363. https://doi.org/10.1016/j.vetpar.2011.07.039.
Dunay, IR, Gajurel, K, Dhakal, R, Liesenfeld, O, Montoya, JG, 2018. Treatment of Fujii, H, Kamiyama, T, Hagiwara, T., 1983. Species and strain differences in sensitivity to
Toxoplasmosis: Historical Perspective, Animal Models, and Current Clinical Practice. Toxoplasma infection among laboratory rodents. Jpn J Med Sci Biol 36 (6), 343–346.
Clin Microbiol Rev 31 (4). https://doi.org/10.1128/CMR.00057-17 e00057-17. https://doi.org/10.7883/yoken1952.36.343.
Edvinsson, B, Lundquist, J, Ljungman, P, et al., 2008. A prospective study of diagnosis of Fux, B., Ferreira, A.M., Cassali, G.D., Tafuri, W.L., Vitor, R.W, 2000. Experimental
Toxoplasma gondii infection after bone marrow transplantation. APMIS 116 (5), toxoplasmosis in BALB/c mice. Prevention of vertical disease transmission by
345–351. treatment and reproductive failure in chronic infection. Memórias do Instituto
Ekman, CCJ, et al., 2012. Case-control study of an outbreak of acute toxoplasmosis in an Oswaldo Cruz 95, 121–126. https://doi.org/10.1590/S0074-02762000000100020.
industrial plant in the state of São Paulo, 54. Revista do Instituto de Medicina Fux, B, Rodrigues, CV, Portela, RW, Silva, NM, Su, C, Sibley, D, Vitor, RW,
Tropical de São Paulo [online], Brazil, pp. 239–244. https://doi.org/10.1590/ Gazzinelli, RT., 2003. Role of cytokines and major histocompatibility complex
S0036-46652012000500001, 2012vn. 5 [Accessed 3 October 2021]. restriction in mouse resistance to infection with a natural recombinant strain (type I-
Elamin, EA, Elias, S, Daugschies, A, Rommel, M., 1992. Prevalence of Toxoplasma gondii III) of Toxoplasma gondii. Infect Immun 71 (11), 6392–6401. https://doi.org/
antibodies in pastoral camels (Camelus dromedarius) in the Butana plains, mid- 10.1128/IAI.71.11.6392-6401.2003. NovPMID: 14573660; PMCID: PMC219541.
Eastern Sudan. Vet Parasitol 43, 171–175. Gajria, B, Bahl, A, Brestelli, J, et al., 2008. ToxoDB: an integrated Toxoplasma gondii
Elmore, SA, Samelius, G., Al-Adhami, B., Huyvaert, K.P., Bailey, L.L., Alisauskas, R.T., database resource. Nucleic Acids Res 36 (Database issue), D553–D556. https://doi.
Gajadhar, A.A., Jenkins, E.J, 2016. Estimating Toxoplasma gondii exposure in arctic org/10.1093/nar/gkm981.
foxes (Vulpes lagopus) while navigating the imperfect world of wildlife serology. Gao, JM, Yi, SQ, Wu, MS, Geng, GQ, Shen, JL, Lu, FL, Hide, G, Lai, DH, Lun, ZR., 2014.
Journal of wildlife diseases 52 (1), 47–56. https://doi.org/10.7589/2015-03-075. Investigation of infectivity of neonates and adults from different rat strains to
England, J H, Bailin, S S, Gehlhausen, J R, Rubin, D.H., 2019. Toxoplasmosis: The Heart Toxoplasma gondii Prugniaud shows both variation which correlates with iNOS and
of the Diagnosis. Open Forum Infectious Diseases Volume 6 (Issue 1). https://doi. Arginase-1 activity and increased susceptibility of neonates to infection. Exp
org/10.1093/ofid/ofy338. Parasitol 149, 47–53. https://doi.org/10.1016/j.exppara.2014.12.008, 2015 Feb.
Faria Junior, GM, Murata, FHA, Lorenzi, HA, Castro, BBP, Assoni, LCP, Ayo, CM, Gao, YM, Ding, H, Lamberton, PHL, Lu, DB., 2016. Prevalence of Toxoplasma gondii in pet
Brandão, CC, de Mattos, LC, 2021. The role of microRNAs in the infection by dogs in mainland China: A meta-analysis. Vet Parasitol 15, 126–130. https://doi.
T. gondii in humans. Front Cell Infect Microbiol 14, 670548. https://doi.org/ org/10.1016/j.vetpar.2016.10.009, 229.
10.3389/fcimb.2021.670548, 11. Gaulin, C, Ramsay, D, Thivierge, K, et al., 2020. Acute Toxoplasmosis among Canadian
Fayer, R, Dubey, JP, Lindsay, DS., 2004. Zoonotic protozoa: from land to sea. Trends Deer Hunters Associated with Consumption of Undercooked Deer Meat Hunted in
Parasitol 20 (11), 531–536. https://doi.org/10.1016/j.pt.2004.08.008. the United States. Emerging Infectious Diseases 26 (2), 199–205. https://doi.org/
Felin, E, Näreaho, A., Fredriksson-Ahomaa, M., 2017. Comparison of commercial ELISA 10.3201/eid2602.191218.
tests for the detection of Toxoplasma antibodies in the meat juice of naturally Gazzinelli, R, Xu, Y, Hieny, S, Cheever, A, Sher, A, 1992. Simultaneous depletion of CD4
infected pigs. Veterinary parasitology 238, 30–34. https://doi.org/10.1016/j. + and CD8+ T lymphocytes is required to reactivate chronic infection with
vetpar.2017.03.012. Toxoplasma gondii. Journal Immunology 1 (1), 175–180. https://doi.org/10.1007/
Fernandes, NS, Guimarães, HR, Amorim, AC, da, S, Reis, MB, Trindade, RA, Melo, ACFL., 978-1-4020-5614-7_1757, 149.
2015. Avaliação Parasitológica de Hortaliças: da Horta ao Consumidor Final. Saúde Gazzonis, AL, Zanzani, SA, Villa, L, Manfredi, MT., 2019. Toxoplasma gondii in naturally
E Pesqui 8 (2), 255–265. infected goats: Monitoring of specific IgG levels in serum and milk during lactation
Ferra, B, Holec-Gąsior, L., Kur, J., 2015. Serodiagnosis of Toxoplasma gondii infection in and parasitic DNA detection in milk. Prev Vet Med 1 (170), 104738. https://doi.org/
farm animals (horses, swine, and sheep) by enzyme-linked immunosorbent assay 10.1016/j.prevetmed.2019.104738.
using chimeric antigens. Parasitology international 64 (5), 288–294. https://doi. Germano, PML, Germano, MIS., Germano PML, Germano MIS. (Org.), 2011.
org/10.1016/j.parint.2015.03.004. Toxoplasmose. Higiene e vigilância sanitária de alimentos, 4.ed. Manole, Barueri,
Ferreira, AI, De Mattos, CC, Frederico, FB, Meira, CS, Almeida Jr, GC, Nakashima, F, pp. 440–448.
Bernardo, CR, Pereira-Chioccola, VL, De Mattos, LC., 2014. Risk factors for ocular Gharekhani, J, Yakhchali, M, Esmaeilnejad, B, Mardani, K, Majidi, G, Sohrabi, A, 2018.
toxoplasmosis in Brazil. Epidemiol Infect 142 (1), 142–148. https://doi.org/ Seroprevalence and Risk Factors of Neospora caninum and Toxoplasma gondii in Small
10.1017/S0950268813000526. Ruminants in Southwest of Iran. Arch Razi Inst 73, 305–310. https://doi.org/
Ferreira, IM, Vidal, JE, de Mattos Cde, C, de Mattos, LC, Qu, D, Su, C, Pereira- 10.1016/j.vetpar.2008.12.022.
Chioccola, VL., 2011. Toxoplasma gondii isolates: multilocus RFLP-PCR genotyping

13
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

Gilbert, RE, Freeman, K, Lago, EG, Bahia-Oliveira, LM, Tan, HK, Wallon, M, Jenkins, EJ, Simon, A., Bachand, N., Stephen, C., 2015. Wildlife parasites in a One Health
Buffolano, W, Stanford, MR, Petersen, E., 2008. European Multicentre Study on world. Trends in parasitology 31 (5), 174–180. https://doi.org/10.1016/j.
Congenital Toxoplasmosis (EMSCOT). Ocular sequelae of congenital toxoplasmosis pt.2015.01.002.
in Brazil compared with Europe. PLoS Negl Trop Dis 13 (8), e277. https://doi.org/ Jensen, M, Brockie, PJ, Maricq, AV., 2012. Wnt signaling regulates experience-
10.1371/journal.pntd.0000277, 2. dependent synaptic plasticity in the adult nervous system. Cell Cycle 15 (14),
Gilot-Fromont, E, Jégo, M, Bonenfant, C, Gibert, P, Rannou, B, Klein, F, Gaillard, JM., 2585–2586. https://doi.org/10.4161/cc.21138, 11.
2012. Immune phenotype and body condition in roe deer: individuals with high Jewell, ML, Frenkel, JK, Johnson, KM, Reed, V, Ruiz, A, 1972. Development of
body condition have different, not stronger immunity. PLoS One 7 (9), e45576. Toxoplasma oocysts in neotropical felidae. Am J Trop Med Hyg 21 (5), 512–517.
https://doi.org/10.1371/journal.pone.0045576. https://doi.org/10.4269/ajtmh.1972.21.512.
Gois, GSS, Branco, N, Franco, RM., 2018. Segurança Alimentar: Padronização de Jokelainen, P., Tagel, M., Mõtus, K., Viltrop, A., Lassen, B., 2017. Toxoplasma gondii
Metodologia para Detecção dos Protozoários potencialmente patogênicos (Giardia seroprevalence in dairy and beef cattle: Large-scale epidemiological study in Estonia.
spp. e Cryptosporidium spp.) em hortaliças de produção orgânica e convencional. Veterinary parasitology 236, 137–143. https://doi.org/10.1016/j.
Rev Trab Iniciaç Cient 26. vetpar.2017.02.014.
Gomes, D., Krawczak, F., Oliveira, C., Ferreira Júnior, Á., Fernandes, É., Lopes, W., Jones, JL, Dargelas, V., Roberts, J., Press, C., Remington, J.S., Montoya, J.G., 2009. Risk
Sevá, A., Gennari, S.M, 2020. Toxoplasma gondii in cattle in Brazil: a review. Revista factors for Toxoplasma gondii infection in the United States. Clin. Infect. Dis. 49,
brasileira de parasitologia veterinaria. = Brazilian journal of veterinary parasitology: 878–884.
Orgao Oficial do Colegio Brasileiro de Parasitologia Veterinaria 29 (1), e015719 Jones, JL, Dubey, JP., 2010. Waterborne toxoplasmosis - Recent developments. Exp
https://doi.org/10.1590/S1984-29612019106. Parasitol 124, 10–25.
Grigg, ME, Ganatra, J, Boothroyd, JC, Margolis, TP., 2001. Unusual abundance of Jones, JL, Dubey, J.P., Souza, W., Belfort, JR., 2014. Epidemiologia da Toxoplasmose. R.,
atypical strains associated with human ocular toxoplasmosis. J Infect Dis 1 (5), comp. Toxoplasmose & Toxoplasma gondii [online]. Rio de Janeiro: Editora Fiocruz,
633–639. https://doi.org/10.1086/322800, 184. 117126. https://doi.org/10.7476/9788575415719.0010. ISBN: 978-85-7541-571-9.
Guo, M, Dubey, JP, Hill, D, Buchanan, RL, Gamble, HR, Jones, JL, Pradhan, AK., 2015. Jones, JL, Kruszon-Moran, D, Wilson, M, McQuillan, G, Navin, T, McAuley, JB, 2001.
Prevalence and risk factors for Toxoplasma gondii infection in meat animals and meat Toxoplasma gondii infection in the United States: seroprevalence and risk factors. Am
products destined for human consumption. J Food Prot 201785 (2), 457–476. J Epidemiol 15 (4), 357–365. https://doi.org/10.1093/aje/154.4.357, 154.
https://doi.org/10.4315/0362-028X.JFP-14-328. Juan-Sallés, C, Prats, N, López, S, Domingo, M, Marco, AJ, Morán, JF, 1997. Epizootic
Gutiérrez-Expósito, D, Arteche-Villasol, N, Vallejo-García, R, Ferreras-Estrada, MC, disseminated toxoplasmosis in captive slender-tailed meerkats (Suricata suricatta).
Ferre, I, Sánchez-Sánchez, R, Ortega-Mora, LM, Pérez, V, Benavides, J, 2020. Vet Pathol 34 (1), 1–7. https://doi.org/10.1177/030098589703400101.
Characterization of Fetal Brain Damage in Early Abortions of Ovine Toxoplasmosis. Jungersen, G, Bille-Hansen, V, Jensen, L, Lind, P., 2001. Transplacental transmission of
Vet Pathol 57 (4), 535–544. https://doi.org/10.1177/0300985820921539. JulEpub Toxoplasma gondii in minipigs infected with strains of different virulence. J Parasitol
2020 May 14. PMID: 32406321. 87 (1), 108–113. https://doi.org/10.1645/0022-3395(2001)087[0108:TTOTGI]2.0.
Habegger de Sorrentino, A, López, R, Motta, P, Marinic, K, Sorrentino, A, Iliovich, E, CO;2.
Rubio, AE, Quarleri, J, Salomón, H, 2005. HLA class II involvement in HIV- Khan, A, Jordan, C, Muccioli, C, Vallochi, AL, Rizzo, LV, et al., 2006. Genetic divergence
associated Toxoplasmic encephalitis development. Clinical 115 (2), 133–137. of Toxoplasma gondii strains associated with ocular toxoplasmosis. Brazil. Emerg
https://doi.org/10.1016/j.clim.2005.01.003. Infect Dis. 12, 942–949.
Hill, DE, Chirukandoth, S, Dubey, JP, 2005. Biology and epidemiology of Toxoplasma Khan, A., Dubey, J.P., Su, C., Ajioka, J.W., Rosenthal, B.M., Sibley, L.D, 2011. Genetic
gondii in man and animals. Anim Health Res Rev 6 (1), 41–61. https://doi.org/ analyses of atypical Toxoplasma gondii strains reveal a fourth clonal lineage in North
10.1079/ahr2005100. PMID: 16164008. America. International journal for parasitology 41 (6), 645–655. https://doi.org/
Hill, DE, Chirukandoth, S, Dubey, JP, Lunney, JK, Gamble, H.R., 2006. Comparison of 10.1016/j.ijpara.2011.01.005.
detection methods for Toxoplasma gondii in naturally and experimentally infected Kianersi, F, Naderi Beni, A, Naderi Beni, Z., 2012. Clinical manifestation and prognosis of
swine. Veterinary parasitology 141 (1-2), 9–17. https://doi.org/10.1016/j. active ocular toxoplasmosis in Iran. Int Ophthalmol 32 (6), 539–545. https://doi.
vetpar.2006.05.008. org/10.1007/s10792-012-9599-0.
Hill, DE, Dubey, JP., 2002. Toxoplasma gondii: transmission, diagnosis and prevention. Kijlstra, A, Jongert, E., 2009. Toxoplasma-safe meat: close to reality? Trends Parasitol 25,
Clin Microbiol Infect 8 (10), 634–640. https://doi.org/10.1046/j.1469- 18–22. https://doi.org/10.1016/j.pt.2008.09.008.
0691.2002.00485.x. Kim, MJ, Jung, BK, Cho, J, Song, H, Pyo, KH, Lee, JM, Kim, MK, Chai, JY, 2016.
Hippólito, DDC, Gomes, AHS, Maia, MM, Meira-Strejevitch, CDS, Kanamura, CT, Lauletta Exosomes Secreted by Toxoplasma gondii-infected L6 cells: Their Effects on Host Cell
Lindoso, JA, Pereira-Chioccola, VL, 2019 Jan. Gene expression profile of cytokines Proliferation and Cell Cycle Changes. Korean J Parasitol 54 (2), 147–154. https://
produced in biopsies from patients with American cutaneous leishmaniasis. Acta doi.org/10.3347/kjp.2016.54.2.147.
Trop 189, 69–75. https://doi.org/10.1016/j.actatropica.2018.09.022. Klaren, VN, Kijlstra, A., 2002. Toxoplasmosis, an overview with emphasis on ocular
Hiramoto, R.M., Mayrbaurl-Borges, M., Galisteo Jr, A.J., Meireles, L.R., Macre, M.S., involvement. Ocul Immunol Inflamm 10 (1), 1–26. https://doi.org/10.1076/
Andrade Jr, H.F., 2001. Infectivity of cysts of the ME-49 Toxoplasma gondii strain in ocii.10.1.1.10330.
bovine milk and homemade cheese. Revista de Saúde Pública 35, 113–118. Kotula, AW, Dubey, JP, Sharar, AK, Andrews, CD, Shen, SK, Lindsay, DS., 1991. Effect of
Hohweyer, J, Cazeaux, C, Travaillé, E, Languet, E, Dumètre, A, Aubert, D, et al., 2016. Freezing on Infectivity of Toxoplasma gondii Tissue Cysts in Pork. J Food Prot 54 (9),
Simultaneous detection of the protozoan parasites Toxoplasma, Cryptosporidium 687–690. https://doi.org/10.4315/0362-028X-54.9.687.
and Giardia in food matrices and their persistence on basil leaves. Food Microbiol Kowal, J, Tkach, M, Théry, C., 2014. Biogenesis and secretion of exosomes. Curr Opin
57, 36–44. Cell Biol 29, 116–125. https://doi.org/10.1016/j.ceb.2014.05.004.
Holland, G.N., 2004. Ocular toxoplasmosis: a global reassessment: part II: disease Krusor, C, Smith, WA, Tinker, MT, Silver, M, Conrad, PA, Shapiro, K, 2015.
manifestations and management. Am J Ophthalmol 137, 1–17. https://doi.org/ Concentration and retention of Toxoplasma gondii oocysts by marine snails
10.1016/j.ajo.2003.10.032 vn. demonstrate a novel mechanism for transmission of terrestrial zoonotic pathogens in
Holman, WL, Vercammenb, M, De Braekeleer, J, Verschueren, H., 2000. Identification of coastal ecosystems. Environ Microbiol 17 (11), 4527–4537. https://doi.org/
a 200- to 300-fold repetitive 529 bp DNA fragment in Toxoplasma gondii, and its use 10.1111/1462-2920.12927.
for diagnostic and quantitative PCR. Inter J Parasitol 30, 69–75. Lachkhem, A., Galal, L., Lahmar, I., Passebosc, K., Riahi, H., Plault, N., 2021. First
Hosseini, SA, Amouei, A, Sharif, M, Sarvi, SH, Galal, L, Javidnia, J, Pagheh, AS, isolation and genotyping of Toxoplasma gondii strains from domestic animals in
Gholami, S, Mizani D Daryani, A., 2019. Human toxoplasmosis: a systematic review Tunisia. Sci Rep 11, 9328. https://doi.org/10.1038/s41598-021-88751-1.
for genetic diversity of Toxoplasma gondii in clinical samples. Epidemiology and Laforet, CK, Deksne, G, Petersen, HH, Jokelainen, P, Johansen, MV, Lassen, B., 2019.
Infection 147, 1–9. https://doi.org/10.1017/S0950268818002947 e36. Toxoplasma gondii seroprevalence in extensively farmed wild boars (Sus scrofa) in
Howe, DK, Sibley, LD., 1995. Toxoplasma gondii comprises three clonal lineages: Denmark. Acta Vet Scand 15 (1), 4. https://doi.org/10.1186/s13028-019-0440-x,
correlation of parasite genotype with human disease. J Infect Dis 172 (6), 61.
1561–1566. https://doi.org/10.1093/infdis/172.6.1561. Lagana, A, Russo, F, Veneziano, D, et al., 2013. Extracellular circulating viral
Hunter, CA, Sibley, LD., 2012. Modulation of innate immunity by Toxoplasma gondii microRNAs: current knowledge and perspectives, 4. Front Genet, p. 120.
virulence effectors. Nature reviews. Microbiology 10 (11), 766–778. https://doi.org/ Lake, R, Hudson, A, Cressey, P, 2020. Risk profile: Toxoplasma gondii in red meat and
10.1038/nrmicro2858. meat products. New Zealand Food Authority. Christchurch: Institute of
Hussain, MA, Stitt, V, Szabo, EA, Nelan, B., 2017. Toxoplasma gondii in the Food Supply. Environmental Science – Research Limited, p. 36. ISBN No: 978-0-908334-11-7.
Pathogens 6, 1–7. Lalonde, LF, Gajadhar, AA., 2016. Detection of Cyclospora cayetanensis, Cryptosporidium
Hutchison, WM, Dunachie, JF, Siim, JC, Work, K., 1969. Life cycle of Toxoplasma gondii. spp., and Toxoplasma gondii on imported leafy green vegetables in Canadian survey.
Br Med J 27 (5686), 806. https://doi.org/10.1136/bmj.4.5686.806-b, 4. Food Waterborne Parasitol 2, 8–14.
Innes, EA, Hamilton, C, Garcia, J.L., Chryssafidis, A., Smith, D., 2019. A one health Langoni, H., Silva, A.V., Bergamaschi, S., Lima, V., 2007. Utilization of modified
approach to vaccines against Toxoplasma gondii. Food and waterborne parasitology agglutination test and indirect immunofluorescent antibody test for the detection of
15, e00053, 10.1016/j.fawpar.2019.e00053. Toxoplasma gondii antibodies in naturally exposed horses. Brazilian Journal of
Ismael, AB, Dimier-Poisson, I, Lebrun, M, Dubremetz, JF, Bout, D, Mevelec, MN., 2006. Veterinary Research and Animal Science 44, 27–32. https://doi.org/10.11606/
Mic1-3 knockout of Toxoplasma gondii is a successful vaccine against chronic and issn.1678-4456.bjvras.2007.26657.
congenital toxoplasmosis in mice. J Infect Dis 15 (8), 1176–1183. https://doi.org/ Lass, A, Pietkiewicz, H, Szostakowska, B, Myjak, P., 2012. The first detection of
10.1086/507706, 194. Toxoplasma gondii DNA in environmental fruits and vegetables samples. Eur J Clin
Jamra, LM, Martins, MC, Vieira M de, P., 1991. Ação do sal de cozinha sobre o Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol 31 (6), 1101–1108.
Toxoplasma gondii. Rev Inst Med Trop Sao Paulo 33, 359–363. https://doi.org/ Lassen, B., Janson, M., Viltrop, A., Neare, K., Hütt, P., Golovljova, I., Tummeleht, L.,
10.1590/S0036-46651991000500004. Jokelainen, P., 2016. Serological Evidence of Exposure to Globally Relevant

14
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

Zoonotic Parasites in the Estonian Population. PloS one 11 (10), e0164142. https:// Marciano, MAM, Andrade, JHF, Meireles, LR., 2018. Avaliação da técnica de ELISA para
doi.org/10.1371/journal.pone.0164142. pesquisa de IgG anti-Toxoplasma gondii em exsudatos de carnes de sol. Brazilian
Lehmann, T., Marcet, P.L., Graham, D.H., Dahl, E.R., Dubey, J.P, 2006. Globalization and Journal of Food Technology 21 (1), 1–6.
the population structure of Toxoplasma gondii. Proceedings of the National Academy Marcilla, A., Martin-Jaular, L., Trelis, M., de Menezes-Neto, A., Osuna, A., Bernal, D.,
of Sciences of the United States of America 103 (30), 11423–11428. https://doi.org/ Fernandez-Becerra, C., Almeida, I.C., Del Portillo, H.A, 2014. Extracellular vesicles
10.1073/pnas.0601438103. in parasitic diseases. Journal of extracellular vesicles 3, 25040. https://doi.org/
Li D, Yang S, Peng H. Nan Fang Yi Ke Da Xue Xue Bao. 2020. Characteristics of exosomes 10.3402/jev.v3.25040.
secreted by Toxoplasma gondii-infected mouse dendritic DC2.4 cells. 30;40(5):727- Mareze, M, Benitez, ADN, Brandão, APD, Pinto-Ferreira, F, Miura, AC, Martins, FDC,
732. doi: 10.12122/j.issn.1673-4254.2020.05.19. Caldart, ET, Biondo, AW, Freire, RL, Mitsuka-Breganó, R, Navarro, IT., 2019.
Li, DL, Zou, WH, Deng, SQ, Peng, HJ., 2019. Analysis of the differential exosomal Socioeconomic vulnerability associated to Toxoplasma gondii exposure in southern
miRNAs of DC2.4 dendritic cells induced by Toxoplasma gondii infection. Int J Mol Brazil. PLoS One 14 (2), e0212375. https://doi.org/10.1371/journal.pone.0212375.
Sci 5 (21), 5506. https://doi.org/10.3390/ijms20215506, 20. Markovich, MP, Shohat, T, Riklis, I, et al., 2014. Seroepidemiology of Toxoplasma gondii
Li, X, Ni, HB, Ren, WX, Jiang, J, Gong, QL, Zhang, XX., 2020. Seroprevalence of infection in the Israeli population. Epidemiol Infect 142 (1), 149–155. https://doi.
Toxoplasma gondii in horses: A global systematic review and meta-analysis. Acta Trop org/10.1017/S0950268813000903.
201, 105222. https://doi.org/10.1016/j.actatropica.2019.105222. Maspi, N, Nayeri, T, Moosazadeh, M, Sarvi, S, Sharif, M, Daryani, A., 2021. Global
Li, Y, Liu, Y, Xiu, F, Wang, J, Cong, H, He, S, Shi, Y, Wang, X, Li, X, Zhou, H, 2018. seroprevalence of Toxoplasma gondii in Camelidae: A systematic review and meta-
Characterization of exosomes derived from Toxoplasma gondii and their functions in analysis. Acta Parasitol 5. https://doi.org/10.1007/s11686-020-00333-9.
modulating immune responses. Int J Nanomedicine 13, 467–477. https://doi.org/ Massie, GN, Ware, MW, Villegas, EN, Black, MW., 2010. Uptake and transmission of
10.2147/IJN.S151110, 19. Toxoplasma gondii oocysts by migratory, filter-feeding fish. Vet Parasitol 11 (3-4),
Lin, MH, Chen, TC, Kuo, TT, Tseng, CC, Tseng, CP., 2000. Real-time PCR for quantitative 296–303. https://doi.org/10.1016/j.vetpar.2010.01.002, 169.
detection of Toxoplasma gondii. J Clin Microbiol 38, 4121–4125. Mattos, CC, Meira, CS, Ferreira, AI, Frederico, FB, Hiramoto, RM, Almeida Jr, GD,
Lind, P., Haugegaard, J., Wingstrand, A., Henriksen, S.A, 1997. The time course of the Mattos, LC, Pereira-Chioccola, VL., 2011. Contribution of laboratory methods in
specific antibody response by various ELISAs in pigs experimentally infected with diagnosing clinically suspected ocular toxoplasmosis in Brazilian patients. Diagn
Toxoplasma gondii. Veterinary parasitology 71 (1), 1–15. https://doi.org/10.1016/ Microbiol Infect Dis. 70 (3), 362–366. https://doi.org/10.1016/j.
s0304-4017(97)00010-1. diagmicrobio.2011.02.002.
Lindsay, DS, Collins, MV, Mitchell, SM, Cole, RA, Flick, GJ, Wetch, CN, Lindquist, A, McAuley, J, Boyer, KM, Patel, D, Mets, M, Swisher, C, Roizen, N, Wolters, C, Stein, L,
Dubey, JP., 2003. Sporulation and survival of Toxoplasma gondii oocysts in seawater. Stein, M, Schey, W, et al., 1994. Early and longitudinal evaluations of treated infants
J Eukaryot Microbiol 50 (Suppl), 687–688. https://doi.org/10.1111/j.1550- and children and untreated historical patients with congenital toxoplasmosis: the
7408.2003.tb00688.x. Chicago Collaborative Treatment Trial. Clin Infect Dis 18 (1), 38–72. https://doi.
Lindsay, DS, Dubey, JP., 2020. Neosporosis, Toxoplasmosis, and Sarcocystosis in org/10.1093/clinids/18.1.38, 1994 JanErratum in: Clin Infect Dis 1994 Oct;19(4):
Ruminants: An Update. Vet Clin North Am Food Anim Pract 36 (1), 205–222. 820.
https://doi.org/10.1016/j.cvfa.2019.11.004. McLeod, R, Estes, RG, Mack, DG, Cohen, H, 1984. Immune response of mice to ingested
Lindstrom, I, Kaddu-Mulindwa, DH, Kironde, F, et al., 2006. Prevalence of latent and Toxoplasma gondii: a model of toxoplasma infection acquired by ingestion. J Infect
reactivated Toxoplasma gondii parasites in HIV-patients from Uganda. Acta Tropica Dis 149 (2), 234–244. https://doi.org/10.1093/infdis/149.2.234.
100 (3), 218–222. Mecca, JN, Meireles, LR, de Andrade Jr., HF, 2011. Quality control of Toxoplasma gondii
Liu, L, Liu, LN, Wang, P, Lv, TT, Fan, YG, Pan, HF, 2017. Elevated seroprevalence of in meat packages: standardization of an ELISA test and its use for detection in rabbit
Toxoplasma gondii in AIDS/HIV patients: A meta-analysis. Acta Trop 176, 162–167. meat cuts. Meat Sci 88 (3), 584–589. https://doi.org/10.1016/j.
https://doi.org/10.1016/j.actatropica.2017.08.001. meatsci.2011.01.016.
Liu Q, Wang ZD, Huang SY, Zhu XQ. 2015. Diagnosis of toxoplasmosis and typing of Meira-Strejevitch, CS, Pereira, IS, Hippólito, DDC, Maia, MM, Cruz, AB, Gava, R, Brandão
Toxoplasma gondii.Parasites Vectors 8. doi 10.1186/s13071-015-0902-6. de Mattos, CC, Frederico, FB, Siqueira, RC, Mattos, LC, Pereira-Chioccola, VL., 2020.
Lv, QY, Zheng, HL, Yang, WH, Liu, GH, 2021. Molecular Detection of Toxoplasma gondii Ocular toxoplasmosis associated with up-regulation of miR-155-5p/miR-29c-3p and
and Neospora caninum in Domestic Ducks in Hunan Province. China. Front Vet Sci. down-regulation of miR-21-5p/miR-125b-5p. Cytokine 127, 154990. https://doi.
15 (8), 649603 https://doi.org/10.3389/fvets.2021.649603. org/10.1016/j.cyto.2020.154990.
Machado, FP, Kmetiuk, LB, Teider-Junior, PI, Pellizzaro, M., Yamakawa, AC, Melo, RPB, Wanderley, FS, Porto, WJN, Pedrosa, CM, Hamilton, CM, de Oliveira, MHGS,
Martins, CM, Biondo, AW, 2019. Seroprevalence of anti-Toxoplasma gondii Ribeiro-Andrade, M, Rêgo, RCDS, Katzer, F, Mota, RA., 2020 Aug. Description of an
antibodies in wild boar (Sus scrofa), hunting and hunting dogs in Brazil. PloS one 14 atypical Toxoplasma gondii isolate from a case of congenital toxoplasmosis in
(10), e0223474. https://doi.org/10.1371/journal.pone.0223474. northeastern Brazil. Parasitol Res 119 (8), 2727–2731. https://doi.org/10.1007/
Macrì, G, Sala, M, Linder, AM, Pettirossi, N, Scarpulla, M., 2009. Comparison of indirect s00436-020-06746-9. Epub 2020 Jun 10. PMID: 32518965.
fluorescent antibody test and modified agglutination test for detecting Toxoplasma Mesquita, RT, Ziegler, AP, Hiramoto, RM, Vidal, JE, Pereira-Chioccola, VL., 2010. Real-
gondii immunoglobulin G antibodies in dog and cat. Parasitol Res 105 (1), 35–40. time quantitative PCR in cerebral toxoplasmosis diagnosis of Brazilian human
https://doi.org/10.1007/s00436-009-1358-4. immunodeficiency virus-infected patients. J Med Microbiol 59, 641–647.
Magalhães, FJ, da Silva, JG, Ribeiro-Andrade, M, Pinheiro, JW, Júnior, Aparecido, Miller, M, Shapiro, K, Murray, MJ, Haulena, M, Raverty, S., 2018. Protozoan Parasites of
Mota, R., 2016 Jul. High prevalence of toxoplasmosis in free-range chicken of the Marine Mammals, 3Ed. CRC Press, p. Pages 46. https://doi.org/10.1201/
Fernando de Noronha Archipelago. Brazil. Acta Trop. 159, 58–61. https://doi.org/ 9781315144931. eBook ISBN9781315144931.
10.1016/j.actatropica.2016.03.034. Epub 2016 Mar 28. PMID: 27032879. Miller, MA, Gardner, IA, Packham, A., Mazet, J.K., Hanni, K.D., Jessup, D., Estes, J.,
Mahami-Oskouei, M, Moradi, M, Fallah, E, Hamidi, F, Asl Rahnamaye Akbari, N., 2017. Jameson, R., Dodd, E., Barr, B.C., Lowenstine, L.J., Gulland, F.M., Conrad, P.A.,
Molecular Detection and Genotyping of Toxoplasma gondii in Chicken, Beef, and 2002. Evaluation of an indirect fluorescent antibody test (IFAT) for demonstration of
Lamb Meat Consumed in Northwestern Iran. Iran J Parasitol 12 (1), 38–45. antibodies to Toxoplasma gondii in the sea otter (Enhydra lutris). The Journal of
Maia, MM, Cruz, AB, Pereira, IS, Taniwaki, NN, Namiyama, GM, Pereira-Chioccola, VL., parasitology 88 (3), 594–599. https://doi.org/10.1645/0022-3395 (2002)088
2021a. Characterization of murine extracellular vesicles and Toxoplasma gondii [0594:EOAIFA]2.0.CO;2.
infection. Parasite Immunol 43. https://doi.org/10.1111/pim.12869. Minbaeva, G, Schweiger, A, Bodosheva, A, Kuttubaev, O, Hehl, AB, Tanner, I, Ziadinov, I,
Maia, MM, Cruz, AB, Taniwaki, NN, Namiyama, GM, Gava, R, Gomes, AHS, Torgerson, PR, Deplazes, P., 2013. Toxoplasma gondii infection in Kyrgyzstan:
Kanamura, CT, Barbo, MLP, Pereira-Chioccola, VL., 2021. Immunization with seroprevalence, risk factor analysis, and estimate of congenital and AIDS-related
extracellular vesicles excreted by Toxoplasma gondii confers protection in murine toxoplasmosis. PLoS Negl Trop Dis 7 (2), e2043. https://doi.org/10.1371/journal.
infection, activating cellular and humoral responses. Int J Parasitol 51 (7), 559–569. pntd.0002043.
https://doi.org/10.1016/j.ijpara.2020.11.010. Minuzzi, CE, Portella, LP, Bräunig, P, et al., 2020. Isolation and molecular
Maleki, B., Ahmadi, N., Olfatifar, M., Gorgipour, M., Taghipour, A., Abdoli, A., characterization of Toxoplasma gondii from placental tissues of pregnant women
Khorshidi, A., Foroutan, M., Mirzapour, A., 2021. Toxoplasma oocysts in the soil of who received toxoplasmosis treatment during an outbreak in southern Brazil. PLoS
public places worldwide: a systematic review and meta-Analysis. Trans. R. Soc. Trop. One 15 (1). https://doi.org/10.1371/journal.pone.0228442, 2020e0228442.
Med. Hyg. 115, 471–481. Published 302020a.
Mangen, MJ, Bouwknegt, M, Friesema, IH, Haagsma, JA, Kortbeek, LM, Tariq, L, Montaner, S, Galiano, A, Trelis, M, Martin-Jaular, L, Del Portillo, HA, Bernal, D,
Wilson, M, van Pelt, W, Havelaar, AH., 2015. Cost-of-illness and disease burden of Marcilla, A., 2014. The Role of Extracellular Vesicles in Modulating the Host
food-related pathogens in the Netherlands. Int J Food Microbiol 196, 84–93. https:// Immune Response during Parasitic Infections. Front Immunol 8 (5), 433. https://doi.
doi.org/10.1016/j.ijfoodmicro.2014.11.022, 2011. org/10.3389/fimmu.2014.00433.
Mantel, PY, Marti, M., 2014. The role of extracellular vesicles in Plasmodium and other Montoya, JG, Liesenfeld, O., 2004. Toxoplasmosis. Lancet. 12 (9425), 1965–1976.
protozoan parasites. Cell Microbiol 16 (3), 344–354. https://doi.org/10.1111/ https://doi.org/10.1016/S0140-6736(04)16412-X, 363.
cmi.12259. Montoya, JG, Remington, JS., 1996. Toxoplasmic chorioretinitis in the setting of acute
Manzardo, C, Del Mar Ortega, M, Sued, O, García, F, Moreno, A, Miró, JM., 2005. Central acquired toxoplasmosis. Clin Infect Dis 23 (2), 277–282. https://doi.org/10.1093/
nervous system opportunistic infections in developed countries in the highly active clinids/23.2.277.
antiretroviral therapy era. J Neurovirol (Suppl 3), 72–82. https://doi.org/10.1080/ Moore, D.P., Regidor-Cerrillo, J., Morrell, E., Poso, M.A., Cano, D.B., Leunda, M.R.,
13550280500513846. Linschinky, L., Odeón, A.C., Odriozola, E., Ortega-Mora, L.M., Campero, C.M, 2008.
Marciano, MA M., Silva, R A, Barbosa, M L, Ferreira, ARS, Pereira-Chioccola, VL., 2020. The role of Neospora caninum and Toxoplasma gondii in spontaneous bovine abortion
Determination of the viability of Toxoplasma gondii oocysts by PCR real-time after in Argentina. Veterinary parasitology 156 (3-4), 163–167. https://doi.org/10.1016/
treatment with propidium monoazide. Revista Do Instituto De Medicina Tropical De j.vetpar.2008.06.020.
São Paulo 62, e84. https://doi.org/10.1590/S1678-9946202062084. Morais, RDAPB, Freire, A. B. C., Barbosa, D. R. L., Silva, L. D. C. T. D., Pinheiro, A. F.,
Costa, S. S. D. & Carmo, E. L. D. 2016. Acute toxoplasmosis outbreak in the

15
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

Municipality of Ponta de Pedras, Marajó archipelago, Pará State, Brazil: clinical, Pereira-Chioccola, VL, Vidal, JE, Su, C, 2009. Toxoplasma gondii infection and cerebral
laboratory, and epidemiological features. Rev Pan-Amaz Saude [online]. vol. 7, n. toxoplasmosis in HIV-infected patients. Future Microbiol 24 (10), 1363–1379.
esp, pp.143-152. ISSN 2176-6215. 10.5123/s2176-62232016000500016. https://doi.org/10.2217/fmb.09.89.
Moreno, B, Collantes-Fernández, E, Villa, A, Navarro, A, Regidor-Cerrillo, J, Ortega- Peyron, F, Wallon, M, Kieffer, F, Garweg, J., 2016. Toxoplasmosis. Infectious Diseases of
Mora, LM., 2012. Occurrence of Neospora caninum and Toxoplasma gondii the Fetus and the Newborn Infant. Elsevier, Philadelphia, PA, USA, pp. 949–1042.
infections in ovine and caprine abortions. Vet. Parasitol. 187, 312–318. https://doi. Avci, Pinar, Karimi, M., Sadasivam, M., Antunes-Melo, W.C., Carrasco, E., Hamblin, M.
org/10.1016/j.vetpar.2011.12.034. R., 2018. In-vivo monitoring of infectious diseases in living animals using
Moskwa, B, Kornacka, A, Cybulska, A, Cabaj, W, Reiterova, K, Bogdaszewski, M, Steiner- bioluminescence imaging. Virulence 9, 28–63. https://doi.org/10.1080/
Bogdaszewska, Z, Bien, J., 2018. Seroprevalence of Toxoplasma gondii and Neospora 21505594.2017.1371897.
caninum infection in sheep, goats, and fallow deer farmed on the same area. J Anim Plaza, J, Dámek, F, Villena, I, Innes, EA, Katzer, F, Hamilton, CM, 2020. Detection of
Sci 4 (6), 2468–2473. https://doi.org/10.1093/jas/sky122, 96. Toxoplasma gondii in retail meat samples in Scotland. Food Waterborne Parasitol 12
Moura, L, Bahia-Oliveira, LMG, Wada, MY, Jones, JL, Tuboi, SH, Carmo, EH, et al., 2006. (20), e00086. https://doi.org/10.1016/j.fawpar.2020.e00086.
Waterborne toxoplasmosis, Brazil, from field to gene. Emerg Infect Dis 12, 326–329. Pomares, C, Devillard, S, Holmes, TH, et al., 2018. Genetic Characterization of
Munoz, M., Liesenfeld, O., Heimesaat, M.M, 2011. Immunology of Toxoplasma gondii. Toxoplasma gondii DNA Samples Isolated From Humans Living in North America: An
Immunological reviews 240 (1), 269–285. https://doi.org/10.1111/j.1600- Unexpected High Prevalence of Atypical Genotypes. J Infect Dis 218 (11),
065X.2010.00992. 1783–1791. https://doi.org/10.1093/infdis/jiy375.
Nau, J., Eller, S.K., Wenning, J., Spekker-Bosker, K.H., Schroten, H., Schwerk, C., Pope, SM, Lässer, C., 2013. Toxoplasma gondii infection of fibroblasts causes the
Hotop, A., Groß, U., Däubener, W., 2017. Experimental Porcine Toxoplasma gondii production of exosome-like vesicles containing a unique array of mRNA and miRNA
Infection as a Representative Model for Human Toxoplasmosis. Mediators of transcripts compared to serum starvation. J Extracell Vesicles 11, 2. https://doi.org/
inflammation, 3260289. https://doi.org/10.1155/2017/3260289. 10.3402/jev.v2i0.22484. PMID: 24363837; PMCID: PMC3862870.
Navarro, IT, Vidotto, O, Giraldi, N, Mitsuka, R., 1992. Resistência do Toxoplasma gondii Portas, TJ., 2010. Toxoplasmosis in macropodids: a review. J Zoo Wildl Med 41 (1), 1–6.
ao cloreto de sódio e aos condimentos em linguiça de suínos. Bol Oficina Sanit https://doi.org/10.1638/2009-0148.1.
Panam 112, 138–143. Previato, M, Frederico, FB, Murata, FH, Siqueira, RC, Barbosa, AP, Silveira-Carvalho, AP,
Ngô, HM, Zhou, Y, Lorenzi, H, 2019. Toxoplasma Modulates Signature Pathways of Meira Cda, S, Pereira-Chioccola, VL, Gava, R, Martins Neto, PP, de Mattos, LC, de
Human Epilepsy. Neurodegeneration & Cancer. Sci Rep. 13, 11496. https://doi.org/ Mattos, CC, 2015. A Brazilian report using serological and molecular diagnosis to
10.1038/s41598-017-10675-6. monitoring acute ocular toxoplasmosis. BMC Res Notes 7 (8), 746. https://doi.org/
Nogareda, F., Le Strat, Y., Villena, I., De Valk, H., Goulet, V., 2014. Incidence and 10.1186/s13104-015-1650-6.
prevalence of Toxoplasma gondii infection in women in France, 1980-2020: model- Priya, J, Calderón, MM, Gilman, RH, et al., 2002. Optimization and evaluation of a PCR
based estimation. Epidemiology and infection 142 (8), 1661–1670. https://doi.org/ assay for detecting toxoplasmic encephalitis in patients with AIDS. J. Clin. Microbiol.
10.1017/S0950268813002756. 40, 4499–4503.
Nogueira, PM, Ribeiro, K, Silveira, AC, Campos, JH, Martins-Filho, OA, Bela, SR, Que, X., Wunderlich, A., Joiner, K.A., Reed, S.L, 2004. Toxopain-1 is critical for infection
Campos, MA, Pessoa, NL, Colli, W, Alves, MJ, Soares, RP, Torrecilhas, AC., 2015. in a novel chicken embryo model of congenital toxoplasmosis. Infection and
Vesicles from different Trypanosoma cruzi strains trigger differential innate and immunity 72 (5), 2915–2921. https://doi.org/10.1128/IAI.72.5.2915-2921.2004.
chronic immune responses. J Extracell Vesicles 26 (4), 28734. https://doi.org/ Quiarim, TM, Maia, MM, Cruz, AB, Taniwaki, NN, Namiyama, GM, Pereira-
10.3402/jev.v4.28734. Chioccola, VL., 2021. Characterization of extracellular vesicles isolated from types I,
Oksanen, A, Asbakk, K, Prestrud, KW, Aars, J, Derocher, AE, Tryland, M, Wiig, O, II and III strains of Toxoplasma gondii. Acta Trop 219, 105915. https://doi.org/
Dubey, JP, Sonne, C, Dietz, R, Andersen, M, Born, EW., 2009. Prevalence of 10.1016/j.actatropica.2021.105915.
antibodies against Toxoplasma gondii in polar bears (Ursus maritimus) from Svalbard Ragozo, A.M.A., Yai, L.E.O., Oliveira, L.N., Dias, R.A., Goncalves, H.C., Azevedo, S.S.,
and East Greenland. J Parasitol 95 (1), 89–94. https://doi.org/10.1645/GE-1590.1. Gennari, S.M, 2009. Isolation of Toxoplasma gondii from goats from Brazil. Journal of
Oliveira, JMB, de Almeida, JC, de Melo, RPB, de Barros, LD, Garcia, JL, Andrade, MR, Parasitology 95 (2), 323–326.
Porto, WJN, Regidor-Cerrillo, J, Ortega-Mora, LM, Oliveira, AADF, Mota, RA., 2018 Ramírez-Flores, CJ, Cruz-Mirón, R, Mondragón-Castelán, ME, González-Pozos, S, Ríos-
May. First description of clonal lineage type II (genotype #1) of Toxoplasma gondii in Castro, E, Mondragón-Flores, R.J., 2019. Proteomic and structural characterization
abortion outbreak in goats. Exp Parasitol 188, 21–25. https://doi.org/10.1016/j. of self-assembled vesicles from excretion/secretion products of Toxoplasma gondii.
exppara.2018.03.008. Epub 2018 Mar 8. PMID: 29526575. Proteomics 30 (208), 103490. https://doi.org/10.1016/j.jprot.2019.103490.
Oliveira, PRF, de Melo, RPB, Sierra, TAO, da Silva, RA, da Silva de Oliveira, JE, de Remington, JS, McLeod, R, Desmonts, G., 1995. Toxoplasmosis. editors. In:
Almeida, BG, Mota, RA., 2021. Investigation of soil contaminated with Toxoplasma Remington, JS, Klein, JO (Eds.), Infectious Diseases of the Fetus and New Borne
gondii oocyst in urban public environment, in Brazil. Comp Immunol Microbiol infant. WB Saunders Company, Philadelphia, pp. 140–267.
Infect Dis 79, 101715. https://doi.org/10.1016/j.cimid.2021.101715. DecEpub Remington, JS, McLeod, R, Thulliez, P, Desmonts, G, 2001. Toxoplasmosis. In:
2021 Oct 27. PMID: 34736127. Remington, JS, Klein, JO (Eds.), In Infectious diseases of the fetus and newborn
Ossani, R.A., Borges, H.A.T., Souza, A.P., Sartor, A.A., Miletti, L.C., Federle, M., infant, 5th ed. WB Saunders Company, Philadelphia, PA, pp. 205–346.
Moura, A.B, 2017. Toxoplasma gondii in milk of naturally infected dairy ewes on west Remington, JS, Thulliez, P, Montoya, JG, 2004. Recent developments for diagnosis of
mesoregion of Santa Catarina state, 69. Arquivo Brasileiro de Medicina Veterinária e toxoplasmosis. Journal of clinical microbiology 42 (3), 941–945.
Zootecnia, Brazil, pp. 1294–1300. Rengifo-Herrera, C, Ortega-Mora, LM, Alvarez-García, G, Gómez-Bautista, M, García-
Oz, HS, Tobin, T., 2012. Atovaquone ameliorate gastrointestinal toxoplasmosis Párraga, D, García-Peña, FJ, Pedraza-Díaz, S., 2012. Detection of Toxoplasma gondii
complications in a pregnancy model. Med Sci Monit 18 (9), BR337–BR345. https:// antibodies in Antarctic pinnipeds. Vet Parasitol 23 (1-2), 259–262. https://doi.org/
doi.org/10.12659/msm.883342. 10.1016/j.vetpar.2012.05.020, 190.
Pan, M, Lyu, C, Zhao, J, Shen, B., 2017. Sixty Years (1957-2017) of Research on Ribas, MP, Almería, S, Fernández-Aguilar, X, De Pedro, G, Lizarraga, P, Alarcia-Alejos, O,
Toxoplasmosis in China-An Overview. Front Microbiol 25 (8), 1825. https://doi.org/ Molina-López, R, Obón, E, Gholipour, H, Temiño, C, Dubey, JP, Cabezón, O., 2018.
10.3389/fmicb.2017.01825. Tracking Toxoplasma gondii in freshwater ecosystems: interaction with the invasive
Paraboni, MLR, Costa, DF, Silveira, C, Gava, R, Pereira-Chioccola, VL, Belfort Jr, R, American mink (Neovison vison) in Spain. Parasitol Res 117 (7), 2275–2281.
Commodaro, AG., 2020. A new strain of Toxoplasma gondii circulating in southern https://doi.org/10.1007/s00436-018-5916-5.
Brazil. J Parasit Dis 44 (1), 248–252. https://doi.org/10.1007/s12639-019-01155-x. Ribeiro-Andrade, M, de Crasto Souza Carvalho, J, Amorim da Silva, R, da Conceição
Patz JA, Hahn, M. B. 2013. Climate change and human health: a One Health approach. Carvalho, M, Nascimento Porto, WJ, Mota, RA, 2019. Inter- and intra-genotype
Current topics in microbiology and immunology, 366, 141–171. doi: 10.1016/j. differences in induced cystogenesis of recombinant strains of Toxoplasma gondii
vetpar.2011.12.036. isolated from chicken and pigs. Exp Parasitol 207, 107775. https://doi.org/10.1016/
Paula, NF.D., Dutra, K.S., Oliveira, A.R.D., Santos, D.O.D., Rocha, C.E.V., Vitor, R.W.D. j.exppara.2019.107775. DecEpub 2019 Oct 16. PMID: 31628896.
A., Santos, R.L, 2020. Host range and susceptibility to Toxoplasma gondii infection in Riemann, HP, Meyer, ME, Theis, JH, Kelso, G, Behymer, DE., 1975. Toxoplasmosis in an
captive neotropical and Old-world primates. Journal of Medical Primatology 49 (4), infant fed unpasteurized goat milk. J Pediatr 87, 573–576.
202–210. https://doi.org/10.1111/jmp.12470. Robert-Gangneux, F, Dardé, ML., 2012. Epidemiology of and diagnostic strategies for
Pelloux, H, Dupouy-Camet, J, Derouin, F, et al., 1997. A multicentre prospective study toxoplasmosis [published correction appears in Clin Microbiol Rev. 2012 Jul;25(3):
for the polymerase chain reaction detection of Toxoplasma gondii DNA in blood 583]. Clin Microbiol Rev. 25 (2), 264–296. https://doi.org/10.1128/CMR.05013-11.
samples from 186 AIDS patients with suspected toxoplasmic encephalitis. AIDS 11, Robert-Gangneux, F, Dupretz, P, Yvenou, C, Quinio, D, Poulain, P, Guiguen, C, 2010.
1888–1890. Clinical relevance of placenta examination for the diagnosis of congenital
Pena, HF, Marvulo, MF, Horta, MC, Silva, MA, Silva, JC, Siqueira, DB, 2011. Isolation toxoplasmosis. Pediatr Infect Dis J 29, 33–38.
and genetic characterization of Toxoplasma gondii from a red-handed howler monkey Rodrigues, J., Veigas, S., Rocha, H., Mateus, T.L., 2018. Toxoplasma gondii na Europa e
(Alouatta belzebul), a jaguarundi (Puma yagouaroundi), and a black-eared opossum em Portugal. Instituto Nacional de Saude Doutor Ricardo Jorge 5 (05) vn.
(Didelphis aurita) from Brazil. Vet Parasitol 10, 377–381. https://doi.org/10.1016/j. Ross, RD, Stec, LA, Werner, JC, Blumenkranz, MS, Glazer, L, Williams, GA., 2001.
vetpar.2010.10.015, 175. Presumed acquired ocular toxoplasmosis in deer hunters. Retina 21 (3), 226–229.
Pena, HFJ, Gennari, SM, Dubey, JP, et al., 2008. Population structure and mouse- https://doi.org/10.1097/00006982-200106000-00005.
virulence of Toxoplasma gondii in Brazil. Int. J. Parasitol. 38, 561–569. Rostami, A, Riahi, SM, Fakhri, Y, Saber, V, Hanifehpour, H, Valizadeh, S, Gholizadeh, M,
Pereira, IS, Maia, MM, da Cruz, AB, Telles, JPM, Vidal, JE, Gava, R, Meira- Pouya, RH, Gamble, HR., 2017. The global seroprevalence of Toxoplasma gondii
Strejevitch, CS, Pereira-Chioccola, VL., 2020. Plasma extracellular microRNAs are among wild boars: A systematic review and meta-analysis. Vet Parasitol 15 (244),
related to AIDS/cerebral toxoplasmosis co-infection. Parasite Immunol 42 (4), 12–20. https://doi.org/10.1016/j.vetpar.2017.07.013.
e12696. https://doi.org/10.1111/pim.12696. Rothova, A, de Boer, JH, Ten Dam-van Loon, NH, Postma, G, de Visser, L, Zuurveen, SJ,
Schuller, M, Weersink, AJ, van Loon, AM, de Groot-Mijnes, JD, 2008. Usefulness of

16
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

aqueous humor analysis for the diagnosis of posterior uveitis. Ophthalmology 115 intestine. Experimental parasitology 125 (4), 380–383. https://doi.org/10.1016/j.
(2), 306–311. https://doi.org/10.1016/j.ophtha.2007.05.014. exppara.2010.03.004.
Ruiz, A, Frenkel, JK., 1980. Toxoplasma gondii in Costa Rican cats. Am J Trop Med Hyg Silva, VO, Maia, MM, Torrecilhas, AC, Taniwaki, NN, Namiyama, GM, Oliveira, KC,
29 (6), 1150–1160. https://doi.org/10.4269/ajtmh.1980.29.1150. Ribeiro, KS, Toledo, MDS, Xander, P, Pereira-Chioccola, VL., 2018. Extracellular
Saavedra, GM, Ortega, YR, 2004. Seroprevalence of Toxoplasma gondii in swine from vesicles isolated from Toxoplasma gondii induce host immune response. Parasite
slaughterhouses in Lima, Peru, and Georgia, U.S.A. J Parasitol 90 (4), 902–904. Immunol 40 (9), e12571. https://doi.org/10.1111/pim.12571.
https://doi.org/10.1645/GE-258R. Simon, A., Rousseau, A.N., Savary, S., Bigras-Poulin, M., Ogden, N.H, 2013. Hydrological
Sacks, JJ, Roberto, RR, Brooks, NF, 1982. Toxoplasmosis infection associated with raw modelling of Toxoplasma gondii oocysts transport to investigate contaminated
goat’s milk. J Am Med Assoc 248, 1728–1732. snowmelt runoff as a potential source of infection for marine mammals in the
Santos PS, Albuquerque, G. R., da Silva, V. M., Martin, A. R., Marvulo, M. F., Souza, S. L., Canadian Arctic. Journal of environmental management 127, 150–161. https://doi.
Ragozo, A. M., Nascimento, C. C., Gennari, S. M., Dubey, J. P., & Silva, J. C. 2011. org/10.1016/j.jenvman.2013.04.031.
Seroprevalence of Toxoplasma gondii in free-living Amazon River dolphins (Inia Sims, TA, Hay, J, Talbot, IC., 1989. An electron microscope and immunohistochemical
geoffrensis) from central Amazon, Brazil. Veterinary parasitology, 183(1-2), study of the intracellular location of Toxoplasma tissue cysts within the brains of
171–173. 10.1016/j.vetpar.2011.06.007. mice with congenital toxoplasmosis. British journal of experimental pathology 70
Santos Silva, AC, de Barros, LD, Barros, VMC, et al., 2020. Occurrence of Atypical and (3), 317–325.
New Genotypes of Toxoplasma gondii in Free-Range Chickens Intended for Human Skinner, LJ, Timperley, AC, Wightman, D, Chatterton, JMW, Ho-Yen, DO., 1990.
Consumption in Brazil. Acta Parasitol 65 (3), 774–778. https://doi.org/10.2478/ Simultaneous diagnosis of toxoplasmosis in goats and goatowner’s family. Scand J
s11686-020-00194-2. Infect Dis 22, 359–361.
Santos, SV, Pena, HFJ, Talebi, MG, Teixeira, RHF, Kanamura, CT, Diaz-Delgado, J, Smith, D, Kannan, G, Coppens, I, Wang, F, Nguyen, HM, Cerutti, A, Olafsson, EB,
Gennari, SM, Catão-Dias, JL., 2018. Fatal toxoplasmosis in a southern muriqui Rimple, PA, Schultz, TL, Mercado Soto, NM, Di Cristina, M, Besteiro, S,
(Brachyteles arachnoides) from São Paulo state, Brazil: Pathological, Carruthers, VB, 2021. Toxoplasma TgATG9 is critical for autophagy and long-term
immunohistochemical, and molecular characterization. J Med Primatol 47 (2), persistence in tissue cysts. Elife 27 (10), e59384. https://doi.org/10.7554/
124–127. https://doi.org/10.1111/jmp.12326. AprEpub 2017 Nov 29. PMID: eLife.59384.
29193137. Smith, NC, Goulart, C, Hayward, JA, Kupz, A, Miller, CM, van Dooren, GG., 2021.
Santos, TR., Faria, GD., Guerreiro, B.M., Dal Pietro, N.H., Lopes, W.D., Silva, H.M., Control of human toxoplasmosis. Int J Parasitol 1 (2-3), 95–121. https://doi.org/
Garcia, J.L., Luvizotto, M.C., Bresciani, K.D., da Costa, A.J., 2016. Congenital 10.1016/j.ijpara.2020.11.001.
Toxoplasmosis in Chronically Infected and Subsequently Challenged Ewes. PloS one Soares, B, Cantos, GA., 2006. Detecção de estruturas parasitárias em hortaliças
11 (10), e0165124. https://doi.org/10.1371/journal.pone.0165124. comercializadas na cidade de Florianópolis, SC, Brasil. Rev Bras Cien Farm 42,
Schares, G, Koethe, M., Bangoura, B., Geuthner, A.-C., Randau, F., Ludewig, M., et al., 455–460.
2018. Toxoplasma gondii infections in chickens - performance of various antibody Sogorb, F, Jamra, LF, Guimaraes, EC., Toxoplasmose em animais de São Paulo, Brasil,
detection techniques in serum and meat juice relative to bioassay and DNA detection 1977. Toxoplasmosis in animals of Sao Paulo, Brazil]. Rev Inst Med Trop Sao Paulo
methods. Int. J. Parasitol. 48, 751–762. https://doi.org/10.1016/j. 19 (3), 191–194. Portuguese. PMID: 601428.
ijpara.2018.03.007. Sommer, R, Rommel, M, Levetzov, R., 1965. Die überlebensdauer von toxoplasmazysten
Schares, G, Vrhovec, MG, Pantchev, N, Herrmann, DC, Conraths, FJ, 2008. Occurrence of in fleisch und fleischzubereitungen. Fleischwirtsch 45, 454–457.
Toxoplasma gondii and Hammondia hammondi oocysts in the faeces of cats from Sroka, J, Karamon, J, Wójcik-Fatla, A, Piotrowska, W, Dutkiewicz, J, Bilska-Zając, E,
Germany and other European countries. Vet Parasitol 25 (1-2), 34–45, 152. Zając, V, Kochanowski, M, Dąbrowska, J, Cencek, T, 2020. Toxoplasma gondii
Schlüter, D, Kaefer, N, Hof, H, et al., 1997. Expression pattern and cellular origin of infection in slaughtered pigs and cattle in Poland: seroprevalence, molecular
cytokines in the normal and Toxoplasma gondii-infected murine brain. Am. J. Pathol. detection and characterization of parasites in meat. Parasit Vectors 4 (1), 223.
150, 1021–1035. https://doi.org/10.1186/s13071-020-04106-1, 13.
Schlüter, D, Kwok, LY, Lütjen, S, et al., 2003. Both lymphotoxin-alpha and TNF are Su, C, Dubey, J.P., 2020. Isolation and genotyping of Toxoplasma gondii strains. Methods
crucial for control of Toxoplasma gondii in the central nervous system. J. Immunol. in Molecular Biology 2071, 49–80.
170 (12), 6172–6182. Su, C, Evans, D, Cole, R, et al., 2003. Recent expansion of Toxoplasma through enhanced
Schoondermark-Van de Ven, E., Melchers, W., Camps, W., Eskes, T., Meuwissen, J., oral transmission. Science 299, 414–416.
Galama, J., 1994. Effectiveness of spiramycin for treatment of congenital Toxoplasma Su, C, Khan, A, Zhou, P, Majumdar, D, Ajzenberg, D, Dardé ML, et al, 2012. Globally
gondii infection in rhesus monkeys. Antimicrobial agents and chemotherapy 38 (9), diverse Toxoplasma gondii isolates comprise six major clades originating from a
1930–1936. https://doi.org/10.1128/AAC.38.9.1930. small number of distinct ancestral lineages. Proc Natl Acad Sci USA 109, 5844-5849.
Schorey, JS, Cheng, Y, Singh, PP, Smith, VL., 2015. Exosomes and other extracellular doi: 10.1073/pnas.1203190109.
vesicles in host-pathogen interactions. EMBO Rep 16 (1), 24–43. https://doi.org/ Subauste, C., 2012. Animal models for Toxoplasma gondii infection. Curr Protoc Immunol.
10.15252/embr.201439363. Chapter 19, 11–23. Unit 19 13.
Schott, KC, Krusor, C, Tinker, MT, Moore, J, Conrad, PA, Shapiro, K, 2016. Concentration Suijkerbuijk, A., van Gils, P.F., Bonačić Marinović, A.A., Feenstra, T.L., Kortbeek, L.M.,
and retention of Toxoplasma gondii surrogates from seawater by red abalone (Haliotis Mangen, M.J., Opsteegh, M., de Wit, G.A., van der Giessen, J., 2018. The design of a
rufescens). Parasitology 143 (13), 1703–1712. https://doi.org/10.1017/ Social Cost-Benefit Analysis of preventive interventions for toxoplasmosis: An
S0031182016001359. example of the One Health approach. Zoonoses and public health 65 (1), 185–194.
Schwab, A, Meyering, SS, Lepene, B, et al., 2015. Extracellular vesicles from infected https://doi.org/10.1111/zph.12417.
cells: potential for direct pathogenesis. Front Microbiol 20, 1132 eCollection 2015. Sullivan Jr, WJ, Jeffers, V., 2012. Mechanisms of Toxoplasma gondii persistence and
Shah, H.A., Huxley, P., Elmes, J., Murray, K.A., 2019. Agricultural land-uses consistently latency. FEMS Microbiol Rev 36 (3), 717–733. https://doi.org/10.1111/j.1574-
exacerbate infectious disease risks in Southeast Asia. Nat. Commun. 10 https://doi. 6976.2011.00305.x.
org/10.1038/s41467-019-12333-z. Sun, X., Wang, Z., Li, J., Wei, F., Liu, Q., 2015. Evaluation of an indirect ELISA using
Shapiro, K, Krusor, C, Mazzillo, FF, Conrad, PA, Largier, JL, Mazet, JA, Silver, MW., recombinant granule antigen GRA1, GRA7 and soluble antigens for serodiagnosis of
2014. Aquatic polymers can drive pathogen transmission in coastal ecosystems. Proc Toxoplasma gondii infection in chickens. Research in veterinary science 100,
Biol Sci 22 (1795), 20141287. https://doi.org/10.1098/rspb.2014.1287, 281. 161–164. https://doi.org/10.1016/j.rvsc.2015.04.011.
Shariatzadeh, SA, Sarvi, S, Hosseini, SA, Sharif, M, Gholami, S, Pagheh, AS, Montazeri, F, Suzuki, Y, Conley, FK, Remington, JS., 1989. Importance of endogenous IFN-gamma for
Nayeri, T, Nakhaei, M, Mikaeili Galeh, T, Daryani, A, 2021. The global prevention of toxoplasmic encephalitis in mice. Journal of Immunology 15 (6),
seroprevalence of Toxoplasma gondii infection in bovines: a systematic review and 2045–2050, 143.
meta-analysis. Parasitology 30, 1–17. https://doi.org/10.1017/ Suzuki, Y, Wong, SY, Grumet, FC, Fessel, J, Montoya, JG, Zolopa, AR, Portmore, A,
S0031182021001116. Schumacher-Perdreau, F, Schrappe, M, Köppen, S, Ruf, B, Brown, BW,
Sharif, M, Amouei, A, Sarvi, S, et al., 2017. Genetic diversity of Toxoplasma gondii isolates Remington, JS., 1996. Evidence for genetic regulation of susceptibility to
from ruminants: A systematic review. Int J Food Microbiol 258, 38–49. https://doi. toxoplasmic encephalitis in AIDS patients. J Infect Dis.
org/10.1016/j.ijfoodmicro.2017.07.007. Tavassoli, M., Esmaeilnejad, B., Malekifard, F., Soleimanzadeh, A., Dilmaghani, M.,
Sibley, LD, Ajioka, JW., 2008. Population structure of Toxoplasma gondii: clonal 2013. Detection of Toxoplasma gondii DNA in Sheep and Goat Milk in Northwest of
expansion driven by infrequent recombination and selective sweeps. Annu. Rev. Iran by PCR-RFLP. Jundishapur Journal of Microbiology 6 (10).
Microbiol. 62, 329–351. Tenter, AM, Heckeroth, AR, Weiss, LM., 2000. Toxoplasma gondii: from animals to
Sibley, LD, Boothroyd, CJ, 1992. Virulent strains of T. gondii comprise a single clonal humans. Int J Parasitol 30 (12-13), 1217–1258. https://doi.org/10.1016/s0020-
lineage. Nature 359, 82–85. 7519(00)00124-7.
Sibley, LD., 2003. Toxoplasma gondii: perfecting an intracellular life style. Traffic 4 (9), Tenter, AM., 2009. Toxoplasma gondii in animals used for human consumption. Mem Inst
581–586. https://doi.org/10.1034/j.1600-0854.2003.00117.x. Oswaldo Cruz 104 (2), 364–369.
Sibley, L.D., Khan, A., Ajioka, J.W., Rosenthal, B.M., 2009. Genetic diversity of Théry, C, Ostrowski, M, Segura, E., 2009. Membrane vesicles as conveyors of immune
Toxoplasma gondii in animals and humans. Philosophical transactions of the Royal responses. Nat Rev Immunol 9 (8), 581–593. https://doi.org/10.1038/nri2567.
Society of London. Series B, Biological sciences 364 (1530), 2749–2761. https://doi. Théry, C, Witwer, KW, Aikawa, E, Alcaraz, MJ, Anderson, JD, Andriantsitohaina, R,
org/10.1098/rstb.2009.0087. et al., 2018. Minimal information for studies of extracellular vesicles 2018
Silva, JG, Alves, BHL, Melo, RPB, Kim, PC P, Neto, OLS, Bezerra, MJG, Mota, RA, 2015. (MISEV2018): a position statement of the International Society for Extracellular
Occurrence of anti-Toxoplasma gondii antibodies and parasite DNA in raw milk of Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 7 (1),
sheep and goats of local breeds reared in Northeastern Brazil. Acta tropica 142, 1535750.
145–148. Thompson, RCA., 2013. Parasite zoonoses and wildlife: One Health, spillover and human
Silva, P, Shiraishi, CS, Silva, AV, Gonçalves, GF, Sant’Ana, D., Araújo, E.J., 2010. activity. Int J Parasitol 43, 1079–1088. https://doi.org/10.1016/j.
Toxoplasma gondii: a morphometric analysis of the wall and epithelial cells of pig ijpara.2013.06.007.

17
R.A.M. de Barros et al. Acta Tropica 231 (2022) 106432

Titilincu, A., Mircean, V., Iovu, A., Cozma, V., 2009. Development of an indirect ELISA Paulo. Brazil. Rev. Inst. Med. Trop.Sao Paulo 50, 209–212. https://doi.org/10.1590/
test using tachyzoite crude antigen for sero-diagnosis of sheep Toxoplasma gondii S0036-46652008000400004.
infection. Bulletin UASVM Veterinary Medicine 66 (2). Vidal, JE, Penalva de Oliveira, AC, Hernandez, AV., 2003. Prognostic factors of clinical
Tkach, M, Théry, C., 2016. Communication by extracellular vesicles: where we are and response in patients with toxoplasmic encephalitis and AIDS in the HAART era.
where we need to go. Cell 10, 1226–1232. J Neurovirol 60, 16–17.
Torrecilhas, AC, Schumacher, RI, Alves, MJ, Colli, W., 2012. Vesicles as carriers of Vidal, JE, Penalva de Oliveira, AC, Pellegrino, D, et al., 2011. Complicações neurológicas
virulence factors in parasitic protozoan diseases. Microbes Infect 14 (15), em pacientes infectados pelo HIV e fatores associados a óbito na era HAART: estudo
1465–1474. https://doi.org/10.1016/j.micinf.2012.07.008. observacional prospectivo no Instituto de Infectologia Emílio Ribas. Serviços de
Torrecilhas AC, Soares R P, Schenkman S, Fernández-Prada C, Olivier M. 2020. Infectologia e Neurologia do Instituto de Infectologia. Bras. J. infec Dis. 15 (suppl.1),
Extracellular Vesicles in Trypanosomatids: Host Cell Communication. Frontiers in 23. https://doi.org/10.1590/S0104-42302010000200027.
cellular and infection microbiology, 10, 602502. 10.3389/fcimb.2020.602502. Vidal, JE, Pereira-Chioccola, VL, 2021. Cerebral toxoplasmosis in people living with
Trocoli Torrecilhas, AC, Tonelli, RR, Pavanelli, WR, da Silva, JS, Schumacher, RI, de HIV/AIDS. In: Martins-Duarte, E.S., Adesse, D. (Eds.), Toxoplasma gondii – Biology
Souza, W, E Silva, NC, de Almeida Abrahamsohn, I, Colli, W, Manso Alves, MJ, 2009. and role in health and disease. Nova Medicine & Health, New York, pp. 277–296.
Trypanosoma cruzi: parasite shed vesicles increase heart parasitism and generate an Vieira, FP, Alves, MG, Martins, LM, et al., 2015. Waterborne toxoplasmosis investigated
intense inflammatory response. Microbes Infect 11 (1), 29–39. https://doi.org/ and analyzed under hydrogeological assessment: new data and perspectives for
10.1016/j.micinf.2008.10.003. further research. Mem Instituto Oswaldo Cruz 110, 929–935.
Tumurjav, B., Terkawi, M.A., Zhang, H., Zhang, G., Jia, H., Goo, Y.K., 2010. Wang, J, Liu, X, Jia, B, et al., 2012. A comparative study of small RNAs in Toxoplasma
Serodiagnosis of ovine toxoplasmosis in Mongolia by an enzyme-linked gondii of distinct genotypes. Parasit Vectors 3, 186, 10.1doi186/1756-3305-5-186.
immunosorbent assay with recombinant Toxoplasma gondii matrix antigen 1. The Weiss, LM, Kim, K (Eds.), 2020. TOXOPLASMA GONDII - The Model Apicomplexan—
Japanese journal of veterinary research 58, 111–119. perspectives and Methods, 3 ed. Elsevier. https://doi.org/10.1016/j.
Twu, O, Johnson, PJ., 2014. Parasite extracellular vesicles: mediators of intercellular ijpara.2003.12.009.
communication. PLoS Pathog 10 (8), e1004289. https://doi.org/10.1371/journal. Wendte, JM, Gibson, AK, Grigg, ME., 2011. Population genetics of Toxoplasma gondii:
ppat.1004289. new perspectives from parasite genotypes in wildlife. Vet Parasitol 182 (1), 96–111.
Unzaga, JM, Moré, G, Bacigalupe, D, Rambeaud, M, Pardini, L, Dellarupe, A, De Felice, L, https://doi.org/10.1016/j.vetpar.2011.07.018.
Gos, ML, Venturini, MC., 2014. Toxoplasma gondii and Neospora caninum infections Wilking, H., Thamm, M., Stark, K., et al., 2016. Prevalence, incidence estimations and
in goat abortions from Argentina. Parasitol. Int. 63, 865–867. https://doi.org/ risk factors of Toxoplasma gondii infection in Germany: a representative, cross-
10.1016/j.parint.2014.07.009. sectional, serological study. Sci Rep 6, 22551. https://doi.org/10.1038/srep22551.
Vallochi, AL, Muccioli, C, Martins, MC, Silveira, C, Belfort Jr, R, et al., 2005. The Witter, R, Pena, HFJ, MO, Maia, de Magalhães, AO, Morgado, TO, Colodel, EM,
genotype of Toxoplasma gondii strains causing ocular toxoplasmosis in humans in Barros, DA, Igarashi, M, Gennari, SM, Pacheco, RC., 2020. Isolation and genotyping
Brazil. Am J Ophthalmol 139, 350–351. of Toxoplasma gondii in the Midwestern Brazil revealed high genetic diversity and
VanWormer, E, Conrad, PA, Miller, MA, Melli, AC, Carpenter, TE, Mazet, JA., 2013. new genotypes. Acta Trop 212, 105681. https://doi.org/10.1016/j.
Toxoplasma gondii, source to sea: higher contribution of domestic felids to terrestrial actatropica.2020.105681. DecEpub 2020 Sep 11. PMID: 32926845.
parasite loading despite lower infection prevalence. Ecohealth 10 (3), 277–289. Wowk, PF, Zardo, ML, Miot, HT., Goldenberg, S, Carvalho, PC, Mörking, PA., 2017.
https://doi.org/10.1007/s10393-013-0859-x. Proteomic profiling of extracellular vesicles secreted from Toxoplasma gondii.
VanWormer, E, Fritz, H, Shapiro, K, Mazet, JA, Conrad, PA., 2013a. Molecules to Proteomics 17 (15-16). https://doi.org/10.1002/pmic.201600477.
modeling: Toxoplasma gondii oocysts at the human-animal-environment interface. Xiao, J, Yolken, RH, 2015. Strain hypothesis of Toxoplasma gondii infection on the
Comp Immunol Microbiol Infect Dis 36 (3), 217–231. https://doi.org/10.1016/j. outcome of human diseases. Acta Physiol (Oxf) 213 (4), 828–845. https://doi.org/
cimid.2012.10.006. MayEpub 2012 Dec 4. PMID: 23218130; PMCID: PMC3779781. 10.1111/apha.12458.
Vargas-Villavicencio, JA, Besné-Mérida, A, Correa, D., 2016. Vertical transmission and Yai, LE, Ragozo, AM, Soares, RM, Pena, HF, Su, C, Gennari, SM, 2009. Genetic diversity
fetal damage in animal models of congenital toxoplasmosis: A systematic review. Vet among capybara (Hydrochaeris hydrochaeris) isolates of Toxoplasma gondii from
Parasitol 223, 195–204. https://doi.org/10.1016/j.vetpar.2016.04.024. Brazil. Vet Parasitol 10 (3-4), 332–337. https://doi.org/10.1016/j.
Vidal, JE, Dauar, RF, Penalva de Oliveira, AC., 2008. Utility of brain biopsy in patients vetpar.2009.03.007, 162.
with acquired immunodeficiency syndrome before and after introduction of highly Ybañez, R., Ybañez, A.P., Nishikawa, Y., 2020. Review on the Current Trends of
active antiretroviral therapy. Neurosurgery 63. Toxoplasmosis Serodiagnosis in Humans. Frontiers in cellular and infection
Vidal, JE, Hernandez, AV, Penalva de Oliveira, AC, Dauar, R, Barboza, SP, Focaccia, R, microbiology 10, 204. https://doi.org/10.3389/fcimb.2020.00204.
2005. Cerebral toxoplasmosis in HIV-Positive patients in Brazil: clinical features and Zakimi, S, Kyan, H, Oshiro, M, et al., 2006. Genetic characterization of GRA6 genes from
predictors of treatment response in the HAART era. AIDS Patient Care STDS 19, Toxoplasma gondii from pigs in Okinawa. Japan. J. Vet. Med. Sci. 68, 1105–1107.
840–848. Zenner, L, Darcy, F, Capron, A, Cesbron-Delauw, MF., 1998. Toxoplasma gondii: kinetics
Vidal, JE, Oliveira, AC., 2013. AIDS-related cerebral toxoplasmosis in São Paulo State, of the dissemination in the host tissues during the acute phase of infection of mice
Brazil: marked improvements in the highly active antiretroviral therapy-era but the and rats. Exp Parasitol 90 (1), 86–94. https://doi.org/10.1006/expr.1998.4301.
challenges continue. Braz J Infect Dis 17, 379–380. https://doi.org/10.1016/j. Zhao, XY, Ewald, SE., 2020. The molecular biology and immune control of chronic
bjid.2012.10.030. Toxoplasma gondii infection. The Journal of clinical investigation 130 (7),
Vidal, JE, Penalva de Oliveira, AC, Fink, MC, et al., 2008. Aids-related progressive 3370–3380. https://doi.org/10.1172/JCI136226.
multifocal leukoencephalopathy: a retrospective study in a referral center in São Zinsstag, J., 2012. Convergence of EcoHealth and One Health. EcoHealth 9 (4), 371–373.
https://doi.org/10.1007/s10393-013-0812.

18

You might also like