Previous Article in Journal
Human–Chelonian Bond in Italy: An Exploratory Study of Pet Turtle and Tortoise Ownership
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Review of Toxoplasmosis: What We Still Need to Do

by
Muhammad Farhab
1,2,†,
Muhammad Waqar Aziz
3,†,
Aftab Shaukat
4,*,
Ming-Xing Cao
1,5,
Zhaofeng Hou
1,2,
Si-Yang Huang
1,2,
Ling Li
1,2 and
Yu-Guo Yuan
1,2,*
1
College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
2
Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
3
Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore 54600, Pakistan
4
College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
5
College of Veterinary Medicine, Shanxi Agricultural University, Taiyuan 030800, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Vet. Sci. 2025, 12(8), 772; https://doi.org/10.3390/vetsci12080772
Submission received: 11 April 2025 / Revised: 24 July 2025 / Accepted: 8 August 2025 / Published: 18 August 2025

Simple Summary

Toxoplasma gondii is the causative agent of toxoplasmosis, a zoonotic disease infecting nearly one-third of humans worldwide. Although often asymptomatic in healthy individuals, it poses serious risks to immunocompromised patients, pregnant women, and certain animal species, leading to severe neurological, ocular, or fetal complications. Transmission occurs through contaminated food/water (oocysts), undercooked meat (tissue cysts), or congenital/transplant routes. Diagnosis relies on serology and PCR, with treatment (e.g., pyrimethamine-sulfadiazine) reserved for high-risk cases. Despite promising antigen candidates, no human vaccine exists due to the parasite’s complex biology; the only licensed vaccine prevents abortions in sheep. This review highlights current knowledge on toxoplasmosis, emphasizing its global health impact and unmet medical needs.

Abstract

Toxoplasma gondii is responsible for the disease toxoplasmosis and has the broadest host range among apicomplexan parasites, as it infects virtually all warm-blooded vertebrates. Toxoplasmosis is a zoonotic and emerging public health concern with considerable morbidity and mortality, especially in the developing world, affecting approximately one-third of the world’s human population. Clinical presentation varies among species, and the infection establishes lifelong chronicity in hosts. Most of the host species (including healthy humans) are asymptomatic on the one hand, it is fatal to marsupials, neotropical primates and some marine mammals on the other hand. In immunocompetent humans, infection is typically asymptomatic, whereas immunocompromised individuals may develop disseminated disease affecting virtually any organ system—most commonly reproductive, cerebral, and ocular systems. Toxoplasmosis spreads by ingestion of food or water contaminated with T. gondii oocysts, consumption of undercooked/raw meat containing tissue cysts, transplacental transmission from mother to fetus, or by receiving infected organ/blood from the infected individual. Toxoplasmosis is mainly diagnosed by serologic tests and polymerase chain reaction (PCR). It is treated with pyrimethamine combined with sulfadiazine or clindamycin, often supplemented with leucovorin, atovaquone, and dexamethasone. Despite having many potent anti-T. gondii antigenic candidates, there is no commercially available vaccine for humans due to many factors, including the complex life cycle of the parasite and its evasion strategies. To date, the only commercially available anti-T. gondii vaccine is for sheep, licensed for veterinary use to prevent ovine abortions. In this review, we have summarized the current understanding of toxoplasmosis.

1. Introduction

The Greek word “Toxoplasma” means “arc-shaped body”, and was named by Nicolle and Manceaux for its arc-shaped tachyzoites [1]. It is an obligate intracellular protozoan that causes toxoplasmosis in warm-blooded animals (including humans, other mammals, and birds) worldwide. Although it can be isolated from cold-blooded animals, it does not parasitize them [2,3,4]. Unlike closely related parasites, which typically have narrow host ranges, Toxoplasma gondii has a remarkably broad distribution. Felids (family Felidae) are the only definitive hosts, and sexual reproduction occurs in their small intestine, resulting in the excretion of oocysts into the environment [2]. Toxoplasma gondii has medical and veterinary importance and is used as a model for apicomplexan organisms. Toxoplasma gondii can be grown and maintained in virtually any warm-blooded cell line and in mice. Additionally, T. gondii is readily amenable to genetic manipulation and has high transfection efficiency [5]. Even though most species and immunocompetent individuals are immune to this infection, the fact that it can infect virtually all mammals has gained increasing attention as an excellent model system to study host–pathogen interactions [6,7]. It infects up to one-third of the world’s human population and is the second most common asymptomatic intrauterine infection worldwide [7].
Over the past decades, there have been several breakthroughs in human and animal toxoplasmosis. The objective of this review is to concisely summarize the current trends in human and animal toxoplasmosis with special emphasis on the epidemiology of the disease, including its global distribution, pathophysiology, clinical presentations, diagnosis, treatment, and preventive measures. It also aims to provide an overview of the present understanding of T. gondii vaccine development and the challenges that are limiting its commercial availability for human use. As far as molecular epidemiology is concerned, we have considered geographical mapping. Toxoplasma gondii PopSet sequences were accessed on 6 June 2025 [8]. From the extensive literature on toxoplasmosis treatment, we drafted a summary of toxoplasmosis. The treatment regimens were specially emphasized on drug selection rather than drug doses, dose intervals, mechanism of action, etc.

2. History

2.1. Etiology

The name “Toxoplasma gondii” was proposed based on its morphology and the host Ctenodactylus gundi (originally misidentified as C. gondi) [9]. In 1937, T. gondii was isolated from guinea pig brains and maintained in the laboratory through serial mouse passage [10]. In 1939, T. gondii was isolated from a fatal case of human infantile encephalitis [11]. This isolate, later designated the RH strain, became the prototype for studying acute toxoplasmosis due to its consistent lethality in laboratory animals. In 1941, T. gondii isolates from humans and animals were established as the same species. In 1992, T. gondii was classified based on infection in mice as Type I, II, III, and atypical. The T. gondii genome was mapped in 2005 [12]. Recent studies reported that the T. gondii Regulator of Cystogenesis 1 gene is required for the formation of T. gondii bradyzoites [9,12,13].

2.2. Historical Background of the Disease in Animals

Toxoplasma gondii was first reported in 1908 in tissue of Ctenodactylus gundi in Tunisia. In 1910 in Italy, there was the first report of fatal toxoplasmosis in a dog that died of acute visceral toxoplasmosis. The first case of toxoplasmosis in a cat was reported in 1942 in the Middletown, New York, USA. Toxoplasmosis in sheep was discovered in 1957 from aborted fetuses. Cattle and horses are resistant to clinical T. gondii, and there is no confirmed case of clinical toxoplasmosis in either of these animals. Cattle are considered resistant to clinical toxoplasmosis and viable T. gondii is rarely isolated from bovine tissues.
In 1954, Hartley et al. [14] observed ovine abortions and isolated T. gondii-like bodies from fetal membrane lesions. They termed these “New Zealand type II abortions”, but cautioned that attributing causality to T. gondii was premature. In 1957, Buxton [15] inoculated sheep with gauze-filtered suspension of fetal cotyledons from the field cases of T. gondii-associated abortion, and then they successfully isolated T. gondii from these inoculated sheep. They proved that the cause of New Zealand Type II Abortion is toxoplasmosis [14,15].
T. gondii was isolated for the first time from sea otters in 2000 [9,12,13]. In 2012, Dubey et al. [16] reported the first confirmed case of acute toxoplasmosis in cattle, which died of acute toxoplasmosis in New Zealand. Diagnosis was confirmed by presence of numerous tissue cysts, which was confirmed by immunohistochemical staining with bradyzoite-specific T. gondii antibodies (BAG1) [16].
Kimble et al. [17] presented the first report of clinical toxoplasmosis in horses. An adult American Quarter Horse gelding with a history of weight loss presented with an acute onset of colic, fever, soft faeces, and elevated liver enzymes. Within and surrounding necrotic areas were free and intrahistiocytic clusters of protozoal tachyzoites. Similar but milder inflammation was evident in the spleen, lungs, and liver. Necrotizing typhlitis was also evident. Immunolabelling for T. gondii was positive and the ultrastructural morphology of the protozoa was compatible with T. gondii [17,18].

2.3. Parasite Morphology and Life Cycle

In 1960, it was discovered that cystic organisms (later termed bradyzoites) resist pepsin and trypsin digestion [19]. In 1970, oocysts were discovered, and both asexual and sexual stages were identified in intestine of cats. In 1972, the asexual enteroepithelial stages were designated as types A–E. The term “bradyzoite”, also called cystozoites, was proposed in 1973 for the cystic organisms. In 1976, it was found that inoculation of tachyzoites (alternatively called trophozoite, feeding form, proliferative form, endodyozoite) to mice leads to tissue cysts formation as early as 3 days, and cats can shed oocysts for up to 18 days from the day of infection. A summary of milestones of T. gondii in animals is presented in Table 1.

2.4. Notable Outbreaks/Epizootics in Domestic and Wild Species

In 1954, New Zealand experienced lamb abortion rates of 15-20%, initially suspected to result from toxoplasmosis [14]. Although Hartley et al. [14] successfully isolated T. gondii from the samples of aborted samples, they were not sure that T. gondii was the actual cause until it was confirmed by Buxton [15], by experimentally infecting sheep in 1957. During the lambing season of 2005, a toxoplasmosis abortion storm occurred in a flock of purebred Suffolk ewes on a farm in Texas, USA. Only 14 healthy lambs were born, and 38 abortuses, mummies, and weak or stillborn lambs were delivered. Another 15 fetuses identified by ultrasound were presumably resorbed or were aborted undetected. In the 2006 lambing season, two of 26 ewes delivered T. gondii-infected lambs. Toxoplasma gondii was isolated from the brain and heart of a lamb from the second ewe. In subsequent seasons, the ewes lambed normally. The results of the present study support the hypothesis that most sheep that have aborted due to T. gondii develop protection against future toxoplasmosis-induced abortion, but the protection is not absolute [83]. From 2015 to 2017, investigations identified 22 ovine abortion outbreaks in 20 Spanish farms, and viable T. gondii were isolated from 11 of these 22 outbreaks [84]. Representative samples were collected from abortion outbreaks (n = 31) and from chronically infected adult sheep at slaughterhouses (n = 50) in different Spanish regions, and bioassayed in mice. As a result, 30 isolates were obtained: 10 from abortion-derived samples and 20 from adult myocardial tissues [84]. Gabardo et al. [85] described an outbreak of T. gondii in a sheep herd in 2010 in Brazil. A total of 40 out of 100 pregnant ewes were aborted in the last month of pregnancy or had stillbirths. The fetal central nervous system has intralesional cysts positive for T. gondii by immunohistochemistry [85]. Recently, a study in Spain described two outbreaks of abortions, one affecting a dairy flock (abortions in 30 of 239 pregnant ewes) and the other affecting a meat flock, abortions in 70 of 210 pregnant ewes [86]. In 1979 [74] reported congenital toxoplasmosis in three flocks of sheep from Tasmania as follows. (i) In December 1973, three does presented premature parturition or stillbirth, and their kids were seropositive for T. gondii through IFAT at 1:16 dilution. No significant reactions were obtained in brucellosis and leptospirosis agglutination tests. (ii) In 1976, there was a case of many abortions or stillbirths and in October–November 1977, five stillbirths from the same flock. Dam was T. gondii IFAT positive with titre of 1:512. (iii) In 1978, abortion in an Angora goat resulted in twin kids. Toxoplasma gondii from one of these kids was successfully isolated through a mouse bioassay.
In 2001, abortions and stillbirths were seen in a pig farm from Jiangxi Province; 42.0% of pigs were affected with 8.0% mortality. Toxoplasma gondii tachyzoites were isolated from many organs under a light microscope, and from mice inoculated with tissues of dead pigs [5]. In another outbreak from a farm in Guangzhou city, sows in late pregnancy aborted. A total of 33.0% of sows were affected with 2.0% mortality. Toxoplasma gondii was recovered from mice inoculated with tissues of a dead sow [5]. In 2004, 19 pigs from a farm containing 260 sows and 960 fattening pigs in Jinchang, Gansu Province, China died. Tachyzoites of T. gondii were found in body fluids of mice inoculated intraperitoneally with ground samples from the heart, liver, spleen, and brain of two sick pigs. In addition, the inoculation of five pigs with T. gondii tachyzoites caused death in two of the pigs [87]. During October 1994, there were four outbreaks of disease with mortality in different pig herds in two different provinces, respectively, Lombardia and Emilia-Romagna regions (North Italy). The morbidity was high (50–60%), and mortality ranged from 10% to 42% [88].
Hector’s dolphins (Cephalorhynchus hectori), a small, endangered coastal species, are endemic to New Zealand. In 2013, Roe et al. [89] found that 7 of 28 (25%) dolphins died due to disseminated toxoplasmosis, including 2 of 3 Maui’s dolphins, a critically endangered sub-species [89]. Toxoplasma gondii infection was detected by Dubey et al. [90] in bottlenose dolphins (Tursiops truncatus) from a sea aquarium in Canada in 2009. All of them were seropositive; among them, two dolphins died. Toxoplasma gondii tissue cysts were identified in histological sections of the brain of one dolphin. Another dolphin did not have visible organisms, but viable T. gondii was isolated by bioassay in mice and cats from its brain and skeletal muscle [90].
During 2001–2015, 14 out of 183 seals diagnosed to be infected by protozoal infections by histopathology, of which (n = 8) were confirmed cases of T. gondii-related mortality [91], including the one described by [14]. Toxoplasma gondii was associated with two cases of suspected cases among these marine mammals [91].
In 1964, McAllister et al. [92] reported an outbreak of toxoplasmosis that killed 44 of a herd of 56 chinchillas and produced four abortions with the loss of a total of five fetuses. A wild mouse thought to be the house mouse (Mus musculus), caught in a trap in the basement housing the chinchillas, was found to be infected with T. gondii [92].
An epizootic of toxoplasmosis occurred among 22 adult and 30 kit black-footed ferrets (Mustela nigripes) maintained under quarantine conditions at the Louisville Zoological Garden (Kentucky, USA) in 1992. Two adults and six kits died with acute disease. Toxoplasmosis was confirmed by immunohistochemical staining. Chronic toxoplasmosis resulted in the death of an additional 13 adult ferrets [93].
Between 2006 and 2010, Budapest Zoo and Botanical Garden of Hungary faced a fatal toxoplasmosis outbreak in Tammar wallabies (Macropus eugenii), which led to the death of six animals. Toxoplasma gondii was confirmed through immunohistochemical examination. In another four specimens, histopathology revealed T. gondii-like organisms (which could not be differentiated from Neospora caninum solely by morphology), and in another 11 animals, toxoplasmosis was the possible cause of death. The current zoo population of 12 Tammar wallabies was tested for T. gondii IgG antibodies by the modified agglutination test (MAT), with negative results [94].
In 2017, Guthrie et al. [95] described an outbreak of fatal toxoplasmosis in nine red-necked wallabies (Macropus rufogriseus) (RNW) [95]. In 2018, Verma et al. [96] isolated T. gondii from brains and/or muscles of 22 of 44 northern sea otters (Enhydra lutris kenyoni) from Washington State, USA. The isolates were bioassayed in mice and viable T. gondii were isolated [96]. In the spring of 1999, a large Wisconsin mink (Mustela vison) farm experienced an outbreak of abortions, stillborn kits, and kit mortality. A total of 7,800/1,976 females lost their entire litter either from abortion or neonatal mortality, respectively. Overall kit mortality was 10,408. Toxoplasmosis was diagnosed by the detection of T. gondii tachyzoites by immunohistochemistry [97].
In 1999, Bouer reported fatal outbreak of toxoplasmosis in a Zoo in Brazil in three wooly monkeys (Lagothrix lagotricha) [98]. Carme et al. [99] reported outbreaks of toxoplasmosis in two episodes in 2001 and 2006 in a captive breeding colony of squirrel monkeys (Saimiri sciureus) in the Institut Pasteur in French Guiana. During both outbreaks, a total of 50 monkeys died and none recovered spontaneously [99]. In 2019, four squirrel monkeys (Saimiri sp.) died in Japan, followed by seven of thirteen dying in South Korea in 2018.
In another outbreak seven howler (Alouatta sp.) monkeys died of disseminated acute toxoplasmosis during 34 days, with intervals of 2–15 days between deaths from Brazil. These monkeys were kept in two side-by-side enclosures, suggesting in-contact transmission [100].
Seven of 30 canaries in an aviary in New Zealand developed ophthalmic problems. Clinically, five birds had unilateral and two birds had bilateral lesions characterized by conjunctivitis, crusty exudates on eyelids, and collapse of the eyeball. Numerous T. gondii tachyzoites were seen in the detached retina and vitreous humor of acutely affected birds. The diagnosis of toxoplasmosis was confirmed by immunohistochemical staining with T. gondii antiserum [101]. Vielmo et al. [102] described a toxoplasmosis outbreak in domestic chickens and guinea fowl in southern Brazil. 22 birds (13 chickens and 9 guinea fowl) from a total of 76 birds (47 domestic chickens and 29 guinea fowl), showed clinical signs of lethargy, anorexia, and neurological signs, and 15 (9 chickens, 6 guinea fowl) died within 24–72 h. Histopathological findings included tissue necrosis with intralesional T. gondii. Immunohistochemistry for T. gondii was positive [102]. In 2004, five lories died of acute toxoplasmosis in an aviary in South Carolina, United States. For the isolation of T. gondii, tissues were bioassayed in mice and viable T. gondii was isolated from all five of five lories [48]. In 2008, there was a report of death of three (out of ten) Nicobar pigeons (Caloenas nicobaria) in an aviary collection in South Africa, without any clinical symptoms. Numerous protozoal tachyzoites were present in all organs and there was strong positive immunohistochemical (IHC) labelling of these organisms for T. gondii [57]. In 2011, three 13-month-old black-footed penguin chicks (Spheniscus demersus) died from acute toxoplasmosis within 24 h of showing central nervous signs [59]. In the middle of June 1952, Erichsen et al. [103] reported an epidemic outbreak of toxoplasmosis in a chicken flock in south-eastern Norway. There was an unusually high mortality among chickens on a farm, without showing signs of illness prior to death. During a period of three months further two dead chickens were diagnosed from the affected farms. From these animals T. gondii was successfully isolated, and it was decided to eradicate the whole flock [103].
A severe outbreak of toxoplasmosis was observed in a hare ranch (Lepus timidus ainu) in Sapporo, Japan. Eight hares out of thirteen had revealed the symptoms and all died. The adult hare suffered in a very high percentage (seven out of eight), compared with that of the young (one out of five). Parasites were demonstrated heavily in the liver, spleen, and mesenteric lymph node by the Giemsa-stained smear. The isolation of the parasites was successful in one case after four successive inoculations to mice [104]. Notable outbreaks/epizootics of toxoplasmosis in domestic and wild species are presented in Table 2.

2.5. Transmission

Congenital T. gondii infection in humans was described in 1939. In 1959, it was found that congenital infections in mice can produce infected offspring for at least 10 generations. In 1965, it was discovered that T. gondii infectivity is associated with cat feces. In 1970, the sexual phase of the parasite in the small intestine of the cat was discovered [24]. The schizogony and gametogony were identified in cat enterocytes and characterized morphologically and biologically. Pigs and mice (and presumably humans) can be infected by ingesting even 1 oocyst, whereas 100 oocysts may not infect cats. Cats can shed millions of oocysts after ingesting only 1 bradyzoite, while ingestion of 100 bradyzoites may not infect mice orally [12].

2.6. Human Toxoplasmosis

The chronology of T. gondii transmission routes was established through key discoveries: congenital infection (1939), carnivorism (1960), and feco-oral transmission (1965) [11].
First report of acquired toxoplasmosis was from a 6-year-old boy in the USA in 1941, and this isolate was given the child’s initials (RH) and became the famous RH strain. Toxoplasma gondii was isolated from the heart, spleen, blood, and other tissues in 1940–1941. In 1956, it was reported that lymphadenopathy was a frequent sign of acquired toxoplasmosis. Acute toxoplasmosis-induced encephalitis was reported in 1983, in which almost all cases resulted from reactivation of chronic infection initiated by the immune suppression due to HIV infection. In 1988, the first postnatal acquired toxoplasmosis presenting ocular disease (retinochoidal scars) was reported.
In 1974, results of the 15-year study demonstrated that (1) early pregnancy infections are more damaging to the fetus; (2) not all maternal infections transmit vertically; (3) women seropositive before pregnancy did not transmit infection to the fetus; and (4) spiramycin reduces transmission but not severity of disease in infants [12] (Table 3).

2.7. Diagnosis, Treatment, and Control

Diagnostic advances began with development of the first serologic anti-T. gondii diagnostic test called the Sabin–Feldman dye test in 1948, which became the gold standard for detecting anti-T. gondii antibodies across species owing to its high sensitivity [30], followed by the development of many antibody-based diagnostics. Detection of T. gondii DNA was achieved, targeting the tachyzoite B1 gene, in 1989 through PCR. In 1941, sulfonamides were reported to be effective against murine toxoplasmosis [31]. In 1958, spiramycin was found to have antitoxoplasmic activity in mice. In 1973, clindamycin was documented as an alternative to sulfonamides [34]. In the 1990s, it was concluded that excluding cats from pig facilities can reduce T. gondii infection in pigs. Vaccination of sheep with a live cyst-less strain of T. gondii reduces neonatal mortality in lambs [36].

3. Life Cycle and Immunology/Immunopathogenesis

3.1. Life Cycle in Definitive Host (Felids)

Sexual reproduction of T. gondii is restricted to felids because their intestines lack delta-6-desaturase, creating a linoleic acid-rich environment that supports parasite development [26,107,108].
Bradyzoites in tissue cysts are the primary infectious form for both feline oocyst production and natural human infection, because they are more acid-resistant [109] and they can survive up to 2 h under gastric conditions. The evolution of bradyzoites is one of the most important strategies adopted by T. gondii to survive within the gastric proteolytic environment [19]. The presence of bradyzoites has been detected even during acute infections, suggesting rapid stage conversion occurs post-infection [110].
Sporozoites may convert to tachyzoites, and tachyzoites may convert to bradyzoite tissue cysts, and when tissue cysts rupture, a few bradyzoites return to the intestinal epithelium to initiate the enteroepithelial cycle; this event is unpredictable. Enteroepithelial stages preceding the formation of gamonts have not been found [111,112,113]. The prepatent period for oocyst excretion exceeds 18 days after ingesting sporulated oocysts. Unsporulated oocysts are nonpathogenic orally to cats [114].
After ingestion, most of the tachyzoites are destroyed by the gastric acidic environment. Pharyngeal-buccal mucosal invasion may occur [110,115], which can lead to excretion of T. gondii oocysts with a prepatent period of 18 days or more, though this occurs less efficiently than with bradyzoite ingestion.

3.1.1. Motility, Invasion, and Egress

Toxoplasma gondii zoites move along a substrate by a gliding process that maintains anteroposterior polarity, maintaining their crescent shape. This locomotion reaches speeds of up to 10 μm/s in vitro [116]. This gliding motility consists of irregular corkscrew-like trajectories [117]. Toxoplasma gondii glide, invade, and egress through an actin-myosin-based motility machinery called the glideosome, which is a multi-protein complex anchored to T. gondii’s inner membrane complex (IMC), comprising Myosin A, actin, Myosin Light Chain 1, Gliding-Associated Proteins, surface adhesins [118,119]. The essential force comes from the myosin motor TgMyoA, which binds to TgACT1 (actin) and propels it unidirectionally toward the parasite’s rear. For attachment to host cells, surface adhesins like TgMIC2 and TgAMA1 engage host receptors, creating traction points. The glideosome then translocates these adhesins rearward, pulling the parasite forward in a substrate-dependent manner [120]. This (active) invasion is completed in less than 20 s, and during invasion, the same machinery drives the parasite into the host, forming a parasitophorous vacuole. The glideosome also facilitates egress by reactivating the actin–myosin motor, to rupture the host cell membrane (lytic egress) to disseminate and infect new cells [120,121,122].

3.1.2. Schizogony

Bradyzoites initiate schizogony upon invading feline intestinal epithelial cells, an asexual reproduction process in which these zoites undergo repeated nuclear divisions without immediate cytokinesis, forming multinucleated schizonts [111]. These schizonts subsequently undergo cytokinesis, producing merozoites. Collectively, this developmental progression from bradyzoites/sporozoites to merozoites via schizont intermediates defines schizogony in T. gondii [123]. The enteroepithelial stages consist of five types, named as Type A, B, C, D, and E and gamonts [114]. All five types are enteroepithelial, but only Types B-E are schizonts, implying Type A to be an earlier stage. Although type B meets schizont criteria, schizogony has not been observed in Types A and B, and only Types C, D, and E undergo schizogony [26]. Toxoplasma gondii schizonts and merozoites are not infective to mice. Type B and C schizonts develop in both intestinal enterocytes and lamina propria, while Types D and E are restricted to enterocytes [124].

3.1.3. Gametogenesis

The sexual cycle starts 2 days after ingestion of tissue cysts by the cat. The gametes originate from gametocytes formed during gametogony, which is initiated by merozoites (formed from types D/E schizonts) [111]. Gamonts localize to the enterocyte apical cytoplasm near villus tips in the ileum [125]. Macrogamonts (female) are subspherical cells, measuring 8 × 6 µm, with a central nucleus and numerous mitochondria and they mature directly into macrogametes [126]. In contrast, microgamonts (male) are ovoid to ellipsoidal, 6 µm long and 2 µm wide, and undergo nuclear division to produce up to 30 motile microgametes. These microgametes bud from the surface of the microgamont, each equipped with two flagella (~10 μm long) and a single mitochondrion [125]. The fusion of microgamete and macrogamete produces the immature unsporulated oocyst, which is released into the intestinal lumen (by rupture of epithelial cells) and excreted in feces [127]. Oocysts can be excreted in cat feces 3–21 days post-infection, patent period to be less than 1 week, and peak excretion at the 7th day on average [24].

3.2. Life Cycle in the Environment

Cat feces contain diploid oocysts that are formed from the fusion of gametes. The multilayered oocyst wall enables survival for over a year under harsh extremes of environment, including mechanical and chemical stress [128]. Under favorable conditions, diploid oocysts sporulate in the environment via meiosis, producing eight haploid sporozoites as follows:

Sporulation

Sporulation of diploid oocysts occurs outside the cat after 24 h, depending on aeration and temperature, as follows: The nucleus of unsporulated oocysts divides twice to form four nuclei; and a second limiting membrane is formed surrounding all four nuclei [129]. The cytoplasmic division results in the formation of two sporoblasts, each with two nuclei [130]. Both sporoblasts (within the oocysts) elongate to be converted to sporocysts. Sporozoite formation begins with the appearance of two dense cytoplasmic plaques (anlagen) at opposite poles of the sporocyst. Each of the two original nuclei undergoes division, producing four nuclei that migrate into the elongating anlagen [131]. Via endodyogeny—a specialized form of internal budding characteristic of apicomplexans—these anlage give rise to two mature sporozoites, totalling four per sporocyst that align peripherally within each sporocyst. In a nutshell, each sporulated oocyst (11 × 13 μm) contains two sporocysts (6 × 8 µm), and each sporocyst contains four sporozoites [115]. Sporulated oocysts are more environmentally resistant (can survive for several months) than the unsporulated oocysts, likely due to the structure of inner layers of the oocysts and sporocysts [113].

3.3. Life Cycle in Intermediate Hosts, Including Humans and Cats

Toxoplasmosis spreads in intermediate hosts (including humans and cats) by ingestion of T. gondii sporulated oocysts from contaminated food or water, by eating undercooked or raw meat containing tissue cysts from chronically infected ones (humans, other mammals, and birds), by receiving the infected organ/blood from the infected individual, through transplacental transmission, accidental (laboratory) exposure [13,110,113,132], and through milk from the infected mother [133].
Proteolytic enzymes, low gastric pH, and parasite-derived proteases degrade the cyst/oocyst wall, releasing bradyzoites from tissue cysts and sporozoites from oocysts [114].
Felines fed bradyzoite tissue cysts will probably have tachyzoites within a day [24]. Excysted sporozoites invade various nucleated cells [134], except erythrocytes (irrespective of the species having nucleated or anucleated erythocytes) [135], and form a parasitophorous vacuole. The rapidly dividing tachyzoite stage of T. gondii replicates through endodyogeny within host cell parasitophorous vacuoles. Two daughter tachyzoites form internally while maintaining the mother cell’s structure, emerging through outer membrane incorporation. This process generates the characteristic rosette clusters observed during acute infection [136]. Tachyzoite replication continues until it reaches a critical mass, which leads to their egress from the vacuole. The host cell is destroyed, and the released tachyzoites infect adjoining cells. Tachyzoites disseminate freely within phagocytic cells or via lymphatics. Tachyzoites can cross epithelial and endothelial barriers. In mice, a sporozoite takes 12 h post-infection to divide into two tachyzoites.
Tachyzoites efficiently invade nearly all nucleated cells of homeotherms, causing acute infection [114,115]. Unlike schizonts in enterocytes, tachyzoites replicate in the lamina propria via endodyogeny (leading to formation of two daughter cells) [124]. This (tachyzoite) is the stage of T. gondii that is mainly cultivated and maintained in human foreskin fibroblast (HFF) cells for in vitro research purposes under Biosecurity Level 2 conditions [4].
Tachyzoites can differentiate into slow-growing bradyzoites within tissue cysts, via endopolygeny, resulting in chronic infection [137]. The bradyzoite tissue cysts vary in size from 5 to 50 µm in diameter [115]. Felines fed tissue cysts will probably have bradyzoite tissue cysts within 7-21 days. They are mostly formed in the brain, but they can also be formed at several other tissues (as the sclera, optic nerve, tongue, etc.) of the intermediate host during chronic infection [12]. Ingesting the tissue cysts leads to the continuation of the sexual cycle and the asexual cycle within the cats, while intermediate hosts support only the asexual cycle [115] as presented in Figure 1.

3.4. Pathogenesis in General

The bradyzoites/sporozoites, released from the ingested tissue cysts or oocysts, penetrate and replicate in the intestinal epithelial cells, and are transformed to tachyzoites. A host may die because of necrosis of the intestine and mesenteric lymph nodes before other organs are severely damaged. Tachyzoites then disseminate to a variety of organs, causing fatal interstitial pneumonia in felids [13,110].
In the immunocompetent host, both the humoral and the cellular immune responses are activated to limit the infection as the activation of antibodies, macrophages, interferon γ, and CD8+ cytotoxic T lymphocytes. These antigen-specific lymphocytes are capable of killing both extracellular parasites and target cells infected with parasites [138,139]. Parasite dissemination initiates the production of IL-12, IL-18, and IFN-γ, leading to Th1 activation by the host, and this immune response is sufficient in almost all immunocompetent individuals to cope with this foreign invader [108,140]. The role of specific cytokines against T. gondii is summarized in Table 4.
As tachyzoites are cleared from the acutely infected host, tissue cysts containing bradyzoites begin to appear in various body tissues of the host. Toxoplasma gondii secretes signaling molecules into infected host cells to modulate host gene expression, metabolism, and immune response [141]. CD8+ T cells and alternatively activated macrophages can kill cysts at least in the murine model, but it requires further validation [142].
Immuno-compromised hosts allow the persistence of tachyzoites and give rise to progressive focal destruction in affected organs [143,144]. All infected individuals harbor cysts containing bradyzoites (but as subclinical infection), bradyzoites within these cysts replicate to a threshold after which the cysts are ruptured, which leads to the liberation of bradyzoites. These bradyzoites then invade other tissues, leading to development of new bradyzoite-containing cysts [145]. This is the probable source of recrudescent infection in immune-compromised individuals and the most likely stimulus for the persistence of antibody titers in the immune-competent host. Although the concept that toxoplasmosis is related to neuropsychiatric conditions is not proven yet [146,147,148,149]. In rodents, the claim that chronic T. gondii infection increases predation also needs further confirmation [150,151].
Table 4. Role of cytokines against T. gondii.
Table 4. Role of cytokines against T. gondii.
CytokineMain FunctionsRef.
INF αInduce other inflammatory proteins.[152]
INFγProvides protection against T. gondii by activation of MΦ, NO, and GTPase signaling. Also induces cell-autonomous immunity, iNOS, and IDO production.[153]
TNFαInvolved in an acute inflammatory response.[153]
IL1βAcute phase response mediator; induces other inflammatory proteins.[154]
IL2Induces growth of T cells and the release of IFNγ, involved in the lytic activity of MΦ and NK cells.[155]
IL4Antagonizes the products of Th1 cells; long exposure leads to chronic toxoplasmosis.[156]
IL5Has a counter-protective role in acute toxoplasmosis and a protective role in chronic toxoplasmosis.[157]
IL6Has a pleiotropic role in immunity, including creating barriers in early ocular toxoplasmosis, enhancing activities of NK cells, and maturation of T/B cells.[158]
IL7Plays a crucial role in the development of memory CD8+ T cells.[159]
IL10Suppress inflammation to prevent T. gondii encephalitis, Controls hyper-inflammation, regulates the protective functioning of CD4+ cells, and plays a suppressive microbicidal function for MΦ and Np.[160]
IL12Central inducer of IFNγ, activates NK cells, CD4 T cells, and CD8 T cells.[161]
IL15Required for optimal role of NK cells, CD8+ cells, and IELs.[162]
IL17AMainly involved in innate immunity by the recruitment of Np IL12, IFNγ, and IL6.[163]
IL18Involved in the production of IFNγ by NK cells and T cells.[164]
IL23Stimulates NK cells and T cells more specifically in the absence of IL-12.[165]
IL27Required for resistance to chronic toxoplasmic encephalitis, induces CXCR3, T-bet, Blimp1, and IL10 expression, inhibit Th17 development.[166]
IL33induces CCL 2 expression (proinflammatory), induces IL-10 production by M2 macrophages (anti-inflammatory).[167]
TGFβAnti-inflammatory role in the brain, eyes, and intestine.[168]
IFN (Interferon), IL (Interleukin), TNF (Tumor Necrosis Factor), TGF (Transforming Growth Factor), MΦ (macrophages), Np (neutrophils), IELs (intraepithelial lymphocytes).

4. Epidemiology of Toxoplasmosis

Toxoplasmosis has a global distribution. The infection rate is much higher in developing and underdeveloped countries compared to developed countries [169]. The highest PopSet counts are reported from China and the United States to be 323 and 283, respectively, while the lowest (1 each) are from Costa Rica and Egypt, as presented in Figure 2. Continent-wise, prevalence is the highest in Africa (61.4%), intermediate in Oceania (38.5%), South America (31.2%), and Europe (29.7%), and the lowest in North America (17.5%) and Asia (16.4%), as presented in Figure 3. Prevalence is influenced by factors such as climatic conditions and cat populations [123,170]. The survival rate of oocysts is strongly affected by several climatic and environmental factors. Prevalence is higher in older people compared to the young. In addition, socio-economic factors also play a significant role in transmission [123].
Toxoplasma gondii is a single species with diverse isolates and strains, categorized into Types I, II, III, and atypical variants based on molecular markers and murine virulence. Type I isolates are considered virulent to outbred mice, whereas Type II and III isolates are not [48]. Although many other types also exist, having comparatively less prevalence or encompassing less geographical region as HG16, Caribbean 2, Chinese 1, Africa 1, and Africa 4. In general, Type I stains are rare, while Type II and Type III have global prevalence (except in Brazil) [5]. The T. gondii strains in Europe and North America are similar to each other. Africa 1 is reported in sub-Saharan Africa [5,171,172,173] and in Denmark. HG16 is prevalent only in France, Caribbean 2 is prevalent only in the Caribbean region, Chinese 1 and Africa 4 are prevalent in China [174]. We have accessed the data of T. gondii prevalence in different species of different countries through https://toxodb.org (accessed on 6 June 2025) and presented in Figure 3. Notably, high PopSet counts are reported from the United States and China (regions with low prevalence), while Africa (with the highest prevalence) reports nearly the lowest PopSet counts (Figure 2 and Figure 3).

5. Clinical Signs

The severity of the disease varies in different species and the infected individual remains a carrier for life. Most host species (including healthy humans) are asymptomatic; on the one hand, it is fatal to marsupials, neotropical primates, and some marine mammals on the other hand [2,94,175,176,177]. Severity depends on the frequency of exposure, inoculum size, life cycle stage, parasite strain, route of infection, host genetics, unrecognized immune deficiencies, geographic location, and variability of immune responses of the affected host [178].

5.1. Clinical Signs of Toxoplasmosis in Humans

Although immunocompetent persons are usually asymptomatic or have mild symptoms, presenting with nonspecific symptoms such as fever, headache, fatigue, and lymphadenopathy, their increased susceptibility to neuropsychiatric disorders such as autism, schizophrenia, anxiety, and Alzheimer’s disease is notable [179,180].
The trimester of gestation significantly influences congenital infection outcomes. Infection with T. gondii before pregnancy confers little or no risk to the fetus except in women who become infected up to 3 months before conception [181]. Of the children infected with congenital toxoplasmosis, seroconversion and transmission to the fetus occurred in more than half of the cases during the third trimester, followed by less than a fourth in the second trimester and <3.5 percent in the first [182]. First-trimester transmission may cause miscarriage, stillbirth, and/or severe neurologic sequelae [182,183,184]. First trimester shows reduced Toll-like receptor expression in trophoblast cells, indicating a reduced immune response of the placenta to intrauterine infection [185]. So, first-trimester infections are associated with more severe fetal damage such as chorioretinitis, hydrocephalus, and cerebral calcifications. Maternal infection in the third trimester often results in asymptomatic newborns. However, if not treated appropriately, these newborns might develop retinochoroiditis and neurologic deficits in childhood or early adulthood [181,182,185]. If the infected individuals are not treated, then chorioretinitis may develop as a sequel [132]. The parasite persists in the brain for the lifetime of infected individuals [186].
In France, most strains isolated from congenital infections are Type II and the severity of congenital toxoplasmosis is related to the trimester of pregnancy when the mother becomes infected. In Brazil, third-trimester maternal infection often leads to severe congenital toxoplasmosis, linked to atypical genotypes [185].
In pregnant women, the placenta is a target tissue for parasite multiplication, and aids in trans-placental transmission [7]. So, as pregnancy progresses, the placenta development increases, and hence the infection rate is low at the earlier stages of pregnancy and high at later stages of pregnancy [123,187,188]. But the outcome of the infection during early gestation is more severe, and vice versa. Without treatment, the rate of fetal transmission during the first, second, and third trimesters averages up to 25%, 50%, and 70% [123]. Treatment of the pregnant woman reduces the incidence and severity of manifestations of congenital infection [189].
Ocular toxoplasmosis causes 35% of chorioretinitis cases in the United States and Europe [190]. Both congenital transmission and postnatal exposure contribute to onset of ocular toxoplasmosis. An infected individual may present clinical signs such as eye pain, scotomas, blurred vision, photophobia, and with macular involvement threatening central vision, extraocular muscle involvement leading to strabismus, and yellow-white cotton-like patches in the posterior retina [191]. Congenital cases show chorioretinal scarring with fibrosis. Immunocompromised individuals develop severe necrotizing retinitis with both tachyzoites and bradyzoite cysts. Eye infections from toxoplasmosis can be an early warning sign that the parasite may soon attack the brain, causing life-threatening encephalitis, warranting neurologic evaluation. Although initial inflammation may resolve, recurrent chorioretinitis frequently causes cumulative retinal damage and secondary glaucoma [192,193].
Clinical signs such as lymphadenopathy may be self-limited or prolonged symptoms. The signs are usually resolved within a few months to more than a year [194]. The lymph nodes mostly involved are the cervical nodes (present as non-tender, discrete, firm, not matted or fixed to contiguous tissues and do not suppurate), mesenteric nodes (pain, fever may be mistaken for appendicitis), and pectoral nodes (may be mistaken for breast cancer) [195,196].
More rarely, seropositive transplant recipients may reactivate their latent infection due to the transplant-related immune suppression, although typically, if a seropositive person receives a transplant from a seropositive donor, there is a rise in IgG antibody titer and development of IgM antibody specific for T. gondii without illness ascribed to the parasite [195]. Toxoplasmosis is a rare but frequently fatal complication in bone marrow transplant recipients, particularly when the donor is seronegative for toxoplasmosis. The lack of donor-derived immunity can result in an 80% mortality rate. Early diagnosis remains challenging, with only 47% of cases identified ante-mortem, and pulmonary or cerebral infections are associated with the poorest outcomes. Given these risks, prophylaxis is strongly recommended for high-risk serodiscordant pairs (donor-negative/recipient-positive) to prevent lethal infection [197].
Individuals with AIDS who are seropositive for T. gondii are at high risk for T. gondii encephalitis (TE), mostly due to recrudescent infection [198] and when the CD4+ T-cell count falls below 100/μL. Other clinical findings include meningoencephalitis (meningeal involvement is uncommon), altered mental status (75%), seizures (33%), motor deficits, cranial nerve palsies, movement disorders, dysmetria, visual-field loss, aphasia, ataxia, hydrocephalus, choreiform movements, choreoathetosis, and necrotizing encephalitis caused by direct invasion by the parasite [198]. Clinical signs of human toxoplasmosis are summarized in Table 5.
Table 5. Clinical manifestations within humans.
Table 5. Clinical manifestations within humans.
System InvolvedPregnant Women
[7]
Fetus/Infant
[199]
Older Children and Adults (Immunocompetent)
[200]
Immunocompromised (HIV)
[201]
Cardiac/Renal Transplant
[202]
Bone Marrow/HSCT Transplantation
[203]
NeurologicalSame as in older children and adultsEncephalitis, epilepsy, psychomotor retardation, microcephaly, cerebral calcification, hydrocephalusEncephalitis, meningoencephalitis, meningitis, fatal brain abscess, seizures, poor cognition/motor functionEncephalitis, hemiparesis, altered mental state, seizures, cranial nerve disturbancesEncephalitis (within 3 months post-transplant)Localized encephalitis, seizures, headache, confusion
OphthalmologicRetinitis, chorioretinitis, peripheral retinal scars, uveitisRetinochoroiditis, vitritis, blurred vision, scotoma, photophobia, strabismus, glaucoma, vision lossRetinitis, retinochoroiditis, branch retinal artery occlusion, risk of permanent vision lossOcular involvementRetinal involvement-
CardiovascularSame as in older children and adultsMyocarditis, pericarditisMyocarditis, pericarditis-Myocardial involvement-
Respiratory PneumoniaDiffuse interstitial pneumoniaFebrile illness with cough, dyspneaPulmonary involvement
Musculoskeletal Myositis, dermatomyositisPolymyositis, dermatomyositis, myositis, myalgias---
Gastrointestinal (GI)/Hepatic JaundicePancreatitis, increased liver enzymes, mesenteric lymphadenopathy, GI pathologies, hepatocellular abnormalities---
Hematologic/SystemicAsymptomatic to illness, cervical lymphadenopathyRash, petechiae, anemia, high mortality riskSepsis-like syndrome, weight loss-FeverFever
Other--Guillain–Barre syndromePsychosis, dementia, anxiety--
Timing/Special NotesCan show all signs of other groupsProgressive manifestations-Brain predominantly infectedReactivation infection (3 months post)Often leads to death
HSCT: hematopoietic stem cell transplantation; GI: gastrointestinal.

5.2. Clinical Signs of Toxoplasmosis in Animals

Table 6 and Table 7 summarize species-specific clinical manifestations of toxoplasmosis in animals, excluding nonsystemic signs (e.g., fever, anorexia).

5.2.1. Clinical Manifestations Within Wildlife

Wildlife susceptibility to T. gondii ranges from resistant carriers to highly vulnerable species. Resistant hosts rarely show symptoms but may shed oocysts (felids) [150] or maintain latent infections (capybaras) [204]. Endangered species such as striped skunks [205], capybaras, and nutria act as resistant reservoir hosts. While most bat species show no disease, juvenile flying-foxes develop acute pneumonia [206]. Rodents range from fatally affected woodchucks to completely resistant nutria [204,207]. Moderately susceptible species (deer) survive with sporadic acute disease [208], while highly susceptible groups (lagomorphs [209], birds of prey [210]) suffer organ-specific necrosis. Extremely vulnerable animals (koalas [211] and otters [212]) often die rapidly from systemic collapse. New World primates show higher susceptibility than Old World primates, rarely surviving the disease [213] (Table 6).
Table 6. Clinical manifestations within wildlife.
Table 6. Clinical manifestations within wildlife.
ClassOrder
Family
Species and Clinical ManifestationsRef.
MammaliaCarnivora
Felidae
  • Wild cats (Felis silvestris): acute infection.
  • Captive pallas’s cats (Otocolobus manul): up to 60% of infected kittens die, repeated transplacental transmission.
  • Sand cats (Felis margarita).
  • Gordon’s wildcat (Felis silvestris gordoni).
[128,214]
Mephitidae (skunks)Striped skunk (Mephitis mephitis): asymptomatic.[205]
UrsidaeGiant panda (Ailuropoda melanoleuca): affects gastrointestinal, and respiratory systems which can lead to death.[62]
Mustelidae
(sea otters)
Southern sea otters (Enhydra lutris nereis): significant mortality with encephalitis. [212]
Rodentia
Sciuridae (squirrels)
  • Red squirrels: fatal acute toxoplasmosis, mimic signs of rabies.
  • Eastern fox squirrel (Sciurus niger): pneumonitis.
  • Eurasian red squirrel (Sciurus vulgaris): necrosis in the spleen, liver, and lungs.
  • Swinhoe’s striped squirrel (Tamiops swinhoei): enteritis.
[66]
Castoridae (beavers)Beaver (Castor canadensis): fatal systemic toxoplasmosis (lymphohistiocytic encephalitis, myocarditis, interstitial pneumonia with multinucleated cells).[215]
SciuridaeWoodchuck (Marmota monax): head tilt, circling, and rapid weight loss. [204,207]
CaviidaeCapybara (Hydrochoerus hydrochaeris): no clinical toxoplasmosis.
EchimyidaeNutria (Myocastor coypus): no clinical toxoplasmosis.
Lagomorpha
Leporidae (rabbits, hares)
  • Domestic rabbits (Oryctolagus cuniculus): die with or without present diarrhea, foci of necrosis of the spleen and liver associated with the massive presence of multiplying tachyzoites.
  • Brown hares (Lepus europaeus): develop fatal, hemorrhagic enteritis, enlargement and hyperemia of mesenteric lymph nodes, splenomegaly, and multiple necrotic lesions in the parenchyma of the liver and other organs, and death.
  • Mountain hare (Lepus timidus): gross lesions and extensive necrotic areas in the small intestine, mesenteric lymph nodes, and liver.
[216]
Eulipotyphla
Talpidae
(moles/Insectivores)
  • Mole (Talpa europaea): fatal toxoplasmosis, affecting the brain.
  • White-toothed shrews (Crocidura russula): affect brains and hearts.
  • Striped field mice (Apodemus agrarius): affect heart.
[217]
Chiroptera
Pteropodidae
(bats)
  • Spectacled flying-fox (Pteropus conspicillatus): acute toxoplasmosis
  • Little red flying-fox (Pteropus scapulatus): acute toxoplasmosis with severe, acute interstitial pneumonia and fibrin present within alveoli in the lungs, multiple foci of gliosis, including gemistocytic astrocytes, at all levels of the cerebrum, cerebellum, and brainstem.
  • Red night bats: no clinical disease.
[206]
Artiodactyla
Cervidae
(deer)
No transplacental infection unless an acute infection occurs during pregnancy. Acute toxoplasmosis and death in mule deer.
  • Reindeer (Rangifer tarandus): enteritis and death.
[218]
Bovidae
(Antelope, nilgai)
  • Pronghorn antelope (Antilocapra americana): acute fatal toxoplasmosis.
  • Rocky Mountain bighorn sheep (Ovis canadensis canadensis): encephalitis. captive nilgais (Boselaphus tragocamelus): abortion and neonatal death.
[50,214]
Non-human primates
Callitrichidae
  • Golden lion tamarins (Leontopithecus rosalia): fatal.
[214,219]
Cercopithecidae (Macaques)
  • Rhesus monkey (Macaca mulatta): congenital toxoplasmosis.
  • Stump-tailed macaques (Macaca arctoides): congenital toxoplasmosis.
  • Cynomolgus monkey (Macaca fascicularis): recurrent toxoplasmic retinochoroiditis.
  • Barbary macaque (Macaca sylvana): concurrent central nervous system toxoplasmosis.
Atelidae
  • Alouatta sp. Diffuse yellowish liver with multifocal petechiae, enlarged spleens with bulging edges and ared pulp expansion, pulmonary edema, intestinal mucosal petechiae and suffusions.
[100]
 ○  Alouatta belzebul: prostration, diarrhea.
[58]
 ○  Alouatta caraya: prostration, inappetence, abdominal distension and pain, intestinal hypomotility.
[220,221]
  • Brachyteles arachnoides: prostrated, laterally recumbent, tachypnea, pyrexia, bilateral pneumonia, cardiovascular collapse, and died.
[222]
  • Alouatta guariba guariba: mild jaundice, depression, anorexia, fever, oliguria, and severe hepatosplenomegaly, hepatomegaly with yellowish discoloration.
[223]
  • Lagothrix lagothricha: apathetic, diarrhoea, dark coloured urine, vomit, death.
[98]
Cebidae
  • Cebus capucinus: paraparesis, interstitial pneumonia, hepaticnecrosis, encephalitis, and enteritis.
[224]
  • Squirrel monkey (Saimiri sciureus): acute fatal. Inflamed liver and spleen, enteritis, villus atrophy, multifocal areas of necrosis along with T. gondii in the lungs, liver, spleen, lymph nodes, adrenal glands, heart, congested and pulmonary edema, respiratory failure.
[5,38,225,226]
Diprotodontia
Macropodidae
Acute fatal, respiratory distress, diarrhoea, neurological disturbances, myocardial hemorrhages and pale streaks, lymphadenomegaly, splenomegaly, adrenal enlargement and reddening, gastrointestinal reddening and ulceration, pancreatic swelling, brain malacia.
  • Black-faced kangaroos (Macropus fuliginosus melanops): Congenital toxoplasmosis.
Tammar wallabies (Macropus eugenii): acute infection, necrosis, enteritis, lymphadenitis, adrenalitis, carditis, myositis, and encephalitis. Severe generalized pulmonary congestion and edema.
[227,228,229,230,231,232]
PhascolarctidaeKoalas (Phascolarctos cinereus): acute fatal, myocardial hemorrhages, and pale streaks.[233,234]
VombatidaeWombats (Vombatus ursinus): respiratory distress, neurological disturbances.[234]
Phalangeridae
(Possums)
Acute fatal, respiratory distress, neurological disturbances, myocardial hemorrhages and pale streaks, splenomegaly, gastrointestinal reddening and ulceration, brain malacia.[234,235,236]
Peramelemorphia
Peramelidae
(Bandicoots)
Neurological disturbances, adrenal enlargement and reddening, pancreatic swelling.
  • Eastern barred bandicoots (Perameles gunnii): acute infection, necrosis, enteritis, lymphadenitis, adrenalitis, carditis, myositis, and encephalitis. Severe generalized pulmonary congestion and edema.
[237,238,239]
Dasyuromorphia
Dasyuridae
Myrmecobiidae
Acute fatal, neurological disturbances, gastrointestinal reddening, and ulceration[5]
Peramelemorphia
Thylacomyidae
Bilby (Macrotis lagotis): neurological disturbances, adrenal enlargement and reddening, pancreatic swelling.[240]
Cetacea
Delphinidae
  • Bottlenose dolphin (Tursiops aduncus): congenital toxoplasmosis.
  • Risso’s dolphin (Grampus griseus): disseminated congenital toxoplasmosis.
[66]
Aves
  • Hawaiian crow (Corvus hawaiiensis): fatal.
  • Black-footed penguin (Spheniscus demersus): fatal, nervous signs.
  • Bald eagle (Haliaeetus leucocephalus): necrotizing myocarditis.
  • Barred owl (Strix varia): hepatitis.
  • Turkeys: fatal systemic toxoplasmosis.
[214,241]

5.2.2. Clinical Manifestations Within Domestic Animals

The cats may remain asymptomatic or develop systemic illness, with rare cases of transplacental/lactogenic transmission [24,177,242]. Dogs demonstrate mild respiratory or hepatic involvement and no evidence of vertical transmission [243]. Ferrets can have congenital infection. Ectothermic pets do not experience natural T. gondii infections, despite being experimentally infectible in laboratory settings [67]. Toxoplasma gondii’s agricultural impact concentrates primarily in small ruminants [244] and swine [245], while most other farm species either resist infection or serve as silent reservoirs, except camels, which may develop acute respiratory disease [246]. Poultry species show decreased egg production, and turkeys harboring tissue cysts in their muscles without clinical signs [12,247,248] (Table 7).
Table 7. Clinical manifestations within domestic animals.
Table 7. Clinical manifestations within domestic animals.
Domestic Animals
Pet animals
Cats (definitive host)Asymptomatic or polypnea, icterus, uveitis and retinochoroiditis, pericardial and abdominal effusions, diffuse necrotizing hepatitis, transplacental and lactogenically.[24,177,242]
DogsRespiratory and hepatic systems, no transplacental infection, resistant to experimental toxoplasmosis.[243]
FerretsCongenital toxoplasmosis, acute and chronic forms.[67]
MinkCan be naturally infected.[69]
Fish, reptiles, amphibiansNot occur in fish, reptiles, or amphibians as natural infection, but they can be experimentally infected.[4,249]
Production Animals
HorsesRelatively resistant to experimental infection, no clinical disease.[17,18]
SwineCan be naturally infected, causing sow abortion (sows abort only once).[245]
CattleRare, no report of zoonotic.[16]
Sheep, goatsAbortion.[244]
BuffalosNo clinical disease.[250]
CamelsAcute toxoplasmosis with dyspnea.[246]
Llamas, alpaca, and vicunasNo clinical disease.[251]
ChickensNo clinical signs, no vertical transmission, decreased egg production.[247]
TurkeysNo clinical signs, having tissue cysts in breast and leg muscles.[248]
Ducks and geeseNo clinical disease.[12]
Clinical signs of serologically positive species, not supported with confirmatory tests of toxoplasmosis, are not listed in this table.

6. Diagnosis of Toxoplasmosis

6.1. Diagnosis of Toxoplasmosis in Animals

Toxoplasmosis is diagnosed by biologic, serologic, or histologic and molecular methods, or some combination of these [252]. Diagnosis in felids can also be performed through coproparasitological examinations [253]. While traditional serological tests such as the indirect hemagglutination assay (IHA), indirect fluorescent antibody test (IFAT), and latex agglutination test (LAT) have been historically used for toxoplasmosis diagnosis, many of these are now considered outdated. Serological testing remains a cornerstone for T. gondii diagnosis in veterinary medicine, serving both clinical and epidemiological applications. While newer molecular methods have emerged, well-established serological assays continue to provide reliable diagnostic solutions. The indirect fluorescent antibody test (IFAT) maintains particular utility due to its ability to differentiate acute and chronic infections through IgM and IgG detection, with reported sensitivity of 85-95% in domestic species. Similarly, the modified agglutination test (MAT) is widely employed in veterinary laboratories, offering excellent specificity (92-98%) and stability of reagents for field surveys. These techniques complement newer diagnostic approaches while remaining indispensable for (1) large-scale seroprevalence studies, (2) routine screening in clinical practice, and (3) situations where tissue sampling is impractical. Their continued use reflects standardized protocols, cost-effectiveness, and proven performance across animal species. Currently, enzyme-linked immunosorbent assay (ELISA) remains widely used due to its reliability and automation potential [173]. However, newer advanced techniques—including chromatographic immunoassays (e.g., lateral flow tests), chemiluminescence assays (CLIAs), and real-time PCR—have improved sensitivity and specificity for detecting T. gondii infection, particularly in congenital and immunocompromised cases [252]. Increased IgM titers (>1:256) are consistent with recent infection. IgG titers must be measured in paired serum samples obtained during the acute and convalescent stages (3–4 weeks apart) and must show at least a fourfold increase/decrease in titer [254,255] (Table 8 and Table 9).
Histological examination may present tachyzoites or bradyzoites in domestic animals, but it is not definitive due to the morphological overlap with other cyst-forming coccidia (Neospora caninum, Hammondia spp.). Thus, histopathology must be combined with T. gondii-specific immunohistochemistry (targeting SAG1/BAG1 antigens) and PCR amplification of conserved genomic targets (B1/REP-529), particularly in food animals where accurate speciation carries zoonotic significance.
Application of specific polymerase chain reaction (PCR) assays allows diagnosis from tissue DNA samples [256]. Isolation of T. gondii from secretions, excretions, and organs by bioassay using laboratory animals is an accurate method of diagnosing animal infection. It has the disadvantage that it is difficult to perform, less sensitive, and unless the T. gondii strain is highly virulent, it requires three weeks before mouse examination yields recognizable T. gondii cysts [257].

6.2. Diagnosis of Toxoplasmosis in Humans

The diagnosis of parasite infections is typically divided into direct methods—such as microscopy, molecular and imaging techniques, and biological isolation—and indirect methods, including serological tests like Sabin–Feldman dye test, IFA, ELISA, and agglutination tests for antibody detection [258].
Microscopy detects tachyzoites or tissue cysts. Tachyzoites are more likely to be detected in acute infection, but they are often cleared before sampling. Conversely, bradyzoite tissue cysts are sparse and unevenly distributed, making them difficult to identify even with the use of specialized stains [198]. The main techniques include the following: (1) Giemsa or H&E staining for tachyzoites and cysts, (2) PAS staining for bradyzoites, and (3) immunohistochemistry (IHC) with T. gondii-specific antibodies (anti-SAG1 for tachyzoites, anti-BAG1 for bradyzoites) [258,259]. This morphological approach is particularly useful for tissue diagnosis in immunocompromised patients but remains less sensitive (30-50%) than PCR or serology [198] due to (a) sampling limitations of focal infections, (b) intermittent parasite presence, and (c) technical challenges in distinguishing T. gondii from similar structures. In general, microscopic detection of T. gondii cystic forms in biological samples from patients with suspected acute toxoplasmosis is difficult. Conversely, rapidly proliferating forms are more likely to be detected.
The Sabin–Feldman dye test, historically vital for toxoplasmosis diagnosis, detects serum antibodies with high sensitivity across species. However, its reliance on live T. gondii tachyzoites posed biosafety risks and technical challenges, leading to its obsolescence [30]. Subsequent serological advances included the indirect immunofluorescent antibody test (IFA), which eliminated the need for live parasites and became a preferred alternative [260].
Modern diagnostics now prioritize enzyme-linked immunosorbent assay (ELISA) based techniques. Unlike older methods, ELISA platforms use purified or recombinant antigens (e.g., SAG1, GRA7) to detect antibodies, enhancing safety, reproducibility, and scalability. Among ELISA variants, IgG ELISA is now the most widely used for chronic infection screening, owing to its ability to quantify long-term antibody responses with high specificity [261]. For acute infection confirmation, testing paired sera (collected 1–2 weeks apart) to detect rising IgG titers remains critical, supplemented by IgM ELISA (to identify recent exposure) and IgG avidity testing (to distinguish acute from past infections). Further advancements include chemiluminescence assays (e.g., Architect, Liaison), which offer automated, high-throughput analysis with improved sensitivity and standardization [257,262].
Table 8. Diagnostic potential of immunoglobulins.
Table 8. Diagnostic potential of immunoglobulins.
AntibodyLife SpanPredictionRef.
IgEShort-term (days to weeks)Acute toxoplasmosis[263]
IgAFew weeksSupports acute/reactivated/congenital infection[264]
IgM 1 week to months/yearsCongenital toxoplasmosis; alone insufficient to establish acute toxoplasmosis[265,266]
IgGLifelong (post-infection)Seroconversion and exposure (timing unclear without avidity testing)[267]
Table 9. Summary of direct and indirect, serological, and salivary methods for detection of T. gondii infection.
Table 9. Summary of direct and indirect, serological, and salivary methods for detection of T. gondii infection.
1 TypeMethod CategorySpecific TechniquesTargetDetected Analyte (Key Reagent)
DirectMicroscopy (MS)Light MS (Giemsa, H&E, PAS)Tachyzoites, tissue cystsTachyzoites, tissue cysts
Immunohistochemistry (IHC)T. gondii antigensFluorophore-labeled anti-T. gondii antibodies
Electron microscopyUltrastructural parasite featuresN/A (morphology only)
BioassayMouse bioassayViable parasitesN/A (relies viable infection)
Cell cultureReplicating tachyoitesN/A
ImagingCT/MRI (CNS lesions)Brain abscesses, calcificationsN/A
Ultrasonography (congenital)Fetal abnormalitiesN/A
MolecularPCR, qPCR, LAMP, etc.DNA regionsDNA Region
NanoparticlePiezoelectric immunoagglutination (PIA)Antigens IgG
1 Plasmonic gold chips (PGC)Antigens (saliva)IgG
Quantum dot-labeled antigen detectionAntigens
ImmunoassaysImmunofluorescence antigen detection (IFA-D)Antigens in tissuesFluorophore-labeled anti-T. gondii antibodies
IndirectSerological Assays (Antibody Detection)Dye test (DT) Live tachyzoiteIgG, IgA, IgM
Modified agglutination test (MAT)Formalin-fixed tachyzoite IgG
Indirect fluorescent antibody test (IFAT)Fixed tachyzoites IgG, IgM
Indirect hemagglutination (IHA)Tanned red blood cells sensitized with soluble antigens IgG
ELISA
-
Whole-cell lysate
Tachyzoite lysate antigens (TLAs)IgG
-
Recombinant antigen
SAG1/GRA7/ROP1 proteinIgG, IgM
-
Multiplex
Multiple antigensIgG, IgM, IgA
-
Avidity-modified
TLA/recombinant antigensIgG (avidity index)
-
Chemiluminescence (CLIA)
Recombinant antigensIgG, IgM
-
Point-of-care
Lateral flow stripsIgG, IgM
Immunosorbent agglutination assay (ISAGA) Anti-human IgM IgM
Latex agglutination test (LAT) antigen-coated latex particles IgG, IgM
Western blotting (WB) Tachyzoite lysate/recombinantIgG, IgM
Immunochromatographic test (ICT) 2 Conjugate or reagent pad IgG, 3 ESA
Avidity test Tachyzoite lysate antigen, recombinant antigens IgG (avidity index)
Antigen DetectionLateral flow assay
1 All methods are serological except PGC, which uses saliva. PGC is not yet validated for animal diagnosis; 2 A conjugate or reagent pad contains antibodies specific to the target analyte conjugated to colored particles (i.e., colloidal gold particles or latex microspheres); 3 ESA: excreted/secreted antigens.
A summary of the molecular techniques is presented in Table 10.

7. Treatment

7.1. Treatment of Toxoplasmosis in Animals

Currently available anti-T. gondii drugs limit the multiplication of the tachyzoites and oocyst shedding, but they cannot eradicate the infection [275]. Anti-coccidial drugs treat acute infection. Other drugs, such as diaminodiphenylsulfone, atovaquone, spiramycin, toltrazuril, ponazuril, and diclazuril may also limit infection [5,276,277]. A summary of drugs used to treat animal toxoplasmosis is presented in Table 11.

7.2. Treatment of Toxoplasmosis in Humans

In 1953, the traditional gold standard for treating human toxoplasmosis was established—combination therapy targeting two enzymes in the folate pathway: dihydrofolate reductase (pyrimethamine; loading dose of 75 mg followed by 25 mg orally per day) and dihydropteroate synthetase (sulfadiazine; 1 g orally every 6 h), commonly called Pyr-Sulf [31]. Their combination is eightfold more effective than either compound alone [282,283]. However, recent studies highlight alternative regimens (e.g., trimethoprim-sulfamethoxazole, clindamycin-based combinations [284], or atovaquone [282]) with comparable efficacy, particularly in sulfa-intolerant patients or ocular toxoplasmosis. These drugs limit the proliferation of tachyzoites and the destruction of host cells. If fetal infection is detected, the combination of pyrimethamine and sulfadiazine is administered to the mother (only after the first 12–14 weeks of pregnancy) to prevent the severity of congenital toxoplasmosis and to the newborn in the postnatal period [283]. Complete blood counts are monitored twice weekly during Pyr-Sulf treatment [285].
In 1958, spiramycin was found to have antitoxoplasmic activity. It has been used prophylactically in pregnant women as it does not cross the placenta. It is generally effective in reducing fetal transmission of toxoplasmosis when administered early following maternal infection [286], whereas delayed initiation is associated with reduced efficacy and potential adverse outcomes, including fetal loss [184].
Long-term treatment of trimethoprim/sulfamethoxazole can prevent recurrent eye infections; nevertheless, approximately 40% of immunocompromised patients discontinue the medication due to sulfa drug toxicity [287]. Consequently, an alternate treatment that has demonstrated good outcomes is the intravitreal injection of dexamethasone and either clindamycin or trimethoprim/sulfamethoxazole [284].
Pyrimethamine may induce folate deficiency, agranulocytosis, Stevens–Johnson syndrome, toxic epidermal necrolysis, and hepatic necrosis [285]. Leucovorin (folinic acid) (10 mg orally each day for at least 3–6 weeks) may also be added with pyrimethamine and sulfadiazine [288,289,290]. It does not inhibit the action of pyrimethamine on T. gondii, as the parasite cannot take up folinic acid at the concentrations achieved in serum. The parasite can take up folate (folic acid), which bypasses dihydrofolate reductase (DHFR) inhibitors or other upstream enzymes [289].
Patients should consume at least 2 L of fluid daily to prevent crystalluria because of sulfadiazine. Fluid intake should be sufficient to ensure a daily urine volume of at least 1.2 L (in adults). Urinary alkalinization may be helpful if urine volume or pH is unusually low [288]. Atovaquone has potent activity against T. gondii in patients with trimethoprim–sulfamethoxazole intolerence, and against cyst forms of T. gondii, and is effective against T. gondii chorioretinitis [282].
In 1973, clindamycin was documented as an alternative for sulfonamide-sensitive patients [34], without loss of efficacy [34,289]. Intravitreal injection of clindamycin and dexamethasone is also being used to treat ocular toxoplasmosis with satisfactory results [284]. It is initially coccidiostatic but becomes coccidiocidal after a few days of treatment. The dosages used against T. gondii are higher than for the treatment of anaerobic infections, for which it is marketed [115].
A combination of azithromycin and clarithromycin is also good for treating toxoplasmosis [289]. Virginiamycin, terbinafine, and triazine derivatives (e.g., diclazuril, toltrazuril, ponazuril) can work against toxoplasmosis [291]. A flow chart of drugs used to treat human toxoplasmosis is presented in Figure 4.
There are many limitations of anti-T. gondii chemotherapy, including recurrence of the disease, and the solution to this problem is the identification of the population that is at more risk of recurrence, and the application of secondary prophylaxis at the time at which the recurrence is most likely to occur. Also, no drug is available to act against the latent stage of the infection. The treatment success rate remains low, as the drugs available to treat T. gondii are active against tachyzoites, and not against bradyzoites [284,292]. Therefore, vaccination against toxoplasmosis could be an effective and appropriate medical prevention [293].

8. Control of Toxoplasmosis

8.1. Vaccines

As discussed earlier, there are limitations of anti-T. gondii chemotherapy, as it may cause the recurrence of the disease, is ineffective against the latent stage of the infection [284], low success rate, may lead to folate deficiency, agranulocytosis, Stevens–Johnson syndrome, toxic epidermal necrolysis, and hepatic necrosis. Therefore, vaccination against toxoplasmosis could be an effective and appropriate preventive measure, especially for specific populations such as pregnant women and HIV patients [284,293].
Despite encouraging results of a live attenuated vaccine (Toxovax®, MSD, New Zealand) developed for sheep to limit their abortion storm [294] and recent advances in genetics [295,296,297], no anti-T. gondii vaccine is available for humans [298]. Challenges in T. gondii vaccine development in humans include, but are not limited to, the complex life cycle of T. gondii, diversity of T. gondii strains, the establishment of latent infection, and T. gondii immune evasion strategies, the difficulty in clinical translation, and lack of optimal adjuvants [293,299]. A summary of proposed different types of vaccines used against toxoplasmosis is presented in Table 12.

8.2. Non-Vaccine Prevention Strategies

8.2.1. Food Safety Measures

Methods to control toxoplasmosis apart from vaccination and chemotherapy include proper cooking (≥73 °C, followed by 3 min rest before eating) and freezing of meat (−12 °C for ≥3 days), which kills tissue cysts in contaminated meat [12,115,304]. Freezing meat overnight in a household freezer before human or animal consumption remains the easiest and most economical method of reducing transmission of T. gondii through meat. Studies constructed thermal curves showing temperatures required to kill T. gondii in infected meat by freezing, cooking, and by gamma irradiation [12].

8.2.2. Personal and Pet Hygiene Practices

With proper hygiene, general interaction with cats (e.g., petting) poses minimal risk. Additional precautions include wearing gloves while gardening, washing hands after handling cats or their belongings, and keeping cats indoors to prevent hunting. Litter boxes should be cleaned daily and pregnant women should avoid cleaning cat litter boxes. Protocols have also been developed for serological screening of pregnant mothers and neonates, but the cost and benefit are poorly documented.

8.2.3. Livestock Management Protocols

On livestock farms, excluding cats is essential to prevent oocyst contamination of feed and water, thereby reducing meat-borne transmission to humans, a significant route of exposure [4]. Effective sanitary planning in animal production is essential to minimize T. gondii infection in livestock, which serves as a key source of human transmission [305]. Farms should implement strict biosecurity measures, including rodent control, proper feed storage (to prevent contamination with cat feces), and restricted access to livestock areas to reduce exposure to infected materials. Pregnant livestock (especially sheep and goats) are highly susceptible, with toxoplasmosis causing abortions and economic losses. Vaccination of sheep (where available) and regular serological monitoring can help identify and manage infected herds [306]. Additionally, farm workers should be educated on hygiene practices, such as handwashing and the use of protective equipment, to prevent zoonotic transmission [307]. Proper disposal of placental and fetal tissues from aborted fetuses is crucial to avoid environmental contamination with T. gondii oocysts [308].

8.2.4. Feline Management Strategies

Controlling the population of stray cats is a critical measure in reducing environmental contamination with T. gondii oocysts, as cats are the definitive hosts of the parasite. Responsible pet ownership programs should emphasize spaying and neutering to prevent overpopulation, alongside public education on proper cat care, including keeping cats indoors to limit their hunting behavior and access to intermediate hosts [309]. Municipalities should support trap-neuter-return (TNR) programs to stabilize feral cat colonies and reduce their numbers [310]. Pet owners should also be encouraged to dispose of cat litter safely—preferably by sealing it in bags before disposal—to prevent oocyst dissemination [311]. Community awareness campaigns can highlight the risks of feeding stray cats near farms or water sources, which may contribute to parasite spread [309].

8.2.5. One Health Implementation

A coordinated One Health strategy, integrating human, animal, and environmental health sectors, is vital for effective toxoplasmosis control. Public health professionals should collaborate with veterinarians on monitoring T. gondii prevalence in both pets and livestock, and with environmental agencies to assess contamination risks in soil and water [312]. Educational initiatives can promote safe food handling (e.g., cooking meat thoroughly, washing produce) and protective measures for pregnant women and immunocompromised individuals. Urban planners and policymakers should implement sanitation improvements, such as proper waste management and water treatment, to reduce environmental oocyst persistence. Joint research efforts can identify high-risk transmission pathways and guide targeted interventions. Cross-sectoral cooperation under One Health ensures strategies for mitigating risks at individual and community levels [313,314].

8.2.6. Environmental Conservation

Toxoplasma gondii exhibits remarkable genetic diversity in the forest due to high biodiversity, which supports extensive parasite recombination, with wild felids—having larger home ranges than domestic cats—contributing to widespread oocyst contamination through their feces. Importantly, deforestation displaces wild felids into peri-domestic areas, significantly increasing environmental contamination risks near human settlements. Prevention of habitat destruction is therefore crucial to maintain ecological barriers and reduce zoonotic transmission. Concurrently, anthropization favors selection of a few domestic-adapted strains, while forest edges create zones where wild and domestic cycles intersect, facilitating strain mixing [315,316,317].

9. Conclusions

The reviewed findings suggest future research should focus on understanding the parasite’s adaptation to diverse hosts, developing screening technologies, and validating animal models. In addition, controlled clinical trials should be conducted on suspected populations. There is a need to integrate omics, molecular biology, and bioinformatics to help demonstrate the genetic diversity of T. gondii strains, which would aid in the development of more sensitive and specific diagnostic tools, and to identify a potent antigenic candidate for vaccine development against toxoplasmosis. Further investigation is needed to comprehend the impact of species specificity and the inhibition of delta-6-desaturase activity on the sexual development of T. gondii in the intestines of non-feline hosts.

Author Contributions

Conceptualization, Y.-G.Y., M.-X.C. and M.F.; software, M.F., M.-X.C., Z.H., S.-Y.H. and M.W.A.; validation and formal analysis, M.F., A.S., Z.H., S.-Y.H. and L.L.; data curation, M.F., A.S., M.W.A. and S.-Y.H.; writing—original draft preparation, M.F., M.W.A. and L.L.; writing—review and editing, M.F., M.W.A., A.S. and Z.H.; visualization, M.F., M.W.A., S.-Y.H. and M.-X.C.; project administration, M.F., M.W.A. and Y.-G.Y.; funding acquisition, M.F., M.W.A., A.S. and Y.-G.Y. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge funding from the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD), Development of a new precise cytosine base editor (JBGS [2021] 025), National Natural Science Foundation of China (Nos. 32373041, W2433064), China Postdoctoral Science Foundation (No. 2020M671615), the 111 Project D18007, and Yangzhou city and Yangzhou University corporation (YZ2023205/2022187).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to express their sincere gratitude to Usman Nazir (College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China) for his valuable technical support and guidance in software utilization and image creation.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Splendore, A. Un Nuovo Protozoa Parassita Deconigli Incontrato Nelle Lesioni Anatomiche d’une Malattia Che Ricorda in Molti Punti Il Kala-Azar Dell’uoma. Nota Preliminare Pel. Rev. Soc. Sci. Sao Paulo 1908, 3, 109–112. [Google Scholar]
  2. Innes, E.A. A Brief History and Overview of Toxoplasma gondii. Zoonoses Public Health 2010, 57, 1–7. [Google Scholar] [CrossRef] [PubMed]
  3. Nayeri, T.; Sarvi, S.; Daryani, A. Toxoplasma gondii in Mollusks and Cold-Blooded Animals: A Systematic Review. Parasitology 2021, 148, 895–903. [Google Scholar] [CrossRef] [PubMed]
  4. Weiss, L.; Kim, K. Toxoplasma gondii: The Model Apicomplexan. Perspectives and Methods, 3rd ed.; Elsevier: London, UK, 2020; ISBN 78-0-12-815041-2. [Google Scholar]
  5. Dubey, J.P. Toxoplasmosis of Animals and Humans, 3rd ed.; CRC Press: Boca Raton, FL, USA; Taylor & Francis Group: Abingdon, UK, 2022; ISBN 978-1-003-19937-3. [Google Scholar]
  6. Khan, A.; Grigg, M.E. Toxoplasma gondii: Laboratory Maintenance and Growth. Curr. Protoc. Microbiol. 2017, 44, 20C. 1.1–20C. 1.17. [Google Scholar] [CrossRef]
  7. Rostami, A.; Riahi, S.M.; Contopoulos-Ioannidis, D.G.; Gamble, H.R.; Fakhri, Y.; Shiadeh, M.N.; Foroutan, M.; Behniafar, H.; Taghipour, A.; Maldonado, Y.A. Acute Toxoplasma Infection in Pregnant Women Worldwide: A Systematic Review and Meta-Analysis. PLoS Neglected Trop. Dis. 2019, 13, e0007807. [Google Scholar] [CrossRef]
  8. Search for Popset Isolate Sequences by Geographic Location. Available online: https://toxodb.org/toxo/app/search/popsetSequence/PopsetByCountry (accessed on 6 June 2025).
  9. Bouaicha, F.; Amairia, S.; Amdouni, Y.; Elati, K.; Bensmida, B.; Rekik, M.; Gharbi, M. Molecular and Serological Detection of Toxoplasma gondii in Two Species of Rodents: Ctenodactylus Gundi (Rodentia, Ctenodactylidae) and Psammomys Obesus (Rodentia, Muridae) From South Tunisia. Vet. Med. Sci. 2025, 11, e70371. [Google Scholar] [CrossRef]
  10. Sabin, A.B.; Olitsky, P.K. Toxoplasma and Obligate Intracellular Parasitism. Science 1937, 85, 336–338. [Google Scholar] [CrossRef]
  11. Wolf, A.; Cowen, D.; Paige, B. Human Toxoplasmosis: Occurrence in Infants as an Encephalomyelitis Verification by Transmission to Animals. Science 1939, 89, 226–227. [Google Scholar] [CrossRef]
  12. Dubey, J.P. The History of Toxoplasma gondii—The First 100 Years. J. Eukaryot. Microbiol. 2008, 55, 467–475. [Google Scholar] [CrossRef]
  13. Sokol-Borrelli, S.L.; Reilly, S.M.; Holmes, M.J.; Orchanian, S.B.; Massmann, M.D.; Sharp, K.G.; Cabo, L.F.; Alrubaye, H.S.; Martorelli Di Genova, B.; Lodoen, M.B. A Transcriptional Network Required for Bradyzoite Development in Toxoplasma gondii Is Dispensable for Recrudescent Disease. Nat. Commun. 2023, 14, 6078. [Google Scholar] [CrossRef]
  14. Hartley, W.J.; Jebson, J.L.; McFarlane, D. New Zealand Type II Abortion in Ewes. Aust. Vet. J. 1954, 30, 216–218. [Google Scholar] [CrossRef]
  15. Buxton, D. Ovine Toxoplasmosis: A Review. J. R. Soc. Med. 1990, 83, 509–511. [Google Scholar] [CrossRef] [PubMed]
  16. Dubey, J.P.; Julian, A.F. First Report of Acute, Visceral, Fatal Toxoplasmosis in a Naturally Infected Calf (Bos taurus). Vet. Parasitol. 2025, 334, 110373. [Google Scholar] [CrossRef] [PubMed]
  17. Kimble, K.M.; Gomez, G.; Szule, J.A.; Dubey, J.P.; Buchanan, B.; Porter, B.F. Systemic Toxoplasmosis in a Horse. J. Comp. Pathol. 2021, 182, 27–31. [Google Scholar] [CrossRef]
  18. Toxoplasma gondii Infection in Horses. A Review—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/18412038/ (accessed on 8 May 2024).
  19. Jacobs, L.; Remington, J.S.; Melton, M.L. The Resistance of the Encysted Form of Toxoplasma gondii. J. Parasitol. 1960, 46, 11–21. [Google Scholar] [CrossRef]
  20. Nicolle, C. Sur Une Infection a Corps de Leishman (on Organismes Voisons) Du Gondi. CR Acad. Sci. 1908, 147, 736. [Google Scholar]
  21. Boothroyd, J.C. Toxoplasma gondii: 25 Years and 25 Major Advances for the Field. Int. J. Parasitol. 2009, 39, 935. [Google Scholar] [CrossRef]
  22. Goldman, M.; Carver, R.K.; Sulzer, A.J. Reproduction of Toxoplasma gondii by Internal Budding. J. Parasitol. 1958, 44, 161–171. [Google Scholar] [CrossRef]
  23. Dubey, J.P.; Miller, N.L.; Frenkel, J.K. The Toxoplasma gondii Oocyst from Cat Feces. J. Exp. Med. 1970, 132, 636–662. [Google Scholar] [CrossRef]
  24. Dubey, J.P.; Frenkel, J.K. Cyst-Induced Toxoplasmosis in Cats. J. Protozool. 1972, 19, 155–177. [Google Scholar] [CrossRef]
  25. Frenkel, J.K. Toxoplasma in and around Us. BioScience 1973, 23, 343–352. [Google Scholar] [CrossRef]
  26. Martorelli Di Genova, B.; Wilson, S.K.; Dubey, J.P.; Knoll, L.J. Intestinal Delta-6-Desaturase Activity Determines Host Range for Toxoplasma Sexual Reproduction. PLoS Biol. 2019, 17, e3000364. [Google Scholar] [CrossRef]
  27. Beverley, J.K.A. Congenital Transmission of Toxoplasmosis through Successive Generations of Mice. Nature 1959, 183, 1348–1349. [Google Scholar] [CrossRef]
  28. Weinman, D.; Chandler, A.H. Toxoplasmosis in Swine and Rodents; Reciprocal Oral Infection and Potential Human Hazard. Proc. Soc. Exp. Biol. Med. 1954, 87, 211–216. [Google Scholar] [CrossRef] [PubMed]
  29. Hutchison, W.M. Experimental Transmission of Toxoplasma gondii. Nature 1965, 206, 961–962. [Google Scholar] [CrossRef] [PubMed]
  30. Reiter-Owona, I.; Petersen, E.; Joynson, D.; Aspöck, H.; Dardé, M.L.; Disko, R.; Dreazen, O.; Dumon, H.; Grillo, R.; Gross, U.; et al. The Past and Present Role of the Sabin-Feldman Dye Test in the Serodiagnosis of Toxoplasmosis. Bull. World Health Organ. 1999, 77, 929–935. [Google Scholar] [PubMed]
  31. Eyles, D.E.; Coleman, N. Antibiotics in the Treatment of Toxoplasmosis. Am. J. Trop. Med. Hyg. 1953, 2, 64–69. [Google Scholar] [CrossRef]
  32. Warren, J.; Sabin, A.B. Effect of Certain Antiprotozoal Drugs on Toxoplasma In Vitro and In Vivo. Proc. Soc. Exp. Biol. Med. 1942, 51, 15–18. Available online: https://journals.sagepub.com/doi/abs/10.3181/00379727-51-13808 (accessed on 7 May 2024). [CrossRef]
  33. Garin, J.P.; Eyles, D.E. [Spiramycin therapy of experimental toxoplasmosis in mice]. Presse Med. 1958, 66, 957–958. [Google Scholar]
  34. Araujo, F.G.; Remington, J.S. Effect of Clindamycin on Acute and Chronic Toxoplasmosis in Mice. Antimicrob. Agents Chemother. 1974, 5, 647–651. [Google Scholar] [CrossRef]
  35. Tabbara, K.F.; O’Connor, G.R. Treatment of Ocular Toxoplasmosis with Clindamycin and Sulfadiazine. Ophthalmology 1980, 87, 129–134. [Google Scholar] [CrossRef]
  36. Effect on Lambing Percentage of Vaccinating Ewes with Toxoplasma gondii—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/16030917/ (accessed on 7 May 2024).
  37. Jokelainen, P.; Nylund, M. Acute Fatal Toxoplasmosis in Three Eurasian Red Squirrels (Sciurus Vulgaris) Caused by Genotype II of Toxoplasma gondii. J. Wildl. Dis. 2012, 48, 454–457. [Google Scholar] [CrossRef]
  38. Anderson, D.C.; McClure, H.M. Acute Disseminated Fatal Toxoplasmosis in a Squirrel Monkey. J. Am. Vet. Med. Assoc. 1982, 181, 1363–1366. [Google Scholar] [CrossRef] [PubMed]
  39. Riemann, H.P.; Fowler, M.E.; Schulz, T.; Lock, A.; Thilsted, J.; Pulley, L.T.; Henrickson, R.V.; Henness, A.M.; Franti, C.E.; Behymer, D.E. Toxoplasmosis in Pallas Cats. J. Wildl. Dis. 1974, 10, 471–477. [Google Scholar] [CrossRef] [PubMed]
  40. Howerth, E.W.; Rodenroth, N. Fatal Systemic Toxoplasmosis in a Wild Turkey. J. Wildl. Dis. 1985, 21, 446–449. [Google Scholar] [CrossRef] [PubMed]
  41. Hartley, W.J.; Dubey, J.P.; Spielman, D.S. Fatal Toxoplasmosis in Koalas (Phascolarctos cinereus). J. Parasitol. 1990, 76, 271–272. [Google Scholar] [CrossRef]
  42. Migaki, G.; Sawa, T.R.; Dubey, J.P. Fatal Disseminated Toxoplasmosis in a Spinner Dolphin (Stenella longirostris). Vet. Pathol. 1990, 27, 463–464. [Google Scholar] [CrossRef]
  43. Geisel, O.; Breuer, W.; Minkus, G.; Hermanns, W. [Toxoplasmosis causing death in a mole (Talpa europaea)]. Berl. Munch. Tierarztl. Wochenschr. 1995, 108, 241–243. [Google Scholar]
  44. Pertz, C.; Dubielzig, R.R.; Lindsay, D.S. Fatal Toxoplasma gondii Infection in Golden Lion Tamarins (Leontopithecus rosalia rosalia). J. Zoo. Wildl. Med. 1997, 28, 491–493. [Google Scholar]
  45. Mikaelian, I.; Dubey, J.P.; Martineau, D. Severe Hepatitis Resulting from Toxoplasmosis in a Barred Owl (Strix varia) from Québec, Canada. Avian Dis. 1997, 41, 738–740. [Google Scholar] [CrossRef]
  46. Cole, R.A.; Lindsay, D.S.; Howe, D.K.; Roderick, C.L.; Dubey, J.P.; Thomas, N.J.; Baeten, L.A. Biological and Molecular Characterizations of Toxoplasma gondii Strains Obtained from Southern Sea Otters (Enhydra Lutris Nereis). J. Parasitol. 2000, 86, 526–530. [Google Scholar] [CrossRef] [PubMed]
  47. Work, T.M.; Massey, J.G.; Rideout, B.A.; Gardiner, C.H.; Ledig, D.B.; Kwok, O.C.; Dubey, J.P. Fatal Toxoplasmosis in Free-Ranging Endangered ’Alala from Hawaii. J. Wildl. Dis. 2000, 36, 205–212. [Google Scholar] [CrossRef] [PubMed]
  48. Dubey, J.P.; Parnell, P.G.; Sreekumar, C.; Vianna, M.C.B.; De Young, R.W.; Dahl, E.; Lehmann, T. Biologic and Molecular Characteristics of Toxoplasma gondii Isolates from Striped Skunk (Mephitis mephitis), Canada Goose (Branta canadensis), Black-Winged Lory (Eos cyanogenia), and Cats (Felis catus). J. Parasitol. 2004, 90, 1171–1174. [Google Scholar] [CrossRef] [PubMed]
  49. Forzán, M.J.; Frasca, S. Systemic Toxoplasmosis in a Five-Month-Old Beaver, (Castor canadensis). J. Zoo. Wildl. Med. 2004, 35, 113–115. [Google Scholar] [CrossRef]
  50. Sedlák, K.; Bártová, E.; Literák, I.; Vodicka, R.; Dubey, J.P. Toxoplasmosis in Nilgais (Boselaphus tragocamelus) and a Saiga Antelope (Saiga tatarica). J. Zoo. Wildl. Med. 2004, 35, 530–533. [Google Scholar] [CrossRef]
  51. Szabo, K.A.; Mense, M.G.; Lipscomb, T.P.; Felix, K.J.; Dubey, J.P. Fatal Toxoplasmosis in a Bald Eagle (Haliaeetus leucocephalus). J. Parasitol. 2004, 90, 907–908. [Google Scholar] [CrossRef]
  52. Honnold, S.P.; Braun, R.; Scott, D.P.; Sreekumar, C.; Dubey, J.P. Toxoplasmosis in a Hawaiian Monk Seal (Monachus schauinslandi). J. Parasitol. 2005, 91, 695–697. [Google Scholar] [CrossRef]
  53. Gerhold, R.W.; Howerth, E.W.; Lindsay, D.S. Sarcocystis Neurona-Associated Meningoencephalitis and Description of Intramuscular Sarcocysts in a Fisher (Martes pennanti). J. Wildl. Dis. 2005, 41, 224–230. [Google Scholar] [CrossRef]
  54. Bangari, D.S.; Mouser, P.; Miller, M.A.; Stevenson, G.W.; Vemulapalli, R.; Thacker, H.L. Toxoplasmosis in a Woodchuck (Marmota monax) and Two American Red Squirrels (Tamiasciurus hudsonicus). J. Vet. Diagn. Investig. 2007, 19, 705–709. [Google Scholar] [CrossRef]
  55. Pas, A.; Dubey, J.P. Fatal Toxoplasmosis in Sand Cats (Felis margarita). J. Zoo. Wildl. Med. 2008, 39, 362–369. [Google Scholar] [CrossRef]
  56. Pas, A.; Dubey, J.P. Seroprevalence of Antibodies to Toxoplasma gondii in Gordon’s Wildcat (Felis Silvestris Gordoni) in the Middle East. J. Parasitol. 2008, 94, 1169. [Google Scholar] [CrossRef]
  57. Las, R.D.; Shivaprasad, H.L. An Outbreak of Toxoplasmosis in an Aviary Collection of Nicobar Pigeons (Caloenas Nicobaria). J. S. Afr. Vet. Assoc. 2008, 79, 149–152. [Google Scholar]
  58. Pena, H.F.J.; Marvulo, M.F.V.; Horta, M.C.; Silva, M.A.; Silva, J.C.R.; Siqueira, D.B.; Lima, P.-A.C.P.; Vitaliano, S.N.; Gennari, S.M. Isolation and Genetic Characterisation of Toxoplasma gondii from a Red-Handed Howler Monkey (Alouatta belzebul), a Jaguarundi (Puma yagouaroundi), and a Black-Eared Opossum (Didelphis aurita) from Brazil. Vet. Parasitol. 2011, 175, 377–381. [Google Scholar] [CrossRef] [PubMed]
  59. Ploeg, M.; Ultee, T.; Kik, M. Disseminated Toxoplasmosis in Black-Footed Penguins (Spheniscus demersus). Avian Dis. 2011, 55, 701–703. [Google Scholar] [CrossRef] [PubMed]
  60. Sangster, C.R.; Gordon, A.N.; Hayes, D. Systemic Toxoplasmosis in Captive Flying-Foxes. Aust. Vet. J. 2012, 90, 140–142. [Google Scholar] [CrossRef] [PubMed]
  61. Mancianti, F.; Nardoni, S.; Mugnaini, L.; Poli, A. Toxoplasma gondii in Waterfowl: The First Detection of This Parasite in Anas Crecca and Anas Clypeata from Italy. J. Parasitol. 2013, 99, 561–563. [Google Scholar] [CrossRef]
  62. Ma, H.; Wang, Z.; Wang, C.; Li, C.; Wei, F.; Liu, Q. Fatal Toxoplasma gondii Infection in the Giant Panda. Parasite 2015, 22, 30. [Google Scholar] [CrossRef]
  63. Donahoe, S.L.; Šlapeta, J.; Knowles, G.; Obendorf, D.; Peck, S.; Phalen, D.N. Clinical and Pathological Features of Toxoplasmosis in Free-Ranging Common Wombats (Vombatus ursinus) with Multilocus Genotyping of Toxoplasma gondii Type II-like Strains. Parasitol. Int. 2015, 64, 148–153. [Google Scholar] [CrossRef]
  64. Fayyad, A.; Kummerfeld, M.; Davina, I.; Wohlsein, P.; Beineke, A.; Baumgärtner, W.; Puff, C. Fatal Systemic Toxoplasma gondii Infection in a Red Squirrel (Sciurus vulgaris), a Swinhoe’s Striped Squirrel (Tamiops swinhoei) and a New World Porcupine (Erethizontidae sp.). J. Comp. Pathol. 2016, 154, 263–267. [Google Scholar] [CrossRef]
  65. Torres-Castro, M.; Noh-Pech, H.; Puerto-Hernández, R.; Reyes-Hernández, B.; Panti-May, A.; Hernández-Betancourt, S.; Yeh-Gorocica, A.; González-Herrera, L.; Zavala-Castro, J.; Puerto, F. First Molecular Evidence of Toxoplasma gondii in Opossums (Didelphis Virginiana) from Yucatan, Mexico. Open Vet. J. 2016, 6, 57. [Google Scholar] [CrossRef]
  66. Kumar, A.; Melotti, J.R.; Cooley, T.M.; Fitzgerald, S.D. Mortality Due to Toxoplasmosis in Suburban Eastern Fox Squirrels (Sciurus Niger) in Michigan, USA. J. Wildl. Dis. 2019, 55, 213–217. [Google Scholar] [CrossRef] [PubMed]
  67. Toxoplasma gondii Infections in Captive Black-Footed Ferrets (Mustela nigripes), 1992–1998: Clinical Signs, Serology, Pathology, and Prevention—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/14733273/ (accessed on 8 May 2024).
  68. Bonser, G.M. Fatal Intussusception of the Canine Caecum Associated with the Presence of Toxoplasma in the Circular Muscle of the Ileum. J. Pathol. Bacteriol. 1950, 62, 650–653. [Google Scholar] [CrossRef] [PubMed]
  69. Toxoplasmosis In Mink—PMC. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1614573/ (accessed on 8 May 2024).
  70. Reed, W.M.; Turek, J.J. Concurrent Distemper and Disseminated Toxoplasmosis in a Red Fox. J. Am. Vet. Med. Assoc. 1985, 187, 1264–1265. [Google Scholar] [CrossRef] [PubMed]
  71. Farrell, R.L.; Docton, F.L.; Chamberlain, D.M.; Cole, C.R. Toxoplasmosis. I. Toxoplasma Isolated from Swine. Am. J. Vet. Res. 1952, 13, 181–185. [Google Scholar]
  72. Hartley, W.J.; Marshall, S.C. Toxoplasmosis as a Cause of Ovine Perinatal Mortality. N. Z. Vet. J. 1957, 5, 119–124. [Google Scholar] [CrossRef]
  73. Pande, P.G.; Shukla, R.R.; Sekariah, P.C. Toxoplasma from the Eggs of the Domestic Fowl (Gallus Gallus). Science 1961, 133, 648. [Google Scholar] [CrossRef]
  74. Munday, B.L.; Mason, R.W. Toxoplasmosis as a Cause of Perinatal Death in Goats. Aust. Vet. J. 1979, 55, 485–487. [Google Scholar] [CrossRef]
  75. Dubey, J.P.; Quinn, W.J.; Weinandy, D. Fatal Neonatal Toxoplasmosis in a Bobcat (Lynx Rufus). J. Wildl. Dis. 1987, 23, 324–327. [Google Scholar] [CrossRef]
  76. Hagemoser, W.A.; Dubey, J.P.; Thompson, J.R. Acute Toxoplasmosis in a Camel. J. Am. Vet. Med. Assoc. 1990, 196, 347. [Google Scholar] [CrossRef]
  77. Dubey, J.P.; Graham, D.H.; Dahl, E.; Hilali, M.; El-Ghaysh, A.; Sreekumar, C.; Kwok, O.C.H.; Shen, S.K.; Lehmann, T. Isolation and Molecular Characterization of Toxoplasma gondii from Chickens and Ducks from Egypt. Vet. Parasitol. 2003, 114, 89–95. [Google Scholar] [CrossRef]
  78. Lloyd, C.; Stidworthy, M.F. Acute Disseminated Toxoplasmosis in a Juvenile Cheetah (Acinonyx Jubatus). J. Zoo. Wildl. Med. 2007, 38, 475–478. [Google Scholar] [CrossRef] [PubMed]
  79. Dubey, J.P.; Casey, S.J.; Zajac, A.M.; Wildeus, S.A.; Lindsay, D.S.; Verma, S.K.; Oliveira, S.; Kwok, O.C.H.; Su, C. Isolation and Genetic Characterization of Toxoplasma gondii from Alpaca (Vicugna Pacos) and Sheep (Ovis Aries). Trop. Anim. Health Prod. 2014, 46, 1503–1507. [Google Scholar] [CrossRef] [PubMed]
  80. Sarkari, B.; Asgari, Q.; Bagherian, N.; Ashkani Esfahani, S.; Kalantari, M.; Mohammadpour, I.; Ashrafmansori, M.; Amerinia, M.; Sabet Sarvestani, F. Molecular and Serological Evaluation of Toxoplasma gondii Infection in Reared Turkeys in Fars Province, Iran. Jundishapur J. Microbiol. 2014, 7, e11598. [Google Scholar] [CrossRef]
  81. Rong, G.; Zhou, H.-L.; Hou, G.-Y.; Zhao, J.-M.; Xu, T.-S.; Guan, S. Seroprevalence, Risk Factors and Genotyping of Toxoplasma gondii in Domestic Geese (Anser Domestica) in Tropical China. Parasit. Vectors 2014, 7, 459. [Google Scholar] [CrossRef] [PubMed]
  82. Jabbari, J.; Hajipour, N.; Hassanzadeh, P.; Ketzisc, J. Detection of Toxoplasma gondii Infection in Buffaloes (Bubalus Bubalis) and Cattle (Bos taurus) at the Tabriz Abattoir, Iran. Vet. Med. Sci. 2024, 10, e1511. [Google Scholar] [CrossRef]
  83. Edwards, J.F.; Dubey, J.P. Toxoplasma gondii Abortion Storm in Sheep on a Texas Farm and Isolation of Mouse Virulent Atypical Genotype T. Gondii from an Aborted Lamb from a Chronically Infected Ewe. Vet. Parasitol. 2013, 192, 129–136. [Google Scholar] [CrossRef]
  84. Fernández-Escobar, M.; Calero-Bernal, R.; Benavides, J.; Regidor-Cerrillo, J.; Guerrero-Molina, M.C.; Gutiérrez-Expósito, D.; Collantes-Fernández, E.; Ortega-Mora, L.M. Isolation and Genetic Characterization of Toxoplasma gondii in Spanish Sheep Flocks. Parasites Vectors 2020, 13, 396. [Google Scholar] [CrossRef]
  85. Gabardo, M.; Oliveira, J.; Ecco, R.; Guedes, R. Outbreak of Ovine Abortion by Toxoplasmosis in Southeastern Brazil. Braz. J. Vet. Pathol. 2013, 6, 37–41. [Google Scholar]
  86. Gutiérrez-Expósito, D.; Tejerina, F.; Gutiérrez, J.; Fernández-Escobar, M.; Ortega-Mora, L.M.; Mantecón, A.R.; Dagleish, M.P.; Pérez, V.; Benavides, J. Direct Economic Losses of Toxoplasma gondii Abortion Outbreaks in Two Spanish Sheep Flocks. Vet. Parasitol. Reg. Stud. Rep. 2021, 26, 100623. [Google Scholar] [CrossRef]
  87. Li, X.; Wang, Y.; Yu, F.; Li, T.; Zhang, D. An Outbreak of Lethal Toxoplasmosis in Pigs in the Gansu Province of China. J. Vet. Diagn. Investig. 2010, 22, 442–444. [Google Scholar] [CrossRef]
  88. Gelmetti, D.; Sironi, G.; Finazzi, M.; Gelmini, L.; Rosignoli, C.; Cordioli, P.; Lavazza, A. Diagnostic Investigations of Toxoplasmosis in Four Swine Herds. J. Vet. Diagn. Investig. 1999, 11, 87–90. [Google Scholar] [CrossRef]
  89. Roe, W.D.; Howe, L.; Baker, E.J.; Burrows, L.; Hunter, S.A. An Atypical Genotype of Toxoplasma gondii as a Cause of Mortality in Hector’s Dolphins (Cephalorhynchus hectori). Vet. Parasitol. 2013, 192, 67–74. [Google Scholar] [CrossRef]
  90. Dubey, J.P.; Mergl, J.; Gehring, E.; Sundar, N.; Velmurugan, G.V.; Kwok, O.C.H.; Grigg, M.E.; Su, C.; Martineau, D. Toxoplasmosis in Captive Dolphins (Tursiops truncatus) and Walrus (Odobenus rosmarus). J. Parasitol. 2009, 95, 82–85. [Google Scholar] [CrossRef] [PubMed]
  91. Barbieri, M.M.; Kashinsky, L.; Rotstein, D.S.; Colegrove, K.M.; Haman, K.H.; Magargal, S.L.; Sweeny, A.R.; Kaufman, A.C.; Grigg, M.E.; Littnan, C.L. Protozoal-Related Mortalities in Endangered Hawaiian Monk Seals Neomonachus Schauinslandi. Dis. Aquat. Organ. 2016, 121, 85–95. [Google Scholar] [CrossRef]
  92. McAllister, R.A. An Outbreak of Toxoplasmosis in an Ontario Chinchilla Herd. Can. J. Comp. Med. Vet. Sci. 1964, 28, 53–56. [Google Scholar] [PubMed]
  93. Burns, R.; Williams, E.S.; O’Toole, D.; Dubey, J.P. Toxoplasma gondii Infection in Captive Black-Footed Ferrets (Mustela nigripes), 1992-1998: Clinical Signs, Serology, Pathology, and Pevention. J. Wildl. Dis. 2003, 39, 787–797. [Google Scholar] [CrossRef] [PubMed]
  94. Sós, E.; Szigeti, A.; Fok, E.; Molnár, V.; Erdélyi, K.; Perge, E.; Biksi, I.; Gál, J. Toxoplasmosis in Tammar Wallabies (Macropus Eugenii) in the Budapest Zoo and Botanical Garden (2006–2010). Acta Vet. Hung. 2012, 60, 361–370. [Google Scholar] [CrossRef]
  95. Guthrie, A.; Rooker, L.; Tan, R.; Gerhold, R.; Trainor, K.; Jiang, T.; Su, C. Newly Described Toxoplasma gondii Strain Causes High Mortality in Red Necked Wallabies (Macropus rufogriseus) in a Zoo. J. Zoo. Wildl. Med. 2017, 48, 694–702. [Google Scholar] [CrossRef]
  96. Verma, S.K.; Knowles, S.; Cerqueira-Cézar, C.K.; Kwok, O.C.; Jiang, T.; Su, C.; Dubey, J.P. An Update on Toxoplasma gondii Infections in Northern Sea Otters (Enhydra Lutris Kenyoni) from Washington State, USA. Vet. Parasitol. 2018, 258, 133–137. [Google Scholar] [CrossRef]
  97. Frank, R.K. An Outbreak of Toxoplasmosis in Farmed Mink (Mustela Vison S.). J. Vet. Diagn. Investig. 2001, 13, 245–249. [Google Scholar] [CrossRef]
  98. Bouer, A.; Werther, K.; Catão-Dias, J.L.; Nunes, A.L.V. Outbreak of Toxoplasmosis in Lagothrix Lagotricha. IJFP 1999, 70, 282–285. [Google Scholar] [CrossRef] [PubMed]
  99. Carme, B.; Ajzenberg, D.; Demar, M.; Simon, S.; Dardé, M.L.; Maubert, B.; de Thoisy, B. Outbreaks of Toxoplasmosis in a Captive Breeding Colony of Squirrel Monkeys. Vet. Parasitol. 2009, 163, 132–135. [Google Scholar] [CrossRef] [PubMed]
  100. Santana, C.H.; de Oliveira, A.R.; Dos Santos, D.O.; Pimentel, S.P.; de Souza, L.D.R.; Moreira, L.G.A.; Braz, H.M.B.; de Carvalho, T.P.; Lopes, C.E.B.; Oliveira, J.B.S.; et al. Genotyping of Toxoplasma gondii in a Lethal Toxoplasmosis Outbreak Affecting Captive Howler Monkeys (Alouatta Sp.). J. Med. Primatol. 2021, 50, 99–107. [Google Scholar] [CrossRef] [PubMed]
  101. Vickers, M.C.; Hartley, W.J.; Mason, R.W.; Dubey, J.P.; Schollam, L. Blindness Associated with Toxoplasmosis in Canaries. J. Am. Vet. Med. Assoc. 1992, 200, 1723–1725. [Google Scholar] [CrossRef]
  102. Vielmo, A.; Pena, H.F.J.; Panziera, W.; Bianchi, R.M.; De Lorenzo, C.; Oliveira, S.; Alves, B.F.; Gennari, S.M.; Pavarini, S.P.; de Barros, C.S.L.; et al. Outbreak of Toxoplasmosis in a Flock of Domestic Chickens (Gallus Gallus Domesticus) and Guinea Fowl (Numida Meleagris). Parasitol. Res. 2019, 118, 991–997. [Google Scholar] [CrossRef]
  103. Erichsen, S.; Harboe, A. Toxoplasmosis in Chickens: I. An Epidemic Outbreak of Toxoplasmosis in a Chicken Flock in South-Eastern Norway. Acta Pathol. Microbiol. Scand. 1953, 33, 56–71. [Google Scholar] [CrossRef]
  104. Shimizu, K. Studies on Toxoplasmosis I: An Outbreak of Toxoplasmosis among Hares (Lepus Timidus Ainu) in Sapporo. Jpn. J. Vet. Res. 1958, 6, 157–166. [Google Scholar] [CrossRef]
  105. Oh, H.; Eo, K.; Gumber, S.; Hong, J.J.; Kim, C.; Lee, H.; Jung, Y.; Kim, J.; Whang, G.; Lee, J.; et al. An Outbreak of Toxoplasmosis in Squirrel Monkeys (Saimiri sciureus) in South Korea. J. Med. Primatol. 2018, 47, 238–246. [Google Scholar] [CrossRef]
  106. Nishimura, M.; Goyama, T.; Tomikawa, S.; Fereig, R.M.; El-Alfy, E.-S.N.; Nagamune, K.; Kobayashi, Y.; Nishikawa, Y. Outbreak of Toxoplasmosis in Four Squirrel Monkeys (Saimiri sciureus) in Japan. Parasitol. Int. 2019, 68, 79–86. [Google Scholar] [CrossRef]
  107. English, E.D.; Striepen, B. The Cat Is out of the Bag: How Parasites Know Their Hosts. PLoS Biol. 2019, 17, e3000446. [Google Scholar] [CrossRef]
  108. Sasai, M.; Yamamoto, M. Innate, Adaptive, and Cell-Autonomous Immunity against Toxoplasma gondii Infection. Exp. Mol. Med. 2019, 51, 1–10. [Google Scholar] [CrossRef]
  109. Kumar, P.; Tomita, T.; Gerken, T.A.; Ballard, C.J.; Lee, Y.S.; Weiss, L.M.; Samara, N.L. A Toxoplasma gondii O-Glycosyltransferase That Modulates Bradyzoite Cyst Wall Rigidity Is Distinct from Host Homologues. Nat. Commun. 2024, 15, 3792. [Google Scholar] [CrossRef] [PubMed]
  110. Lyons, R.E.; McLeod, R.; Roberts, C.W. Toxoplasma gondii Tachyzoite-Bradyzoite Interconversion. Trends Parasitol. 2002, 18, 198–201. [Google Scholar] [CrossRef]
  111. Dubey, J.P. Schizogony and Gametogony of Oocyst-Deficient T-263 Strain of Toxoplasma gondii. Vet. Parasitol. 2017, 245, 160–162. [Google Scholar] [CrossRef] [PubMed]
  112. Benavides, J.; Maley, S.; Pang, Y.; Palarea, J.; Eaton, S.; Katzer, F.; Innes, E.A.; Buxton, D.; Chianini, F. Development of Lesions and Tissue Distribution of Parasite in Lambs Orally Infected with Sporulated Oocysts of Toxoplasma gondii. Vet. Parasitol. 2011, 179, 209–215. [Google Scholar] [CrossRef] [PubMed]
  113. Shapiro, K.; Bahia-Oliveira, L.; Dixon, B.; Dumètre, A.; de Wit, L.A.; VanWormer, E.; Villena, I. Environmental Transmission of Toxoplasma gondii: Oocysts in Water, Soil and Food. Food Waterborne Parasitol. 2019, 15, e00049. [Google Scholar] [CrossRef]
  114. Attias, M.; Teixeira, D.E.; Benchimol, M.; Vommaro, R.C.; Crepaldi, P.H.; De Souza, W. The Life-Cycle of Toxoplasma gondii Reviewed Using Animations. Parasites Vectors 2020, 13, 588. [Google Scholar] [CrossRef]
  115. Dubey, J.P.; Lindsay, D.S.; Speer, C. Structures of Toxoplasma gondii Tachyzoites, Bradyzoites, and Sporozoites and Biology and Development of Tissue Cysts. Clin. Microbiol. Rev. 1998, 11, 267–299. [Google Scholar] [CrossRef]
  116. Boucher, L.E.; Bosch, J. The Apicomplexan Glideosome and Adhesins—Structures and Function. J. Struct. Biol. 2015, 190, 93–114. [Google Scholar] [CrossRef]
  117. Kumar, A.; Vadas, O.; Dos Santos Pacheco, N.; Zhang, X.; Chao, K.; Darvill, N.; Rasmussen, H.Ø.; Xu, Y.; Lin, G.M.-H.; Stylianou, F.A.; et al. Structural and Regulatory Insights into the Glideosome-Associated Connector from Toxoplasma gondii. eLife 2023, 12, e86049. [Google Scholar] [CrossRef]
  118. Jacot, D.; Frénal, K.; Marq, J.-B.; Sharma, P.; Soldati-Favre, D. Assessment of Phosphorylation in Toxoplasma Glideosome Assembly and Function. Cell Microbiol. 2014, 16, 1518–1532. [Google Scholar] [CrossRef]
  119. Opitz, C.; Soldati, D. “The Glideosome”: A Dynamic Complex Powering Gliding Motion and Host Cell Invasion by Toxoplasma gondii. Mol. Microbiol. 2002, 45, 597–604. [Google Scholar] [CrossRef] [PubMed]
  120. Keeley, A.; Soldati, D. The Glideosome: A Molecular Machine Powering Motility and Host-Cell Invasion by Apicomplexa. Trends Cell Biol. 2004, 14, 528–532. [Google Scholar] [CrossRef] [PubMed]
  121. Heintzelman, M.B. Gliding Motility in Apicomplexan Parasites. Semin. Cell Dev. Biol. 2015, 46, 135–142. [Google Scholar] [CrossRef]
  122. Kato, K. How Does Toxoplama Gondii Invade Host Cells? J. Vet. Med. Sci. 2018, 80, 1702–1706. [Google Scholar] [CrossRef]
  123. Nasiru Wana, M.; Mohd Moklas, M.A.; Watanabe, M.; Nordin, N.; Zasmy Unyah, N.; Alhassan Abdullahi, S.; Ahmad Issa Alapid, A.; Mustapha, T.; Basir, R.; Roslaini, A.M. A Review on the Prevalence of Toxoplasma gondii in Humans and Animals Reported in Malaysia from 2008–2018. Int. J. Environ. Res. Public Health 2020, 17, 4809. [Google Scholar] [CrossRef] [PubMed]
  124. Jiang, Y.; Shi, Y.; Xue, Y.; Hu, D.; Song, X. AP2XII-1 and AP2XI-2 Suppress Schizogony Gene Expression in Toxoplasma gondii. Int. J. Mol. Sci. 2024, 25, 5527. [Google Scholar] [CrossRef]
  125. Tomasina, R.; Francia, M.E. The Structural and Molecular Underpinnings of Gametogenesis in Toxoplasma gondii. Front. Cell Infect. Microbiol. 2020, 10, 608291. [Google Scholar] [CrossRef]
  126. Ferguson, D.J.; Hutchison, W.M.; Siim, J.C. The Ultrastructural Development of the Macrogamete and Formation of the Oocyst Wall of Toxoplasma gondii. Acta Pathol. Microbiol. Scand. B 1975, 83, 491–505. [Google Scholar] [CrossRef]
  127. Dubey, J.P.; Cerqueira-Cézar, C.K.; Murata, F.H.A.; Kwok, O.C.H.; Yang, Y.R.; Su, C. All about Toxoplasmosis in Cats: The Last Decade. Vet. Parasitol. 2020, 283, 109145. [Google Scholar] [CrossRef]
  128. Hatam-Nahavandi, K.; Calero-Bernal, R.; Rahimi, M.T.; Pagheh, A.S.; Zarean, M.; Dezhkam, A.; Ahmadpour, E. Toxoplasma gondii Infection in Domestic and Wild Felids as Public Health Concerns: A Systematic Review and Meta-Analysis. Sci. Rep. 2021, 11, 9509. [Google Scholar] [CrossRef]
  129. Dubey, J.P.; Ferreira, L.R.; Martins, J.; Jones, J.L. Sporulation and Survival of Toxoplasma gondii Oocysts in Different Types of Commercial Cat Litter. J. Parasitol. 2011, 97, 751–754. [Google Scholar] [CrossRef] [PubMed]
  130. Ferguson, D.J.; Birch-Andersen, A.; Siim, J.C.; Hutchison, W.M. Ultrastructural Studies on the Sporulation of Oocysts of Toxoplasma gondii. I. Development of the Zygote and Formation of the Sporoblasts. Acta Pathol. Microbiol. Scand. B 1979, 87B, 171–181. [Google Scholar] [CrossRef] [PubMed]
  131. Hill, D.; Dubey, J.P. Toxoplasma gondii: Transmission, Diagnosis and Prevention. Clin. Microbiol. Infect. 2002, 8, 634–640. [Google Scholar] [CrossRef] [PubMed]
  132. Deganich, M.; Boudreaux, C.; Benmerzouga, I. Toxoplasmosis Infection during Pregnancy. Trop. Med. Infect. Dis. 2022, 8, 3. [Google Scholar] [CrossRef] [PubMed]
  133. Holec-Gąsior, L.; Sołowińska, K. Detection of Toxoplasma gondii Infection in Small Ruminants: Old Problems, and Current Solutions. Animals 2023, 13, 2696. [Google Scholar] [CrossRef]
  134. Zhang, Y.; Lai, B.S.; Juhas, M.; Zhang, Y. Toxoplasma gondii Secretory Proteins and Their Role in Invasion and Pathogenesis. Microbiol. Res. 2019, 227, 126293. [Google Scholar] [CrossRef]
  135. Malkwitz, I.; Berndt, A.; Zhang, R.; Daugschies, A.; Bangoura, B. Replication of Toxoplasma gondii in Chicken Erythrocytes and Thrombocytes Compared to Macrophages. Parasitol. Res. 2017, 116, 123–131. [Google Scholar] [CrossRef]
  136. Clough, B.; Frickel, E.-M. The Toxoplasma Parasitophorous Vacuole: An Evolving Host-Parasite Frontier. Trends Parasitol. 2017, 33, 473–488. [Google Scholar] [CrossRef]
  137. Zhang, N.-Z.; Chen, J.; Wang, M.; Petersen, E.; Zhu, X.-Q. Vaccines against Toxoplasma gondii: New Developments and Perspectives. Expert Rev. Vaccines 2013, 12, 1287–1299. [Google Scholar] [CrossRef]
  138. Tomita, T.; Guevara, R.B.; Shah, L.M.; Afrifa, A.Y.; Weiss, L.M. Secreted Effectors Modulating Immune Responses to Toxoplasma gondii. Life 2021, 11, 988. [Google Scholar] [CrossRef]
  139. Castaño, P.; Fernández, M.; Regidor-Cerrillo, J.; Fuertes, M.; Horcajo, P.; Ferre, I.; Ferreras, M.C.; Ortega-Mora, L.M.; Pérez, V.; Benavides, J. Peripheral and Placental Immune Responses in Sheep after Experimental Infection with Toxoplasma gondii at the Three Terms of Gestation. Vet. Res. 2019, 50, 66. [Google Scholar] [CrossRef]
  140. Sana, M.; Rashid, M.; Rashid, I.; Akbar, H.; Gomez-Marin, J.E.; Dimier-Poisson, I. Immune Response against Toxoplasmosis—Some Recent Updates RH: Toxoplasma gondii Immune Response. Int. J. Immunopathol. Pharmacol. 2022, 36, 03946320221078436. [Google Scholar] [CrossRef]
  141. Lima, T.S.; Lodoen, M.B. Mechanisms of Human Innate Immune Evasion by Toxoplasma gondii. Front. Cell Infect. Microbiol. 2019, 9, 103. [Google Scholar] [CrossRef]
  142. Sasai, M.; Pradipta, A.; Yamamoto, M. Host Immune Responses to Toxoplasma gondii. Int. Immunol. 2018, 30, 113–119. [Google Scholar] [CrossRef]
  143. Sanchez, S.G.; Besteiro, S. The Pathogenicity and Virulence of Toxoplasma gondii. Virulence 2021, 12, 3095–3114. [Google Scholar] [CrossRef] [PubMed]
  144. Bando, H.; Murata, Y.; Han, Y.; Sugi, T.; Fukuda, Y.; Bzik, D.J.; Fox, B.A.; Kato, K. Toxoplasma gondii Chitinase-like Protein TgCLP1 Regulates the Parasite Cyst Burden. Front. Cell Infect. Microbiol. 2024, 14, 1359888. [Google Scholar] [CrossRef] [PubMed]
  145. Yang, Y.; Xin, S.; Murata, F.H.A.; Cerqueira-Cézar, C.K.; Kwok, O.C.H.; Su, C.; Dubey, J.P. Recent Epidemiologic, Clinical, Subclinical and Genetic Diversity of Toxoplasma gondii Infections in Bats. Res. Vet. Sci. 2021, 140, 193–197. [Google Scholar] [CrossRef] [PubMed]
  146. Milne, G.; Webster, J.P.; Walker, M. Toxoplasma gondii: AnUnderestimated Threat? Trends Parasitol. 2020, 36, 959–969. [Google Scholar] [CrossRef]
  147. Henriquez, S.A.; Brett, R.; Alexander, J.; Pratt, J.; Roberts, C.W. Neuropsychiatric Disease and Toxoplasma gondii Infection. Neuroimmunomodulation 2009, 16, 122–133. [Google Scholar] [CrossRef]
  148. Yang, Z.; Chen, J.; Zhang, C.; Peng, H. Pathological Mechanisms of Glial Cell Activation and Neurodegenerative and Neuropsychiatric Disorders Caused by Toxoplasma gondii Infection. Front. Microbiol. 2024, 15, 1512233. [Google Scholar] [CrossRef]
  149. Tyebji, S.; Seizova, S.; Hannan, A.J.; Tonkin, C.J. Toxoplasmosis: A Pathway to Neuropsychiatric Disorders. Neurosci. Biobehav. Rev. 2019, 96, 72–92. [Google Scholar] [CrossRef]
  150. Tong, W.H.; Pavey, C.; O’Handley, R.; Vyas, A. Behavioral Biology of Toxoplasma gondii Infection. Parasit. Vectors 2021, 14, 77. [Google Scholar] [CrossRef] [PubMed]
  151. Vyas, A. Mechanisms of Host Behavioral Change in Toxoplasma gondii Rodent Association. PLoS Pathog. 2015, 11, e1004935. [Google Scholar] [CrossRef] [PubMed]
  152. Li, Y.; Zhang, Y.; Xia, N.; Zhou, T.; Shen, B. Antitumor Effects of a Toxoplasma Mutant Lacking Lactate Dehydrogenases. Parasitol. Res. 2021, 120, 3335–3339. [Google Scholar] [CrossRef] [PubMed]
  153. Involvement of MyD88 in Host Defense and the Down-Regulation of Anti-Heat Shock Protein 70 Autoantibody Formation by MyD88 in Toxoplasma gondii-Infected Mice—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/12435148/ (accessed on 7 May 2024).
  154. T Cell Suppression in Vitro During Toxoplasma gondii Infection Is the Result of IL-2 Competition Between Tregs and T Cells Leading to Death of Proliferating T Cells—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/24117537/ (accessed on 7 May 2024).
  155. The Murine C-Rel Proto-Oncogene Encodes Two mRNAs the Expression of Which Is Modulated by Lymphoid Stimuli—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/2204017/ (accessed on 7 May 2024).
  156. Akbar, H.; Dimier-Poisson, I.; Moiré, N. Role of CD4+ Foxp3+ Regulatory T Cells in Protection Induced by a Live Attenuated, Replicating Type I Vaccine Strain of Toxoplasma gondii. Infect. Immun. 2015, 83, 3601–3611. [Google Scholar] [CrossRef]
  157. Mitchell, D.M.; Ravkov, E.V.; Williams, M.A. Distinct Roles for IL-2 and IL-15 in the Differentiation and Survival of CD8+ Effector and Memory T Cells. J. Immunol. 2010, 184, 6719–6730. [Google Scholar] [CrossRef]
  158. Benedetto, N.; Auriault, C.; Darcy, F.; Lando, D.; Watier, H.; Capron, A. Effect of rIFN-Gamma and IL-2 Treatments in Mouse and Nude Rat Infections with Toxoplasma gondii. Eur. Cytokine Netw. 1991, 2, 107–114. [Google Scholar]
  159. A Single Polymorphic Amino Acid on Toxoplasma gondii Kinase ROP16 Determines the Direct and Strain-Specific Activation of Stat3—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/19901082/ (accessed on 7 May 2024).
  160. Possibilities for Immunomodulation in Congenital Toxoplasmosis—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/27357341/ (accessed on 7 May 2024).
  161. TGF-Β1 Levels and Intraocular Tissue Alterations in Mice Infected with a Virulent Type I RH Toxoplasma gondii Strain—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/26773166/ (accessed on 7 May 2024).
  162. The Role of IL-27 in the Development of T-Cell Responses during Parasitic Infections—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/15546389/ (accessed on 7 May 2024).
  163. IL-6-Dependent Spontaneous Proliferation Is Required for the Induction of Colitogenic IL-17-Producing CD8+ T Cells—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/18426983/ (accessed on 7 May 2024).
  164. Transforming Growth Factor-Beta Inhibits Interleukin-12-Induced Production of Interferon-Gamma by Natural Killer Cells: A Role for Transforming Growth Factor-Beta in the Regulation of T Cell-Independent Resistance to Toxoplasma gondii—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/7737303/ (accessed on 7 May 2024).
  165. Langrish, C.L.; Chen, Y.; Blumenschein, W.M.; Mattson, J.; Basham, B.; Sedgwick, J.D.; McClanahan, T.; Kastelein, R.A.; Cua, D.J. IL-23 Drives a Pathogenic T Cell Population That Induces Autoimmune Inflammation. J. Exp. Med. 2005, 201, 233–240. [Google Scholar] [CrossRef]
  166. Tong, X.; Chen, S.; Zheng, H.; Huang, S.; Lu, F. Increased IL-27/IL-27R Expression in Association with the Immunopathology of Murine Ocular Toxoplasmosis. Parasitol. Res. 2018, 117, 2255–2263. [Google Scholar] [CrossRef]
  167. Clark, J.T.; Christian, D.A.; Gullicksrud, J.A.; Perry, J.A.; Park, J.; Jacquet, M.; Tarrant, J.C.; Radaelli, E.; Silver, J.; Hunter, C.A. IL-33 Promotes Innate Lymphoid Cell-Dependent IFN-γ Production Required for Innate Immunity to Toxoplasma gondii. Elife 2021, 10, e65614. [Google Scholar] [CrossRef] [PubMed]
  168. Zhao, M.; Zhang, H.; Liu, X.; Jiang, Y.; Ren, L.; Hu, X. The Effect of TGF-β on Treg Cells in Adverse Pregnancy Outcome upon Toxoplasma gondii Infection. Front. Microbiol. 2017, 8, 901. [Google Scholar] [CrossRef] [PubMed]
  169. Ayub, I.; Sani, S.S.; Zaman, A.; Shahid, M.R.; Bashir, W.; Ijaz, M.; Zafar, S.; Rafique, A. Prevalence of Toxoplasma gondii in Human and Animals and Its Economic Impacts: A Review. Pak. Veter-J. 2024, 45, 1–9. [Google Scholar] [CrossRef]
  170. Tenter, A.M.; Heckeroth, A.R.; Weiss, L.M. Toxoplasma gondii: From Animals to Humans. Int. J. Parasitol. 2000, 30, 1217–1258. [Google Scholar] [CrossRef]
  171. Döşkaya, M.; Caner, A.; Ajzenberg, D.; Değirmenci, A.; Dardé, M.-L.; Can, H.; Erdoğan, D.D.; Korkmaz, M.; Üner, A.; Güngör, Ç. Isolation of Toxoplasma gondii Strains Similar to Africa 1 Genotype in Turkey. Parasitol. Int. 2013, 62, 471–474. [Google Scholar] [CrossRef]
  172. Molan, A.; Nosaka, K.; Hunter, M.; Wang, W. Global Status of Toxoplasma gondii Infection: Systematic Review and Prevalence Snapshots. Trop. Biomed. 2019, 36, 898–925. [Google Scholar]
  173. Ceylan, C.; Sevinc, F.; Ceylan, O. Serostatus of Small Ruminant Toxoplasmosis and Neosporosis throughout the Southeastern Anatolia Region of Türkiye. Pak. Vet. J. 2024, 44, 917–923. [Google Scholar] [CrossRef]
  174. Jokelainen, P.; Murat, J.B.; Nielsen, H.V. Direct Genetic Characterization of Toxoplasma gondii from Clinical Samples from Denmark: Not Only Genotypes II and III. Eur. J. Clin. Microbiol. Infect. Dis. 2018, 37, 579–586. [Google Scholar] [CrossRef]
  175. Kalogeropoulos, D.; Sakkas, H.; Mohammed, B.; Vartholomatos, G.; Malamos, K.; Sreekantam, S.; Kanavaros, P.; Kalogeropoulos, C. Ocular Toxoplasmosis: A Review of the Current Diagnostic and Therapeutic Approaches. Int. Ophthalmol. 2022, 42, 295–321. [Google Scholar] [CrossRef]
  176. Fabiani, S.; Caroselli, C.; Menchini, M.; Gabbriellini, G.; Falcone, M.; Bruschi, F. Ocular Toxoplasmosis, an Overview Focusing on Clinical Aspects. Acta Trop. 2022, 225, 106180. [Google Scholar] [CrossRef]
  177. Calero-Bernal, R.; Gennari, S.M. Clinical Toxoplasmosis in Dogs and Cats: An Update. Front. Vet. Sci. 2019, 6, 54. [Google Scholar] [CrossRef] [PubMed]
  178. Smith, D.F.Q.; Casadevall, A. On the Relationship between Pathogenic Potential and Infective Inoculum. PLoS Pathog. 2022, 18, e1010484. [Google Scholar] [CrossRef] [PubMed]
  179. Flegr, J.; Horáček, J. Negative Effects of Latent Toxoplasmosis on Mental Health. Front. Psychiatry 2020, 10, 1012. [Google Scholar] [CrossRef] [PubMed]
  180. Al Malki, J.S.; Hussien, N.A.; Al Malki, F. Maternal Toxoplasmosis and the Risk of Childhood Autism: Serological a Nd Molecular Small-Scale Studies. BMC Pediatr. 2021, 21, 133. [Google Scholar] [CrossRef] [PubMed]
  181. Chaudhry, S.A.; Gad, N.; Koren, G. Toxoplasmosis and Pregnancy. Can. Fam. Physician 2014, 60, 334–336. [Google Scholar]
  182. Hampton, M.M. Congenital Toxoplasmosis: A Review. Neonatal Netw. 2015, 34, 274–278. [Google Scholar] [CrossRef]
  183. Robert-Gangneux, F.; Dardé, M.-L. Epidemiology of and Diagnostic Strategies for Toxoplasmosis. Clin. Microbiol. Rev. 2012, 25, 264–296. [Google Scholar] [CrossRef]
  184. Bollani, L.; Auriti, C.; Achille, C.; Garofoli, F.; De Rose, D.U.; Meroni, V.; Salvatori, G.; Tzialla, C. Congenital Toxoplasmosis: The State of the Art. Front. Pediatr. 2022, 10, 894573. [Google Scholar] [CrossRef]
  185. Dubey, J.P.; Murata, F.H.A.; Cerqueira-Cézar, C.K.; Kwok, O.C.H.; Villena, I. Congenital Toxoplasmosis in Humans: An Update of Worldwide Rate of Congenital Infections. Parasitology 2021, 148, 1406–1416. [Google Scholar] [CrossRef]
  186. Blanchard, N.; Dunay, I.R.; Schlüter, D. Persistence of Toxoplasma gondii in the Central Nervous System: A Fine-tuned Balance between the Parasite, the Brain and the Immune System. Parasite Immunol. 2015, 37, 150–158. [Google Scholar] [CrossRef]
  187. Kong, P.; Gupta, N. Phospholipid Biogenesis in the Apicomplexan Parasites Eimeria Falciformis and Toxoplasma gondii. Ph.D. Thesis, Humboldt-Universität zu Berlin, Berlin, Germany, 2017. [Google Scholar]
  188. Nitzsche, R.; Günay-Esiyok, Ö.; Tischer, M.; Zagoriy, V.; Gupta, N. A Plant/Fungal-Type Phosphoenolpyruvate Carboxykinase Located in the Parasite Mitochondrion Ensures Glucose-Independent Survival of Toxoplasma gondii. J. Biol. Chem. 2017, 292, 15225–15239. [Google Scholar] [CrossRef] [PubMed]
  189. Wallon, M.; Liou, C.; Garner, P.; Peyron, F. Congenital Toxoplasmosis: Systematic Review of Evidence of Efficacy of Treatment in Pregnancy. Bmj 1999, 318, 1511–1514. [Google Scholar] [CrossRef] [PubMed]
  190. Miyagaki, M.; Zong, Y.; Yang, M.; Zhang, J.; Zou, Y.; Ohno-Matsui, K.; Kamoi, K. Ocular Toxoplasmosis: Advances in Toxoplasma gondii Biology, Clinical Manifestations, Diagnostics, and Therapy. Pathogens 2024, 13, 898. [Google Scholar] [CrossRef]
  191. Smith, J.R.; Ashander, L.M.; Arruda, S.L.; Cordeiro, C.A.; Lie, S.; Rochet, E.; Belfort, R.J.; Furtado, J.M. Pathogenesis of Ocular Toxoplasmosis. Prog. Retin. Eye Res. 2021, 81, 100882. [Google Scholar] [CrossRef] [PubMed]
  192. Fernández, C.; Jaimes, J.; Ortiz, M.C.; Ramírez, J.D. Host and Toxoplasma gondii Genetic and Non-Genetic Factors Influencing the Development of Ocular Toxoplasmosis: A Systematic Review. Infect. Genet. Evol. 2016, 44, 199–209. [Google Scholar] [CrossRef]
  193. Grigg, M.E.; Dubey, J.P.; Nussenblatt, R.B. Ocular Toxoplasmosis: Lessons from Brazil. Am. J. Ophthalmol. 2015, 159, 999–1001. [Google Scholar] [CrossRef]
  194. Ventura, L.; Zanelli, M.; Zizzo, M.; Sanguedolce, F.; Martino, G.; Castro Ruiz, C.; Annessi, V.; Ascani, S. Toxoplasma Cyst Detection in Piringer-Kuchinka Lymphadenitis. Report of Two Cases and Literature Review. Pathologica 2021, 113, 126–130. [Google Scholar] [CrossRef]
  195. Derouin, F.; Pelloux, H.; ESCMID Study Group on Clinical Parasitology. Prevention of Toxoplasmosis in Transplant Patients. Clin. Microbiol. Infect. 2008, 14, 1089–1101. [Google Scholar] [CrossRef]
  196. Bledsoe, J.R.; Hasserjian, R.P. Toxoplasma Lymphadenitis. In Hematopathology; Springer: Cham, Switzerland, 2020; pp. 495–500. ISBN 3-319-95308-7. [Google Scholar]
  197. Mele, A.; Paterson, P.; Prentice, H.; Leoni, P.; Kibbler, C. Toxoplasmosis in Bone Marrow Transplantation: A Report of Two Cases and Systematic Review of the Literature. Bone Marrow Transplant. 2002, 29, 691–698. [Google Scholar] [CrossRef]
  198. Loscalzo, J.; Fauci, A.; Kasper, D.; Hauser, S.; Longo, D.; Jameson, J.L. Harrison’s Principles of Internal Medicine, 21st ed.; McGraw-Hill: New York, NY, USA; Health Professions Division: Fort Lauderdale, FL, USA, 2022. [Google Scholar]
  199. Moncada, P.A.; Montoya, J.G. Toxoplasmosis in the Fetus and Newborn: An Update on Prevalence, Diagnosis and Treatment. Expert Rev. Anti-Infect. Ther. 2012, 10, 815–828. [Google Scholar] [CrossRef]
  200. Weiss, L.M.; Dubey, J.P. Toxoplasmosis: A History of Clinical Observations. Int. J. Parasitol. 2009, 39, 895–901. [Google Scholar] [CrossRef]
  201. Basavaraju, A. Toxoplasmosis in HIV Infection: An Overview. Trop. Parasitol. 2016, 6, 129–135. [Google Scholar] [CrossRef]
  202. Khurana, S.; Batra, N. Toxoplasmosis in Organ Transplant Recipients: Evaluation, Implication, and Prevention. Trop. Parasitol. 2016, 6, 123–128. [Google Scholar] [CrossRef] [PubMed]
  203. Martino, R.; Maertens, J.; Bretagne, S.; Rovira, M.; Deconinck, E.; Ullmann, A.J.; Held, T.; Cordonnier, C.; European Group for Blood and Marrow Transplantation Infectious Diseases Working Party. Toxoplasmosis after Hematopoietic Stem Cell Transplantation. Clin. Infect. Dis. 2000, 31, 1188–1194. [Google Scholar] [CrossRef] [PubMed]
  204. Truppel, J.H.; Reifur, L.; Montiani-Ferreira, F.; Lange, R.R.; de Castro Vilani, R.G.D.; Gennari, S.M.; Thomaz-Soccol, V. Toxoplasma gondii in Capybara (Hydrochaeris Hydrochaeris) Antibodies and DNA Detected by IFAT and PCR. Parasitol. Res. 2010, 107, 141–146. [Google Scholar] [CrossRef] [PubMed]
  205. Quirk, T.; Dubey, J.P. Experimental Toxoplasma gondii Infection in Striped Skunk (Mephitis mephitis). J. Parasitol. 2008, 94, 761–763. [Google Scholar] [CrossRef]
  206. Jensen, S.K.; Aars, J.; Lydersen, C.; Kovacs, K.M.; Åsbakk, K. The Prevalence of Toxoplasma gondii in Polar Bears and Their Marine Mammal Prey: Evidence for a Marine Transmission Pathway? Polar Biol. 2010, 33, 599–606. [Google Scholar] [CrossRef]
  207. Galeh, T.M.; Sarvi, S.; Montazeri, M.; Moosazadeh, M.; Nakhaei, M.; Shariatzadeh, S.A.; Daryani, A. Global Status of Toxoplasma gondii Seroprevalence in Rodents: A Systematic Review and Meta-Analysis. Front. Vet. Sci. 2020, 7, 461. [Google Scholar] [CrossRef]
  208. Nava, M.; Corlatti, L.; Formenti, N.; Trogu, T.; Pedrotti, L.; Gugiatti, A.; Lanfranchi, P.; Luzzago, C.; Ferrari, N. Parasite-Mediated Manipulation? Toxoplasma gondii Infection Increases Risk Behaviour towards Culling in Red Deer. Biol. Lett. 2023, 19, 20230292. [Google Scholar] [CrossRef]
  209. French, S.K.; Pearl, D.L.; Stevens, B.; Peregrine, A.S.; Jardine, C.M. Morbidity and Mortality in Ontario Rodents and Lagomorphs: A 30-Year Retrospective Review. J. Wildl. Dis. 2021, 57, 874–883. [Google Scholar] [CrossRef]
  210. Karadjian, G.; Laboutière, L.; Chevillot, A.; Voisinot, A.; Blaizot, A.; Puech, M.-P.; Aubert, D.; Risco-Castillo, V.; Blaga, R.; Vallée, I. Toxocara Cati and Toxoplasma gondii in French Birds of Prey. J. Wildl. Dis. 2022, 58, 373–379. [Google Scholar] [CrossRef]
  211. Canfield, P.J.; Hartley, W.J.; Dubey, J.P. Lesions of Toxoplasmosis in Australian Marsupials. J. Comp. Pathol. 1990, 103, 159–167. [Google Scholar] [CrossRef]
  212. Shapiro, K.; VanWormer, E.; Packham, A.; Dodd, E.; Conrad, P.A.; Miller, M. Type X Strains of Toxoplasma gondii Are Virulent for Southern Sea Otters (Enhydra Lutris Nereis) and Present in Felids from Nearby Watersheds. Proc. Biol. Sci. 2019, 286, 20191334. [Google Scholar] [CrossRef]
  213. Epiphanio, S.; Sinhorini, I.L.; Catão-Dias, J.L. Pathology of Toxoplasmosis in Captive New World Primates. J. Comp. Pathol. 2003, 129, 196–204. [Google Scholar] [CrossRef] [PubMed]
  214. Denk, D.; De Neck, S.; Khaliq, S.; Stidworthy, M.F. Toxoplasmosis in Zoo Animals: A Retrospective Pathology Review of 126 Cases. Animals 2022, 12, 619. [Google Scholar] [CrossRef] [PubMed]
  215. Jordan, C.N.; Kaur, T.; Koenen, K.; DeStefano, S.; Zajac, A.M.; Lindsay, D.S. Prevalence of Agglutinating Antibodies to Toxoplasma gondii and Sarcocystis Neurona in Beavers (Castor Canadensis) from Massachusetts. J. Parasitol. 2005, 91, 1228–1229. [Google Scholar] [CrossRef] [PubMed]
  216. Almeria, S.; Murata, F.H.; Cerqueira-Cézar, C.K.; Kwok, O.C.; Shipley, A.; Dubey, J.P. Epidemiological and Public Health Significance of Toxoplasma gondii Infection in Wild Rabbits and Hares: 2010–2020. Microorganisms 2021, 9, 597. [Google Scholar] [CrossRef]
  217. Wilson, A.G.; Lapen, D.R.; Mitchell, G.W.; Provencher, J.F.; Wilson, S. Interaction of Diet and Habitat Predicts Toxoplasma gondii Infection Rates in Wild Birds at a Global Scale. Glob. Ecol. Biogeogr. 2020, 29, 1189–1198. [Google Scholar] [CrossRef]
  218. Bouchard, É.; Sharma, R.; Bachand, N.; Gajadhar, A.A.; Jenkins, E.J. Pathology, Clinical Signs, and Tissue Distribution of Toxoplasma gondii in Experimentally Infected Reindeer (Rangifer Tarandus). Int. J. Parasitol.: Parasites Wildl. 2017, 6, 234–240. [Google Scholar] [CrossRef]
  219. Fatal Acute Toxoplasmosis in Three Golden Lion Tamarins (Leontopithecus Rosalia)—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/9638627/ (accessed on 8 May 2024).
  220. Reis Amendoeira, M.R.; Arruda, I.F.; Moreira, S.B.; Ubiali, D.G.; da Silva Barbosa, A.; Jesus Pena, H.F.; Barbosa Pereira, A.H.; Nascimento da Silveira, C.; Bonifácio, T.F.; Clemes, Y.S.; et al. Isolation and Genetic Characterization of Toxoplasma gondii from a Captive Black-and-Gold Howler Monkey (Alouatta Caraya Humboldt, 1812) in Brazil. Int. J. Parasitol. Parasites Wildl. 2022, 19, 187–190. [Google Scholar] [CrossRef]
  221. Moreira, S.B.; Pereira, A.H.B.; Pissinatti, T.D.A.; Arruda, I.F.; de Azevedo, R.R.M.; Schiffler, F.B.; Amendoeira, M.R.R.; Dos Santos, A.F.A.; Pissinatti, A.; Ubiali, D.G. Subacute Multisystemic Toxoplasmosis in a Captive Black-and-Gold Howler Monkey (Alouatta caraya) Indicates Therapy Challenging. J. Med. Primatol. 2022, 51, 392–395. [Google Scholar] [CrossRef] [PubMed]
  222. Santos, S.V.; Pena, H.F.J.; Talebi, M.G.; Teixeira, R.H.F.; Kanamura, C.T.; Diaz-Delgado, J.; Gennari, S.M.; Catão-Dias, J.L. Fatal Toxoplasmosis in a Southern Muriqui (Brachyteles arachnoides) from São Paulo State, Brazil: Pathological, Immunohistochemical, and Molecular Characterization. J. Med. Primatol. 2018, 47, 124–127. [Google Scholar] [CrossRef] [PubMed]
  223. Grandi, F.; Monteiro, L.N.; Salgado, B.S.; Rocha, R.M.; Nonogaki, S.; Rocha, N.S. What Is Your Diagnosis? Hepatosplenomegaly in a Howler Monkey (Alouatta fusca). Vet. Clin. Pathol. 2012, 41, 433–434. [Google Scholar] [CrossRef] [PubMed]
  224. Fiorello, C.V.; Heard, D.J.; Heller, H.L.B.; Russell, K. Medical Management of Toxoplasma Meningitis in a White-Throated Capuchin (Cebus capucinus). J. Zoo. Wildl. Med. 2006, 37, 409–412. [Google Scholar] [CrossRef]
  225. Cedillo-Peláez, C.; Rico-Torres, C.P.; Salas-Garrido, C.G.; Correa, D. Acute Toxoplasmosis in Squirrel Monkeys (Saimiri sciureus) in Mexico. Vet. Parasitol. 2011, 180, 368–371. [Google Scholar] [CrossRef]
  226. Inoue, M. Acute Toxoplasmosis in Squirrel Monkeys. J. Vet. Med. Sci. 1997, 59, 593–595. [Google Scholar] [CrossRef]
  227. (PDF) Toxoplasmosis in Wild Eastern Barred Bandicoots, Perameles gunnii. Available online: https://www.researchgate.net/publication/236942612_Toxoplasmosis_in_wild_eastern_barred_bandicoots_Perameles_gunnii (accessed on 8 May 2024).
  228. Attwood, H.D.; Woolley, P.A.; Rickard, M.D. Toxoplasmosis in Dasyurid Marsupials. J. Wildl. Dis. 1975, 11, 543–551. [Google Scholar] [CrossRef]
  229. Dubey, J.P.; Murata, F.H.A.; Cerqueira-Cézar, C.K.; Kwok, O.C.H.; Su, C.; Grigg, M.E. Recent Aspects on Epidemiology, Clinical Disease, and Genetic Diversity of Toxoplasma gondii Infections in Australasian Marsupials. Parasites Vectors 2021, 14, 301. [Google Scholar] [CrossRef]
  230. Portas, T.J. Toxoplasmosis in Macropodids: A Review. J. Zoo Wildl. Med. 2010, 41, 1–6. [Google Scholar] [CrossRef]
  231. Hartley, M.P. Toxoplasma gondii Infection in Two Common Wombats (Vombatus ursinus). Aust. Vet. J. 2006, 84, 107–109. [Google Scholar] [CrossRef]
  232. Hill, N.J.; Dubey, J.P.; Vogelnest, L.; Power, M.L.; Deane, E.M. Do Free-Ranging Common Brushtail Possums (Trichosurus Vulpecula) Play a Role in the Transmission of Toxoplasma gondii within a Zoo Environment? Vet. Parasitol. 2008, 152, 202–209. [Google Scholar] [CrossRef]
  233. Taggart, P.L.; Fancourt, B.A.; Boardman, W.S.J.; Peacock, D.E.; Caraguel, C.G.B. Infection Pressure is necessary, but not sufficient by Itself, to Explain Toxoplasma gondii Seroprevalence in Intermediate Host Species. J. Parasitol. 2021, 107, 554–561. [Google Scholar] [CrossRef]
  234. Taggart, P.L.; Fancourt, B.A.; Fabijan, J.; Peacock, D.E.; Speight, K.N.; Caraguel, C.G.B.; McAllister, M.M. No Evidence of Toxoplasma gondii Exposure in South Australian Koalas (Phascolarctos cinereus). J. Parasitol. 2019, 105, 638–641. [Google Scholar] [CrossRef] [PubMed]
  235. Zhu, B.Y.; Hartigan, A.; Reppas, G.; Higgins, D.P.; Canfield, P.J.; Slapeta, J. Looks Can Deceive: Molecular Identity of an Intraerythrocytic Apicomplexan Parasite in Australian Gliders. Vet. Parasitol. 2009, 159, 105–111. [Google Scholar] [CrossRef] [PubMed]
  236. Hillman, A.E.; Ash, A.L.; Kristancic, A.R.; Elliot, A.D.; Lymbery, A.J.; Robertson, I.D.; Thompson, R.C.A. Validation of Various Parasite Detection Tests for Use in the Australian Marsupials Quenda (Isoodon obesulus) and Brushtail Possums (Trichosurus vulpecula). J. Vet. Diagn. Investig. 2017, 29, 64–75. [Google Scholar] [CrossRef] [PubMed]
  237. Adriaanse, K.; Lynch, M.; Sutherland, D.; Traub, R.; Lowe, J.; Hufschmid, J. Toxoplasma gondii Does Not Inhibit the Assisted Colonization of Eastern Barred Bandicoots (Perameles gunnii) to Phillip Island, Victoria, Australia. J. Wildl. Dis. 2024, 60, 116–125. [Google Scholar] [CrossRef]
  238. Lynch, M.; Liyanage, K.L.D.T.D.; Stent, A.; Sutherland, D.R.; Coetsee, A.; Adriaanse, K.; Jabbar, A.; Hufschmid, J. Assessing the Impact of Toxoplasma gondii in Endangered Eastern Barred Bandicoots (Perameles gunnii) on Phillip and French Islands. J. Wildl. Dis. 2025, 61, 654–662. [Google Scholar] [CrossRef]
  239. Liyanage, K.L.D.T.D.; Lynch, M.; Omotainse, O.S.; Su, C.; Hufschmid, J.; Jabbar, A. Molecular Detection and Characterisation of Toxoplasma gondii in Eastern Barred Bandicoots (Perameles gunnii) in Victoria, Australia. Int. J. Parasitol. Parasites Wildl. 2025, 27, 101071. [Google Scholar] [CrossRef]
  240. Randel, C.J.; Vanherweg, W.J. Survey for Select Pathogens in the Desert Kit Fox (Vulpes Macrotis Arsipus) in California, USA. J. Wildl. Dis. 2022, 58, 631–635. [Google Scholar] [CrossRef]
  241. Morozińska-Gogol, J. The Presence of Toxoplasma gondii in the Terrestrial and Marine Environments and Its Importance for Public Health. Ann. Parasitol. 2021, 67, 137–149. [Google Scholar] [CrossRef]
  242. Rahimi, M.T.; Daryani, A.; Sarvi, S.; Shokri, A.; Ahmadpour, E.; Teshnizi, S.H.; Mizani, A.; Sharif, M. Cats and Toxoplasma gondii: A Systematic Review and Meta-Analysis in Iran. Onderstepoort J. Vet. Res. 2015, 82, e1–e10. [Google Scholar] [CrossRef]
  243. Toxoplasma gondii Infections in Dogs: 2009-2020—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/33160144/ (accessed on 8 May 2024).
  244. Mumtaz, T.; Awan, U.A.; Mushtaq, A.; Afzal, M.S.; Mahmood, T.; Wasif, S.; Ali, A.; Ajmal, K.; Mohamed, T.; Muhammad, A.; et al. Prevalence of Toxoplasmosis in Sheep and Goats in Pakistan: A Systematic Review and Meta-Analysis. Pathogens 2022, 11, 1331. [Google Scholar] [CrossRef]
  245. A Review of Toxoplasmosis in Pigs—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/3518210/ (accessed on 8 May 2024).
  246. Agriculture|Free Full-Text|Seroprevalence and Potential Risk Factors of Toxoplasma gondii in Dromedary Camels. Available online: https://www.mdpi.com/2077-0472/13/1/129 (accessed on 8 May 2024).
  247. Toxoplasma gondii Infections in Chickens (Gallus Domesticus): Prevalence, Clinical Disease, Diagnosis and Public Health Significance—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/19744305/ (accessed on 8 May 2024).
  248. Zaki, L.; Olfatifar, M.; Ghaffarifar, F.; Eslahi, A.V.; KarimiPourSaryazdi, A.; Taghipour, A.; Hamidianfar, N.; Badri, M.; Jokelainen, P. Global Prevalence of Toxoplasma gondii in Birds: A Systematic Review and Meta-Analysis. Parasite Epidemiol. Control 2024, 25, e00350. [Google Scholar] [CrossRef]
  249. Marino, A.M.F.; Giunta, R.P.; Salvaggio, A.; Castello, A.; Alfonzetti, T.; Barbagallo, A.; Aparo, A.; Scalzo, F.; Reale, S.; Buffolano, W.; et al. Toxoplasma gondii in Edible Fishes Captured in the Mediterranean Basin. Zoonoses Public Health 2019, 66, 826–834. [Google Scholar] [CrossRef]
  250. de Barros, L.D.; Garcia, J.L.; Bresciani, K.D.S.; Cardim, S.T.; Storte, V.S.; Headley, S.A. A Review of Toxoplasmosis and Neosporosis in Water Buffalo (Bubalus Bubalis). Front. Vet. Sci. 2020, 7, 455. [Google Scholar] [CrossRef]
  251. Basso, W.; Sollberger, E.; Schares, G.; Küker, S.; Ardüser, F.; Moore-Jones, G.; Zanolari, P. Toxoplasma gondii and Neospora Caninum Infections in South American Camelids in Switzerland and Assessment of Serological Tests for Diagnosis. Parasit. Vectors 2020, 13, 256. [Google Scholar] [CrossRef]
  252. Liu, Q.; Wang, Z.-D.; Huang, S.-Y.; Zhu, X.-Q. Diagnosis of Toxoplasmosis and Typing of Toxoplasma gondii. Parasites Vectors 2015, 8, 292. [Google Scholar] [CrossRef]
  253. da Silva, Y.H.; Campos, D.R.; Lima, G.A.C.; Quintal, J.P.; Guimarães, B.G.; do Rêgo, G.M.M.; de Avelar, B.R.; Intrieri, J.d.M.; Correia, T.R.; Scott, F.B. Prevalence of Gastrointestinal Parasites in Domestic Cats (Felis catus) Diagnosed by Different Coproparasitological Techniques in the Municipality of Seropédica, Rio de Janeiro. Rev. Bras. Parasitol. Vet. 2023, 32, e006223. [Google Scholar] [CrossRef] [PubMed]
  254. Mikita, K.; Maeda, T.; Ono, T.; Miyahira, Y.; Asai, T.; Kawana, A. The Utility of Cerebrospinal Fluid for the Molecular Diagnosis of Toxoplasmic Encephalitis. Diagn. Microbiol. Infect. Dis. 2013, 75, 155–159. [Google Scholar] [CrossRef] [PubMed]
  255. Schaefer, J.J.; Kirchgessner, M.S.; Whipps, C.M.; Mohammed, H.O.; Bunting, E.M.; Wade, S.E. Prevalence of Antibodies to Toxoplasma gondii in White-Tailed Deer (Odocoileus virginianus) in New York State, USA. J. Wildl. Dis. 2013, 49, 940–945. [Google Scholar] [CrossRef] [PubMed]
  256. Dubey, J.P.; Carpenter, J.L. Histologically Confirmed Clinical Toxoplasmosis in Cats: 100 Cases (1952–1990). J. Am. Vet. Med. Assoc. 1993, 203, 1556–1566. [Google Scholar] [CrossRef]
  257. Dubey, J.P.; Darrington, C.; Tiao, N.; Ferreira, L.R.; Choudhary, S.; Molla, B.; Saville, W.J.A.; Tilahun, G.; Kwok, O.C.H.; Gebreyes, W.A. Isolation of Viable Toxoplasma gondii from Tissues and Feces of Cats from Addis Ababa, Ethiopia. J. Parasitol. 2013, 99, 56–58. [Google Scholar] [CrossRef]
  258. Ybañez, R.H.D.; Ybañez, A.P.; Nishikawa, Y. Review on the Current Trends of Toxoplasmosis Serodiagnosis in Humans. Front. Cell Infect. Microbiol. 2020, 10, 204. [Google Scholar] [CrossRef]
  259. Mcauley, J.B.; Jones, J.L.; Singh, K. Toxoplasma. In Manual of Clinical Microbiology; John Wiley & Sons: Hoboken, NJ, USA, 2015; pp. 2373–2386. [Google Scholar]
  260. Carvalho, N.O.R.; Gaspar, R.C.; Lima, S.G.; da Silva Zanini, D.; Rodrigues, N.J.L.; Guimarães, V.Y.; da Silva, A.V.; Rahal, S.C.; Henrique, A.L.P.; Fornazari, F.; et al. Comparison Between Indirect Fluorescent Antibody Test and Modified Agglutination Test for Detecting Anti-Toxoplasma gondii IgG Antibodies in Neotropical Primates. J. Med. Primatol. 2025, 54, e70002. [Google Scholar] [CrossRef] [PubMed]
  261. Dong, H.; Zhang, J.; Wang, Q.; Shen, Y.; Zhou, B.; Dai, L.; Zhu, W.; Sun, H.; Xie, X.; Xie, H.; et al. Development of an Indirect ELISA for Detecting Toxoplasma gondii IgG Antibodies Based on a Recombinant TgIMP1 Protein. PLoS Negl. Trop. Dis. 2024, 18, e0012421. [Google Scholar] [CrossRef] [PubMed]
  262. Iqbal, J.; Khalid, N. Detection of Acute Toxoplasma gondii Infection in Early Pregnancy by IgG Avidity and PCR Analysis. J. Med. Microbiol. 2007, 56, 1495–1499. [Google Scholar] [CrossRef] [PubMed]
  263. Wong, S.Y.; Hajdu, M.P.; Ramirez, R.; Thulliez, P.; McLeod, R.; Remington, J.S. Role of Specific Immunoglobulin E in Diagnosis of Acute Toxoplasma Infection and Toxoplasmosis. J. Clin. Microbiol. 1993, 31, 2952–2959. [Google Scholar] [CrossRef]
  264. Olariu, T.R.; Blackburn, B.G.; Press, C.; Talucod, J.; Remington, J.S.; Montoya, J.G. Role of Toxoplasma IgA as Part of a Reference Panel for the Diagnosis of Acute Toxoplasmosis during Pregnancy. J. Clin. Microbiol. 2019, 57, e01357-18. [Google Scholar] [CrossRef]
  265. Naot, Y.; Remington, J.S. An Enzyme-Linked Immunosorbent Assay for Detection of IgM Antibodies to Toxoplasma gondii: Use for Diagnosis of Acute Acquired Toxoplasmosis. J. Infect. Dis. 1980, 142, 757–766. [Google Scholar] [CrossRef]
  266. Dhakal, R.; Gajurel, K.; Pomares, C.; Talucod, J.; Press, C.J.; Montoya, J.G. Significance of a Positive Toxoplasma Immunoglobulin M Test Result in the United States. J. Clin. Microbiol. 2015, 53, 3601–3605. [Google Scholar] [CrossRef]
  267. Teimouri, A.; Mohtasebi, S.; Kazemirad, E.; Keshavarz, H. Role of Toxoplasma gondii IgG Avidity Testing in Discriminating between Acute and Chronic Toxoplasmosis in Pregnancy. J. Clin. Microbiol. 2020, 58, e00505-20. [Google Scholar] [CrossRef]
  268. Brenier-Pinchart, M.P.; Morand-Bui, V.; Fricker-Hidalgo, H.; Equy, V.; Marlu, R.; Pelloux, H. Adapting a Conventional PCR Assay for Toxoplasma gondii Detection to Real-Time Quantitative PCR Including a Competitive Internal Control. Parasite 2007, 14, 149–154. [Google Scholar] [CrossRef]
  269. Lin, M.-H.; Chen, T.-C.; Kuo, T.; Tseng, C.-C.; Tseng, C.-P. Real-Time PCR for Quantitative Detection of Toxoplasma gondii. J. Clin. Microbiol. 2000, 38, 4121–4125. [Google Scholar] [CrossRef]
  270. Kong, Q.-M.; Lu, S.-H.; Tong, Q.-B.; Lou, D.; Chen, R.; Zheng, B.; Kumagai, T.; Wen, L.-Y.; Ohta, N.; Zhou, X.-N. Loop-Mediated Isothermal Amplification (LAMP): Early Detection of Toxoplasma gondii Infection in Mice. Parasites Vectors 2012, 5, 2. [Google Scholar] [CrossRef]
  271. Ajzenberg, D.; Bañuls, A.-L.; Tibayrenc, M.; Dardé, M.L. Microsatellite Analysis of Toxoplasma gondii Shows Considerable Polymorphism Structured into Two Main Clonal Groups. Int. J. Parasitol. 2002, 32, 27–38. [Google Scholar] [CrossRef] [PubMed]
  272. Shwab, E.K.; Zhu, X.-Q.; Majumdar, D.; Pena, H.F.; Gennari, S.M.; Dubey, J.P.; Su, C. Geographical Patterns of Toxoplasma gondii Genetic Diversity Revealed by Multilocus PCR-RFLP Genotyping. Parasitology 2014, 141, 453–461. [Google Scholar] [CrossRef] [PubMed]
  273. Guo, Z.-G.; Johnson, A.M. DNA Polymorphisms Associated with Murine Virulence of Toxoplasma gondii Identified by RAPD-PCR. In Toxoplasma gondii; Springer: Berlin/Heidelberg, Germany, 1996; pp. 17–26. [Google Scholar]
  274. Costa, J.-M.; Cabaret, O.; Moukoury, S.; Bretagne, S. Genotyping of the Protozoan Pathogen Toxoplasma gondii Using High-Resolution Melting Analysis of the Repeated B1 Gene. J. Microbiol. Methods 2011, 86, 357–363. [Google Scholar] [CrossRef] [PubMed]
  275. Murata, Y.; Sugi, T.; Weiss, L.M.; Kato, K. Identification of Compounds That Suppress Toxoplasma gondii Tachyzoites and Bradyzoites. PLoS ONE 2017, 12, e0178203. [Google Scholar] [CrossRef]
  276. Moré, G.; Venturini, M.C.; Pardini, L.; Unzaga, J.M. Toxoplasma. In Parasitic Protozoa of Farm Animals and Pets; Springer: Berlin/Heidelberg, Germany, 2018; pp. 149–168. [Google Scholar]
  277. WOAH. WOAH Terrestrial Manual; WOAH: Paris, France, 2018. [Google Scholar]
  278. Arafa, F.M.; Osman, D.H.; Tolba, M.M.; Rezki, N.; Aouad, M.R.; Hagar, M.; Osman, M.; Said, H. Sulfadiazine Analogs: Anti-Toxoplasma in Vitro Study of Sulfonamide Triazoles. Parasitol. Res. 2023, 122, 2353–2365. [Google Scholar] [CrossRef]
  279. Toxoplasmosis in Animals—Generalized Conditions. Available online: https://www.msdvetmanual.com/generalized-conditions/toxoplasmosis/toxoplasmosis-in-animals (accessed on 7 May 2024).
  280. Halonen, S.K.; Weiss, L.M. Toxoplasmosis. Handb. Clin. Neurol. 2013, 114, 125–145. [Google Scholar] [CrossRef]
  281. Greene, C.E.; Cook, J.R.; Mahaffey, E.A. Clindamycin for Treatment of Toxoplasma Polymyositis in a Dog. J. Am. Vet. Med. Assoc. 1985, 187, 631–634. [Google Scholar] [CrossRef] [PubMed]
  282. Dunay, I.R.; Gajurel, K.; Dhakal, R.; Liesenfeld, O.; Montoya, J.G. Treatment of Toxoplasmosis: Historical Perspective, Animal Models, and Current Clinical Practice. Clin. Microbiol. Rev. 2018, 31, e00057-17. [Google Scholar] [CrossRef] [PubMed]
  283. Petersen, E.; Schmidt, D.R. Sulfadiazine and Pyrimethamine in the Postnatal Treatment of Congenital Toxoplasmosis: What Are the Options? Expert Rev. Anti-Infect. Ther. 2003, 1, 175–182. [Google Scholar] [CrossRef] [PubMed]
  284. De-La-Torre, A.; Stanford, M.; Curi, A.; Jaffe, G.J.; Gomez-Marin, J.E. Therapy for Ocular Toxoplasmosis. Ocul. Immunol. Inflamm. 2011, 19, 314–320. [Google Scholar] [CrossRef]
  285. Alday, P.H.; Doggett, J.S. Drugs in Development for Toxoplasmosis: Advances, Challenges, and Current Status. Drug Des. Dev. Ther. 2017, 11, 273–293. [Google Scholar] [CrossRef]
  286. Montoya, J.G.; Remington, J.S. Clinical Practice: Management of Toxoplasma gondii Infection during Pregnancy. Clin. Infect. Dis. 2008, 47, 554–566. [Google Scholar] [CrossRef]
  287. Wang, Z.-D.; Liu, H.-H.; Ma, Z.-X.; Ma, H.-Y.; Li, Z.-Y.; Yang, Z.-B.; Zhu, X.-Q.; Xu, B.; Wei, F.; Liu, Q. Toxoplasma gondii Infection in Immunocompromised Patients: A Systematic Review and Meta-Analysis. Front. Microbiol. 2017, 8, 389. [Google Scholar] [CrossRef]
  288. Kavanagh, O.N. Alkalising Agents in Urinary Tract Infections: Theoretical Contraindications, Interactions and Synergy. Ther. Adv. Drug Saf. 2022, 13, 20420986221080794. [Google Scholar] [CrossRef]
  289. Brunton, L.; Lazo, J.; Parker, K. Goodman & Gilman’s The Pharmacological Basis of Therapeutics, 11th ed.; McGraw Hill Professional: New York, NY, USA, 2005; ISBN 978-0-07-160891-6. [Google Scholar]
  290. Zuther, E.; Johnson, J.J.; Haselkorn, R.; McLeod, R.; Gornicki, P. Growth of Toxoplasma gondii Is Inhibited by Aryloxyphenoxypropionate Herbicides Targeting Acetyl-CoA Carboxylase. Proc. Natl. Acad. Sci. USA 1999, 96, 13387–13392. [Google Scholar] [CrossRef]
  291. Tilley, M.; Fichera, M.E.; Jerome, M.E.; Roos, D.S.; White, M.W. Toxoplasma gondii Sporozoites Form a Transient Parasitophorous Vacuole That Is Impermeable and Contains Only a Subset of Dense-Granule Proteins. Infect. Immun. 1997, 65, 4598–4605. [Google Scholar] [CrossRef]
  292. Malik, M.A.; Sajid, M.S.; Abbas, R.Z.; Aleem, M.T.; Anjum, F.R.; Khan, A.; Farhab, M.; Maqbool, M.; Zeeshan, M.; Hussain, K. Anthelmintic Drug Resistance in Livestock: Current Understanding and Future Trends. In Parasitic Helminths and Zoonoses-From Basic to Applied Research; IntechOpen: London, UK, 2022; ISBN 1-80355-568-8. [Google Scholar]
  293. Zhang, Y.; Li, D.; Lu, S.; Zheng, B. Toxoplasmosis Vaccines: What We Have and Where to Go? NPJ Vaccines 2022, 7, 131. [Google Scholar] [CrossRef]
  294. Innes, E.A.; Bartley, P.M.; Rocchi, M.; Benavidas-Silvan, J.; Burrells, A.; Hotchkiss, E.; Chianini, F.; Canton, G.; Katzer, F. Developing Vaccines to Control Protozoan Parasites in Ruminants: Dead or Alive? Vet. Parasitol. 2011, 180, 155–163. [Google Scholar] [CrossRef] [PubMed]
  295. Li, L.; Farhab, M.; Kyaw, P.O.; Xia, X.-X.; Cai, H.-Q.; Zhang, T.; Cao, M.-X.; Li, J.-G.; Yuan, Y.-G. Targeted Gene Knock-Out of Fel D1 in Fetal Fibroblasts Using CRISPR–Cas9: Implications for Cat Allergies. Animals 2025, 15, 927. [Google Scholar] [CrossRef] [PubMed]
  296. Yuan, Y.-G.; Liu, S.-Z.; Farhab, M.; Lv, M.-Y.; Zhang, T.; Cao, S.-X. Genome Editing: An Insight into Disease Resistance, Production Efficiency, and Biomedical Applications in Livestock. Funct. Integr. Genom. 2024, 24, 81. [Google Scholar] [CrossRef] [PubMed]
  297. Li, L.; Zhang, T.; Farhab, M.; Xia, X.-X.; Reza, A.M.M.T.; Kyaw, P.O.; Chen, F.; Aly Sayed Ismail, E.; Xue, G.; Zhong, P.; et al. Comprehensive Analysis of circRNAs and lncRNAs Involvement in the Development of Skeletal Muscle in Myostatin-Deficient Rabbits. Anim. Biotechnol. 2025, 36, 2465624. [Google Scholar] [CrossRef]
  298. Innes, E.A.; Hamilton, C.; Garcia, J.L.; Chryssafidis, A.; Smith, D. A One Health Approach to Vaccines against Toxoplasma gondii. Food Waterborne Parasitol. 2019, 15, e00053. [Google Scholar] [CrossRef]
  299. Khan, M.S.; Buzdar, S.A.; Hussain, R.; Alouffi, A.; Aleem, M.T.; Farhab, M.; Javid, M.A.; Akhtar, R.W.; Khan, I.; Almutairi, M.M. Cobalt Iron Oxide (CoFe2O4) Nanoparticles Induced Toxicity in Rabbits. Vet. Sci. 2023, 10, 514. [Google Scholar] [CrossRef]
  300. Chu, K.-B.; Quan, F.-S. Advances in Toxoplasma gondii Vaccines: Current Strategies and Challenges for Vaccine Development. Vaccines 2021, 9, 413. [Google Scholar] [CrossRef]
  301. Innes, E.A.; Bartley, P.M.; Maley, S.; Katzer, F.; Buxton, D. Veterinary Vaccines against Toxoplasma gondii. Memórias Do Inst. Oswaldo Cruz 2009, 104, 246–251. [Google Scholar] [CrossRef]
  302. Jongert, E.; Roberts, C.W.; Gargano, N.; Förster-Waldl, E.; Petersen, E. Vaccines against Toxoplasma gondii: Challenges and Opportunities. Memórias Do Inst. Oswaldo Cruz 2009, 104, 252–266. [Google Scholar] [CrossRef]
  303. Liu, Q.; Singla, L.D.; Zhou, H. Vaccines against Toxoplasma gondii: Status, Challenges and Future Directions. Hum. Vaccines Immunother. 2012, 8, 1305–1308. [Google Scholar] [CrossRef]
  304. Dubey, J.P. Toxoplasmosis–a Waterborne Zoonosis. Vet. Parasitol. 2004, 126, 57–72. [Google Scholar] [CrossRef]
  305. Stelzer, S.; Basso, W.; Benavides Silván, J.; Ortega-Mora, L.M.; Maksimov, P.; Gethmann, J.; Conraths, F.J.; Schares, G. Toxoplasma gondii Infection and Toxoplasmosis in Farm Animals: Risk Factors and Economic Impact. Food Waterborne Parasitol. 2019, 15, e00037. [Google Scholar] [CrossRef]
  306. Kuruca, L.; Belluco, S.; Vieira-Pinto, M.; Antic, D.; Blagojevic, B. Current Control Options and a Way towards Risk-Based Control of Toxoplasma gondii in the Meat Chain. Food Control 2023, 146, 109556. [Google Scholar] [CrossRef]
  307. Teweldemedhin, M.; Gebremichael, A.; Geberkirstos, G.; Hadush, H.; Gebrewahid, T.; Asgedom, S.W.; Gidey, B.; Asres, N.; Gebreyesus, H. Seroprevalence and Risk Factors of Toxoplasma gondii among Pregnant Women in Adwa District, Northern Ethiopia. BMC Infect. Dis. 2019, 19, 327. [Google Scholar] [CrossRef] [PubMed]
  308. Kurth, K.; Jiang, T.; Muller, L.; Su, C.; Gerhold, R.W. Toxoplasma gondii Contamination at an Animal Agriculture Facility: Environmental, Agricultural Animal, and Wildlife Contamination Indicator Evaluation. Int. J. Parasitol. Parasites Wildl. 2021, 16, 191–198. [Google Scholar] [CrossRef] [PubMed]
  309. Peris, M.P.; Planas, S.; Langa, J.; Laborda, A.; Castillo, J.A.; Gracia, M.J. Seroprevalence of Zoonotic Pathogens in Stray Cats in an Urban Area of Northeast Spain. Vet. Parasitol. Reg. Stud. Rep. 2024, 53, 101052. [Google Scholar] [CrossRef] [PubMed]
  310. Montoya, A.; García, M.; Gálvez, R.; Checa, R.; Marino, V.; Sarquis, J.; Barrera, J.P.; Rupérez, C.; Caballero, L.; Chicharro, C.; et al. Implications of Zoonotic and Vector-Borne Parasites to Free-Roaming Cats in Central Spain. Vet. Parasitol. 2018, 251, 125–130. [Google Scholar] [CrossRef]
  311. Kochanowsky, J.A.; Koshy, A.A. Toxoplasma gondii . Curr. Biol. 2018, 28, R770–R771. [Google Scholar] [CrossRef]
  312. Aguirre, A.A.; Longcore, T.; Barbieri, M.; Dabritz, H.; Hill, D.; Klein, P.N.; Lepczyk, C.; Lilly, E.L.; McLeod, R.; Milcarsky, J.; et al. The One Health Approach to Toxoplasmosis: Epidemiology, Control, and Prevention Strategies. Ecohealth 2019, 16, 378–390. [Google Scholar] [CrossRef]
  313. Huertas-López, A.; Álvarez-García, G.; Sánchez-Sánchez, R.; Cantos-Barreda, A.; Ibáñez-López, F.J.; Martínez-Subiela, S.; Cerón, J.J.; Martínez-Carrasco, C. A Systematic Review and Meta-Analysis of the Serological Diagnosis of Toxoplasma gondii Infection Highlight the Lack of a One Health Integrative Research. Res. Vet. Sci. 2023, 155, 137–149. [Google Scholar] [CrossRef]
  314. İnci, A.; Sohel, M.H.; Babür, C.; Uslu, S.; Karademir, G.K.; Yürük, M.; Düzlü, Ö.; Kızgın, A.D.; Yıldırım, A. An Overview of One Health Concept Focusing on Toxoplasmosis. Turk. Parazitol. Derg. 2023, 47, 256–274. [Google Scholar] [CrossRef]
  315. Gilot-Fromont, E.; Lélu, M.; Dardé, M.-L.; Richomme, C.; Aubert, D.; Afonso, E.; Mercier, A.; Gotteland, C.; Villena, I. The Life Cycle of Toxoplasma gondii in the Natural Environment. In Toxoplasmosis: Recent Advances; InTech: Houston TX, USA, 2012; pp. 3–36. ISBN 978-953-51-0746-0. [Google Scholar]
  316. Mercier, A.; Devillard, S.; Ngoubangoye, B.; Bonnabau, H.; Bañuls, A.-L.; Durand, P.; Salle, B.; Ajzenberg, D.; Dardé, M.-L. Additional Haplogroups of Toxoplasma gondii out of Africa: Population Structure and Mouse-Virulence of Strains from Gabon. PLoS Negl. Trop. Dis. 2010, 4, e876. [Google Scholar] [CrossRef]
  317. Dakroub, H.; Sgroi, G.; D’Alessio, N.; Russo, D.; Serra, F.; Veneziano, V.; Rea, S.; Pucciarelli, A.; Lucibelli, M.G.; De Carlo, E.; et al. Molecular Survey of Toxoplasma gondii in Wild Mammals of Southern Italy. Pathogens 2023, 12, 471. [Google Scholar] [CrossRef]
Figure 1. Life cycle of T. gondii. Created in BioRender. Farhab, M. (2025) https://BioRender.com/n4n4txj.
Figure 1. Life cycle of T. gondii. Created in BioRender. Farhab, M. (2025) https://BioRender.com/n4n4txj.
Vetsci 12 00772 g001
Figure 2. Epidemiology of T. gondii. Countries with more PopSet, including humans and animals, are depicted in darker red, while those with fewer reported cases are indicated with a faint red color. Countries shown in the map with the same color as the background indicate that no PopSet data was available from these countries in ToxoDB. Data collected from [8].
Figure 2. Epidemiology of T. gondii. Countries with more PopSet, including humans and animals, are depicted in darker red, while those with fewer reported cases are indicated with a faint red color. Countries shown in the map with the same color as the background indicate that no PopSet data was available from these countries in ToxoDB. Data collected from [8].
Vetsci 12 00772 g002
Figure 3. Global distribution of T. gondii groups defined by microsatellite typing using 15MS.
Figure 3. Global distribution of T. gondii groups defined by microsatellite typing using 15MS.
Vetsci 12 00772 g003
Figure 4. A flow chart of drugs used to treat human toxoplasmosis.
Figure 4. A flow chart of drugs used to treat human toxoplasmosis.
Vetsci 12 00772 g004
Table 1. Milestones in T. gondii research in animals.
Table 1. Milestones in T. gondii research in animals.
YearFindingRef.
History
1909Name “Toxoplasma gondii” proposed [20]
1937Successful isolation of viable T. gondii [10]
1992T. gondii was classified as Type I, II, III, and atypical[12]
2005Gene mapping achieved[12,21]
Parasite Morphology and Life Cycle
1928Cyst recognized [4]
1958Tachyzoites division by endodyogeny described[22]
1960Bradyzoites resistance to digestive enzymes recognized [19]
1970Oocyst described[23]
1970Asexual and sexual stages were reported in the intestine of cats[24]
1972Asexual enteroepithelial stages were designated as types A–E[24]
1973The term “bradyzoite”, also called cystozoites, was proposed for the cystic organisms[25]
1973Term tachyzoite proposed (tachy = fast, zoite = life) [25]
1988Term tissue cyst proposed [12]
2019T. gondii sexual reproduction is associated with delta-6-desaturase gene[26]
2023ROCY1 gene activates T. gondii bradyzoite formation[13]
Transmission
1959Congenital transmission was observed in mice for up to 10 generations [27]
1954Transmission by carnivorism explained[28]
1965T. gondii infectivity is associated with cat feces [29]
1970–
1972
Sexual life cycle to defined only in felines, including excretion of oocysts only by felids[12]
2008Congenital transmission found in a large wild animal species, the white-tailed deer [12]
Diagnosis
1958Sabin–Feldman dye test[30]
1968IgM antibody detection from cord blood or infant serum for detecting congenital toxoplasmosis proposed[12]
1965Direct agglutination test (DAT) in humans and other animals [12]
1969 Detection of IgM in cord blood through the indirect fluorescent antibody test and ELISA [12]
1989Detection of T. gondii DNA was achieved, using the B1 gene from tachyzoite, through PCR[12]
Treatment
1942Sulfonamides were reported to be effective against murine toxoplasmosis[31,32]
1953Combined therapy with sulfonamides and pyrimethamine was discovered to have satisfactory results in treating toxoplasmosis in humans[31]
1958Spiramycin found to have anti-T. gondii activity in mice[33]
1973Clindamycin was documented to have anti-T. gondii effects[34,35]
Prevention and Control
1972Susceptible populations should avoid contact with oocysts[12]
1990sKeeping cats out of pig farms can reduce T. gondii infection in pigs [12]
1995Vaccination of sheep to reduce neonatal mortality in lambs is made available commercially[36]
First Confirmed Cases of Toxoplasmosis in Animals
1908First report of T. gondii (bradyzoites) in tissue (cyst) of Ctenodactylus gundi, in Tunisia[1]
1914Eurasian red squirrels (Sciurus vulgaris) [37]
1916Howler monkey (Alouatta seinculus) [38]
1951Squirrel monkeys (Saimiri sciureus) in the Philadelphia Zoo [38]
1974Pallas’s cats (Otocolobus manul) [39]
1985Wild turkeys from Blairsville, Georgia, USA [40]
1990Koalas (Phascolarctos cinereus); an arboreal marsupial, Sydney, Australia [41]
1990Dolphins (Stenella longirostris) [42]
1995Common mole (Talpa europaea) from Germany [43]
1997Golden lion tamarins (Leontopithecus rosalia) [44]
1997Barred owl (Strix varia) from Quebec, Canada [45]
2000Sea otters (Enhydra lutris nereis) [46]
2000Hawaiian crow (Corvus hawaiiensis) [47]
2004Striped skunk (Mephitis mephitis); T. gondii genotype III from Mississippi, USA [48]
2004Beaver (Castor canadensis) [49]
2004Captive nilgais (Boselaphus tragocamelus)[50]
2004Saiga antelope (Saiga tatarica) [50]
2004Bald eagle (Haliaeetus leucocephalus) from New Hampshire [51]
2004Canada goose (Branta canadensis); T. gondii genotype III from Mississippi, USA[48]
2004Black-winged lory (Eos cyanogenia); T. gondii genotype III from South Carolina, USA[48]
2005Hawaiian monk seal[52]
2005Fisher (Martes pennanti) from Garrett County, Maryland, USA [53]
2007American Red squirrels (Tamiasciurus hudsonicus) from New York, USA[54]
2007Woodchuck (Marmota monax) from New York, USA[54]
2008Sand cats (F. margarita) from Sharjah, UAE[55]
2008Gordon’s wildcat (Felis silvestris gordoni) from Sharjah, UAE [56]
2008Nicobar pigeons from South Africa [57]
2011Red-handed howler monkey (Alouatta belzebul)[58]
2011Black-footed penguin (Spheniscus demersus) [59]
2012Flying-foxes (megachiropteran bats)[60]
2013Northern shoveller (Anas clypeata) from Tuscany, Italy [61]
2013Common teal (Anas crecca) from Tuscany, Italy [61]
2015Giant panda (Ailuropoda melanoleuca) from Zhengzhou, China [62]
2015Wombats (Vombatus ursinus) [63]
2016Swinhoe’s striped squirrel (Tamiops swinhoei) from Germany [64]
2016Opossums (Didelphis virginiana) from Yucatan, Mexico [65]
2019Eastern fox squirrels (Sciurus niger) [66]
Domestic Animals
Pet animals
1908Domestic rabbits (Oryctolagus cuniculus) from Brazil [1,20]
1932Ferrets (Mustela putorius furo) from New Zealand [67]
1942Cats from the Middletown, New York, USA[12]
1950Dogs [68]
1956Mink[69]
1985Red foxes (Vulpes vulpes)[70]
Production Animals
1952Swine [71]
1957Sheep [72]
1961Chickens [73]
1979Goats [74]
1987Bobcat (Lynx rufus) from Ronan, Montana, USA[75]
1990Camels [76]
2003Ducks [77]
2007Cheetah (Acinonyx jubatus) from Dubai, UAE[78]
2014Alpaca [79]
2014Turkeys[80]
2014Geese from Hainan, China[81]
2021Horses[17,18]
2024Buffalos [82]
2025Cattle[16]
Table 2. Notable outbreaks/epizootics in domestic and wild species.
Table 2. Notable outbreaks/epizootics in domestic and wild species.
YearAnimal SpecieAffected/TotalRegionRef
1952ChickensHigh mortality (flock eradicated)South-Eastern Norway[103]
1992Canaries7/30 with ophthalmic lesionsNew Zealand[101]
2004Lories5/5 diedColumbia, South Carolina, USA[48]
2008Nicobar pigeons3/10 diedSouth Africa[57]
2011Black-footed penguins3 chicks diedNetherlands[59]
2019Chickens and guinea fowl15/76 diedViamão, Rio Grande do Sul, Southern Brazil[102]
1973Goats3 does congenital toxoplasmosis (Flock 1)Tasmania, Australia[74]
19775 stillbirths (Flock 2)
1978An abortion having twin kids (Flock 3)
1954Sheep15–20% abortion rateWellington, New Zealand[14,15]
200538 abortions + 15 resorbedTexas, USA[83]
20062 of 26 infected lambsTexas, USA[83]
201030/239 abortions in a dairy flock Palencia, Spain[86]
70/210 abortions in a meat flockSegovia, Spain
201040/100 pregnant ewes abortedSerro, Minas Gerais State, Brazil[85]
2015–
2018
146/242 from 11 abortion episodesSpain[84]
213/342 from two slaugheterhouses
1999monkeysWooly monkeys3 diedBrazil[98]
2001 and
2006
Squirrel monkeys
(Saimiri sciureus)
50 monkeys died and none recovered spontaneouslyFrench Guiana[99]
20187 of 13 deadSeoul, South Korea[105]
20194 diedHokkaido, Japan[106]
2020Howler monkeys
(Alouatta sp.)
7 died Brazil[100]
1994Pigs50–60% morbidity, 10–42% mortality
200/800 pigs died
Mantova, Lombardia, Italy[88]
810/2080 pigs diedModena, Emilia-Romagna, Italy
~31/345 pigs died
34/80 pigs diedMantova, Lombardia, Italy
200142% affected, 8% mortalityChina[88]
200133% affected, 2% mortalityChina[5]
200419/260 sows diedJinchang, Gansu Province, China[87]
Marine
2001–
2015
Hawaiian Monk Seals (Neomonachus Schauinslandi)8/183 confirmed T. gondiiHawaii, USA[91]
2009Bottlenose dolphins2 died (all seropositive)Canada[90]
2013Hector’s dolphins7/28 died (25%)New Zealand[89]
2015–2019Northern sea otters (Enhydra lutris kenyoni)22/44 infectedWashington, USA[96]
Miscellaneous
1957Hares (Lepus timidus ainu)8/13 diedSapporo, Japan[104]
1964Chinchillas44/56 died + 4 abortionsNot specified[92]
1992Black-footed ferrets (Mustela nigripes) 8 acute deaths + 13 chronicKentucky, USA[93]
1999Mink10,408 kits diedWisconsin, USA[97]
2006–2010Tammar wallabiesDeaths; 6 confirmed + 11 suspectedBudapest, Hungary[94]
2017Red-necked wallabies9 diedVirginia, USA[95]
Table 3. Milestones in T. gondii research in humans.
Table 3. Milestones in T. gondii research in humans.
YearFindingRef.
1939First isolate of T. gondii from human.[11,12]
1939First report of congenital transmission demonstrated in human.[11]
1940T. gondii isolated in heart, spleen, and other tissues of humans.[12]
1941Report of acquired toxoplasmosis, whose isolate became the famous RH strain.[12]
1941T. gondii isolated from blood of humans.[12]
1953Combined therapy with sulfonamides and pyrimethamine was discovered to have satisfactory results in treating toxoplasmosis in humans.[31]
1958Spiramycin has been used prophylactically in pregnant women. [33]
1960Transmission by carnivorism reported.[11]
1965Feco-oral transmission reported.[11]
1972Susceptible populations should avoid contact with oocysts.[12]
1974(1) An infection acquired during early pregnancy is more damaging to the fetus.[12]
(2) Not all women who acquired the infection transmitted it to the fetus.
(3) Women seropositive before pregnancy did not transmit infection to the fetus.
(4) Treatment with spiramycin reduced congenital transmission, but not clinical disease in infants.
1976The RH strain has lost the capacity to produce oocysts in cats.[12]
1979The first human toxoplasmosis outbreak described, was through oocyst inhalation/ingestion.[12]
1983Reported fatal acute toxoplasmosis-induced encephalitis, almost all of whom were HIV infected individuals.[12]
1995Canadian waterborne outbreak of toxoplasmosis.[12]
2006T. gondii outbreak in Brazil.[12]
Table 10. Summary of molecular methods for detection of T. gondii infection.
Table 10. Summary of molecular methods for detection of T. gondii infection.
Molecular Methods PurposeDNA Target RegionsRef.
Conventional PCR Species confirmationB1 gene, 529-bp repetitive element, 18S rDNA gene, SAG1, SAG2, and GRA1 [268,269]
Real-time PCR B1 gene, 529-bp repetitive element, 18S rDNA gene, SAG1
LAMP 529-bp repetitive element, B1, SAG1, SAG2, GRA1, oocyst wall protein genes [270]
Microsatellite analysis GenotypingTUB2, W35, TgM-A, B18, B17; M33, IV.1, XI.1, M48, M102, N60, N82, AA, N61, and N83[271]
Multilocus sequence typing BTUB, SAG2, GRA6, and SAG3[272]
PCR-RFLP SAG1, SAG2, SAG3, BTUB, GRA6, c22-8, c29-2, L358, PK1 and Apico[272,273]
RAPD-PCR Genomic DNA
High-resolution melting (HRM) analysis B1 gene[274]
Table 11. Drugs used to treat animal toxoplasmosis.
Table 11. Drugs used to treat animal toxoplasmosis.
Sr.DrugsDose *SpecieRef.
1Sulfadiazine15–25 mg/kgall animals[278,279]
Pyrimethamine0.44 mg/kg[280]
2Trimethoprim-sulfamethoxazole15 mg/kgdogs and cats[279]
3Clindamycin10–12.5 mg/kgdogs[281]
25–50 mg/kgcats[127]
* PO: Administered orally, every 12 h for 4 weeks.
Table 12. Types of proposed vaccines used against toxoplasmosis.
Table 12. Types of proposed vaccines used against toxoplasmosis.
Sr.Vaccine TypeDescriptionAdvantagesLimitationRef.
1ESA vaccinesExcretory–secretory antigens (ESAs) are used as antigenic agents Reduces parasitemia of highly virulent strains.
Propranolol and alum-adjuvanted ESE vaccine improve the protective effect of ESA and extend the survival time of mice.
Does not protect against all strains[293]
2Live attenuated vaccines
e.g.,
modified T. gondii strain (S48) vaccine
ΔHAP2 parasites by CRISPR/Cas9
ΔCDPK2 and ΔADSL knock-out vaccine
T. gondii is attenuated through gamma radiation, chemical treatment and passages.Currently, live attenuated vaccine is the most effective anti-T. gondii vaccine.
If oral live attenuated vaccines are prepared, they can mimic natural T. gondii infection and induce host cellular and humoral immunity against T. gondii without causing disease.
Short shelf life, may revert to virulence[294]
3Subunit vaccines
e.g.,
rTgHSP70 subunit vaccine
rROP18 and rCDPK6 combined with PLG subunit vaccine
Immunogenic parts of T. Gondii.Multi-epitope subunit vaccines with different T-cell and B-cell epitopes are more promising for research.Poor immunogenicity, requires carrier[298]
4DNA vaccines
e.g.,
ROP, GRA, MIC, and SAG antigen vaccines
Made by antigenic proteins such as rhoptry proteins (ROPs), microsomal proteins (MICs), and surface antigens (SAGs).Inexpensive, easy to administer, and induces a strong immune response.Poor immunogenicity in large animals, may trigger antibodies against the DNA vector[137,298]
5Multiple epitope vaccines
SAPNs vaccine and SAPNs scaffolded peptide epitopes vaccines
Antigenic peptide epitopes from SAG1, SAG2C, GRA6, GRA5 are used as antigenic agents.Generates stronger Th1 responses and allows epitope optimization.Poor immunogenicity, short half-life, may cause immune tolerance[300]
6mRNA vaccines
e.g.,
TgNTPase-II-LNP vaccine
mRNA-stimulating antigenic protein.Safe, no risk of gene recombination.Short intracellular half-life, not very stable in vivo[301]
7Carbohydrate-based vaccinesT. gondii proteins tagged with surface carbohydrates to stabilize and transport them, eliciting carbohydrate-specific antibodies.TLR-2 and TLR-4 can recognize T. gondii GPI.Poor immunogenicity, autoimmunity[302]
8Exosome vaccines
e.g.,
DC2.4 exosome vaccine
Exosomes from T. gondii, and DC2.4 cell-derived exosomes as antigenic agents.Induce humoral and cell-mediated immunity.Poor biocompatibility[303]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Farhab, M.; Aziz, M.W.; Shaukat, A.; Cao, M.-X.; Hou, Z.; Huang, S.-Y.; Li, L.; Yuan, Y.-G. Review of Toxoplasmosis: What We Still Need to Do. Vet. Sci. 2025, 12, 772. https://doi.org/10.3390/vetsci12080772

AMA Style

Farhab M, Aziz MW, Shaukat A, Cao M-X, Hou Z, Huang S-Y, Li L, Yuan Y-G. Review of Toxoplasmosis: What We Still Need to Do. Veterinary Sciences. 2025; 12(8):772. https://doi.org/10.3390/vetsci12080772

Chicago/Turabian Style

Farhab, Muhammad, Muhammad Waqar Aziz, Aftab Shaukat, Ming-Xing Cao, Zhaofeng Hou, Si-Yang Huang, Ling Li, and Yu-Guo Yuan. 2025. "Review of Toxoplasmosis: What We Still Need to Do" Veterinary Sciences 12, no. 8: 772. https://doi.org/10.3390/vetsci12080772

APA Style

Farhab, M., Aziz, M. W., Shaukat, A., Cao, M.-X., Hou, Z., Huang, S.-Y., Li, L., & Yuan, Y.-G. (2025). Review of Toxoplasmosis: What We Still Need to Do. Veterinary Sciences, 12(8), 772. https://doi.org/10.3390/vetsci12080772

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop