Skip to main content
American Journal of Alzheimer's Disease and Other Dementias logoLink to American Journal of Alzheimer's Disease and Other Dementias
. 2014 Jan 10;29(3):205–214. doi: 10.1177/1533317513517049

Possible Link Between Toxoplasma Gondii and the Anosmia Associated With Neurodegenerative Diseases

Joseph Prandota 1,
PMCID: PMC10852608  PMID: 24413543

Abstract

Toxoplasma gondii is an intracellular protozoan infecting 30% to 50% of global human population. Recently, it was suggested that chronic latent neuroinflammation caused by the parasite may be responsible for the development of several neurodegenerative diseases manifesting with the loss of smell. Studies in animals inoculated with the parasite revealed cysts in various regions of the brain, including olfactory bulb. Development of behavioral changes was paralleled by the preferential persistence of cysts in defined anatomic structures of the brain, depending on the host, strain of the parasite, its virulence, and route of inoculation. Olfactory dysfunction reported in Alzheimer’s disease, multiple sclerosis, and schizophrenia was frequently associated with the significantly increased serum anti–T gondii immunoglobulin G antibody levels. Damage of the olfactory system may be also at least in part responsible for the development of depression because T gondii infection worsened mood in such patients, and the olfactory bulbectomized rat serves as a model of depression.

Keywords: cerebral toxoplasmosis, neurodegeneration, impaired smell, anosmia, olfaction, autoimmune diseases, depression


The olfactory route for various infectious and/or toxic agents may initiate or exacerbate classical neurodegenerative and autoimmune diseases, especially in persons with genetic predisposition. 19 Several authors showed that many neurologic and neurodegenerative abnormalities are first demonstrable in the olfactory system with loss of smell (anosmia) up to 10 years before the onset of cognitive or motor dysfunction. 7 Neuroinflammation is a common feature of these diseases mostly emerging in the elderly individuals 9 and marked by activated glial cells that secrete numerous pro- and anti-inflammatory cytokines and other neurobiomediators. 10 For example, an exacerbation of Alzheimer’s symptoms lasting for few months following a systemic infection was also capable of elevating serum interleukin (IL) 1β. 11 Recently, it was suggested that chronic T gondii infection may be the key infectious agent responsible for triggering and development of several neurodegenerative diseases associated with an increased generation of several pro- and anti-inflammatory cytokines, including IL-1β. 1214

Toxoplasmosis is one of the most frequent infections affecting both healthy and immunocompromised humans with approximately 6 billion people infected. 15,16 During its life cycle, the pathogen interacts with approximately 3000 host genes or proteins, and many of them represent an extensive Toxoplasma gondii host–pathogen interactome enrichment in several psychiatric and neurological diseases. 17 At present, in immunocompetent individuals T gondii infection is believed to be asymptomatic, 18,19 but an increasing body of literature strongly suggests that the parasite is slowly emerging as a global health threat, 16,1922 especially in neurodegenerative diseases. Seroprevalence of the parasite measured by specific serum anti–toxo immunoglobulin G (IgG) antibodies varies widely in different countries depending on diagnostic tests used, environmental and socioeconomic conditions, including eating habits, health-related practices, and host susceptibility (Table 1). All these factors considerably hinder attempts to establish clear-cut connections between the highly prevalent infection of T gondii and the development of neurological diseases that are heralded by anosmia. In 1994, the National Health Interview Survey data obtained from 42 000 US households showed a 1.4% prevalence of self-reported olfactory dysfunction exponentially increasing with age. 24 Pregnancy is one of potential risk factors for olfactory disorders, 25 and the relationship between development of these abnormalities and chronic latent T gondii infection may be supported by the fact that at that time hormonal storm markedly affecting cellular and humoral immunity of pregnant woman may also exacerbate latent toxoplasmosis and increase the risk of congenital infection in the fetus. At present, one cannot exclude that isolated/syndromic congenital anosmia 26 is due to perinatal infection with the parasite, especially that structural differences in the brains of individuals with congenital anosmia are extending well beyond olfactory bulb and tract, including the piriform and orbitofrontal cortices. 27,28 Thus, from the first days/weeks of life, these neuroinflammatory processes may play an important role in the progress of pathophysiological abnormalities developing in the brain that finally lead to the olfactory system dysfunction also in neurodegenerative diseases.

Table 1.

Seroprevalence of Toxoplasma Gondii–Specific IgG Antibodies in Pregnant Women in a Selected Number of Countries.a

Study Location Year Prevalence (%)
Mexico 2006 6.1
United Kingdom 2005 9.1
Norway 1998 10.9
Bangladesh 1997 11.18
India 1999 11.6
Thailand 1998 13.1
Sweden 1999 14
Finland 1992 20.3
Denmark 1993 27.4
Turkey 2005 30.1
The Nederlands 2004 40.5
Polandb 2012 40.6
Switzerland 1995 46.1
France 1996 54.3
West Indies 2006 57
Germany 1999 63.2
DRSTP 2007 75.2

Abbreviations: DRSTP, Democratic Republic of Sao Tome and Principe; IgG, immunoglobulin G.

a Adapted from Elsheikha 23 ; with own modification.

b In Poland, between 2004 and 2012, the mean seroprevalence of IgG antibodies increased with age among 8281 pregnant women analyzed (mean age 26.7 vs 28.7 years; P < .001) with a yearly seroconversion rate of 0.8%. 24

Toxoplasma gondii tachyzoites may invade different types of brain cells including neurons, astrocytes, microglial cells, and Purkinje cells in the cerebellum. Intracellular tachyzoites manipulate signaling pathways and several signs for transduction mechanisms involved in apoptosis, immune cell maturation, and antimicrobial effectors functions. 29 It was demonstrated that in neurons infected by T gondii not only parasitic cysts but also the host cell cytoplasm and some axons were stained positive for the parasite antigens, thus supporting the notion that it may interfere with neuronal function. 20,30 It must be noted that in mice (at day 60 postinoculation with the parasite type II ME49 strain), a calculation of total cyst number per brain volume of various regions of the brain revealed that although cyst number decreased in cortex, thalamus, hippocampus, and striatum, their number slightly increased in olfactory bulb, hypothalamus, cerebellum, and brain stem. 30,31 The development of behavioral changes was paralleled by the preferential persistence of cysts in defined anatomic structures of the brain, 3032 depending on the host, strain of the parasite, its virulence, and the route of inoculation. 3234 Localization of T gondii cysts in different brain regions and cell types in both embryonal and adult animal brain tissues are presented in Tables 2 to 7. Immunochemistry study revealed that all major parts of neurons including the soma, dendrites, and axons harbored cysts, whereas intraneuronal T gondii antigen was present in the cytoplasm of cyst harboring neurons, and the parasite antigen–positive axons could be followed over long distances. 31 Astrocyte interactions with neuronal cysts were frequently observed. 33 Exposure of lipopolysaccharide (LPS) to neurons in the central nervous system (CNS) induced strong neurodegeneration in vivo and in vitro in substantia nigra and midbrain dopaminergic neurons 4951 as well as in hippocampal and cortical neurons. 52,53 Similar neuronal cell death was also reported in the enteric nervous system (ENS). 54,55 It was found that the increased production of nitric oxide (NO) by inducible nitric oxide synthase was a major cause of cell death in LPS-treated cell cultures. 49,52 Toxoplasma gondii infection of different host brain cells was associated with an enhanced generation of various cytokines, including interferon (IFN) γ, tumor necrosis factor (TNF) α, IL-1β, NO, and reactive oxygen/nitrogen species 56,57 as well as with an increased production of many neurotic biomolecules (Table 8). These molecular disturbances could affect the sense of smell also in children with autism, 13,64 Asperger’s syndrome, 65 and migraine patients 66,67 and result in olfactory impairment along with age 6870 (Table 9). This reasoning may be supported by the progressive decline in the levels of serum heat shock protein (HSP) 60 and HSP70 with age, whereas HSP70 antibody levels tend to increase (Table 10). On the other hand, it is known that host-derived HSPs play an important role in the development of innate immune defense against T gondii infection. 72 It must be noted that different strains of T gondii induced several constellations of cytokine responses 73 important for the development of various clinical signs and symptoms in the infected host. Virulence of the parasite has been linked with strain-dependent distinct dendritic cell responses and reduced number of activated CD8+ T cells. 74 In animals, oral/peritoneal inoculation with T gondii genotypes I to III resulted in atrophy or hypoplasia of some segments of the gastrointestinal tract and death/hypertrophy of part of myenteric neurons. 7577 Similar morphometric abnormalities of the ENS may be responsible for the development of gastrointestinal tract dysfunction reported in patients with autism, inflammatory bowel and/or autoimmune diseases, and in many other gastrointestinal tract disturbances. 55 Glial cells in the ENS appear to be very similar in origin, gross morphology, and ultrastructure to astrocytes of the CNS and bear similar relationships with neuronal cell bodies and processes to peripheral Schwann cells. 78 All these abnormalities in the brain and other organs associated with chronic T gondii infection strongly suggest that similar neuroinflammatory processes are also taking place in the olfactory system, leading to its progressing damage.

Table 2.

Preferential Localization of Toxoplasma Gondii ME49 Strain Cysts in Different Regions of Murine Brain at 2 and 6 Months Postinoculation.a

Brain Region 2 Months After Inoculationb 6 Months After Inoculationc
Cerebral cortex 34 57
Hippocampus 10 25
Thalamus 5
Hypothalamus 6 3
Amygdala 25 9
Caudate putamen 12 6
Cerebellum 8

a Adapted from to Melzer et al 33 with own modification.

b Total number of cysts observed = 67.

c Total number of cysts observed = 32.

Table 7.

Toxoplasma Gondii Infection and Cyst Formation in Primary Cultures of Cells of the Central Nervous System.a

Parameters Astrocytes Microglial Cells Neurons Refs
Relative efficiency of infection 100a 50b 5-15b 3942
Cytokine release IL-1, IL-6, GM-CSF IL-10, IL-6, TNF-α TNF-α 4345
Cyst formation Yes Limited Yes 42,45
Size of cysts Large (∼50 μm) Small (∼10 μm) Small (∼ 10 μm) 43
Size of brain cells 100 μm (harbor several dozen tachyzoites) 5-10 μm 10-15 μmc (contain only few tachyzoites) 42
Effect of IFN-γ and TNF-α Encystation Parasite killing ? 39,44 46
Inducible NOS Low High Yesd 47,48

Abbreviations: GM-CSF, granulocyte-macrophage colony-stimulating factor; IL, interleukin; IFN, interferon; NOS, nitric oxide synthase; TNF, tumor necrosis factor.

a Adapted from Fagard et al 38 with own modification.

b Lüder et al 37 found that in rats only 30% of microglial cells were infected with Toxoplasma gondii, whereas 10% of neurons and astrocytes were invaded. Besides, parasites showed low replication rates, with only 1 or 2 degenerated parasites in 93% of the parasitophorous vacuole.

c Cerebellar granular neurons and pyramidal hippocampal neurons (when the size of a cell doubles, its volume increases 8-fold). It must be noted that T gondii size is 2 to 4 μm.

d Not documented for T gondii infection. ? Not established yet.

Table 8.

Possible Consequences on Neurons of Cytokines and Biomolecules Secreted Upon Toxoplasma Gondii Infection.a

Cell Type Secreted Biomolecules Neurotic
Astrocyte IL-6
GM-CSF
TNF-α ±
IL-1β
Arachidonic acid +
Macrophage IL-12
NO ±
Microglial cells RNI +
NO ±
H2O2 +
IFN-γ
Glutamate +
Neuron NO ±
TNF-α, ±
Glutamate +
Natural killer cell IFN-γ
T cell PAF +
IL-4
IFN-γ
IL-10

Abbreviations: RNI, reactive oxygen intermediates; PAF, platelet-activating factor; GM-CSF, granulocyte-macrophage colony-stimulating factor; IL, interleukin; IFN, interferon; NO, nitric oxide; TNF, tumor necrosis factor.

a Adapted from Fagard et al 38 with own modification. It must be noted that Toxoplasma gondii infection caused a significant increase in dopamine metabolism in neural cells, which may lead to psychobehavioral changes in humans infected with toxoplasmosis. 58 Dopamine concentrations were 14% higher in the brain of mice with chronic infections than in controls. 59 In addition, induction of indoleamine 2,3-dioxygnase expression and decreased levels of tryptophan and increased formation of kynurenine were found in the brain, lungs, and serum of mice infected with the parasite. 60 Moreover, dopamine stimulated tachyzoite proliferation in human fibroblast and primary neonatal rat astrocyte cell cultures, 61 thus further enhancing the harmful effects of the parasite on the brain function. In addition, chronic latent T gondii infection is associated with overproduction of various cytokines, and it was postulated that cytokines may induce changes in mood and behavior, leading to depressive illness in man. 62,63

Table 9.

Percentage of Toxoplasma Gondii–Positive Individuals Among 214 Nonpsychiatrically Affected Controls Depending on Age Analyzed During a Large Epidemiologic Study of 869 Psychiatric Patients.a

Age, years
Percentage T. gondii Positive 18-20 21-25 26-30 31-35 36-40 41-45 46-50 51-55 56-60 61-65 66-70 71-75
100 100
80 80 80
60 58 58 60
40 35 32 40 39 38
20 19
0

a Adapted from Hinze-Selch et al. 70 In the control individuals 45 years or younger recruited from the same geographical region as the psychiatric patients admitted to the hospital, the serofrequency of Toxoplasma gondii infection ranged between 20% and 40% without any systematic age effect, whereas in the individuals older than 45 years, the serofrequency systematically increased with age from about 40% to almost 100%. 70

Table 10.

Changes in Serum Heat Shock Protein (HSP) and anti-HSP Antibody Levels in Aging.a

Age group, years
<40 40-69 70-78 ≥90
HSP60, ng/mLb 759 (239-1356) 383 (145-777) 221 (49-547) 294 ns (117-361)
HSP70, ng/mL 400 (60-1520) 80 (40-315) 50 (0-270) 20c (0-245)
Positive samplesd 10/13; 77% 14/16; 88% 14/20; 70% 8/11; 73%
Anti-hHSP60, U/mL 482 (427-603) 439 (370-491) 389 (234-548) 577 ns (486-783)
Anti-hHSP70, U/mL 115 (102-144) 143 (130-172) 232 (134-269) 191 ns (146-267)
Anti-mHSP65, U/mL 119 (48-267) 201 (142-291) 138 (107-226) 268e (181-507)

a Adapted from Rea et al 71 with own modification.

b Data are presented as medians with interquartile ranges in parentheses.

c P = .02.

d All 60 serum samples contained detectable levels of HSP60 and anti-hHSP60, anti-HSP70, and anti-HSP65 antibodies. The numbers of samples with detectable levels of HSP70 is indicated.

e P = .03, nonsignificant versus <40 age group (independent samples t test on log-transformed data).

Table 3.

Distribution of Reactivation Foci in the Central Nervous System After Acute Toxoplasma Gondii Infection in Mice.a

Cerebrumb
Mice no. Left Right Gray Matter White Matterc Cerebellumd Meningese
M1f 8 3 11 1 1 0
M2 20 19 37 2 0 0
M3 33 0 26 7 0 0
M4 3 5 5 3 0 0
M5 14 6 11 5 0 4
M6 19 29 24 22 0 2
M7 4 2 3 3 0 0
M8 13 15 17 10 2 3
M9 18 14 22 8 0 2
M10 4 6 10 0 0 0
Mean (SD) 13.5 (9.3) 9.7 (9.0) 15.2 (9.6) 7.2 (7.3) 0.3 (0.7) 1.0 (1.4)

Abbreviation: SD, standard deviation.

a Adapted from Dellacasa-Lindberg et al 35 with own modification.

b Number of foci detected in the left cerebral hemisphere.

c Markedly fewer number of foci was detected in gray matter than that in white matter (P < .01).

d Significantly fewer number of foci was found in the cerebellum than that in cerebrum (P < .001).

e Significantly fewer number of foci were found in the meningeal areas than in cerebrum (P < .001). Dellacasa-Lindberg et al 35 suggested that there was a striking resemblance in the distribution of parasitic lesions during acute toxoplasmic encephalitis in human and murine infections.

f Ten BALB/c mice (M1-10) were infected intraperitoneally with 5 × 104 freshly egressed Toxoplasma gondii tachyzoites. Mice were subjected to dexamethasone treatment and killed upon detection of central nervous system infection.

Table 4.

The Number of Cysts Load in Brain Tissue and Congenital Transmission Rate From the Offspring.a

Group of mice Number of Cysts in Brain Congenital Toxoplasma gondii Transmission Rate (%)
Early-stage infection 224 ± 59b,c (n = 18) 94.74
Intermediate-stage infection 202 ± 44b (n = 19) 90.48
Late-stage infection 134 ± 31 (n = 22) 91.67

Abbreviations: ANOVA, analysis of variance; SD, standard deviation.

a Adapted from Wang et al 36 with own modification. Each group of mice was infected with 5 cysts of Toxoplasma gondii by oral inoculation on the 5th, 10th, and 15th day after gestation. Results are expressed as the mean number of cysts collected from each group ± SD, generated by using one-way ANOVA. The congenital T gondii transmission rate is shown at the age of 12 weeks after birth in the offspring from the infected group.

b P < .01 versus late-stage infection.

c P > .05 versus intermediate-stage infection group.

Table 5.

Infection Rates of Different Cell Types From Embryonal Rat Cortices (E15) After In Vitro Infection With Toxoplasma Gondii Tachyzoites.a

Cell Type mAb for Host Cell Identification Frequency of Cell Typeb Rate of T gondii Infectionb
Neurons Anti-Nf 200 kDa 88.0 ± 1.3% 9.5 ± 1.1%
Astrocytes Anti-GFAP 7.9 ± 2.1% 9.7 ± 3.3%
Microglia Anti-CD71 4.1 ± 0.9% 31.5 ± 5.9%

Abbreviations: mAB, monoclonal antibody; GFAP, glial filament acidic protein; SD, standard deviation.

a Adapted from Lüder et al 37 with own modification.

b Determined 48 hours postinfection by double immmunofluorescence (at least 100 parasitophorous vacuoles were examined for each determination). Data represent means ± SD from 3 independent experiments.

Table 6.

Replication and Morphology of Toxoplasma Gondii in Different Cell Types From Embryonal Rat Cortices (E15).a

Intracellular Replication (% PV)b Neurons Astrocytes Microglia
1-2 parasites/PV 66 ± 2.6 67.2 ± 5.0 93.1 ± 4.5
4-8 parasites/PV 30 ± 2 26.2 ± 1.2 8.9 ± 4.5
16-32 parasites/PV 4.3 ± 1.5 5.3 ± 2.9 0
>32 parasites/PV 0 1.3 ± 1.2 0
Morphology of T gondii Normal Normal Often degenerated

Abbreviations: PV, parasitophorous vacuole; SD, standard deviation.

a Adapted from Lüder et al 37 with own modification.

b Determined 48 hours postinfection by double immunofluorescence (at least 100 PV were examined for each determination). Data represent means ± SD from 3 independent experiments.

Xiao et al 79 showed that in male mice, infection with the parasite led mainly to modulation of genes associated with olfactory function, such as downregulation of the number of olfactory receptors and dopamine receptor D4. However, general olfactory tests and sensorimotor gating were normal in both male and female infection. 79,80 The discrepancy between the findings in rodents and impaired sense of smell reported in the patients with Alzheimer’s disease, as well as in the individuals with various autoimmune diseases having chronic T gondii infection, may be at least in part explained by the markedly greater morphometric parameters of rhinencephalon in animals (lobus olfactorius) than in humans (bulbus olfactorius), 81 which must be clearly associated with a considerably smaller extent of the olfactory tissue subjected to neuroinflammatory destruction. 82 It must be emphasized that low olfactory bulb volumes have been found in patients with schizophrenia (left and right bulb) and their first-degree relatives (right bulb) as compared with healthy individuals (Table 11). 83 In 1 study, the significant atrophy was also reported in 43.9% of 150 patients with systemic lupus erythematous (SLE), with progression of reduction in right and left hippocampal volumes related to disease duration (P < .001). 84 Moreover, patients with neuropsychiatric SLE had amygdala damage. 85 In patients with Parkinson’s disease, olfactory loss was considered as a marked early symptom that correlated with the progression of the disease, 86 and parkinsonian symptoms have been observed as an initial manifestation in a Japanese patient with acquired immunodeficiency syndrome and T gondii infection. 87 Olfactory dysfunction has also been reported in HIV-infected and AIDS individuals, 88,89 in patients with Alzheimer’s disease, 90 in patients with Down’s syndrome, 91 in patients with multiple sclerosis, 92 in patients with SLE, 93 in patients with schizophrenia 94 and their relatives, 95 and during several pregnancies, 96 that is, the clinical entities with significantly increased serum anti–T gondii IgG antibody levels compared with healthy controls. 23,97104 Furthermore, the above-mentioned disturbed brain regions were consistently more infected than other sites in animals with toxoplasmosis. 3133

Table 11.

Olfactory Bulb Volumes in Patients With Schizophrenia, First-Degree Relatives, and Healthy Controls.a

Volumes, mm3
Left Bulb Right Bulb
Group Mean SD Mean SD
Patients (n = 11) 70.82b 11.77 70.18c 14.11
Control individuals (n = 20) 81.62 16.91 85.97 13.75
Relatives (n = 19) 83.51 17.96 75.41d 13.56

Abbreviations: MANOVA, multivariate analysis of variance; SD, standard deviation.

a Adapted from Turetsky et al 83 with own modification.

b Significant difference between patients and relatives (MANOVA, P < .05, 2-tailed). c Significant difference between patients and controls (P < .05).

d Significant difference between relatives and controls (P < .05).

Depression is highly prevalent in various medical conditions, including infectious, autoimmune, and neurodegenerative diseases. It seems that damage of the olfactory system is at least in part responsible also for development of depression because it was found that T gondii infection worsened mood in pregnant women, 105 female veterans, 106 older persons, 107 and patients with multiple sclerosis. 108,109 Higher incidence of depression also preceded the onset of Parkinson’s disease, 110 and the olfactory bulbectomized rat is usually serving as a model of depression. 111 Moreover, depression was reported in a sample of patients with obsessive–compulsive disorder, 112 and an important role of the parasite was suggested in the pathogenesis of this clinical entity. 113 Also, patients with recurrent mood disorders with history of suicide attempt had higher T gondii antibody titers than nonsuicide attempters (P = .004). 114 It must be added that antidepressants act on the host immune system causing neuroendocrine alterations associated with an increased generation of several bioneurotic molecules (Table 12). 115 Thus, the relationship between chronic latent T gondii infection and brain damage resulting in the development of depression should be seriously taken into consideration. In such patients, treatment of the infestation together with estimation of clinical course of depression would be helpful in more beneficial modification of actual therapeutic regimens. This suggestion is in line with the finding that the antipsychotic haloperidol and the mood stabilizer valproic acid most effectively inhibited the parasite growth in vitro with synergistic activity. 29

Table 12.

Antidepressant Effects on the Host Immune System.a

Antidepressant Source and Type of Effector Cells Neuroendocrine Alterations
Fluvoxamine, reboxetine, imipramine Murine glia cells ↓ NO levels after IFN-γ stimulation
Amitryptyline, nortriptyline Rat glia cells ↓ IL-1 and TNF-α after LPS stimulation
Venlafaxine Rat encephalogenic T-cell clones, splenocytes, peritoneal macrophages ↓ IL-12, TNF-α, and IFN-γ
Imipramine, mianserin, clomipramine, sertraline, and citalopram Human peripheral white blood cells ↓ Proinflammatory cytokines; ↑ anti-inflammatory cytokines
Imipramine, venlafaxine, fluoxetine Healthy human whole blood treatment resistant ↓ IL-10
Sertraline, citalopram, fluoxetine, fluvoxamine, paroxetine Patients with depression ↓ TNF-α, CRP, and leukocyte count
Bupropion, mirtazapine, citalopram, paroxetine, venlafaxine Patients with depression ↓ IL-6, TGF-β
Sertraline Patients with depression ↓ IL-12, ↑ IL-4, TGF-β
Desipramine and fluoxetine Rats ↓ IDO activity

Abbreviations: CRP, C-reactive protein; IDO, indoleamine 2,3-dioxygenase; TGF-β, transforming growth factor-β; LPS, lipopolysaccharide; IL, interleukin; ↓, decrease; ↑, increase ;TNF, tumor necrosis factor ;.NO, nitric oxide; IFN, interferon.

a Adapted from Antonioli et al 115 with own modification.

Interestingly, T gondii infection can convert the rodents’ natural aversion to cat odors into attraction, 82,116,117 probably because of altered neuronal activity in limbic brain regions that is necessary for innate defensive behavior associated with the activation of adjacent sexual arousal pathways 79,80 and generation of various neurotransmitters. 115 It should be noted that a surface SAG1 antigen of T gondii combined with nontoxic mutants of cholera toxin and enterotoxin (powerful mucosal adjuvants) administered intranasally in mice provided a beneficial high-level protection after virulent challenge infection with the parasite cysts. 118 In addition, treatment with monoclonal antibody against IL-6 resulted in a remarkable decrease in inflammation and numbers of cysts in the brain of animals with toxoplasmic encephalitis. 119 This beneficial effect may be partly explained by the fact that IL-6 enhances intracellular replication of the parasite acting through interactions with IFN-γ and TNF-α molecular activities. 120 Unfortunately, despite development of many serological and molecular methods in recent years, diagnosis of toxoplasmosis still faces difficulties because most of the commercially available tests are not fully specific and sensitive, representing wide variations in accuracy due to the fact that the parasite exhibits several protein and LPS antigens depending on its virulence, strain type, infection stage (tachyzoites, bradyzoites, oocysts), innate and/or acquired host immunity, and so on. 121123

In summary, damage of the olfactory system caused by chronic latent T gondii infection may affect olfactory bulb volume and various olfactory functions, therefore being responsible for the smell impairment in patients with several neuropsychiatric and/or autoimmune diseases. In addition, it seems that damage of the olfactory system may also be at least in part responsible for development of depression, which is frequently observed in those individuals.

Footnotes

The author declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author received no financial support for the research, authorship, and/or publication of this article.

References

  • 1. Mullol J, Alobid I, Marino-Sanchez F, et al. Furthering the understanding of olfaction, prevalence of loss of smell and risk factors: a population-based survey (OLFACT study). BMJ Open. 2012;2(6):e001256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Majde JA. The potential role of common olfactory challenges in the initiation of neuroinflammation. In: Gemma C, ed. Neuroinflammation. Pathogenesis, Mechanisms and Management. New York, NY: Nova Science Publishers; 2012:237–244. [Google Scholar]
  • 3. Moscavitch SD, Szyper-Kravitz M, Shoenfeld Y. Autoimmune pathology accounts for common manifestations in a wide range of neuro-psychiatric disorders: The olfactory and immune system interrelationship. Clin Immunol. 2009;130(3):235–243. [DOI] [PubMed] [Google Scholar]
  • 4. Prandota J. Autoimmune hepatitis associated with the odor of fish food proteins. A causal relationship or just a mere association? A case report. Allergol Immunopathol (Madr). 2002;30(6):331–337. [DOI] [PubMed] [Google Scholar]
  • 5. Prandota J. Autoimmune hepatitis and odour of fish food proteins. Allergol Immunopathol (Madr). 2003;31(1):56. [DOI] [PubMed] [Google Scholar]
  • 6. Prandota J. Odour of aquarium fish food proteins as a probable environmental agent causing autoimmune hepatitis in a 15-years-old boy. Pediatr Pol. 2002;77(4):337–341. [Google Scholar]
  • 7. Doty RL. Studies of human olfaction from the University of Pennsylvania Smell and Taste Center. Chem Senses. 1997;22(5):565–586. [DOI] [PubMed] [Google Scholar]
  • 8. Majde JA. Neuroinflammation resulting from covert brain invasion by common viruses –A potential role in local and global neurodegeneration. Med Hypotheses. 2010;75(2):204–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Mattson MP. Infectious agents and age-related neurodegenerative disorders. Ageing Res Rev. 2004;3(1):105–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Perricone C, Shoenfeld N, Agmon-Levin N, et al. Smell and autoimmunity: a comprehensive review. Clinic Rev Allerg Immunol. 2013;45(1):87–96. doi:10.1007/s12016-012-8343-x. [DOI] [PubMed] [Google Scholar]
  • 11. Holmes C, El-Okl M, Williams AL, Cunningham C, Wilcockson D, Perry VH. Systemic infection, interleukin 1b, and cognitive decline in Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2003;74(6):788–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Prandota J. Neuropathological changes and clinical features of autism spectrum disorder participants are similar to that reported in congenital and chronic cerebral toxoplasmosis in humans and mice. Res Autism Spectr Disord. 2010;4(2):103–118. [Google Scholar]
  • 13. Prandota J. Autism spectrum disorders may be due to cerebral toxoplasmosis associated with chronic neuroinflammation causing persistent hypercytokinemia that resulted in an increased lipid peroxidation, oxidative stress, and depressed metabolism of endogenous and exogenous substances. Res Autism Spectr Disord. 2010;4(2):119–155. [Google Scholar]
  • 14. Prandota J. Metabolic, immune, epigenetic, endocrine and phenotypic abnormalities found in individuals with autism spectrum disorders, Down syndrome and Alzheimer disease may be caused by congenital and/or acquired chronic cerebral toxoplasmosis. Res Autism Spectr Disord. 2011;5(1):14–59. [Google Scholar]
  • 15. Klaren VN, Kijstra A. Toxoplasmosis, an overview with emphasis on ocular involvement. Ocul Immunol Inflamm. 2002;10(1):1–26. [DOI] [PubMed] [Google Scholar]
  • 16. Furtado JM, Smith JR, Belfort R, Jr, Gattey D, Winthrop KL. Toxoplasmosis: a global threat. J Glob Infect Dis. 2011;3(3):281–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Carter CJ. Toxoplasmosis and polygenic disease susceptibility genes: extensive Toxoplasma gondii host/pathogen interactome enrichment in nine psychiatric or neurological disorders. J Pathog. 2013;(2013). doi:10.1155/2013/965046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Halonen SK, Weiss LM. Toxoplasmosis. In: Aminoff MJ, Boller F, Swaab DF, eds. Neuroparasitology and Tropical Neurology, Handbook of Clinical Neurology Series. Amsterdam, Netherlands: Elsevier BV; 2013:125–145. doi:10.1016/B978-0-444-440-3.00008-X. [Google Scholar]
  • 19. McLeod R, Van Tubbergen C, Montoya JG, Petersen E. Human toxoplasma infection. In: Weiss LM, Kim K, eds. Toxoplasma Gondii. 2nd ed. Amsterdam, Netherlands: Academic Press; 2014:99–159. [Google Scholar]
  • 20. Torgerson R, Mastroiacovo P. The global burden of congenital toxoplasmosis: a systematic review. Bull World Health Organ. 2013;91(7):501–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Nissapatorn V. Toxoplasmosis: a silent threat in Southeast Asia. J Parasitol. 2007;2(1):1–12. [Google Scholar]
  • 22. Flegr J, Prandota J, Sovičková M, Israili ZH. Toxoplasmosis–A global threat. Correlation between latent toxoplasmosis and specific disease burden–a WHO data-based study of 29 European and 59 non-European countries. (2013). Manuscript sent for publication. [Google Scholar]
  • 23. Elsheikha HM. Congenital toxoplasmosis: priorities for further health promotion action. Public Health. 2008;122(4):335–353. [DOI] [PubMed] [Google Scholar]
  • 24. Nowakowska D, Wujcicka W, Sobala W, Spiewak E, Gaj Z, Wilczyński J. Age-associated prevalence of Toxoplasma gondii in 8281 pregnant women in Poland between 2004 and 2012. Epidemiol Infect. 2013;31:1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Hoffman HJ, Ishill EK, Macturk RH. Age-related changes in the prevalence of smell/taste problems among the United States adult population. Ann NY Acad Sci. 1998;855(1):716–722. [DOI] [PubMed] [Google Scholar]
  • 26. Karstensen HG, Tommerup N. Isolated and syndromic forms of congenital anosmia. Clin Genet. 2012;81(3):210–215. [DOI] [PubMed] [Google Scholar]
  • 27. Barresi M, Ciurleo R, Giacoppo S, et al. Evaluation of olfactory dysfunction in neurodegenerative diseases. J Neurol Sci. 2012;323(1-2):16–24. [DOI] [PubMed] [Google Scholar]
  • 28. Frasnelli J, Fark T, Lehmann J, Gerber J, Hummel T. Brain structure is changed in congenital ansomia. NeuroImage. 2013;83:1074–1080. doi:10.1016/j.neuroimage.2013.07.070. [DOI] [PubMed] [Google Scholar]
  • 29. Fond G, Capdevielle D, Macgregor A, et al. Toxoplasma gondii: a potential role in the genesis of psychiatric disorders [in French]. Encephale. 2013;39(1):38–43. [DOI] [PubMed] [Google Scholar]
  • 30. Conley FK, Jenkins KA. Immunohistological study of the anatomic relationship of Toxoplasma antigens to the inflammatory response in the brains of mice chronically infected with Toxoplasma gondii. Infect Immun. 1981;31(3):1184–1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Haroon F, Händel U, Angenstein F, et al. Toxoplasma gondii actively inhibits neuronal function in chronically infected mice. PLoS One. 2012;7(4): e35516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Berenreiterova M, Flegr J, Kubena AA, Nemec P. The distribution of Toxoplasma gondii cysts in the brain of a mouse with latent toxoplasmosis. Implications of the behavioral manipulation hypothesis. PLoS One. 2011;6(12):e28925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Melzer TC, Cranston HJ, Weiss LM, Halonen SK. Host cell preference of Toxoplasma gondii cysts in murine brain: a confocal study. J Neuroparasitol. 2010;1. doi:10.4303/jnp/N100505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Ferguson DJ, Hutchison WM. An ultrastructural study of the early development and tissue cyst formation of Toxoplasma gondii in the brains of mice. Parasitol Res. 1987;73(6):483–491. [DOI] [PubMed] [Google Scholar]
  • 35. Dellacasa-Lindberg I, Fuks JM, Arrighi RB, et al. Migratory activation of primary cortical microglia upon infection with Toxoplasma gondii. Infect Immun. 2011;79(8):.046–3052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Wang T, Liu M, Gao XJ, Zhao ZJ, Chen XG, Lun ZR. Toxoplasma gondii: the effects of infection at different stages of pregnancy on the offspring of mice. Exp Parasitol. 2011;127(1):107–112. [DOI] [PubMed] [Google Scholar]
  • 37. Lüder CG, Giraldo-Velasquez M, Sendtner M, Gross U. Toxoplasma gondii in primary rat CNS cells: differential contribution of neurons, astrocytes, and microglial cells for the intracerebral development and stage differentiation. Exp Parasitol. 1999;93(1):23–32. [DOI] [PubMed] [Google Scholar]
  • 38. Fagard R, Van Tan H, Creuzet C, Pelloux H. Differential development of Toxoplasma gondii in neural cells. Parasitol Today. 1999;15(12):504–507. [DOI] [PubMed] [Google Scholar]
  • 39. Chao CC, Anderson WR, Hu S, Gekker G, Martella A, Peterson PK. Activated microglia inhibit multiplication of Toxoplasma gondii via a nitric oxide mechanism. Clin Immunol Immunopathol. 1993;67(2):178–183. [DOI] [PubMed] [Google Scholar]
  • 40. Peterson PK, Gekker G, Hu S, Chao CC. Intracellular survival and multiplication of Toxoplasma gondii in astrocytes. J Infect Dis. 1. 993;168(6):1472–1478. [DOI] [PubMed] [Google Scholar]
  • 41. Creuzet C, Robert F, Roisin MP, et al. Neurons in primary culture are less efficiently infected by Toxoplasma gondii than glial cells. Parasitol Res. 1998;84(1):25–30. [DOI] [PubMed] [Google Scholar]
  • 42. Halonen SK, Lyman WD, Chiu FC. Growth and development of Toxoplasma gondii in human neurons and astrocytes. J Neuropathol Exp Neurosci. 1996;55(11):1150–1156. [DOI] [PubMed] [Google Scholar]
  • 43. Jones TC, Bienz KA, Erb P. In vitro cultivation of Toxoplasma gondii cysts in astrocytes in the presence of gamma interferon. Infect Immun. 1986;51(1):147–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Fischer HG, Nitzgen B, Reichmann G, Gross U, Hadding U. Host cells of Toxoplasma gondii encystation in infected primary culture from mouse brain. Parasitol Res. 1997;83:637–641. [DOI] [PubMed] [Google Scholar]
  • 45. Fischer HG, Nitzgen B, Reichmann G, Hadding U. Cytokine responses induced by Toxoplasma gondii in astrocytes and microglial cells. Eur J Immunol. 1997;27(6):1539–1548. [DOI] [PubMed] [Google Scholar]
  • 46. Däubener W, Remscheid C, Nockemann S, et al. Anti-parasitic effector mechanisms in human brain tumor cells: role of interferon-gamma and tumor necrosis-alpha. Eur J Immunol. 1996:26(2):487–492. [DOI] [PubMed] [Google Scholar]
  • 47. Minc-Golomb D, Tsarfaty I, Schwartz JP. Expression of inducible nitric oxide synthase by neurons following exposure to endotoxin and cytokine. Brit J Pharmacol. 1994;112(3):720–722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Minamitani M, Tanaka J, Suzuki Y. Pathomechanism of cerebral hypoplasia in experimental toxoplasmosis in murine fetuses. Early Human Dev. 1996;44(1):37–50. [DOI] [PubMed] [Google Scholar]
  • 49. Shibata H, Katsuki H, Nishiwaki M, Kume T, Kaneko S, Akaike A. Lipopolysaccharide-induced dopaminergic cell death in rat midbrain slice culture: role of inducible nitric oxide synthase and protection by indomethacin. J Neurochem. 2003;86(5):1201–1212. [DOI] [PubMed] [Google Scholar]
  • 50. Castano A, Herrera AJ, Cano J, Machado A. Lipopolysaccharide intranigral injection induces inflammatory reaction and damage in nigrostriatal dopaminergic system. J Neurochem. 1998;70(4):1584–1592. [DOI] [PubMed] [Google Scholar]
  • 51. Herrera AJ, Castano A, Venero JL, Cano J, Machado A. The single intranigral injection of LPS as a new model for studying the selective effects of inflammatory reactions on dopaminergic system. Neurobiol Dis. 2000;7(4):429–447. [DOI] [PubMed] [Google Scholar]
  • 52. Lee P, Son D, Lee J, Kim YS, Kim H, Kim SY. Excessive production of nitric oxide induces the neuronal cell death in lipopolysaccharide-treated rat hippocampal slice culture. Neurosci Lett. 2003;349(1):33–36. [DOI] [PubMed] [Google Scholar]
  • 53. Picot L, Chevalier S, Mezghani-Abdelmoula S, et al. Cytotoxic effects of the lipopolysaccharide from Pseudomonas fluorescence on neurons and glial cells. Microb Pathog. 2003;35(3):95–106. [DOI] [PubMed] [Google Scholar]
  • 54. Arciszewski M, Pierzynowski S, Ekbald E. Lipopolysaccharide induces cell death in cultured porcine myenteric neurons. Dig Dis Sci. 2005;50(9):1661–1668. [DOI] [PubMed] [Google Scholar]
  • 55. Prandota J. Gastrointestinal tract abnormalities in autism, inflammatory bowel disease and many other clinical entities may be due to T. gondii infection. Sci Rep. 2012;1:4. doi:10.4172/scientificreports.256. [Google Scholar]
  • 56. Butcher BA, Kim L, Johnson PF, Denkers EY. Toxoplasma gondii tachyzoites inhibit proinflammatory cytokine induction in infected macrophages by preventing nuclear translocation of the transcription factor NF-kappa B. J Immunol. 2001;167(4):193–2201. [DOI] [PubMed] [Google Scholar]
  • 57. Suzuki Y. Immunopathogenesis of cerebral toxoplasmosis. J Infect Dis. 2002;186(suppl 2): S234–S240. [DOI] [PubMed] [Google Scholar]
  • 58. Prandovszky E, Gaskell E, Martin H, Dubey JP, Webster JP, McConkey GA. The neurotropic parasite Toxoplasma gondii increases dopamine metabolism. PLoS One. 2011;6(9):e23866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Stibbs HH. Changes in brain concentrations of catecholamines and indoleamines in Toxoplasma gondii infected mice. Ann Trop Med Parasitol. 1985;79(2):153–157. [DOI] [PubMed] [Google Scholar]
  • 60. Silva NM, Rodrigues CV, Santoro MM, Reis LFL, Alvarez-Leite JI, Gazzinelli RT. Expression of indoleamine 2,3-dioxygenase, tryptophan degradation and kynurenine formation during in vivo infection with Toxoplasma gondii: induction of endogenous gamma interferon and requirement of interferon regulatory factor 1. Infect Immun. 2002;70(2):859–868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Strobl JS, Goodwin DG, Rzigalinski BA, Lindsay DS. Dopamine stimulates propagation of Toxoplasma gondii tachyzoites in human fibroblast and primary neonatal rat astrocyte cell cultures. J Parasitol. 2012;98(6):1296–1298. [DOI] [PubMed] [Google Scholar]
  • 62. Pollak Y, Yirmiya R. Cytokine-induced changes in mood and behavior: implications for depression due to a general medical condition, immunotherapy and antidepressive treatment. Int J Neuropsychopharmacol. 2002;5(4):389–399. [DOI] [PubMed] [Google Scholar]
  • 63. Dunn AJ, Swiergiel AH, de Beaurepaire R. Cytokines as mediators of depression: what can we learn from animal studies? Neurosci Biobehav Rev. 2005;29(4-5):891–909. [DOI] [PubMed] [Google Scholar]
  • 64. Dudova I, Vodicka J, Havlovicova M, Sedlacek Z, Urbanek T, Hrdlicka M. Odor detection threshold, but not odor identification, is impaired in children with autism. Eur Child Adolesc Psychiatry. 2011;20(7):333–340. [DOI] [PubMed] [Google Scholar]
  • 65. Suzuki Y, Critchley HD, Rowe A, Howlin P, Murphy DG. Impaired olfactory identification in Asperger’s syndrome. J Neuropsychiatry Clin Neurosci. 2003;15(1):105–107. [DOI] [PubMed] [Google Scholar]
  • 66. Sjöstrand C, Savic I, Laudon-Meyer E, Hillert L, Lodin K, Waldenlind E. Migraine and olfactory stimuli. Curr Pain Headache Rep. 2010;14(3):244–251. [DOI] [PubMed] [Google Scholar]
  • 67. Prandota J. Recurrent headache as the main symptom of acquired cerebral toxoplasmosis in nonhuman immunodeficiency virus-infected subjects with no lymphadenopathy. The parasite may be responsible for the neurogenic inflammation postulated as a cause of different types of headaches. Am J Ther. 2007;14(1):63–105. [DOI] [PubMed] [Google Scholar]
  • 68. Schubert CR, Cruickshanks KJ, Murphy C, et al. Olfactory impairment in adults: the beaver dam experience. Ann NY Acad Sci. 2009;1170:531–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Murphy C, Schubert CR, Cruickshanks KJ, Klein BE, Klein R, Nondahl DM. Prevalence of olfactory impairment in older adults. JAMA. 2002;288(18):2307–2312. [DOI] [PubMed] [Google Scholar]
  • 70. Hinze-Selch D, Däubener W, Erdag S, Wilms S. The diagnosis of a personality disorder increases the likelihood for seropositivity to Toxoplasma gondii in psychiatric patients. Folia Parasitol (Praha). 2010;57(2):129–135. [DOI] [PubMed] [Google Scholar]
  • 71. Rea IM, McNerlan S, Pockley AG. Serum heat shock protein and anti-heat shock protein antibody levels in aging. Exp Gerontol. 2001;36(2):341–352. [DOI] [PubMed] [Google Scholar]
  • 72. Hisaeda H, Himeno K. The role of host-derived heat shock protein in immunity against Toxoplasma gondii infection. Parasitol Today. 1997;13(12):465–468. [DOI] [PubMed] [Google Scholar]
  • 73. Araujo FG, Slifer T. Different strains of Toxoplasma gondii induce different cytokine responses in CBA/Ca mice. Infect Immun. 2003;71(7):4171–4174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Tait ED, Jordan KA, Dupont CD, et al. Virulence of Toxoplasma gondii is associated with distinct dendritic cell responses and reduced numbers of activated CD8+ T cells. J Immunol. 2010;185(3):1502–1512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Zaniolo LM, da Silva AV, Sant'Ana Dde M, Araújo EJ. Toxoplasma gondii infection causes morphological changes in caecal myenteric neurons. Exp Parasitol. 2012;130(2):103–109. [DOI] [PubMed] [Google Scholar]
  • 76. Silva LS, Sartori AL, Zaniolo LM, da Silva AV, Sant'Ana Dde M, Araújo EJ. Toxoplasma gondii: myenteric neurons of intraperitoneally inoculated rats show quantitative and morphometric alterations. Exp Parasitol. 2011;129(1):5–10. [DOI] [PubMed] [Google Scholar]
  • 77. Odorizzi L, Moreira NM, Gonçalves GF, da Silva AV, Sant'ana Dde M, Araújo EJ. Quantitative and morphometric changes of subpopulations of myenteric neurons in swines with toxoplasmosis. Auton Neurosci. 2010;155(1-2):68–72. [DOI] [PubMed] [Google Scholar]
  • 78. Cabarrocas J, Savidge TC, Liblau RS. Role of enteric glial cells in inflammatory bowel disease. Glia. 2003;41(1):81–93. [DOI] [PubMed] [Google Scholar]
  • 79. Xiao J, Kannan G, Jones-Brando L, et al. Sex-specific changes in gene expression and behavior induced by chronic Toxoplasma infection in mice. Neuroscience. 2012;206:39–48. [DOI] [PubMed] [Google Scholar]
  • 80. House PK, Vyas A, Sapolsky R. Predator cat odors activate sexual arousal pathways in brains of Toxoplasma gondii infected rats. PLoS One. 2011;6(8):e23277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Vos H, R Herrlinger R. (1964) Taschenbuch der Anatomie. PZWL, Warsaw 1974, part III, p. 50.
  • 82. Vyas A, Kim SK, Sapolsky R. The effects of toxoplasma infection on rodent behavior are dependent on dose of the stimulus. Neuroscience. 2007;148(2):342–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Turetsky BI, Moberg PJ, Arnold SE, Doty RL, Gur RE. Low olfactory bulb volume in first-degree relatives of patients with schizophrenia. Am J Psychiatry. 2003;160(4):703–708. [DOI] [PubMed] [Google Scholar]
  • 84. Appenzeller S, Carnewalle AD, Li LM, Constallat LT, Cendes F. Hippocampal atrophy in systemic lupus erythematosus. Ann Rheum. 2006;65(12):1585–1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Emmer BJ, van der Grond J, Steup-Beekman GM, Huizinga TWJ, van Buchem MA. Selective involvement of the amygdala in systemic lupus erythematosus. PLoS Med. 2006;3(12):e499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Haehner A, Boesveldt S, Berendse HW, et al. Prevalence of smell loss in Parkinson’s disease—a multicenter study. Parkinsonism Relat Disord. 2009;15(7):490–494. [DOI] [PubMed] [Google Scholar]
  • 87. Murakami T, Nakajima M, Nakamura T, et al. Parkinsonian symptoms as an initial manifestation in a Japanese patient with acquired immunodeficiency syndrome and Toxoplasma infection. Internal Med. 2000;39(12):1111–1114. [DOI] [PubMed] [Google Scholar]
  •  88. Graham CS, Graham BG, Bartlett JA, Heald AE, Schiffman SS. Taste and smell losses in HIV infected patients. Physiol Behav. 1995;58(2):287–293. [DOI] [PubMed] [Google Scholar]
  •  89. Zucco GM, Ingegneri G. Olfactory deficits in HIV-infected patients with and without AIDS dementia complex. Physiol Behav. 2004;80(5):669–674. [DOI] [PubMed] [Google Scholar]
  •  90. Mesholam RI, Moberg PJ, Mahr RN, Doty RL. Olfaction in neurodegenerative disease: a meta-analysis of olfactory functioning in Alzheimer’s and Parkinson’s diseases. Arch Neurol. 1998;55(1):84–90. [DOI] [PubMed] [Google Scholar]
  •  91. Murphy C, Jinich S. Olfactory dysfunction in Down’s syndrome. Neurobiol Aging. 1996;7(4):631–637. [DOI] [PubMed] [Google Scholar]
  •  92. Zivadinov R, Zorzon M, Monti Bragadin L, Pagliaro G, Cazzato G. Olfactory loss in multiple sclerosis. J Neurol Sci. 1999;168(2):127–130. [DOI] [PubMed] [Google Scholar]
  •  93. Stojanovich L, Zandman-Goddard G, Pavlovich S, Sikanich N. Psychiatric manifestations in systemic lupus erythematosus. Autoimmun Rev. 2007;6(6):421–426. [DOI] [PubMed] [Google Scholar]
  •  94. Moberg PJ, Doty RL, Turetsky BI, et al. Olfactory identification deficits in schizophrenia: correlation with duration of illness. Am J Psychiatry. 1997;154(7):1016–1018. [DOI] [PubMed] [Google Scholar]
  •  95. Kopala LC, Good KP, Morrison K, Bassett AS, Alda M, Honer WG. Impaired olfactory identification in relatives of patients with familial schizophrenia. Am J Psychiatry. 2001;158(8):1286–1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  •  96. Wohlgemuth C, Beinder E, Ochsenbein-Kölbie N, Hummel T. Changes in olfactory function with several pregnancies? Swiss Med Wkly. 2008;138(31-32):466–469. [DOI] [PubMed] [Google Scholar]
  •  97. Kusbeci OY, Miman O, Yaman N, Aktepe OC, Yazar S. Could Toxoplasma gondii have any role in Alzheimer’s disease. Alzheimer Dis Assoc Disord. 2011;25(1):1–3. [DOI] [PubMed] [Google Scholar]
  •  98. Miman O, Kusbeci OY, Aktepe OC, Cetinkaya Z. The probable relation between Toxoplasma gondii and Parkinson’s disease. Neurosci Lett. 2010;475(3):129–131. [DOI] [PubMed] [Google Scholar]
  •  99. Koseoglu E, Yazar S, Koc I. Is Toxoplasma gondii a causal agent in migraine? Am J Med Sci. 2009;338(2):120–122. [DOI] [PubMed] [Google Scholar]
  • 100. Emelia O, Amal RN, Ruzanna ZZ, et al. Seroprevalence of anti-Toxoplasma gondii IgG antibody in patients with schizophrenia. Trop Biomed. 2012;29(1):151–159. [PubMed] [Google Scholar]
  • 101. Torrey EF, Bartko JJ, Lun ZR, Yolken RH. Antibodies to Toxoplasma gondii in patients with schizophrenia: A meta-analysis. Schizophr Bull. 2007;33(3):729–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Oksenhendler E, Charreau I, Tournerie C, Mahamoudu A, Carbon C, Aboulker JP. Toxoplasma gondii infection in advanced HIV infection. AIDS. 1994;8(4):483–487. [DOI] [PubMed] [Google Scholar]
  • 103. Arnson Y, Amital H, Guiducci S, et al. The role of infections in the immunopathogenesis of systemic sclerosis—evidence from serological studies. Ann NY Acad Sci. 2009;1173:627–632. [DOI] [PubMed] [Google Scholar]
  • 104. Wilcox MH, Powell RJ, Pugh SF, Balfour AH. Toxoplasmosis and systemic lupus erythematosus. Ann Rheum Dis. 1990;49(4):254–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Gröer MW, Yolken RH, Xiao JC, et al. Prenatal depression and anxiety in Toxoplasma gondii-positive women. Am J Obst Gynecol. 2011;204(5):e1–e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Radford A, Williams SN, Kane B, Groer M. Relationship of Toxoplasma gondii antibody titers and dysphoric moods in female veterans [abstract]. Brain Behav Immun. 2012;26 (Suppl 1):S23. [Google Scholar]
  • 107. Penninx BW, Kritchevsky SB, Yaffe K, et al. Inflammatory markers and depressed mood in older persons: results from the Health, Aging and Body Composition study. Biol Psychiatry. 2003;54(5):566–572. [DOI] [PubMed] [Google Scholar]
  • 108. Chwastiak L, Ehde DM, Gibbons LE, Sullivan M, Bowen JD, Kraft GH. Depressive symptoms and severity of illness in multiple sclerosis: epidemiologic study of a large community sample. Am J Psychiatry. 2002;159(11):1862–1868. [DOI] [PubMed] [Google Scholar]
  • 109. Beiske AG, Svensson E, Sandanger I, et al. Depression and anxiety amongst multiple sclerosis patients. Eur J Neurol. 2008;15(3):239–245. [DOI] [PubMed] [Google Scholar]
  • 110. Leentjens AF, Van den Akker M, Metsemakers JF, Lousberg R, Verhey FR. Higher incidence of depression preceding the onset of Parkinson’s disease: a register study. Mov Disord. 2003;18(4):414–418. [DOI] [PubMed] [Google Scholar]
  • 111. Song C, Leonard BE. The olfactory bulbectomised rat as a model of depression. Neurosci Biobehav Rev. 2005;29(4-5):627–647. [DOI] [PubMed] [Google Scholar]
  • 112. Diaconu G, Turecki G. Obsessive-compulsive personality disorder and suicidal behavior: evidence for a positive association in a sample of depressed patients. J Clin Psychiatry. 2009;70(11):1551–1556. [DOI] [PubMed] [Google Scholar]
  • 113. Miman O, Mutlu EA, Ozcan O, Atambay M, Karlidag R, Unal S. Is there any role of Toxoplasma gondii in the etiology of obsessive-compulsive disorder? Psychiatry Res. 2010;177(1-2):263–265. [DOI] [PubMed] [Google Scholar]
  • 114. Arling TA, Yolken RH, Lapidus M, et al. Toxoplasma gondii antibody titers and history of suicide attempts in patients with recurrent mood disorders. J Nerv Ment Dis. 2009;197(12):905–908. [DOI] [PubMed] [Google Scholar]
  • 115. Antonioli M, Rybka J, Carvalho LA. Neuroimmune endocrine effects of antidepressants. Neuropsychiatr Dis Treat. 2012;8:65–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Webster JP, McConkey GA. Toxoplasma gondii-altered host behavior: clues as to mechanism of action. Folia Parasitol. 2010;57(2):95–104. [DOI] [PubMed] [Google Scholar]
  • 117. Kannan G, Moldovan K, Xiao JC, Yolken RH, Jones-Brando L, Pletnikov MV. Parasite strain-dependent effects of Toxoplasma gondii on mouse behaviour. Folia Parasitol (Praha). 2010;57(2):151–155. [DOI] [PubMed] [Google Scholar]
  • 118. Bonenfant C, Dimier-Poisson I, Velge-Roussel F, et al. Intranasal immunization with SAG1 and nontoxic mutant heat-labile enterotoxins protects mice against Toxoplasma gondii. Infect Immun. 2001;69(3):1605–1612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Suzuki Y, Yang Q, Conley FK, Abrams JS, Remington JS. Antibody against interleukin-6 reduces inflammation and numbers of cysts in brains of mice with toxoplasmic encephalitis. Infect Immun. 1994;62(7):2773–2778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Beaman MH, Hunter CA, Remington JS. Enhancement of intracellular replication of Toxoplasma gondii by IL-6. Interactions with IFN-gamma and TNF-alpha. J Immunol. 1994;153(10):4583–4587. [PubMed] [Google Scholar]
  • 121. Prandota J. Increased generation of antibodies and autoantibodies directed against brain proteins in patients with autism and their families may be caused by T. gondii infection. Maternal and fetal microchimerisms probably play an important role in these processes acting as a “Trojan horse” in dissemination of the parasite. In: Gemma C. (ed.), Neuroinflammation. Pathogenesis, Mechanisms, and Management. New York, NY: Nova Science Publishers; 2012:447–638. [Google Scholar]
  • 122. Holec-Gasior L. Toxoplasma gondii recombinant antigens as tools for serodiagnosis of human toxoplasmosis. Current status of studies. Clin Vac Immunol. 2013;20(9):1343–1351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Magi B, Magliorini L. Western blotting for the diagnosis of congenital toxoplasmosis. New Microbiol. 2011;34(1):93–95. [PubMed] [Google Scholar]

Articles from American Journal of Alzheimer's Disease and Other Dementias are provided here courtesy of SAGE Publications

RESOURCES