Skip to content
BY-NC-ND 3.0 license Open Access Published by De Gruyter November 22, 2017

Positive association between Toxoplasma gondii IgG serointensity and current dysphoria/hopelessness scores in the Old Order Amish: a preliminary study

  • Abhishek Wadhawan , Aline Dagdag , Allyson Duffy , Melanie L. Daue , Kathy A. Ryan , Lisa A. Brenner , John W. Stiller , Toni I. Pollin , Maureen W. Groer , Xuemei Huang , Christopher A. Lowry , Braxton D. Mitchell and Teodor T. Postolache EMAIL logo
From the journal Pteridines

Abstract

Toxoplasma gondii (T. gondii) IgG seropositivity and serointensity have been previously associated with suicidal self-directed violence (SSDV). Although associations with unipolar depression have also been investigated, the results have been inconsistent, possibly as a consequence of high heterogeneity. We have now studied this association in a more homogeneous population, [that is (i.e.) Old Order Amish (OOA)] with previously reported high T. gondii seroprevalence. In 306 OOA with a mean age of 46.1±16.7 years, including 191 (62.4%) women in the Amish Wellness Study, we obtained both T. gondii IgG titers (by enzyme-linked immunosorbent assay [ELISA]), and depression screening questionnaires (Patient Health Questionnaire [PHQ-9] [n=280] and PHQ-2 [n=26]). Associations between T. gondii IgG and dysphoria/hopelessness and anhedonia scores on depression screening questionnaires were analyzed using multivariable linear methods with adjustment for age and sex. Serointensity was associated with both current dysphoria/hopelessness (p=0.045) and current combined anhedonia and dysphoria/hopelessness (p=0.043), while associations with simple anhedonia and past/lifelong (rather than current) phenotypes were not significant. These results indicate the need for larger longitudinal studies to corroborate the association between dysphoria/hopelessness and T. gondii IgG-titers. Current hopelessness is a known risk factor for SSDV which responds particularly well to cognitive behavioral therapy, and may be a focused treatment target for T. gondii-positive individuals at high-risk for SSDV.

Introduction

Toxoplasma gondii (T. gondii) is a protozoan parasite that can infect most warm-blooded animals, including humans. More than 30% of the world’s population carries T. gondii [1], with infection rates as high as 77% to as low as about 0.8% [2]. Toxoplasma gondii seroprevalence differs significantly between countries, diverse topographical regions within the same country and individuals of different ethnicities that live in similar regions [2], [3], [4]. Approximately, 11% of the United States population aged 6 years or older are infected with T. gondii [5].

Homoeothermic mammals, such as cattle and humans, serve as intermediate hosts for T. gondii. Its definitive hosts are members of the Felidae family, which includes household cats [3]. Toxoplasma gondii has three infectious stages (tachyzoites, sporozoites and bradyzoites). The sporozoites have the capability of infecting the intermediate hosts, but not the definitive hosts while the tachyzoites and bradyzoites have the capability to infect both the intermediate and definitive hosts [3], [6]. Tissue cysts contain bradyzoites and are predominantly found in skeletal and cardiac muscles, and neuronal tissues, e.g. brain and eye [7]. Cysts may also be located within other organs, e.g. kidneys, lungs and liver [7]. In immunosuppressed hosts (e.g. individuals with human immunodeficiency virus [HIV]/acquired immunodeficiency syndrome [AIDS]), the bradyzoites may convert into tachyzoites that penetrate other local and distant cells [8].

Toxoplasma gondii can be transmitted from mother to fetus during acute infection of a seronegative mother [9] or through reactivation of T. gondii infection during pregnancy [10]. The most common mode of infection in humans is through ingestion of contaminated food or water (e.g. ingestion of tissue cysts in raw or undercooked meat from intermediate hosts such as pigs, goats, cattle or oocyst in contaminated water). Rare, but medically relevant, ways of transmission include transplantation of an organ or blood transfusion from T. gondii-infected individuals [3], [11]. Acute infection with T. gondii typically produces a flu-like response that is often self-limiting in the immunocompetent host [6], [12], but the outcome of it is not a cure, rather a chronic latent infection develops with a potential for reactivation [6].

Multiple reports suggest links between chronic T. gondii infection and mental illnesses, including schizophrenia [13], [14], [15], [16], [17], [18], [19], [20], [21] and bipolar disorder [14], [22], [23], [24], [25]. Chronic T. gondii infection has also been associated with depression in psychiatric patients [26], women Veterans [27] and pregnant women [28]. However, several other studies found no association between T. gondii infection and depression [23], [29], [30], [31], [32]. One potential source of this discrepancy may be the heterogeneity of samples studied. Some of the heterogeneous risk factors in the study participants could be differences in lifestyles, co-occurring substance use disorders, discrete symptoms of depression being differentially associated with T. gondii infection, varying genetic vulnerability of the subjects to depression linked with T. gondii infection, infection with distinct strains of T. gondii or involvement of different mechanisms of infection with T. gondii (e.g. oocyst vs. tissue cyst), with all factors potentially having variable associations with depression.

The current study aimed to minimize the effects of possible confounders by investigating possible associations between T. gondii infection and two dominant symptoms of depression (i.e. dysphoria/hopelessness and anhedonia) in a sample of Old Order Amish (OOA), a population that is relatively homogeneous with similar lifestyles, socioeconomic status and cultural practices, has a low prevalence of substance use and a relatively high prevalence of T. gondii infection [33], [34]. We hypothesized that serological markers of T. gondii infection are positively associated with symptoms of depression, specifically, dysphoria/hopelessness and anhedonia. To our knowledge, this is the first time that relationships between specific symptoms of depression and T. gondii seropositivity and serointensity have been investigated.

Materials and methods

The protocol was approved by the University of Maryland Baltimore Institutional Review Board. Individuals were participants who had been previously enrolled in the Amish Wellness Study conducted by the University of Maryland School of Medicine. The Wellness Study is a community-based program that includes an assessment of cardio-metabolic health (e.g. lipids, blood pressure, glycemia) as well as a broad panel of lifestyle factors collected from medical and family history questionnaires, and mood and sleep questionnaires. The Wellness Study recruitment process involves the participants aged 18 years and older being approached in their home by one of the registered nurses from the University of Maryland Amish Research Clinic and an Amish liaison. If the participant was interested, the nurse explained the study and obtained informed consent. The participants completed a panel of questionnaires at the home visit during which they were enrolled, and then received a clinic visit at either the Amish Research Clinic or in the Amish Wellness Mobile Clinic, where they underwent a clinical protocol that included a fasting blood draw. Toxoplasma gondii antibodies were subsequently measured from this sample.

Depression symptoms were obtained from the study participants using a mental health screening questionnaire. First, we used a questionnaire based on the Patient Health Questionnaire-9 (PHQ-9) [35], called the Amish Mood Questionnaire (n=280), which was later replaced by a questionnaire based on the Patient Health Questionnaire-2 (PHQ-2) [36], called the Mental Health Depression Questionnaire (n=26). Each questionnaire asked: “during the last month have you had little feelings of interest or pleasure?”, which represented anhedonia, and “during the last month have you often been bothered by feeling down, depressed, or hopeless?”, which represented dysphoria/hopelessness. If the participants answered yes to both of these questions then they were categorized as currently experiencing both anhedonia and dysphoria/hopelessness. The questionnaires also asked if the participant had experienced either of these feelings in the past. Studies have reported that PHQ-9 has a sensitivity ranging from 80% to 81.3% and specificity ranging from 85.3% to 92% [37], [38] and the PHQ-2 sensitivity ranges from 83% to 89.3% and its specificity ranges from 75.9% to 92% [36], [38].

Participants’ blood samples were tested for the presence of T. gondii IgG antibodies using an enzyme-linked immunosorbent assay (ELISA) (IBL International, Männedorf, Switzerland), which tests for the levels of immunoglobulin G (IgG) to whole tachyzoites. Standards for validation were employed for all assays. A preset cut-off value as stated by the kit manufacturer was used to define the status of the samples. Seropositivity was determined based on the IgG titer concentration (serointensity) of the antibodies. Participants with concentrations greater than or equal to 12 IU/mL were deemed seropositive. When the ELISA results indicated an equivocal concentration of T. gondii antibody (8–12 IU/mL), we repeated the ELISA. When the second ELISA remained in the equivocal range or showed a level less than 8 IU/mL, the result was considered negative. If the second ELISA showed a concentration in the positive range (>12 IU/mL), the result was considered positive. We did not measure IgM antibodies in these samples.

We included participants with both available T. gondii plasma titers and symptoms of depression (n=306, 62.4% women; ages 18–87 years with a mean age of 46.1±16.7 years). The demographic characteristics of the study population have been summarized in Table 1.

Table 1:

Demographic characteristics of the sample.

VariablesTotal sample (n=306)Seropositive for T. gondii IgG (n, %=141, 46%)Seronegative for T. gondii IgG (n, %=165, 54%)
Age, mean±SD, years46.1±16.750.7±16.242.2±16.1
Gender, males, n (%)115 (37.6%)60 (42.6%)55 (33.3%)
Current dysphoria/hopelessness, present, n (%)26 (9.2%)16 (12.7%)10 (6.3%)
Current anhedonia, present, n (%)20 (7%)12 (9.6%)8 (5%)
Current anhedonia and dysphoria/hopelessness, present, n (%)16 (6%)11 (9.3%)5 (3.4%)
Current anhedonia or dysphoria/hopelessness, present, n (%)30 (10.4%)17 (13.3%)13 (8.1%)
Ever anhedonia or dysphoria/hopelessness, present, n (%)62 (21.5%)33 (25.4%)29 (18.2%)
Past anhedonia or dysphoria/hopelessness, present, n (%)52 (17.5%)27 (19.8%)25 (15.5%)
Toxoplasma IgG antibody titers, mean±SD, IU/mL47.8±104.399.8±136.63.4±2.8
Log Toxoplasma IgG antibody titers, mean±SD2.4±1.94.1±10.9±0.9

Logistic regression models were applied using depressive symptoms as dependent binary variables, and seropositivity or log-transformed serointensity as independent variables, adjusting for the covariates age and sex. The criterion two-tailed α was set at 0.05. No Bonferroni corrections for multiple comparisons were applied, considering the limited statistical power, given the need for adjustment for age (strong confounder based on cumulative effects) and sex (different exposure mechanisms to T. gondii and different vulnerability to depression). Statistics were completed using SAS/STAT 14.2 (Cary, NC, USA).

Results

All results presented are with adjustment for age and sex.

Serointensity (titers)

We found significant associations between serointensity and current dysphoria/hopelessness: p=0.045, odds ratio (OR)=1.26, 95% confidence interval (CI): [1.01, 1.58], and current combined anhedonia and dysphoria/hopelessness: p=0.043, OR=1.34, 95% CI: [1.01, 1.79].

There were no significant associations between serointensity and current anhedonia: p=0.170, OR=1.19, 95% CI: [0.93, 1.54], current anhedonia or dysphoria/hopelessness: p=0.147, OR=1.17, 95% CI: [0.95, 1.45], past anhedonia or dysphoria/hopelessness: p=0.872, OR=1.01, 95% CI: [0.86, 1.20], and ever-experienced anhedonia or dysphoria/hopelessness: p=0.428, OR=1.07, 95% CI: [0.91, 1.25].

Seropositivity

A low-grade significance/statistical trend was found for the association between seropositivity and current dysphoria/hopelessness: p=0.058, OR=2.31, 95% CI: [0.97, 5.50]. We also found a statistical trend for the association between seropositivity and current combined anhedonia and dysphoria/hopelessness: p=0.056, OR=2.99, 95% CI: [0.97, 9.15].

There were no significant associations between seropositivity and current anhedonia: p=0.181, OR=1.91, 95% CI: [0.74, 4.93], current anhedonia or dysphoria/hopelessness: p=0.164, OR=1.75, 95% CI: [0.80, 3.86], ever-experienced anhedonia or dysphoria/hopelessness: p=0.136, OR=1.56, 95% CI: [0.87, 2.79] and past anhedonia or dysphoria/hopelessness: p=0.303, OR=1.39, 95% CI: [0.74, 2.59]. These results are summarized in Table 2.

Table 2:

Associations between T. gondii IgG (seropositivity and serointensity) and cardinal symptoms of depression (anhedonia and dysphoria/hopelessness).

VariablesOdds ratio estimates adjusted for age and sex95% CI adjusted for age and sexp-Values adjusted for age and sex
Current dysphoria/hopelessness ~ seropositivity2.31[0.97, 5.50]0.058a
Current dysphoria/hopelessness ~ log_serointensity1.26[1.01, 1.58]0.045b
Current anhedonia ~ seropositivity1.91[0.74, 4.93]0.181
Current anhedonia ~ log_serointensity1.19[0.93, 1.54]0.170
Current anhedonia and dysphoria/hopelessness ~ seropositivity2.99[0.97, 9.15]0.056a
Current anhedonia and dysphoria/hopelessness ~ log_serointensity1.34[1.01, 1.79]0.043b
Current anhedonia or dysphoria/hopelessness ~ seropositivity1.75[0.80, 3.86]0.164
Current anhedonia or dysphoria/hopelessness ~ log_serointensity1.17[0.95, 1.45]0.147
Past anhedonia or dysphoria/hopelessness ~ seropositivity1.39[0.74, 2.59]0.303
Past anhedonia or dysphoria/hopelessness ~ log_serointensity1.01[0.86, 1.20]0.872
Ever anhedonia or dysphoria/hopelessness ~ seropositivity1.56[0.87, 2.79]0.136
Ever anhedonia or dysphoria/hopelessness ~ log_serointensity1.07[0.91, 1.25]0.428
  1. p-Values, odds ratio estimates and confidence intervals, generated by multi-variable logistic regression models with the presence of symptoms of depression as dependent variables and T. gondii seropositivity and serointensity as independent variables, accounting for sex and age. aStatistical trend (p<0.10). bSignificant (p<0.05). ~, related to.

Discussion

We found that serointensity for T. gondii was significantly associated with current dysphoria/hopelessness, and with current combined anhedonia and dysphoria/hopelessness, while associations with anhedonia scores alone, and lifelong symptoms, were not significant. There was also a marginal statistical trend for associations between T. gondii seropositivity and a combination of current dysphoria/hopelessness and anhedonia.

The results of our study point toward an association between certain cardinal depressive symptoms and anti-T. gondii IgG antibodies, which is consistent with the results of some previous studies in psychiatric patients [26], women Veterans [27] and women evaluated for prenatal depression [28].

Psychotropic medications have various degrees of anti-T. gondii activity in vitro [39]. This may be potentially clinically relevant considering a recent study that found that T. gondii seropositive patients with bipolar disorder, who were treated with medications with high in vitro anti-T. gondii activity, had fewer episodes of depression than those who were treated with medications with lower in-vitro anti-T. gondii activity [40]. Another clinically relevant report describes a T. gondii-seropositive patient with refractory symptoms of depression, who improved with antidepressant treatment only after being treated for toxoplasmosis [41].

However, our results are inconsistent with other studies that found no associations between T. gondii infection and symptoms of depression [14], [23], [29], [30], [31], [32], [42], [43], [44], [45], [46]. Many of these studies had limitations in terms of not adjusting for confounders such as occupation, heritability, living environment, cultural practices, socioeconomic status and substance use disorders. The current study has likely minimized these confounders by evaluating the OOA, which is a relatively homogeneous population.

Toxoplasma gondii has also been previously associated with suicidal behavior, an important cause of death in patients with depression. Specifically, several coauthors of the current study first reported an association between T. gondii IgG antibody titers and a history of suicide attempts in individuals with recurrent mood disorders [47]. In addition, the seropositivity rate for anti-T. gondii IgG antibodies was significantly higher in psychiatric inpatients with a history of attempted suicide [48]. We have further confirmed this association between suicide attempts and T. gondii infection in patients with schizophrenia in Germany, in recent suicide attempters admitted to a psychiatry ward in Sweden, and in a large cohort of Danish mothers [49], [50], [51]. Recently, a study in patients with mental illness identified T. gondii IgM titers, rather than T. gondii IgG titers, being associated with suicide attempts [52]. However, the same group presented data at a recent professional meeting confirming T. gondii IgG titers as being predictively associated with death by suicide (unpublished observation).

Moreover, identification of specific clinical, endophenotypic or biomarker-based subgroups, may lead to individualized suicide prevention efforts. For instance, T. gondii-seropositive patients with increased hopelessness, a more robust predictor of suicide than syndromal depression [53], may differ in prognosis and therapeutic response relative to other patients.

Other unmeasured clinical traits may be important as well in the interface between T. gondii and suicidal behavior. For instance, impulsivity and aggression have been recognized as endophenotypes for suicidal behavior, and their presence has been associated with an increased risk of suicide [54]. Research suggests that T. gondii infection can lead to impaired regulation of impulse control, which includes risk-taking behavior and violence [55]. Relationships between chronic infection with T. gondii and greater impulsivity in younger psychiatrically healthy males, as well as greater trait reactive aggression among females, have been reported [56]. Coccaro et al. [57] reported an association between greater impulsivity and aggression scores with T. gondii seropositivity in patients with intermittent explosive disorder.

We are not aware of other studies that have been conducted using principal component analysis on various symptoms of depression in the context of associations with T. gondii infection. For practical reasons, we only chose to examine two symptoms of depression (dysphoria/hopelessness and anhedonia) in our study. The heavy schedule of staff and participant involvement in the Amish Wellness Study did not allow for more extensive interviewing, and the PHQ-9-based evaluation had to be changed to the shorter PHQ-2-based instrument (retaining only the anhedonia and dysphoria/hopelessness questions). Nevertheless, previous reports showed that asking even a solitary question related to dysphoria/hopelessness has a sensitivity for major depression of about 85–90%, which increases to 95% upon addition of another question related to anhedonia [36]. Most importantly, both dysphoria/hopelessness and anhedonia have been recognized as risk factors for suicide in depressed patients [58]. In addition, symptoms of dysphoria/hopelessness as well as anhedonia have been well modeled in animal studies [54], allowing application of research domain criteria principles to suicide research.

The top candidate mechanism underlying the association between depression and T. gondii includes perturbation of the serotonergic system in the brain of depressed individuals [59]. The biosynthesis of serotonin begins with the metabolism of tryptophan, which is the precursor amino acid for this neurotransmitter [60]. Tryptophan is metabolized through two pathways, which involve production of either serotonin or kynurenine. The rate-limiting enzymes involved in the metabolism of tryptophan toward kynurenine metabolites are indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) [61]. Toxoplasma gondii infection in hosts with intact immunity results in the release of interferon-γ (IFN-γ), which further leads to upregulation of IDO. Conversion of tryptophan into kynurenine by IDO ultimately hinders T. gondii replication by depleting tryptophan that is necessary for the development of T. gondii [62]. However, Engin et al. [63] reported that during infection with T. gondii, breakdown of tryptophan may be induced via other unknown pathways as well, which may involve interactions between TDO, IDO and nitric oxide (NO), with NO possibly inhibiting complete exhaustion of tryptophan in acute infection with T. gondii. Studies, including ours, in participants with depressive disorders, have previously linked increased plasma levels of kynurenine [64] and its metabolite quinolinic acid [65], [66] to a history of suicide attempts [67].

How could kynurenine production lead to symptoms of depression and suicidal behavior? One possibility is that kynurenine can act as an immunosuppressant [68], [69], [70], [71] and thus, might promote reactivation of T. gondii intermittently. We previously reported that in patients with schizophrenia, only those who had both high kynurenine levels along with T. gondii seropositivity were at a higher risk of having a history of suicide attempts [72]. This brings up the possibility that higher kynurenine levels actuate the increased risk for suicidal self-directed violence (SSDV) by reactivation of latent T. gondii [72]. Another hypothesis is the excitotoxic N-methyl-D-aspartate (NMDA) receptor-mediated effects of quinolinic acid, a metabolite of kynurenine produced in the glial cells within the brain [61]. Both conceptual arguments and data support the hypothesis that depression may be induced via stimulation of NMDA receptors [73], [74], [75], [76], [77]. Dysfunction in NMDA glutamate receptors may also be implicated in the pathophysiology of suicide [75] as interventional studies, using intravenous ketamine (an NMDA antagonist) in patients with major depression, have demonstrated a rapid improvement in depression scores of these patients [78], [79] and a reduction in their suicidal ideation [80].

In contrast to dysphoria/hopelessness, we did not find a significant association between anhedonia scores and T. gondii serointensity. Anhedonia is defined as the inability to feel pleasure, which may be precipitated by a relative dopamine deficiency in the brain [81]. We hypothesize that the T. gondii-innate enzymatic capability to produce dopamine [82] may lead to a selective protection from anhedonia in T. gondii-linked depression, a hypothesis to be tested in future studies.

There are certain limitations to our study. Due to its cross-sectional nature, we cannot establish causality. Reverse causality has not been ruled out – it is possible that a higher T. gondii seropositivity in OOA with depressive symptoms (current dysphoria/hopelessness) could be the consequence, rather than the cause of depression. A possible explanation for depression causing increased exposure to T. gondii and higher T. gondii seropositivity includes less than optimal self-care (such as hand washing, vegetable cleaning and preparation of meat products) in depressed individuals.

Also, as our sample size was somewhat limited, we cannot rule out the possibility of a type II error with regard to the negative findings such as, the absence of associations between T. gondii and anhedonia (in contrast to dysphoria/hopelessness), and past symptoms (in contrast to present complaints). We did not use collateral information or standardized structured diagnostic instruments. Our PHQ-9 and PHQ-2 questionnaires were modified after feedback from members of the Amish community, and thus, the scores and thresholds on those questionnaires cannot be directly used to estimate diagnosis in the absence of thorough validation. Under any circumstances, meta-analyses have reported that both PHQ-9 and PHQ-2 scales cannot be used to clinically diagnose depression but they are useful for screening and referral for further clinical evaluation [37], [38].

There is a possibility that the participants might under-acknowledge and underreport certain symptoms of depression depending on how recently the episodes of low mood occurred, thus leading to recall or reporting bias. While there are a number of strengths in recruiting OOA, the Amish believe in limiting the expression of personal emotions. They value stoicism and expressing gratefulness rather than complaining, respond to hardships of daily life by yielding to divine providence and accepting it as God’s will (Gelassenheit). They often attribute the resolution of past symptoms to divine intervention [33] and once past symptoms resolve, they may be particularly prone to underreport them. Therefore, finding significant associations between T. gondii and “current, but not past”, hopelessness may be a result of a culturally based bias, and might not be generalizable to the non-OOA.

Nevertheless, we believe that the strengths of the study outweigh its limitations. Specifically, our study sample consisted of OOA, known to have a very low prevalence of substance use (which could be a major confounder in studies of depression), and a great deal of homogeneity in terms of food consumption, meal preparation, hygienic practices, social supports and occupation [33]. Recall and reporting biases notwithstanding, the Amish respond to questionnaires with well-known honesty and thoroughness that characterize the OOA culture [33], thereby increasing the reliability of the responses. Our past pencil-and-paper surveys of the Amish suggest that the OOA participants take the time to understand the questions being asked and to answer them with thoughtfulness and consistency [83]. Of major importance, the higher rate of seropositivity for T. gondii allows performing multivariable analyses with smaller sample sizes than in the general US population.

The findings from our study have a number of implications. Certain targeted treatment approaches might be able to benefit individual patients having both markers of chronic T. gondii infection and dysphoria/hopelessness, with/without anhedonia. For instance, cognitive behavioral therapy (CBT) has shown promise in reducing dysphoria/hopelessness in depressed patients [84], [85]. Brief supportive-expressive psychodynamic psychotherapy has also been shown to lower the serum levels of pro-inflammatory cytokines such as tumor necrosis factor (TNF) and interleukin 6 (IL-6) [86], that might play a mediating role after T. gondii reactivation in chronically infected individuals, and have been implicated in the metabolism of neurotransmitters, activation of neuroendocrine pathways, altered neuroplasticity [87] and behavioral manifestations seen in depressed individuals [88]. In T. gondii-seropositive patients with elevated markers of inflammation, clinical trials on anti-inflammatory drugs such as celecoxib [89], infliximab [90], or other cytokine inhibitors and anti-cytokine monoclonal antibodies [91], may lead to novel interventions for refractory or persistently suicidal depression. It is likely that treatment with anti-inflammatory agents may not be helpful for all cases of depression as inflammation is likely involved in only a sub-group of depressed patients that might be easily identified using molecular markers of inflammation [92], [93]. Anti-parasitic interventions aimed at preventing reactivation of T. gondii infection may also have a role in refractory and suicidal depression in T. gondii-seropositive patients with intermittent recurrent elevations in titers, suggesting reactivation. Furthermore, primary strategies to prevent T. gondii infection such as education about food preparation, food storage, avoiding eating raw or undercooked meat and hand hygiene prior to food preparation or meals could prove useful in lowering the morbidity, economic and emotional burden of depression, and possibly, its premature mortality through suicide.

Acknowledgments

This work was supported by Distinguished Investigator Award from the American Foundation for Suicide Prevention (Postolache, PI, Rujescu, co-I, DIG 1-162-12), with additional funding from the Mid-Atlantic Nutrition Obesity Research Center Pilot NORC grant (Postolache, PI), a subaward of the parent grant P30 DK072488 (Mitchell, PI) and from the Joint Institute for Food Safety and Applied Nutrition/UMD, through the cooperative agreement FDU.001418 (Postolache, PI). This study was also supported by the VA Merit Review CSR&D grant 1I01 CX001310-01A1 (Postolache, PI). Additional support for the writing of this manuscript was provided by the Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC, Denver, CO, USA) and by the Veterans Integrated Service Network 5, MIRECC (Baltimore, MD, USA). We thank the staff of the Amish Research Clinic of the University of Maryland for their overall support and the trainees of the Mood and Anxiety Program for their help with references, mailings and data management. The authors thank Alexandra T. Dagdag for her assistance in making corrections to the galley proof PDF of this article. The views, opinions and findings contained in this article belong to the authors and should not be construed as an official position of the NIH, American Foundation for Suicide Prevention, US Food and Drug Administration or the US Department of Veterans Affairs.

  1. Conflict of interest statement: The authors declare that they have no conflict of interest related to the publication of this paper.

References

1. Dubey JP. Toxoplasmosis of animals and humans, 2nd ed. Boca Raton, Florida: CRC Press, 2009.Search in Google Scholar

2. Pappas G, Roussos N, Falagas ME. Toxoplasmosis snapshots: global status of Toxoplasma gondii seroprevalence and implications for pregnancy and congenital toxoplasmosis. Int J Parasitol 2009;39:1385–94.10.1016/j.ijpara.2009.04.003Search in Google Scholar PubMed

3. Tenter AM, Heckeroth AR, Weiss LM. Toxoplasma gondii: from animals to humans. Int J Parasitol 2000;30:1217–58.10.1016/S0020-7519(00)00124-7Search in Google Scholar PubMed

4. Cook A, Holliman R, Gilbert R, Buffolano W, Zufferey J, Petersen E, et al. Sources of toxoplasma infection in pregnant women: European multicentre case-control study commentary: congenital toxoplasmosis – further thought for food. Br Med J 2000;321:142–7.10.1136/bmj.321.7254.142Search in Google Scholar

5. Centers for Disease Control and Prevention (CDC). Parasites – Toxoplasmosis (toxoplasma infection). Epidemiology and risk factors. 2017 [updated March 1, 2017]. Available from: https://www.cdc.gov/parasites/toxoplasmosis/epi.html.Search in Google Scholar

6. Hill DE, Chirukandoth S, Dubey JP. Biology and epidemiology of Toxoplasma gondii in man and animals. Anim Health Res Rev 2005;6:41–61.10.1079/AHR2005100Search in Google Scholar PubMed

7. Dubey JP, Lindsay DS, Speer CA. Structures of Toxoplasma gondii tachyzoites, bradyzoites, and sporozoites and biology and development of tissue cysts. Clin Microbiol Rev 1998;11:267–99.10.1128/CMR.11.2.267Search in Google Scholar PubMed

8. Dubey JP. Advances in the life cycle of Toxoplasma gondii. Int J Parasitol 1998;28:1019–24.10.1016/S0020-7519(98)00023-XSearch in Google Scholar PubMed

9. Peyron F, Mc Leod R, Ajzenberg D, Contopoulos-Ioannidis D, Kieffer F, Mandelbrot L, et al. Congenital toxoplasmosis in France and the United States: one parasite, two diverging approaches. PLoS Negl Trop Dis 2017;11:e0005222.10.1371/journal.pntd.0005222Search in Google Scholar PubMed

10. Halonen SK, Weiss LM. Toxoplasmosis. Handb Clin Neurol 2013;114:125–45.10.1016/B978-0-444-53490-3.00008-XSearch in Google Scholar PubMed

11. Dubey JP, Jones JL. Toxoplasma gondii infection in humans and animals in the United States. Int J Parasitol 2008;38:1257–78.10.1016/j.ijpara.2008.03.007Search in Google Scholar PubMed

12. Montoya JG, Liesenfeld O. Toxoplasmosis. Lancet 2004;363:1965–76.10.1016/S0140-6736(04)16412-XSearch in Google Scholar PubMed

13. Fuller Torrey E, Rawlings R, Yolken RH. The antecedents of psychoses: a case-control study of selected risk factors. Schizophr Res 2000;46:17–23.10.1016/S0920-9964(99)00237-6Search in Google Scholar PubMed

14. Sutterland AL, Fond G, Kuin A, Koeter MW, Lutter R, van Gool T, et al. Beyond the association. Toxoplasma gondii in schizophrenia, bipolar disorder, and addiction: systematic review and meta-analysis. Acta Psychiatr Scand 2015;132:161–79.10.1111/acps.12423Search in Google Scholar PubMed

15. Torrey EF, Bartko JJ, Lun ZR, Yolken RH. Antibodies to Toxoplasma gondii in patients with schizophrenia: a meta-analysis. Schizophr Bull 2007;33:729–36.10.1093/schbul/sbl050Search in Google Scholar PubMed PubMed Central

16. Torrey EF, Bartko JJ, Yolken RH. Toxoplasma gondii and other risk factors for schizophrenia: an update. Schizophr Bull 2012;38:642–7.10.1093/schbul/sbs043Search in Google Scholar PubMed PubMed Central

17. Yolken RH, Dickerson FB, Fuller Torrey E. Toxoplasma and schizophrenia. Parasite Immunol 2009;31:706–15.10.1111/j.1365-3024.2009.01131.xSearch in Google Scholar PubMed

18. Niebuhr DW, Millikan AM, Cowan DN, Yolken R, Li Y, Weber NS. Selected infectious agents and risk of schizophrenia among U.S. military personnel. Am J Psychiatry 2008;165:99–106.10.1176/appi.ajp.2007.06081254Search in Google Scholar PubMed

19. Amminger GP, McGorry PD, Berger GE, Wade D, Yung AR, Phillips LJ, et al. Antibodies to infectious agents in individuals at ultra-high risk for psychosis. Biol Psychiatry 2007;61:1215–7.10.1016/j.biopsych.2006.09.034Search in Google Scholar PubMed

20. Mortensen PB, Norgaard-Pedersen B, Waltoft BL, Sorensen TL, Hougaard D, Torrey EF, et al. Toxoplasma gondii as a risk factor for early-onset schizophrenia: analysis of filter paper blood samples obtained at birth. Biol Psychiatry 2007;61:688–93.10.1016/j.biopsych.2006.05.024Search in Google Scholar PubMed

21. Brown AS, Schaefer CA, Quesenberry CP Jr, Liu L, Babulas VP, Susser ES. Maternal exposure to toxoplasmosis and risk of schizophrenia in adult offspring. Am J Psychiatry 2005;162:767–73.10.1176/appi.ajp.162.4.767Search in Google Scholar PubMed

22. de Barros JL, Barbosa IG, Salem H, Rocha NP, Kummer A, Okusaga OO, et al. Is there any association between Toxoplasma gondii infection and bipolar disorder? A systematic review and meta-analysis. J Affect Disord 2017;209:59–65.10.1016/j.jad.2016.11.016Search in Google Scholar PubMed

23. Pearce BD, Kruszon-Moran D, Jones JL. The relationship between Toxoplasma gondii infection and mood disorders in the third National Health and Nutrition Survey. Biol Psychiatry 2012;72:290–5.10.1016/j.biopsych.2012.01.003Search in Google Scholar PubMed PubMed Central

24. Hamdani N, Daban-Huard C, Lajnef M, Richard JR, Delavest M, Godin O, et al. Relationship between Toxoplasma gondii infection and bipolar disorder in a French sample. J Affect Disord 2013;148:444–8.10.1016/j.jad.2012.11.034Search in Google Scholar PubMed

25. Oliveira J, Kazma R, Le Floch E, Bennabi M, Hamdani N, Bengoufa D, et al. Toxoplasma gondii exposure may modulate the influence of TLR2 genetic variation on bipolar disorder: a gene-environment interaction study. Int J Bipolar Disord 2016;4:11.10.1186/s40345-016-0052-6Search in Google Scholar PubMed PubMed Central

26. Alvarado-Esquivel C, Sanchez-Anguiano LF, Hernandez-Tinoco J, Berumen-Segovia LO, Torres-Prieto YE, Estrada-Martinez S, et al. Toxoplasma gondii infection and depression: a case-control seroprevalence study. Eur J Microbiol Immunol (Bp) 2016;6: 85–9.10.1556/1886.2016.00010Search in Google Scholar PubMed PubMed Central

27. Duffy AR, Beckie TM, Brenner LA, Beckstead JW, Seyfang A, Postolache TT, et al. Relationship between Toxoplasma gondii and mood disturbance in women veterans. Mil Med 2015;180:621–5.10.7205/MILMED-D-14-00488Search in Google Scholar PubMed

28. Groer MW, Yolken RH, Xiao JC, Beckstead JW, Fuchs D, Mohapatra SS, et al. Prenatal depression and anxiety in Toxoplasma gondii-positive women. Am J Obstet Gynecol 2011;204:433.e1–7.10.1016/j.ajog.2011.01.004Search in Google Scholar PubMed PubMed Central

29. Alvarado-Esquivel C, Martinez-Martinez AL, Sanchez-Anguiano LF, Hernandez-Tinoco J, Castillo-Orona JM, Salas-Martinez C, et al. Lack of association between Toxoplasma gondii exposure and depression in pregnant women: a case-control study. BMC Infect Dis. 2017;17:190.10.1186/s12879-017-2292-1Search in Google Scholar PubMed PubMed Central

30. Flegr J, Hodny Z. Cat scratches, not bites, are associated with unipolar depression – cross-sectional study. Parasit Vectors 2016;9:8.10.1186/s13071-015-1290-7Search in Google Scholar PubMed PubMed Central

31. Gale SD, Brown BL, Berrett A, Erickson LD, Hedges DW. Association between latent toxoplasmosis and major depression, generalised anxiety disorder and panic disorder in human adults. Folia Parasitol (Praha) 2014;61:285–92.10.14411/fp.2014.038Search in Google Scholar PubMed

32. Gale SD, Berrett AN, Brown B, Erickson LD, Hedges DW. No association between current depression and latent toxoplasmosis in adults. Folia Parasitol (Praha) 2016;63.10.14411/fp.2016.032Search in Google Scholar PubMed

33. Kraybill DB, Johnson-Weiner KM, Nolt SM. The Amish. Baltimore, Maryland: The John Hopkins University Press, 2013.10.56021/9781421409146Search in Google Scholar

34. Hill D, Coss C, Dubey JP, Wroblewski K, Sautter M, Hosten T, et al. Identification of a sporozoite-specific antigen from Toxoplasma gondii. J Parasitol 2011;97:328–37.10.1645/GE-2782.1Search in Google Scholar PubMed

35. Kroenke K, Spitzer RL, Williams JBW. The PHQ-9: validity of a brief depression severity measure. J Gen Intern Med 2001;16:606–13.10.1046/j.1525-1497.2001.016009606.xSearch in Google Scholar PubMed

36. Kroenke K, Spitzer RL, Williams JB. The patient health questionnaire-2: validity of a two-item depression screener. Med Care 2003;41:1284–92.10.1097/01.MLR.0000093487.78664.3CSearch in Google Scholar PubMed

37. Gilbody S, Richards D, Brealey S, Hewitt C. Screening for depression in medical settings with the Patient Health Questionnaire (PHQ): a diagnostic meta-analysis. J Gen Intern Med 2007;22:1596–602.10.1007/s11606-007-0333-ySearch in Google Scholar PubMed

38. Mitchell AJ, Yadegarfar M, Gill J, Stubbs B. Case finding and screening clinical utility of the Patient Health Questionnaire (PHQ-9 and PHQ-2) for depression in primary care: a diagnostic meta-analysis of 40 studies. BJPsych Open 2016;2:127–38.10.1192/bjpo.bp.115.001685Search in Google Scholar PubMed

39. Jones-Brando L, Torrey EF, Yolken R. Drugs used in the treatment of schizophrenia and bipolar disorder inhibit the replication of Toxoplasma gondii. Schizophr Res 2003;62:237–44.10.1016/S0920-9964(02)00357-2Search in Google Scholar PubMed

40. Fond G, Boyer L, Gaman A, Laouamri H, Attiba D, Richard JR, et al. Treatment with anti-toxoplasmic activity (TATA) for toxoplasma positive patients with bipolar disorders or schizophrenia: a cross-sectional study. J Psychiatr Res 2015;63:58–64.10.1016/j.jpsychires.2015.02.011Search in Google Scholar PubMed

41. Kar N, Misra B. Toxoplasma seropositivity and depression: a case report. BMC Psychiatry 2004;4:1.10.1186/1471-244X-4-1Search in Google Scholar PubMed PubMed Central

42. Cetinkaya Z, Yazar S, Gecici O, Namli MN. Anti-Toxoplasma gondii antibodies in patients with schizophrenia – preliminary findings in a Turkish sample. Schizophr Bull 2007;33:789–91.10.1093/schbul/sbm021Search in Google Scholar PubMed PubMed Central

43. Hinze-Selch D, Daubener W, Eggert L, Erdag S, Stoltenberg R, Wilms S. A controlled prospective study of toxoplasma gondii infection in individuals with schizophrenia: beyond seroprevalence. Schizophr Bull 2007;33:782–8.10.1093/schbul/sbm010Search in Google Scholar PubMed PubMed Central

44. Hamidinejat H, Ghorbanpoor M, Hosseini H, Alavi SM, Nabavi L, Jalali MH, et al. Toxoplasma gondii infection in first-episode and inpatient individuals with schizophrenia. Int J Infect Dis 2010;14:e978–81.10.1016/j.ijid.2010.05.018Search in Google Scholar PubMed

45. Massa NM, Duncan E, Jovanovic T, Kerley K, Weng L, Gensler L, et al. Relationship between Toxoplasma gondii seropositivity and acoustic startle response in an inner-city population. Brain Behav Immun 2017;61:176–83.10.1016/j.bbi.2016.11.021Search in Google Scholar PubMed PubMed Central

46. Flegr J, Escudero DQ. Impaired health status and increased incidence of diseases in Toxoplasma-seropositive subjects – an explorative cross-sectional study. Parasitology 2016;143:1974–89.10.1017/S0031182016001785Search in Google Scholar PubMed

47. Arling TA, Yolken RH, Lapidus M, Langenberg P, Dickerson FB, Zimmerman SA, et al. Toxoplasma gondii antibody titers and history of suicide attempts in patients with recurrent mood disorders. J Nerv Ment Dis 2009;197:905–8.10.1097/NMD.0b013e3181c29a23Search in Google Scholar PubMed

48. Yagmur F, Yazar S, Temel HO, Cavusoglu M. May Toxoplasma gondii increase suicide attempt-preliminary results in Turkish subjects? Forensic Sci Int 2010;199:15–7.10.1016/j.forsciint.2010.02.020Search in Google Scholar PubMed

49. Okusaga O, Langenberg P, Sleemi A, Vaswani D, Giegling I, Hartmann AM, et al. Toxoplasma gondii antibody titers and history of suicide attempts in patients with schizophrenia. Schizophr Res 2011;133:150–5.10.1016/j.schres.2011.08.006Search in Google Scholar PubMed

50. Zhang Y, Traskman-Bendz L, Janelidze S, Langenberg P, Saleh A, Constantine N, et al. Toxoplasma gondii immunoglobulin G antibodies and nonfatal suicidal self-directed violence. J Clin Psychiatry 2012;73:1069–76.10.4088/JCP.11m07532Search in Google Scholar PubMed

51. Pedersen MG, Mortensen PB, Norgaard-Pedersen B, Postolache TT. Toxoplasma gondii infection and self-directed violence in mothers. Arch Gen Psychiatry 2012;69:1123–30.10.1001/archgenpsychiatry.2012.668Search in Google Scholar PubMed

52. Dickerson F, Wilcox HC, Adamos M, Katsafanas E, Khushalani S, Origoni A, et al. Suicide attempts and markers of immune response in individuals with serious mental illness. J Psychiatr Res 2017;87:37–43.10.1016/j.jpsychires.2016.11.011Search in Google Scholar PubMed

53. Beck AT. Hopelessness as a predictor of eventual suicide. Ann N Y Acad Sci 1986;487:90–6.10.1111/j.1749-6632.1986.tb27888.xSearch in Google Scholar PubMed

54. Gould TD, Georgiou P, Brenner LA, Brundin L, Can A, Courtet P, et al. Animal models to improve our understanding and treatment of suicidal behavior. Transl Psychiatry 2017;7:e1092.10.1038/tp.2017.50Search in Google Scholar PubMed PubMed Central

55. Sugden K, Moffitt TE, Pinto L, Poulton R, Williams BS, Caspi A. Is Toxoplasma gondii infection related to brain and behavior impairments in humans? Evidence from a population-representative birth cohort. PLoS One 2016;11:e0148435.10.1371/journal.pone.0148435Search in Google Scholar PubMed PubMed Central

56. Cook TB, Brenner LA, Cloninger CR, Langenberg P, Igbide A, Giegling I, et al. “Latent” infection with Toxoplasma gondii: association with trait aggression and impulsivity in healthy adults. J Psychiatr Res 2015;60:87–94.10.1016/j.jpsychires.2014.09.019Search in Google Scholar PubMed

57. Coccaro EF, Lee R, Groer MW, Can A, Coussons-Read M, Postolache TT. Toxoplasma gondii infection: relationship with aggression in psychiatric subjects. J Clin Psychiatry 2016;77:334–41.10.4088/JCP.14m09621Search in Google Scholar PubMed

58. Fawcett J, Scheftner WA, Fogg L, Clark DC, Young MA, Hedeker D, et al. Time-related predictors of suicide in major affective disorder. Am J Psychiatry 1990;147:1189–94.10.1176/ajp.147.9.1189Search in Google Scholar PubMed

59. Owens MJ, Nemeroff CB. Role of serotonin in the pathophysiology of depression: focus on the serotonin transporter. Clin Chem 1994;40:288–95.10.1093/clinchem/40.2.288Search in Google Scholar PubMed

60. Birdsall TC. 5-Hydroxytryptophan: a clinically-effective serotonin precursor. Altern Med Rev 1998;3:271–80.Search in Google Scholar PubMed

61. Vecsei L, Szalardy L, Fulop F, Toldi J. Kynurenines in the CNS: recent advances and new questions. Nat Rev Drug Discov 2013;12:64–82.10.1038/nrd3793Search in Google Scholar PubMed

62. Notarangelo FM, Wilson EH, Horning KJ, Thomas MA, Harris TH, Fang Q, et al. Evaluation of kynurenine pathway metabolism in Toxoplasma gondii-infected mice: implications for schizophrenia. Schizophr Res 2014;152:261–7.10.1016/j.schres.2013.11.011Search in Google Scholar PubMed PubMed Central

63. Engin AB, Dogruman-Al F, Ercin U, Celebi B, Babur C, Bukan N. Oxidative stress and tryptophan degradation pattern of acute Toxoplasma gondii infection in mice. Parasitol Res 2012;111:1725–30.10.1007/s00436-012-3015-6Search in Google Scholar PubMed

64. Sublette ME, Galfalvy HC, Fuchs D, Lapidus M, Grunebaum MF, Oquendo MA, et al. Plasma kynurenine levels are elevated in suicide attempters with major depressive disorder. Brain Behav Immun 2011;25:1272–8.10.1016/j.bbi.2011.05.002Search in Google Scholar PubMed PubMed Central

65. Brundin L, Sellgren CM, Lim CK, Grit J, Palsson E, Landen M, et al. An enzyme in the kynurenine pathway that governs vulnerability to suicidal behavior by regulating excitotoxicity and neuroinflammation. Transl Psychiatry 2016;6:e865.10.1038/tp.2016.133Search in Google Scholar PubMed

66. Erhardt S, Lim CK, Linderholm KR, Janelidze S, Lindqvist D, Samuelsson M, et al. Connecting inflammation with glutamate agonism in suicidality. Neuropsychopharmacology 2013;38:743–52.10.1038/npp.2012.248Search in Google Scholar PubMed

67. Bay-Richter C, Linderholm KR, Lim CK, Samuelsson M, Traskman-Bendz L, Guillemin GJ, et al. A role for inflammatory metabolites as modulators of the glutamate N-methyl-D-aspartate receptor in depression and suicidality. Brain Behav Immun 2015;43:110–7.10.1016/j.bbi.2014.07.012Search in Google Scholar PubMed

68. Bauer TM, Jiga LP, Chuang J-J, Randazzo M, Opelz G, Terness P. Studying the immunosuppressive role of indoleamine 2,3-dioxygenase: tryptophan metabolites suppress rat allogeneic T-cell responses in vitro and in vivo. Transpl Int 2005;18:95–100.10.1111/j.1432-2277.2004.00031.xSearch in Google Scholar PubMed

69. Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara GB. Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase. J Exp Med 2002;196:459–68.10.1084/jem.20020121Search in Google Scholar

70. Mellor AL, Baban B, Chandler P, Marshall B, Jhaver K, Hansen A, et al. Cutting edge: induced indoleamine 2,3 dioxygenase expression in dendritic cell subsets suppresses T cell clonal expansion. J Immunol 2003;171:1652–5.10.4049/jimmunol.171.4.1652Search in Google Scholar PubMed

71. Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med 1999;189:1363–72.10.1084/jem.189.9.1363Search in Google Scholar PubMed

72. Okusaga O, Duncan E, Langenberg P, Brundin L, Fuchs D, Groer MW, et al. Combined Toxoplasma gondii seropositivity and high blood kynurenine–Linked with nonfatal suicidal self-directed violence in patients with schizophrenia. J Psychiatr Res 2016;72:74–81.10.1016/j.jpsychires.2015.10.002Search in Google Scholar

73. Skolnick P, Layer RT, Popik P, Nowak G, Paul IA, Trullas R. Adaptation of N-methyl-D-aspartate (NMDA) receptors following antidepressant treatment: implications for the pharmacotherapy of depression. Pharmacopsychiatry 1996;29:23–6.10.1055/s-2007-979537Search in Google Scholar PubMed

74. Trullas R, Skolnick P. Functional antagonists at the NMDA receptor complex exhibit antidepressant actions. Eur J Pharmacol 1990;185:1–10.10.1016/0014-2999(90)90204-JSearch in Google Scholar PubMed

75. Nowak G, Ordway GA, Paul IA. Alterations in the N-methyl-D-aspartate (NMDA) receptor complex in the frontal cortex of suicide victims. Brain Res 1995;675:157–64.10.1016/0006-8993(95)00057-WSearch in Google Scholar PubMed

76. Krystal JH, Sanacora G, Blumberg H, Anand A, Charney DS, Marek G, et al. Glutamate and GABA systems as targets for novel antidepressant and mood-stabilizing treatments. Mol Psychiatry 2002;7(Suppl 1):S71–80.10.1038/sj.mp.4001021Search in Google Scholar PubMed

77. Zarate CA, Quiroz J, Payne J, Manji HK. Modulators of the glutamatergic system: implications for the development of improved therapeutics in mood disorders. Psychopharmacol Bull 2002;36:35–83.Search in Google Scholar PubMed

78. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry 2000;47:351–4.10.1016/S0006-3223(99)00230-9Search in Google Scholar PubMed

79. Zarate CA Jr, Singh JB, Carlson PJ, Brutsche NE, Ameli R, Luckenbaugh DA, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry 2006;63:856–64.10.1001/archpsyc.63.8.856Search in Google Scholar PubMed

80. DiazGranados N, Ibrahim LA, Brutsche NE, Ameli R, Henter ID, Luckenbaugh DA, et al. Rapid resolution of suicidal ideation after a single infusion of an N-methyl-D-aspartate antagonist in patients with treatment-resistant major depressive disorder. J Clin Psychiatry 2010;71:1605–11.10.4088/JCP.09m05327bluSearch in Google Scholar PubMed PubMed Central

81. Berrios GE. The history of mental symptoms: descriptive psychopathology since the nineteenth century. Cambridge, UK: Cambridge University Press, 1996.10.1017/CBO9780511526725Search in Google Scholar

82. Prandovszky E, Gaskell E, Martin H, Dubey JP, Webster JP, McConkey GA. The neurotropic parasite Toxoplasma gondii increases dopamine metabolism. PLoS One 2011;6:e23866.10.1371/journal.pone.0023866Search in Google Scholar PubMed PubMed Central

83. Patel F, Postolache N, Mohyuddin H, Vaswani D, Balis T, Raheja UK, et al. Seasonality patterns of mood and behavior in the Old Order Amish. Int J Disabil Hum Dev 2012;12:53–60.10.1515/ijdhd-2012-0127Search in Google Scholar PubMed PubMed Central

84. Handley TE, Kay-Lambkin FJ, Baker AL, Lewin TJ, Kelly BJ, Inder KJ, et al. Incidental treatment effects of CBT on suicidal ideation and hopelessness. J Affect Disord 2013;151: 275–83.10.1016/j.jad.2013.06.005Search in Google Scholar PubMed

85. Gudmundsdottir RM, Thome M. Evaluation of the effects of individual and group cognitive behavioural therapy and of psychiatric rehabilitation on hopelessness of depressed adults: a comparative analysis. J Psychiatr Ment Health Nurs 2014;21:866–72.10.1111/jpm.12157Search in Google Scholar PubMed

86. Del Grande da Silva G, Wiener CD, Barbosa LP, Goncalves Araujo JM, Molina ML, San Martin P, et al. Pro-inflammatory cytokines and psychotherapy in depression: results from a randomized clinical trial. J Psychiatr Res 2016;75:57–64.10.1016/j.jpsychires.2016.01.008Search in Google Scholar PubMed

87. Raison CL, Capuron L, Miller AH. Cytokines sing the blues: inflammation and the pathogenesis of depression. Trends Immunol 2006;27:24–31.10.1016/j.it.2005.11.006Search in Google Scholar PubMed PubMed Central

88. Schiepers OJ, Wichers MC, Maes M. Cytokines and major depression. Prog Neuro-Psychopharmacol Biol Psychiatry 2005;29:201–17.10.1016/j.pnpbp.2004.11.003Search in Google Scholar PubMed

89. Kohler O, Benros ME, Nordentoft M, Farkouh ME, Iyengar RL, Mors O, et al. Effect of anti-inflammatory treatment on depression, depressive symptoms, and adverse effects: a systematic review and meta-analysis of randomized clinical trials. JAMA Psychiatry 2014;71:1381–91.10.1001/jamapsychiatry.2014.1611Search in Google Scholar PubMed

90. Raison CL, Rutherford RE, Woolwine BJ, Shuo C, Schettler P, Drake DF, et al. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA Psychiatry 2013;70:31–41.10.1001/2013.jamapsychiatry.4Search in Google Scholar PubMed PubMed Central

91. Kappelmann N, Lewis G, Dantzer R, Jones PB, Khandaker GM. Antidepressant activity of anti-cytokine treatment: a systematic review and meta-analysis of clinical trials of chronic inflammatory conditions. Mol Psychiatry 2016.10.1016/j.bbi.2017.07.023Search in Google Scholar PubMed

92. Wium-Andersen MK, Orsted DD, Nielsen SF, Nordestgaard BG. Elevated C-reactive protein levels, psychological distress, and depression in 73, 131 individuals. JAMA Psychiatry 2013;70: 176–84.10.1001/2013.jamapsychiatry.102Search in Google Scholar PubMed

93. Mondelli V, Ciufolini S, Belvederi Murri M, Bonaccorso S, Di Forti M, Giordano A, et al. Cortisol and inflammatory biomarkers predict poor treatment response in first episode psychosis. Schizophr Bull 2015;41:1162–70.10.1093/schbul/sbv028Search in Google Scholar PubMed PubMed Central


Article note:

The findings of this study were presented at the “36th International Winter-Workshop on Clinical, Chemical and Biochemical Aspects of Pteridines and Related Topics” in Innsbruck, Austria on February 23rd, 2017, and were also published as an abstract in Pteridines for the above annual workshop. The findings of this study were also presented as a poster at the “72nd Annual Scientific Convention and Meeting” of the Society of Biological Psychiatry on May 18, 2017, in San Diego, California, USA, and were also published as an abstract in the supplement to Biological Psychiatry for this annual meeting.


Received: 2017-9-8
Accepted: 2017-9-30
Published Online: 2017-11-22
Published in Print: 2017-12-20

©2017 Walter de Gruyter GmbH, Berlin/Boston

This article is distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Downloaded on 24.4.2024 from https://www.degruyter.com/document/doi/10.1515/pterid-2017-0019/html
Scroll to top button