jdm

Journal of Diabetes & Metabolism

ISSN - 2155-6156

Review Article - (2013) Volume 4, Issue 2

T. gondii Infection Acquired during Pregnancy and/or after Birth may be Responsible for Development of both Type 1 and 2 Diabetes Mellitus

Joseph Prandota*
Department of Social Pediatrics, Faculty of Health Sciences, Wroclaw Medical University, Poland
*Corresponding Author: Joseph Prandota, Faculty of Health Sciences, Department of Social Pediatrics, Wroclaw Medical University, 5 Bartla Str., Wroclaw, Poland Email:

Abstract

Recently, it was suggested that maternal T and potentially B cells transferred during pregnancy and/or the breast milk feeding and their encounter with the antigen in mesenteric lymph nodes might play a role in development of type 1 diabetes mellitus (T1DM). T. gondii infection during gestation and/or after birth may be responsible for development of both T1DM and T2DM in children, adolescents and adults because: a) maternal microchimerism in peripheral blood was demonstrated to be significantly higher in patients with T1DM compared to unaffected siblings and healthy subjects, b) transmission of T. gondii as a Trojan horse in various types of eukaryotic cells, including T and B lymphocytes, c) swallowing by the fetus of amniotic fluid containing infected leukocytes and other cells, d) elimination of T. gondii in the breast milk during lactation, e) involvement of mesenteric lymph nodes after oral infection with the parasite, and f) damage of the myenteric neurons during infection with the parasite in both the animals with streptozotocin-induced diabetes and diabetic patients. Moreover, a significantly lower occurrence of antibodies against T. gondii found in the sera of patients with T1DM compared with their first-degree family members or healthy controls may be due to their T and/or B cell exhaustion perspective caused by chronic infection with the parasite. This suggestion may be supported by the finding that latent toxoplasmosis was associated with markedly reduced lymphocyte B-cell counts responsible for production of antibodies, markedly lower serum IgG, IgM, and IgA levels, and a significant suppression of IL-2. On the other hand, patients with T2DM had increased anti-T. gondii antibodies significantly more frequently than respective controls. Impaired vascular endothelial function characteristic for the patients with diabetes mellitus may be at least in part due to the preferential T. gondii infection of endothelial cells. Vitamin D and minocycline exerted beneficial effects on development and clinical course of diabetes mellitus probably because of their immunomodulatory and antitoxoplasmatic activities. These data strongly suggest that the parasite play an important role in development of both types of diabetes mellitus.

Keywords: Diabetes mellitus; T. gondii infection; Toxoplasmosis; Microchimerism; Diabetic ketoacidosis; Amyloid deposition; Vitamin D deficiency; Diabetes comorbidities; Diabetes termal therapy; Cold stress; Glucose-6-dehydrogenase deficiency; Indoleamine 2,3-dioxygenase; Minocycline

Introduction

Diabetes mellitus is characterized by persistent hyperglycemia with disturbances of carbohydrate, fat and protein metabolism that results from abnormalities in insulin secretion, action or both [1]. About 350 million people across the globe are estimated to have diabetes [2], and type 2 diabetes mellitus (T2DM) accounts roughly 90% of all diagnosed cases [1]. Diabetes has a prevalence of 2-5% in most Western countries, and is rapidly increasing in Asiatic countries due to changes in dietary habits during the last years [3]. From 1980 through 2010, the number of Americans with diagnosed diabetes has more than tripled (from 5.6 to 20.9 million) [4], and in the UK insulin use in children and adolescents increased significantly from 1.08 per 1000 children in 1998 to 1.98 in 2005 (p<0.001) [5]. Type 1 diabetes (T1DM), previously called insulindependent diabetes mellitus or juvenile-onset diabetes, accounts for about 10% of all diagnosed cases, and usually affects children and young adults, although disease onset can occur at any age. Risk factors may be autoimmune, genetic, and/or environmental [6].

In T1DM, pancreatic islet β cells are destroyed as a result of autoimmune processes causing severe insulin deficiency. Without insulin, blood glucose concentration increase with glucose uptake into muscle (energy), and to the liver (storage of glycogen), and hepatic gluconeogenesis also continues unabated, while ketone bodies and keto-acids are accumulating, finally leading to acute metabolic crises [7]. In T2DM, increases in insulin resistance lead to enhanced demand for insulin generation, beta cell hypertrophy, beta cell damage and fibrosis caused by excessive ROS/RNI production and/or other molecular pathomechanisms, with further reductions in insulin secretion [7].

T. gondii is a protozoan parasite known to infect animals, birds and mammals, including about 30-50% of the world human immunocompetent population who have chronic asymptomatic infection and harbor parasite cysts especially in the central nervous system [8,9]. In Europe, North America, and Africa, there are three dominant clonal lineages of T. gondii called type I (e.g. RH and GT1), type II (ME49), and type III (VEG), which differ in prevalence, virulence, migratory capacity within the host, and ability to convert to the bradyzoite cyst phase [10]. Geoepidemiological prevalence of the parasite varies depending on the world region, ranging from as low as 4% in some areas of the Far East, through 10-30% in the US, and from 10% to up to 60% in European regions with high consumption of raw food, such as France [11-13].

T. gondii can infect and replicate in virtually any nucleated host cells and specifically increase the levels of key host microRNAs [14]. IFN-γ- mediated immune responses control the parasite in both phagocytic and non-phagocytic cells through at least six different mechanisms depending on the types of cells responding to this cytokine. Such effector functions involve: 1) mechanisms mediated by IFN-γ responsive gene family proteins, including IGTP (an essential mediator of specialized antimicrobial activities of IFN-γ), which may be involved in the processing and trafficking of cytokines and/or antigens; 2) production of NO by inducible NO synthase (iNOS); 3) production of various cytokines (TNF-α, IFN-γ, IL-1β, etc.); 4) tryptophan degradation by indoleamine 2,3-dioxygenase and tryptophan 2,3-dioxygenase; 5) limiting the availability of intracellular iron to the parasite, and 6) production of reactive oxygen/nitrogen species/intermediates (ROS/ RNI) [15,16].

Immunocompetent hosts infected with T. gondii must develop a powerful immune response that has to be under tight control [17] and persistently maintained during their lifetime in all infected tissues to avoid life-threatening toxoplasmic encephalitis after reactivation of latent parasites [18,19].

Possible Association between Diabetes Mellitus, Autoimmunity and T. gondii Infection

A significantly lower occurrence of antibodies against T. gondii and some viruses was reported in the sera of patients with T1DM compared with their first-degree family members or healthy controls [20]. This very interesting finding may be caused by the impaired innate immune state capacity of those individuals caused by chronic infection with the parasite (probably acquired early during prenatal life), which is primarily responsible for the prolific production of autoantibodies due to various autoimmune condition. Host cellmediated immune responses are suppressed during chronic infection with T. gondii, and this was associated with significant suppression of IL-2, IFN-γ, and markedly lower levels of IgG1, IgG2a, IgG2b, IgG3, IgA, and IgM [21] (Tables 1 and 2). This reasoning may be supported by the fact that latent toxoplasmosis was associated with markedly reduced lymphocyte B-cell counts responsible for production of antibodies [22], and is consistent with the current studies on CD8 T cell deficiency and probable exhaustion perspective observed during chronic toxoplasmosis [23].

Infectious agent T1DM FM Controls p value
T. gondii 5.4 24.4 40.0 0.001
EBV-VCA (IgG anti-VCA) 82.1 92.6 92.8 0.04
EBV-EBNA (IgG anti-EBNA) 71.4 89.3 90.7 0.001
CMV 69.6 79.7 92.9 0.001
HP 55.1 78.3 80.7 0.01
CMV: Cytomegalovirus; EBV: Ebstein-Barr Virus; EBV-VCA: EBV Viral Capsid Antigen; EBV-EBNA: EB Nuclear Antigen; HP: Helicobacter Pylori

Table 1: Percentages of T1DM patients, their first-degree family members (FM) and healthy controls with antibodies against T. gondii and some other infectious agents (according to Krause et al. [20]; with own modification).

Autoantibody T1DM FM Controls p value
Antigliadin IgG 31.0 8.2 1.4 0.001
Antitissue transglutaminase IgG 3.5 0 0 0.03
Anticentromere 3.6 0.8 0 0.06

Table 2: Percentages of autoantibodies associated with various autoimmune condition in T1DM patients,their first-degree Family Members (FM) and healthy controls (according to Krause et al. [20]; with own modification).

Gokce et al. [24] studied seropositivity rate of anti-T. gondii antibodies in 807 persons (351 men, 456 women; mean age 52.8 ± 14.01 SD, range 15-88 yrs) with T2DM and found that IgG antibodies were present significantly more frequently as compared with 250 healthy controls (110 men, 140 women; mean age 51.94 ± 13.44, range 18-75 yrs) (p<0.001). IgM antibodies were found in 19 of the patients with diabetes mellitus and only in 4 controls (2.4 vs. 1.6%, p=0.3) [24] (Table 3). These findings may be supported by the geoepidemiology of autoimmune diseases which demonstrates that genetic individual susceptibility interacted with lifestyle and environmental factors, such as socioeconomic status, nutritional habits, environmental pollutants, and viral, bacterial and parasitic infections, acting as triggering and/ or protective agents [25-29]. For example, helminths were found to be protective in T1DM, autoimmune encephalitis, and ulcerative colitis probably via an induction of proinflammatory responses (TH1 cytokines) and a concomitant development of type 2 Th cell line adaptive immunity [25,26]. Shapira et al. [25] suggested that T. gondii infection can initiate a pathogenic process that may eventually result in clinically overt autoimmunity because serum anti-toxoplasma antibodies IgG were positive in 42% of 1514 European patients with 11 different autoimmune diseases as compared with 29% of controls (p<0.0001). In addition, ATxA IgM were more prevalent in the patients with anti-phospholipid syndrome (p<0.01), systemic sclerosis (p<0.05), and inflammatory bowel disease (p<0.05) than in controls [25]. Maternal microchimerism was found in the peripheral blood of patients with T1DM and pancreatic islet beta cell microchimerism [30]. It was demonstrated that this event leads to the production of IL-2, a proinflammatory cytokin, in IL-2 knockout mice [31]. Several autoimmune diseases due to maternal/fetal microchimerism were presented in table 4. Some of these diseases were probably associated with transmission of the parasite in various nucleated cells as a Trojan horse, because the percentage of T. gondii positive persons increases with age (Table 5).

Duration of T2DM (yrs) Number of IgG-positive individuals Number of IgG-negative individuals
0-5 39 (16.8) 193 (83.2)
6-10 149 (51.6) 140 (48.4)
> 11 269 (94.1) 17 (5.9)
Values in parentheses denote percentages

Table 3: Relationship between the seropositivity rate of anti-T. gondii antibodies analyzed in 807 individuals with T2DM and duration of the disease (according to Gokce et al. [24]; with own modification).

Disease Female/male ratio Tissue source References
Systemic sclerosis ≥ 8:1 Peripheral blood cells [33,34]
Juvenile idiopathic inflammatory myopathy 3:1 Sorted CD4+ or CD8+ peripheral blood cells [35,36]
Systemic lupus erythematosus 5:1 Peripheral blood nucleated cells [37]
Sjögren syndrome 9:1 Peripheral blood whole nucleated cells [37]
Primary biliary cirrhosis 14:1 Peripheral blood nucleated cells [38]
Hashimoto’s thyroiditis 20:1 Thyroid tissue [39]
Graves’ disease 8:1 Thyroid tissue [40]
Lichen planus 2:1 Peripheral blood nucleated cells [41]
Polymorphic eruptions of pregnancy pregnancy   [42]
Interestingly, maternal microchimerism was found in the peripheral blood of patients with T1DM and pancreatic islet beta cell microchimerism [43]. It was demonstrated that this bioevent also leads to the production of IL-2,a proinflammatory cytokine,in IL-2 knockout mice [44]

Table 4: Autoimmune diseases associated with fetal and/or maternal microchimerism (according to Klonisch and Drouin [32]; with own modification).

Percent
T. gondii positive
Age (yrs)                      
  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                        
In the control individuals 45 yrs old or younger recruited from the same geographical region as the psychiatric patients admitted to the hospital, serofrequency of T. gondii infection ranged between 20 and 40% without any systematic age effect, whereas in the individuals older than 45 yrs serofrequency systematically increased with age from about 40% to almost 100% [45]

Table 5: Percentage of T. gondii positive individuals among 214 nonpsychiatrically affected controls depending on age analyzed during a large epidemiologic study of 869 psychiatric patients [45,46].

It should be emphasized that insulin and D-glucose had a doseresponsive mitogenic effect on intracellular T. gondii replication and development in 3T3-L1 cells. In vitro insulin concentrations between 10-2 and 10-1 μg/ml combination of 4.5 g/l D-glucose in DMEM (Dulbecco’s Modified Eagle Medium) gave maximum stimulus to T. gondii replication [44]. In the absence of D-glucose, insulin had comparably less effect on the parasite growth than two of their combination. D-glucose markedly affected the tachyzoite replication and appeared to be indispensable for maintaining the host 3T3-L1 cells [44]. Thus, the additive/synergistic effect of insulin plus D-glucose on multiplication of the parasite in pancreatic islet beta-cells may be at least in part responsible first for inducing insulitis, and then progressing to diabetes, as well as for triggering development of various autoimmune defense reactions of the host.

In summary, there is a strong laboratory, pathophysiologic and clinical evidence supporting suggestion about the association between chronic latent T. gondii infection and both development of diabetes mellitus and several concomitant autoimmune diseases.

Similarities between Genetic Contribution of HLADQ Molecules to the Development of Diabetes and Genetically-Dependent Outcome of T. gondii Infection

Diabetes

In humans, T1DM is associated with genes encoding the MHC, particularly the class II molecule DQA1*0301/DQB1*0302 [47-50]. About 90% of patients with T1DM express HLA-DQ8/DR4 or HLADQ2/ DR3 biomolecules [48]. Geogenetically, in Chinese population DQA1*0501, DQA1*0501, DQB1*0201, and DQB1*0302 were the susceptible alleles (all p<0.005) are relevant to T1DM, which is not totally the same as non-Chinese populations [51]. It must be emphasized that in the patients with the T1DM-associated DQB1*0302-DRB1*04 haplotype, maternal microchimerism was found more often when the haplotype was paternally (70%) rather than maternally transmitted (14%) [30]. This finding is in agreement with the suggestion that not only the inherited but also non-inherited maternal HLA haplotypes may influence the risk for development of T1DM [52].

In mice, Wen et al. [50] provided direct in vivo evidence for the contribution of HLA-DQ molecules to the development of diabetes. They found that substitution of HLA-DQA1*0301/DQB1*0302 for murine MHC class II provoked autoimmune diabetes in nondiabetes- prone rat insulin promoter RIP-B7-1 C57BL/6 mice. Rajagopalan et al. [53] showed that spontaneous diabetes occurred in RIP-B7-1 transgenic mice expressing transgenic HLA-DR3 or HLA– DQ8 molecules and the incidence of the disease was comparable between the two (approximately 30% in either sex up to 50 weeks of age). However, Kudva et al. [54] found that in NOD mice lacking endogenous class II molecules, transgenic expression of HLA-DR3 and HLA-DQ8 associated with predisposition to T1DM alone was not sufficient to induce spontaneous diabetes. It should be noted that the induction of immunodominant, protective CD8+ T cell responses to T. gondii infection requires proteolysis by the endoplasmic reticulum aminopeptidase associated with antigen processing (ERAAP) in the endoplasmic reticulum [55]. Although a key function for ERAAP is shaping many precursor peptides to the appropriate length for presentation by MHC class I molecules, this pathomechamism may also partly participate in the immune processes reported in mice by Kudva et al. [54]. This reasoning is supported by the finding that T. gondii infection caused downregulation of MHC class II molecules and inability to upregulate class I molecules in murine macrophages [56-58].

T. gondii

T. gondii tachyzoite division is composed of single G1, S, mitosis /cytokinesis phases with infectious daughters formed following each nuclear cycle [59,60]. Consequently, replicating parasites, which can divide 5 to 6 times in a single host cell, are continuously infective when mechanically liberated from host cells [59]. Gaji et al. [59] showed that tachyzoites preferentially egress and invade in the G1 phase of the parasite cell cycle, thus demonstrating functional coordination between the cell cycle and intercellular transmission. The parasite rapidly alters the expression of many mRNAs soon after cell invasion to intracellular replication [59].

Mack et al. [61] demonstrated a cause and effect relationship between human MHC genes and resistance to T. gondii infection and associated inflammatory processes. They found that in Caucasians, the DQ3 gene frequency was significantly higher in infected infants with hydrocephalus (0.783) than infected infants without hydrocephalus (0.444) or published controls (0.487). Consistent with the observed association between DQ3 and hydrocephalus in human infants, was the finding in the murine model that the DQ3 (DQ8; DQB1*0302) gene protected less than DQ1 (DQ6; DQB1*0601) [61].

Dubey et al. [62] studied pathogenesis of T. gondii oocysts in HLA transgenic mice infected with different doses of the parasite strains of different genotypes derived from several countries. It was found that the decreasing order of infectivity and pathogenicity was the following: mice C57BL/6 background IFN-γ gene knock out, HLA-A*1101, HLA-A*0201, HLA-B*0702, Swiss Webster, C57/black, and BALB/c. Mice fed as few as 1 oocyst of type I and several atypical strains died of acute toxoplasmosis within 21 days post inoculation, while some T. gondii type II, and III strains were less virulent [62]. In North America, T. gondii serotype II and NE-II caused congenital toxoplasmosis, and prematurity and severe disease at birth was related with the parasite NE-II serotype [63]. This serotype was also associated with rural residence, lower socioeconomic status and Hispanic ethnicity (p<0.01- 0.001) [63].

In summary, literature data indicate that there are considerable similarities of genetic contribution between chronic latent T. gondii infection, parasite strain affecting infectivity and pathogenicity, its developmental stage, dose, route of infection, innate immune state of the host, and local environmental factors, and development of diabetes mellitus.

Disturbances of Innate Immunity in Patients with Diabetes Important for Triggering Development of the Disease

Han et al. [64] showed an overall depressed immunity in longterm patients with T1DM, and markedly increased gene expression for IFN-γ, IL-4 and IL-10 mRNA levels in new-onset patients compared to at-risk and long-term types of T1DM patients.

Clinical investigations performed in diabetic patients and experimental studies in diabetic rats and mice showed defects of neutrophil chemotactic, phagocytic and microbicidal activities [65]. Also, there were many other abnormalities, including decreased microvascular response to inflammatory mediators, reduced protein leakage and edema formation, decreased mast cell degranulation, impairment of neutrophil adhesion to the endothelium and migration to the site of inflammation, production of ROS, reduced release of cytokines and prostaglandins by neutrophils, increased leukocyte apoptosis, and reduction in lymph node retention capacity [66]. Endothelial dysfunction appeared to precede the development of overt hyperglycemia in the patients with T2DM [66]. Metabolic routes by which hyperglycemia is linked to neutrophil dysfunction included the advanced protein glycosylation reaction, the polyol pathway, oxygen free radical formation, the NO-cyclic guanosine-3’-5’ monophosphate pathway, and the glycolytic and glutaminolytic pathways [65].

Diabetes causes marked changes in function and metabolism of neutrophils, e.g. glutamine oxidation and glutaminase activity are markedly decreased in neutrophils from diabetic rats [67] and glutamine plays an important role in protein (as an amino acid source), lipid (by NADPH production) and nucleotide synthesis (by purine and pyrimidine production), and in NADPH oxidase activity [68]. It must be emphasized that the tachyzoite stage of T. gondii, responsible for an acute infection, rapidly metabolizes glucose via glycolysis [69,70]. Blume et al. [70] demonstrated however that glucose is nonessential for T. gondii tachyzoites because host-derived glucose and its transporter in the parasite are dispensable by glutaminolysis. Thus, eventually increased requirements for glutamine and competition for this amino acid between T. gondii and neutrophils (and probably other cells) may result in diminished sources of glutamine and development of disturbances in maintaining regular metabolic and immune processes in many host cells. Moreover, this amino acid raises the in vitro bacterial killing activity and the rate of ROS generation by neutrophils [71,72], and delays spontaneous apoptosis of these cells [73].

Defective phagocytosis and decreased activity of intracellular killing of bacteria and Candida [74-76] (probably including also T. gondii) in neutrophils, are examples of the impaired mechanisms responsible for the increased susceptibility to infections in the patients with advanced stages of DM.

Rodacki et al. [77] found that the onset period of T1DM is marked by a slight reduction in blood NK cells, but in some patients the cells were unusually activated, as estimated by increased IFN-γ expression). NK cells patients with in long-standing T1DM had a markedly lower expression of p30/p46 NK-activating receptor molecules compared with control individuals, and this abnormality may be rather consequence than cause of the disease [77]. T1DM follows an immunologically mediated destruction of the pancreatic cells and autoreactive T-cells play a pivotal role in this process. NK cells are also potentially involved in this process, given their ability to kill target cells and interact with antigen-presenting cells and T-cells [78-80]. Indeed, NK cells can lyse islet cells in vitro [81,82]. Because NK cells are a major source of IFN-γ, their pathophysiological impact on the disease is to modulate the aggressiveness of the immune attack and the rate of its progression from insulitis to overt diabetes [77].

It was reported that pancreatic β-cell destructive insulitis was associated with increased expression of several proinflammatory cytokines (IL-1, TNF-α, TNF-β, INF-α, INF-γ, IL-2, and IL-12), whereas non-destructive (benign) insulitis was linked with the expression of antiinflammatory cytokines (IL-4 and IL-10) and TGF-β [83] (Table 6). Proinflammatory cytokines such as IL-1, TNF-α, TNF-β and IFN-γ may be directly cytotoxic to pancreatic β-cells by inducing NO and ROS in these cells. These data are in agreement with the findings that systemic administration of a wide variety of cytokines demonstrated to prevent development of IDDM in NOD mice and/or BB rats depending on the dose and frequency of administration [83].

  Proinflammatory Type 1 cytokines Type 2 cytokines Type 3
  cytokines     cytokine
  IL-1 TNF-α IFN-α IL-12 IFN-γ TNF-β IL-2 IL-4 IL-6 IL-10 TGF-β
NOD mice ++ ++ 0 ++ ++ ++ ++ + ++ + +
BB rats ++ ++ ++ ++ ++ ? ++ 0 ? + +
Humans 0 nd ++ ? ++ ? nd 0 0 ? ?
++: cytokine presence correlates with β-cell destructive insulitis; +: cytokine presence correlates with benign insulitis; 0: cytokine presence does not correlate with either destructive or benign insulitis; nd: not detected;
?: not reported; NOD: Non-Obese Diabetic mice; BB: Biobreeding rats; Humans: pancreas of humans with IDDM

Table 6: Correlations of cytokines expressed in islets with β-cell destructive or benign insulitis (according to Rabinovitch [83]; with own modification).

Lajoix et al. [84] demonstrated the presence of neuronal NO synthase (nNOS) in rat pancreatic islets and INS-1 cells. Electron microscopic study showed that nNOS was mainly localized in insulin secretory granules and to a lesser extent in the mirochondria and the nucleus. It appeared that β-cell nNOS exerted, like brain nNOS, two catalytic activities: a NO production and a NOS monooxidative reductase activity [84]. Kröncke et al. [85] found that NO is extremely toxic for islet cells and even in the absence of other macrophagegenerated potentially toxic products can rapidly and completely kill these cells.

Proinflammatory cytokines are increasingly thought to contribute to beta-cell dysfunction and death not only in T1DM, but also in the progression of T2DM. It was established that pancreatic beta cells, as well as neural cells, can be destroyed by several toxic agents and noxious stimuli, such as for example: 1) ROS (H2O2, O2 -, HO-) and NO, 2) cytokines (TNF, IL1-β, IFN-γ), 3) hyperglycemia and 4) islet amyloid polypeptide [86]. Cytokines can alter intracellular calcium levels by depleting calcium from the endoplasmic reticulum (ER) and by increasing calcium influx from the extracellular space [87]. Depleting ER calcium leads to protein misfolding and activation of the ER stress response. Disrupting intracellular calcium may also affect organelles, including the mitochondria and the nucleus, and as a chronic condition, cytokine-induced calcium disruption may lead to beta-cell death in both T1DM and T2DM [87]. Host cell mitochondria and ER have an intimate relationship with T. gondii due to their recruitment to and association with the parasitophorous vacuole membrane (Table 7) [88]. Moreover, discharge of adhesive proteins T. gondii apical storage organelles (micronemes) is stimulated by contact with host cells and this process is regulated by increases in intracellular calcium within the parasite [89]. In addition, gliding of the parasite is controlled by secretion of microneme proteins and factors that alter calcium fluctuation in the cytosol, while chelation of intracellular calcium blocked parasite motility [90].

Sample treatment Sample size
(n)
SvPVM
Mean (SE)
SvM
Mean (SE)
SvER
Mean (SE)
Percentage
PVM-mitochondrial association
Percentage
PVM-ER association
Untreated,
4 hrs
31 3.20 (0.16) 0.58 (0.07) 1.78 (0.22) 18 56
Nocodazole,
4 hrs
31 3.70 (0.17) 0.33 (0.08) 1.79 (0.15) 9 47
Untreated,
20 hrs
30 2.70 (0.19) 0.61 (0.06) 0.80 (0.13) 23 30
Nocodazole,
20 hrs
30 3.22 (0.19) 0.81 (0.09) 1.21 (0.20) 25 38
Pyrimethamine
20 hrs
30 2.96 (0.14) 0.79 (0.09) 1.00 (0.14) 27 34
ER: Endoplasmic Reticulum. PVM: Parasitophorous Vacuole Membrane. The extent of PVM-organelle association was measured at 4 hrs and 20 hrs post-infection. The specific parameters measured are the Surface to Volume ratios (Sv) of the PVM (SvPVM), PVM-associated mitochondria (SvM) and PVM-associated Endoplasmic Reticulum (SvER), all relative to the volume of the PV. The mean surface densities and the Standard Error (SE) for the sample size measured are presented. The extent of PVM-mitochondrial and PVM-ER association was represented as a percentage of their surface densities (SvM and SvER) relative to SvPVM. The values for the mean surface densities and the percentage association were rounded off to the level of significance indicated

Table 7: Morphometric analysis of T. gondii PVM-host organelle association (acc. to Sinai et al. [88]; with own modification).

Experimental studies indicated that disruption of endothelial insulin signaling through the activity of protein kinase C-β and NFκB reduces NO availability, and Tabit et al. [91] observed 1.7- fold higher basal eNOS phosphorylation at serine 1177 in patients with diabetes (p=0.007). Nitrotyrosine levels were higher in diabetic patients indicating endothelial oxidative stress, and protein kinase C-β expression was higher in those patients and was associated with lower flow-mediated dilation (r=-0.541, p=0.02) [91]. In T. gondii, calcium-dependent protein kinases (serine/threonine kinases) are key mediators of signaling [92], and a protein kinase C receptor 1 was identified in tachyzoites [93]. Proliferation of the parasite is dependent on its ability to invade host cells, which is mediated partly by calciumdependent protein kinase 1 [94]. cAMP-dependent protein kinase plays an important role in the growth of tachyzoites [95], and a plantlike calcium-dependent protein kinase in T. gondii is required for optimal in vitro growth, regulates microneme secretion when parasites are intracellular and its egress from host cells [96].

Leem and Koh [97] suggested that impaired mitochondrial function and ER stress are closely associated with pancreatic β cell dysfunction and peripheral insulin resistance, and each of these factors contributes to the development of T2DM [98-103]. ROS generation is thought to act as local messengers between the ER and mitochondria [103] and many ROS sources and targets are localized to the ER and mitochondria [104]. NO signals the ER stress response via inhibition of mitochondrial respiration because in NO-generating cells the respiratory chain is disrupted [105] and NO can bind to cytochrome c oxidase and inhibit the enzyme, in competition with oxygen [106]. In addition, NO mediates cytokine-induced (IL-1β, TNF-α, and IFN-γ) inhibition of insulin secretion by human islets of Langerhans through generation of iron-nitrosyl complexes that inactivate enzymes, such as aconitase and ribonucleotide reductase [107-109], while on the other hand, IL-1β and TNF induce NO formation and accumulation of cyclic GMP in pancreatic β cells [110,111].

Weaver et al. [112] showed that stimulation of human donor islets with a cocktail of inflammatory cytokines (TNF-α, IL-1β, and IFN-γ) significantly induced NADPH oxidase-1 (NOX-1) gene expression (p<0.05), and concomitantly induced loss of islet glucose stimulated insulin response (p<0.05), elevated expression of MCP-1 (p<0.01), increased cellular ROS production, and induced cell death [112]. Recently, the role of NOX in mitochondrial dysregulation in diabetes was reported [113], and NOX-1 participated in ROS-dependent cell death of Caco2 cells [114].

T1DM results from the destruction of insulin-producing pancreatic beta cells by a beta cell-specific autoimmune process leading to absolute insulin deficiency. Beta cell autoantigens, macrophages, dendritic cells (DC), B lymphocytes, and T lymphocytes have been found to be involved in the pathogenesis of autoimmune diabetes [115-117]. Beta cell autoantigens are thought to be released by cellular turnover or damage, then processed, and finally presented to T helper cells by antigen-presenting cells. Macrophages and DC are the first cell types to infiltrate the pancreatic islets. Naive CD4+ T cells can be activated by IL-12, a proinflammatory cytokine, released from macrophages and DC. The CD4+ T cells secrete IFN-γ and IL-2, and IFN-γ activates other resting macrophages, which release IL-1β, TNF-α, and free radicals, all toxic to pancreatic beta cells if produced in excess [118]. Beta cell antigen-specific CD8+ T cells activated by IL-2 produced by the activated TH1 CD4+ T cells differentiate into cytotoxic T cells and are recruited into the pancreatic islets, finally leading to the destruction of beta cells [115-117]. In addition, beta cells can also be damaged by granzymes and perforin released from CD8+ cytotoxic T cells, and by cytokines and reactive oxygen/nitrogen species generated by activated macrophages accumulated in the islets. Thus, activated macrophages, TH1 CD4+ T cells, and beta cell-cytotoxic CD8+ T cells act synergistically to destroy beta cells, resulting in the development of autoimmune T1DM [115-117].

Abscisic acid (ABA) is a membrane-permeant hormone rapidly produced and released from human islets stimulated with high glucose concentrations, which regulates several important physiologic functions related to stress [119]. ABA acts as an endogenous proinflammatory cytokine in human granulocytes because it activates many functions of these cells including phagocytosis, migration, production of ROS and NO [119,120]. Nanomolar ABA concentrations increase glucosestimulated insulin secretion from human and murine pancreatic β cells, and the paracrine production of the hormone by activated granulocytes and monocytes suggests that ABA may also be involved in dysregulation of insulin release during inflammation of pancreatic islets [119,120].

ABA has been detected as a product of human granulocytes [120,121], pancreatic islet cells [119], and other cell types [122]. Nagamune et al. [121] demonstrated production of ABA also by T. gondii. ABA induced the release of intracellular calcium stores via the generation of cAMP ribose [121], controlled calcium signaling within the parasite, and calcium was responsible for several critical bioevents related to T. gondii, including its motility, secretion, cell invasion, and egress. It appeared that ABA-mediated calcium signaling controls the decision between lytic and chronic stage growth, a developmental switch that is crucial in pathogenicity and transmission of the parasite [121]. Parasite- or host cell-derived ABA represents a potential initiating agent of calcium mediated host cell autophagy [123]. The above-presented processes, with possible involvement of T. gondii infection, may be at least in part responsible for the increased β-cell apoptosis and deficit of β cells development found in pancreatic tissue from 124 autopsies in humans with T2DM and in mouse model of type 2 diabetes [124,125].

Pathologic or radiologic evidence of pancreatitis was noted with T. gondii, especially in HIV-infected patients or in other immunocompromised hosts. The majority of these individuals had no clinical symptoms of pancreatitis and infection was demonstrated histologically during postmortem examination [126].

Immunomodulatory Effects of Latent Toxoplasmosis in Animals and Humans

Animals

Kankova et al. [127] showed that mice in the early phase of latent T. gondii infection had transiently increased serum IL-12 levels and decreased generation of IL-10. There was also a decreased production of NO by stimulated macrophages, diminished generation of IL-2 and IL-4, and a markedly lower proliferative activity of splenocytes compared with controls in the early and also in the late phases of the infection, which suggested that immunosuppression processes play an important role during latent toxoplasmosis [127].

Tachyzoites of T. gondii stimulate production of IL-12 [128-131] and this proinflammatory cytokine activates NK cells and T cells to produce IFN-γ that is crucial for host resistance [132-136]. IFN-γ and TNF-α act synergistically to mediate killing of tachyzoites by macrophages and the combination of these two cytokines results in a greatly enhanced production of free oxygen radicals and NO, both of which can affect parasite killing [128,136-138], although NO and its metabolites appear to be the primary effectors. NO is produced as a result of iNOS activation, which is dependent on activation of NFκB [139]. Gomez-Marin [140] obtained evidence of NO generation not only in the host cells, but also in T. gondii [141,142], which has its own cNOS producing 2-6 μmol of nitrites that could be essential in intracellular signaling. The NO defensive mechanism, where levels of nitrites can reach 120 μmol or more, is probably toxic for human and mice tissues [140].

T. gondii induces overproduction of IFN-γ and other proinflammatory cytokines which may contribute to host tissue injury and death [143,144]. IFN-γ-induced antitoxoplasmatic activity is mediated by NO and indeed, induction of iNOS was demonstrated during the parasite replication in murine macrophages [145]. IFN-γ combined with TNF-α activated macrophages to produce increased quantities of RNI that are involved in the control of parasite replication [138] (Table 8).

Cytokines Sources Functions
IL-12 Dendritic cells [147-149] Promotes T cell proliferation and differentiation [134,150]
  Neutrophils [151-153] Promotes NK cell responses [129,154]
  Inflammatory monocytes [153,155] Promotes IFN-γ production [122,149]
IFN-γ NK cells [137,145,154] Promotes iNOS expression [158]
  CD4+ T cells [159] Promotes p47 GTP-ase-mediated killing of T. gondii [160,161]
  CD8+ T cells [159] Promotes tryptophan degradation [135,162-164]
TNF-α Neutrophils [151,165] Promotes macrophage activation [166]
  Dendritic cells [165] Promotes control of parasite in non-hematopoietic cells [167]
  Macrophages [168]  
  Microglia [169] Promotes iNOS expression [138,170-172]
  T cells [173]  
IL-6 Monocytes [174] Necessary for optimal neutrophil responses [175]
  Astroglia [176] Necessary for optimal IFN-γ responses [175]
  Stromal cells [177]  
  Retinal pigment epithelial cells [178]  
LT-α Lymphocytes [179] Necessary for normal secondary lymphoid architecture [180]
    Necessary for optimal antibody and IFN-γ responses early during infection [138]
    Necessary for optimal expression of iNOS [170]
IL-10 NK cells [181] Inhibits CD4+ T cell-mediated pathology [182]
  Macrophages [183]  
  CD4+ T cells [184]  
  CD8+ T cells [183]  
IL-27 Antigen-presenting cells [185] Inhibits IL-17 production [186]
    Inhibits IL-2 production [187]
    Promotes IL-10 production [188]
    Promotes PD-L1 expression [189]
CD40L
(surface protein)
Expressed on T cells [190] Promotes TH1 responses in humans [191]
    Promotes iNOS expression [192]
    Promotes xenophagic killing of T. gondii [193]
Numbers in parentheses denote reference nos

Table 8: Cytokines necessary for survival during T. gondii infection (according to Dupont et al. [146]; with own modification).

Seabra et al. [194] found that NO was produced by monocytederived macrophages only if cultured in the presence of macrophagecolony- stimulating factor. Monocyte-derived or peritoneal macrophages infected with T. gondii presented lower iNOS expression, had a marked reduction in NO production, and only viable parasites caused partial inhibition of this process [194].

Lüder et al. [195] showed that infection of primary bone marrowderived macrophages or monocyte/macrophage RAW264.7 cells with a mouse-avirulent T. gondii strain markedly decreased NO production that had been induced by activation with either IFN-γ or bacterial lipopolysaccharide (LPS), or IFN-γ plus LPS. The down-regulation of NO production by the parasite enabled its considerable replication in macrophages activated with IFN-γ or LPS alone. iNOS transcripts induced by IFN-γ alone or in combination with LPS were also dosedependently down-regulated after infection of RAW264.7 cells with T. gondii [195].

Infection of mice with T. gondii elicits a dominant TH1 cytokine response involving IFN-γ, IL-12, IL-1β, and TNF-α. TNF-α induction has a serious impact on the parasite-induced pathology at early stages of infection. TH2-associated cytokines, such as IL-4 and IL-10, appear relatively late after infection, and may limit immune pathology [196]. To resolve acute infection, IFN-γ induces indoleamine 2,3 dioxygenase (IDO) release, resulting in tryptophan degradation and kynurenic acid accumulation [197]. IDO activated T cells and blocked their conversion into TH17-like T cells [198]. Tryptophan depletion is thought to be responsible for suppression of the growth of the acute stage tachyzoites. Kynurenic acid accumulation in the brain could potentially alter dopamine metabolism due to its NMDA antagonistic property [199-201] (Figures 1-3).

diabetes-metabolism-tryptophan-metabolism

Figure 1: Various pathways of the essential amino acid tryptophan metabolism. About 99% of the dietary tryptophan is metabolized along the kynurenine pathway (red arrows). Alternative pathways are the conversion of tryptophan to 5-hydroxytryptamine (5-HT) and then to melatonin, or to tryptamine and then to the kynuramines (or kynurenamines). N1-acetyl-5-methoxykynuramine is a metabolite deriving from melatonin by mechanisms involving free radicals, exhibits potent antioxidant properties exceeding those of its direct precursor N1-acetyl-N2-formyl-5-methoxykynuramine, a biogenic amine generated through either an enzymatic or a chemical reaction (free radicals) pathway. 3-HAO, 3-hydroxyanthranilate oxidase; IDO, indoleamine 2,3-dioxygenase; KAT, Kynurenine aminotransferase; MAO, monoamine oxidase; QPRT, quinolinic-acid phosphoribosyl transferase; TDO, tryptophan 2,3-dioxygenase [200].

diabetes-metabolism-indoleamine

Figure 2: Interrelationships between indoleamine 2,3-dioxygenase (IDO) and nitric oxide synthase (NOS) in macrophages or glial cells, and the potential interactions with neurons by means of N-methyl-D-aspartate (NMDA)-receptor-induced nitric-oxide (NO) formation. Arg, arginine; 3-HAA, 3-hydroxyanthranilic acid; 3-HK, 3-hydroxykynurenine; IFN-γ, interferon-γ; IL, interleukin; Kyn, kynurenine; KynA, kynurenic acid; LPS, lipopolysaccharide; mRNA, messenger RNA; iNOS, inducible nitric-oxide synthase; TGF-β, transforming growth factor-β; TNF-α, tumor necrosis factor-α; Trp, tryptophan; xA, xanthurenic acid. The broken lines represent possible reactions [200].

diabetes-metabolism-mononuclear-phagocytes

Figure 3: Possible model for NO-mediated regulation of IDO in IFN-γ-primed mononuclear phagocytes. NOS, nitric-oxide synthase; IDO, indoleamine 2,3-dioxygensae, L-Arg, L-arginine; L-Trp, L-tryptophan; IFN-γ, interferon-; NO, nitric oxide; Kyn, kynurenine; 3-HAA, 3-hydroxyanthranilic acid; QA, quinolinic acid; SNP, sodium nitroprusside; GTN, glyceryl trinitrate; SNAP, S-nitroso-N-acetylpenicillamine; DEANO, diethylaminodinitric oxide. SNP, DEANO, and SNAP release NO extracellularly, while GTN is thought to release NO intracellularly [201].

In mice, T. gondii infection caused a significantly increased formation of RNI probably due to elevated serum NO concentrations [143], and a significantly higher serum kynurenine/tryptophan ratio compared with control animals (p<0.05). The authors suggested that increased free radical toxicity may cause elevation in tissue malondialdehyde (MDA) levels arising from lipid peroxidation in T. gondii-infected mice, while unchanged serum MDA concentrations might indicate the increased oxidative stress due to the parasite infection restricted to intracellular area [143].

Wang et al. [123] presented evidence that T. gondii induces host cell autophagy in both HeLa cells and primary fibroblasts by a mechanism dependent on calcium, and that it exploits the nutritive function of host autophagy to enhance its proliferation. Autophagy is a lysosomal selfdigestion process essential for cellular homeostasis, differentiation and survival, which protects organisms against a wide range of pathologies, including infection, neurodegeneration, ageing. (Nb. autophagy is considered a basis for the health promoting effects of vitamin D because of at least two functions, the induction of cancer cell death and the clearance of Mycobacterium tuberculosis in macrophages [202]). Autophagy-dependent T. gondii growth correlates with autophagydependent consumption of host cell mass that is dependent on parasite consumption. In macrophages infected with the parasite, fusion of the parasitophorous vacuole with lysosomes can be induced in an autophagy-dependent manner when host cell anti-parasitic function is activated via CD40 [203]. Parasite-induced autophagy is dependent on calcium sigaling and on abscisic acid. Autophagy as a component of host defense may be upregulated by proinflammatory factors, such as LPS [204] and IFN-γ [205]. The parasite is able to actively sequester host cell lysosome-derived vesicles, thereby potentially gaining access to their content [206]. T. gondii reside in host cell vacuoles which resist typical phagosome-lysosome fusion because phagosome acidification necessary for microbicidal effect is blocked by intracellular parasite [207]. Extracellular Toxoplasma is highly susceptible to acidic pH conditions, indicating that the acidification block is important for intracellular survival of the parasite [207]. Thus, it seems that hypoglycemia-induced neuronal death, increased superoxide production and microglia activation reported in patients with diabetes, and is not a simple result of glucose deprivation, but instead the end result of a multifactorial process [208]. These changes may at least in part reflect a defense reaction of the host against chronic latent T. gondii infection through increasing systemic and local acidification of the tissues.

Humans

Flegr and Striz [22] found that in 128 analyzed male patients the prevalence of T. gondii infection was 10.9% which contrasted with 23.7% in 312 female patients and 20-30% in general population reported in Prague (Czech Republic). The male patients with latent T. gondii infection had significantly decreased leukocyte, NK-cell and monocyte counts while the T. gondii-positive women had increased these values, as compared with controls. The B-cell counts were markedly reduced in both men and women with toxoplasmosis [22]. Karaman et al. [209] found a significantly higher serum NO levels in patients with latent toxoplasmosis as compared with seronegative controls, and Dzitko et al. [210] demonstrated markedly increased serum prolactin (a strong immunomodulator) levels in women with latent T. gondii infection. Prolactin has been show to enhance production of IFN-γ, IL-12, and IL-10, but not of TNF-α, in a stimulus specific manner [211]. In vitro preincubation of tachyzoites with recombinant human prolactin resulted in a significant reduction (up to 36.15%) in replication abilities of the parasite, and the inhibition of replication was caused by a limited capacity of the parasites to penetrate host’s cells as demonstrated by the reduced number of infected cells [212]. More detailed effects of chronic T. gondii infection concerning modulation of human innate immunity and metabolism were presented in other works [213-216]. Tables 9 and 10 summarized host cell-mediated evasion strategy against infection with the parasite.

Evasion strategy Consequence(s) Molecular mechanism(s) Parasite effect References
Induction of IL-10 Decreased TH1 response;
Deactivation of macrophages
Independent of PGE2 Indirect Indirect [218,219]
Induction of TGF-β Reduced TNF-α production by macrophages;
reduced IFN-γ production by NK cells
Antagonizes IL-12 Indirect [130,220,221]
IFN-γ/-β upregulation Reduced IFN-γ levels and splenocyte proliferation   Indirect [222]
Inhibition of TNF- α
and IL-12 production
Deactivation of macrophages;
inhibition of TH1 responses
Reduced phosphorylation of p65/RelA; defective nuclear import of NF-kB; IL-10-independent STAT3 phosphorylation Direct [15,223-226]
Decreased IL-12 production by DCs Inhibition of TH1 responses LXA4-mediated downregulation
of CCR5
Indirect [227,228]
Blockade of MHC
class II upregulation
Defective antigen presentation to CD4+ T cells Reduced activity of CIITA and IRF-1 promoters Direct [56,229]
Inhibition of NO production Defective antiparasitic activity Inhibition of iNOS transcription Direct [194,195]
Inhibition of NO production in microglia Reduced antiparasitic activity Secretion of PGE2,
IL-10 and TGF-β
Indirect [230,231]
Inhibition of p47 GTPases   Reduced transcription   [232]
Significant suppression of IL-2, IFN-γ (but not IL-10). Markedly lower levels of IgG1, IgG2a, IgG2b, IgG3, IgA, IgM Suppressed cytokine and immunoglobulin secretions by murine splenic lymphocytes   Indirect [21]
CIITA: Master Regulator of Major Histocompatibility Complex Class II Transcription; CCR5: CC Chemokine Receptor; DCs: Dendritic Cells; iNOS: Inducible Nitric Oxide Synthase; IRF-1: Interferon Regulatory Factor-1; LXA: Lipoxin A4; MHC: Major Histocompatibility Complex Molecules; PGE2: Prostaglandin E2; TGF-β: Transforming Growth Factor-β. Proliferation of T . gondii in inflammatory macrophages was associated with diminished ROS production in host cells [233]. In young children with congenital toxoplasmosis specific T cell response to the parasite antigens was impaired and such hyporesponsiveness has been restored during childhood. The acquisition of functional T cell response was disease-unrelated and indistinguishable
in terms of strength, epitope specificity, and cytokine profile from the corresponding responses in immunocompetent adults with asymptomatic acquired T. gondii infection [234].
In pregnant mice, T. gondii infection caused a decrease of CD4+CD25+-regulatory T cells [235]. It must be noted that peripheral blood leukocytes (PBL) from healthy children older than 36 mths responded to several stimuli at levels comparable to those of PBL from adults, but surprisingly, cord blood leukocytes appeared to be more efficient in antigen-presenting function than PBL from children younger than 13 months [236].

Table 9: Partial downregulation of cell-mediated immune responses after infection with T. gondii (Lang, Gro & Lüder [217]; with own modification).

Evasion strategy Consequence(s) Molecular mechanism(s) Parasite effect References
Apoptosis of CD4+ cells T-cell unresponsiveness Cell death by neglect Indirect [237]
Apoptosis of leukocytesa Unrestricted parasite replication and host death Upregulation of Fas and FasL;
TNF-dependent mechanisms
Indirect [238-240]
Inhibition of apoptosis in parasite-positive cells Blockade of host cell suicide; avoidance of CTL- and NK-mediated cytotoxicity Inhibition of cytochrome c-release; upregulation of anti-apoptotic molecules
Interference with caspase activation; degradation of PARP (?)
Direct [241-245]      
[246]
CTL: Cytotoxic T lymphocyte; Fas: Receptor; FasL: Fas Ligand (a cell surface molecule belonging to TNF family and death factor, which binds to its receptor Fas, thus inducing apoptosis of Fas-bearing cells); NK: Natural Killer cells; PARP, Poly(ADP-Ribose) Polymerase. aT. gondii delayed neutrophil apoptosis by inducing granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor secretion by the parasite-infected human fibroblasts. Although neutrophils are unable to kill T. gondii, this can retard their division time from the usual 6-8 hrs cycle to a 24 hrs cycle and this enhanced neutrophil survival may contribute to the robust proinflammatory response elicited in the pathogen-infected host cells [247].

Table 10:Suppression of immune responses to T. gondii by parasite-triggered modulation of host cell apoptosis (acc. to Lang, Gro β & Lüder [231]; with own modification).

In the sera of 37 IgG-seropositive patients with T. gondii infection, Karaman et al. [209] demonstrated significantly increased malondialadehyde (MDA) and NO concentrations, and a decrease in glutathione activity as compared with healthy controls. It was also found a markedly higher MDA levels (p<0.001) paralleled with significantly decreased concentrations of glutathione peroxidase (p<0.0188) and tocopherol fractions (alpha, gamma and lambda) (p<0.001) in T. gondii seropositive than in seronegative blood donors [248,249]. These significant alterations in redox status between the two groups of blood donors indicate that chronic T. gondii infection is associated with oxidative stress because MDA is arising from the lipid peroxidation and is an indicator of oxidative stress, glutathione defends cells against oxidative damage by ROS and peroxidase, and tocopherol is an antioxidant [249,250]. The increased NO concentrations can be associated with the stimulation of the cell-mediated immune system in these individuals reflecting a defense of the host against the infection with the parasite. This may be supported by the finding that NO is a major effector molecule of macrophage cytotoxicity against T. gondii, and its production by macrophages is catalyzed by a cytokine-inducible form of the NO synthase positively controlled by TNF-α, IFN-γ and IL-2 [178]. Other cell types, such as endothelial cells and hepatocytes, display a similar capacity for NO generation in response to cytokine stimulation [251].

Baltaci and Mogulkoc [252] reported that in rats T. gondii infection caused also a significant increase of plasma leptin concentrations (p<0.01), with no change in body weight of the animals. In the obese individuals, including diabetics, this biomarker exerts a proinflammatory activity, and its structure is similar to that of IL-2, an important proinflammatory cytokine (Table 11).

Parameters Obese children Controls p
Leptin (ng/ml)a 19.9 ± 7.4 7.9 ± 5.1 <0.001
IL-1β (pg/ml) 33 ± 8.9 3.6 ± 1 <0.001
IL-2 (U/l) 0.4 ± 0.1 0.9 ± 0.1 <0.01
IL-6 (pg/ml) 45.2 ± 11.8 13.1 ± 3.9 <0.001
TNF-α (pg/ml) 9.2 ± 2.3 3.9 ± 1 <0.001
E-selectine (ng/ml) 78 ± 38 59 ± 29 <0.01
hsCRP (mg/l) 4.1 ± 4.8 0.9 ± 1.5 <0.001
Results are mean ± SD; CRP: C-Reactive Protein; hs: high-sensitivity; aLeptin has the structure similar to that of IL-2 and may activate the innate immune system and shift the cognate immune system toward a predominance of a proinflammatory TH1 T cell population while reducing the regulatory TH2 phenotype [255]. Leptin treatment was found to increase energy expenditure (oxygen consumption),as well as increased thermogenic marker uncoupling protein-1 and type II deiodinase mRNA levels 1.7- and 3-fold,respectively,in mice [256].

Table 11: Serum proinflammatory cytokines and leptin concentrations in obese children at prepubertal age compared with healthy children of the same age (according to Aygun et al. [253] and Kapiotis et al. [254]; with own modification).

Maternal Microchimerism (Mmc) in the Circulation and Tissues of Mothers’ Immune-Competent Children with T1DM. Pancreatic Islet β Cell Microchimerism may Play an Important Role in Development of T1DM because of Possible Intracellular Transmission of T. gondii Infection as a Trojan Horse to Mother’s Progeny

Pregnant women infected with T. gondii for the first time can transmit the infection to their fetuses across the placenta. The risk of congenital infection is lowest (10-25%) when acute maternal toxoplasmosis occurs during the first trimester and highest (60-90%) when it occurs during the third trimester [257]. In primarily infected pregnant woman invasion of the placenta by tachyzoites that then multiply within placenta cells, may cross the placenta, and enter the fetal gastrointestinal tract with amniotic fluid, invade circulation and/ or fetal tissue [258]. Passage of erythrocytes between mother and fetus is a well-documented phenomenon and it was reported that maternal erythrocytes have been found in 10-80% of all newborns [259]. Also, materno-fetal passage of leukocytes and platelets has been demonstrated [260].

Investigations of MMc in subsets of whole blood from 31 healthy adult women showed that 39% (12/31) of probands had MMc in at least one cellular subset [261]. MMc was demonstrated in T lymphocytes in 25% and B lymphocytes in 14%, while monocyte/macrophages had MMc in 16% and NK cells in 28%, thus demonstrating that MMc cells are detectable in the peripherial blood of many healthy individuals [261,262].

Diabetics

Zhou et al. [263] demonstrated that two independent pathways of maternal cell transmission to offspring, i.e. transplacental passage during pregnancy and breast milk-feeding after birth.

Bidirectional trafficking of maternal and fetal cells occurs during almost all pregnancies resulting in the persistence of low levels of these cells in the mother and/or her offspring for several decades after delivery [264-267]. The cell types exchanged between mother and fetus include leukocytes and T cells [268-271] in addition to progenitors of different line-ages [267,272], such as hematopoietic [267] or mesenchymal stem cells [273] and/or endothelial progenitors [274]. It was suggested that maternal T and potentially B cells transferred during pregnancy or lactation might play a role in the development of T1DM [275].

Roy et al. [275] believe that fetal microchimeric cells did not seem to influence the level of islet inflammation in mothers despite their antibeta cell specificity. They showed that fetal lymphoid progenitor cells enter the maternal thymus and develop into double positive and single positive thymocyte [270]. A possible role of microchimerism in the pathogenesis of some autoimmune diseases has been suggested because microchimeric cells may differentiate into many lineages in various tissues finally inducing diverse pathophysiologic processes during the host lifetime [276] (Table 4). For example, proliferation of these cells in maternal tissue environment may be at least in part responsible for development of autoimmune thyroid diseases because of a significant prevalence of anti-IgG T. gondii antibodies present in those individuals [25,277,278], as well as papillary thyroid cancer induction [279]. Fetal microchimerism may take place during pregnancy starting from the 4th to 6th week of gestation [280]. This traffic of cells is primarily composed by immune cells, such as T and B-lymphocytes, monocytes, and NK cells, including hematopoietic stem cells CD34+ and CD34+/38+ committed to early B and T-cells with the capacity for multilineage differentiation [281]. The number of fetal progenitor cells circulating in the blood of pregnant women, has been estimated to be 0-2/mL, but in normal second-trimester pregnancies may vary from 1 to 6 cells per ml of maternal venous blood [280]. At 36th week of gestation, 100% of pregnant women have fetal cells in their circulation and after delivery and 30-50% of healthy women have detectable fetal cells in their blood from 4 weeks to decades after delivery [279,282] (Table 12). Interestingly, in mice maternal background strain and strain differences between the mother and father significantly affected fetalmaternal trafficking (both the number of fetal cells and the relative distribution of cell types in maternal organs) more than maternal immune competence [284].

Extravillous cytotrophoblasts
Nucleated erythroblasts
Platelets
Mesenchymal stem cells
CD34+ hematopoietic stem/progenitor cells
CD34+ and CD38+ lymphoid progenitors
CD19+ and IgM+ B lymphocyte precursor cells
CD8+ T cells
CD4+, CD25high and FOXP3+ regulatory T-cells
CD45+ leukocytes
CD3+ and CD14+ mononuclear cells
CD56+ and CD16+ natural killer cells
Maternal cells [263] and soluble maternal HLA are also transferred in breast milk [283]

Table 12: Different types of cells involved in fetal-maternal trafficking (according to Klonisch and Drouin [32]; with own modification).

Chimeric maternal cells (e.g. hematopoietic cells) have been found in human fetal [285] (including a second trimester fetus [286]), newborn [287] and infant tissues [288]. It must be emphasized that T1DM has been mostly related to maternal cell microchimerism [289]. Nelson et al. [30] reported markedly higher levels of maternal microchimeric cells in the peripheral blood of 94 patients with T1DM, ranging from 3 to 28 years of age, than 54 unaffected siblings as well as 24 unrelated healthy individuals studied.

Recently, patients with juvenile diabetes were found to have increased levels of maternal cell microchimerism when compared to their unaffected siblings. Nelson et al. [30] reported that maternal microchimerism (MMc) levels, expressed as the genome equivalent per 100,000 proband cells, were markedly higher in T1DM patients’ circulation than unaffected siblings and healthy individuals. In the patients with the T1DM-associated DQB1*0302-DRB1*04 haplotype, MMc was found more often when the haplotype was paternally (70%) rather than maternally transmitted (14%). Female islet β cells (presumed maternal) formed 0.39-0.96% of the total, whereas female hematopoietic cells were very rare [30]. There are also strong suggestions of a prominent role of the maternal perinatal environment in the predisposition to juvenile diabetes among the offspring [290,291] (Tables 12 and 13). Fetal DNA in maternal organs also has been detected in animals with streptozocin-induced diabetes [292].

Group of animals Days of pregnancy Pancreas Liver Kidney
Controls 6 ND ND ND
  13 ND ND ND
  19 ND ND ND
Diabetics 6 64.2 ± 6.8 9.7 ± 0.8 13.5 ± 3.2
  13 45.4 ± 5.1 13.3 ± 1.2 18.8 ± 3.5
  19 49.8 ± 7.5 10.7 ± 1.4 10.1 ± 2.9
Data are expressed as mean ± SE. Results represent the amount of fetal DNA per 1 x 105 maternal (total) genome equivalents

Table 13: Fetal DNA in maternal organs at day 30 after streptozotocin-induced diabetes (200 mg/kg for 2 days) in mice (according to Sunami et al. [292]; with own modification).

In humans, fetal cells have been detected for decades in the maternal blood and tissues affected by both autoimmune and nonautoimmune diseases, and these cells had morphological features of both hematopoietic and non-hematopietic tissue lineages cells [293-295]. In mice, 30 days after STZ-induced diabetes fetal cells were detected in the maternal bone marrow, pancreas, liver and kidney (Table 13). Histological examination showed differentiated fetal cells within the pancreatic acinar cells, hepatocytes and tubular epithelial cells, and their morphological appearance was undistinguishable from their maternal counterparts and constant over the long term after delivery [293].

In pancreas samples obtained from 6 males with T1DM Vanzyl et al. [296] found that the frequency of MMc was significantly higher (range 0.32-0.80%, mean 0.58%) than in 4 male controls (0.24-0.50%, mean 0.38%; p=0.05). Interestingly, clusters of 2-3 MMc were occasionally found in the pancreases, particularly T1DM pancreases, suggesting replication of these cells [296]. In all pancreas samples analyzed MMc were identified within islets that appear to be insulin positive but noninsulin staining MMc were also observed in the exocrine tissue [296]. In a separate analysis of pancreatic autopsy material from 4 males with T1DM they demonstrated that female cells in the pancreases of these male individuals were often positive for insulin. It was suggested that maternal cells could contribute to endocrine function in offspring [30,297]. It appeared that in all pancreases analyzed, female (presumed maternal) cells have been identified and immunochemistry showed insulin-producing XX beta cells in males. VanZyl and Gillespie [262] suggested that some cells that transfer from mother to child must therefore be adult stem cells with the capacity to differentiate into islet beta cells. Because these cells are 50% genetically distinct from the host, it could be a trigger for autoimmune attack, but so far there is no evidence to support this hypothesis [262]. On the other hand, this idea may however be in agreement with the fact that T. gondii is transmitted as a Trojan horse in many hematopietic cells, and therefore these cells could be the focus of lymphocytic attack and selectively destroyed in T1DM pancreas. This reasoning may be consistent with the findings that islets in T1DM pancreas are often present in a lobular pattern, with islets completely intact on one lobe and totally destroyed on an adjacent lobe [259,260].

T. gondii infection

T. gondii can actively infect any nucleated cell type, including cells from the immune system. Persson et al. [298] observed that a large number of NK cells have been infected by the parasite early after intraperitoneal inoculation of T. gondii into C57BL/6 mice. It appeared that one mechanism of NK cell infection involved NK-cell-mediated targeting of infected dendritic cells (DC), and infected NK cells were not efficiently targeted by other NK cells. It was suggested that rapid transfer of the parasite from infected DC to effector NK cells may contribute to sequestration of T. gondii and shielding from immune recognition shortly after infection [298]. It must be noted that NK cells do not posses intracellular killing pathway, such as for example NO, therefore NK cell infection may provide a niche in which the parasites proliferate and promote its persistence in a less hostile environment [298]. Table 14 presented in vitro differences in division rates of intracellular T. gondii tachyzoites in various types of cells [300-309].

Cell type Parasite division rate   Mechanism References
  Unprimed IFN-γ primed    
Hemopoietic        
Lymphocyte S     [285]
Neutrophil S     [285-287]
Adherent monocyte S     [287-290]
Nonadherent monocyte R R ROS; not TS [285,291]
Dendritic cell R     [285]
Alveolar macrophage R S Partly TS [155]
Peritoneal macrophage R S   [292]
Monocyte-derived macrophage R S ROS; not RNI [155,156,157,288,290,292,293]
Nonhematopietic        
Neuron S     [294]
Foreskin fibroblast R S TS [154,295]
Umbilical vein endothelial cell R S TS or ROS; not RNI [290,296]
Retinal pigment epithelal cell R S TS [297]
Fetal astrocyte R S RNI [294,298]
Fetal microglial cell R R   [299]
R: Rapid; S: Slow; RNI: Reactive Nitrogen Intermediates; ROS: Reactive Oxygen Species; TS: Tryptophan Starvation

Table 14: Division rate of intracellular T. gondii tachyzoites in primary human cells in vitro (according to Channon et al. [299]; with own modification).

The risk of infection with the parasite is 0.1% to 1% of all pregnancies [314]. All eukaryotic cells may function as systemic parasite transporters. Maternal microchimerism was found to be more common than fetal microchimerism (40% vs. 15%, p=0.05) [315]. In the mouse model, the effect of maternofetal transmission of T. gondii after oral infection was measured by the mortality rate in the mother, the fetus and the neonate [316]. When the infection preceded the mating, the percentage of neonates who died ranged from to 52% to 74%. In contrast, when the mating preceded the infection, these percentages were much more elevated ranging from 74% to 96% [316]. Weight and Carding [10] showed that T. gondii upregulated intercellular junction-associated proteins, such as intercellular adhesion molecule 1 (ICAM-1), in MDCK II and BeWo cells [317]. The parasite influenced the cellular distribution of occludin to transmigrate the intestinal epithelium. ICAM-1 is expressed on leukocytes and endothelial cells and binding to its ligand (leukocyte function-associated antigen-1) activates leukocyte transmigration via actin-cytoskeletal rearrangements [318,319]. Migration is most common in type I strains and tachyzoites that move between epithelial cells, via the paracellular pathway, and do so without affecting the integrity of the monolayer [258,317]. It must be also added that NO mediates IFN-γ-induced hyperpermeability, dilates tight junctions and depletes ATP in cultured human intestinal epithelial monolayers [320,321].

Studies in animals performed by Chen et al. [322] showed that multipotent mesenchymal stromal cells were transferred to rat maternal venous blood. They trafficked via VEGF-A and integrin-dependent pathways across the placenta, engrafted in various fetal organs, and persisted in offspring for at least 12 weeks. Silveira et al. [323] found that T. gondii may circulate in the peripheral blood of recently and chronically infected immunocompetent individuals with or without ocular lesions. In the patients with active ocular lesions and positive anti-toxoplasma IgG but negative IgM, propagation of the infection may still occur through the blood [323].

T. gondii can be also transmitted via breast milk [324,325], and achlorhydria observed in human newborn infants during the first 10 days of life [326] may favor infectivity with various forms of the parasite, such as oocysts, tachyzoites (invasive forms), and/or tissue cysts filled with bradyzoites [327]. It was demonstrated that breastfeeding seems to influence the development of diabetes in non-obese diabetic (NOD) neonates [328], because NOD progeny from NOD mothers deficient in T and B cells failed to develop diabetes [329]. Given the absence of activity of autoantibodies in this process [330,331], it seems that maternal T and predominantly B cells transferred during gestation or lactation might play a role in development of autoimmune diabetes. Roy et al. [275] found that in mice maternal T cells reactive against the endocrine pancreas of their progeny markedly increased islet infiltration. Specific maternal microchimeric T cells targeting fetal antigens in beta cells may also constitute evidence of their predisposing role in development of autoimmune T1DM. It must be emphasized that the maternal environment during gestation and a genetic predisposition to diabetes also play an important role in future development of T1DM in the offspring [290,332].

Possible Link between a Specific Role of T Cell Membrane Metalloproteinases (MMPS) in Development of Diabetes and T. Gondii Infection-Associated Positive Modulation of Macrophages Migration by Increasing Expression of Matrix Metalloproteinases and Decreasing CD44 Receptor at Cell Surface

Diabetes

MMPs, a family of secreted zinc proteases capable of degrading collagen and other matrix components, may participate in a wide variety of pathophysiological responses, including inflammatory pocesses, embryonic development, and cell apoptosis [333,334]. MMP-2 appeared to be critical in rat-pancreatic islet development and is activated between embryological days 17 and 19, and TGF-β is responsible for islet morphoghenesis by regulating MMP-2 expression [335], On the other hand, tissue inhibitor of metalloproteinase-1 (TIMP-1) prevents cytokine-induced dysfunction and cytotoxicity in pancreatic islets and β-cells, and in addition to inhibiting MMP-2 and MMP-9 activity may also inhibit cytokine-mediated apoptosis in various cell lines. TIMP-1 mediated these effects by inhibiting cytokineinduced activation of NK-κB [333]. Several studies demonstrated that pancreatic islets contain detectable amounts of MMPs and TIMPs [333]. In the children and adolescents with T1DM, Florys et al. [336] found serum MMP-2 as well as TIMP-1 and TIMP-2 levels significantly higher than in controls (p<0.01, p<0.02, and p<0.001, respectively), and a strong positive correlation was noted between MMP-2 and TIMP-2 (r=0.8, p<0.0001).

Ridnour et al. [337] showed a dose-dependent, biphasic regulatory effect of NO on the activity of MMPs (MMP-1, -9, and 13) secreted from murine macrophages. Low exogenous NO perturbed MMP/ tissue TIMP-1 levels by enhancing MMP activity and suppressing the endogenous inhibitor TIMP-1. Exposure of purified latent MMP-9 to exogenous NO demonstrated a concentration-dependent activation and inactivation of the enzyme, which occurred at higher NO flux. They suggested that NO regulation of MMP-9 secreted from macrophages might occur by RNI-mediated protein modification [337].

Zhou et al. [338] demonstrated that MMPs contribute to pancreatic islet fibrosis and insulin insufficiency in Zucker diabetic fatty rats. In both male and female rats, the authors found marked increases the mRNAs encoding proteases and extracellular matrix components that are associated with fibrosis and tissue remodelling. The mRNAs for MMP-2 (>10-fold increase in activity), -12, -14 were sharply increased with the onset of islet dysfunction and development of diabetes [338].

The pathogenesis of T1DM begins with the activation of autoimmune T killer cells and is followed by their homing into the pancreatic islets where they directly contact and subsequently destroy insulin-producing β cells [339]. Autoreactive IS CD8+ T killer cells transmigrated from bloodstream through pancreatic microvessels endothelial cell barrier and into the islets of Langerhans, are specific for islet-derived insulin antigen [339,340]. The cell surface-associated signalling and adhesion CD44 receptor and other adhesion receptors including selectins, cadherins, immunoglobulin superfamily cell adhesion molecules, such as VCA, ICAM-1, and -2) and integrins, which are expressed in both T cells and endothelial cells, contribute to the adhesion of T cells to the endothelium [341]. The high affinity interactions of T cell CD44 with its abundant endothelial ligand, hyaluronian, are essential for firm adhesion and the subsequent transmigration events [342,343]. T cell membrane type-1 MMP (MT1- MMP) regulates the functionality of CD44 (a marker of activated T cells) and thus control the rate at which T cells home into the pancreatic islets, finally affecting the severity of the disease [339]. CD44 is heavily glycosylated and its glycosylation negatively regulates oligomerization, the movement of CD44 across the plasma membrane, and recognition of hyaluronian [339,344].

T. gondii infection

The parasite is able to infect macrophages and dendritic cells for dispersal throughout the body. Seipel et al. [345] found that T. gondii infection positively modulated the macrophages migratory molecular complex by increasing MMPs, CD44 and alpha(v)beta(3) integrin. Migration in Matrigel™ of infected macrophages was augmented after 48 hrs of infection, and inhibition of MMPs abolished this process. T. gondii infection also induced a decrease of CD44 receptor at cell surface and increased secretion of active MMP-9. Infected macrophages showed increased expression of MT1-MMP and ADAM10 (a disintegrin and metalloproteinase membrane10) MMPs [345]. Expression and function of several MMPs in experimental animals and humans during T. gondii infection were presented in table 15 [346-350].

Model Mmpa-MMPb/TIMPb expression patternc Functional data/associations Comments References
1. In vitro        
T. gondii-infected THP-1 cells Mmp-2,-9, Timp-2↓, Mt1-mmp↑
proMMP-2, proMMP-9, TIMP-2↓
proMT1-MMP↑, actMT1-MMP↑
  MT1-MMP activation by T. gondii infection probably explains parasite dissemination and access to immune-priviledged sites. [347]
T. gondii-infected (macrophage-like) RAW 264.7 cells ActMMP-9↑ in infected supernatants
MT1-MMP↑ in T. gondii-infected cells
ADAM-10↑ in T. gondii-infected cells
MMP inhibitor I → Abolished invasiveness of T. gondii-infected macrophages over 3D ECM
MMPs might facilitate the access of infected leukocytes to immune-privileged sites
Increasing levels of MT1-MMP→ shedding of CD44→, a docking molecule for MMP-9 [345]
2. In vivo        
Ileum tissue from T. gondii-infected mice Mmp-2,-9/MMP-2,-9↑ in small intestine
ActMMP-2↑ in small intestine
IL-23p19KO; mice
Mmp-2,-9/MMP-2,-9↓
No actMMP-2
→ Significantly reduced intestinal pathology
MMP-2KO mice (compared to MMP-9KO and WT mice)
→ Protected against the development of intestinal immunopathology and early death; MMP-2 mediates immunopathology in T. gondii-infected ileitis.
Treatment with gelatinase inhibitors (doxycycline and MMPI RO28-2653)
→ Ameliorated intestinal pathology
Treatment with gelatinase inhibitors protects mice against T- gondii-induced immunopathology
Selective blockage of gelatinoses may be a safe and effective strategy in prevention and treatment of intestinal inflammation [348]
Brain tissue from T. gondii-infected mice Mmp-8, -10 and TIMP-1↑ in brain
MMP-8↑ in brain infiltrating CD4+/CD8+ T cells
MMP-10↑ in brain infiltrating CD4+ T cells
TIMP-1↑ in CNS-resident astrocytes and in brain infiltrating CD4+/CD8+ T cells
TIMP-1 deficient mice → Little morphological changes in tissue structure
→ ↑ CD4+ T cells in brain
→ Reduced parasite burden in brain
TIMP-1 is associated with inhibition of pathogen clearance without development of adverse pathology→
MMP-8 and –10 production by brain-infiltrating T cells implies a role for MMPs in brain tissue penetration; TIMP-1 is associated with inhibition of pathogen clearance [349]
3. Clinical studies        
Serum from pregnant women with or without T. gondii infection vs. healthy non-pregnant women MMP-12 - > associated with ↑ elastin degradation products;
Pregnant women with toxoplasmosis > healthy pregnant women > healthy non-pregnant women
  Interaction between MMP-12 and elastin in the serum of infected pregnant women suggests MMP-12 mediates damage to elastin and contributes to T. gondii-associated pathology during pregnancy [350]
↑↓: Increased or decreased levels; ADAM: A Disintegrin And Metalloprotease; actMMP: activated MMP; CNS: Central Nervous System; ECM: Extracellular Matrix; KO: Knockout; MMP: Matrix Metalloproteinase; MT-MMP: Membrane-Type Matrix Metalloproteinase; TIMP: Tissue Inhibitor Of Metallopproteinases; aMmp/Timp refers to mRNAs; bMMP/TIMP refers to proteins. WT: Wild Type.

Table 15: Expression and function of MMPs/TIMPs/ADAMs in toxoplasmosis (T. gondii infection) (acc. to Geurts et al. [346]; with own modification).

T. gondii tachyzoites store toxolysin 4 (TLN4), an extensively processed putative metalloproteinase, in the micronemes and secrete it in response to elevated calcium, suggesting a possible role in cell invasion [351]. TLN4 is initially synthesized as a large precursor (~260 kDa) that is extensively processed into multiple proteolytic fragments within the parasite secretory system, and at least some of these fragments remain associated in a large molecular complex [351].

Niehaus et al. [352] found that T. gondii glycosylphosphatidylinositols (GPIs) induced MMP-9 in human macrophage-like THP-1 cells via TLR2/4-dependent mechanism and the degradation of human extracellular galectin-3 (a substrate for MMPs-2 and -9, and a lectin specific for β-galactosides, which binds to both the glycan and lipid moieties of the parasite GPIs). It must be added that the parasite activated TLR2/4 receptors and induced a NF-κB-dependent production of TNF-α in macrophages [353,354].

Disturbed Carbohydrate Metabolism in Diabetes Mellitus. T. gondii Infection also Significantly Affects Glycolysis, Gluconeogensis and Tricarboxylic Acid (TCA) Cycle

Animals

Glycolysis, gluconeogenesis and the TCA cycle are central pathways of the carbohydrate metabolism which need to be tightly regulated depending on the cellular demand for energy, reducing power and precursors for biosynthesis pathways. Diabetes mellitus is a metabolic disorder characterized by chronic hyperglycemia with disturbances of carbohydrate, fat and protein metabolism [1].

Palsamy and Subramanian [355] found altered activities of the key enzymes of carbohydrate metabolism, such as hexokinase, pyruvate kinase, lactate dehydrogenase, glucose-6-phosphatase, fructose-1,6-biphosphatase, glucose-6-phosphate dehydrogenase, glycogen synthase and glycogen phosphorylase in liver and kidney tissues of streptozocin-nicotinamide-induced diabetic rats. During diabetic conditions, the glycogen levels, glycogen synthase activity and responsiveness to insulin signaling were diminished and glycogen phosphorylase activity was markedly increased [356] (Table 16). Glucose-6-phosphatase, a key enzyme in the homeostatic regulation of blood glucose level is critical in providing glucose mainly to the liver and kidney during diabetes, prolonged fasting or starvation [357]. It catalyzes the dephosphorylation of glucose-6-phosphate to free glucose as the terminal step in gluconeogenesis and glycogenolysis. This reaction occurs in the lumen of the endoplasmic reticulum and the enzyme complex is composed of a glucose-6-phosphate transporter that transport glucose-6-phosphate from the cytoplasm into the lumen of the endoplasmic reticulum and a glucose-6-phosphatase catalytic subunit that hydrolyzes the glucose-6-phosphate to glucose and phosphate [358]. Glucose-6-phosphate dehydrogenase catalyzes the first and rate-limited step of the hexose monophosphate shunt and produces NADPH needed for the maintenance of reduced glutathione and reductive biosynthesis [359]. NADPH is essential for both the production of ROS, such as superoxide anions and NO and the elimination of these radicals via glutathione peroxidase and catalase in hepatic as well as extrahepatic tissues [360]. It has been shown that modest changes in glucose-6-phosphate dehydrogenase activity itself have significant effects on cell growth and cell death in a variety of cell types [361,362].

Groups Glycogen Glycogen synthase Glycogen phosphorylase
Controls 60.83 ± 1.35 842.17 ± 10.91 658.33 ± 11.46
Diabetic rats 19.50 ± 2.35a 525.67 ± 14.39a 897.50 ± 28.26a
Units are expressed as: mg/g wet tissue for glycogen, μmoles of UDP formed/h/mg protein for glycogen synthase and μmoles Pi liberated/h/mg protein for glycogen phosphorylase. Values are given as mean ± SEM, for groups of 6 rats in each.
aValues statistically significant at p<0.05

Table 16: Level of glycogen content and activities of glycogen synthase and glycogen phosphorylase in liver tissues of streptozotocin-nicotinamide-induced diabetic rats (according to Palsamy and Subramanian [355]; with own modification).

Suhail and Rizvi [363] analyzed activities of the key glycolytic enzymes of red blood cells, i.e. hexokinase (HK), phosphofruktokinase (PFK) and pyruvate kinase (PK) and found that they were significantly (p<0.01) increased in patients with T1DM. It was suggested that the increased activity of the enzymes might be due to a greater proportion of young erythrocytes in diabetic individuals because of a shortened red cell life span as compared to healthy persons [363]. However, Sitzmann [364] measured the activity of the same three enzymes in 119 children suffering from juvenile T1DM and reported that the values were mildly diminished both in the group with good state of metabolic control and in those with bad control compared with the respective reference values [364]. This discrepancy may be at least in part explained by the exposure time of these patients to T. gondii infection, its intensity, cell/ tissue invasiveness of the parasite, its strain, etc. This suggestion may be supported by marked modulations of activities of enzymes participating in the metabolic pathways during glycolysis, gluconeogenesis and TCA-cycle found in T. gondii tachyzoites and bradyzoites [365-367] (Tables 17-19). However, it appeared that the level of mRNA upregulation of some enzymes important for carbohydrate metabolism was only moderate, therefore other mechanisms, such as an increase in gluconeogenesis enzyme activity or uptake of glucose-6-phosphate from the host cell by the hexose transporter [377] may also take place [365]. T. gondii utilizes host sugars for energy and to generate glycoconjugate that are important for its survival and virulence. The parasite glucose transporter is proficient in transporting mannose, galactose and fructose besides glucose, and serves as a major hexose transporter at its plasma membrane [70].

Protein name Gene name Change in expression Microarray experiment
↓Aldolasea A and B ↓ALDOA, ALDOB ↓↑
↓ATP synthasea beta subunit ↓ATP5D ↓↑
↓Cytochrome c oxidase subunit Vib ↓COX6B ↓↓ ↓↑
↓Dimethyl arginine dimethyl aminohydrolase ↓DDAH1 ↓↓ ↓↓
↓Enolasea 1 ↓ENO1 ↓M ↓↑
↓Glyceraldehyde 3 phosphate dehydrogenasea ↓GAPDH ↓↑ ↓↑
↓Phosphoglycerate kinasea 1 ↓PGK1 ↓M ↓↑
↓Protein disulfide isomerase ↓P4HB ↓↑ ↓↑
↓Pyruvate kinase M2 isozyme ↓PKM2 ↓M
↓Thioredoxin domain 5 isoform 2 ↓TXNDC5 ↓↑
↓Triose phosphate isomerase ↓TPI1 ↓M ↓↑
Host cell proteins were designated as being downregulated in expression (↓), upregulated (↑), or modulated (M). aThese host cell proteins also changed expression in the brains of patients with mild cognitive impairement, early AD, or AD [369]. It must be noted that T. gondii tachyzoites are thought to rely upon both glycolysis and the tricarboxylic acid cycle, while bradyzoites are largely dependent upon glycolysis [364,371]. Although tachyzoites utilize both glycolysis and oxidative phosphorylation to obtain energy, glycolysis seems to be the predominant pathway for ATP synthesis in the bradyzoite [372,373]. Moreover, ENO2 and lactate dehydrogenase1 are only found in tachyzoites, while ENO1 and lactate dehydrogenase 2 are exclusively expressed in bradyzoites [374,375]. Silencing of tachyzoite ENO2 altered nuclear targeting of bradyzoite ENO1 in T. gondii [376,377].

Table 17: Selected modifications in the proteomes of human foreskin fibroblasts infected with T. gondii: proteins implicated in glycolysis Nelson et al. [368]; with own modification).

Enzyme Gene accession numbers from ToxoDataBase a
Glucosephosphate-Mutase I (GPM1) 76.m00002
Glucosephosphate-Mutase II (GPM2) 641.m00009
Fructose-Biphosphatase I (FBP1) 20.m03907
Fructose-Biphosphatase II (FBP2) 50.m00005
PEP-Carboxykinase I (PEP-CK1) 80.m00002
PEP-Carboxykinase II (PEP-CK2) 80.m02252
Pyruvate-Carboxylase (Pyc) 76.m01567
In contrast to most of the other T. gondii gluconeogenesis genes,namely fructose-biphosphatase,glucosophosphate-mutase and PEP-carboxykinase, which are encoded twice in the genome,pyruvate-carboxylase is only encoded once and the enzyme is localized inside the single mitochondrion,while the remaining reactions of gluconeogenesis are typically cytosolic [365]. a(www.toxodb.org/toxo/home.jsp)

Table 18: Gene prediction for irreversible steps in gluconeogenesis in T. gondii (according to Fleige et al. [365]; with own modification).

Enzyme Gene accession numbers from ToxoDataBase a Localization
Citrate Synthase I (CS1) 59.m03414 Mitochondrion
Citrate Synthase II (CS2) 20.m03767  
Citrate Synthase III (CS3) 42.m03311  
Aconitase (ACN) 42.m03524 Mitochondrion
Isocitrate-Dehydrogenase I (IDH1) 583.moo674 Mitochondrion
Isocitrate-Dehydrogenase II (IDH2) 57.m00028  
a-Ketoglutarate-Dehydrogenase E1 (OGDH E1) 49.m03397  
Dihydrolipoly-Transacetylase (OGDH E2) 38.m00017  
Dihydrolipoly-Transacetylase (OGDH E3) 20.m03954  
Succinyl-coa-synthetase alpha (Scsa) 80.m00087 Mitochondrion
Succinyl-coa-synthetase (ATP) (Scsb) 583.m00592 Mitochondrion
Fumarase (FUM) 57.m01846  
Malate-Dehydrogenase (MDH) 641.m00168 Mitochondrion
FAD Malate-Dehydrogenase (MDH-FAD) 80.m00006 Mitochondrion
Branched-Chain a-Ketoglutarate-Dehydrogenase E1 (BCOGDH-E1) 49.m00028  
It seems that tachyzoites, but not bradyzoites, posses a functional tricarboxylic acid cycle because in cell homogenates of tachyzoites, activities of Succinate Dehydrogenase (SDH) and NADP+-dependent isocitrate dehydrogenase were detected, while no SDH activity could be found in bradyzoites [365,372] a(www.toxodb.org/toxo/home.jsp)

Table 19: Gene prediction for putative tricarboxylic acid cycle-associated enzymes in T. gondii and their localization (according to Fleige et al. [365]; with own modification).

T. gondii infection

The parasite posses a complete glycolytic pathway [366], as well as all enzymes for the TCA cycle and mitochondrial electron transports chain [365-368]. Tachyzoites are thought to rely upon both glycolysis and the TCA cycle, unlike the bradyzoites, which are thought to be largely dependent upon glycolysis [370]. Several enzymes of the glycolytic pathway have been shown to be modulated also during differentiation [370,378], with some showing stage-specific isoforms, such as enolase and lactate dehydrogenase (LDH) [379]. LDH1 and LDH2 genes and their products have been implicated in the control of a metabolic flux during parasite differentiation, and LDH knockdown parasites exhibited variable growth in either the tachyzoite or the bradyzoite stage, as compared with the parental parasites [380].

Denton et al. [372] found that both developmental stages of T. gondii (i.e. tachyzoites and bradyzoites) contained high activities of phosphofructokinase (specific for pyrophosphate rather than ATP), pyruvate kinase and lactate dehydrogenase (LDH), suggesting that energy metabolism in both forms of the parasite may center around a high glycolytic flux linked to lactate production. The significantly higher activity of the latter two enzymes in bradyzoites suggested that lactate production is particularly important in this developmental form. It should be noted that LDH is a terminal glycolytic enzyme that plays an indispensable role in the interconversion of pyruvate to lactate to yield energy under anaerobic conditions [378] and the reaction occurs in both cytosolic and mitochondrial compartments [357]. Both parasite forms contained low activities of NAPD(+)-linked isocitrate dehydrogenase. The results were consistent with the bradyzoites lacking a functional TCA cycle and respiratory chain [372]. Moreover, T. gondii uses PP(i) in place of ATP as an energy donor in at least two reactions: the glycolytic PP(i)-dependent phosphofructokinase and V-H(+)-PPase [vacuolar H(+)-translocating PPase (pyrophosphatase)] [381]. Pace et al. [381] showed that overexpression of cytosolic pyrophosphatase (TgPPase) in extracellular tachyzoites led to a 6-fold decrease in the cytosolic concentration of PP(i) relative to wild-type strain RH tachyzoites. This reduction of PP(i) was associated with a higher glycolytic flux in the overexpressing mutants. In addition to elevated glycolytic flux, TgPPase-overexpressing tachyzoites also possessed higher ATP concentrations relative to wild-type RH parasites. The authors believed that PP(i) had a significant regulatory role in glycolysis and, potentially, other downstream processes that regulate growth and cell division [381].

Glycolysis is a major source for T. gondii motility [366,373]. In the presence of glucose, a carbon source for both glycolysis and oxidative phosphorylation, a robust movement in a majority of the tachyzoites was observed, while omission of glucose resulted in a drastic decrease in the fraction of mobile parasites [71.6 ± 7.4% vs. 2.7 ± 2.9% (SD)]. Host cell egress and invasion induce marked relocations of glycolytic enzymes in T. gondii tachyzoites [366,373]. In the parasite, Coppin et al. [373] identified several genes and proteins associated with amylopectin synthesis or degradation and glucose metabolism, including different forms of certain glycolytic enzymes, which are stage-specifically expressed during tachyzoite-bradyzoite interconversion. This interconversion is central to the pathogenic process and sometimes is associated with recrudescence of infection observed especially in the patients with AIDS. Biochemical analysis revealed that the glycolytic metabolite lactate is an inhibitory component of resistant for conversion cells, and upregulation of glycolysis in permissive cells through the addition of glucose or by overexpression of the host cell kinase, Akt (an intracellular ligand), was sufficient to convert cells from a permissive to a resistant phenotype [382]. In chronic infection with the parasite, the bradyzoites are located within tissue cysts that are enclosed by the wall containing specific lectin binding sugars, while the bradyzoites have accumulated large amounts of amylopectin, the storage polysaccharide of glucose [373].

Pomel et al. [367] demonstrated that the glycolytic enzymes of T. gondii tachyzoites undergoes a striking translocation from the parasites’ cytoplasm to their pellicles upon the parasite egress from host cells. Specifically, the glycolytic enzymes are relocated to the cytoplasmic face of the inner membrane complex as well as to the space between the plasma membrane and inner membrane complex. The glycolytic enzymes remain pellicle-associated during extended incubations of parasites in the extracellular millieu and do not revert to a cytoplasmic location until well after parasite have completed invasion of new host cells [367]. Translocation of glycolytic enzymes to and from the parasite pellicles occurred in response to changes in extracellular [K+] experienced during egress and invasion, a signal that requires changes of [Ca2+] concentration in the parasite during egress [367].

Nelson et al. [368] found a considerable up-regulation of the glycolytic pathway in T. gondii-infected cell because six of the ten enzymes involved in glycolysis showed evident modification, with either an increase or modulation (aldolase A and B, glyceraldehyde 3 phosphate dehydrogenase, phosphoglycerate kinase, enolase, and pyruvate kinase), and one which showed a decrease in expression (triose phosphate isomerase) [368,383]. Increased glycolysis results in the production of both ATP and pyruvate, which enters most biosynthetic processes in the cell and may considerably affect oxidative phosphorylation (modulation of ATP synthase beta subunit, cytochrome c oxidase subunit Vib, and inorganic pyrophosphatase) (Tables 17 and 20). It should be noted that Kimata and Tanabe [384] found a markedly reduced invasion rate of T. gondii observed in ATP-depleted chick embryo erythrocytes, and the rate was restored in ATP-restored cells. This indicated that T. gondii invasion was dependent on the ATP level of the erythrocytes). It was also demonstrated that during T. gondii infection, six proteins involved in carbohydrate metabolism were modulated in the infected cell (aldose reductase, aldehyde dehydrogenase 1A3, aldehyde dehydrogenase X, hexoaminidase B, phosphoenoylpyruvate carboxykinase, and 6-phosphogluconolactonase) [368]. Infection with T. gondii resulted also in dysregulation of the host cell cycle by promoting the G1-to-S transition in infected human foreskin fibroblasts [385].

Protein name Gene name Change in expression Microarray experiment
6-Phosphogluconolactonase PGLS
Acetyl coenzyme A acyltransferase ACAT1 M
Adenylate kinase 2 isoform a AK2
Adenylate dehydrogenase 1A3 ALDH1A3
Aldehyde dehydrogenase X ALDH1B1
Aldose reductase  AKR1B1
Carbonyl reductasea CBR1  
Cathepsin B CTSD
Coproporphyrinogen oxidase CPOX  
Glutathione synthetase GSS  
Glutathione-S-transferasea chain A GSTA2
Phosphoenolpyruvate carboxykinase pck1  
Protein-L-isoaspartate O-methyltransferase PCMT1  
Pyridoxine 5’-phosphate oxidase PNPO  
Pyrophosphatase (inorganic) PPA1  
Host cell proteins were designated as being down regulated in expression (↓), upregulated (↑), or modulated (M). Modulated proteins had expression altered across several isoforms on the same gel using the Amersham difference gel electrophoresis, and this probably indicated a posttranslational modification event [354]. aThe host cell proteins, which also changed expression in the brains of patients with mild cognitive impairement, early AD, or AD [369]

Table 20: Selected modifications in the proteomes of human foreskin fibroblasts infected with T. gondii: proteins implicated in metabolism (Nelson et al. [368]; with own modification).

Fleige et al. [365] showed that in T. gondii all analyzed TCA cycle enzymes were localized in the mitochondrion, including both isoforms of malate dehydrogenase (Table 19). The TCA cycle metabolizes acetyl-CoA into CO2, thereby generating ATP and reducing power, which typically enters the respiratory chain. The authors suggested that tachyzoites, but not bradyzoites, posses a functional TCA cycle [365]. Moreover, data on the parasite carbohydrate metabolism by localizing enzymes for glycogenesis and amylopectin synthesis were provided (Table 21). It was found that reactions of gluconeogenesis are mainly cystosolic, including PEP-carboxykinase-I and both isoforms of fructose bisphosphatase and glucosephosphate-mutase, while pyruvate-carboxylase is localized in the single mitochondria [366] (Table 18). In vitro, bradyzoites displayed a 2-fold upregulation of fructose-biphosphate I and glucosephosphate-mutase I (5-fold) and II (also 5-fold) compared with tachyzoites. This moderate upregulation of gluconeogenesis genes was likely contributing to satisfying the increased demand of glucose-6-phosphate for amylopectin synthesis, which take place during bradyzoite development [365] (Table 21). However, since the level of mRNA upregulation is only moderate, other mechanisms, such as an increase in gluconeogenesis enzyme activity or uptake of glucose-6-phosphate from the host cell by the hexose transporter [377] might also take place [365]. All these findings strongly suggest that chronic T. gondii infection plays an important role in carbohydrate metabolism disturbances characteristic for T1DM and T2DM.

Enzymes Bradyzoite Tachyzoite
Actin
UDP-glucose phosphorylase
Starch (glycogen) synthase
Branching enzyme 1 ↑↑
Branching enzyme 2 ↑↑
Isoamylase ↑↑
D-enzyme (α-1,4-glucanotransferase)
α-glucan 0
α-amylase 0
α-glucosidase ↑↑ 0
Glycogenin
R1 protein
(α-glucan water dikinase)
0
Debranching enzyme
↑↑: Markedly increased gene expression; ↑: Increased expression; ↑: Weak expression; 0: No gene present; T. gondii enzymes were identified at the genome Web site: htttp://www.toxodb.org. The parasite genome encodes two fructose 1,6-biphosphatase isoenzymes, a single pyruvate-carboxylase, and two PEP-carboxykinases. The conversion from glucose-6-phosphate into glucose-1-phosphate,which forms the link between amylopectin metabolism and gluconeogenesis, is catalysed by two isoforms of glucosephosphate-mutase [365]. The following soluble tachyzoite antigenic proteins have been identified: a putative protein disulfide isomerase, Hsp60, Hsp70, a pyruvate kinase, a putative glutamate dehydrogenase, a coronin, a protein kinase C receptor 1, a malate dehydrogenase, a major surface antigen 1, an uridine phosphorylase, and a peroxiredoxin [93]

Table 21: Bradyzoite and tachyzoite stage-specifically expressed genes coding the enzymes involved in T. gondii amylopectin metabolism (Coppin et al. [386]; with own modification).

N-Linked Glycosylation of Proteins by T. Gondii may Participate in the Increased Generation of HbA1c Characteristic for Diabetes Mellitus

Diabetes

In 1968, Rahbar [387] first discovered the association of increased HbA1c with diabetes mellitus [388]. HbA1c results from glucose condensing nonenzymatically with the N-terminal valine residue of the hemoglobin beta chain [389], and the HbA to HbA1c conversion takes place during the entire life span of the red blood cells, so the HbA1c concentration is higher in old red cells than in new red cells [389]. It must be noted that a significantly higher incidence of major congenital anomalies occurred in the offspring of women who had elevated glycosylated hemoglobin levels in the early part of their pregnancy [390]. This finding may be consistent with the severity of anomalies observed in newborn infants with congenital T. gondii infection.

Gould et al. [391] found that erythrocyte 2,3-diphosphoglycerate, a catalyst of glycation, was elevated in high compared with low glycators (5.61 ± 0.26 vs. 4.81 ± 0.24 mmol/l, p<0.001), and mean centile glycated hemoglobin was positively correlated with intra-erythrocyte pH (r=0.55, p<0.05).

Merino-Torres et al. [392] found that hemoglobin glycosylation index (HGI) is not related with blood glucose. In addition, the percentage of self-monitored blood glucose was the same for high glycosylators (HGI<0) as for low glycosylators (HGI>0). Moreover, Lachin et al. [393] showed that glycation index was not an independent risk factor for microvascular complications, and effect of this bioparameter on the risk was wholly explained by the associated level of A1c. Therefore, it was suggested that HGI should not be used to estimate risk of complications or guide therapy [393]. These findings may be consistent with an important role of T. gondii infection being a potential risk factor in development of diabetes.

T. gondii infection

The post-translational modification of proteins by the addition of N or O-linked oligosaccharides is common in most eukaryotic cells. They are added onto proteins either during their transport into the endoplasmic reticulum (in the case of O-linked oligosaccharides), or during their transport through the Golgi complex (in the case of the O-linked oligosaccharides) [394]. Protein glycosylation plays a critical role in the interaction of various pathogens with their host cells and organisms [395], and the correct folding of proteins and their export from the endoplasmic reticulum [396], as well as the correct intracellular targeting of proteins [397]. Luk et al. [394] showed that N-glycosylation is a common post-translational modification of proteins in T. gondii essential for the survival of the parasite and its viability, but the structure of these glycans differs substantially from that usually encountered in animals, plants and other unicellular organisms. The parasite synthesizes a large number of proteins with N and O-linked glycans that are found throughout the secretory pathway of tachyzoites. T. gondii tachyzoites contain at least 11 major and multiple minor N-glycosylated proteins, but so far only two have been reported to be glycosylated. The presence of N-linked glycans on GAP50, the membrane anchor for the myosin XIV motor complex in the parasite and gp23, which is a GPI-anchored Toxoplasma surface protein , have been demonstrated [384]. It was found that tachyzoite surface glycoprotein gp23 has N-linked glycans in the hybrid-type glycans composed of at least N-acetylgalactosamine, N-acetylglucosamine and mannose. T. gondii microsomes have the ability of to synthesize in vitro a glycosylated lipid-bound high mannose structure that is assumed to be identical with the common precursor for N-glycosylation in eukaryotes [398].

Garenaux et al. [399] demonstrated that T. gondii independently transfers endogenous truncated as well as host-derived N-glycans onto its own proteins, and suggested that the parasite scavenges N-glycosylation intermediates from the host cells to compensate for the rapid evolution of its biosynthetic pathway, which is primarily devoted to modification of proteins with glycosylphosphatidylinositol rather than N-glycans. In a similar way, T. gondii has been shown to mobilize selected host lipids to fulfill its high metabolic requirements during proliferation [400-402]. These exchanges could be facilitated by the close association of the parasitophorous vacuole (PV) membrane with the host endoplasmic reticulum (and mitochondria), where the early steps of N-glycan biosynthesis take place [399]. It must be emphasized that the PV membrane surrounding intracellular T. gondii functions as a molecular sieve allowing exchange of molecules up to 1300-1900 Da between the host cell cytoplasm and the parasitophorous vacuolar space [403], i.e. protein antigens consisting of about 9-12 amino acids with a mean molecular weight of approximately 160 Da.

Finally, Fauquenoy et al. [404] demonstrated that N-glycans are required for efficient binding of T. gondii to gliding partners, because the parasite motility and host cell entry was severely impaired in the unglycosylated GAP50 mutants. In addition, N-glycosylation was found to be a prerequisite for GAP50 transport from the endoplasmic reticulum to the Golgi apparatus and for its subsequent delivery into the inner complex membrane. Thus, it seems that T. gondii uses N-glycosylation intermediates from the host cell for its own metabolic processes and such metabolic intervention may interfere with the rate of HbA1c generation in diabetics.

T. Gondii Infection may be atleast in Part Responsible for Development of Glucose-6-Dehydrogenase (G6PD) Deficiency, and Probably Participate in the Enhancing Diabetic Ketoacidosis Severity

G6PD deficiency, the most common enzyme deficiency and an X-linked inherited disorder, is affecting over 400 million people world-wide, and causes several diseases, including neonatal hyperbilirubinemia, with acute and chronic hemolysis, although persons with this condition may be asymptomatic.

Tabbara et al. [405] performed serology testing for the presence of T. gondii and also analyzed frequency of glucose-6-phosphate dehydrogenase (G6PD) deficiency in 91 blood donor male volunteers aged 17 to 52 years. They found that 53 (58%) individuals were G6P dehydrogenase deficient. In addition, it appeared that 31 (58.5%) G6PD deficient subjects had positive titers for T. gondii as compared to 9 (24%) G6PD normal persons (p<0.002). It was suggested that G6PD deficiency increased the risk for the parasite infection by 2.5 fold probably due to decreased killing effect of phagocytic cells [405,406]. In addition, Gupta et al. [407] demonstrated that the parasite uses its secretory apparatus to modify lipids in the PVM and host cell membranes, because secreted T. gondii soluble phosphatidylserine decarboxylase reduced externalized phosphatidylserine on host cells enabling evasion of phagocytosis. The presence of increased serum anti-T. gondii IgM and IgG titers and G6PD deficiency were also reported in a 5-years-old male child [408].

Carette et al. [409] described two patients, one with ketosis-prone type 2 diabetes and the other with T1DM and ketoacidosis, who developed hemolysis during acute decompensation of the disease states. It appeared that both these patients had G6PD deficiency. It was suggested that this abnormality might also be facilitated by the erythrocyte depletion of glutathione, an important antioxidant, which is observed in the patients with diabetes [409,410].

Sobngwi et al. [411] demonstrated that G6PD deficiency alone is not a causative factor of ketosis-prone diabetes because they found a high (20.3%) prevalence of G6PD deficiency also in individuals without G6PD gene mutation, which may suggest a novel pathomechanism predisposing to ketosis-prone diabetes. In addition, in an adolescent with G6PD and T1DM it was found that disorders of hemolysis reduce the exposure time of hemoglobin to glucose, resulting in a falsely low hemoglobin A1c level discordant with blood glucose measurements [412]. Xu et al. [413] found that chronic hyperglycemia caused inhibition of G6PD activity via decreased expression and increased phosphorylation of G6PD, which therefore increased oxidative stress. In cultured cells high glucose concentrations caused activation of protein kinase A (PKA) and subsequent phosphorylation and inhibition of G6PD activity and hence decreased NADPH generation [414]. One may suggest that in the patients with ketosis-prone diabetes, a concomitant infection with T. gondii exerted similar effects on PKA activity and phosphorylation of the enzyme (Table 22), which could potentiate harmful effects of high glucose levels, thus finally aggravating clinical course of the disease. This reasoning may be supported by the findings that mononuclear leukocytes from obese patients with T2DM also have reduced activity of 6GPD, hexokinase, and 6-phosphofructokinase [419], and an increased prevalence of proliferative retinopathy was found in the patients deficient in G6PD [420]. In addition, moderate upregulation of gluconeogenesis genes was likely to contribute to the increased demand of glucose-6-phosphate for amylopectin synthesis (this reflects process of gluconeogenesis in contrast to glycolysis, i.e. degradation of this storage material), which takes place during bradyzoite development [365] (Table 21). However, since the level of mRNA upregulation was only moderate, other mechanisms, such as an increase in gluconeogenesis enzyme activity or uptake of glucose- 6-phosphate from the host cell by the hexose transporter [363] might also take place [361]. This may suggest that T. gondii infection of the red blood cells is able to filch this enzyme from the host cells for its own metabolic requirements, thus leading to the enhancement of G6PD erythrocyte deficiency and aggravation of diabetes state described above by Sobngwi et al. [411]. These reasoning is consistent with the finding of Usher-Smith et al. [421] that one of several factors associated with the presence of diabetic ketoacidosis at diagnosis of diabetes in children and young adults was a preceding infection (OR 3.41, CI 0.94 to 10.47).

Protein (localization) Phosphorylation status in infected cells Functional category
Caldesmon Unphosphorylated Mitosis
Calreticulin Unphosphorylated Protein folding
Nucleobindin Unphosphorylated Protein fate
Protein disulphide isomerase Unphosphorylated Structural
Thyroid hormone binding protein Unphosphorylated Signal transduction/cellular communication
Chaperonin HSP60 (mitochondria) Phosphorylated Energy metabolism
Lamin A protein (mitochondria) Phosphorylated Unknown
Vimentin (mitochondria) Phosphorylated Structural
T. gondii kinase activity is involved in phosphorylation of host IκBα and this unusual mechanism can be utilized in manipulating the NF-κB pathway [415]. Moreover, a novel parasite kinase activity at the T. gondii parasitophorus vacuole membrane is also capable of phosphorylating host IκB [416]. There is biochemical evidence for the presence of an oxidative phosphorylation and functional respiratory chain in the mitochondrion of tachyzoites [417]. Recently, cAMP dependent protein kinase important for the tachyzoite growth was identified in the parasite [95], and protein phosphorylation is a key event in the process of T. gondii-host cell interaction [418]. Changes in the proteomes of human foreskin fibroblasts following infection with T. gondii included, e.g. protein kinase C (delta binding protein) (modulated),protein kinase NYD-Sp9 (↑, up-regulated in expression), protein serine/threonine kinase (↓,down-regulated),glutathione synthetase (↑), glutathione-S-transferase chain A (↓), as well as several enzymes involved in glycolysis (Table 17) [368].

Table 22: Proteins undergoing a change in phosphorylation state following T. gondii infection (Nelson et al. [368]; with own modification).

T. Gondii Infection may be Associated with Amyloid Deposition in the Pancreatic Islet Beta Cells. The Presence of Lactoferrin (Lf) in Amyloid Fibrils Characteristic for Patients with T2DM Probably Reflects Host Defense Reaction to the Parasite Latent Infection

Patients with diabetes

Amyloidosis is a disorder of protein metabolism in which normally soluble autologous proteins are deposited in tissues as abnormal insoluble fibrils, causing structural and functional disruptions [422]. In mouse model of type 2 diabetes evidence was presented for role of islet amyloid formation rather than direct action of amyloid [125]. Amyloid fibril formation is the hallmark of T2DM and amyloid fibrils deposit in the extracellular space and generally co-localize with the glycosaminoglycans (GAGs) of the basement membrane, a specialized component of the extracellular matrix that mainly is built of collagen and GAGs. GAGs have been shown to accelerate formation of amyloid fibrils in vitro for a number of protein systems [423]. Monsellier et al. [423] found that the GAGs acceleration effect was mainly governed by three parameters that account for about 75% of the observed experimental variability: the GAG sulfation state, the solute molarity, and the ratio of protein and GAG molar concentrations. GAGs are long unbranched polysaccharides that often occur as O- or N-linked chains of proteoglycans, with the exception of hyaluronic acid existing in a free form. Naturally occurring GAGs include heparin, heparin sulfate, dermatan sulfate, keratan sulfate, chondroitin sulfate and hyaluronic acid [423]. GAGs have been found intimately associated with all types of amyloid deposits in vivo so far analyzed, which may suggest that they play a pivotal role in amyloidogenesis [424-426] because they display an ability to promote fibrillogenesis in vitro for a number of protein or peptide systems [427-430]. For example, heparan sulfate and HS proteoglycans accumulation with amyloid β-peptide deposits was found in AD and Tg2576 mice [431]. In addition, for ordered protein to fibrillate, its unique and rigid structure has to be destabilized and partially unfolded, while on the other hand, fibrillogenesis of a natively unfolded protein involves the formation of partially folded conformation, i.e. partial folding rather than unfolding [432]. Moreover, the AD-associated amyloid β-protein exerted potent antimicrobial activity against eight common and clinically relevant microorganisms [433]. Lf, a component of amyloid fibrils in pancreatic islet β-cells, was found to have strong anti-toxoplasmatic activity [434]. In addition, long-term lactation exerted a protective effect on the development of T2DM in women with recent gestational diabetes mellitus [435], at least in part probably because the breast-milk contains increased amounts of Lf [434].

Amyloid formation and aberrant protein aggregations play a role in a range of human diseases including T2DM, Alzheimer’s diseases and Parkinson’s disease [436-438] (Table 23). Human islet amyloid polypeptide (IAPP, or amylin) is the major protein component of the pancreatic islet amyloid associated with T2DM [124,440-443] and IAPP induced toxicity is believed to contribute to the loss of β-cell mass associated with the late stages of T2DM. This amorphous proteinaceous material originally described as hyaline has tinctorial properties of amyloid [444]. Islet amyloid polypeptide is a 37 aminoacid, beta-cell peptide with is co-stored and co-released with insulin [445,446].

Diseases Protein that forms toxic oligomers Cell lost
T2DM islet amyloid polypeptide b-cells
Alzheimer’s diseasea b-amyloid protein cortical neurons
Parkinson’s diseasea synuclein dopaminergic neurons
Prion encephalopathy/
transmissible spongiform encephalopathies
prion cortical neurons
Amyotrophic lateral sclerosis mutant superoxide dysmutase motor neurons
Polyglutamine/Huntington’s disease Huntington’s polyglutamine pyramidal neurons
The islet amyloid is derived from islet amyloid polypeptide (IAPP,amylin),a protein co-expressed and co-secreted with insulin by pancreatic β-cells. aIt must be noted that anti-IgG T. gondii antibodies were found to be significantly increased in both Alzheimer’s and Parkinson’s diseases as compared with controls (p<0.001)

Table 23: The common molecular basis of islet amyloidogenic proteins-related T2DM and neurodegenerative diseases (according to Haataja et al. [439]; with own modification).

Amyloid deposition in pancreatic islet is one of the most common pathological features of T2DM found in at least one islet at postmortem in more than 90% of diabetic patients, but also demonstrated in about 15% of elderly (>60 years old) non-diabetic individuals [445,447,448]. In human diabetes, islet amyloid can affect less than 1% and up to 80% of islet mass indicating that islet amyloidosis largely results from diabetes-related pathologies. Interestingly, in the aged rats vitamin D administration mitigated age-related cognitive decline through the modulation of proinflammatory state, increased beta amyloid clearance and decreased amyloid burden [449].

Islet amyloid polypeptide (amylin) is secreted from pancreatic beta-cells, converted to amyloid, and its immunoreactivity was localized to beta-cell lysosomes. Pharmacological doses of islet amyloid polypeptide were found to inhibit insulin secretion as well as insulin action on peripheral tissues (insulin resistance) [450].

T. gondii infection

Tachyzoite forms of the parasite grow within an intracellular vacuole surrounded by host’s cell mitochondria and endoplasmic reticulum [88] suggesting that the rapid replication and propagation of the tachyzoites may imply essential metabolites supplied by these organelles of the infected cells [451]. Bradyzoites transform the vacuolar membrane to an envelope called a cyst-wall which loses the ability to bind to mitochondria and endoplasmic reticulum of the infected cells [452]. Tables 24 and 25 are presented with number of organelle and inclusion bodies in different forms of the protozoan, which contain various substances, including amylopectin, probably important for generation of the amyloid present in diabetic islet cells. Guerardel et al. [454] found that T. gondii synthesizes amylopectin following changes in the environmental conditions and this storage polysaccharide differs from glycogen and starch in terms of glucan chain length (Table 21). The authors demonstrated that the origin of the host cell can affect the physiology and some key metabolism of the parasite. Using HepG2 cells they discovered that the culture medium of growing intracellular tachyzoites turned yellow rapidly, compared to that of tachyzoites growing in non-transformed human foreskin fibroblast cells. This lead to the acidification of the culture medium from pH 7-7.5 to pH 6-6.5 and this change in the pH probably was a consequence of glycolysis responsible for the major energetic metabolism driving ATP synthesis in the parasite, and the elevation of lactic acid level [454]. It is known that the culture medium acidification causes an accumulation of large amounts of amylopectin (10-20 mg of amylopectin per 2×1010 parasites). Nutrient starvation occuring during this process may also be involved in amylopectin biosynthesis and accumulation. In another intracellular apicomplexan, Eimeria spp., depletion of amylopectin by incubation of the parasite at 37°C or 41°C lead to the impairment of these functions [455,456]. In addition, Ryley et al. [457] incubated Eimeria tenella sporozoites anaerobically, and correlated the decrease in amylopectin stores of the parasite with production of lactic acid plus lesser amounts of CO2 and glycerol.

Stage Mean no. (range) of:
  Rhoptries Micronemes Dense granules Amylopectin Lipid
Sporozoite 5.9 (2-11) 55 (40-78) 9.4 (5-15) 7.8 (3-13) 1.25 (1-3)
Tachyzoite 6.7 (2-11) 25 (19-38) 9.1 (5-17) 2.4 (1-6) 0.6 (0-2)
Bradyzoite 5.5 (2-8) 75.5 (36-112) 2.7 (1-5) 21.8 (7-38) 0
Sporozoites were freshly excysted from 34-day-old oocysts. Tachyzoites were obtained from the peritoneum of an IFN-γ knockout mouse 8 days after inoculation of tissue cysts. Bradyzoites were from cysts in the brains of mice at 8 months after inoculation of oocysts. It must be noted that tachyzoites and bradyzoites could both be present in the same parasitophorus vacuole indicating that stage conversion from tachyzoite to bradyzoite is asynchronous. Numbers represent means that were obtained by counting all organelles or inclusion bodies in 20 longitudinal sections of each type of zoite; ranges are given in parentheses

Table 24: Relative numbers of organelle and inclusion bodies in sporozoites, tachyzoites, and bradyzoites of the VEG strain of T. gondii (according to Dubey et al. [453]; with own modifications).

Stage Mean size (nm) (range) in:
  Dense granules Amylopectin Lipid
Sporozoite 208 (175-250) 356 (200-460) 388 (200-550)
Tachyzoite 244 (133-334) 201 (103-333) 224 (150-400)
Bradyzoite 181 (167-201) 358 (192-603) 0
For the source of these infectious stages, (Table 23). It must be noted that tachyzoites and bradyzoites could both be present in the same parasitophorus vacuole indicating that stage conversion from tachyzoite to bradyzoite is asynchronous

Table 25: Relative sizes of inclusion bodies in sporozoites, tachyzoites, and bradyzoites of the VEG strain of T. gondii (according to Dubey et al. [453],with own modifications).

Sawesi et al. [458] found that in mice T. gondii infection resulted in highly increased extracellular levels of glycosaminoglycans, including hyaluronan and chondroitin sulfate A, suggesting a role of these substances in the general defense mechanism of the host cells. This suggestion is consistent with the finding that cell surface heparan sulfate (HS) and glycans, which contain sialic acid, have been shown to act as potential receptors for the parasite [459-461]. Loss of HS chains or sialic acid from cellular glycoconjugates resulted in marked reduction of T. gondii infection in vitro [460]. Inhibition of glycosaminoglycanmediated amyloid formation by islet amyloid polypeptide and pro- IAPP processing intermediates reported by Meng and Raleigh [436], as well as the presence of Lf in amyloid fibrils and deposits in the cornea, seminal vesicles, and brain [462], are in agreement with the abovepresented findings.

Nilsson and Dobson [462] found a highly amyloidogenic region of Lf (sequence NAGDVAFV) that forms amyloid fibrils at pH 7.4 when incubated at 37°C. Although full-length Lf does not by itself form amyloid fibrils, the protein binds to the peptide fibrils and the binding constitutes a selective interaction with the NAGDVAFV fibrils. The Lf appears to coat the peptide fibril surface to form mixed peptide/protein fibrils, but there was no formation of Lf-only fibrils. It was suggested that such process could be generally important during formation of amyloid fibrils in vivo because the identification of both full-length protein and protein fragments was common in ex vivo amyloid deposits [462].

Ando et al. [463] reported that in three patients with corneal amyloidosis electrophoresis of amyloid fibrils revealed Lf with and without sugar chains, and N-terminal sequence analysis demonstrated full-length Lf and a truncated tripeptide of N-terminal amino acids, Gly-Arg-Arg. Carboxymethylated wild-type Lf formed amyloid fibrils in vitro. Lf gene analysis in the 3 patients revealed a Glu561Asp mutation, and a compound heterozygote of Ala11Thr and Glu561Asp mutations in 1 patient. Heterozygotic Glu561Asp mutation was found in 44.8% of healthy Japanese volunteers, suggesting that the mutation may not be an essential mutation for amyloid formation (p=0.104), and that Lf was this precursor protein [463].

Lf is a cationic iron-binding glycoprotein belonging to the transferrin family, which accumulates in the amyloid deposits in the brain in neurodegenerative diseases, such as AD and Pick’s disease. Iwamaru et al. [464] showed that bovine Lf inhibited amyloidogenic isoform of cellular prion protein (PrP(Sc)) accumulation in scrapieinfected cells in a time- and dose-dependent manner. Lf mediated the cell surface retention of normal cellular prion protein by diminishing its internalization and was capable of interacting with it as well as with PrP(Sc). In addition, Lf partially inhibited the formation of proteaseresistant prion protein. Lf present in the breast milk also protected against T. gondii infection [420], and current evidence indicate that a short duration of breastfeeding may constitute a risk factor for development of T1DM later in life [465].

Gastrointestinal Sensory-Motor Dysfunction Reported in Patients with Diabetes Mellitus may be at least in Part Caused by T. Gondii Infection

Diabetic patients

Gastrointestinal disturbances are common in diabetic patients and the entire gastrointestinal tract (GT) may be involved [466-469]. In patients with diabetic autonomic neuropathy (DAN), a number of abnormal conditions have been reported in different segments of the GT, such as esophagus (dysmotility), stomach (dysmotility, delayed emptying) and small and large intestine (dysmotility, delayed transit, bacterial overgrowth and diarrhea) [470] (Table 26). At present, it is believed that DAN is the major factor in the pathogenesis of these GT abnormalities and prolonged hyperglicemia play an important role in development of DAN through the glycation of the enteric nervous system [466,484-486]. Morphological and biomechanical changes as well as alterations of the enteric nervous system in experimental animals and patients with diabetes mellitus are presented in tables 27 and 28.

GT segment Motility disorders References
Esophagus Increased amplitude and number of peristaltic contractions [471-474]
  Increased number of spontaneous and non-propagated contractions [475]
  Decreased amplitude of lower esophageal sphincter pressure [471]
  Multi-peaked contractions [476, 477]
Stomach Decreased antral IMMC [478]
  Decreased postprandial antral activity and the number of antral contractions [479]
  Pyloric dysmotility [480]
Small intestine Decreased or increased frequency and amplitude of the antropyloroduodenal contractions [481]
  Increased duration of MMC cycle [482]
  Early recurrence of the MMC and clusters of contractile activity [483]
IMMC: Inter-digestive Migrating Motor Complex; MMC: Migrating Motor Complex

Table 26: Disorders of GT motility in patients with DM (according to Zhao et al. [466]; with own modification).

Morphological changes Biomechanical changes
Increased intestinal weight, length, weight per unit length Decreased opening angle and residual strain in duodenum
Increased surface area of mucosa Increased opening angle and residual strain in jejunum and ileum
Increased number of goblet cells per villus Increased circumferential stiffness of the intestinal wall
Increased smooth muscle mass Increased longitudinal stiffness of the intestinal wall
Increased different layer thickness Decreased stress relaxation of small intestine
Increased proliferating cell nuclear antigen  
Decreased volume of interstitial cells of Cajal  

Table 27: Morphological and biomechanical changes of small intestine in diabetic patients (according to Zhao et al. [466]; with own modification)

GT segment Type of abnormality Species References
Esophagus, stomach, intestine Loss of ICCs Human, mouse, rat [488]
Esophagus, stomach, intestine Diabetic gastroenteropathy Human, mouse, rat [489]
Stomach Gastroparesis, oxidative stress Mouse [490]
Stomach Gastroparesis, regional injury of ICCs Rat [491]
Stomach Gastroparesis Human [492]
Stomach Gastroparesis Human [493]
Stomach, intestine Oxidative stress Human, mouse, rat [494]
Duodenum Loss of enteric neurons Rat [495]
Duodenum, jejunum,
ileum, colon
Region-specific nitrergic neuronal loss, gastrointestinal motility disorders Rat [496]
Duodenum, cecum Loss of enteric neurons Rat [497]
Jejunum Decreased NO responsiveness and nNOS protein expression Rat [498]
Ileum Loss of enteric neurons Rat [499]
Small intestine Loss of enteric neurons, gastrointestinal motility disorders Human, mouse, rat [500]
Colon Loss of enteric neurons, gastrointestinal motility disorders, increased oxidative stress Human [501]
Colon Reduction in GFAP and neurotrophins Rat [502]
GT: Gastrointestinal Tract; ICCs, Interstitial Cells of Cajal; nNOS, Neuronal NO Synthase; GFAP, Glial Fibrillary Acidic Protein

Table 28: Alterations of the enteric nervous system found in experimental and clinical diabetes mellitus (according to Bagyanszki and Bodi,[487]; with own modification).

Diabetic rats

In animals, streptozotocin-induced model of diabetes was associated with development of serious pathophysiological abnormalities, including morphological changes in the gastrointestinal tract and quantitative alterations of the myenteric plexus neurons and other cells. For example, seven days after diabetes induction by streptozotocin, a significant reduction in the circumference and area of the colon (Table 29), size of the myenteric neurons and their nuclei (Table 30), and a decrease in the overall neuronal population as compared with controls, were demonstrated [504]. These changes were associated at least in part with marked body weight loss by the animals with diabetes [504]. Also, the number of myenteric plexus neurons per calculated area on the small and large stomach curvatures in diabetic rats examined after two months were found to be significantly decreased in diabetics compared with controls rats (Table 31). In addition, in the ileum, the relative percentage of the NADH-diaphorase positive myenteric neurons with the smaller area of cell body size in the ileum of diabetic rats increased and these with larger area decreased, in contrast to that found in control animals (Table 32). Izbeki et al. [496] also found that in the jejunum, ileum and colon of rats with streptozotocin-induced diabetes, both the total and the nitrergic neuronal cell number decreased significantly, while in the duodenum only the number of nitrergic neurons decreased. De Mello et al. [495] also demonstrated a significant reduction in density of the duodenal myenteric neurons stained with HuC/HuD compared with controls (18.6 and 19.77%, respectively, p<0.001). The density of nNOS neurons was also lower than that of controls (8.62 and 7.30%, respectively), but the difference did not attain statistical significance, which may suggest that the nitrergic neurons are less sensitive to acute diabetes induced by streptozotocin [495]. On the other hand, the density of NADHd-positive neurons in these groups was markedly higher than in control animals indicating that acute diabetes increases the activity of respiratory chain enzymes on these neurons, enhancing their staining [495] (Table 33).

Parameters Controls (n=8) Diabetics (n=8)
Length (cm) 16.33 ± 1.57 14.49 ± 0.61
Weight (g) 2.90 ± 0.42 2.51 ± 0.11
Circumference (cm) 2.60 ± 0.15 2.05 ± 0.10a
Area (cm2) 42.58 ± 5.04 29.91 ± 2.27a
aValues significantly different compared with controls (P < 0.05)

Table 29: Length, weight, circumference, and area of the colon in streptozotocin-induced diabetic rats (according to Furlan et al. [503]; with own modification).

Parameters Controls (n=8) Diabetics (n=8)
Total profile of the cell body (mm2) 219.20 ± 4.99 193.60 ± 4.32b
Nucleus profile (mm2) 81.88 ± 1.57 76.77 ± 1.31a
% of the cell profile occupied by the nucleus 39.89 ± 0.46 42.28 ± 0.42b
*Values significantly different compared with controls (ap< 0.05; bp< 0.001)

Table 30: Areas of cell body and nucleus profiles of the proximal colon myenteric neurons in streptozotocin-induced diabetic rats (according to Furlan et al. [503]; with own modification).

Rat No. Small curvature   Large curvature  
  Controls Diabetics Controls Diabetics
1 7743 4827 1331 962
2 6495 5544 1661 1172
3 8127 5172 2269 1770
4 9050 5018 1524 1032
5 7854 5784 1535 1458
Mean 7854 5269 1664 1279
SD 917 390 358 334
P   0.007319   0.008835

Table 31: Number of neurons per 11.6 mm2 areas of the small and large stomach curvatures in streptozotocin-induced diabetic rats (according to Fregonesi et al. [504]; with own modification).

Area of cell body size (μm2) Absolute (F) and Controls F relative (%)
%
frequency of Diabetics F neurons/group
%
<100 14 2.8 24 4.8
100-200 186 37.2 311 62.2
201-300 212 42.4 144 28.8
301-400 74 14.8 21 4.2
401-500 11 2.2 - -
> 501 3 0.6 - -
Total 500 100 500 100

Table 32: Changes of absolute (F) and relative (%) frequency of NADH-diaphorase positive myenteric neurons classified according to the area of their cellular body profiles in the ileum of diabetic rats (n=5 per group) 15 weeks after induction of the disease with streptozotocin (according to Alves et al. [505]; with own modification).

Technique Group of animals Density of neurons
HU Controls (n=5) 1472.80 ± 179.14
  Diabetics (n=5) 1198.80 ± 237.24a
  Insulin-treated rats (n=5) 1181.60 ± 179.53a
NADH Controls (n=5) 631.80 ±  5.18
  Diabetics (n=4) 820.0 ± 8.13a
  Insulin-treated rats  (n=4) 987.5 ± 19.69a
NOS Controls (n=5) 454.80 ± 59.41
  Diabetics (n=5) 415.60 ± 109.30
  Insulin-treated rats (n=5) 421.60 ± 48.22
Results are expressed as mean ± SD; ap<0.001. HuC/HuD,anti-human neuronal protein HuC/HuD identifies all neuronal cell bodies in the ganglion; NADHd, diaphorase positive

Table 33: Density of neurons (neurons/11.07 mm2) reactive to the HuC/HuD and nNOS immunohistochemical techniques and NADHd histochemistry of the myenteric plexus of the duodenum in adult rats with experimental acute diabetes (according to de Mello et al. [495]; with own modification).

T. gondii infection

Immunochemistry studies performed by Haroon et al. [506] showed that in infected neurons of chronically infected BALB/c mice upon oral infection with T. gondii cysts, not only parasitic cysts but also the host cell cytoplasm and some axons stained positive for the parasite antigen suggesting that parasitic proteins might directly interfere with neuronal function. It appeared that both bradyzoites and tachyzoites functionally silence infected neurons because the activitydependent uptake of the potassium analogue thallium was reduced in cysts harboring neurons, and the percentage of nonfunctional neurons increased over time in these animals. In vitro, live cell calcium (Ca2+) imaging investigations revealed that tachyzoites actively manipulated Ca2+ signaling upon glutamate stimulation leading either to hyper- or hyporesponsive neurons, and depleted Ca2+ stores in the endoplasmic reticulum [506].

In rats, after oral infection with T. gondii oocysts, Sant’Ana et al. [507] found a marked reduction in the number of goblet cells producing neutral mucins (PAS+) and sulphomucins (AB pH 1.0) as compared with control animals, and these changes reflected production of a more fluid mucous (Table 34). The number of vasoactive intestinal peptide (VIP-IR) submucosal neurons as well as the area of the VIPIR neuronal cell bodies also decreased significantly compared with control rats (667 ± 6.98 vs. 856 ± 14.89 per 1.74 mm2 of the jejunum, p<0.05, and 317.29 ± 9.28 vs. 404.24 ± 11.10 μm2, p<0.05, respectively). All these abnormalities indicated that oral T. gondii infection caused alterations in the chemical composition of the intestinal mucous and reduction in the number of submucosal neurons associated with atrophy of the remaining neurons in this cell subpopulation [507]. In this context, it is suggested that the morphological changes reported in duodenal atresia (a well known neonatal intestinal disease), such as the neuronal cells decreased in number and size, the circular musculature moderately-to-severely hypertrophic, and the interstitial cells of Cajal decreased even around the myenteric plexus [508], were due to chronic latent T. gondii infection. Also inflammatory lesions involving esophagus, stomach and duodenum frequently observed in neonates may be caused by the infection with the protozoan acquired prenatally [509-511]. Recently an association between T. gondii infection and development of abdominal hernia has been proposed [512]. More detailed gastrointestinal disturbances caused by chronic infection with the parasite in experimental animals and patients with several neurodegenerative diseases have been described elsewhere [434].

Animals IELs Goblet cells    
  HE PAS+ AB 1.0+ AB 2.5+
Controls 7.80 ± 1.65 21.30 ± 3.29 9.71 ± 1.22 7.10 ± 1.12
Infected rats 8.90 ± 1.42 18.60 ± 2.18a 5.70 ± 1.79a 6.20 ± 1.73
Values are means ± SD; aResults significantly different compared with controls (P <0.05); AB 1+ or 2.5+, Alcian blue pH 1 or 2.5; HE: Hematoxylin/Eosin; IELs: Intraepithelial Lymphocytes; PAS: Periodic Acid Shiff

Table 34: Changes in proportion of IELs and goblet cells/100 epithelial cells in the jejunum mucous tunica of rats 36 days post oral infection with 500 sporulated genotype 2 T. gondii ME-49 strain oocysts (according to Sant’Ana et al. [507]; with own modification).

Possible Links between Elevated Plasma Levels of TGF-ß1 in both T1DM and T2DM, and T. gondii Infection. Dual Role: TGF-ß Inhibits Inflammation and Autoimmunity, and Increases Intracellular Parasite Replication

Diabetes

TGF-β is a multi-functional cytokine with anti-inflammatory activities, such as inhibition of proliferation, maturation and/or activation of macrophages, lymphocytes and NK cells [513-516]. Smart et al [517] reported that TGF-β signaling is crucial for establishing and maintaining defining features of mature pancreatic β cells. Several authors demonstrated significantly increased plasma TGF-β1 levels in patients with NIDDM (7.9 ± 1.0 ng/ml vs. 3.1 ± 0.4 ng/ml, P<0.001; correlation with glycosylated hemoglobin (r2 = 0.42, P<0.001), women with T2DM, and women with prior history of gestational diabetes mellitus as compared with control individuals [518,519]. Elevated TGF-β1 concentrations were associated with retinopathy and neuropathy. It was interpreted as an anti-inflammatory response as these patients were known to have sublinical inflammation presumably triggered by hyperglycemia [519], although there might be also a concomitant T. gondii infection. Smoker diabetic patients showed also much higher plasma and urinary TGF-β1 levels than non-smoker diabetic individuals (12.6 ± 4.9 vs. 7.7 ± 4.7 ng/ml, P < 0.001; 27.5 ± 16.0 vs. 15.3 ± 6.3 ng/mg urinary creatinine, P = 0.01; and 15.3 ± 6.3 vs. 8.1 ± 4.4 ng/mg urinary creatinine, P<0.02, respectively) compared with control subjects [520]. These are important findings because TGF-β system mediated also diabetic renal hypertrophy and fibrosis build-up due to the extracellular matrix production [521,522], and TGF-β1 was found to induce vaginal tissue fibrosis in animal model [523], while the beta cell hypertrophy, beta cell damage and fibrosis, with reduction in insulin secretion, is characteristic for patients with T2DM [7]. In this context, a significant increase of plasma TGF-β1 levels caused by latent T. gondii infection may play a key role in development of pancreatic islet beta cell abnormalities found in T2DM.

Tonkin & Haskins [524] demonstrated that regulatory T cells (Tregs) transfer causes a reduction in the number of effector TH1 T cells and macrophages, and also inhibits effector T cell cytokine and chemokine production. Transfection of effector T cells with a dominant negative TGF-β receptor showed that in vivo suppression of diabetes by TGF-β-induced Tregs is TGF-β-dependent [524]. Anti-islet autoimmunity can be inhibited by transfer of „natural” CD4+CD25+ Tregs [525-528], or by induced Tregs which upregulate the Treg transcription factor Foxp3 after activation of CD4 T cells in the presence of TGF-β [524,529,530]. However, in pregnant mice, the infection of T. gondii caused the decrease of CD4+CD25+-regulatory T cells [235].

TGF-β plays a critical role in the suppression of lymphocyte proliferation and differentiation therefore preventing hazardous autoimmune responses, and its immunosuppressive effects are mediated through the inhibition of TNF-α and IL-1 [531] and blocking the induction of adhesion molecules like ICAM-1 and VCAM-1 [519,532,533]. Filisetti & Candolfi [534] reported that TGF-β is well known for its immunosuppressive action on leukocyte cell lines. This cytokine was found to be an antagonist of TNFα, TNF-β, IFN-γ and IL-2 [130,221]. The antiinflammatory action of TGF-β control development of immunopathological processes related to TH1 immune response in the brain [535] and the intestines [536]. However, TGF-β was reported to increase in vitro replication of T. gondii in retinal cells, suggesting that this cytokine may be involved also in immunopathological phenomena [537]. Elevated expression of TGF-β in vitreous, retina and retinal pigment epithelium has been correlated closely with retinal fibrosis and choroidal neovascularization [513]. Thus, the development of pancreatic islet β cells fibrosis characteristic for the patients with T2DM may be associated with the increased levels of this cytokine due to T. gondii infection because TGF-β belongs to biomediators favouring growth of the parasite [538].

T. gondii infection

Normal pregnancy is characterized by a preferentially TH2 immune response, with the production of antiinflammatory cytokines, such as IL-4, IL-5, IL-10 and TGF-β by both maternal and fetal cells [539-543]. Host protection to T. gondii infection involves TH1 type immune response of inflammatory cells, lymphocytes and macrophages with enhanced production of IFN-γ, TNF-α, and IL-1β [534,544]. Activated macrophages by IFN-γ inhibit parasite replication through a number of potent microbicidal mechanisms such as oxidative [545] and non-oxidative [546] mechanisms as well as the induction by IFN-γ of IDO that degrades tryptophan, which is required for the parasite replication [547]. Barbosa et al. [539] showed that in contrast with HeLa cells, treatments with IL-10 or TGF-β1 induced a considerable augmentation in both T. gondii intracellular replication and invasion into BeWo cells. BeWo trophoblasts were unable to control replication of the parasite even in the presence of exogenous IFN-γ [548]. In addition, treatment with IFN-γ alone or associated with IL-10 or TGF-β1 increased the same parameters in BeWo cells, whereas the opposite effect was observed in HeLa cells. When endogenous IL-10 or TGF-β was blocked, both BeWo and HeLa cells were able to control the parasite infection only in the presence of IFN-γ. It was suggested that the higher susceptibility of BeWo cells to T. gondii may be due to immunomodulation mechanisms, suggesting the role of trophoblast cells in maintaining a placental microenvironment favourable to pregnancy may facilitate the infection into the placental tissues [539].

Hunter et al. [130] demonstrated that TGF-β antagonizes the ability of IL-12 to stimulate production of IFN- by splenocytes from SCID mice, and suggested a role for TGF-β in regulation of T cellindependent resistance to T. gondii. Malipiero et al. [549] found that TGF-β is a potent deactivator of polymorphonuclear leukocytes (PMN) and macrophages since it suppresses the production of ROS, RNI and IL-1. TGF-β impairs expression of L-selectin on PMN and L-selectin is known to be essential for PMN recruitment in bacterial meningitis. On peripheral monocytes TGF-β is chemotactic, enhances phagocytosis, activates the production of cytokines – IL-1, TNF-β, and leads to increased expression of several integrin receptors. On tissue macrophages including microglia, the cytokine was found to inhibit phagocytosis and the production TNF-α, IL-1, IL-6, ROS, and to induce increased expression of IL-1 receptor antagonist [531,549-552].

Activation of macrophages plays an important role in the host resistance against intracellular pathogens. Langermans et al. [221] found that the IFN-γ-induced toxoplasmastatic activity of macrophages was inhibited by TGF- (mean fold increase = 6.3), which was also found for the IFN-γ-induced production of TNF-α, RNI and PGE2 by macrophages. It was found that PGE2, which has macrophage deactivating properties, was not involved in the inhibition of macrophage activation by TGF-β. It appeared that inhibition of TNF- production was a key factor in the TGF-β-induced suppression of macrophage activation with respect to toxoplasmastatic activity and RNI production [221].

Bogdan & Nathan [553] found that TGF-β can induce resting human monocytes to produce TNF, IL-1, and IL-6. It was found that IL-10 was about 25-fold more potent suppressor of LPS-induced TNF production by mouse macrophages than was TGF-β. TGF-β suppressed TNF release on a translational level. TGF-β, IL-4, and IL-10 have been shown to have strong macrophage-deactivating effects.

Seabra et al. [554]. Activated macrophages control growth by NO production. However, T. gondii active invasion inhibits NO production, allowing parasite persistence. The mechanism used by T. gondii to inhibit NO production persisting in activated macrophages depends on phosphatidylserine exposure. TGF-β1 led to iNOS degradation, actin filament (F-actin) depolymerization, and lack of NF-κB in the nucleus [554].

Nagineni et al. [513] reported that in human retinal pigment epithelial cultures TGF-β enhanced parasite replication. Soluble extracts of T. gondii stimulated secretion of both TGF-β1 and TGF-β2 significantly. T. gondii infection completely inhibited secretion of the active form of TGF-β2. Finally, Malipiero et al. [555] found that endogenous TGF-β suppresses host defense against pathogen infection also in the central nervous system. Thus, it seems that the increased levels of plasma TGF-β reported in the patients with T1DM and T2DM exert both beneficial and harmful effects because although this cytokine is an important regulator of pancreatic islet development and has antiinflammatory and immunosuppresive activities, at the same time increases T. gondii replication in the host cells with further development of various pathophysiological irregularities.

Beneficial Effect of Thermal Therapy on Glycemic Control in the Patients with T2DM may be Associated with Stage Conversion of T. gondii Tachyzoites to Bradyzoites and Increased Generation of NO by Endothelial NOS

Recently, it was reported that regular thermal therapy might promote insulin sensitivity while boosting expression of eNOS [556], and that control of glycemia was improved in the patients with T2DM receiving regular hot tub treatment [557]. Moreover, it was demonstrated that vascular endothelial constitutive isoform of NO synthase (eNOS) has been induced in cultured endothelial cells and cardiomyocytes exposed to mild heat (42°C) [558,559]. Endothelial eNOS mRNA expression and NO production also augmented about 40% in hamsters given daily hot treatments (15 min of infrared sauna) that increased core temperature by about 1°C [560,561]. In the patients with coronary risk factors who underwent daily sauna treatment for two weeks (15 min of 60°C infrared sauna followed by 30 min covered with blankets) endothelium-dependent vasodilation was found to increase markedly [562]. Recently, it was also found that in mice thermal induction and overexpression of hsp72 might counter high fat diet-induced insulin resistance [563]. Several studies [564,565] showed that a physiologically relevant hyperthermia (39°C) selectively induced constitutive hs-hsp70 (hsc70) in H9c2 cardiac myoblasts and conferred oxidative protection. Hsc70-enriched cells exhibited a marked resistance to oxidative challenge, including exposure to hydrogen peroxide, hydroxyl radical, and hypoxia/reoxygenation [565]. These are important findings because, for example, the seropositivity rate for anti-T. gondii IgG antibodies among patients with chronic heart failure was significantly higher than in healthy volunteers (68% vs. 36%, (P < 0.05), respectively) [566], and infection with the protozoan is associated with oxidative stress [209].

An increase in heat shock proteins 60, 70 and 90 (Hsp90), formation in T. gondii was demonstrated in bradyzoites on conditions which induce stage conversion, including increased temperature [567-569], and Hsp60 contributed to protection against the parasite infection [570]. Moreover, HSPs have been found to play a key role in the induction of a cellular immune response, including activation of NK cells by HSP70, which comprise 5-20% of peripheral blood mononuclear cells and are important in the control of bacteria, parasites and viruses [571]. Febrile temperatures (41°C) resulted in a synergistic increase in Hsp90 and Hsp70 synthesis induction [572,573], and a temperature-controlled shift from oligomeric complexes to smaller species with increasing temperature was found for small HSPs with low molecular mass of about 12-43 kDa [574] (Table 35).

RH tachyzoites, ME49 strain of T. gondii. ESP molecular weight (kDa)a Temp. (°C) 4   25   37   42
110     + +
97     + +
86     + (Tg386) +
80     + (Tg485) +
70 + + +  
60     + +
54 + + +  
42     + (Tg786) +
40     + +
36 + + + (Tg378)  
30 + + +  
28 + + + (Tg556)  
26 + + +  
22 + + +  
19 + + +  
aThe RH strain of T. gondii was maintained by peritoneal passages in Balb/c mice. For positive reference serum, mouse was infected with ME49 strain of the parasite for 8 weeks until the animal had brain cysts postmortem. The molecular mass of 15 ESP was estimated at 37°C. Among them,110, 97, 86, 80, 60, 42 and 40 kDa proteins were released temperature-dependently, while those of 70,54,36,30,28,26,22,and 19 kDa were released temperature-independently as low as 4°C. Five ESP of 86, 80, 42, 36 and 28 kDa reacted with monoclonal antibodies: Tg378 and Tg556 clones were detected 36 kDa and 28kDa proteins, respectively, in dense granules with involvement into parasitophorus vacuole, and these ESP were released regardless of temperature and time. Tg386 clone labeled presumably micronemal structure in tachyzoites, Tg485 clone labeled surface membrane protein; while Tg786 clone labeled probably rhoptry in the apical portion. ESP by Tg786 clone was released continuously with increment, whereas those by Tg378 and Tg556 clones were ceased to release after 3 and 4 hrs changes, respectively

Table 35: Profile of Excretory/Secretory Proteins (ESP) released from purified tachyzoites of T. gondii incubated for 1 hr at different temperatures and then analyzed by monoclonal antibodies (Son and Nam [575]; with own modification).

Thus, the beneficial effects of thermal therapy reported in patients with T2DM may be associated with the changes in the stage of T. gondii during latent chronic pancreatic toxoplasmosis because bradyzoites to tachyzoites interconversion can be due to a variety of factors, including temperature-dependent HSPs induction (Table 36). This reasoning may be supported by the following findings: a) in mice a mutagenized strain tachyzoites of T. gondii showed variable growth at temperatures between 34 and 39°C, and inability to grow at 40°C, which correlated with a loss of virulence [583]; b) the reports on the parasite-induced changes in human behavior, including decreased psychomotor performance in individuals with latent asymptomatic toxoplasmosis [584]; and c) the fact that T. gondii may circulate also in the peripheral blood of immunocompetent persons with acute and chronic toxoplasmosis [323]. Virulence of the parasite is associated with distinct dendritic cell responses and reduced numbers of activated CD8+ T cells [585]. It seems, therefore, that marked changes in the innate immune state associated with an improvement of balance between various proinflammatory and antiinflammatory cytokines and other inflammation mediators caused by fever probably resulted in T. gondii tachyzoite (representing a subacute persistent stage of cerebral toxoplasmosis) bradyzoite (representing a chronic stage) interconversion and/or apoptosis in tachyzoites, finally beneficially affecting glycemic control of some patients with T2DM.

Tachyzoite to bradyzoite conversion Bradyzoite to tachyzoite conversion
High pHa Lack of NO
Low pHa Lack of IFN-γ
Heat shockb Lack of TNF-α
Mitochondrial inhibitionc Lack of IL-12
Presence of NOc Lack of T cells
Elevation of both cAMP and cGMPd Induction of a variety of HSPs,including HSP70, is associated with bradyzoite transitione
aOne cannot exclude that the differences in therapeutic efficacy between valproic acid and sodium valproate used in several neuropsychiatric diseases were related to acidic or alkaline target local tissue conditions induced by these two pharmaceutical forms of a drug (low doses vs. high doses, respectively) [577,578]. bPhysiologically relevant circumstances that could play a role in stage conversion in vivo include heat shock through a fever [576]. cNO overproduction in ASD individuals [579] is an inhibitor of mitochondrial function [106]. dStress-induced elevation of cAMP could play a role in bradyzoite induction because addition of cAMP or cGMP to tachyzoites can stimulate stage conversion [580]. PLK, a T. gondii ME49 clonal strain able to differentiate in vitro, exhibited a rise in cAMP in response to bradyzoite inducing conditions, but elevation of cAMP under the same conditions was not evident in RH, a strain that does not differentiate well [580]. It must be emphasized that inducers of oxidative stress (nb. a state characteristic for autistic patients) also have been demonstrated to cause parasite encystment in vitro [581,582]

Table 36: Factors associated with tachyzoite and bradyzoite interconversion (according to Lyons et al. [576]; with own modification).

Vitamin D Deficiency is Linked with Development of Diabetes Mellitus. Protective Role of Vitamin D may be Partly Due to its Immunomodulatory and Antitoxoplasmatic Activities

Diabetes

Sørensen et al. [586] reported a trend toward a higher risk of T1DM with the lower serum levels of vitamin D during pregnancy (the odds of the disease was more than 2-fold higher for the offspring of women with the lowest levels of 25-OH D compared with the offspring of those with levels above the upper quartile).

Evidence exists that patients with T1DM and T2DM have a higher incidence of hypovitaminosis D [587], and vitamin D deficiency has been associated with increased risk of T1DM. Vitamin D deficiency in early life accelerates T1DM in non-obese diabetic mice [588]. Children and adults need at least 1000 IU of vitamin D per day to prevent deficiency when there is inadequate sun exposure [589]. Interestingly, BCG vaccinated infants were almost six times (CI: 1.8-18.6) more likely to have sufficient plasma vitamin D concentrations than unvaccinated infants [590]. It appeared that Mycobacterium tuberculosis purified protein induced a significant increase of several immune factors in adolescents, including IFN-γ, TNF-α, IL-2, IL-6, IL-10, IL-17, GMCSF, MIP1α, and IP-10 when compared to paired samples taken prior to BCG vaccination (P<0.0025) [591].

Moreover, intranasal vaccination with mycobacterial 65-kD heat shock protein (HSP) prevented development of insulitis and diabetes in non-obese diabetic mice [592], and DNA vaccine containing the mycobacterial hsp65 gene protected mice from streptozotocin-induced insulitis and diabetes [593]. These findings may be explained by the important role of HSPs acting as molecular chaperones in protection from and pathogenesis of infectious diseases [434,568,594]. Hisaeda & Himeno [595] showed that the expression of host-derived 65 kDa HSP was crucial in directing host immune system to achieve protective immunity against infection with T. gondii. A relationship was found between the biomolecule expression on/in host macrophages and development of immune defense against the parasite, regardless of differences in strains and forms of the protozoan (Tables 37 and 38).

Infected parasite Host statusa Resistance to infectionb Expression of HSP65c
Beverly strain Bradyzoite Non-immune + +
Bradyzoite Immune with sonicated T. gondii ++ ++
Tachyzoite
After in vivo passage
Non-immune - -
RH strain      
Tachyzoite Non-immune - -
Tachyzoite Live-vaccinated with a low dose of the Beverly strain ++ ++
aBALB/c mice were used as hosts; bSymbols used here represent resistance to infection; +: resistant; ++: very resistant; -: susceptible; cSymbols used here represented levels of HSP65 expression as follows: +: strong; ++: very strong; -: none

Table 37: Relationship between HSP65 expression and resistance to infection with T. gondii (according to Hisaeda and Himeno [595]).

Hosta Expression of HSP65b Resistance to infectionc
CB17 scid/scid - -
   + Fetal thymus graft + +
   + Fetal liver cell transfer ++ ++
BALB/c nu/nu ± -
BALB/c +/+ ++ ++
   + Anti-TcR αβ +++ +++d
   + Anti-TcR γδ - -
aTcR: T-cell receptor; bSymbols represent as follows: -: none; ±: very weak; +: weak; ++: strong; +++: Very strong; cInfection with bradyzoites of the Beverly strain of T. gondii. Symbols represent as follows: -: Susceptible; +: weakly resistant; ++: resistant; +++: strongly resistant; “++” here (also expression level) is comparable with “+” in Table 34. dEspecially at the early phase of infection. Scid, severe combined immunodeficiency; nu/nu, nude mice

Table 38: Requirements of T cells for HSP65 expression (according to Hisaeda and Himeno [595]; with own modification).

Vitamin D treatment has been shown to improve, and even prevent, development and/or clinical course of T1DM in both humans and in animal models [596]. Pancreatic islet insulin-producing betacells as well as numerous cell types of the immune system express the vitamin D receptor and vitamin D-binding protein. Some organs have the capacity to metabolize 25-hydroxyvitamin D to its active form 1,25-dihydroxyvitamin D, which has a potent immunomodulatory activity that also enhances the production and secretion of several hormones, including insulin [589]. Pharmacologic doses of 1,25(OH)2D prevented insulitis and T1DM in nonobese diabetic mice [597] and other models of T1DM, possibly by immune modulation, such as for example, increased monocyte differentiation to macrophages, thus increasing their cytotoxic activity, reduced the antigen-presenting activity of macrophages to lymphocytes, prevented dendritic cell maturation, decreased proliferation of activated lymphocytes, inhibited T lymphocyte-mediated immunoglobulin synthesis in lymphocyte B cells, delayed-type hypersensitivity reactions, and generation and activity of NK cells [598-603], as well as by direct effects on beta-cell function. It should be noted that vitamin D deficiency was found to be associated with retinopathy in children and adolescents with T1DM [604]. The prevalence of this clinical entity was higher as compared with the vitamin D sufficient patients (18 vs. 9%, P = 0.02; OR 2.12 [95% CI 1.03-4.33]), and was dependent of diabetes duration (1.13, 1.05-1.23), and HbA1c levels (1.24, 1.02-1.50) [604]. This is an important finding because retinopathy and other abnormalities were frequent complications reported in individuals with ocular toxoplasmosis [605-607], and cataracts have been demonstrated also in both streptozotocin-induced diabetic rats [608] and T1DM pediatric population [609]. Tedesco et al. [607] demonstrated free parasites in the retinal vasculature, the presence of mononuclear inflammatory infiltrate and parasites in the vasculature of choroids in infected eyes. It was suggested that the increased levels of histamine found in the retina and choroid of diabetic rats may enhance permeability of local vascular bed and participate in development of diabetic ocular complications, including lens opacities [610].

T2DM involves impaired pancreatic β cell function, insulin resistance and inflammation [611]. In T2DM, several disturbances in concentrations of systemic inflammation mediators have been demonstrated, including proinflammatory cytokines and other factors: IL-2, IL-6, IL-12, IFN-γ, TNF- and TNF-β, C-reactive protein, and plasminogen activator inhibitor-1 [603,612,613]. Vitamin D supplementation can increase insulin sensitivity and decrease inflammation in the patients with T2DM [587]. Studies showed that hypovitaminosis D was associated with an enhanced inflammatory response manifested as significantly increased serum TNF-α, IL-6, and CRP levels in healthy [613-616] and obese persons [617,618]. Moreover, in some clinical states associated with inflammation, vitamin D supplementation caused a marked decrease in serum levels of these proinflammatory factors [619,620] and an increase in antiinflammatory cytokine IL-10 concentration [621]. 1,25(OH)2D3, vitamin D3 analog, exerted direct action on purified mouse Langerhans cells reducing IL-10 production and enhancing the production of IL-6 and IL-12p40 upon activation by CD40 ligation [622]. In addition, 1,25(OH)2D3 upregulated the production of IL-1β, CCL3, CCL4, and CCL5. The generation of TH2-type chemokines, represented by CL17 and CCL22 was inhibited, whereas IFN-γ-triggered production of TH1- type chemokines represented by CXCL9, CXCL10, and CXCL11, was increased [622]. It was reported that daily intake of 2000 UI vitamin D was associated with improved β cell function [623]. Moreover, results of a cross-sectional analysis showed that patients with serum 25OH D ≥ 80 nmol/l levels had reduced risk of developing T2DM when compared to those individuals who had ≤ 37 nmol/l [624].

Vitamin D is important for insulin synthesis and release because of the presence of both 1-α-hydroxylase and vitamin D receptor in pancreatic β cells [587,625]. Vitamin D is also involved in insulin sensitivity by controlling calcium flux through the membrane in both β cells and peripheral insulin-target tissues [611,626]. The opening of voltage-sensitive Ca2+ channels allows Ca2+ uptake by β-cell, thereby contributing to secretion of insulin [97]. Evidence exists that vitamin D has a potential antimicrobial activity and therefore may reduce the risk of various infections through multiple mechanisms [627]. Dendritic cells (DCs) are primary targets for the immunomodulatory activity of 1,25(OH)2D3, as indicated by inhibited DCs differentiation and maturation, leading to down-regulated expression of MHC-II, costimulatory molecules and IL-12. Inhibition of this proinflammatory cytokine production by 1,25-(OH)2D3 is associated with downregulation of NF-κB protein levels in activated lymphocytes [628,629]. 1,25-(OH)2D3 dose-dependently inhibited LPS-induced cytokines production in PBMC modulating intracellular calcium [630]. In addition, this active metabolite may protect against oxidative injuries caused by the NO burst because it dose-dependently inhibited iNOS messenger RNA expression of the LPS-stimulated RAW 264.7 cells and also significantly reduced the gaseous NO release and OONOproduction [631]. Moreover, 1,25(OH)2D3 enhances IL-10 production and promotes DCs apoptosis [632], as well as increases PGE2 production by monocytes, a mechanism which partially accounts for the antiproliferative effect of 1,25-(OH)2D3 on lymphocytes [633]. This metabolite has a direct effect on naive CD4+ T cells to enhance development of TH2 cells [634], increases regulatory T-cells and arrests autoimmune diabetes in NOD mice [635]. These actions emphasize the plethora of general benefits of vitamin D and its active metabolite in immunomodulating and antimicrobial mechanisms, thus favoring the host in courtailing present and imminent infections.

T. gondii

In acute toxoplasmosis, 1,25(OH)2D3 reduced survival rate of infected mice compared to untreated animals, and significantly decreased serum IFN-γ and IL-12p40 concentrations indicating inhibition of TH1-type cytokines, as well as reduced CD4+ T lymphocyte and splenocyte counts, thus enhancing host sensitivity to T. gondii infection [636]. Surprisingly, no increase in parasite load was observed in the organs, which suggested an inhibitory effect of 1,25(OH)2D3 at a cellular level [636], like previously it was reported for Plasmodium faciparum [637]. Further studies showed that treatment with vitamin D dose-dependently inhibited both in vivo and in vitro growth of T. gondii intracellularly, possibly by limiting tachyzoite proliferation within the parasitophorous vacuole because of activity at the cellular level [627,638]. Ghaffarifar et al. [639] demonstrated that in RPMI 1640 cell culture vitamin D3 (1000 IU) similarly like IFN-γ (100 IU) significantly decreased proliferation of T. gondii (RH stran) tachyzoites per infected peritoneal macrophage of BALB/c mice as compared with control animals (Table 39). It should be noted that the in vitro inhibiting effect of vitamin D3 alone on tachyzoite proliferation, as well as the increase of NO generation by macrophages, were more distinct than the respective effects of IFN-γ [639]. This emphasizes the importance of NO activity against T. gondii tachyzoites in the infected cells (Table 40), despite the fact that T. gondii partially inhibits NO production of activated murine macrophage [194]. 1,25(OH)2D3 also induced NO synthesis and suppressed growth of Mycobacterium tuberculosis, another intracellular microorganism, in a human macrophage-link cell line [640]. These findings are consistent with the beneficial effects of pretreatment with 1,25(OH)2D3 (0.5 μg/kg for 2 days) on various tissue pathological changes caused by peritoneal administration of oocysts in mice and histologically examined after seven days post inoculation [638]. It appeared that 1,25(OH)2D3 reduced tissue damage and parasite load in situ, and in particular the difference of the number of parasites per 1 mg of standardized tissue DNA was significant in the spleen (Table 41). Thus, vitamin D immunomodulatory and antitoxoplasmatic activities were probably at least in part responsible for inhibition of diabetes development in both patients with T1DM and T2DM.

Experiment No. Controls Solventa Vit D3
(1000 IU)
IFN-γ
(100 IU)
Vit D3(1000 IU) plus IFN-γ (100 IU)
1 3.01 ± 0.14 2.93 ± 0.16 2.49 ± 0.19b 2.6 ± 0.2b 2.37 ± 0.19b
2 3.15 ± 0.12 3.03 ± 0.16 2.74 ± 0.16 2.5 ± 0.15b 2.58 ± 0.13b
3 3.05 ± 0.15 3.04 ± 0.14 2.82 ± 0.17 2.57 ± 0.16b 2.69 ± 0.2b
4 3.16 ± 0.14 3.0 ± 0.14 2.39 ± 0.19b 2.59 ± 0.2b 2.03 ± 0.19b
Numbers of tachyzoites are given as a mean ± SD; aEthanol 95; bStatistically significant differences compared with controls (P 0.05)

Table 39: Effect of vitamin D3 and IFN-γ on proliferation of T. gondii (RH strain) tachyzoites per infected peritoneal macrophage of BALB/c mice after incubation for 96 hrs in RPMI1640 cells culture (according to Ghaffarifar et al. [639]; with own modification).

Experiment No. Controls Solventa Vit D3
(1000 IU)
IFN-γ (100 IU) Vit D3 (1000 IU) plus IFN-γ (100 IU)
1 109 ± 8.02 108.2 ± 12.45 165 ± 11.30b 146 ± 7.22b 187.8 ± 9.82b
2 108 ± 9.46 108.9 ± 6.93 121.2 ± 6.68 139.5 ± 5.76b 136.2 ± 10.21b
3 109.6 ± 7.35 108.2 ± 4.96 139 ± 7.01b 146 ± 4.93b 146.9 ± 9.62b
4 109 ± 7.03 108.6 ± 4.26 166 ± 7.01b 146.2 ± 5.60b 191.5 ± 9.62b
Values are given as mean ± SD. aEthanol 95. bStatistically significant results compared with controls (P ≤ 0.05). NO production was estimated as a nitrite release from infected macrophages (μM/ml)

Table 40: Effect of vitamin D3 and IFN-γ on NO production by peritoneal macrophages of BAL: B/c mice infected with T. gondii (RH strain) after incubation for 24 hrs in RPMI1640 cells culture (according to Ghaffarifar et al. [639]; with own modification).

Tissue Pathology No treatment Treatment with Vit D3
Lung Alveolar macrophages 1 0
  Inflammatory foci 2 1
Liver Inflammatory foci 3 2
  Hemorrhage 2 0
  Mitosis 1 0
Small intestine Inflammatory infiltrates 1 0
  Necrotic mucosal cells 2 1
Brain Presence of the parasite 2 0
Spleen Granulocytes 2 1
Histopathologic examination of the tissues was performed 7 days post inoculation. Numbers are based on severity of the lesions (0, no lesion, 1, mild, 2, slight, 3, moderate changes) and the total was divided the number of animals in the group. Also, in vitro studies with incubated intestinal epithelial cells showed a significant dose-dependent inhibition of intracellular T. gondii tachyzoites (RH strain, type I) proliferation at 10-7 mol/l of 1,25(OH)2D3 concentration

Table 41: Effect of pretreatment with 1,25(OH)2D3 (0.5 μg/kg/2 days) on tissue pathology caused by T. gondii avirulent ME49 strain infection with 20 cysts administered intraperitoneally in BALB/c mice (according to Rajapakse et al. [638]; with own modification).

Beneficial Role of the Increased Indoleamine 2,3-Dioxygenase (IDO) Activity for both Diabetes and T. gondii Infection Prevention/Treatment

Diabetes

IDO may exert an immunoregulatory function and has the capacity to affect the course of various infections, autoimmunity, cancer and transplantations [6]. Increasing evidence support suggestions that IDO may delay the onset and progression of autoimmune diseases, e.g. IDO expressing NK cells contributed and promoted acceptance of rat liver allograft [641]. Jalili et al. [642] demonstrated the long survival and viability of syngeneic islets exposed to IDO-expressing fibroblasts within the composite grafts in a diabetic animal model. It was also found that transient up-regulation of IDO in dendritic cells by human chorionic gonadotropin down-regulated autoimmune diabetes [643]. Nb. it should be noted that IDO production by human dendritic cells results in the inhibition of T cell proliferation [644]. In addition, human chorionic gonadotrophin administration markedly inhibited T1DM onset in NOD female mice in an IDO-dependent fashion. Also, it was found that a defect in tryptophan catabolism impaired tolerance in NOD mice [643], and previously it was reported that IFN-γ blocks the growth of T. gondii in human fibroblasts by inducing the host cells to degrade tryptophan [161]. Moreover, functional IDO was induced when human islet were treated with IFN-γ [643], and otherwise it is known that IFN-γ is the key cytokine responsible for development of immune defense against T. gondii in all infected tissues and cells, including the central nervous system [161,231]. IDO is induced in the mouse brain in response to peripheral administration of LPS and superantigen [645], although LPS induction of IDO is mediated dominantly by an IFN-γ-independent mechanism [646]. These findings are important for diabetic patients with T. gondii infection because their metabolic-cytokine responses to a second immunological challenge might be excessive [647]. Recently, Fallarino et al. [648] provided promising evidence for treatment of T1DM using IDO expressing encapsulated Sertoli cells, and it appeared that IDO mediated TLR9- driven protection from experimental autoimmune diabetes induced in C57BL/6 mice by streptozotocin. In wild type animals, the disease was accompanied by up-regulation of IDO in pancreatic lymph nodes and would be greatly exacerbated by in vivo administration of an IDO inhibitor [648,649].

It should be emphasized that the increased IDO activity causes acceleration of tryptophan metabolism that results in enhanced generation of melatonin (Figure 1), a neuroimmunomodulator produced also by the pineal gland, retina, gut and immunocompetent cells including both bone marrow cells [650] and lymphocytes [651]. Melatonin is a free radical and peroxynitrite scavenger [652-654], and exerts anti-inflammatory effects, including inhibition of NF-κB activation [653,655,656], prevention of iNOS expression and direct inhibition of catalytic activity of NOS [657], decrease malondialdehyde production and increase glutathione peroxidase activity [658-660]. Also two melatonin metabolites, N1-acetyl-N2- formyl-5-methoxykynuramine and N1-acetyl-5-methoxykynuramine, exerted potent anti-inflammatory and antioxidant effects [661,662]. The protective biological effects of melatonin on oxidative stress and inflammatory processes in both T1DM and T2DM, are important for reducing pancreatic β-cell damage caused by persistent autoimmune state and excessive production of proinflammatory cytokines during insulitis, eventually associated with chronic T. gondii infection. Interestingly, maternally administered melatonin differentially regulated LPS-induced proinflammatory and anti-inflammatory cytokines in maternal serum, amniotic fluid, fetal liver and brain [663].

On the other hand, melatonin was found to decrease insulin secretion specifically both in vitro/in vivo [664-666] and type 2 diabetic rats, as well as patients, exhibited decreased melatonin levels, whereas the levels in type 1 rats were increased [666]. Peschke et al. [666] suggested that catecholamines, which decrease insulin levels and stimulate melatonin synthesis, control insulin-melatonin interactions because the amines were increased in T1DM but were diminished in T2DM. This difference may be at least in part explained by the increased protective/defensive body requirements for this hormone in T2DM that resulted in its shortage in the host, as compared with T1DM, time of exposition to triggering factor(s) and underlying molecular and metabolic disturbances in these two clinical entities.

Finally, calcium ion is necessary for insulin exocytosis and -cell glycolysis, both processes important in signaling circulating glucose concentration [667]. Calcium also plays an essential role in T. gondii motility, enhanced invasion of the parasite to host cells and its increased intracellular replication [668,669]. Therefore, the competition for calcium may contribute to its shortage in islet β-cells and participate in the development of hypoinsulinemia.

T. gondii infection

Increased levels of many tryptophan metabolites have been reported in several neurodegenerative disorders and were postulated to be secondary to induction of IDO and other enzymes of the L-tryptophan-kynurenine pathway. Inhibition of the increased IDO activity significantly exacerbated diseases scores and neuronal cell death in various central nervous system disorders [670]. Moreover, L-tryptophan degradation by IDO might have an important role in IFN-γ-induced antimicrobial effects [671]. Local accumulation of kynurenine metabolites, in particular, quinolinic acid, following IDO induction may represent a potentially detrimental event because quinolinic acid is a potent excitotoxin, and its overproduction has been linked to neuronal damage occurring in brain inflammation [672], initiation of lipid peroxidation [673], and development of disturbances in gluconeogenesis in the liver [674]. Spekker et al [675] proposed that IDO is responsible for the suppression of Neospora caninum growth, and other studies on T. gondii suggested that its growth could be contained when certain immune cells including dendritic cells were actively expressing IDO [163]. Moreover, L-tryptophan-L-kynurenine pathway metabolism accelerated by T. gondii infection was found to be abolished in IFN-γ-gene-deficient mice, and an antitoxoplasmatic mechanism of cross-regulation between iNOS and IDO that may vary among tissues, was demonstrated [671] (Figure 1). Thus, IDO and iNOS are involved in the immunomodulatory roles of IFN-γ, and evidence suggests that these functionally cross-regulated pathways [671] may play an important role in prediction and treatment of autoimmune diseases, particularly T1DM [6]. Dominant control of the regulatory T cells functional status and blocking their conversion into TH17-like T cells in response to inflammatory stimuli may markedly contribute to these bioactions [191].

Prevention of Hypoglycemia-Induced Neuronal Death by Minocycline may be Partly Associated with its Antimicrobial Activity against T. gondii Infection

Hypoglycemia and minocycline

Recently, Won et al [208] demonstrated that minocycline treatment markedly reduced neuronal death induced by hypoglycemia and cognitive impairment associated with this clinical state was also significantly prevented. Hypoglycemia was reported even in the patients with T1DM and T2DM who were strictly monitoring their blood glucose levels [208,676,677], and may be associated with serious complications, such as deterioration of mental efficiency, focal neurological deficits, seizure, and neuronal death [678,679]. Interestingly, it was suggested that hypoglycemia-induced neuronal death is not only a result of markedly impaired glucose supply to the brain [680,681], but also other contributing factors are involved, including sustained stimulation of glutamate receptors, NADPH oxidase activation with enhanced ROS generation, and extracellular zinc release [682-684], which plays an important role in mediating positively gene expression and production of cytokines IL-2 and IFN-γ in the Th1 cell line and negatively TNF-α, IL-1, and IL-8 in the monocyte-macrophage cell line [685]. In experimentally induced toxoplasmosis, zinc added to diet stimulated cellular immunity, increased CD8 and total number of lymphocytes [686].

T. gondii infection

Minocycline, a tetracycline broad-spectrum antibiotic, was developed as an antimicrobial drug for treatment of various infectious diseases, including T. gondii infection [687,688]. This antibiotic showed bacteriostatic effects limiting the growth of bacteria, inhibited production of proinflammatory cytokines, MMP-9 and activation of peripheral/central immunocompetent cells, including T cells, macrophages, and microglia [689]. Mice chronically infected with a low virulent strain Me49 of the parasite showed a significant reduction in the number of brain cysts after three weeks of treatment with 50 mg/ kg per day of minocycline. Therapy of the infected animals with the chemotherapeutic (100 mg/kg/day for 12 days) increased their survival and cure rates [687]. Moreover, in acute and chronic neurological disease animal models, including Parkinson’s [690], Huntington’s [691] and Alzheimer’s disease [692], as well as in human clinical trials minocycline had also neuroprotective, antiinflammatory and antiapoptotic properties [693-696]. These effects were thought to arise through the inhibition of microglial activation, iNOS, COX-2 expression and modulation of cytokine expression and release [208]. Moreover, minocycline was found to exert an inhibitory effect on TNFand IFN-γ production by stimulated T cells [697]. In contrast to the effect on T cells, addition of minocycline to LPS-stimulated monocytes led to a dose-dependent increase in TNF-α and IL-6 production. These results indicated that minocycline exerted differential effects on the regulation of cytokine production by T cells and monocytes. Given the pleiotropic effects of minocycline, it was suggested that the immunostimulatory effect on monocytes might counteract its beneficial properties in the treatment of several forms of chronic inflammation [697]. Several authors found a possible relationship between T. gondii infection and etiology of Parkinson’s disease [698], schizophrenia [699,700], Alzheimer’s disease [701], and obsessive-compulsive disorder [702], and minocycline was found to exert beneficial effects in the treatment of schizophrenia [703,704], as well as in patients with toxoplasmic encephalitis [705]. Thus, it seems that preventive effect of minocycline in the hypoglycemia-induced neuronal death was at least in part associated with its antiparasitic activity.

Prevention of Hypoglycemia-Induced Neuronal Death by Hypothermia may be Caused by Cold Stress-Related Modulation of Immune Responses Associated with Enhanced Activity Directed against T. gondii Infection

Hypoglycemia and hypothermia

Shin et al. [706] demonstrated that hypothermia prevented neuronal death induced by hypoglycemia. It was suggested that hypoglycemiainduced microglial activation resulted from the brain infiltration with peripheral and local inflammatory cells, enhanced release of several proinflammatory/neurotoxic substances (various cytokines, chemokines, NO, ROS) associated with morphological changes of microglial cells [707,708], and finally brain tissue inflammation [709,710]. The beneficial effect of cold stress on antibody production was found to be mediated via several nervous system-derived factors, such as glucocorticoids, catecholamines, and/or neuropeptides [711]. Shanks & Kusnecov [712] demonstrated stress-induced enhancement of B cell antibody responses to KLH (a keyhole limpet hemocyanin, a potent antigen in reaginic sensitization of mice and rats) in BALB/cByJ mice and B lymphocytes expressed the β2-adrenergic receptor, and binding of norepinephrine to this receptor was necessary to maintain optimal TH2 cell-dependent antibody generation in vivo [713]. Moreover, norepinephrine appeared to be necessary for optimal IgG and IgM responses [713]. Experimental studies revealed adrenergic modulation of insulin sensitivity, i.e. drugs with β-adrenomimetic activity, such as epinephrine, novodrin, partusisten, α-adrenomimetic - phenylephrine, and one-hour immobilization stress enhanced insulin sensitivity in animals [714,715]. Interestingly, Keijzers et al. [716] suggested that caffeine might decrease insulin sensitivity in healthy humans possibly as a result of elevated plasma epinephrine levels. It seems, however, that the significant increases of epinephrine and norepinephrine (P < 0.0005 and P < 0.02, respectively) [716] reflected rather a defense reaction of the host, because Graham et al. [717] demonstrated that caffeine ingestion elevated insulin response in humans during an oral glucose tolerance test. The above-presented adrenergic modulation of insulin sensitivity in experimental animals is consistent with this reasoning.

In mice, chronic cold stress induced a regulatory phenotype in macrophages, characterized by diminished phagocytic ability, decreased TNF-α and IL-6 and increased IL-10 production [718]. Resting macrophages stimulated spleen cells to produce regulatory cytokines, and an immunosuppressive state that impaired cold stressed mice to control Trypanosoma cruzi proliferation [718]. These regulatory effects correlated with an increase in macrophage expression of 11β-hydroxysteroid dehydrogenase, an enzyme that converts inactive glucocorticosteroid into its active form [718]. Moreover, in experimental animals, exposure to cold significantly decreased insulin secretion induced by arginine, butyrate and tolbutamide, and the release of insulin by pancreatic β-cells appeared to be inhibited through stimulation of α-adrenergic receptors [719,720].

Hypothermia and T. gondii infection

Several authors showed that cold stress profoundly modulated immune response and the outcome of infections because many mediators released during physical stress, such as glucocorticoids and catecholamines, influenced the growth and multiplication of pathogens, including T. gondii [711,721-724]. Cold stress can suppress [725,726] or enhance [727,728] immunity by inducing and/or enhancing the synthesis of specific proteins with relevant roles in transcription, translation, and recombination [729,730]. Aviles & Monroy [711] showed the increased serum specific anti-T. gondii IgG (but not IgM) levels in infected and in infected and stressed mice that underwent cold stress (the animals were kept in cold water, 1 ± 0.5°C for 5 min each day for 8 days) in the chronic phase of infection with the parasite. It appeared that cold stress modulated not only the physiologic processes of the host but also affected the pattern of antibody production against T. gondii antigens during longer parasite persistence in the circulation [711]. In mice with chronic phase of infection, cold water stress caused strong antibody response manifesting as the 5-kDa antigen on the surface of tachyzoites (not present in bradyzoite preparations), with significant diagnostic potential [731]. A similar protein (4-5kDa) was identified in human serum samples that reacted with IgM upon primary infection with the parasite [732]. Otherwise it is known that B cells (responsible for the production of antibodies) play an important role in resistance to persistent T. gondii infection, especially in the brain and lung [733].

Catecholamines act through adrenergic receptors and suppress the activation of TH1 responses and stimulate TH2 immune responses in antigen presenting cells and TH1 cells [734-738]. Recent data however suggest that catecholamines play an important role in the induction of stress-induced (tailshock stress) proinflammatory cytokines and that β-adrenoceptors are critical for tissue (peripheral) IL-1β induction, while both α- and β-adrenoceptors contribute to the induction of plasma (systemic) cytokines [739]. Catecholamines also regulate host innate immune responses under stress situations. In vitro studies have shown that the treatment of human umbilical vein endothelial cells with α1-adrenergic receptor agonists inhibited multiplication of T. gondii [725], and norepinephrine influenced the growth and the production of virulence-associated factors in gram-negative bacteria [740,741]. Recently, it was found that cold water stress decreased parasite burden, expression of chemokines and their receptors in intestinal epithelial cells in vitro and in vivo [722], and down-regulated expression of TLR- 2, -4, -9, and -11 in these cells [742]. Moreover, increased survival was found in mice infected orally with T. gondii and subjected to cold water stress as compared with controls [743]. In contrast, such stress had deleterious outcomes when mice were infected intraperitoneally [721]. These findings may support our suggestion that cold stressinduced host innate immune modulations during latent chronic T. gondii infection also may be partly responsible for the prevention of hypoglycemia-induced neuronal death by hypothermia.

Impaired Vascular Endothelial Function and Abnormal Metabolic Fate of NO in the Patients with Diabetes Mellitus may be Due to the Preferential T. gondii Infection of Endothelial Cells

Endothelial dysfunction with reduced bioavailability of NO characteristic for the patients with diabetes mellitus play an important role in the development of diabetic vasculopathy [744,745], but has been linked also to atherogenesis and non-diabetic glomerulosclerosis [746,747]. It is interesting that the effect of intensive glycemic control on levels of markers of inflammation in patients with T1DM in the diabetes control and complications trial appeared to be not good enough [748]. Thus, these discrepancies might be at least partly explained by the preferential T. gondii infection of the vascular endothelial cells.

Diabetes mellitus

Hamed et al. [749] reported that the endothelial progenitor cells (EPCs) from diabetic patients generated more O2 -, had higher NAD(P)H oxidase and superoxide dismutase activities, but lower NO bioavailability, as compared with healthy individuals. The function of EPCs, which are key cells in vascular repair, is impaired in diabetes mellitus, and NO and ROS can regulate their function. Plasma glucose and HbA1c levels in the diabetic patients were correlated negatively with the NO production from their EPCs [749]. NO is a biologically active unstable radical that is synthesized in vascular endothelial cells by eNOS, and its bioavailability depends on the balance between its production and inactivation rates [750]. Exposure of EPCs to high glucose concentrations increases NAD(P)H oxidase activity which results in increased O2 - generation and reduced NO bioavailability because O2 - inactivates NO and uncouples eNOS [751]. It was demonstrated that NO bioavailability and the in vivo reendothelialization capacity of ECs from diabetic patients can be restored by inactivating NAD(P)H oxidase [752].

Milsom et al. [753] found preferential binding of endogenous and exogenous NO to glycosylated deoxy-hemoglobin and consequently an abnormal metabolic fate of NO in patients with T1DM. They showed that NO-hemoglobin binding was increased at a HbA1c concentrations greater than 8.5% compared with 5.9% (P < 0.01). In blood from diabetic patients, added NO was metabolized mainly to nitrosyl hemoglobin and plasma nitrosothiols, with a 2-fold increase in nitrosyl hemoglobin observed across all NO levels (P < 0.05), and these preferential increases correlated positively with HbA1c concentrations [753].

Cellek et al. [754] demonstrated NO-dependent morphological degeneration and functional nitrergic nerves impairment in diabetes mellitus, and reduced activity and protein amount of neuronal NOS in penile tissue of diabetic rats. Similar degenerative process of the peripheral nerves was reported in diabetic men [755]. Administration of L-NAME (NG-nitro-L-arginine methyl ester), the inhibitor of NO synthase, appeared to be protective in this condition [754]. The atrophy of the nitrergic neurons in the enteric nervous system of the gastrointestinal tract has been documented also during chronic T. gondii infection in experimental animals and men, streptozotocininduced diabetes, and in diabetic patients [434], because the parasite shows tropism to nerve cells [756]. These morphologic and functional abnormalities probably reflect a defense activity of the innate immune system of the host in response to the presence of the parasite, resulting in an increased level of NO produced by the iNOS [757,758]. NO is generated together with several proinflammatory and anti-inflammatory cytokines and chemokines during the invasion by T. gondii, which induces severe inflammatory process at the site of infection. However, excessive amounts of NO are cytotoxic not only for the parasite but also for the host cells [757], because this cytotoxic molecule inhibited the mitochondrial and nuclear enzymes [759]. On the other hand, however, inhibition of iNOS exacerbated chronic cerebral toxoplasmosis in T. gondii-susceptible C57BL/6 mice, although did not reactivate the latent disease in T. gondii-resistant BALB/c mice [760].

Glucose enters into the glycolytic pathway by phosphorylation to glucose-6-phosphate accomplished by hexokinase and glucokinase, with ATP required as phosphate donor. Glucose-6-phosphate is an important compound, being at the junction of glycolysis, gluconeogenesis, the hexose monophosphate shunt, glycogenesis, and glycogenolysis [761]. As in many reactions involving phosphorylation, magnesium must be present [761], and patients with T2DM frequently have hypomagnesemia, hypertriacylglycerolemia in association with enhanced HbA1c levels, retinopathy and neuropathy [762,763]. Glycolysis proceeds by the oxidation of glyceraldehyde-3-phosphate to 1,3-diphosphoglycerate, and glyceraldehyde-3-phosphate dehydrogenase is the enzyme responsible for the oxidation [761]. In many biological systems, nitrosation reactions transferring NO+ from NO donor to a protein S group affect protein function and Mohr et al [764] demonstrated that NO-induced S-glutathionylation led to inactivation of glyceraldehyde-3-phosphate dehydrogenase, thus finally affecting metabolism of carbohydrates and lipid acids. In short, gluconeogenesis is the process whereby glucose is formed from noncarbohydrate metabolic substrates such as pyruvate, lactate, or amino acids, mainly alanine. This metabolic pathway occurs predominantly in the liver and kidney and is essential for the production of glucose during prolonged fasting when glucose stores have been depleted (Figures 4 and 5) [765,766].

diabetes-metabolism-glyceroneogenesis

Figure 4: Glyceroneogenesis generates G3P (glycerol-3-phosphate) instead of glucose. Gluconeogenesis (broken arrows) and glyceroneogenesis (more frequently broken arrows) share several steps in common with glycolysis (solid arrows) because most of the reactions are readily reversible (two direction arrows). Three kinase reactions (HK, hexokinase; PFK, phosphofructokinase; and PK, pyruvate kinase) in glycolysis are not reversible and require separate enzymes for gluconeogenesis (PC, pyruvate carboxylase; PEPCK, phosphoenolpyruvate carboxykinase; F1,6-Bpase, fructose-1,6-bisphosphatase; and G6Pase, glucose-6-phosphatase) and glyceroneogenesis (PC, PEPCK). PEP, phosphoenolpyruvate; DHAP, dihydroxyacetone phosphate; GAP, glyceraldehyde-3-phosphate; OAA, oxaloacetate; TCA, tricarboxylic acid cycle (Krebs cycle) [765].

diabetes-metabolism-adipocytes

Figure 5: Simplified process of glyceroneogenesis in adipocytes and its regulation by PEPCK-C. Lipolysis releases glycerol and fatty acids from triglyceride stores in fat cells. Fasting increases PEPCK-C production and fatty acid release is restrained due to increased glyceroneogenesis and reesterification in triacylglycerols, which requires glycerol-3-phosphate formation from non-carbohydrate precursors [766].

T. gondii infection

In congenital toxoplasmosis, infection of endothelial cells lining the umbilical cord and the placental blood vessels by the parasite is potentially the major transmission route to the fetus [767]. T. gondii invades and proliferates in HUVEC where it resides in parasitophorous vacuoles (PV), and at the time of PV formation, the cell surface anionic sites and fucose residues (human endothelial cells contain exposed fucose residues) are excluded, while HLA class I molecules are present only on a minority of the parasite-containing vacuoles. When the parasite invades the host cells, a PV sheltering are formed because the PV membrane contains proteins (ROP2) anchoring the mitochondria of the host cells to the external surface [768]. T. gondii cyst wall also has anionic sites and this negative charge is mainly produced by phospholipids [769]. The protozoan uses the mitochondria of the host cell to escape from the action of lysosome enzymes. Once within the PV, the parasite multiplies even in phagocytic cells until complete destruction of the host cell [768]. The formation of PV prevents it from merging with the lysosome – a strategy of the protozoan developed to protect its growth within the host cells. During the intracellular life cycle of T. gondii there is no fusion of host cell lysosome with PV, however lysosome-phagosome fusion and parasite destruction occur when fixed or antibody-coated live parasites are internalized by macrophages [770].

Cortez et al. [771] found that HUVEC activated with INF-γ inhibited T. gondii infection and multiplication by 67.5% and 91.0%, respectively. After 4 hrs, 10.2% of INF-γ-activated HUVEC exhibited phagosomelysosome fusion, and NAD(P)H oxidase present at the plasma membrane of activated HUVEC was internalized together with the parasite in 38% of the cells. This may suggest that NAD(P)H oxidase may participate in a mechanism by which INF-γ-activated HUVEC inhibit T. gondii multiplication [771]. This suggestion and the fact that diabetic patients had increased NAD(P)H oxidase activity [749] are consistent with the finding that two forms of lactate dehydrogenase LDH1 and LDH2 from T. gondii were inhibited competitively by gossypol and gossylic iminolactone, and this correlated with specific inhibition of tachyzoites growth in human foreskin fibroblast cultures [772]. Since the bradyzoite LDH2 was more sensitive in vitro to these compounds than the tachyzoite LDH1, it is likely that the growth of bradyzoites in cysts would be inhibited by gossypol and gossylic iminolactone as well [772]. Gossypol and derivatives are aldose reductase (polyol dehydrogenase) inhibitors [773], and gossypol, a natural product from cotton seed, is a non-selective competitive inhibitor of NADH binding to LDH with Ki value 1.4 μM for LDH1 [774]. There was an association between the erythrocyte aldose reductase activity and the complications of diabetes mellitus [775,776]. It was reported that the patients with T1DM who had erythrocyte aldose reductase activity greater the mean ± 2 SD of the found in non-diabetic controls were four times more likely to have diabetic complication than non-diabetic individuals (P < 0.0005) [775]. In order to explain this relationship it should be noted that aldose reductase is a member a family of NADPH-dependent oxidoreductases, present in several human tissues that reduces glucose to sorbitol. In animal models there is evidence that the production of sorbitol is associated with the development of diabetic complications, including neuropathy, nephropathy, cataracts and retinopathy. In hyperglycemia, hexokinase is saturated and the fraction metabolized by aldose reductase increases which involves the sorbitol or polyol metabolic pathway resulting in accumulations of sorbitol and fructose [776]. Chandra et al. [777] showed that increasing NO availability inhibits aldose reductase, thus preventing sorbitol accumulation, whereas inhibiting NO synthesis promotes the activation of the rate-limiting enzyme. Inhibitors of aldose reductase have been developed to reduce the incidence or slow the progression of the major complications of diabetes mellitus. Flavonoids were found to possess aldose reductase inhibition and antioxidant activities in vitro as well as inhibition in the accumulation of sorbitol in the tissues of streptozotocin-induced diabetic rats [778].

Canedo-Solares et al. [779] demonstrated that invasion of both human microvascular endothelial cells (HMEC-1) and umbilical vein endothelial cells (HUVECs) by T. gondii RH and ME49 strains increased along with time. HMEC-1 cells were more susceptible to infection with the parasite than HUVECs, and ME49 parasites were faster than RH ones and this may be related to their ability to survive out of the cell and to the fact that T. gondii is more invasive during G1-S phase. In addition, both HMEC-1 and HUVECs showed higher number of parasitic vacuoles per cell when infected by ME49 tachyzoites than by RH protozoan, i.e. ~ 30 vs. 20 at 4 hrs, respectively [779]. These differences may also partly be explained by various division rates of intracellular T. gondii tachyzoites documented in vitro in many primary human cells, including endothelial cells (Table 14). Interestingly, compared to RH tachyzoites, ME49 tachyzoites induced a stronger upregulation of ICAM-1 in the brain vascular endothelial cells and an earlier and stronger IL-6 and MCP-1 secretion by these cells. T. gondii type I and II strains induced similar migration patterns of antigen-presenting cells but the infected antigen-presenting cells showed a more intensive migration compared to lymphocytes (4.63% vs. 0.6% of all cells) across the blood-brain barrier [780]. It was suggested that the parasite modulated gene expression of the brain vascular endothelial cells to promote its own migration through the blood-brain barrier as a Trojan horse [780]. Knight et al. [781] reported that the exposure of the rat retinal vascular endothelial cells to T. gondii infection after 2 hrs resulted in change of expression of approximately 6% of genes, including those involved in cell structure, protein and vesicle trafficking, cell-cycle regulation, transcriptional and translational machinery, and apoptosis. Infection of BUVECs with tachyzoites also enhanced transcription of several genes and induced adhesion of polymorphonuclear neutrophil cells (PMN) to both infected and noninfected BUVECs within one cell layer, suggesting parasite-induced paracrine activation of these cells and an effective role of PMN in development of the innate immunity to the parasites [782,783].

T. gondii is the common cause of posterior uveitis and a recurrent necrotizing retinochoroiditis worldwide, which may lead to a permanent loss of visual acuity. Tachyzoites spread throughout the body through the blood stream and lymphatics, but preferentially encyst in the eye and other parts of the CNS. Furtado et al. [784] found that human monocyte-derived dendritic cells infected with T. gondii tachyzoites transmigrated in larger numbers across stimulated human retinal endothelium than uninfected dendritic cells (P ≤ 0.0004). Antibody blockade of ICAM-1, VCAM-1, and activated leukocyte cell adhesion molecule inhibited transmigration, while chemokines CCL21 or CXCL10 increased this process, which might be important for development of a novel therapeutic approach [784]. Zamora et al. [785] reported that T. gondii tachyzoites invade human retinal endothelial cells (HREC), which were more susceptible to infection with the parasite than other subpopulations human dermal endothelial cells. The enhanced susceptibility of human retinal vascular endothelium to infection by T. gondii may be related to preferential binding of tachyzoites to the retinal vascular endothelial surface, relative ease of penetration into the cell, rate of replication within the cell and/or cell response to infection, as compared with aortic (55% more), umbilical vein (33%) and dermal (34%) endothelial cells [786]. It appeared that growth of the tachyzoites was approximately 2.8-fold higher in retinal endothelium than in foreskin fibroblasts [786]. Binding of the parasite to host cells is partially mediated by interaction with sulfated glycosaminoglycans (GAGs). Table 42 summarized the blocking effect of several soluble sulfated proteoglycans on attachment of T. gondii parasites to human host cells from a variety of lineages [787]. Heparin, heparin sulfate, chondroitin sulfate showed a negative charged sulfate groups on the molecules-mediated dose dependent inhibition in the average number of parasites attached to the substrate and gliding [787]. It was found that low concentrations of GAGs increased invasion of human fibroblasts while mutant CHO cells lacking in cell surface sulfated proteoglycans were less susceptible to infection [788].

Cell type Cell line IC50 (mg/ml)a          
    Heparin CSC CSA Dextran sulfated Dextran Fucoidine
Endothelial HUV-EC-Cb 1.8 ± 0.2 1.6 ± 0.4 2.9 ± 1.5 1.0 ± 0.4 9.7 ± 3.4 4.1 ± 1.8
Epithelial HEp-2b 5.5 ± 1.6 4.5 ± 0.4 6.3 ± 2.3 5.7 ± 2.3 14.1 ± 2.1 19.5 ± 0.2
Fibroblast HFF 4.8 ± 2.0 4.1 ± 1.4 7.3 ± 1.7 2.2 ± 0.4 > 20 > 20
Glial or astrocyte U373b 3.0 ± 1.9 3.0 ± 0.8 1.1 ± 0.4 2.1 ± 0.4 7.4 ± 3.8 16.0 ± 3.7
Macrophage U937b 2.0 ± 0.3 1.8 ± 0.8 2.6 ± 1.5 6.7 ± 2.6 7.0 ± 2.7 13.2 ± 4.9
Melanocyte G361b 4.0 ± 0.3 2.0 ± 0.1 3.0 ± 1.9 4.6 ± 0.4 8.8 ± 4.7 0.3 ± 0.1
Averagec   3.5 ± 1.4 2.8 ± 1.1 3.9 ± 2.2 3.7 ± 2.1 11.2 ± 4.6 11.7 ± 6.2
aMean ± SE, n = 2 or 3; bObtained from the American Type Culture collection; cAverage values (± SE) for all cell types tested. dA sulfated L-fucose oligosaccharide. eThe synthetic polyanion. IC50: Inhibitory Concentration; CSC: Chondroitin sulfate C; CSA: Chondroitin Sulfate A; Several heparin binding proteins were identified in lysates of T. gondii based on their ability to agglutinate red blood cells in a heparin-sensitive manner [460].

Table 42: Inhibition by soluble glycosaminoglycans of T. gondii attachment to various mammalian cells (acc. Carruthers et al. [459]; with own modification).

Brunton et al. [789] found that pretreatment of rat retinal vascular endothelial cells infected with T. gondii RH strain tachyzoites, with IFN-γ, TNF or IL-1β resulted in a significant decrease in the parasite replication, and this inhibition appeared to be independent of NO production while L-tryptophan catabolism may have a role in this IFN-γ-mediated process. When the ovine umbilical vein endothelial cells were pretreated with IFN-γ, a high degree of inhibition of T. gondii replication was observed with the effect being dose-dependent, with a maximum IFN-γ activity of 625 U/ml (range 0.15-1250 U/ml) [790]. Studies of Däubener et al [163] confirmed that indoleamine 2,3-dioxygenase induction contributed to the antiparasitic effector mechanism inducible in human brain microvascular endothelial cells (HBMEC) by IFN-γ and TNF-α, because this enzyme was strongly induced in HBMEC, and its activity was enhanced by co-stimulation with TNF-α, while the addition of excess amounts of tryptophan to the HBMEC cultures resulted in a complete abrogation of the antitoxoplasmatic effect. The mechanism of inhibition of T. gondii in HUVEC activated by IFN-γ was found to be different from that present in mouse macrophages and human fibroblasts [310-313]. For example, Ji et al [791] demonstrated that exogenous NO triggered egress of T. gondii tachyzoites from infected peritoneal macrophages obtained from C57BL/6 mice, which then underwent necrosis. Moreover, addition of the calcium ionophore A23187 resulted in an increased release of merozoites from mature T. gondii meronts grown in cultured primary bovine umbilical vein endothelial cells (BUVECs) [792]. The extent and time course of the release was dependent on both, maturity of the meronts and concentration of the calcium ionophore because survival of parasitized host cells and the parasites themselves should be dependent on ion balances, especially on extra- and intracellular calcium concentrations [792].

Dimier & Bout [793] showed that when HUVEC were pretreated with IL-1β and TNF-α concentrations ranging from 1 to 100 U/ml, a dose-dependent inhibition of the intracellular parasite replication was observed. In addition, Benedetto et al. [724] demonstrated that pretreatment of HUVEC with an α-adrenergic resulted in a high degree of intracellular killing of T. gondii in these cells. Moreover, α-adrenergics activated HUVEC, and induced a marked dosedependent toxoplasmastatic activity. Also a significant positive correlation was observed between the toxoplasmastatic activity and release of NO2 - during the activation phase before infection with the parasite, although this effect was not present during the infection phase.

Diabetes Mellitus Comorbidities

Possible association between diabetes, epilepsy and T. gondii infection

The prevalence of active epilepsy in European countries varies between 3.3 and 7.1 per 1000 for the age range 0 to 90 yrs, with a peak prevalence of 9 to 11.6 per 1000 in adults > 50 yrs of age, and 2.1 to 4.1 per 1000 in children and adolescence [794,795]. Ramakrishnan & Appleton [796] found that 6 of 285 children aged less than 16 yrs with T1DM had epilepsy giving a prevalence of 21/1000, which is approximately six times greater than the prevalence of epilepsy in general population of children in UK. Caletta et al. [797] found that among their cohort of 10 children suffering from both T1D and epilepsy, 5 was had generalized epilepsy and 5 was diagnosed with focal disease. Seizures caused by hypoglycemia in the patients with T1DM are quite frequent events with 18.2 to 62 per 100 patient years depending on age, type of treatment, and residual insulin secretion [798,799].

Schober et al. [794] found that children and adolescents with T1DM had an increased prevalence of epileptic seizures. There was also an association between epilepsy and diabetic ketoacidosis in these patients, and the risk of ketoacidosis was almost double in the patients with epilepsy compared with patients with diabetes alone. It must be noted that the frequency of severe hypoglycemia was lower in the patients treated with antiepileptics [800]. This may suggest that T. gondii infection was an important triggering agent because for example valproic acid, an epileptic drug, exerted strong antitoxoplasmatic effect (Table 43). It must be noted that uptake of the drug by bovine brain microvessel endothelial cells has been well documented [802], and these cells represent frequent targets of the parasite in human host (Table 42). In addition, Glodek-Brzozowska et al. [803] analysed 784 children with T1DM and found that 8 of them (aged 7.5 to 18 yrs) had epilepsy, as a concomitant disease, with local or generalized abnormal changes in EEG. The patients had seizure types of different morphology, including petit mal, partial, myoclonic, or tonic-clonic seizures, and 7/8 patients received treatment with valproic acid as the main anticonvulsant. Poor control of diabetes was observed in 4 children during acute seizure attacks [804]. Moreover, Schober et al. [794] reported that the increased risk of this abnormality cannot be caused by carbonic anhydrase inhibitors, as previously suggested [805], because treatment with other antiepileptic medications also was associated with metabolic acidosis, which was observed even in patients without anticonvulsive treatment. The patients with epilepsy were younger at onset of diabetes and shorter as compared with those without epilepsy [794].

Drug Solvent ID50a
(mg/ml)
TD50b
(mg/ml)
TIc
Valproic acid ethanol 4.5 62.4 13.9
Sodium valproate ethanol 4.1 52 12.7
Carbamazepine ethanol 72 100 1.3
Litium carbonate 1 N HCl > 100 > 100  
Haloperidol ethanol 5.6 103 18.4
9-OH-Risperidone tartaric acid 20.1 134 6.7
Risperidone tartaric acid 74 129 1.7
Fluphenazine HCl Toxo CGM 3.5 17.9 5.1
Clozapine ethanol 5.8 20 3.4
Olanzapine DMSO 33.2 100 3.0
Chlorpromazine HCl DMSO 2.6 6 2.3
Quetiapine fumarate DMSO 18.6 33 1.8
Trimethoprim DMSO 5.3 63.8 12.1
aMedian inhibitory dose, a measure of tachyzoite inhibition; bMedian toxicity dose,a measure of cytotoxicity; cTherapeutic index, a measure of efficacy determined by TD50/ID50 ratio. DMSO, dimethylsulfoxide; Toxo CGM, Toxoplasma cell growth medium. Valproic acid at a concentration of 1 μg/ml inhibited 7% of the tachyzoites and trimethoprim at 3.2 μg/ml produced 2% inhibition, but the combination of these two compounds at those concentrations resulted in a potentiating effect inhibiting 55% of the tachyzoites

Table 43: Drugs tested for in vitro activity against T. gondii (according to Jones- Brando et al. [801]; with own modification).

Palmer [806] showed a highly significant association between the seroprevalence rates for chronic T. gondii infection and prevalence rates of cryptogenic epilepsy (P < 0.001) (log-odds ratio of 4.8, CI 2.6 to 7.8). Three children aged 5 to 11 years with Landau-Kleffner syndrome (aphasia plus epilepsy; the speech disorders appeared after epileptic seizures started) and EEG abnormalities, also had increased titers of IgG antibodies against T. gondii [807]. Furthermore, Stommel et al. [804] suggested that chronic T. gondii infection with brain cysts may be a cause of cryptogenic epilepsy because they found a statistically significant 59% elevation of the parasite antibodies among these patients as compared to controls (P=0.013).

Interestingly, it was suggested [808] that the syndrome of epilepsia partialis continua represented symptoms of hyperglycemia and was the first symptom leading to the diagnosis of diabetes mellitus. However, the majority of the 22 patients analyzed had evidence of localized structural brain lesions and therefore it seemed that the hyperglycemia was not the main cause of the epileptic abnormalities [808].In a German population-based case-control study including 366 glioma and 381 meningioma cases, and 1494 controls Berg-Beckhoff et al. [809] found the positive association between epilepsy and particularly glioma suggesting that epilepsy is an early symptom of the disease. As the association was observed also for epilepsies occurring more than a decade before the diagnosis of glioma, the authors suggested that this might indicate an etiological role of epilepsy, or a relatively long preclinical state [809]. This finding and reasoning is consistent with the recent suggestion that chronic latent T. gondii infection of the central nervous system may be responsible for development of ependymoma and glioma [810].

Possible association between diabetes, migraine/other type of headaches and T. gondii infection

The prevalence of migraine and tension-type headache in the patients with diabetes mellitus was found to be higher as compared with controls without diabetes [811-813]. The disease was more common amongst teenagers with migraine compared with those without migraine [814]. Also, within a headache clinic population Tietjen et al. [815] identified diabetes mellitus as a migraine comorbidity constellation. However, in a large population-based cross-sectional study performed in Norway, Aamodt et al. [811] found the inverse relationship between migraine and diabetes. Prevalence odds ratios of migraine was lower amongst persons with diabetes compared with those without diabetes, the OR being 0.4 (95% CI: 0.2-0.9) for T1DM, and 0.7 (CI: 0.5-0.9) for T2DM. It appeared that OR of headache were lower amongst those with duration of diabetes ≥ 13 yrs compared with those who were diagnosed with diabetes during the last 3 years, OR 06 (0.4-0.9) [811].

Split & Szydlowska [813] reported that amongst their cohort of 154 patients with T2DM, 95 (61.7%) had migraine, and 32 (20.8%) tension-type headaches. In 50 individuals, the onset of migraine headaches occurred after diagnosis of diabetes, while in 45 individuals migraine was diagnosed before the onset of diabetes. Interestingly, in this group of patients the onset of diabetes caused a significant increase in the average number of headache days per year. In the control group of 106 persons, migraine was diagnosed in 17 (16%) subjects, while 28 (26.4%) individuals suffered from tension-type headache [813]. Recently, Cavestro et al. [816] showed that both blood glucose and insulin levels were higher in migraineurs than in healthy controls (P < 0.0001) suggesting altered metabolism of insulin in this clinical entity. Moreover, Okada et al. [817] demonstrated higher levels of lactic acid and pyruvic acid in the plasma of patients with migraine as compared with controls, which indicated mitochondrial function abnormalities in carbohydrate and fats metabolism, including tricarboxylic acid cycle. In addition, insulin sensitivity was found to be impaired in the patients with migraine as compared with normal individuals (P < 0.001) [818,819]. Therefore, its seems that there is a relationship between migraine/other types of headache and T1DM/T2DM, especially that many patients with recurrent headaches/migraine were found to be seropositive for T. gondii [820,821] and about two billion of people worldwide are chronically infected with the parasite.

Possible association between T1DM, autism spectrum disorders (ASD) and T. gondii infection

Freeman et al. [822] reported an increased prevalence of ASD in their group of 984 pediatric patients with T1DM in Toronto (Canada) than in general population (0.9% [95% CI: 0.3-1.5 vs. 0.34-0.67]). The median age at diagnosis of ASD was 4.8 yrs (range 3.3-6.8), while that for T1DM was 8.2 years (range 0.8-13.5). In mothers of patients with autism, Comi et al. [823] reported higher incidence of autoimmune diseases compared with controls, and specifically autoantibodies implicated in autoimmune thyroid disorders also have been found with an increased prevalence in the patients with T1DM [824-826]. Although the studies performed by Harjutsalo & Tuomilehto [826] in northern Finland did not support their suggestion about the link between T1DM and ASD, Iafusco et al. [828] in their cohort of patients with T1DM (aged <14 years) from several Italian centers of pediatric diabetology (0.72% [0.69-0.75] found a pattern similar to that observed by Freeman et al. [822]. They suggested that these differences may be linked to the incidence of diabetes, and emphasized that, for example, Sardinia (Italy) has a very high incidence of T1DM epidemiology (42.4/100 [95% CI: 40.5-44.4]), while the peninsular Italy has on overall incidence similar to other Mediterranean regions (8.4/100 [7.9-8.9]; only 2 of 1373 analyzed patients aged < 14 years from Sardinia were diagnosed with autism [828]. These discrepancies emphasize importance of geoepidemiological differences in the incidence of diabetes and the fact that some environmental factors play an important role in development of both T1DM and ASD in the same person. Recently, a significantly lower occurrence of seropositive titers against the parasite was found in the patients with T1DM and their close family members [20] as compared with healthy controls. Similar results were obtained in 83 autistic children aged 1-18 yrs (mean age 6 yrs) in whom only two patients had positive serum anti-T. gondii IgG levels (unpublished results, Magdalena Cubala-Kucharska, personal information, November 2012).These unexpected findings may be explained by the suppression of cytokine IL-2 generation, decreased activation of lymphocyte B cells responsible for immunoglobulin secretion and markedly lower levels of immunoglobulins IgG, IgA, and IgM due to T. gondii infection [21]. In addition, persistent and prolific primary autoimmune-induced generation of many antibodies characteristic for the patients with ASD directed against own proteins (the so called a perpetuum mobile-like biomachinery [434,828] may be associated with their exhausted secondary innate and acquired immune responses directed against foreign T. gondii antigens [23], in which host-endoplasmic reticulum-parasitophorous vacuole interaction provides a route of entry for antigen cross-presentation in T. gondiiinfected dendritic cells [829,15]. This explanation may be at least in part responsible for the markedly low occurrence of specific antibodies directed against the parasite found in the autistic children, as well as for the positive relationship found between the proportion of children who received the recommended vaccines by age 2 years and the prevalence of autism or speech/language impairment across the U.S. population [831].

Patients with autism have significantly increased NO production [579,832-834]. Higher plasma nitrite and nitrate (NOx) levels were also found in children with autism compared with controls [833]. Higher NOx concentrations demonstrated in red blood cells of autistic patients compared to age- and sex-matched normal controls, along with enzymatic evidence of NO-related oxidative stress [832], were associated with mitochondrial dysfunction [835]. The induction of a high-output inducible enzyme NOS (iNOS) is triggered primarily by IFN-γ, in combination with TNF- and IL-1β, or endotoxin [579,836,837]. It must be noted that autistic children showed enhanced production of the cytokines IFN-γ, TNF-α and IL-1 compared to controls [838,839]. Given the key role of these cytokines in the induction of iNOS that resulted in the elevated NO generation in autism one may suggest that iNOS was also involved in the enhanced NO production in diabetes mellitus due to a profuse secretion of these proinflammatory cytokines in this clinical entity (Table 6) [83-87]. Recently, it was demonstrated in vitro that exogenous NO released by different doses of sodium nitroferricyanide (III) dehydrate could trigger egress of T. gondii tachyzoites from infected peritoneal macrophages collected from C57BL/6 mice, which then underwent necrosis [840]. This finding is very important because it may represent a novel approach for treatment of neuroinflammation caused by the parasites located in astrocytes and other eukaryotic cells.

Possible association between T1DM, celiac disease (CD) and T. gondii infection

CD occurs in children and adolescents with T1D with the prevalence ranging from 4.4 to 11.1% (mean 8%) as compared with 0.5% in general population [840-842]. The mechanism of association of these two diseases involves HLA genotypes DR3-DQ2 and DR4-DQ8 (T1DM), and DR3-DQ2 (CD) [840]. It is striking that T1DM and celiac disease share 13/52 (25%) risk loci outside the HLA gene complex (http://www.t1dbase.org) [843]. Both diseases T1DM and CD have an abnormal small intestinal immune response with inflammation and a variable grade of enteropathy [842]. Patients with T1DM often have only few and mild symptoms of CD or are asymptomatic (silent), and rarely present with severe manifestation of CD. The mean age at diagnosis of classical CD is usually around 2-3 yrs, while the age for T1DM is 7-8 yrs. In the patients with T1DM, diabetes is usually diagnosed first, CD precedes diabetes onset only in 10-25% of individuals [840,844,845].

Cronin et al. [846] reported that of 177 patients attending a seizure clinic, four individuals had celiac disease (1 in 44). In a control group of 488 pregnant women only 2 samples were positive for celiac disease (1 in 244). In previous hospital records, 16 patients (10 F/6 M) suffered from both celiac disease and epilepsy (mean age at diagnosis of epilepsy was 23 yrs (range from < 1 to 67); mean age at diagnosis of celiac disease was 28.5 yrs (range < 1 to 73 yrs) [846].

Sandberg-Bennich et al. [847] demonstrated that the most evident risk factor for development of celiac disease was associated with neonatal infections (OR =1.52, confidence limits 1.19; 1.95). T. gondii infection may therefore play an important role in triggering development of both diabetes and celiac disease, especially that the increased percentage of antigliadin IgG antibodies was found to be associated with T1DM [20] (Table 2). Moreover, Rostami Nejad et al. [848] found that amongst 827 pregnant women, 154 (31%) and 58 (7%) of them had positive total IgG and IgM for T. gondii serology (blood sample were taken at mean pregnancy duration of 5.5 months). In addition, 27 women (mean age 27 yrs; mean pregnancy duration 4.8 months) were simultaneously diagnosed with celiac disease, and 16 out of 27 (59%) had infection with the parasite as compared with 257 out of 800 (32%) non-celiac disease pregnant women (OR = 3.07, 95% confidence limits 1.4-6.7) (P = 0.04). The authors suggested that celiac disease during pregnancy increase the risk of T. gondii infection [848]. Critical analysis of the literature data suggests however that on the contrary, chronic latent infection with the parasite increases the risk of celiac disease development [810]. This reasoning is strongly supported by the recent report of Severance et al. [849] that in mice receiving a standard wheat-based rodent chow, peroral, intraperitoneal and prenatal T. gondii exposure launched a highly significant generation of anti-gluten IgG antibodies in all infected animals compared to uninfected controls (P ≤ 0.00001). Perorally-infected females showed higher concentrations of anti-gluten IgG than males (P ≤ 0.009) indicating that the parasite-generated gastrointestinal infection led to a marked anti-gluten response in a sex-dependent manner [849]. These findings may be explained by the facts that: 1) transepithelial migration of T. gondii is linked to its active motility and virulence [850], 2) involves an interaction of human ICAM-1 with the parasite adhesin MIC2 resulting in its immunoprecipitation [317], and 3) the parasite targets the paracellular pathway to invade the intestinal epithelium and affects epithelial tight junction-associated proteins [10], thus finally affecting host intestinal wall permeability. Beneficial ameliorative effects of breastfeeding and human colostrum, which contains large quantities of lactoferrin, administered in neonatal autistic rats with celiac disease, may support the above-presented reasoning [851].

Concluding Remarks

Maternal and/or fetal-derived islet infiltration by the cells infected with T. gondii may induce increased production of proinflammatory cytokines, such as TNF-α, IL-1β, and IFN-γ, which can result in development of insulitis. They also modify the antigen presenting cells toward a more immunogenic phenotype, thus triggering the offspring T cells to react against pancreatic self antigens, like it was found during induction of diabetes by viral or bacterial infections. This reasoning is consistent with the reports that elimination of maternally transmitted autoantibodies prevented development of diabetes in nonobese diabetic mice, and that oral administration of autoantigen induced autoimmune diabetes. Moreover, in vitro studies showed that insulin and D-glucose had a dose-responsive mitogenic effect on intracellular T. gondii replication and development in 3T3-L1 cells. One cannot therefore exclude that the additive/synergistic effect of insulin/glucose on multiplication of the parasite in pancreatic islet beta-cells may be at least in part responsible for triggering autoimmune defense reaction of the host that induce insulitis, and finally cause diabetes. TGF-β, a multifunctional cytokine, also was found to favour growth of the parasite through suppressing IFN-γ toxoplasmastatic activity. The pancreatic islet β cells hypertrophy and fibrosis characteristic for the patients with T2DM may be caused by the increased levels of TGF-β due to T. gondii infection because the elevated expression of the cytokine in vitreous, retina and retinal pigment epithelium was closely correlated with retinal fibrosis and choroidal neovascularization. In addition, TGF-β played a key role of in mediating diabetic renal hypertrophy, tubulointerstitial and vaginal tissue fibrosis. Increased membrane metalloproteinases secretion induced by the parasite may also contribute to the pancreatic islet fibrosis. Diabetes caused marked changes in the function and metabolism of neutrophils, for example glutamine oxidation and glutaminase activity were markedly decreased in the neutrophils from diabetic rats, and glutamine plays an important role in protein (as an amino acid source), lipid (by NADPH production) and nucleotide synthesis (by purine and pyrimidine production), as well as in NADPH oxidase activity. It must be emphasized that the tachyzoite stage of T. gondii, responsible for an acute infection, rapidly metabolizes glucose via glycolysis However, it was also demonstrated that glucose was nonessential for T. gondii tachyzoites because hostderived glucose and its transporter in the parasite were dispensable by glutaminolysis. Thus, eventually increased requirements for glutamine and competition for this amino acid between T. gondii and neutrophils (and probably other cells) may result in diminished sources of glutamine and development of disturbances in maintaining regular metabolic and immune processes in many host cells. Moreover, this amino acid raises the in vitro bacterial killing activity and the rate of ROS generation by neutrophils, and delays spontaneous apoptosis of these cells. These findings may be further supported by the morphological abnormalities of myenteric neurons in experimental animals during Toxoplasma gondii infection, including hypertrophy/atrophy of the neurons and changes in the cell body areas depending on the parasite genotype, its form, dose, route of inoculation, animals studied, and part of the gastrointestinal tract involved. Virulence of the parasite was also associated with distinct dendritic cell responses and reduced numbers of activated CD8+ T cells. Impaired vascular endothelial function and abnormal metabolic fate of NO in patients with diabetes mellitus may be at least in part due to the preferential T. gondii infection of endothelial cells. Finally, vitamin D and minocycline exerted beneficial effects on development and clinical course of diabetes mellitus probably because of their immunomodulating and antitoxoplasmatic activities. In addition, a strong correlation was found between the HSP65 expression and protection against T. gondii infection, suggesting that this protein significantly contributed to development of the host defense system. It seems therefore that infection with this protozoan plays an important role in the etiopathogenesis of both types of diabetes.

References

  1. American Diabetes Association (2008) Diagnosis and classification of diabetes mellitus. Diabetes Care 31 Suppl 1: S55-60.
  2. Danaei G, Finucane MM, Lu Y, Singh GM, Cowan MJ, et al. (2011) National, regional, and global trends in fasting plasma glucose and diabetes prevalence since 1980: systematic analysis of health examination surveys and epidemiological studies with 370 country-years and 2·7 million participants. Lancet 378: 31-40.
  3. Contreras F, Rivera M, Vasquez J, De la Parte MA, Velasco M (2000) Diabetes and hypertension physiopathology and therapeutics. J Hum Hypertens 14 Suppl 1: S26-S31.
  4. http://cdc.gov/diabetes/statistics/prev/national/figpersons.htm. Accessed Dec 15, 2012
  5. Hsia Y, Neubert AC, Rani F, Viner RM, Hindmarsh PC, et al. (2009) An increase in the prevalence of type 1 and 2 diabetes in children and adolescents: results from prescription data from a UK general practice database. Br J Clin Pharmacol 67: 242-249.
  6. Baban B, Penberthy WT, Mozaffari MS (2010) The potential role of indoleamine 2,3 dioxygenase (IDO) as a predictive and therapeutic target for diabetes treatment: a mythical truth. EPMA J 1: 46-55.
  7. Boucher BJ (2011) Vitamin D insufficiency and diabetes risks. Curr Drug Targets 12: 61-87.
  8. Dubey JP, Beattie CP (1988) Toxoplasmosis in animals and man CRC Press. Boca Raton, FL, pp.1-220.
  9. Dubey JP (1998) Advances in the life cycle of Toxoplasma gondii. Int J Parasitol 28: 1019-1024.
  10. Weight CM, Carding SR (2012) The protozoan pathogen Toxoplasma gondii targets the paracellular pathway to invade the intestinal epithelium. Ann N Y Acad Sci 1258: 135-142.
  11. Munoz M, Liesenfeld O, Heimesaat MM (2011) Immunology of Toxoplasma gondii. Immunol Rev 240: 269-285.
  12. Dubey JP, Jones JL (2008) Toxoplasma gondii infection in humans and animals in the United States. Int J Parasitol 38: 1257-1278.
  13. Halos L, Thébault A, Aubert D, Thomas M, Perret C, et al. (2010) An innovative survey underlining the significant level of contamination by Toxoplasma gondii of ovine meat consumed in France. Int J Parasitol 40: 193-200.
  14. Zeiner GM, Norman KL, Thomson JM, Hammond SM, Boothroyd JC (2010) Toxoplasma gondii infection specifically increases the levels of key host microRNAs. PLoS One 5: e8742.
  15. Butcher BA, Kim L, Johnson PF, Denkers EY (2001) Toxoplasma gondii tachyzoites inhibit proinflammatory cytokine induction in infected macrophages by preventing nuclear translocation of the transcription factor NF-kB. J Immunol 167: 2193-2201.
  16. Suzuki Y (2002) Immunopathogenesis of cerebral toxoplasmosis. J Infect Dis 186 Suppl 2: S234-240.
  17. Aliberti J (2005) Host persistence: exploitation of anti-inflammatory pathways by Toxoplasma gondii. Nat Rev Immunol 5: 162-170.
  18. Israelski DM, Remington JS (1993) Toxoplasmosis in the non-AIDS immunocompromised host. Curr Clin Top Infect Dis 13: 322-356.
  19. Bhopale GM (2003) Pathogenesis of toxoplasmosis. Comp Immunol Microbiol Infect Dis 26: 213-222.
  20. Krause I, Anaya JM, Fraser A, Barzilai O, Ram M, et al. (2009) Anti-infectious antibodies and autoimmune-associated autoantibodies in patients with type I diabetes mellitus and their close family members. Ann N Y Acad Sci 1173: 633-639.
  21. Guk SM, Kook J, Jeon YH, Choi JH, Han ET, et al. (2005) Suppressed cytokine and immunoglobulin secretions by murine splenic lymphocytes infected in vitro with Toxoplasma gondii tachyzoites. J Parasitol 91: 467-470.
  22. Flegr J, Stríž I (2011) Potential immunomodulatory effects of latent toxoplasmosis in humans. BMC Infect Dis 11: 274.
  23. Bhadra R, Khan IA (2012) Redefining chronic toxoplasmosis--a T cell exhaustion perspective. PLoS Pathog 8: e1002903.
  24. Gokce C, Yazar S, Bayram F, Gundogan K, Yaman O, et al. (2008) Anti-Toxoplasma gondii antibodies in type 2 diabetes. Natl Med J India 21: 51.
  25. Shapira Y, Agmon-Levin N, Selmi C, Petrikova J, Barzilaj O, et al. (2012) Prevalence of anti-toxoplasma antibodies in patients with autoimmune diseases. J Autoimmunity 39: 112-116.
  26. Zandman-Goddard G, Shoenfeld Y (2009) Parasitic infection and autoimmunity. Lupus 18: 1144-1148.
  27. Shapira Y, Agmon-Levin N, Shoenfeld Y (2010) Defining and analyzing geoepidemiology and human autoimmunity. J Autoimmun 34: J168-177.
  28. Shapira Y, Agmon-Levin N, Shoenfeld Y (2010) Geoepidemiology of autoimmune rheumatic diseases. Nat Rev Rheumatol 6: 468-476.
  29. Clin Rev Allergy Immunol, et al. (2012) Geographical differences in autoantibodies and anti-infectious agents antibodies among healthy adults. . Clin Rev Allergy Immunol 42:154-163.
  30. Nelson JL, Gillespie KM, Lambert NC, Stevens AM, Loubiere LS, et al. (2007) Maternal microchimerism in peripheral blood in type 1 diabetes and pancreatic islet beta cell microchimerism. Proc Natl Acad Sci U S A 104: 1637-1642.
  31. Wrenshall LE, Stevens ET, Smith DR, Miller JD (2007) Maternal microchimerism leads to the presence of interleukin-2 in interleukin-2 knock out mice: implications for the role of interleukin-2 in thymic function. Cell Immunol 245: 80-90.
  32. Klonisch T, Drouin R (2009) Fetal-maternal exchange of multipotent stem/progenitor cells: microchimerism in diagnosis and disease. Trends Mol Med 15: 510-518.
  33. Artlett CM, Smith JB, Jimenez SA (1998) Identification of fetal DNA and cells in skin lesions from women with systemic sclerosis. N Engl J Med 338: 1186-1191.
  34. Artlett CM, Cox LA, Ramos RC, Dennis TN, Fortunato RA, et al. (2002) Increased microchimeric CD4+ T lymphocytes in peripheral blood from women with systemic sclerosis. Clin Immunol 103: 303-308.
  35. Reed AM, Picornell YJ, Harwood A, Kredich DW (2000) Chimerism in children with juvenile dermatomyositis. Lancet 356: 2156-2157.
  36. Artlett CM, Ramos R, Jiminez SA, Patterson K, Miller FW, et al. (2000) Chimeric cells of maternal origin in juvenile idiopathic inflammatory myopathies. Childhood Myositis Heterogeneity Collaborative Group. Lancet 356: 2155-2156.
  37. Abbud Filho M, Pavarino-Bertelli EC, Alvarenga MP, Fernandes IM, Toledo RA, et al. (2002) Systemic lupus erythematosus and microchimerism in autoimmunity. Transplant Proc 34: 2951-2952.
  38. Endo T, Pelster B, Piekarski G (1981) Infection of murine peritoneal macrophages with Toxoplasma gondii exposed to ultraviolet light. Z Parasitenkd 65: 121-129.
  39. Klintschar M, Schwaiger P, Mannweiler S, Regauer S, Kleiber M (2001) Evidence of fetal microchimerism in Hashimoto's thyroiditis. J Clin Endocrinol Metab 86: 2494-2498.
  40. Ando T, Imaizumi M, Graves PN, Unger P, Davies TF (2002) Intrathyroidal fetal microchimerism in Graves' disease. J Clin Endocrinol Metab 87: 3315-3320.
  41. Vabres P, Malinge MC, Larrègue M, Bonneau D (2002) Microchimerism from a dizygotic twin in juvenile ulcerative lichen planus. Lancet 359: 1861-1862.
  42. Aractingi S, Berkane N, Bertheau P, Le Goué C, Dausset J, et al. (1998) Fetal DNA in skin of polymorphic eruptions of pregnancy. Lancet 352: 1898-1901.
  43. Hayashida M, Nishimoto Y, Matsuura T, Takahashi Y, Kohashi K, et al. (2007) The evidence of maternal microchimerism in biliary atresia using fluorescent in situ hybridization. J Pediatr Surg 42: 2097-2101.
  44. Muraji T, Hosaka N, Irie N, Yoshida M, Imai Y, et al. (2008) Maternal microchimerism in underlying pathogenesis of biliary atresia: quantification and phenotypes of maternal cells in the liver. Pediatrics 121: 517-521.
  45. Hinze-Selch D, Däubener W, Erdag S, Wilms S (2010) The diagnosis of a personality disorder increases the likelihood for seropositivity to Toxoplasma gondii in psychiatric patients. Folia Parasitol (Praha) 57: 129-135.
  46. Zhu S, Lai DH, Li SQ, Lun ZR (2006) Stimulative effects of insulin on Toxoplasma gondii replication in 3T3-L1 cells. Cell Biol Int 30: 149-153.
  47. Todd JA, Acha-Orbea H, Bell JI, Chao N, Fronek Z, et al. (1988) A molecular basis for MHC class II--associated autoimmunity. Science 240: 1003-1009.
  48. Todd JA (1990) Genetic control of autoimmunity in type 1 diabetes. Immunol Today 11: 122-129.
  49. Acha-Orbea H, McDevitt HO (1987) The first external domain of the nonobese diabetic mouse class II I-A beta chain is unique. Proc Natl Acad Sci U S A 84: 2435-2439.
  50. Wen L, Wong FS, Tang J, Chen NY, Altieri M, et al. (2000) In vivo evidence for the contribution of human histocompatibility leukocyte antigen (HLA)-DQ molecules to the development of diabetes. J Exp Med 191: 97-104.
  51. Zhang XM, Wang HY, Luo YY, Ji LN (2009) HLA-DQ, DR allele polymorphism of type 1 diabetes in the Chinese population: a meta-analysis. Chin Med J (Engl) 122: 980-986.
  52. Akesson K, Carlsson A, Ivarsson SA, Johansson C, Weidby BM, et al. (2009) The non-inherited maternal HLA haplotype affects the risk for type 1 diabetes. Int J Immunogenet 36: 1-8.
  53. Rajagopalan G, Kudva YC, Chen L, Wen L, David CS (2003) Autoimmune diabetes in HLA-DR3/DQ8 transgenic mice expressing the co-stimulatory molecule B7-1 in the beta cells of islets of Langerhans. Int Immunol 15: 1035-1044.
  54. Kudva YC, Rajagopalan G, Raju R, Abraham RS, Smart M, et al. (2002) Modulation of insulitis and type 1 diabetes by transgenic HLA-DR3 and DQ8 in NOD mice lacking endogenous MHC class II. Hum Immunol 63: 987-999.
  55. Blanchard N, Gonzales F, Schaeffer M, Joncker NT, Cheng T, et al. (2008) Immunodominant, protective response to the parasite T. gondii requires antigen processing in the endoplasmic reticulum. Nat Immunol 9: 937-944.
  56. Lüder CG, Lang T, Beuerle B, Gross U (1998) Down-regulation of MHC class II molecules and inability to up-regulate class I molecules in murine macrophages after infection with Toxoplasma gondii. Clin Exp Immunol 112: 308-316.
  57. Lüder CG, Walter W, Beuerle B, Maeurer MJ, Gross U (2001) Toxoplasma gondii down-regulates MHC class II gene expression and antigen presentation by murine macrophages via interference with nuclear translocation of STAT1alpha. Eur J Immunol 31: 1475-1484.
  58. Lüder CG, Lang C, Giraldo-Velasquez M, Algner M, Gerdes J, et al. (2003) Toxoplasma gondii inhibits MHC class II expression in neural antigen-presenting cells by down-regulating the class II transactivator CIITA. J Neuroimmunol 134: 12-24.
  59. Gaji RY, Behnke MS, Lehmann MM, White MW, Carruthers VB (2011) Cell cycle-dependent, intercellular transmission of Toxoplasma gondii is accompanied by marked changes in parasite gene expression. Mol Microbiol 79: 192-204.
  60. Radke JR, Striepen B, Guerini MN, Jerome ME, Roos DS, et al. (2001) Defining the cell cycle for the tachyzoite stage of Toxoplasma gondii. Mol Biochem Parasitol 115: 165-175.
  61. Mack DG, Johnson JJ, Roberts F, Roberts CW, Estes RG, et al. (1999) HLA-class II genes modify outcome of Toxoplasma gondii infection. Int J Parasitol 29: 1351-1358.
  62. Dubey JP, Ferreira LR, Martins J, McLeod R (2012) Oral oocyst-induced mouse model of toxoplasmosis: effect of infection with Toxoplasma gondii strains of different genotypes, dose, and mouse strains (transgenic, out-bred, in-bred) on pathogenesis and mortality. Parasitology 139: 1-13.
  63. McLeod R, Boyer KM, Lee D, Mui E, Wroblewski K, et al. (2012) Prematurity and severity are associated with Toxoplasma gondii alleles (NCCCTS, 1981-2009). Clin Infect Dis 54: 1595-1605.
  64. Han D, Leyva CA, Matheson D, Mineo D, Messinger S, et al. (2011) Immune profiling by multiple gene expression analysis in patients at-risk and with type 1 diabetes. Clin Immunol 139: 290-301.
  65. Alba-Loureiro TC, Munhoz CD, Martins JO, Cerchiaro GA, Scavone C, et al. (2007) Neutrophil function and metabolism in individuals with diabetes mellitus. Braz J Med Biol Res 40: 1037-1044.
  66. Hadi HA, Suwaidi JA (2007) Endothelial dysfunction in diabetes mellitus. Vasc Health Risk Manag 3: 853-876.
  67. Alba-Loureiro TC, Hirabara SM, Mendonça JR, Curi R, Pithon-Curi TC (2006) Diabetes causes marked changes in function and metabolism of rat neutrophils. J Endocrinol 188: 295-303.
  68. Newsholme P, Lima MM, Procopio J, Pithon-Curi TC, Doi SQ, et al. (2003) Glutamine and glutamate as vital metabolites. Braz J Med Biol Res 36: 153-163.
  69. Ohsaka A, Yoshikawa K, Hagiwara T (1982) 1H-NMR spectroscopic study of aerobic glucose metabolism in Toxoplasma gondii harvested from the peritoneal exudate of experimentally infected mice. Physiol Chem Phys 14: 381-384.
  70. Blume M, Rodriguez-Contreras D, Landfear S, Fleige T, Soldati-Favre D, et al. (2009) Host-derived glucose and its transporter in the obligate intracellular pathogen Toxoplasma gondii are dispensable by glutaminolysis. Proc Natl Acad Sci U S A 106: 12998-13003.
  71. Curi TC, De Melo MP, Palanch AC, Miyasaka CK, Curi R (1998) Percentage of phagocytosis, production of O2-, H2O2 and NO, and antioxidant enzyme activities of rat neutrophils in culture. Cell Biochem Funct 16: 43-49.
  72. Ogle CK, Ogle JD, Mao JX, Simon J, Noel JG, et al. (1994) Effect of glutamine on phagocytosis and bacterial killing by normal and pediatric burn patient neutrophils. JPEN J Parenter Enteral Nutr 18: 128-133.
  73. Pithon-Curi TC, Schumacher RI, Freitas JJ, Lagranha C, Newsholme P, et al. (2003) Glutamine delays spontaneous apoptosis in neutrophils. Am J Physiol Cell Physiol 284: C1355-1361.
  74. Kaneshige H, Endoh M, Tomino Y, Nomoto Y, Sakai H, et al. (1982) Impaired granulocyte function in patients with diabetes mellitus. Tokai J Exp Clin Med 7: 77-80.
  75. Wilson RM, Reeves WG (1986) Neutrophil phagocytosis and killing in insulin-dependent diabetes. Clin Exp Immunol 63: 478-484.
  76. Davidson NJ, Sowden JM, Fletcher J (1984) Defective phagocytosis in insulin controlled diabetics: evidence for a reaction between glucose and opsonising proteins. J Clin Pathol 37: 783-786.
  77. Rodacki M, Svoren B, Butty V, Besse W, Laffel L, et al. (2007) Altered natural killer cells in type 1 diabetic patients. Diabetes 56: 177-185.
  78. Colucci F, Caligiuri MA, Di Santo JP (2003) What does it take to make a natural killer? Nat Rev Immunol 3: 413-425.
  79. Baxter AG, Smyth MJ (2002) The role of NK cells in autoimmune disease. Autoimmunity 35: 1-14.
  80. Johansson S, Berg L, Hall H, Höglund P (2005) NK cells: elusive players in autoimmunity. Trends Immunol 26: 613-618.
  81. MacKay P, Jacobson J, Rabinovitch A (1986) Spontaneous diabetes mellitus in the Bio-Breeding/Worcester rat. Evidence in vitro for natural killer cell lysis of islet cells. J Clin Invest 77: 916-924.
  82. Nakamura N, Woda BA, Tafuri A, Greiner DL, Reynolds CW, et al. (1990) Intrinsic cytotoxicity of natural killer cells to pancreatic islets in vitro. Diabetes 39: 836-843.
  83. Rabinovitch A (1998) An update on cytokines in the pathogenesis of insulin-dependent diabetes mellitus. Diabetes Metab Rev 14: 129-151.
  84. Lajoix AD, Reggio H, Chardès T, Péraldi-Roux S, Tribillac F, et al. (2001) A neuronal isoform of nitric oxide synthase expressed in pancreatic beta-cells controls insulin secretion. Diabetes 50: 1311-1323.
  85. Kröncke KD, Rodriguez ML, Kolb H, Kolb-Bachofen V (1993) Cytotoxicity of activated rat macrophages against syngeneic islet cells is arginine-dependent, correlates with citrulline and nitrite concentrations and is identical to lysis by the nitric oxide donor nitroprusside. Diabetologia 36: 17-24.
  86. Adeghate E, Parvez SH (2000) Nitric oxide and neuronal and pancreatic beta cell death. Toxicology 153: 143-156.
  87. Ramadan JW, Steiner SR, O'Neill CM, Nunemaker CS (2011) The central role of calcium in the effects of cytokines on beta-cell function: implications for type 1 and type 2 diabetes. Cell Calcium 50: 481-490.
  88. Sinai AP, Webster P, Joiner KA (1997) Association of host cell endoplasmic reticulum and mitochondria with the Toxoplasma gondii parasitophorous vacuole membrane: a high affinity interaction. J Cell Sci 110 : 2117-2128.
  89. Sibley LD (2010) How apicomplexan parasites move in and out of cells. Curr Opin Biotechnol 21: 592-598.
  90. Nagamune K, Moreno SN, Chini EN, Sibley LD (2008) Calcium regulation and signaling in apicomplexan parasites. Subcell Biochem 47: 70-81.
  91. Tabit CE, Shenouda SM, Holbrook M, Fetterman JL, Kiani S, et al. (2012) Protein Kinase-C Beta Contributes to Impaired Endothelial Insulin Signaling in Humans with Diabetes Mellitus. Circulation 127: 86-95 .
  92. Billker O, Lourido S, Sibley LD (2009) Calcium-dependent signaling and kinases in apicomplexan parasites. Cell Host Microbe 5: 612-622.
  93. Ma GY, Zhang JZ, Yin GR, Zhang JH, Meng XL, et al. (2009) Toxoplasma gondii: proteomic analysis of antigenicity of soluble tachyzoite antigen. Exp Parasitol 122: 41-46.
  94. Johnson SM, Murphy RC, Geiger JA, DeRocher AE, Zhang Z, et al. (2012) Development of Toxoplasma gondii calcium-dependent protein kinase 1 (TgCDPK1) inhibitors with potent anti-toxoplasma activity. J Med Chem 55: 2416-2426.
  95. Kurokawa H, Kato K, Iwanaga T, Sugi T, Sudo A, et al. (2011) Identification of Toxoplasma gondii cAMP dependent protein kinase and its role in the tachyzoite growth. PLoS One 6: e22492.
  96. McCoy JM, Whitehead L, van Dooren GG, Tonkin CJ (2012) TgCDPK3 Regulates Calcium-Dependent Egress of Toxoplasma gondii from Host Cells. PLoS Pathog 8: e1003066.
  97. Leem J, Koh EH (2012) Interaction between mitochondria and the endoplasmic reticulum: implications for the pathogenesis of type 2 diabetes mellitus. Exp Diabetes Res 2012: 242984.
  98. Lowell BB, Shulman GI (2005) Mitochondrial dysfunction and type 2 diabetes. Science 307: 384-387.
  99. Patti ME, Corvera S (2010) The role of mitochondria in the pathogenesis of type 2 diabetes. Endocr Rev 31: 364-395.
  100. Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, et al. (2004) Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 306: 457-461.
  101. Eizirik DL, Cardozo AK, Cnop M (2008) The role for endoplasmic reticulum stress in diabetes mellitus. Endocr Rev 29: 42-61.
  102. Giorgi C, De Stefani D, Bononi A, Rizzuto R, Pinton P (2009) Structural and functional link between the mitochondrial network and the endoplasmic reticulum. Int J Biochem Cell Biol 41: 1817-1827.
  103. Csordás G, Hajnóczky G (2009) SR/ER-mitochondrial local communication: calcium and ROS. Biochim Biophys Acta 1787: 1352-1362.
  104. Malhotra JD, Kaufman RJ (2007) Endoplasmic reticulum stress and oxidative stress: a vicious cycle or a double-edged sword? Antioxid Redox Signal 9: 2277-2293.
  105. Xu W, Liu L, Charles IG, Moncada S (2004) Nitric oxide induces coupling of mitochondrial signalling with the endoplasmic reticulum stress response. Nat Cell Biol 6: 1129-1134.
  106. Brown GC (2001) Regulation of mitochondrial respiration by nitric oxide inhibition of cytochrome c oxidase. Biochim Biophys Acta 1504: 46-57.
  107. Corbett JA, Sweetland MA, Wang JL, Lancaster JR Jr, McDaniel ML (1993) Nitric oxide mediates cytokine-induced inhibition of insulin secretion by human islets of Langerhans. Proc Natl Acad Sci U S A 90: 1731-1735.
  108. Cunningham JM, Green IC (1994) Cytokines, nitric oxide and insulin secreting cells. Growth Regul 4: 173-180.
  109. Cetkovic-Cvrlje M, Eizirik DL (1994) TNF-alpha and IFN-gamma potentiate the deleterious effects of IL-1 beta on mouse pancreatic islets mainly via generation of nitric oxide. Cytokine 6: 399-406.
  110. Corbett JA, Wang JL, Hughes JH, Wolf BA, Sweetland MA, et al. (1992) Nitric oxide and cyclic GMP formation induced by interleukin 1 beta in islets of Langerhans. Evidence for an effector role of nitric oxide in islet dysfunction. Biochem J 287 : 229-235.
  111. Corbett JA, Wang JL, Sweetland MA, Lancaster JR Jr, McDaniel ML (1992) Interleukin 1 beta induces the formation of nitric oxide by beta-cells purified from rodent islets of Langerhans. Evidence for the beta-cell as a source and site of action of nitric oxide. J Clin Invest 90: 2384-2391.
  112. Weaver JR, Holman TR, Imai Y, Jadhav A, Kenyon V, et al. (2012) Integration of pro-inflammatory cytokines, 12-lipoxygenase and NOX-1 in pancreatic islet beta cell dysfunction. Mol Cell Endocrinol 358: 88-95.
  113. Syed I, Kyathanahalli CN, Jayaram B, Govind S, Rhodes CJ, et al. (2011) Increased phagocyte-like NADPH oxidase and ROS generation in type 2 diabetic ZDF rat and human islets: role of Rac1-JNK1/2 signaling pathway in mitochondrial dysregulation in the diabetic islet. Diabetes 60: 2843-2852.
  114. Kim KA, Kim JY, Lee YA, Song KJ, Min D, et al. (2011) NOX1 participates in ROS-dependent cell death of colon epithelial Caco2 cells induced by Entamoeba histolytica. Microbes Infect 13: 1052-1061.
  115. Yoon JW, Jun HS (2005) Autoimmune destruction of pancreatic beta cells. Am J Ther 12: 580-591.
  116. Yoon JW, Jun HS (2001) Cellular and molecular pathogenic mechanisms of insulin-dependent diabetes mellitus. Ann N Y Acad Sci 928: 200-211.
  117. Yoon JW, Jun HS, Santamaria P (1998) Cellular and molecular mechanisms for the initiation and progression of beta cell destruction resulting from the collaboration between macrophages and T cells. Autoimmunity 27: 109-122.
  118. Wachlin G, Augstein P, Schröder D, Kuttler B, Klöting I, et al. (2003) IL-1beta, IFN-gamma and TNF-alpha increase vulnerability of pancreatic beta cells to autoimmune destruction. J Autoimmun 20: 303-312.
  119. Bruzzone S, Bodrato N, Usai C, Guida L, Moreschi I, et al. (2008) Abscisic acid is an endogenous stimulator of insulin release from human pancreatic islets with cyclic ADP ribose as second messenger. J Biol Chem 283: 32188-32197.
  120. Bruzzone S, Moreschi I, Usai C, Guida L, Damonte G, et al. (2007) Abscisic acid is an endogenous cytokine in human granulocytes with cyclic ADP-ribose as second messenger. Proc Natl Acad Sci U S A 104: 5759-5764.
  121. Nagamune K, Hicks LM, Fux B, Brossier F, Chini EN, et al. (2008) Abscisic acid controls calcium-dependent egress and development in Toxoplasma gondii. Nature 451: 207-210.
  122. Li HH, Hao RL, Wu SS, Guo PC, Chen CJ, et al. (2011) Occurrence, function and potential medicinal applications of the phytohormone abscisic acid in animals and humans. Biochem Pharmacol 82: 701-712.
  123. Wang Y, Weiss LM, Orlofsky A (2009) Host cell autophagy is induced by Toxoplasma gondii and contributes to parasite growth. J Biol Chem 284: 1694-1701.
  124. Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, et al. (2003) Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52: 102-110.
  125. Butler AE, Janson J, Soeller WC, Butler PC (2003) Increased beta-cell apoptosis prevents adaptive increase in beta-cell mass in mouse model of type 2 diabetes: evidence for role of islet amyloid formation rather than direct action of amyloid. Diabetes 52: 2304-2314.
  126. Parenti DM, Steinberg W, Kang P (1996) Infectious causes of acute pancreatitis. Pancreas 13: 356-371.
  127. Kankova S, Holan V, Zajicova A, Kodym P, Flegr J (2010) Modulation of immunity in mice with latent toxoplasmosis – the experimental support for the immunosuppression hypothesis of Toxoplasma-induced changes in reproduction of mice and humans. Parasitol Res 107: 1421-1427.
  128. Miller CM, Smith NC, Johnson AM (1999) Cytokines, nitric oxide, heat shock proteins and virulence in Toxoplasma. Parasitol Today 15: 418-422.
  129. Gazzinelli RT, Hieny S, Wynn TA, Wolf S, Sher A (1993) Interleukin 12 is required for the T-lymphocyte-independent induction of interferon gamma by an intracellular parasite and induces resistance in T-cell-deficient hosts. Proc Natl Acad Sci U S A 90: 6115-6119.
  130. Hunter CA, Bermudez L, Beernink H, Waegell W, Remington JS (1995) Transforming growth factor-beta inhibits interleukin-12-induced production of interferon-gamma by natural killer cells: a role for transforming growth factor-beta in the regulation of T cell-independent resistance to Toxoplasma gondii. Eur J Immunol 25: 994-1000.
  131. Sher A, Sousa CR (1998) Ignition of the type 1 response to intracellular infection by dendritic cell-derived interleukin-12. Eur Cytokine Netw 9: 65-68.
  132. Suzuki Y, Orellana MA, Schreiber RD, Remington JS (1988) Interferon-gamma: the major mediator of resistance against Toxoplasma gondii. Science 240: 516-518.
  133. Pfefferkorn ER, Eckel M, Rebhun S (1986) Interferon-gamma suppresses the growth of Toxoplasma gondii in human fibroblasts through starvation for tryptophan. Mol Biochem Parasitol 20: 215-224.
  134. Gazzinelli RT, Wysocka M, Hayashi S, Denkers EY, Hieny S, et al. (1994) Parasite-induced IL-12 stimulates early IFN-gamma synthesis and resistance during acute infection with Toxoplasma gondii. J Immunol 153: 2533-2543.
  135. Däubener W, Remscheid C, Nockemann S, Pilz K, Seghrouchni S, et al. (1996) Anti-parasitic effector mechanisms in human brain tumor cells: role of interferon-gamma and tumor necrosis factor-alpha. Eur J Immunol 26: 487-492.
  136. Sibley LD, Adams LB, Fukutomi Y, Krahenbuhl JL (1991) Tumor necrosis factor-alpha triggers antitoxoplasmal activity of IFN-gamma primed macrophages. J Immunol 147: 2340-2345.
  137. Sher A, Oswald IP, Hieny S, Gazzinelli RT (1993) Toxoplasma gondii induces a T-independent IFN-gamma response in natural killer cells that requires both adherent accessory cells and tumor necrosis factor-alpha. J Immunol 150: 3982-3989.
  138. Langermans JA, Van der Hulst ME, Nibbering PH, Hiemstra PS, Fransen L, et al. (1992) IFN-gamma-induced L-arginine-dependent toxoplasmastatic activity in murine peritoneal macrophages is mediated by endogenous tumor necrosis factor-alpha. J Immunol 148: 568-574.
  139. Xie QW, Kashiwabara Y, Nathan C (1994) Role of transcription factor NF-kappa B/Rel in induction of nitric oxide synthase. J Biol Chem 269: 4705-4708.
  140. Gómez-Marín JE (2000) No NO production during human Toxoplasma infection. Parasitol Today 16: 131.
  141. Ghigo D, Todde R, Ginsburg H, Costamagna C, Gautret P, et al. (1995) Erythrocyte stages of Plasmodium falciparum exhibit a high nitric oxide synthase (NOS) activity and release an NOS-inducing soluble factor. J Exp Med 182: 677-688.
  142. Paveto C, Pereira C, Espinosa J, Montagna AE, Farber M, et al. (1995) The nitric oxide transduction pathway in Trypanosoma cruzi. J Biol Chem 270: 16576-16579.
  143. Engin AB, Dogruman-Al F, Ercin U, Celebi B, Babur C, et al. (2012) Oxidative stress and tryptophan degradation pattern of acute Toxoplasma gondii infection in mice. Parasitol Res 111: 1725-1730.
  144. Liesenfeld O, Kang H, Park D, Nguyen TA, Parkhe CV, et al. (1999) TNF-alpha, nitric oxide and IFN-gamma are all critical for development of necrosis in the small intestine and early mortality in genetically susceptible mice infected perorally with Toxoplasma gondii. Parasite Immunol 21: 365-376.
  145. Adams LB, Hibbs JB Jr, Taintor RR, Krahenbuhl JL (1990) Microbiostatic effect of murine-activated macrophages for Toxoplasma gondii. Role for synthesis of inorganic nitrogen oxides from L-arginine. J Immunol 144: 2725-2729.
  146. Dupont CD, Christian DA, Hunter CA (2012) Immune response and immunopathology during toxoplasmosis. Semin Immunopathol 34: 793-813.
  147. Liu CH, Fan YT, Dias A, Esper L, Corn RA, et al. (2006) Cutting edge: dendritic cells are essential for in vivo IL-12 production and development of resistance against Toxoplasma gondii infection in mice. J Immunol 177: 31-35.
  148. Hou B, Benson A, Kuzmich L, DeFranco AL, Yarovinsky F (2011) Critical coordination of innate immune defense against Toxoplasma gondii by dendritic cells responding via their Toll-like receptors. Proc Natl Acad Sci U S A 108: 278-283.
  149. Mashayekhi M, Sandau MM, Dunay IR, Frickel EM, Khan A, et al. (2011) CD8a(+) dendritic cells are the critical source of interleukin-12 that controls acute infection by Toxoplasma gondii tachyzoites. Immunity 35: 249-259.
  150. Wilson DC, Matthews S, Yap GS (2008) IL-12 signaling drives CD8+ T cell IFN-gamma production and differentiation of KLRG1+ effector subpopulations during Toxoplasma gondii Infection. J Immunol 180: 5935-5945.
  151. Bliss SK, Marshall AJ, Zhang Y, Denkers EY (1999) Human polymorphonuclear leukocytes produce IL-12, TNF-alpha, and the chemokines macrophage-inflammatory protein-1 alpha and -1 beta in response to Toxoplasma gondii antigens. J Immunol 162: 7369-7375.
  152. Whitmarsh RJ, Gray CM, Gregg B, Christian DA, May MJ, et al. (2011) A critical role for SOCS3 in innate resistance to Toxoplasma gondii. Cell Host Microbe 10: 224-236.
  153. Bliss SK, Zhang Y, Denkers EY (1999) Murine neutrophil stimulation by Toxoplasma gondii antigen drives high level production of IFN-gamma-independent IL-12. J Immunol 35: 2081-2088.
  154. Hunter CA, Subauste CS, Van Cleave VH, Remington JS (1994) Production of gamma interferon by natural killer cells from Toxoplasma gondii-infected SCID mice: regulation by interleukin-10, interleukin-12, and tumor necrosis factor alpha. Infect Immun 62: 2818-2824.
  155. Mordue DG, Sibley LD (2003) A novel population of Gr-1+-activated macrophages induced during acute toxoplasmosis. J Leukoc Biol 74: 1015-1025.
  156. Johnson LL, VanderVegt FP, Havell EA (1993) Gamma interferon-dependent temporary resistance to acute Toxoplasma gondii infection independent of CD4+ or CD8+ lymphocytes. Infect Immun 61: 5174-5180.
  157. Denkers EY, Gazzinelli RT, Martin D, Sher A (1993) Emergence of NK1.1+ cells as effectors of IFN-gamma dependent immunity to Toxoplasma gondii in MHC class I-deficient mice. J Exp Med 178: 1465-1472.
  158. Gazzinelli R, Xu Y, Hieny S, Cheever A, Sher A (1992) Simultaneous depletion of CD4+ and CD8+ T lymphocytes is required to reactivate chronic infection with Toxoplasma gondii. J Immunol 149:175-180.
  159. Howard JC, Hunn JP, Steinfeldt T (2011) The IRG protein-based resistance mechanism in mice and its relation to virulence in Toxoplasma gondii. Curr Opin Microbiol 14: 414-421.
  160. Taylor GA, Collazo CM, Yap GS, Nguyen K, Gregorio TA, et al. (2000) Pathogen-specific loss of host resistance in mice lacking the IFN-gamma-inducible gene IGTP. Proc Natl Acad Sci U S A 97: 751-755.
  161. Pfefferkorn ER (1984) Interferon gamma blocks the growth of Toxoplasma gondii in human fibroblasts by inducing the host cells to degrade tryptophan. Proc Natl Acad Sci U S A 81: 908-912.
  162. Murray HW, Szuro-Sudol A, Wellner D, Oca MJ, Granger AM, et al. (1989) Role of tryptophan degradation in respiratory burst-independent antimicrobial activity of gamma interferon-stimulated human macrophages. Infect Immun 57: 845-849.
  163. Däubener W, Spors B, Hucke C, Adam R, Stins M, et al. (2001) Restriction of Toxoplasma gondii growth in human brain microvascular endothelial cells by activation of indoleamine 2,3-dioxygenase. Infect Immun 69: 6527-6531.
  164. Bennouna S, Bliss SK, Curiel TJ, Denkers EY (2003) Cross-talk in the innate immune system: neutrophils instruct recruitment and activation of dendritic cells during microbial infection. J Immunol 171: 6052-6058.
  165. Chang HR, Grau GE, Pechère JC (1990) Role of TNF and IL-1 in infections with Toxoplasma gondii. Immunology 69: 33-37.
  166. Yap GS, Sher A (1999) Effector cells of both nonhemopoietic and hemopoietic origin are required for interferon (IFN)-gamma- and tumor necrosis factor (TNF)-alpha-dependent host resistance to the intracellular pathogen, Toxoplasma gondii. J Exp Med 189: 1083-1092.
  167. Li ZY, Manthey CL, Perera PY, Sher A, Vogel SN (1994) Toxoplasma gondii soluble antigen induces a subset of lipopolysaccharide-inducible genes and tyrosine phosphoproteins in peritoneal macrophages. Infect Immun 62: 3434-3440.
  168. Schlüter D, Meyer T, Strack A, Reiter S, Kretschmar M, et al. (2001) Regulation of microglia by CD4+ and CD8+ T cells: selective analysis in CD45-congenic normal and Toxoplasma gondii-infected bone marrow chimeras. Brain Pathol 11: 44-55.
  169. Deckert-Schlüter M, Bluethmann H, Rang A, Hof H, Schlüter D (1998) Crucial role of TNF receptor type 1 (p55), but not of TNF receptor type 2 (p75), in murine toxoplasmosis. J Immunol 160: 3427-3436.
  170. Schlüter D, Kwok LY, Lütjen S, Soltek S, Hoffmann S, et al. (2003) Both lymphotoxin-alpha and TNF are crucial for control of Toxoplasma gondii in the central nervous system. J Immunol 170: 6172-6182.
  171. Gazzinelli RT, Eltoum I, Wynn TA, Sher A (1993) Acute cerebral toxoplasmosis is induced by in vivo neutralization of TNF-alpha and correlates with the down-regulated expression of inducible nitric oxide synthase and other markers of macrophage activation. J Immunol 151: 3672-3681.
  172. Schlüter D, Kaefer N, Hof H, Wiestler OD, Deckert-Schlüter M (1997) Expression pattern and cellular origin of cytokines in the normal and Toxoplasma gondii-infected murine brain. Am J Pathol 150: 1021-1035.
  173. Pelloux H, Pernod G, Ricard J, Renversez TC, Ambroise-Thomas P (1994) Interleukin-6 is secreted by human monocytes after stimulation with anti-Toxoplasma gondii sera. J Infect Dis 169: 1181-1182.
  174. Jebbari H, Roberts CW, Ferguson DJ, Bluethmann H, Alexander J (1998) A protective role for IL-6 during early infection with Toxoplasma gondii. Parasite Immunol 20: 231-239.
  175. Fischer HG, Nitzgen B, Reichmann G, Hadding U (1997) Cytokine responses induced by Toxoplasma gondii in astrocytes and microglial cells. Eur J Immunol 27: 1539-1548.
  176. Suzuki Y, Rani S, Liesenfeld O, Kojima T, Lim S, et al. (1997) Impaired resistance to the development of toxoplasmic encephalitis in interleukin-6-deficient mice. Infect Immun 65: 2339-2345.
  177. Chou DB, Sworder B, Bouladoux N, Roy CN, Uchida AM, et al. (2012) Stromal-derived IL-6 alters the balance of myeloerythroid progenitors during Toxoplasma gondii infection. J Leukoc Biol 92: 123-131.
  178. Nagineni CN, Detrick B, Hooks JJ (2000) Toxoplasma gondii infection induces gene expression and secretion of interleukin 1 (IL-1), IL-6, granulocyte-macrophage colony-stimulating factor, and intercellular adhesion molecule 1 by human retinal pigment epithelial cells. Infect Immun 68: 407-410.
  179. Browning JL, Sizing ID, Lawton P, Bourdon PR, Rennert PD, et al. (1997) Characterization of lymphotoxin-alpha beta complexes on the surface of mouse lymphocytes. J Immunol 159: 3288-3298.
  180. De Togni P, Goellner J, Ruddle NH, Streeter PR, Fick A, et al. (1994) Abnormal development of peripheral lymphoid organs in mice deficient in lymphotoxin. Science 264: 703-707.
  181. Perona-Wright G, Mohrs K, Szaba FM, Kummer LW, Madan R, et al. (2009) Systemic but not local infections elicit immunosuppressive IL-10 production by natural killer cells. Cell Host Microbe 6: 503-512.
  182. Gazzinelli RT, Wysocka M, Hieny S, Scharton-Kersten T, Cheever A, et al. (1996) In the absence of endogenous IL-10, mice acutely infected with Toxoplasma gondii succumb to a lethal immune response dependent on CD4+ T cells and accompanied by overproduction of IL-12, IFN-gamma and TNF-alpha. J Immunol 157: 798-805.
  183. Deckert-Schlüter M, Buck C, Weiner D, Kaefer N, Rang A, et al. (1997) Interleukin-10 downregulates the intracerebral immune response in chronic Toxoplasma encephalitis. J Neuroimmunol 76: 167-176.
  184. Jankovic D, Kullberg MC, Feng CG, Goldszmid RS, Collazo CM, et al. (2007) Conventional T-bet(+)Foxp3(-) Th1 cells are the major source of host-protective regulatory IL-10 during intracellular protozoan infection. J Exp Med 204: 273-283.
  185. Pflanz S, Timans JC, Cheung J, Rosales R, Kanzler H, et al. (2002) IL-27, a heterodimeric cytokine composed of EB13 and p28 protein, induces proliferation of naive CD4(+) T cells. Immunity 16: 779-790.
  186. Stumhofer JS, Laurence A, Wilson EH, Huang E, Tato CM, et al. (2006) Interleukin 27 negatively regulates the development of interleukin 17-producing T helper cells during chronic inflammation of the central nervous system. Nat Immunol 7: 937-945.
  187. Villarino A, Hibbert L, Lieberman L, Wilson E, Mak T, et al. (2003) The IL-27R (WSX-1) is required to suppress T cell hyperactivity during infection. Immunity 19: 645-655.
  188. Stumhofer JS, Silver JS, Laurence A, Porrett PM, Harris TH, et al. (2007) Interleukins 27 and 6 induce STAT3-mediated T cell production of interleukin 10. Nat Immunol 8: 1363-1371.
  189. Hirahara K, Ghoreschi K, Yang XP, Takahashi H, Laurence A, et al. (2012) Interleukin-27 priming of T cells controls IL-17 production in trans via induction of the ligand PD-L1. Immunity 36: 1017-1030.
  190. Banchereau J, Bazan F, Blanchard D, Brière F, Galizzi JP, et al. (1994) The CD40 antigen and its ligand. Annu Rev Immunol 12: 881-922.
  191. Subauste CS, Wessendarp M, Sorensen RU, Leiva LE (1999) CD40-CD40 ligand interaction is central to cell-mediated immunity against Toxoplasma gondii: patients with hyper IgM syndrome have a defective type 1 immune response that can be restored by soluble CD40 ligand trimer. J Immunol 162: 6690-6700.
  192. Jana M, Liu X, Koka S, Ghosh S, Petro TM, et al. (2001) Ligation of CD40 stimulates the induction of nitric-oxide synthase in microglial cells. J Biol Chem 276: 44527-44533.
  193. Portillo JA, Okenka G, Reed E, Subauste A, Van Grol J, et al. (2010) The CD40-autophagy pathway is needed for host protection despite IFN-Γ-dependent immunity and CD40 induces autophagy via control of P21 levels. PLoS One 5: e14472.
  194. Seabra SH, de Souza W, DaMatta RA (2002) Toxoplasma gondii partially inhibits nitric oxide production of activated murine macrophages. Exp Parasitol 100: 62-70.
  195. Lüder CG, Algner M, Lang C, Bleicher N, Gross U (2003) Reduced expression of the inducible nitric oxide synthase after infection with Toxoplasma gondii facilitates parasite replication in activated murine macrophages. Int J Parasitol 33: 833-844.
  196. Prandovszky E, Gaskell E, Martin H, Dubey JP, Webster JP, et al. (2011) The neurotropic parasite Toxoplasma gondii increases dopamine metabolism. PLoS One 6: e23866.
  197. Silva NM, Rodrigues CV, Santoro MM, Reis LF, Alvarez-Leite JI, et al. (2002) Expression of indoleamine 2,3-dioxygenase, tryptophan degradation, and kynurenine formation during in vivo infection with Toxoplasma gondii: induction by endogenous gamma interferon and requirement of interferon regulatory factor 1. Infect Immun 70: 859-868.
  198. Baban B, Chandler PR, Sharma MD, Pihkala J, Koni PA, et al. (2009) IDO activates regulatory T cells and blocks their conversion into Th17-like T cells. J Immunol 183: 2475-2483.
  199. Mortensen PB, Nørgaard-Pedersen B, Waltoft BL, Sørensen TL, Hougaard D, et al. (2007) Early infections of Toxoplasma gondii and the later development of schizophrenia. Schizophr Bull 33: 741-744.
  200. Stone TW, Darlington LG (2002) Endogenous kynurenines as targets for drug discovery and development. Nat Rev Drug Discov 1: 609-620.
  201. Thomas SR, Mohr D, Stocker R (1994) Nitric oxide inhibits indoleamine 2,3-dioxygenase activity in interferon-gamma primed mononuclear phagocytes. J Biol Chem 269: 14457-14464.
  202. Høyer-Hansen M, Nordbrandt SP, Jäättelä M (2010) Autophagy as a basis for the health-promoting effects of vitamin D. Trends Mol Med 16: 295-302.
  203. Andrade RM, Wessendarp M, Gubbels MJ, Striepen B, Subauste CS (2006) CD40 induces macrophage anti-Toxoplasma gondii activity by triggering autophagy-dependent fusion of pathogen-containing vacuoles and lysosomes. J Clin Invest 116: 2366-2377.
  204. Xu Y, Jagannath C, Liu XD, Sharafkhaneh A, Kolodziejska KE, et al. (2007) Toll-like receptor 4 is a sensor for autophagy associated with innate immunity. Immunity 27: 135-144.
  205. Gutierrez MG, Master SS, Singh SB, Taylor GA, Colombo MI, et al. (2004) Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell 119: 753-766.
  206. Coppens I, Dunn JD, Romano JD, Pypaert M, Zhang H, et al. (2006) Toxoplasma gondii sequesters lysosomes from mammalian hosts in the vacuolar space. Cell 125: 261-274.
  207. Sibley LD, Weidner E, Krahenbuhl JL (1985) Phagosome acidification blocked by intracellular Toxoplasma gondii. Nature 315: 416-419.
  208. Won SJ, Kim JH, Yoo BH, Sohn M, Kauppinen TM, et al. (2012) Prevention of hypoglycemia-induced neuronal death by minocycline. J Neuroinflammation 9: 225.
  209. Karaman U, Celik T, Kiran TR, Colak C, Daldal NU (2008) Malondialdehyde, glutathione, and nitric oxide levels in Toxoplasma gondii seropositive patients. Korean J Parasitol 46: 293-295.
  210. Dzitko K, Malicki S, Komorowski J (2008) Effect of hyperprolactinaemia on Toxoplasma gondii prevalence in humans. Parasitol Res 102: 723-729.
  211. Matalka KZ (2003) Prolactin enhances production of interferon-gamma, interleukin-12, and interleukin-10, but not of tumor necrosis factor-alpha, in a stimulus-specific manner. Cytokine 21: 187-194.
  212. Dzitko K, Gatkowska J, Plocinski P, Dziadek B, Dlugonska H (2010) The effect of prolactin (PRL) on the growth of Toxoplasma gondii tachyzoites in vitro. Parasitol Res 107: 199-204.
  213. Prandota J (2009) Neuropathological changes and clinical features of austism spectrum disorder participants are similar to that reported in congenital and chronic cerebral toxoplasmosis in humans and mice. Res Autism Spectr Disord 4: 103-118.
  214. Prandota J (2010) 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 4: 119-155.
  215. Prandota J (2012) Idiopathic intracranial hypertension may be caused by reactivation of latent cerebral toxoplasmosis probably because of disturbances in the host and/or Toxoplasma gondii immune defense mechanisms. Effect of various medications and biologic agents. In: Gemma C (ed). Neuroinflammation. Pathogenesis, Mechanisms, and Management. Nova Science Publishers, New York: 273-336.
  216. Prandota J (2012) Idiopathic intracranial hypertension may be caused by reactivation of latent cerebral toxoplasmosis probably because of disturbances in the host and/or Toxoplasma gondii immune defense mechanisms. Effect of various diseases and clinical states. In: Gemma C (ed). Neuroinflammation: Pathogenesis, Mechanisms and Management. Nova Science Publishers, New York: 337-343.
  217. Lang C, Gross U, Lüder CG (2007) Subversion of innate and adaptive immune responses by Toxoplasma gondii. Parasitol Res 100: 191-203.
  218. Khan IA, Matsuura T, Kasper LH (1995) IL-10 mediates immunosuppression following primary infection with Toxoplasma gondii in mice. Parasite Immunol 17: 185-195.
  219. Neyer LE, Grunig G, Fort M, Remington JS, Rennick D, et al. (1997) Role of interleukin-10 in regulation of T-cell-dependent and T-cell-independent mechanisms of resistance to Toxoplasma gondii. Infect Immun 65: 1675-1682.
  220. Bermudez LE, Covaro G, Remington J (1993) Infection of murine macrophages with Toxoplasma gondii is associated with release of transforming growth factor beta and downregulation of expression of tumor necrosis factor receptors. Infect Immun 61: 4126-4130.
  221. Langermans JA, Nibbering PH, Van Vuren-Van der Hulst ME, Van Furth R (2001) Transforming growth factor-ß suppresses interferon-γ-induced toxoplasmastatic activity in murine macrophages by inhibition of tumour necrosis factor-a production. Parasite Immunol 23: 169-175.
  222. Diez B, Galdeano A, Nicolas R, Cisterna R (1989) Relationship between the production of interferon-alpha/beta and interferon-gamma during acute toxoplasmosis. Parasitology 99 Pt 1: 11-15.
  223. Butcher BA, Denkers EY (2002) Mechanism of entry determines the ability of Toxoplasma gondii to inhibit macrophage proinflammatory cytokine production. Infect Immun 70: 5216-5224.
  224. Shapira S, Speirs K, Gerstein A, Caamano J, Hunter CA (2002) Suppression of NF-κB activation by infection with Toxoplasma gondii. J Infect Dis 185: S66-S72.
  225. Shapira S, Harb OS, Margarit J, Matrajt M, Han J, et al. (2005) Initiation and termination of NF-kappaB signaling by the intracellular protozoan parasite Toxoplasma gondii. J Cell Sci 118: 3501-3508.
  226. Butcher BA, Kim L, Panopoulos AD, Watowich SS, Murray PJ, et al. (2005) IL-10-independent STAT3 activation by Toxoplasma gondii mediates suppression of IL-12 and TNF-alpha in host macrophages. J Immunol 174: 3148-3152.
  227. Aliberti J, Hieny S, Reis e Sousa C, Serhan CN, Sher A (2002) Lipoxin-mediated inhibition of IL-12 production by DCs: a mechanism for regulation of microbial immunity. Nat Immunol 3: 76-82.
  228. Aliberti J, Serhan C, Sher A (2002) Parasite-induced lipoxin A4 is an endogenous regulator of IL-12 production and immunopathology in Toxoplasma gondii infection. J Exp Med 196: 1253-1262.
  229. McKee AS, Dzierszinski F, Boes M, Roos DS, Pearce EJ (2004) Functional inactivation of immature dendritic cells by the intracellular parasite Toxoplasma gondii. J Immunol 173: 2632-2640.
  230. Rozenfeld C, Martinez R, Figueiredo RT, Bozza MT, Lima FR, et al. (2003) Soluble factors released by Toxoplasma gondii-infected astrocytes down-modulate nitric oxide production by gamma interferon-activated microglia and prevent neuronal degeneration. Infect Immun 71: 2047-2057.
  231. Rozenfeld C, Martinez R, Seabra S, Sant'anna C, Gonçalves JG, et al. (2005) Toxoplasma gondii prevents neuron degeneration by interferon-gamma-activated microglia in a mechanism involving inhibition of inducible nitric oxide synthase and transforming growth factor-beta1 production by infected microglia. Am J Pathol 167: 1021-1031.
  232. Butcher BA, Greene RI, Henry SC, Annecharico KL, Weinberg JB, et al. (2005) p47 GTPases regulate Toxoplasma gondii survival in activated macrophages. Infect Immun 73: 3278-3286.
  233. Shrestha SP, Tomita T, Weiss LM, Orlofsky A (2006) Proliferation of Toxoplasma gondii in inflammatory macrophages in vivo is associated with diminished oxygen radical production in the host cell. Int J Parasitol 36: 433-441.
  234. Guglietta S, Beghetto E, Spadoni A, Buffolano W, Del Porto P, et al. (2007) Age-dependent impairment of functional helper T cell responses to immunodominant epitopes of Toxoplasma gondii antigens in congenitally infected individuals. Microbes Infect 9: 127-133.
  235. Ge YY, Zhang L, Zhang G, Wu JP, Tan MJ, et al. (2008) In pregnant mice, the infection of Toxoplasma gondii causes the decrease of CD4+CD25+ -regulatory T cells. Parasite Immunol 30: 471-481.
  236. Clerici M, DePalma L, Roilides E, Baker R, Shearer GM (1993) Analysis of T helper and antigen-presenting cell functions in cord blood and peripheral blood leukocytes from healthy children of different ages. J Clin Invest 91: 2829-2836.
  237. Khan IA, Matsuura T, Kasper LH (1996) Activation-mediated CD4+ T cell unresponsiveness during acute Toxoplasma gondii infection in mice. Int Immunol 8: 887-896.
  238. Liesenfeld O, Kosek JC, Suzuki Y (1997) Gamma interferon induces Fas-dependent apoptosis of Peyer's patch T cells in mice following peroral infection with Toxoplasma gondii. Infect Immun 65: 4682-4689.
  239. Gavrilescu LC, Denkers EY (2001) IFN-gamma overproduction and high level apoptosis are associated with high but not low virulence Toxoplasma gondii infection. J Immunol 167: 902-909.
  240. Mordue DG, Monroy F, La Regina M, Dinarello CA, Sibley LD (2001) Acute toxoplasmosis leads to lethal overproduction of Th1 cytokines. J Immunol 167: 4574-4584.
  241. Nash PB, Purner MB, Leon RP, Clarke P, Duke RC, et al. (1998) Toxoplasma gondii-infected cells are resistant to multiple inducers of apoptosis. J Immunol 160: 1824-1830.
  242. Goebel S, Lüder CG, Gross U (1999) Invasion by Toxoplasma gondii protects human-derived HL-60 cells from actinomycin D-induced apoptosis. Med Microbiol Immunol 187: 221-226.
  243. Payne TM, Molestina RE, Sinai AP (2003) Inhibition of caspase activation and a requirement for NF-kappaB function in the Toxoplasma gondii-mediated blockade of host apoptosis. J Cell Sci 116: 4345-4358.
  244. Molestina RE, Payne TM, Coppens I, Sinai AP (2003) Activation of NF-kappaB by Toxoplasma gondii correlates with increased expression of antiapoptotic genes and localization of phosphorylated IkappaB to the parasitophorous vacuole membrane. J Cell Sci 116: 4359-4371.
  245. Keller P, Schaumburg F, Fischer SF, Häcker G, Gross U, et al. (2006) Direct inhibition of cytochrome c-induced caspase activation in vitro by Toxoplasma gondii reveals novel mechanisms of interference with host cell apoptosis. FEMS Microbiol Lett 258: 312-319.
  246. Goebel S, Gross U, Lüder CG (2001) Inhibition of host cell apoptosis by Toxoplasma gondii is accompanied by reduced activation of the caspase cascade and alterations of poly(ADP-ribose) polymerase expression. J Cell Sci 114: 3495-3505.
  247. Channon JY, Miselis KA, Minns LA, Dutta C, Kasper LH (2002) Toxoplasma gondii induces granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor secretion by human fibroblasts: implications for neutrophil apoptosis. Infect Immun 70: 6048-6057.
  248. Abousamra Sr NK (2008) Toxoplasma gondii antibodies and associated oxidative stress among asymptomatic blood donors. Ann Emerg Med 52: S138.
  249. Elsheikha HM, El-Motayam MH, Abouel-Nour MF, Morsy AT (2009) Oxidative stress and immune-suppression in Toxoplasma gondii positive blood donors: implications for safe blood transfusion. J Egypt Soc Parasitol 39: 421-428.
  250. Mihara M, Uchiyama M (1978) Determination of malonaldehyde precursor in tissues by thiobarbituric acid test. Anal Biochem 86: 271-278.
  251. Oswald IP, Wynn TA, Sher A, James SL (1994) NO as an effector molecule of parasite killing: modulation of its synthesis by cytokines. Comp Biochem Physiol Pharmacol Toxicol Endocrinol 108: 11-18.
  252. Baltaci AK, Mogulkoc R (2012) Plasma leptin levels in rats with induced Toxoplasma gondii infection. Bratisl Lek Listy 113: 67-69.
  253. Aygun AD, Gungor S, Ustundag B, Gurgoze MK, Sen Y (2005) Proinflammatory cytokines and leptin are increased in serum of prepubertal obese children. Mediators Inflamm 2005: 180-183.
  254. Kapiotis S, Holzer G, Schaller G, Haumer M, Widhalm H, et al. (2006) A proinflammatory state is detectable in obese children and is accompanied by functional and morphological vascular changes. Arterioscler Thromb Vasc Biol 26: 2541-2546.
  255. Lord GM, Matarese G, Howard JK, Bloom SR, Lechler RI (2002) Leptin inhibits the anti-CD3-driven proliferation of peripheral blood T cells but enhances the production of proinflammatory cytokines. J Leukoc Biol 72: 330-338.
  256. Asensio CS, Arsenijevic D, Lehr L, Giacobino JP, Muzzin P, et al. (2008) Effects of leptin on energy metabolism in beta-less mice. Int J Obes (Lond) 32: 936-942.
  257. Jones JL, Lopez A, Wilson M, Schulkin J, Gibbs R (2001) Congenital toxoplasmosis: a review. Obstet Gynecol Surv 56: 296-305.
  258. Barragan A, Sibley LD (2003) Migration of Toxoplasma gondii across biological barriers. Trends Microbiol 11: 426-430.
  259. Khosrotehrani K, Stroh H, Bianchi DW, Johnson KL (2003) Combined FISH and immunolabelling on paraffin-embedded tissue sections for the study of microchimerism. Biotechniques 34: 242-244.
  260. Foulis AK, Liddle CN, Farquharson MA, Richmond JA, Weir RS (1986) The histopathology of the pancreas in type 1 (insulin-dependent) diabetes mellitus: a 25-year review of deaths in patients under 20 years of age in the United Kingdom. Diabetologia 29: 267-274.
  261. Loubière LS, Lambert NC, Flinn LJ, Erickson TD, Yan Z, et al. (2006) Maternal microchimerism in healthy adults in lymphocytes, monocyte/macrophages and NK cells. Lab Invest 86: 1185-1192.
  262. vanZyl B, Gillespie KM (2009) Microchimerism in type 1 diabetes. Curr Diab Rep 9: 125-129.
  263. Zhou L, Yoshimura Y, Huang Y, Suzuki R, Yokoyama M, et al. (2000) Two independent pathways of maternal cell transmission to offspring: through placenta during pregnancy and by breast-feeding after birth. Immunology 101: 570-580.
  264. Krabchi K, Gros-Louis F, Yan J, Bronsard M, Massé J, et al. (2001) Quantification of all fetal nucleated cells in maternal blood between the 18th and 22nd weeks of pregnancy using molecular cytogenetic techniques. Clin Genet 60: 145-150.
  265. Maloney S, Smith A, Furst DE, Myerson D, Rupert K, et al. (1999) Microchimerism of maternal origin persists into adult life. J Clin Invest 104: 41-47.
  266. Nguyen Huu S, Dubernard G, Aractingi S, Khosrotehrani K (2006) Feto-maternal cell trafficking: a transfer of pregnancy associated progenitor cells. Stem Cell Rev 2: 111-116.
  267. Bianchi DW, Zickwolf GK, Weil GJ, Sylvester S, DeMaria MA (1996) Male fetal progenitor cells persist in maternal blood for as long as 27 years postpartum. Proc Natl Acad Sci U S A 93: 705-708.
  268. Lambert NC, Lo YM, Erickson TD, Tylee TS, Guthrie KA, et al. (2002) Male microchimerism in healthy women and women with scleroderma: cells or circulating DNA? A quantitative answer. Blood 100: 2845-2851.
  269. Burastero SE, Galbiati S, Vassallo A, Sabbadini MG, Bellone M, et al. (2003) Cellular microchimerism as a lifelong physiologic status in parous women: an immunologic basis for its amplification in patients with systemic sclerosis. Arthritis Rheum 48: 1109-1116.
  270. Khosrotehrani K, Leduc M, Bachy V, Nguyen Huu S, Oster M, et al. (2008) Pregnancy allows the transfer and differentiation of fetal lymphoid progenitors into functional T and B cells in mothers. J Immunol 180: 889-897.
  271. Leduc M, Aractingi S, Khosrotehrani K (2009) Fetal-cell microchimerism, lymphopoiesis, and autoimmunity. Arch Immunol Ther Exp (Warsz) 57: 325-329.
  272. Lee ES, Bou-Gharios G, Seppanen E, Khosrotehrani K, Fisk NM (2010) Fetal stem cell microchimerism: natural-born healers or killers? Mol Hum Reprod 16: 869-878.
  273. O’Donoghue K, Chan J, De La FJ, Kennea N, Sandison A, et al. (2004) Microchimerism in female bone marrow and bone decades after fetal mesenchymal stem-cell trafficking in pregnancy. Lancet 364: 179-182.
  274. Nguyen Huu S, Oster M, Uzan S, Chareyre F, Aractingi S, et al. (2007) Maternal neoangiogenesis during pregnancy partly derives from fetal endothelial progenitor cells. Proc Natl Acad Sci U S A 104: 1871-1876.
  275. Roy E, Leduc M, Guegan S, Rachdi L, Kluger N, et al. (2011) Specific maternal microchimeric T cells targeting fetal antigens in β cells predispose to auto-immune diabetes in the child. J Autoimmun 36: 253-262.
  276. Sarkar K, Miller FW (2004) Possible roles and determinants of microchimerism in autoimmune and other disorders. Autoimmunity Rev 3: 454-463.
  277. Tozzoli R, Barzilai O, Ram M, Villalta D, Bizzaro N, et al. (2008) Infections and autoimmune thyroid diseases: parallel detection of antibodies against pathogens with proteomic technology. Autoimmun Rev 8: 112-115.
  278. Wasserman EE, Nelson K, Rose NR, Rhode C, Pillion JP, et al. (2009) Infection and thyroid autoimmunity: A seroepidemiologic study of TPOaAb. Autoimmunity 42: 439-446.
  279. Fugazzola L, Cirello V, Beck-Peccoz P (2010) Fetal cell microchimerism in human cancers. Cancer Lett 287: 136-141.
  280. Khosrotehrani K, Bianchi DW (2003) Fetal cell microchimerism: helpful or harmful to the parous woman? Curr Opin Obstet Gynecol 15: 195-199.
  281. Khosrotehrani K, Johnson KL, Cha DH, Salomon RN, Bianchi DW (2004) Transfer of fetal cells with multilineage potential to maternal tissue. JAMA 292: 75-80.
  282. Khosrotehrani K, Bianchi DW (2005) Multi-lineage potential of fetal cells in maternal tissue: a legacy in reverse. J Cell Sci 118: 1559-1563.
  283. Molitor ML, Haynes LD, Jankowska-Gan E, Mulder A, Burlingham WJ (2004) HLA class I noninherited maternal antigens in cord blood and breast milk. Hum Immunol 65: 231-239.
  284. Kallenbach LR, Bianchi DW, Peter I, Stroh H, Johnson KL (2011) Maternal background strain influences fetal-maternal trafficking more than maternal immune competence in mice. J Reprod Immunol 90: 188-194.
  285. Jonsson AM, Uzunel M, Götherström C, Papadogiannakis N, Westgren M (2008) Maternal microchimerism in human fetal tissues. Am J Obstet Gynecol 198: 325.
  286. Götherstrom C, Johnsson AM, Mattsson J, Papadogiannakis N, Westgren M (2005) Identification of maternal hematopoietic cells in a 2nd-trimester fetus. Fetal Diagn Ther 20: 355-358.
  287. Srivatsa B, Srivatsa S, Johnson KL, Bianchi DW (2003) Maternal cell microchimerism in newborn tissues. J Pediatr 142: 31-35.
  288. Stevens AM, Hermes HM, Kiefer MM, Rutledge JC, Nelson JL (2009) Chimeric maternal cells with tissue-specific antigen expression and morphology are common in infant tissues. Pediatr Dev Pathol 12: 337-346.
  289. Fugazzola L, Cirello V, Beck-Peccoz P (2012) Microchimerism and endocrine disorders. J Clin Endocrinol Metab 97: 1452-1461.
  290. Larsson K, Elding-Larsson H, Cederwall E, Kockum K, Neiderud J, et al. (2004) Genetic and perinatal factors as risk for childhood type 1 diabetes. Diabetes Metab Res 20: 429-437.
  291. Dahlquist GG, Patterson C, Soltesz G (1999) Perinatal risk factors for childhood type 1 diabetes in Europe. The EURODIAB Substudy 2 Study group. Diabetes Care 22: 1698-1702.
  292. Sunami R, Komuro M, Yuminamochi T, Hoshi K, Hirata S (2010) Fetal cell microchimerism develops through the migration of fetus-derived cells to the maternal organs early after implantation. J Reprod Immunol 84: 117-123.
  293. Johnson KL, Samura O, Nelson JL, McDonnell M d WM, Bianchi DW (2002) Significant fetal cell microchimerism in a nontransfused woman with hepatitis C: Evidence of long-term survival and expansion. Hepatology 36: 1295-1297.
  294. Lambert NC, Pang JM, Yan Z, Erickson TD, Stevens AM, et al. (2005) Male microchimerism in women with systemic sclerosis and healthy women who have never given birth to a son. Ann Rheum Dis 64: 845-848.
  295. Srivatsa B, Srivatsa S, Johnson KL, Samura O, Lee SL, et al. (2001) Microchimerism of presumed fetal origin in thyroid specimens from women: a case-control study. Lancet 358: 2034-2038.
  296. Vanzyl B, Planas R, Ye Y, Foulis A, de Krijger RR, et al. (2010) Why are levels of maternal microchimerism higher in type 1 diabetes pancreas? Chimerism 1: 45-50.
  297. Rust DW, Bianchi DW (2009) Microchimerism in endocrine pathology. Endocr Pathol 20: 11-16.
  298. Persson CM, Lambert H, Vutova PP, Dellacasa-Lindberg I, Nederby J, et al. (2009) Transmission of Toxoplasma gondii from infected dendritic cells to natural killer cells. Infect Immun 77: 970-976.
  299. Channon JY, Seguin RM, Kasper LH (2000) Differential infectivity and division of Toxoplasma gondii in human peripheral blood leukocytes. Infect Immun 68: 4822-4826.
  300. Nakao M, Konishi E (1991) Proliferation of Toxoplasma gondii in human neutrophils in vitro. Parasitology 103 Pt 1: 23-27.
  301. Wilson CB, Remington JS (1979) Activity of human blood leukocytes against Toxoplasma gondii. J Infect Dis 140: 890-895.
  302. Delemarre FG, Stevenhagen A, Kroon FP, van Eer MY, Meenhorst PL, et al. (1994) Effect of IFN-gamma on the proliferation of Toxoplasma gondii in monocytes and monocyte-derived macrophages from AIDS patients. Immunology 83: 646-650.
  303. McLeod R, Bensch KG, Smith SM, Remington JS (1980) Effects of human peripheral blood monocytes, monocyte-derived macrophages, and spleen mononuclear phagocytes on Toxoplasma gondii. Cell Immunol 54: 330-350.
  304. Murray HW, Rubin BY, Carriero SM, Harris AM, Jaffec EA (1985) Human mononuclear phagocyte antiprotozoal mechanisms: oxygen-dependent vs oxygen-independent activity against intracellular Toxoplasma gondii. J Immunol 34: 1982-1988.
  305. Fadul CE, Channon JY, Kasper LH (1995) Survival of immunoglobulin G-opsonized Toxoplasma gondii in nonadherent human monocytes. Infect Immun 63: 4290-4294.
  306. Wilson CB, Westall J (1985) Activation of neonatal and adult human macrophages by alpha, beta, and gamma interferons. Infect Immun 49: 351-356.
  307. Anderson SE, Bautista S, Remington JS (1976) Induction of resistance to Toxoplasma gondii in human macrophages by soluble lymphocyte products. J Immunol 117: 381-387.
  308. Halonen SK, Lyman WD, Chiu FC (1996) Growth and development of Toxoplasma gondii in human neurons and astrocytes. J Neuropathol Exp Neurol 55: 1150-1156.
  309. Pfefferkorn ER, Guyre PM (1984) Inhibition of growth of Toxoplasma gondii in cultured fibroblasts by human recombinant gamma interferon. Infect Immun 44: 211-216.
  310. Woodman JP, Dimier IH, Bout DT (1991) Human endothelial cells are activated by IFN-gamma to inhibit Toxoplasma gondii replication. Inhibition is due to a different mechanism from that existing in mouse macrophages and human fibroblasts. J Immunol 147: 2019-2023.
  311. Nagineni CN, Pardhasaradhi K, Martins MC, Detrick B, Hooks JJ (1996) Mechanisms of interferon-induced inhibition of Toxoplasma gondii replication in human retinal pigment epithelial cells. Infect Immun 64: 4188-4196.
  312. Peterson PK, Gekker G, Hu S, Chao CC (1995) Human astrocytes inhibit intracellular multiplication of Toxoplasma gondii by a nitric oxide-mediated mechanism. J Infect Dis 171: 516-518.
  313. Chao CC, Gekker G, Hu S, Peterson PK (1994) Human microglial cell defense against Toxoplasma gondii. The role of cytokines. J Immunol 152: 1246-1252.
  314. Stray-Pedersen B (1993) Toxoplasmosis in pregnancy. Baillieres Clin Obstet Gynaecol 7: 107-137.
  315. Sunku CC, Gadi VK, de Laval de Lacoste B, Guthrie KA, Nelson JL (2010) Maternal and fetal microchimerism in granulocytes. Chimerism 1: 11-14.
  316. Hafid J, BelleteB, Flori P, Sawadogo P, Boyer Y, et al. (2005) Materno-foetal transmission of murine toxoplasmosis after oral infection. Am J Immunol 1: 1-5.
  317. Barragan A, Brossier F, Sibley LD (2005) Transepithelial migration of Toxoplasma gondii involves an interaction of intercellular adhesion molecule 1 (ICAM-1) with the parasite adhesin MIC2. Cell Microbiol 7: 561-568.
  318. Etienne-Manneville S, Manneville JB, Adamson P, Wilbourn B, Greenwood J, et al. (2000) ICAM-1-coupled cytoskeletal rearrangements and transendothelial lymphocyte migration involve intracellular calcium signaling in brain endothelial cell lines. J Immunol 165: 3375-3383.
  319. Marlin SD, Springer TA (1987) Purified intercellular adhesion molecule-1 (ICAM-1) is a ligand for lymphocyte function-associated antigen 1 (LFA-1). Cell 51: 813-819.
  320. Salzman AL, Menconi MJ, Unno N, Ezzell RM, Casey DM, et al. (1995) Nitric oxide dilates tight junctions and depletes ATP in cultured Caco-2BBe intestinal epithelial monolayers. Am J Physiol 268: G361-373.
  321. Unno N, Menconi MJ, Smith M, Fink MP (1995) Nitric oxide mediates interferon-gamma-induced hyperpermeability in cultured human intestinal epithelial monolayers. Crit Care Med 23: 1170-1176.
  322. Chen CP, Lee MY, Huang JP, Aplin JD, Wu YH, et al. (2008) Trafficking of multipotent mesenchymal stromal cells from maternal circulation through the placenta involves vascular endothelial growth factor receptor-1 and integrins. Stem Cells 26: 550-561.
  323. Silveira C, Vallochi AL, da Silva UR, Muccioli C, Holland GN, et al. (2010) Toxoplasma gondii in the peripheral blood of patients with acute and chronic toxoplasmosis. Br J Ophthalmol.
  324. Camossi LG, Greca-Júnior H, Corrêa AP, Richini-Pereira VB, Silva RC, et al. (2011) Detection of Toxoplasma gondii DNA in the milk of naturally infected ewes. Vet Parasitol 177: 256-261.
  325. Hiramoto RM, Mayrbaurl-Borges M, Galisteo AJ Jr, Meireles LR, Macre MS, et al. (2001) Infectivity of cysts of the ME-49 Toxoplasma gondii strain in bovine milk and homemade cheese. Rev Saude Publica 35: 113-118.
  326. McLeod R, Remington JS (1992) Toxoplasmosis. In: RE Behrman, RM Kliegman, WE Nelson, VC Vaughan, eds. Nelson Textbook of Pediatrics. 14th ed. Philadelphia, WB Saunders: 883-892.
  327. Remington JS, McLeod R, Thulliez P, Desmonts G (2001) Toxoplasmosis. In: Remington JS, Klein JO, eds. Infectious diseases of the fetus and newborn infant (5th edn) Toronto; WB Saunders: 205-346.
  328. Washburn LR, Dang H, Tian J, Kaufman DL (2007) The postnatal maternal environment influences diabetes development in nonobese diabetic mice. J Autoimmun 28: 19-23.
  329. Greeley SA, Katsumata M, Yu L, Eisenbarth GS, Moore DJ, et al. (2002) Elimination of maternally transmitted autoantibodies prevents diabetes in nonobese diabetic mice. Nat Med 8: 399-402.
  330. Koczwara K, Bonifacio E, Ziegler AG (2004) Transmission of maternal islet antibodies and risk of autoimmune diabetes in offspring of mothers with type 1 diabetes. Diabetes 53: 1-4.
  331. Stanley HM, Norris JM, Barriga K, Hoffman M, Yu L, et al. (2004) Is presence of islet autoantibodies at birth associated with development of persistent islet autoimmunity? The Diabetes Autoimmunity Study in the Young (DAISY). Diabetes Care 27: 497-502.
  332. Gillespie KM, Bain SC, Barnett AH, Bingley PJ, Christie MR, et al. (2004) The rising incidence of childhood type 1 diabetes and reduced contribution of high-risk HLA haplotypes. Lancet 364: 1699-1700.
  333. Han X, Sun Y, Scott S, Bleich D (2001) Tissue inhibitor of metalloproteinase-1 prevents cytokine-mediated dysfunction and cytotoxicity in pancreatic islets and beta-cells. Diabetes 50: 1047-1055.
  334. Herren B, Levkau B, Raines EW, Ross R (1998) Cleavage of beta-catenin and plakoglobin and shedding of VE-cadherin during endothelial apoptosis: evidence for a role for caspases and metalloproteinases. Mol Biol Cell 9: 1589-1601.
  335. Miralles F, Battelino T, Czernichow P, Scharfmann R (1998) TGF-beta plays a key role in morphogenesis of the pancreatic islets of Langerhans by controlling the activity of the matrix metalloproteinase MMP-2. J Cell Biol 143: 827-836.
  336. Florys B, Głowińska B, Urban M, Peczynska J (2006) [Metalloproteinases MMP-2 and MMP-9 and their inhibitors TIMP-1 and TIMP-2 levels in children and adolescents with type 1 diabetes]. Endokrynol Diabetol Chor Przemiany Materii Wieku Rozw 12: 184-189.
  337. Ridnour LA, Windhausen AN, Isenberg JS, Yeung N, Thomas DD, et al. (2007) Nitric oxide regulates matrix metalloproteinase-9 activity by guanylyl-cyclase-dependent and -independent pathways. Proc Natl Acad Sci U S A 104: 16898-16903.
  338. Zhou YP, Madjidi A, Wilson ME, Nothhelfer DA, Johnson JH, et al. (2005) Matrix metalloproteinases contribute to insulin insufficiency in Zucker diabetic fatty rats. Diabetes 54: 2612-2619.
  339. Savinov AY, Strongin AY (2009) Matrix metalloproteinases, T cell homing and beta-cell mass in type 1 diabetes. Vitam Horm 80: 541-562.
  340. Savinov AY, Burn P (2010) Interference with islet-specific homing of autoreactive T cells: an emerging therapeutic strategy for type 1 diabetes. Drug Discov Today 15: 531-539.
  341. Constantin G, Majeed M, Giagulli C, Piccio L, Kim JY, et al. (2000) Chemokines trigger immediate beta2 integrin affinity and mobility changes: differential regulation and roles in lymphocyte arrest under flow. Immunity 13: 759-769.
  342. Nandi A, Estess P, Siegelman M (2004) Bimolecular complex between rolling and firm adhesion receptors required for cell arrest; CD44 association with VLA-4 in T cell extravasation. Immunity 20: 455-465.
  343. Lesley J, Hyman R, Kincade PW (1993) CD44 and its interaction with extracellular matrix. Adv Immunol 54: 271-335.
  344. Katoh S, Zheng Z, Oritani K, Shimozato T, Kincade PW (1995) Glycosylation of CD44 negatively regulates its recognition of hyaluronan. J Exp Med 182: 419-429.
  345. Seipel D, Oliveira BC, Resende TL, Schuindt SH, Pimentel PM, et al. (2010) Toxoplasma gondii infection positively modulates the macrophages migratory molecular complex by increasing matrix metalloproteinases, CD44 and alpha v beta 3 integrin. Vet Parasitol 169: 312-319.
  346. Geurts N, Opdenakker G, Van den Steen PE (2012) Matrix metalloproteinases as therapeutic targets in protozoan parasitic infections. Pharmacol Ther 133: 257-279.
  347. Buache E, Garnotel R, Aubert D, Gillery P, Villena I (2007) Reduced secretion and expression of gelatinase profile in Toxoplasma gondii-infected human monocytic cells. Biochem Biophys Res Commun 359: 298-303.
  348. Muñoz M, Heimesaat MM, Danker K, Struck D, Lohmann U, et al. (2009) Interleukin (IL)-23 mediates Toxoplasma gondii-induced immunopathology in the gut via matrixmetalloproteinase-2 and IL-22 but independent of IL-17. J Exp Med 206: 3047-3059.
  349. Clark RT, Nance JP, Noor S, Wilson EH (2011) T-cell production of matrix metalloproteinases and inhibition of parasite clearance by TIMP-1 during chronic Toxoplasma infection in the brain. ASN Neuro 3: e00049.
  350. Chou PH, Lai SC (2011) Elevated concentrations of matrix metalloproteinase-12 and elastin degradation products in the sera of pregnant women infected with Toxoplasma gondii. Ann Trop Med Parasitol 105: 225-231.
  351. Laliberté J, Carruthers VB (2011) Toxoplasma gondii toxolysin 4 is an extensively processed putative metalloproteinase secreted from micronemes. Mol Biochem Parasitol 177: 49-56.
  352. Niehus S, Elass E, Coddeville B, Guérardel Y, Schwarz RT, et al. (2012) Glycosylphosphatidylinositols of Toxoplasma gondii induce matrix metalloproteinase-9 production and degradation of galectin-3. Immunobiology 217: 61-64.
  353. Debierre-Grockiego F, Azzouz N, Schmidt J, Dubremetz JF, Geyer H, et al. (2003) Roles of glycosylphosphatidylinositols of Toxoplasma gondii. Induction of tumor necrosis factor-alpha production in macrophages. J Biol Chem 278: 32987-32993.
  354. Debierre-Grockiego F, Campos MA, Azzouz N, Schmidt J, Bieker U, et al. (2007) Activation of TLR2 and TLR4 by glycosylphosphatidylinositols derived from Toxoplasma gondii. J Immunol 179: 1129-1137.
  355. Palsamy P, Subramanian S (2009) Modulatory effects of resveratrol on attenuating the key enzymes activities of carbohydrate metabolism in streptozotocin-nicotinamide-induced diabetic rats. Chem Biol Interact 179: 356-362.
  356. Parker G, Taylor R, Jones D, McClain D (2004) Hyperglycemia and inhibition of glycogen synthase in streptozotocin-treated mice: role of O-linked N-acetylglucosamine. J Biol Chem 279: 20636-20642.
  357. Bouché C, Serdy S, Kahn CR, Goldfine AB (2004) The cellular fate of glucose and its relevance in type 2 diabetes. Endocr Rev 25: 807-830.
  358. Chou JY, Matern D, Mansfield BC, Chen YT (2002) Type I glycogen storage diseases: disorders of the glucose-6-phosphatase complex. Curr Mol Med 2: 121-143.
  359. Wagle A, Jivraj S, Garlock GL, Stapleton SR (1998) Insulin regulation of glucose-6-phosphate dehydrogenase gene expression is rapamycin-sensitive and requires phosphatidylinositol 3-kinase. J Biol Chem 273: 14968-14974.
  360. Park J, Choe SS, Choi AH, Kim KH, Yoon MJ, et al. (2006) Increase in glucose-6-phosphate dehydrogenase in adipocytes stimulates oxidative stress and inflammatory signals. Diabetes 55: 2939-2949.
  361. Tian WN, Braunstein LD, Pang J, Stuhlmeier KM, Xi QC, et al. (1998) Importance of glucose-6-phosphate dehydrogenase activity for cell growth. J Biol Chem 273: 10609-10617.
  362. Tian WN, Braunstein LD, Apse K, Pang J, Rose M, et al. (1999) Importance of glucose-6-phosphate dehydrogenase activity in cell death. Am J Physiol 276: C1121-1131.
  363. Suhail M, Rizvi S (1989) Effect of type I (insulin-dependent) diabetes mellitus on key glycolytic enzymes of red blood cells. Acta Diabetol Lat 26: 315-320.
  364. Sitzmann FC (1984) [Erythrocyte metabolism in type I diabetes mellitus (key enzymes of glycolysis)]. Padiatr Padol 19: 303-309.
  365. Fleige T, Pfaff N, Gross U, Bohne W (2008) Localisation of gluconeogenesis and tricarboxylic acid (TCA)-cycle enzymes and first functional analysis of the TCA cycle in Toxoplasma gondii. Int J Parasitol 38: 1121-1132.
  366. Fleige T, Fischer K, Ferguson DJ, Gross U, Bohne W (2007) Carbohydrate metabolism in the Toxoplasma gondii apicoplast: localization of three glycolytic isoenzymes, the single pyruvate dehydrogenase complex, and a plastid phosphate translocator. Eukaryot Cell 6: 984-996.
  367. Pomel S, Luk FC, Beckers CJ (2008) Host cell egress and invasion induce marked relocations of glycolytic enzymes in Toxoplasma gondii tachyzoites. PLoS Pathog 4: e1000188.
  368. Nelson MM, Jones AR, Carmen JC, Sinai AP, Burchmore R, et al. (2008) Modulation of the host cell proteome by the intracellular apicomplexan parasite Toxoplasma gondii. Infect Immun 76: 828-844.
  369. Butterfield DA, Lange ML (2009) Multifunctional roles of enolase in Alzheimer's disease brain: beyond altered glucose metabolism. J Neurochem 111: 915-933.
  370. Tomavo S (2001) The differential expression of multiple isoenzyme forms during stage conversion of Toxoplasma gondii: an adaptive developmental strategy. Int J Parasitol 31: 1023-1031.
  371. Xia D, Anderson SJ, Jones AR, Prieto JH, Yates JR, et al. (2008) The proteome of Toxoplasma gondii: integration with the genome provides novel insights into gene expression and annotation. Genome Biol 9: R116.
  372. Denton H, Roberts CW, Alexander J, Thong KW, Coombs GH (1996) Enzymes of energy metabolism in the bradyzoites and tachyzoites of Toxoplasma gondii. FEMS Microbiol Lett 137: 103-108.
  373. Coppin A, Dzierszinski F, Legrand S, Mortuaire M, Ferguson D, et al. (2003) Developmentally regulated biosynthesis of carbohydrate and storage polysaccharide during differentiation and tissue cyst formation in Toxoplasma gondii. Biochimie 85: 353-361.
  374. Dzierszinski F, Mortuaire M, Dendouga N, Popescu O, Tomavo S (2001) Differential expression of two plant-like enolases with distinct enzymatic and antigenic properties during stage conversion of the protozoan parasite Toxoplasma gondii. J Mol Biol 309: 1017-1027.
  375. Ferguson DJ, Parmley SF, Tomavo S (2002) Evidence for nuclear localisation of two stage-specific isoenzymes of enolase in Toxoplasma gondii correlates with active parasite replication. Int J Parasitol 32: 1399-1410.
  376. Holmes M, Liwak U, Pricop I, Wang X, Tomavo S, et al. (2010) Silencing of tachyzoite enolase 2 alters nuclear targeting of bradyzoite enolase 1 in Toxoplasma gondii. Microbes Infect 12: 19-27.
  377. Joët T, Holterman L, Stedman TT, Kocken CH, Van Der Wel A, et al. (2002) Comparative characterization of hexose transporters of Plasmodium knowlesi, Plasmodium yoelii and Toxoplasma gondii highlights functional differences within the apicomplexan family. Biochem J 368: 923-929.
  378. Radke JR, Behnke MS, Mackey AJ, Radke JB, Roos DS, et al. (2005) The transcriptome of Toxoplasma gondii. BMC Biol 3: 26.
  379. Yang S, Parmley SF (1997) Toxoplasma gondii expresses two distinct lactate dehydrogenase homologous genes during its life cycle in intermediate hosts. Gene 184: 1-12.
  380. Al-Anouti F, Tomavo S, Parmley S, Ananvoranich S (2004) The expression of lactate dehydrogenase is important for the cell cycle of Toxoplasma gondii. J Biol Chem 279: 52300-52311.
  381. Pace DA, Fang J, Cintron R, Docampo MD, Moreno SN (2011) Overexpression of a cytosolic pyrophosphatase (TgPPase) reveals a regulatory role of PP(i) in glycolysis for Toxoplasma gondii. Biochem J 440: 229-240.
  382. Weilhammer DR, Iavarone AT, Villegas EN, Brooks GA, Sinai AP, et al. (2012) Host metabolism regulates growth and differentiation of Toxoplasma gondii. Int J Parasitol 42: 947-959.
  383. Blader IJ, Manger ID, Boothroyd JC (2001) Microarray analysis reveals previously unknown changes in Toxoplasma gondii-infected human cells. J Biol Chem 276: 24223-24231.
  384. Kimata I, Tanabe K (1982) Invasion by Toxoplasma gondii of ATP-depleted and ATP-restored chick embryo erythrocytes. J Gen Microbiol 128: 2499-2501.
  385. Molestina RE, El-Guendy N, Sinai AP (2008) Infection with Toxoplasma gondii results in dysregulation of the host cell cycle. Cell Microbiol 10: 1153-1165.
  386. Coppin A, Varré JS, Lienard L, Dauvillée D, Guérardel Y, et al. (2005) Evolution of plant-like crystalline storage polysaccharide in the protozoan parasite Toxoplasma gondii argues for a red alga ancestry. J Mol Evol 60: 257-267.
  387. Rahbar S (1968) An abnormal hemoglobin in red cells of diabetics. Clin Chim Acta 22: 296-298.
  388. Nanji AA, Pudek MR (1983) Glycosylated hemoglobins: a review. Can Fam Physician 29: 564-568.
  389. Bunn HF, Haney DN, Kamin S, Gabbay KH, Gallop PM (1976) The biosynthesis of human hemoglobin A1c. Slow glycosylation of hemoglobin in vivo. J Clin Invest 57: 1652-1659.
  390. Miller E, Hare JW, Cloherty JP, Dunn PJ, Gleason RE, et al. (1981) Elevated maternal hemoglobin A1c in early pregnancy and major congenital anomalies in infants of diabetic mothers. N Engl J Med 304: 1331-1334.
  391. Gould BJ, Davie SJ, Yudkin JS (1997) Investigation of the mechanism underlying the variability of glycated haemoglobin in non-diabetic subjects not related to glycaemia. Clin Chim Acta 260: 49-64.
  392. Merino-Torres JF, Fajardo-Montanana C, Ferrer-Garcia JC, Pinon-Selles F (2003) Hemoglobin glycosylation index is not related with blood glucose. J Diabetes Complications 17: 249-253.
  393. Lachin JM, Genuth S, Nathan DM, Rutledge BN (2007) The hemoglobin glycation index is not an independent predictor of the risk of microvascular complications in the Diabetes Control and Complications Trial. Diabetes 56: 1913-1921.
  394. Luk FC, Johnson TM, Beckers CJ (2008) N-linked glycosylation of proteins in the protozoan parasite Toxoplasma gondii. Mol Biochem Parasitol 157: 169-178.
  395. Varki A, Cummings R, Esko J, Freeze H, Hart GW, Marth J (eds). Essentials of Glycobiology. Cold Spring Harbour Laboratory Press. Cold Spring Harbour: 1999.
  396. Helenius A, Aebi M (2004) Roles of N-linked glycans in the endoplasmic reticulum. Annu Rev Biochem 73: 1019-1049.
  397. Varki A (1993) Biological roles of oligosaccharides: all of the theories are correct. Glycobiology 3: 97-130.
  398. Odenthal-Schnittler M, Tomavo S, Becker D, Dubremetz JF, Schwarz RT (1993) Evidence for N-linked glycosylation in Toxoplasma gondii. Biochem J 291 : 713-721.
  399. Garénaux E, Shams-Eldin H, Chirat F, Bieker U, Schmidt J, et al. (2008) The dual origin of Toxoplasma gondii N-glycans. Biochemistry 47: 12270-12276.
  400. Coppens I, Sinai AP, Joiner KA (2000) Toxoplasma gondii exploits host low-density lipoprotein receptor-mediated endocytosis for cholesterol acquisition. J Cell Biol 149: 167-180.
  401. Charron AJ, Sibley LD (2002) Host cells: mobilizable lipid resources for the intracellular parasite Toxoplasma gondii. J Cell Sci 115: 3049-3059.
  402. Coppens I, Joiner KA (2001) Parasitehost cell interactions in toxoplasmosis: new avenues for intervention? Expert Rev Mol Med 2001: 1-20.
  403. Schwab JC, Beckers CJ, Joiner KA (1994) The parasitophorous vacuole membrane surrounding intracellular Toxoplasma gondii functions as a molecular sieve. Proc Natl Acad Sci U S A 91: 509-513.
  404. Fauquenoy S, Hovasse A, Sloves PJ, Morelle W, Dilezitoko Alayi T, et al. (2011) Unusual N-glycan structures required for trafficking Toxoplasma gondii GAP50 to the inner membrane complex regulate host cell entry through parasite motility. Mol Cell Proteomics 10: M111.
  405. Tabbara KF, Sharara NA, Al-Momen AK (2001) Toxoplasmosis in a group of glucose-6-phosphate dehydrogenase deficient patients. Saudi Med J 22: 330-332.
  406. Vives Corrons JL, Feliu E, Pudjades MA, Cardellach F, Rozman C, et al. (1982) Severe G6PD deficiency associated with chronic hemolytic anemia, granulocyte dysfunction and increased susceptibility to infection: description of a new molecular variant (G6PD Barcelona). Blood 59: 482-434.
  407. Gupta N, Hartmann A, Lucius R, Voelker DR (2012) The obligate intracellular parasite Toxoplasma gondii secretes a soluble phosphatidylserine decarboxylase. J Biol Chem 287: 22938-22947.
  408. Nunes AA (2009) Patient with toxoplasmosis and glucose-6-phosphate dehydrogenase deficiency: a case report. Cases J 2: 8826.
  409. Carette C, Dubois-Laforgue D, Gautier JF, Timsit J (2011) Diabetes mellitus and glucose-6-phosphate dehydrogenase deficiency: from one crisis to another. Diabetes Metab 37: 79-82.
  410. Nwose EU, Jelinek HF, Richards RS, Kerr PG (2007) Erythrocyte oxidative stress in clinical management of diabetes and its cardiovascular complications. Br J Biomed Sci 64: 35-43.
  411. Sobngwi E, Gautier JF, Kevorkian JP, Villette JM, Riveline JP, et al. (2005) High prevalence of glucose-6-phosphate dehydrogenase deficiency without gene mutation suggests a novel genetic mechanism predisposing to ketosis-prone diabetes. J Clin Endocrinol Metab 90: 4446-4451.
  412. Danzig JA, Moser JT, Belfield P, Alter CA (2011) Glucose-6-phosphate dehydrogenase deficiency diagnosed in an adolescent with type 1 diabetes mellitus and hemoglobin A1c discordant with blood glucose measurements. J Pediatr 158: 849-851.
  413. Xu Y, Osborne BW, Stanton RC (2005) Diabetes causes inhibition of glucose-6-phosphate dehydrogenase via activation of PKA, which contributes to oxidative stress in rat kidney cortex. Am J Physiol Renal Physiol 289: F1040-1047.
  414. Zhang Z, Apse K, Pang J, Stanton RC (2000) High glucose inhibits glucose-6-phosphate dehydrogenase via cAMP in aortic endothelial cells. J Biol Chem 275: 40042-40047.
  415. Molestina RE, Sinai AP (2005) Host and parasite-derived IKK activities direct distinct temporal phases of NF-kappaB activation and target gene expression following Toxoplasma gondii infection. J Cell Sci 118: 5785-5796.
  416. Molestina RE, Sinai AP (2005) Detection of a novel parasite kinase activity at the Toxoplasma gondii parasitophorus vacuole membrane capable of phosphorylating host IkBa. Cell Microbiol 7: 351-362.
  417. Vercesi AE, Rodrigues CO, Uyemura SA, Zhong L, Moreno SN (1998) Respiration and oxidative phosphorylation in the apicomplexan parasite Toxoplasma gondii. J Biol Chem 273: 31040-31047.
  418. Ferreira S, De Carvalho TM, De Suza W (2003) Protein phosphorylation during the process of interaction of Toxoplasma gondii with host cells. J Submicrosc Cytol Pathol 35: 245-252.
  419. Muggeo M, Moghetti P, Querena M, Cacciatori V, Zoppini G, et al. (1993) Mononuclear leukocytes from obese patients with type II diabetes have reduced activity of hexokinase, 6-phosphofructokinase and glucose-6-phosphate dehydrogenase. Horm Metab Res 25: 160-164.
  420. Cappai G, Songini M, Doria A, Cavallerano JD, Lorenzi M (2011) Increased prevalence of proliferative retinopathy in patients with type 1 diabetes who are deficient in glucose-6-phosphate dehydrogenase. Diabetologia 54: 1539-1542.
  421. Usher-Smith JA, Thompson MJ, Sharp SJ, Walter FM (2011) Factors associated with the presence of diabetic ketoacidosis at diagnosis of diabetes in children and young adults: a systematic review. BMJ 343: d4092.
  422. Ando Y (2003) [Analyses of pathogenesis and therapeutic approaches for hereditary amyloidosis]. Rinsho Byori 51: 530-535.
  423. Monsellier E, Ramazzotti M, Taddei N, Chiti F (2010) A computational approach for identifying the chemical factors involved in the glycosaminoglycans-mediated acceleration of amyloid fibril formation. PLoS One 5: e11363.
  424. Ancsin JB (2003) Amyloidogenesis: historical and modern observations point to heparan sulfate proteoglycans as a major culprit. Amyloid 10: 67-79.
  425. Aleksandrescu AT (2005) Amyloid accomplices and enforcers. Protein Sci 14: 1-12.
  426. Park K, Verchere CB (2001) Identification of a heparin binding domain in the N-terminal cleavage site of pro-islet amyloid polypeptide. Implications for islet amyloid formation. J Biol Chem 276: 16611-16616.
  427. Suk JY, Zhang F, Balch WE, Linhardt RJ, Kelly JW (2006) Heparin accelerates gelsolin amyloidogenesis. Biochemistry 45: 2234-2242.
  428. Meng F, Abedini A, Song B, Raleigh DP (2007) Amyloid formation by pro-islet amyloid polypeptide processing intermediates: examination of the role of protein heparan sulfate interactions and implications for islet amyloid formation in type 2 diabetes. Biochemistry 46: 12091-12099.
  429. Bravo R, Arimon M, Valle-Delgado JJ, García R, Durany N, et al. (2008) Sulfated polysaccharides promote the assembly of amyloid beta(1-42) peptide into stable fibrils of reduced cytotoxicity. J Biol Chem 283: 32471-32483.
  430. Motamedi-Shad N, Monsellier E, Torrassa S, Relini A, Chiti F (2009) Kinetic analysis of amyloid formation in the presence of heparan sulfate: faster unfolding and change of pathway. J Biol Chem 284: 29921-29934.
  431. O'Callaghan P, Sandwall E, Li JP, Yu H, Ravid R, et al. (2008) Heparan sulfate accumulation with Abeta deposits in Alzheimer's disease and Tg2576 mice is contributed by glial cells. Brain Pathol 18: 548-561.
  432. Uversky VN (2008) Amyloidogenesis of natively unfolded proteins. Curr Alzheimer Res 5: 260-287.
  433. Soscia SJ, Kirby JE, Washicosky KJ, Tucker SM, Ingelsson M, et al. (2010) The Alzheimer's disease-associated amyloid beta-protein is an antimicrobial peptide. PLoS One 5: e9505.
  434. Prandota J (2012) Gastrointestinal tract abnormalities in autism, inflammatory bowel disease and many other clinical entities may be due to T. gondii infection. Scientific Reports 1.
  435. Ziegler AG, Wallner M, Kaiser I, Rossbauer M, Harsunen MH, et al. (2012) Long-term protective effect of lactation on the development of type 2 diabetes in women with recent gestational diabetes mellitus. Diabetes 61: 3167-3171.
  436. Meng F, Raleigh DP (2011) Inhibition of glycosaminoglycan-mediated amyloid formation by islet amyloid polypeptide and proIAPP processing intermediates. J Mol Biol 406: 491-502.
  437. Selkoe DJ (2004) Cell biology of protein misfolding: the examples of Alzheimer's and Parkinson's diseases. Nat Cell Biol 6: 1054-1061.
  438. Haataja L, Gurlo T, Huang CJ, Butler PC (2008) Islet amyloid in type 2 diabetes, and the toxic oligomer hypothesis. Endocrine Rev 29: 303-316.
  439. Sladek R, Rocheleau G, Rung J, Dina C, Shen L, et al. (2007) A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 445: 881-885.
  440. Jack Jr L, Boseman L, Vinicor F (2004) Aging Americans and diabetes. A public health and clinical response. Geriatrics 59: 14-17.
  441. Klöppel G, Löhr M, Habich K, Oberholzer M, Heitz PU (1985) Islet pathology and the pathogenesis of type 1 and type 2 diabetes mellitus revisited. Surv Synth Pathol Res 4: 110-125.
  442. Gepts W, Lecompte PM (1981) The pancreatic islets in diabetes. Am J Med 70: 105-115.
  443. EHRLICH JC, RATNER IM (1961) Amyloidosis of the islets of Langerhans. A restudy of islet hyalin in diabetic and non-diabetic individuals. Am J Pathol 38: 49-59.
  444. Clark A, Nilsson MR (2004) Islet amyloid: a complication of islet dysfunction or an aetiological factor in Type 2 diabetes? Diabetologia 47: 157-169.
  445. Jaikaran ET, Clark A (2001) Islet amyloid and type 2 diabetes: from molecular misfolding to islet pathophysiology. Biochim Biophys Acta 1537: 179-203.
  446. Röcken C, Linke RP, Saeger W (1992) Immunohistology of islet amyloid polypeptide in diabetes mellitus: semi-quantitative studies in a post-mortem series. Virchows Arch A Pathol Anat Histopathol 421: 339-344.
  447. Clark A, Wells CA, Buley ID, Cruickshank JK, Vanhegan RI, et al. (1988) Islet amyloid, increased A-cells, reduced B-cells and exocrine fibrosis: quantitative changes in the pancreas in type 2 diabetes. Diabetes Res 9: 151-159.
  448. Briones TL, Darwish H (2012) Vitamin D mitigates age-related cognitive decline through the modulation of pro-inflammatory state and decrease in amyloid burden. J Neuroinflammation 9: 244.
  449. Höppener JW, Verbeek JS, de Koning EJ, Oosterwijk C, van Hulst KL, et al. (1993) Chronic overproduction of islet amyloid polypeptide/amylin in transgenic mice: lysosomal localization of human islet amyloid polypeptide and lack of marked hyperglycaemia or hyperinsulinaemia. Diabetologia 36: 1258-1265.
  450. Yahiaoui B, Dzierszinski F, Bernigaud A, Slomianny C, Camus D, et al. (1999) Isolation and characterization of a subtractive library enriched for developmentally regulated transcripts expressed during encystation of Toxoplasma gondii. Mol Biochem Parasitol 99: 223-235.
  451. de Melo EJ, de Carvalho TU, de Souza W (1992) Penetration of Toxoplasma gondii into host cells induces changes in the distribution of the mitochondria and the endoplasmic reticulum. Cell Struct Funct 17: 311-317.
  452. Dubey JP, Lindsay DS, Speer CA (1998) Structures of Toxoplasma gondii tachyzoites, bradyzoites, and sporozoites and biology and development of tissue cysts. Clin Microbiol Rev 11: 267-299.
  453. Guérardel Y, Leleu D, Coppin A, Liénard L, Slomianny C, et al. (2005) Amylopectin biogenesis and characterization in the protozoan parasite Toxoplasma gondii, the intracellular development of which is restricted in the HepG2 cell line. Microbes Infect 7: 41-48.
  454. Augustine PC (1980) Effects of storage time and temperature on amylopectin levels and oocyst production of Eimeria meleagrimitis oocysts. Parasitology 81: 519-524.
  455. Nakai Y, Ogimoto K (1983) Relationship between amylopectin and viability of Eimeria tenella sporozoite. Nihon Juigaku Zasshi 45: 127-129.
  456. Ryley JF, Bentley M, Manners DJ, Stark JR (1969) Amylopectin, the storage polysaccharide of the Coccidia Eimeria brunetti and E. tenella. J Parasitol 55: 839-845.
  457. Sawesi O, Spillmann D, Lundén A, Wernersson S, Åbrink M (2010) Serglycin-independent release of active mast cell proteases in response to Toxoplasma gondii infection. J Biol Chem 285: 38005-38013.
  458. Carruthers V, Boothroyd JC (2007) Pulling together: an integrated model of Toxoplasma cell invasion. Curr Opinion Microbiol 10: 83-89.
  459. Jacquet A, Coulon L, De Neve J, Daminet V, Haumont M, et al. (2001) The surface antigen SAG3 mediates the attachment of Toxoplasma gondii to cell-surface proteoglycans. Mol Biochem Parasitol 116: 35-44.
  460. Monteiro VG, Soares CP, de Souza W (1998) Host cell surface sialic acid residues are involved on the process of penetration of Toxoplasma gondii into mammalian cells. FEMS Microbiol Lett 164: 323-327.
  461. Nilsson MR, Dobson CM (2003) In vitro characterization of lactoferrin aggregation and amyloid formation. Biochemistry 42: 375-382.
  462. Ando Y, Nakamura M, Kai H, Katsuragi S, Terazaki H, et al. (2002) A novel localized amyloidosis associated with lactoferrin in the cornea. Lab Invest 82: 757-766.
  463. Iwamaru Y, Shimizu Y, Imamura M, Murayama Y, Endo R, et al. (2008) Lactoferrin induces cell surface retention of prion protein and inhibits prion accumulation. J Neurochem 107: 636-646.
  464. Patelarou E, Girvalaki C, Brokalaki H, Patelarou A, Androulaki Z, et al. (2012) Current evidence on the associations of breastfeeding, infant formula, and cow's milk introduction with type 1 diabetes mellitus: a systematic review. Nutr Rev 70: 509-519.
  465. Zhao J, Frøkjaer JB, Drewes AM, Ejskjaer N (2006) Upper gastrointestinal sensory-motor dysfunction in diabetes mellitus. World J Gastroenterol 12: 2846-2857.
  466. Gentile S, Turco S, Oliviero B, Torella R (1998) The role of autonomic neuropathy as a risk factor of Helicobacter pylori infection in dyspeptic patients with type 2 diabetes mellitus. Diabetes Res Clin Pract 42: 41-48.
  467. Folwaczny C, Riepl R, Tschöp M, Landgraf R (1999) Gastrointestinal involvement in patients with diabetes mellitus: Part I (first of two parts). Epidemiology, pathophysiology, clinical findings. Z Gastroenterol 37: 803-815.
  468. Verne GN, Sninsky CA (1998) Diabetes and the gastrointestinal tract. Gastroenterol Clin North Am 27: 861-874, vi-vii.
  469. Horowitz M, Samsom M (2004) Gastrointestinal function in diabetes mellitus. Chichester: John Wiley & Sons Ltd: 1-337.
  470. Murray FE, Lombard MG, Ashe J, Lynch D, Drury MI, et al. (1987) Esophageal function in diabetes mellitus with special reference to acid studies and relationship to peripheral neuropathy. Am J Gastroenterol 82: 840-843.
  471. Clouse RE, Lustman PJ, Reidel WL (1986) Correlation of esophageal motility abnormalities with neuropsychiatric status in diabetics. Gastroenterology 90: 1146-1154.
  472. Stewart IM, Hosking DJ, Preston BJ, Atkinson M (1976) Oesophageal motor changes in diabetes mellitus. Thorax 31: 278-283.
  473. Hollis JB, Castell DO, Braddom RL (1977) Esophageal function in diabetes mellitus and its relation to peripheral neuropathy. Gastroenterology 73: 1098-1102.
  474. Keshavarzian A, Iber FL, Nasrallah S (1987) Radionuclide esophageal emptying and manometric studies in diabetes mellitus. Am J Gastroenterol 82: 625-631.
  475. Loo FD, Dodds WJ, Soergel KH, Arndorfer RC, Helm JF, et al. (1985) Multipeaked esophageal peristaltic pressure waves in patients with diabetic neuropathy. Gastroenterology 88: 485-491.
  476. Borgström PS, Olsson R, Sundkvist G, Ekberg O (1988) Pharyngeal and oesophageal function in patients with diabetes mellitus and swallowing complaints. Br J Radiol 61: 817-821.
  477. Dooley CP, el Newihi HM, Zeidler A, Valenzuela JE (1988) Abnormalities of the migrating motor complex in diabetics with autonomic neuropathy and diarrhea. Scand J Gastroenterol 23: 217-223.
  478. Ishiguchi T, Tada H, Nakagawa K, Yamamura T, Takahashi T (2002) Hyperglycemia impairs antro-pyloric coordination and delays gastric emptying in conscious rats. Auton Neurosci 95: 112-120.
  479. Mearin F, Camilleri M, Malagelada JR (1986) Pyloric dysfunction in diabetics with recurrent nausea and vomiting. Gastroenterology 90: 1919-1925.
  480. Fraser R, Fried M, Beglinger C (1993) [Assessment of gastric emptying]. Schweiz Med Wochenschr Suppl 54: 15-21.
  481. Samsom M, Jebbink RJ, Akkermans LM, van Berge-Henegouwen GP, Smout AJ (1996) Abnormalities of antroduodenal motility in type I diabetes. Diabetes Care 19: 21-27.
  482. Jebbink HJ, Bravenboer B, Akkermans LM, van Berge-Henegouwen GP, Smout AJ (1993) Relationships between dyspeptic symptoms and gastrointestinal motility in patients with type 1 (insulin-dependent) diabetes mellitus. Diabetologia 36: 948-954.
  483. Cellek S, Foxwell NA, Moncada S (2003) Two phases of nitrergic neuropathy in streptozotocin-induced diabetic rats. Diabetes 52: 2353-2362.
  484. Cellek S, Qu W, Schmidt AM, Moncada S (2004) Synergistic action of advanced glycation end products and endogenous nitric oxide leads to neuronal apoptosis in vitro: a new insight into selective nitrergic neuropathy in diabetes. Diabetologia 47: 331-339.
  485. King RH (2001) The role of glycation in the pathogenesis of diabetic polyneuropathy. Mol Pathol 54: 400-408.
  486. Bagyánszki M, Bódi N (2012) Diabetes-related alterations in the enteric nervous system and its microenvironment. World J Diabetes 3: 80-93.
  487. Ordög T (2008) Interstitial cells of Cajal in diabetic gastroenteropathy. Neurogastroenterol Motil 20: 8-18.
  488. Ordög T, Hayashi Y, Gibbons SJ (2009) Cellular pathogenesis of diabetic gastroenteropathy. Minerva Gastroenterol Dietol 55: 315-343.
  489. Choi KM, Gibbons SJ, Nguyen TV, Stoltz GJ, Lurken MS, et al. (2008) Heme oxygenase-1 protects interstitial cells of Cajal from oxidative stress and reverses diabetic gastroparesis. Gastroenterology 135: 2055-2064, 2064.
  490. Wang XY, Huizinga JD, Diamond J, Liu LW (2009) Loss of intramuscular and submuscular interstitial cells of Cajal and associated enteric nerves is related to decreased gastric emptying in streptozotocin-induced diabetes. Neurogastroenterol Motil 21: 1095-1095e92.
  491. Hasler WL (2011) Gastroparesis: pathogenesis, diagnosis and management. Nat Rev Gastroenterol Hepatol 8: 438-453.
  492. Tang DM, Friedenberg FK (2011) Gastroparesis: approach, diagnostic evaluation, and management. Dis Mon 57: 74-101.
  493. Kashyap P, Farrugia G (2011) Oxidative stress: key player in gastrointestinal complications of diabetes. Neurogastroenterol Motil 23: 111-114.
  494. de Mello ST, de Miranda Neto MH, Zanoni JN, Furlan MM (2009) Effects of insulin treatment on HuC/HuD, NADH diaphorase, and nNOS-positive myoenteric neurons of the duodenum of adult rats with acute diabetes. Dig Dis Sci 54: 731-737.
  495. Izbéki F, Wittman T, Rosztóczy A, Linke N, Bódi N, et al. (2008) Immediate insulin treatment prevents gut motility alterations and loss of nitrergic neurons in the ileum and colon of rats with streptozotocin-induced diabetes. Diabetes Res Clin Pract 80: 192-198.
  496. Zanoni JN, Tronchini EA, Moure SA, Souza ID (2011) Effects of L-glutamine supplementation on the myenteric neurons from the duodenum and cecum of diabetic rats. Arq Gastroenterol 48: 66-71.
  497. Zandecki M, Vanden Berghe P, Depoortere I, Geboes K, Peeters T, et al. (2008) Characterization of myenteric neuropathy in the jejunum of spontaneously diabetic BB-rats. Neurogastroenterol Motil 20: 818-828.
  498. Pereira RV, Tronchini EA, Tashima CM, Alves EP, Lima MM, et al. (2011) L-glutamine supplementation prevents myenteric neuron loss and has gliatrophic effects in the ileum of diabetic rats. Dig Dis Sci 56: 3507-3516.
  499. Nezami BG, Srinivasan S (2010) Enteric nervous system in the small intestine: pathophysiology and clinical implications. Curr Gastroenterol Rep 12: 358-365.
  500. Chandrasekharan B, Anitha M, Blatt R, Shahnavaz N, Kooby D, et al. (2011) Colonic motor dysfunction in human diabetes is associated with enteric neuronal loss and increased oxidative stress. Neurogastroenterol Motil 23: 131-138, e26.
  501. Liu W, Yue W, Wu R (2010) Effects of diabetes on expression of glial fibrillary acidic protein and neurotrophins in rat colon. Auton Neurosci 154: 79-83.
  502. Furlan MMDP, Molinari SL, de Miranda Neto MH (2002) Morphoquantitative effects of acute diabetes on the myenteric neurons of the proximal colon of adult rats. Arq Neuropsiquiatr 60: 576-581.
  503. Fregonesi CEPT, de Miranda Neto MH, Molinari SL, Zanoni JN (2001) Quantitative study of the myenteric plexus of the stomach of rats with streptozotocin-induced diabetes. Arq Neuropsiquiatr 59: 50-53.
  504. Alves AM, Alves EP, Fregonesi CE, Defani MA, Stabille SR, et al. (2006) Morphoquantitative aspects of NADH-diaphorase myenteric neurons in the ileum of diabetic rats treated with acetyl-L-carnitine. Anat Histol Embryol 35: 13-18.
  505. Haroon F, Händel U, Angenstein F, Goldschmidt J, Kreutzmann P, et al. (2012) Toxoplasma gondii actively inhibits neuronal function in chronically infected mice. PLoS One 7: e35516.
  506. Sant'Ana DM, Góis MB, Zanoni JN, da Silva AV, da Silva CJ, et al. (2012) Intraepithelial lymphocytes, goblet cells and VIP-IR submucosal neurons of jejunum rats infected with Toxoplasma gondii. Int J Exp Pathol 93: 279-286.
  507. Alatas FS, Masumoto K, Esumi G, Nagata K, Taguchi T (2011) Potential significance of abnormalities in the interstitial cells of Cajal, smooth muscle, and the enteric nervous system, proximal and distal to the obstructed site of duodenal atresia. J Pediatr Gastroenterol Nutr.
  508. de Boissieu D, Bargaoui K, Sakiroglu O, Francoual C, Dupont C, et al. (1989) [Esophagogastroduodenitis in the newborn. Apropos of 32 cases]. Arch Fr Pediatr 46: 711-715.
  509. Boukthir S, Fetni I, M'Rad S, Bennour F, Barsaoui S (2002) [Esophagogastroduodenitis in the newborn. Report of 90 cases]. Tunis Med 80: 18-20.
  510. de Boissieu D, Dupont C, Barbet JP, Bargaoui K, Badoual J (1994) Distinct features of upper gastrointestinal endoscopy in the newborn. J Pediatr Gastroenterol Nutr 18: 334-338.
  511. Alvarado-Esquivel C, Estrada-Martínez S (2011) Toxoplasma gondii infection and abdominal hernia: evidence of a new association. Parasit Vectors 4: 112.
  512. Nagineni CN, Detrick B, Hooks JJ (2002) Transforming growth factor-beta expression in human retinal pigment epithelial cells is enhanced by Toxoplasma gondii: a possible role in the immunopathogenesis of retinochoroiditis. Clin Exp Immunol 128: 372-378.
  513. Letterio JJ, Roberts AB (1998) Regulation of immune responses by TGF-beta. Annu Rev Immunol 16: 137-161.
  514. Wahl SM (1992) Transforming growth factor beta (TGF-beta) in inflammation: a cause and a cure. J Clin Immunol 12: 61-74.
  515. Horwitz DA, Gray JD, Ohtsuka K, Hirokawa M, Takahashi T (1997) The immunoregulatory effects of NK cells: the role of TGF-beta and implications for autoimmunity. Immunol Today 18: 538-542.
  516. Smart NG, Apelqvist AA, Gu X, Harmon EB, Topper JN, et al. (2006) Conditional expression of Smad7 in pancreatic beta cells disrupts TGF-beta signaling and induces reversible diabetes mellitus. PLoS Biol 4: e39.
  517. Pfeiffer A, Middelberg-Bisping K, Drewes C, Schatz H (1996) Elevated plasma levels of transforming growth factor-beta 1 in NIDDM. Diabetes Care 19: 1113-1117.
  518. Yener S, Demir T, Akinci B, Bayraktar F, Kebapcilar L, et al. (2007) Transforming growth factor-beta 1 levels in women with prior history of gestational diabetes mellitus. Diabetes Res Clin Pract 76: 193-198.
  519. Flores L, Näf S, Hernáez R, Conget I, Gomis R, et al. (2004) Transforming growth factor beta at clinical onset of Type 1 diabetes mellitus. A pilot study. Diabet Med 21: 818-822.
  520. Chen S, Hong SW, Iglesias-de la Cruz MC, Isono M, Casaretto A, et al. (2001) The key role of the transforming growth factor-beta system in the pathogenesis of diabetic nephropathy. Ren Fail 23: 471-481.
  521. Chen S, Jim B, Ziyadeh FN (2003) Diabetic nephropathy and transforming growth factor-beta: transforming our view of glomerulosclerosis and fibrosis build-up. Semin Nephrol 23: 532-543.
  522. Park K, Ryu SB, Park YI, Ahn K, Lee SN, et al. (2001) Diabetes mellitus induces vaginal tissue fibrosis by TGF-beta 1 expression in the rat model. J Sex Marital Ther 27: 577-587.
  523. Tonkin DR, Haskins K (2009) Regulatory T cells enter the pancreas during suppression of type 1 diabetes and inhibit effector T cells and macrophages in a TGF-beta-dependent manner. Eur J Immunol 39: 1313-1322.
  524. Wu AJ, Hua H, Munson SH, McDevitt HO (2002) Tumor necrosis factor-alpha regulation of CD4+CD25+ T cell levels in NOD mice. Proc Natl Acad Sci U S A 99: 12287-12292.
  525. Tang Q, Henriksen KJ, Bi M, Finger EB, Szot G, et al. (2004) In vitro-expanded antigen-specific regulatory T cells suppress autoimmune diabetes. J Exp Med 199: 1455-1465.
  526. You S, Belghith M, Cobbold S, Alyanakian MA, Gouarin C, et al. (2005) Autoimmune diabetes onset results from qualitative rather than quantitative age-dependent changes in pathogenic T-cells. Diabetes 54: 1415-1422.
  527. Tarbell KV, Petit L, Zuo X, Toy P, Luo X, et al. (2007) Dendritic cell-expanded, islet-specific CD4+ CD25+ CD62L+ regulatory T cells restore normoglycemia in diabetic NOD mice. J Exp Med 204: 191-201.
  528. Weber SE, Harbertson J, Godebu E, Mros GA, Padrick RC, et al. (2006) Adaptive islet-specific regulatory CD4 T cells control autoimmune diabetes and mediate the disappearance of pathogenic Th1 cells in vivo. J Immunol 176: 4730-4739.
  529. Tonkin DR, He J, Barbour G, Haskins K (2008) Regulatory T cells prevent transfer of type 1 diabetes in NOD mice only when their antigen is present in vivo. J Immunol 181: 4516-4522.
  530. Suzumura A, Sawada M, Yamamoto H, Marunouchi T (1993) Transforming growth factor-beta suppresses activation and proliferation of microglia in vitro. J Immunol 151: 2150-2158.
  531. Shrikant P, Lee SJ, Kalvakolanu I, Ransohoff RM, Benveniste EN (1996) Stimulus-specific inhibition of intracellular adhesion molecule-1 gene expression by TGF-beta. J Immunol 157: 892-900.
  532. Winkler MK, Benveniste EN (1998) Transforming growth factor-beta inhibition of cytokine-induced vascular cell adhesion molecule-1 expression in human astrocytes. Glia 22: 171-179.
  533. Filisetti D, Candolfi E (2004) Immune response to Toxoplasma gondii. Ann Ist Super Sanita 40: 71-80.
  534. Schlüter D, Bertsch D, Frei K, Hübers SB, Wiestler OD, et al. (1998) Interferon-gamma antagonizes transforming growth factor-beta2-mediated immunosuppression in murine Toxoplasma encephalitis. J Neuroimmunol 81: 38-48.
  535. Buzoni-Gatel D, Debbabi H, Mennechet FJ, Martin V, Lepage AC, et al. (2001) Murine ileitis after intracellular parasite infection is controlled by TGF-beta-producing intraepithelial lymphocytes. Gastroenterology 120: 914-924.
  536. Connor TB Jr, Roberts AB, Sporn MB, Danielpour D, Dart LL, et al. (1989) Correlation of fibrosis and transforming growth factor-beta type 2 levels in the eye. J Clin Invest 83: 1661-1666.
  537. Sarciron ME, Gherardi A (2000) Cytokines involved in Toxoplasmic encephalitis. Scand J Immunol 52: 534-543.
  538. Barbosa BF, Silva DA, Costa IN, Mineo JR, Ferro EA (2008) BeWo trophoblast cell susceptibility to Toxoplasma gondii is increased by interferon-gamma, interleukin-10 and transforming growth factor-beta1. Clin Exp Immunol 151: 536-545.
  539. Saito S (2000) Cytokine network at the feto-maternal interface. J Reprod Immunol 47: 87-103.
  540. Luppi P (2003) How immune mechanisms are affected by pregnancy. Vaccine 21: 3352-3357.
  541. Raghupathy R (2001) Pregnancy: success and failure within the Th1/Th2/Th3 paradigm. Semin Immunol 13: 219-227.
  542. Schäfer-Somi S (2003) Cytokines during early pregnancy of mammals: a review. Anim Reprod Sci 75: 73-94.
  543. Denkers EY, Gazzinelli RT (1998) Regulation and function of T-cell-mediated immunity during Toxoplasma gondii infection. Clin Microbiol Rev 11: 569-588.
  544. Hughes HP (1988) Oxidative killing of intracellular parasites mediated by macrophages. Parasitol Today 4: 340-347.
  545. Ding AH, Nathan CF, Stuehr DJ (1988) Release of reactive nitrogen intermediates and reactive oxygen intermediates from mouse peritoneal macrophages. Comparison of activating cytokines and evidence for independent production. J Immunol 141: 2407-2412.
  546. Pfefferkorn ER, Rebhun S, Eckel M (1986) Characterization of an indoleamine 2,3-dioxygenase induced by gamma-interferon in cultured human fibroblasts. J Interferon Res 6: 267-279.
  547. Oliveira JG, Silva NM, Santos AA, Souza MA, Ferreira GL, et al. (2006) BeWo trophoblasts are unable to control replication of Toxoplasma gondii, even in the presence of exogenous IFN-gamma. Placenta 27: 691-698.
  548. Malipiero U, Koedel U, Pfister HW, Levéen P, Bürki K, et al. (2006) TGFbeta receptor II gene deletion in leucocytes prevents cerebral vasculitis in bacterial meningitis. Brain 129: 2404-2415.
  549. Tsunawaki S, Sporn M, Ding A, Nathan C (1988) Deactivation of macrophages by transforming growth factor-beta. Nature 334: 260-262.
  550. Turner M, Chantry D, Katsikis P, Berger A, Brennan FM, et al. (1991) Induction of the interleukin 1 receptor antagonist protein by transforming growth factor-beta. Eur J Immunol 21: 1635-1639.
  551. Kim DS, Park SH, Park KC (2004) Transforming growth factor-beta1 decreases melanin synthesis via delayed extracellular signal-regulated kinase activation. Int J Biochem Cell Biol 36: 1482-1491.
  552. Bogdan C, Nathan C (1993) Modulation of macrophage function by transforming growth factor beta, interleukin-4, and interleukin-10. Ann N Y Acad Sci 685: 713-739.
  553. Seabra SH, de Souza W, Damatta RA (2004) Toxoplasma gondii exposes phosphatidylserine inducing a TGF-beta1 autocrine effect orchestrating macrophage evasion. Biochem Biophys Res Commun 324: 744-752.
  554. Malipiero U, Koedel U, Pfister W, Fontana A (2007) Bacterial meningitis: the role of transforming growth factor-Beta in innate immunity and secondary brain damage. Neurodegener Dis 4: 43-50.
  555. McCarty MF, Barroso-Aranda J, Contreras F (2009) Regular thermal therapy may promote insulin sensitivity while boosting expression of endothelial nitric oxide synthase--effects comparable to those of exercise training. Med Hypotheses 73: 103-105.
  556. Hooper PL (1999) Hot-tub therapy for type 2 diabetes mellitus. N Engl J Med 341: 924-925.
  557. Ilangovan G, Osinbowale S, Bratasz A, Bonar M, Cardounel AJ, et al. (2004) Heat shock regulates the respiration of cardiac H9c2 cells through upregulation of nitric oxide synthase. Am J Physiol Cell Physiol 287: C1472-1481.
  558. Harris MB, Blackstone MA, Ju H, Venema VJ, Venema RC (2003) Heat-induced increases in endothelial NO synthase expression and activity and endothelial NO release. Am J Physiol Heart Circ Physiol 285: H333-340.
  559. Ikeda Y, Biro S, Kamogawa Y, Yoshifuku S, Eto H, et al. (2001) Repeated thermal therapy upregulates arterial endothelial nitric oxide synthase expression in Syrian golden hamsters. Jpn Circ J 65: 434-438.
  560. Ikeda Y, Biro S, Kamogawa Y, Yoshifuku S, Eto H, et al. (2005) Repeated sauna therapy increases arterial endothelial nitric oxide synthase expression and nitric oxide production in cardiomyopathic hamsters. Circ J 69: 722-729.
  561. Imamura M, Biro S, Kihara T, Yoshifuku S, Takasaki K, et al. (2001) Repeated thermal therapy improves impaired vascular endothelial function in patients with coronary risk factors. J Am Coll Cardiol 38: 1083-1088.
  562. Chung J, Nguyen AK, Henstridge DC, Holmes AG, Chan MH, et al. (2008) HSP72 protects against obesity-induced insulin resistance. Proc Natl Acad Sci U S A 105: 1739-1744.
  563. Su CY, Chong KY, Chen J, Ryter S, Khardori R, et al. (1999) A physiologically relevant hyperthermia selectively activates constitutive hsp70 in H9c2 cardiac myoblasts and confers oxidative protection. J Mol Cell Cardiol 31: 845-855.
  564. Chong KY, Lai CC, Lille S, Chang C, Su CY (1998) Stable overexpression of the constitutive form of heat shock protein 70 confers oxidative protection. J Mol Cell Cardiol 30: 599-608.
  565. Yazar S, Gur M, Ozdogru I, Yaman O, Oguzhan A, et al. (2006) Anti-Toxoplasma gondii antibodies in patients with chronic heart failure. J Med Microbiol 55: 89-92.
  566. Mineo JR, Silva NM, Gazzinelli RT, Silva DA, Ferro EA, et al. (1998) Toxoplasma gondii conversion of bradyzoite to tachyzoite stage-specific antigens in vivo. Keystone Symposium on Molecular Cell Biology, 115 (abstr. no. 116).
  567. Weiss LM, Ma YF, Takvorian PM, Tanowitz HB, Wittner M (1998) Bradyzoite development in Toxoplasma gondii and the hsp70 stress response. Infect Immun 66: 3295-3302.
  568. Echeverria PC, Figueras MJ, Vogler M, Kriehuber T, de Miguel N, et al. (2010) The Hsp90 co-chaperone p23 of Toxoplasma gondii: Identification, functional analysis and dynamic interactome determination. Mol Biochem Parasitol 172: 129-140.
  569. Himeno K, Hisaeda H (1996) Contribution of 65-kDa heat shock protein induced by gamma and delta T cells to protection against Toxoplasma gondii infection. Immunol Res 15: 258-264.
  570. Multhoff G (2009) Activation of natural killer cells by heat shock protein 70. 2002. Int J Hyperthermia 25: 169-175.
  571. Ciavarra RP, Simeone A (1990) T lymphocyte stress response. I. Induction of heat shock protein synthesis at febrile temperatures is correlated with enhanced resistance to hyperthermic stress but not to heavy metal toxicity or dexamethasone-induced immunosuppression. Cell Immunol 129: 363-376.
  572. Angelidis CE, Lazaridis I, Pagoulatos GN (1999) Aggregation of hsp70 and hsc70 in vivo is distinct and temperature-dependent and their chaperone function is directly related to non-aggregated forms. Eur J Biochem 259: 505-512.
  573. Lentze N, Aquilina JA, Lindbauer M, Robinson CV, Narberhaus F (2004) Temperature and concentration-controlled dynamics of rhizobial small heat shock proteins. Eur J Biochem 271: 2494-2503.
  574. Son ES, Nam HW (2001) Detection and characterization of excretory/secretory proteins from Toxoplasma gondii by monoclonal antibodies. Korean J Parasitol 39: 49-56.
  575. Lyons RE, McLeod R, Roberts CW (2002) Toxoplasma gondii tachyzoite-bradyzoite interconversion. Trends Parasitol 18: 198-201.
  576. Kinze S, Clauss M, Reuter U, Wolf T, Dreier JP, et al. (2001) Valproic acid is effective in migraine prophylaxis at low serum levels: a prospective open-label study. Headache 41: 774-778.
  577. Lenaerts M, Bastings E, Sianard J, Schoenen J (1996) Sodium valproate in severe migraine and tension-type headache: an open study of long-term efficacy and correlation with blood levels. Acta Neurol Belg 96: 126-129.
  578. Sweeten TL, Posey DJ, Shankar S, McDougle CJ (2004) High nitric oxide production in autistic disorder: a possible role for interferon-gamma. Biol Psychiatry 55: 434-437.
  579. Kirkman LA, Weiss LM, Kim K (2001) Cyclic nucleotide signaling in Toxoplasma gondii bradyzoite differentiation. Infect Immun 69: 148-153.
  580. Soête M, Camus D, Dubremetz JF (1994) Experimental induction of bradyzoite-specific antigen expression and cyst formation by the RH strain of Toxoplasma gondii in vitro. Exp Parasitol 78: 361-370.
  581. Bohne W, Heesemann J, Gross U (1994) Reduced replication of Toxoplasma gondii is necessary for induction of bradyzoite-specific antigens: a possible role for nitric oxide in triggering stage conversion. Infect Immun 62: 1761-1767.
  582. Radke JR, Guerini MN, White MW (2000) Toxoplasma gondii: characterization of temperature-sensitive tachyzoite cell cycle mutants. Exp Parasitol 96: 168-177.
  583. Havlícek J, Gasová ZG, Smith AP, Zvára K, Flegr J (2001) Decrease of psychomotor performance in subjects with latent 'asymptomatic' toxoplasmosis. Parasitology 122: 515-520.
  584. Tait ED, Jordan KA, Dupont CD, Harris TH, Gregg B, et al. (2010) Virulence of Toxoplasma gondii is associated with distinct dendritic cell responses and reduced numbers of activated CD8+ T cells. J Immunol 185: 1502-1512.
  585. Sørensen IM, Joner G, Jenum PA, Eskild A, Torjesen PA, et al. (2012) Maternal serum levels of 25-hydroxy-vitamin D during pregnancy and risk of type 1 diabetes in the offspring. Diabetes 61: 175-178.
  586. Takiishi T, Gysemans C, Bouillon R, Mathieu C (2010) Vitamin D and diabetes. Endocrinol Metab Clin North Am 39: 419-446, table of contents.
  587. Giulietti A, Gysemans C, Stoffels K, van Etten E, Decallonne B, et al. (2004) Vitamin D deficiency in early life accelerates Type 1 diabetes in non-obese diabetic mice. Diabetologia 47: 451-462.
  588. Holick MF (2008) Diabetes and the vitamin d connection. Curr Diab Rep 8: 393-398.
  589. Lalor MK, Floyd S, Gorak-Stolinska P, Weir RE, Blitz R, et al. (2011) BCG vaccination: a role for vitamin D? PLoS One 6: e16709.
  590. Smith SG, Lalor MK, Gorak-Stolinska P, Blitz R, Beveridge NE, et al. (2010) Mycobacterium tuberculosis PPD-induced immune biomarkers measurable in vitro following BCG vaccination of UK adolescents by multiplex bead array and intracellular cytokine staining. BMC Immunol 11: 35.
  591. Zhu AH, Jin L, Liu JJ, Liu MY, Lv AJ, et al. (2011) [Intranasal vaccination with mycobacterial 65-kD heat-shock protein can prevent insulitis and diabetes in non-obese diabetic mice]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 27: 1165-1168.
  592. Santos RR Jr, Sartori A, Lima DS, Souza PR, Coelho-Castelo AA, et al. (2009) DNA vaccine containing the mycobacterial hsp65 gene prevented insulitis in MLD-STZ diabetes. J Immune Based Ther Vaccines 7: 4.
  593. Zügel U, Kaufmann SH (1999) Role of heat shock proteins in protection from and pathogenesis of infectious diseases. Clin Microbiol Rev 12: 19-39.
  594. Hisaeda H, Himeno K (1997) The role of host-derived heat-shock protein in immunity against Toxoplasma gondii infection. Parasitol Today 13: 465-468.
  595. Palomer X, González-Clemente JM, Blanco-Vaca F, Mauricio D (2008) Role of vitamin D in the pathogenesis of type 2 diabetes mellitus. Diabetes Obes Metab 10: 185-197.
  596. Mathieu C, Waer M, Casteels K, Laureys J, Bouillon R (1995) Prevention of type I diabetes in NOD mice by nonhypercalcemic doses of a new structural analog of 1,25-dihydroxyvitamin D3, KH1060. Endocrinology 136: 866-872.
  597. Luong K, Nguyen LTH, Nguyen DNP (2005) The role of vitamin D in protecting type 1 diabetes mellitus. Diabetes Metab Res Rev 21: 338-346.
  598. Snijder M, van Dam R, Visser M, Deeg D, Seidell J, et al. (2006) To: Mathieu C, Gysemans C, Giulietti A, Bouillon R (2005) Vitamin D and diabetes. Diabetologia 48:1247-1257. Diabetologia 49: 217-218.
  599. Boucher BJ, Mannan N, Noonan K, Hales CN, Evans SJ (1995) Glucose intolerance and impairment of insulin secretion in relation to vitamin D deficiency in east London Asians. Diabetologia 38: 1239-1245.
  600. Zittermann A (2003) Vitamin D in preventive medicine: are we ignoring the evidence? Br J Nutr 89: 552-572.
  601. Hewison M, Gacad MA, Lemire J, Adams JS (2001) Vitamin D as a cytokine and hematopoetic factor. Rev Endocr Metab Disord 2: 217-227.
  602. Mauricio D, Mandrup-Poulsen T, Nerup J (1996) Vitamin D analogues in insulin-dependent diabetes mellitus and other autoimmune diseases: a therapeutic perspective. Diabetes Metab Rev 12: 57-68.
  603. Kaur H, Donaghue KC, Chan AK, Benitez-Aguirre P, Hing S, et al. (2011) Vitamin D deficiency is associated with retinopathy in children and adolescents with type 1 diabetes. Diabetes Care 34: 1400-1402.
  604. Remington JS, Desmonts G (1990) Toxoplasmosis. In Remington JS, Klein JO (eds.) Infectious Diseases of the Fetus and Newborn Infant. The WB Saunders Co, Philadelphia, 89-195.
  605. Tedesco RC, Smith RL, Corte-Real S, Calabrese KS (2004) Ocular toxoplasmosis: the role of retinal pigment epithelium migration in infection. Parasitol Res 92: 467-472.
  606. Delair E, Latkany P, Noble AG, Rabiah P, McLeod R, et al. (2011) Clinical manifestations of ocular toxoplasmosis. Ocul Immunol Inflamm 19: 91-102.
  607. Sasaki K, Kuriyama H, Yeh LI, Fukuda M (1983) Studies on diabetic cataract in rats induced by streptozotocin. I. Photodocumentation of lens opacification. Ophthalmic Res 15: 185-190.
  608. Wilson ME Jr, Levin AV, Trivedi RH, Kruger SJ, Elliott LA, et al. (2007) Cataract associated with type-1 diabetes mellitus in the pediatric population. J AAPOS 11: 162-165.
  609. Grałek M, Fogel W, Chmielecki C (1991) [Does histamine participate in diabetic ocular complications?]. Klin Oczna 93: 337-339.
  610. Chagas CE, Borges MC, Martini LA, Rogero MM (2012) Focus on vitamin D, inflammation and type 2 diabetes. Nutrients 4: 52-67.
  611. Kolb H, Mandrup-Poulsen T (2005) An immune origin of type 2 diabetes? Diabetologia 48: 1038-1050.
  612. Lemire JM (1995) Immunomodulatory actions of 1,25-dihydroxyvitamin D3. J Steroid Biochem Mol Biol 53: 599-602.
  613. Jablonski KL, Chonchol M, Pierce GL, Walker AE, Seals DR (2011) 25-Hydroxyvitamin D deficiency is associated with inflammation-linked vascular endothelial dysfunction in middle-aged and older adults. Hypertension 57: 63-69.
  614. Peterson CA, Heffernan ME (2008) Serum tumor necrosis factor-alpha concentrations are negatively correlated with serum 25(OH)D concentrations in healthy women. J Inflamm (Lond) 5: 10.
  615. Ngo DT, Sverdlov AL, McNeil JJ, Horowitz JD (2010) Does vitamin D modulate asymmetric dimethylarginine and C-reactive protein concentrations? Am J Med 123: 335-341.
  616. Dobnig H, Pilz S, Scharnagl H, Renner W, Seelhorst U, et al. (2008) Independent association of low serum 25-hydroxyvitamin d and 1,25-dihydroxyvitamin d levels with all-cause and cardiovascular mortality. Arch Intern Med 168: 1340-1349.
  617. Bellia A, Garcovich C, D’Adamo M, Lombardo M, Tesauro M, et al. (2011) Serum 25-hydroxyvitamin D levels are inversely associated with systemic inflammation in severe obese subjects. Intern Emerg Med.
  618. Borazan A, Ustün H, Cefle A, Sekitmez N, Yilmaz A (2003) Comparative efficacy of oral and intravenous calcitriol treatment in haemodialysis patients: effects on serum biochemistry and cytokine levels. J Int Med Res 31: 489-496.
  619. Inanir A, Ozoran K, Tutkak H, Mermerci B (2004) The effects of calcitriol therapy on serum interleukin-1, interleukin-6 and tumour necrosis factor-alpha concentrations in post-menopausal patients with osteoporosis. J Int Med Res 32: 570-582.
  620. Schleithoff SS, Zittermann A, Tenderich G, Berthold HK, Stehle P, et al. (2006) Vitamin D supplementation improves cytokine profiles in patients with congestive heart failure: a double-blind, randomized, placebo-controlled trial. Am J Clin Nutr 83: 754-759.
  621. Fujita H, Asahina A, Komine M, Tamaki K (2007) The direct action of 1alpha,25(OH)2-vitamin D3 on purified mouse Langerhans cells. Cell Immunol 245: 70-79.
  622. Mitri J, Dawson-Hughes B, Hu FB, Pittas AG (2011) Effects of vitamin D and calcium supplementation on pancreatic β cell function, insulin sensitivity, and glycemia in adults at high risk of diabetes: the Calcium and Vitamin D for Diabetes Mellitus (CaDDM) randomized controlled trial. Am J Clin Nutr 94: 486-494.
  623. Brock KE, Huang WY, Fraser DR, Ke L, Tseng M, et al. (2011) Diabetes prevalence is associated with serum 25-hydroxyvitamin D and 1,25-dihydroxyvitamin D in US middle-aged Caucasian men and women: a cross-sectional analysis within the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. Br J Nutr 106: 339-344.
  624. Pittas AG, Dawson-Hughes B (2010) Vitamin D and diabetes. J Steroid Biochem Mol Biol 121: 425-429.
  625. Wolden-Kirk H, Overbergh L, Christesen HT, Brusgaard K, Mathieu C (2011) Vitamin D and diabetes: its importance for beta cell and immune function. Mol Cell Endocrinol 347: 106-120.
  626. Youssef DA, Miller CW, El-Abassi AM, Cutchins DC, Cutchins C, et al. (2011) Antimicrobial implications of vitamin D. DermatoEndocrinol 3: 220-229.
  627. Yu XP, Bellido T, Manolagas SC (1995) Down-regulation of NF-kappa B protein levels in activated human lymphocytes by 1,25-dihydroxyvitamin D3. Proc Natl Acad Sci U S A 92: 10990-10994.
  628. D'Ambrosio D, Cippitelli M, Cocciolo MG, Mazzeo D, Di Lucia P, et al. (1998) Inhibition of IL-12 production by 1,25-dihydroxyvitamin D3. Involvement of NF-kappaB downregulation in transcriptional repression of the p40 gene. J Clin Invest 101: 252-262.
  629. Giovannini L, Panichi V, Migliori M, De Pietro S, Bertelli AA, et al. (2001) 1,25-dihydroxyvitamin D(3) dose-dependently inhibits LPS-induced cytokines production in PBMC modulating intracellular calcium. Transplant Proc 33: 2366-2368.
  630. Chang JM, Kuo MC, Kuo HT, Hwang SJ, Tsai JC, et al. (2004) 1-alpha,25-dihydroxyvitamin D3 regulates inducible nitric oxide synthase messenger RNA expression and nitric oxide release in macrophage-like RAW 264.7 cells. J Lab Clin Med 143: 14-22.
  631. van Etten E, Mathieu C (2005) Immunoregulation by 1,25-dihydroxyvitamin D3: basic concepts. J Steroid Biochem Mol Biol 97: 93-101.
  632. Koren R, Ravid A, Rotem C, Shohami E, Liberman UA, et al. (1986) 1,25-Dihydroxyvitamin D3 enhances prostaglandin E2 production by monocytes. A mechanism which partially accounts for the antiproliferative effect of 1,25(OH)2D3 on lymphocytes. FEBS Lett 205: 113-116.
  633. Boonstra A, Barrat FJ, Crain C, Heath VL, Savelkoul HF, et al. (2001) 1alpha,25-Dihydroxyvitamin d3 has a direct effect on naive CD4(+) T cells to enhance the development of Th2 cells. J Immunol 167: 4974-4980.
  634. Gregori S, Giarratana N, Smiroldo S, Uskokovic M, Adorini L (2002) A 1alpha,25-dihydroxyvitamin D(3) analog enhances regulatory T-cells and arrests autoimmune diabetes in NOD mice. Diabetes 51: 1367-1374.
  635. Rajapakse R, Mousli M, Pfaff AW, Uring-Lambert B, Marcellin L, et al. (2005) 1,25-Dihydroxyvitamin D3 induces splenocyte apoptosis and enhances BALB/c mice sensitivity to toxoplasmosis. J Steroid Biochem Mol Biol 96: 179-185.
  636. Vial HJ, Thuet MJ, Philippot JR (1982) Inhibition of the in vitro growth of Plasmodium falciparum by D vitamins and vitamin D-3 derivatives. Mol Biochem Parasitol 5: 189-198.
  637. Rajapakse R, Uring-Lambert B, Andarawewa KL, Rajapakse RP, Abou-Bacar A, et al. (2007) 1,25(OH)2D3 inhibits in vitro and in vivo intracellular growth of apicomplexan parasite Toxoplasma gondii. J Steroid Biochem Mol Biol 103: 811-814.
  638. Ghaffarifar F, Pour MA, Sharifi Z, Asl AD, Al-Kawaz E (2010) The effect of vitamin D3 alone and mixed with IFN-g on tachyzoites of Toxoplasma gondii (RH strain) proloferation and nitric oxide (NO) production in infected macrophages of BALB/C mice. Iranian J Parasitol 5: 48-56.
  639. Rockett KA, Brookes R, Udalova I, Vidal V, Hill AV, et al. (1998) 1,25-Dihydroxyvitamin D3 induces nitric oxide synthase and suppresses growth of Mycobacterium tuberculosis in a human macrophage-like cell line. Infect Immun 66: 5314-5321.
  640. Wang C, Tay SS, Tran GT, Hodgkinson SJ, Allen RD, et al. (2010) Donor IL-4-treatment induces alternatively activated liver macrophages and IDO-expressing NK cells and promotes rat liver allograft acceptance. Transpl Immunol 22: 172-178.
  641. Jalili RB, Forouzandeh F, Moeenrezakhanlou A, Rayat GR, Rajotte RV, et al. (2009) Mouse pancreatic islets are resistant to indoleamine 2,3 dioxygenase-induced general control nonderepressible-2 kinase stress pathway and maintain normal viability and function. Am J Pathol 174: 196-205.
  642. Ueno A, Cho S, Cheng L, Wang J, Hou S, et al. (2007) Transient upregulation of indoleamine 2,3-dioxygenase in dendritic cells by human chorionic gonadotropin downregulates autoimmune diabetes. Diabetes 56: 1686-1693.
  643. Hwu P, Du MX, Lapointe R, Do M, Taylor MW, et al. (2000) Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of T cell proliferation. J Immunol 164: 3596-3599.
  644. Lestage J, Verrier D, Palin K, Dantzer R (2002) The enzyme indoleamine 2,3-dioxygenase is induced in the mouse brain in response to peripheral administration of lipopolysaccharide and superantigen. Brain Behav Immun 16: 596-601.
  645. Fujigaki S, Saito K, Sekikawa K, Tone S, Takikawa O, et al. (2001) Lipopolysaccharide induction of indoleamine 2,3-dioxygenase is mediated dominantly by an IFN-gamma-independent mechanism. Eur J Immunol 31: 2313-2318.
  646. Arsenijevic D, Girardier L, Seydoux J, Pechere JC, Garcia I, et al. (1998) Metabolic-cytokine responses to a second immunological challenge with LPS in mice with T. gondii infection. Am J Physiol 274: E439-445.
  647. Fallarino F, Volpi C, Zelante T, Vacca C, Calvitti M, et al. (2009) IDO mediates TLR9-driven protection from experimental autoimmune diabetes. J Immunol 183: 6303-6312.
  648. Saxena V, Ondr JK, Magnusen AF, Munn DH, Katz JD (2007) The countervailing actions of myeloid and plasmacytoid dendritic cells control autoimmune diabetes in the nonobese diabetic mouse. J Immunol 179: 5041-5053.
  649. Conti A, Conconi S, Hertens E, Skwarlo-Sonta K, Markowska M, et al. (2000) Evidence for melatonin synthesis in mouse and human bone marrow cells. J Pineal Res 28: 193-202.
  650. Carrillo-Vico A, Lardone PJ, Fernández-Santos JM, Martín-Lacave I, Calvo JR, et al. (2005) Human lymphocyte-synthesized melatonin is involved in the regulation of the interleukin-2/interleukin-2 receptor system. J Clin Endocrinol Metab 90: 992-1000.
  651. Allegra M, Reiter RJ, Tan DX, Gentile C, Tesoriere L, et al. (2003) The chemistry of melatonin's interaction with reactive species. J Pineal Res 34: 1-10.
  652. Ortiz GG, Reiter RJ, Zúñiga G, Melchiorri D, Sewerynek E, et al. (2000) Genotoxicity of paraquat: micronuclei induced in bone marrow and peripheral blood are inhibited by melatonin. Mutat Res 464: 239-245.
  653. Tan DX, Chen LD, Poeggeler B (1993) Melatonin: a potent, endogenous hydroxyl radical scavenger. Endocrine J 1: 57-60.
  654. Chuang JI, Mohan N, Meltz ML, Reiter RJ (1996) Effect of melatonin on NF-kappa-B DNA-binding activity in the rat spleen. Cell Biol Int 20: 687-692.
  655. Lezoualc’h F, Sagara Y, Holsboer F, Behl C (1998) High constitutive NF-kappa-B activity mediates resistance to oxidative stress in neuronal cells. J Neurosci 18: 3224-3232.
  656. Pozo D, Reiter RJ, Calvo JR, Guerrero JM (1997) Inhibition of cerebellar nitric oxide synthase and cyclic GMP production by melatonin via complex formation with calmodulin. J Cell Biochem 65: 430-442.
  657. Yavuz O, Cam M, Bukan N, Guven A, Silan F (2003) Protective effect of melatonin on b-cell damage in streptozotocin-induced diabetes in rats. Acta Histochem 105: 261-266.
  658. Rosales-Corral S, Tan DX, Reiter RJ, Valdivia-Velázquez M, Martínez-Barboza G, et al. (2003) Orally administered melatonin reduces oxidative stress and proinflammatory cytokines induced by amyloid-beta peptide in rat brain: a comparative, in vivo study versus vitamin C and E. J Pineal Res 35: 80-84.
  659. Chahbouni M, Escames G, Venegas C, Sevilla B, García JA, et al. (2010) Melatonin treatment normalizes plasma proinflammatory cytokines and nitrosative/oxidative stress in patients suffering from Duchenne muscular dystrophy. J Pineal Res 48: 282-289.
  660. Mayo JC, Sainz RM, Tan DX, Hardeland R, Leon J, et al. (2005) Anti-inflammatory actions of melatonin and its metabolites, N1-acetyl-N2-formyl-5-methoxykynuramine (AFMK) and N1-acetyl-5-methoxykynuramine (AMK), in macrophages. J Neuroimmunol 165: 139-149.
  661. Tan DX, Manchester LC, Burkhardt S, Sainz RM, Mayo JC, et al. (2001) N1-acetyl-N2-formyl-5-methoxykynuramine, a biogenic amine and melatonin metabolite, functions as a potent antioxidant. FASEB J 15: 2294-2296.
  662. Xu DX, Wang H, Ning H, Zhao L, Chen YH (2007) Maternally administered melatonin differentially regulates lipopolysaccharide-induced proinflammatory and anti-inflammatory cytokines in maternal serum, amniotic fluid, fetal liver, and fetal brain. J Pineal Res 43: 74-79.
  663. Peschke E, Peschke D, Hammer T, Csernus V (1997) Influence of melatonin and serotonin on glucose-stimulated insulin release from perifused rat pancreatic islets in vitro. J Pineal Res 23: 156-163.
  664. Peschke E, Schucht H, Mühlbauer E (2010) Long-term enteral administration of melatonin reduces plasma insulin and increases expression of pineal insulin receptors in both Wistar and type 2-diabetic Goto-Kakizaki rats. J Pineal Res 49: 373-381.
  665. Peschke E, Hofmann K, Pönicke K, Wedekind D, Mühlbauer E (2012) Catecholamines are the key for explaining the biological relevance of insulin-melatonin antagonisms in type 1 and type 2 diabetes. J Pineal Res 52: 389-396.
  666. Boucher BJ (1998) Inadequate vitamin D status: does it contribute to the disorders comprising syndrome 'X'? Br J Nutr 79: 315-327.
  667. Pezzella N, Bouchot A, Bonhomme A, Pingret L, Klein C, et al. (1997) Involvement of calcium and calmodulin in Toxoplasma gondii tachyzoite invasion. Eur J Cell Biol 74: 92-101.
  668. Lovett JL, Sibley LD (2003) Intracellular calcium stores in Toxoplasma gondii govern invasion of host cells. J Cell Sci 116: 3009-3016.
  669. Fujigaki H, Saito K (2007) Inhibition of increased indoleamine 2,3-dioxygenase activity exacerbates neuronal cell death in various CNS disorders. Int Congress Series 1304:314-323.
  670. Fujigaki S, Saito K, Takemura M, Maekawa N, Yamada Y, et al. (2002) L-tryptophan-L-kynurenine pathway metabolism accelerated by Toxoplasma gondii infection is abolished in gamma interferon-gene-deficient mice: cross-regulation between inducible nitric oxide synthase and indoleamine-2,3-dioxygenase. Infect Immun 70: 779-786.
  671. Heyes MP, Saito K, Crowley JS, Davis LE, Demitrack MA, et al. (1992) Quinolinic acid and kynurenine pathway metabolism in inflammatory and non-inflammatory neurological disease. Brain 115 : 1249-1273.
  672. Rios C, Santamaria A (1991) Quinolinic acid is a potent lipid peroxidant in rat brain homogenates. Neurochem Res 16: 1139-1143.
  673. Veneziale CM, Walter P, Kneer N, Lardy HA (1967) Influence of L-tryptophan and its metabolites on gluconeogenesis in the isolated, perfused liver. Biochemistry 6: 2129-2138.
  674. Spekker K, Czesla M, Ince V, Heseler K, Schmidt SK, et al. (2009) Indoleamine 2,3-dioxygenase is involved in defense against Neospora caninum in human and bovine cells. Infect Immun 77: 4496-4501.
  675. Cryer PE (2004) Diverse causes of hypoglycemia-associated autonomic failure in diabetes. N Engl J Med 350: 2272-2279.
  676. Davis EA, Jones TW (1998) Hypoglycemia in children with diabetes: incidence, counterregulation and cognitive dysfunction. J Pediatr Endocrinol Metab 11 Suppl 1: 177-182.
  677. Langan SJ, Deary IJ, Hepburn DA, Frier BM (1991) Cumulative cognitive impairment following recurrent severe hypoglycaemia in adult patients with insulin-treated diabetes mellitus. Diabetologia 34: 337-344.
  678. Ryan CM, Atchison J, Puczynski S, Puczynski M, Arslanian S, et al. (1990) Mild hypoglycemia associated with deterioration of mental efficiency in children with insulin-dependent diabetes mellitus. J Pediatr 117: 32-38.
  679. Auer RN, Siesjö BK (1993) Hypoglycaemia: brain neurochemistry and neuropathology. Baillieres Clin Endocrinol Metab 7: 611-625.
  680. Suh SW, Hamby AM, Swanson RA (2007) Hypoglycemia, brain energetics, and hypoglycemic neuronal death. Glia 55: 1280-1286.
  681. Wieloch T (1985) Hypoglycemia-induced neuronal damage prevented by an N-methyl-D-aspartate antagonist. Science 230: 681-683.
  682. Suh SW, Garnier P, Aoyama K, Chen Y, Swanson RA (2004) Zinc release contributes to hypoglycemia-induced neuronal death. Neurobiol Dis 16: 538-545.
  683. Suh SW, Gum ET, Hamby AM, Chan PH, Swanson RA (2007) Hypoglycemic neuronal death is triggered by glucose reperfusion and activation of neuronal NADPH oxidase. J Clin Invest 117: 910-918.
  684. Bao B, Prasad AS, Beck FW, Godmere M (2003) Zinc modulates mRNA levels of cytokines. Am J Physiol Endocrinol Metab 285: E1095-1102.
  685. Tasçi S, Sengil AZ, Altindis M, Arisoy K (1995) The effect of zinc supplementation in experimentally induced Toxoplasma gondii infection. J Egypt Soc Parasitol 25: 745-751.
  686. Chang HR, Comte R, Piguet PF, Pechère JC (1991) Activity of minocycline against Toxoplasma gondii infection in mice. J Antimicrob Chemother 27: 639-645.
  687. Chang HR, Comte R, Pechère JC (1990) In vitro and in vivo effects of doxycycline on Toxoplasma gondii. Antimicrob Agents Chemother 34: 775-780.
  688. Meyer U (2011) Anti-inflammatory signaling in schizophrenia. Brain Behav Immun 25: 1507-1518.
  689. Du Y, Ma Z, Lin S, Dodel RC, Gao F, et al. (2001) Minocycline prevents nigrostriatal dopaminergic neurodegeneration in the MPTP model of Parkinson's disease. Proc Natl Acad Sci U S A 98: 14669-14674.
  690. Chen M, Ona VO, Li M, Ferrante RJ, Fink KB, et al. (2000) Minocycline inhibits caspase-1 and caspase-3 expression and delays mortality in a transgenic mouse model of Huntington disease. Nat Med 6:797-801.
  691. Ryu JK, Franciosi S, Sattayaprasert P, Kim SU, McLarnon JG (2004) Minocycline inhibits neuronal death and glial activation induced by beta-amyloid peptide in rat hippocampus. Glia 48: 85-90.
  692. Yrjänheikki J, Keinänen R, Pellikka M, Hökfelt T, Koistinaho J (1998) Tetracyclines inhibit microglial activation and are neuroprotective in global brain ischemia. Proc Natl Acad Sci U S A 95: 15769-15774.
  693. Yrjänheikki J, Tikka T, Keinänen R, Goldsteins G, Chan PH, et al. (1999) A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc Natl Acad Sci U S A 96: 13496-13500.
  694. Elewa HF, Hilali H, Hess DC, Machado LS, Fagan SC (2006) Minocycline for short-term neuroprotection. Pharmacotherapy 26: 515-521.
  695. Metz LM, Zhang Y, Yeung M, Patry DG, Bell RB, et al. (2004) Minocycline reduces gadolinium-enhancing magnetic resonance imaging lesions in multiple sclerosis. Ann Neurol 55: 756.
  696. Kloppenburg M, Brinkman BM, de Rooij-Dijk HH, Miltenburg AM, Daha MR, et al. (1996) The tetracycline derivative minocycline differentially affects cytokine production by monocytes and T lymphocytes. Antimicrob Agents Chemother 40: 934-940.
  697. Miman O, Kusbeci OY, Aktepe OC, Cetinkaya Z (2010) The probable relation between Toxoplasma gondii and Parkinson's disease. Neurosci Lett 475: 129-131.
  698. Okusaga O, Langenberg P, Sleemi A, Vaswani D, Giegling I, et al. (2011) Toxoplasma gondii antibody titers and history of suicide attempts in patients with schizophrenia. Schizophr Res 133: 150-155.
  699. Emelia O, Amal RN, Ruzanna ZZ, Shahida H, Azzubair Z, et al. (2012) Seroprevalence of anti-Toxoplasma gondii IgG antibody in patients with schizophrenia. Trop Biomed 29: 151-159.
  700. Prandota J (2011) 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 5: 14-59
  701. Miman O, Mutlu EA, Ozcan O, Atambay M, Karlidag R, et al. (2010) Is there any role of Toxoplasma gondii in the etiology of obsessive-compulsive disorder? Psychiatry Res 177: 263-265.
  702. Miyaoka T (2008) Clinical potential of minocycline for schizophrenia. CNS Neurol Disord Drug Targets 7: 376-381.
  703. Levkovitz Y, Mendlovich S, Riwkes S, Braw Y, Levkovitch-Verbin H, et al. (2010) A double-blind, randomized study of minocycline for the treatment of negative and cognitive symptoms in early-phase schizophrenia. J Clin Psychiatry 71: 138-149.
  704. Fung HB, Kirschenbaum HL (1996) Treatment regimens for patients with toxoplasmic encephalitis. Clin Ther 18: 1037-1056.
  705. Shin BS, Won SJ, Yoo BH, Kauppinen TM, Suh SW (2010) Prevention of hypoglycemia-induced neuronal death by hypothermia. J Cereb Blood Flow Metab 30: 390-402.
  706. Möller JC, Klein MA, Haas S, Jones LL, Kreutzberg GW, et al. (1996) Regulation of thrombospondin in the regenerating mouse facial motor nucleus. Glia 17: 121-132.
  707. Vilhardt F (2005) Microglia: phagocyte and glia cell. Int J Biochem Cell Biol 37: 17-21.
  708. Wang Q, Tang XN, Yenari MA (2007) The inflammatory response in stroke. J Neuroimmunol 184: 53-68.
  709. Scholz M, Cinatl J, Schädel-Höpfner M, Windolf J (2007) Neutrophils and the blood-brain barrier dysfunction after trauma. Med Res Rev 27: 401-416.
  710. Aviles HO, Monroy FP (2001) Toxoplasma gondii: cold stress-induced modulation of antibody responses. Exp Parasitol 99: 89-96.
  711. Shanks N, Kusnecov AW (1998) Differential immune reactivity to stress in BALB/cByJ and C57BL/6J mice: in vivo dependence on macrophages. Physiol Behav 65: 95-103.
  712. Kohm AP, Sanders VM (1999) Suppression of antigen-specific Th2 cell-dependent IgM and IgG1 production following norepinephrine depletion in vivo. J Immunol 162: 5299-5308.
  713. VolchegorskiÄ­ IA, Kostin IuK, Skobeleva NA, Lifshits RI (1993) [Experimental study of adrenergic modulation of insulin sensitivity]. Probl Endokrinol (Mosk) 39: 36-40.
  714. Kostin IK, Volchegorskii IA, Lifshits RI, Grigorevskikh VS (1992) Insulin sensitivity during use of adrenoblockers and adrenomimetics. Patol Fiziol Eksp Ter 2: 46-48.
  715. Keijzers GB, De Galan BE, Tack CJ, Smits P (2002) Caffeine can decrease insulin sensitivity in humans. Diabetes Care 25: 364-369.
  716. Graham TE, Sathasivam P, Rowland M, Marko N, Greer F, et al. (2001) Caffeine ingestion elevates plasma insulin response in humans during an oral glucose tolerance test. Can J Physiol Pharmacol 79: 559-565.
  717. Sesti-Costa R, Ignacchiti MD, Chedraoui-Silva S, Marchi LF, Mantovani B (2012) Chronic cold stress in mice induces a regulatory phenotype in macrophages: correlation with increased 11β-hydroxysteroid dehydrogenase expression. Brain Behav Immun 26: 50-60.
  718. Oda S, Ikuta M, Kuhara T, Ohneda A, Sasaki Y (1995) Insulin and glucagon secretion in goats (Capra hircus Linnaeus) exposed to cold. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 111: 303-307.
  719. Oda S, Ohtomo Y, Ohneda A, Sasaki Y, Tsuda T (1986) Adrenergic modulation of pancreatic glucagon and insulin secretion in goats. Comp Biochem Physiol A Comp Physiol 84: 723-728.
  720. Aviles H, Monroy FP (2001) Immunomodulatory effects of cold stress on mice infected intraperitoneally with a 50% lethal dose of Toxoplasma gondii. Neuroimmunomodulation 9: 6-12.
  721. Gopal R, Birdsell D, Monroy FP (2011) Regulation of chemokine responses in intestinal epithelial cells by stress and Toxoplasma gondii infection. Parasite Immunol 33: 12-24.
  722. Banerjee SK, Aviles H, Fox MT, Monroy FP (1999) Cold stress-induced modulation of cell immunity during acute Toxoplasma gondii infection in mice. J Parasitol 85: 442-447.
  723. Benedetto N, Folgore A, Ferrara C, Molitierno M, Galdiero F (1997) Effects of alpha-adrenergic agonists on Toxoplasma gondii replication in human umbilical vein endothelial cells. Pathol Biol (Paris) 45: 9-18.
  724. Cheng GJ, Morrow-Tesch JL, Beller DI, Levy EM, Black PH (1990) Immunosuppression in mice induced by cold water stress. Brain Behav Immun 4: 278-291.
  725. Kizaki T, Oh-ishi S, Ohno H (1996) Acute cold stress induces suppressor macrophages in mice. J Appl Physiol 81: 393-399.
  726. Jain S, Bruot BC, Stevenson JR (1996) Cold swim stress leads to enhanced splenocyte responsiveness to concanavalin A, decreased serum testosterone, and increased serum corticosterone, glucose, and protein. Life Sci 59: 209-218.
  727. Giberson PK, Kim CK, Hutchinson S, Yu W, Junker A, et al. (1997) The effect of cold stress on lymphocyte proliferation in fetal ethanol-exposed rats. Alcohol Clin Exp Res 21: 1440-1447.
  728. Chevrier-Miller M, Morange M, Arrigo AP, Pinto M (1996) Low temperature enhancement of reporter genes expression directed by human immunodeficiency virus type 1 long terminal repeat. Biochem Biophys Res Commun 228: 695-703.
  729. Nishiyama H, Higashitsuji H, Yokoi H, Itoh K, Danno S, et al. (1997) Cloning and characterization of human CIRP (cold-inducible RNA-binding protein) cDNA and chromosomal assignment of the gene. Gene 204: 115-120.
  730. Thompson EG, Aviles HO, Monroy FP (2008) Antibodies in cold stressed mice recognize a surface protein in Toxoplasma gondii tachyzoites. J Parasitol 94: 114-118.
  731. Tomavo S, Couvreur G, Leriche MA, Sadak A, Achbarou A, et al. (1994) Immunolocalization and characterization of the low molecular weight antigen (4-5 kDa) of Toxoplasma gondii that elicits an early IgM response upon primary infection. Parasitology 108 : 139-145.
  732. Kang H, Remington JS, Suzuki Y (2000) Decreased resistance of B cell-deficient mice to infection with Toxoplasma gondii despite unimpaired expression of IFN-gamma, TNF-alpha, and inducible nitric oxide synthase. J Immunol 164: 2629-2634.
  733. Chrousos GP (1995) The hypothalamic-pituitary-adrenal axis and immune-mediated inflammation. N Engl J Med 332: 1351-1362.
  734. Madden KS, Sanders VM, Felten DL (1995) Catecholamine influences and sympathetic neural modulation of immune responsiveness. Annu Rev Pharmacol Toxicol 35: 417-448.
  735. Elenkov IJ, Wilder RL, Chrousos GP, Vizi ES (2000) The sympathetic nerve--an integrative interface between two supersystems: the brain and the immune system. Pharmacol Rev 52: 595-638.
  736. Elenkov IJ, Chrousos GP (2002) Stress hormones, proinflammatory and antiinflammatory cytokines, and autoimmunity. Ann N Y Acad Sci 966: 290-303.
  737. Padgett DA, Glaser R (2003) How stress influences the immune response. Trends Immunol 24: 444-448.
  738. Johnson JD, Campisi J, Sharkey CM, Kennedy SL, Nickerson M, et al. (2005) Catecholamines mediate stress-induced increases in peripheral and central inflammatory cytokines. Neuroscience 135: 1295-1307.
  739. Lyte M, Ernst S (1993) Alpha and beta adrenergic receptor involvement in catecholamine-induced growth of gram-negative bacteria. Biochem Biophys Res Commun 190: 447-452.
  740. Lyte M, Arulanandam BP, Frank CD (1996) Production of Shiga-like toxins by Escherichia coli O157:H7 can be influenced by the neuroendocrine hormone norepinephrine. J Lab Clin Med 128: 392-398.
  741. Gopal R, Birdsell D, Monroy FP (2008) Regulation of toll-like receptors in intestinal epithelial cells by stress and Toxoplasma gondii infection. Parasite Immunol 30: 563-576.
  742. Monroy FP, Banerjee SK, Duong T, Aviles H (1999) Cold stress-induced modulation of inflammatory responses and intracerebral cytokine mRNA expression in acute murine toxoplasmosis. J Parasitol 85: 878-886.
  743. Porta M, La Selva M, Molinatti P, Molinatti GM (1987) Endothelial cell function in diabetic microangiopathy. Diabetologia 30: 601-609.
  744. Jensen T, Bjerre-Knudsen J, Feldt-Rasmussen B, Deckert T (1989) Features of endothelial dysfunction in early diabetic nephropathy. Lancet 1: 461-463.
  745. Perassolo MS, Almeida JC, Steemburgo T, Dall’Alba V, de Mello VDF, et al. (2008) Endothelial dysfunction and serum fatty acid composition in patients with type 2 diabetes mellitus. Metabolism 57: 1167-1172.
  746. Hopfner RL, Gopalakrishnan V (1999) Endothelin: emerging role in diabetic vascular complications. Diabetologia 42: 1383-1394.
  747. Schaumberg DA, Glynn RJ, Jenkins AJ, Lyons TJ, Rifai N, et al. (2005) Effect of intensive glycemic control on levels of markers of inflammation in type 1 diabetes mellitus in the diabetes control and complications trial. Circulation 111: 2446-2453.
  748. Hamed S, Brenner B, Aharon A, Daoud D, Roguin A (2009) Nitric oxide and superoxide dismutase modulate endothelial progenitor cell function in type 2 diabetes mellitus. Cardiovasc Diabetol 8: 56.
  749. Wattanapitayakul SK, Weinstein DM, Holycross BJ, Bauer JA (2000) Endothelial dysfunction and peroxynitrite formation are early events in angiotensin-induced cardiovascular disorders. FASEB J 14: 271-278.
  750. Thum T, Fraccarollo D, Schultheiss M, Froese S, Galuppo P, et al. (2007) Endothelial nitric oxide synthase uncoupling impairs endothelial progenitor cell mobilization and function in diabetes. Diabetes 56: 666-674.
  751. Sorrentino SA, Bahlmann FH, Besler C, Muller M, Schulz S, et al. (2007) Oxidant stress impairs in vivo reendothelialization capacity of endothelial progenitor cells from patients with type 2 diabetes mellitus: restoration by the peroxisome proliferator-activated receptor-gamma agonist rosiglitazone. Circulation 116: 163-173.
  752. Milsom AB, Jones CJ, Goodfellow J, Frenneaux MP, Peters JR, et al. (2002) Abnormal metabolic fate of nitric oxide in type 1diabetes mellitus. Diabetologia 45: 1515-1522.
  753. Cellek S, Rodrigo J, Lobos E, Fernández P, Serrano J, et al. (1999) Selective nitrergic neurodegeneration in diabetes mellitus - a nitric oxide-dependent phenomenon. Br J Pharmacol 128: 1804-1812.
  754. Faerman I, Glocer L, Fox D, Jadzinsky MN, Rapaport M (1974) Impotence and diabetes. Histological studies of the autonomic nervous fibers of the corpora cavernosa in impotent diabetic males. Diabetes 23: 971-976.
  755. Hulinska D, Sykora J, Zastera M (1993) The effect of cyclosporine A on the development of Toxoplasma gondii in the muscles and brain. Ultrastructural study. Bratisl Lek Listy 94: 241-248.
  756. Brunet LR (2001) Nitric oxide in parasitic infections. Int Immunopharmacol 1: 1457-1467.
  757. Chandrasekharan B, Srinivasan S (2007) Diabetes and the enteric nervous system. Neurogastroenterol Motil 19: 951-960.
  758. Kasper L, Courret N, Darche S, Luangsay S, Mennechet F, et al. (2004) Toxoplasma gondii and mucosal immunity. Int J Parasitol 34: 401-409.
  759. Schlüter D, Deckert-Schlüter M, Lorenz E, Meyer T, Röllinghoff M, et al. (1999) Inhibition of inducible nitric oxide synthase exacerbates chronic cerebral toxoplasmosis in Toxoplasma gondii-susceptible C57BL/6 mice but does not reactivate the latent disease in T. gondii-resistant BALB/c mice. J Immunol 162: 3512-3518.
  760. Kurahashi N (1987) Correction: Common AIDS-Related Problems. West J Med 146: 489.
  761. Srinivasan AR, Niranjan G, Kuzhandai Velu V, Parmar P, et al. (2012) Status of serum magnesium in type 2 diabetes mellitus with particular reference to serum triacylglycerol levels. Diabetes Metab Syndr 6: 187-189.
  762. Dasgupta A, Sarma D, Saikia UK (2012) Hypomagnesemia in type 2 diabetes mellitus. Indian J Endocrinol Metab 16: 1000-1003.
  763. Mohr S, Hallak H, de Boitte A, Lapetina EG, Brüne B (1999) Nitric oxide-induced S-glutathionylation and inactivation of glyceraldehyde-3-phosphate dehydrogenase. J Biol Chem 274: 9427-9430.
  764. Beale EG, Hammer RE, Antoine B, Forest C (2004) Disregulated glyceroneogenesis: PCK1 as a candidate diabetes and obesity gene. Trends Endocrinol Metab 15: 129-135.
  765. Forest C, Tordjman J, Glorian M, Duplus E, Chauvet G, et al. (2003) Fatty acid recycling in adipocytes: a role for glyceroneogenesis and phosphoenolpyruvate carboxykinase. Biochem Soc Trans 31: 1125-1129.
  766. Stumbo AC, Barbosa HS, Carvalho TM, Porto LC, Carvalho Ld (2002) Anionic sites, fucose residues and class I human leukocyte antigen fate during interaction of Toxoplasma gondii with endothelial cells. Mem Inst Oswaldo Cruz 97: 517-522.
  767. Sinai AP, Joiner KA (2001) The Toxoplasma gondii protein ROP2 mediates host organelle association with the parasitophorous vacuole membrane. J Cell Biol 154: 95-108.
  768. Guimarães EV, Acquarone M, de Carvalho L, Barbosa HS (2007) Anionic sites on Toxoplasma gondii tissue cyst wall: expression, uptake and characterization. Micron 38: 651-658.
  769. Weiss LM, Kim K (2007) Toxoplasma gondii: the model apicomplexan. Perspectives and methods. The Ultrastructure of Toxoplasma gondii. Elsevier, Rio de Janeiro: 317-340.
  770. Cortez E, Stumbo AC, de Carvalho TM, Barbosa HS, Carvalho L (2005) NAD(P)H-oxidase presence in Toxoplasma gondii tachyzoite vacuole during interaction with IFN-gamma-activated human endothelial cells. J Parasitol 91: 1052-1057.
  771. Dando C, Schroeder ER, Hunsaker LA, Deck LM, Royer RE, et al. (2001) The kinetic properties and sensitivities to inhibitors of lactate dehydrogenases (LDH1 and LDH2) from Toxoplasma gondii: comparisons with pLDH from Plasmodium falciparum. Mol Biochem Parasitol 118: 23-32.
  772. Deck LM, Vander Jagt DL, Royer RE (1991) Gossypol and derivatives: a new class of aldose reductase inhibitors. J Med Chem 34: 3301-3305.
  773. Yu Y, Deck JA, Hunsaker LA, Deck LM, Royer RE, et al. (2001) Selective active site inhibitors of human lactate dehydrogenases A4, B4, and C4. Biochem Pharmacol 62: 81-89.
  774. Hamada Y, Kitoh R, Raskin P (1993) Association of erythrocyte aldose reductase activity with diabetic complications in type 1 diabetes mellitus. Diabet Med 10: 33-38.
  775. Tomlinson DR (1993) Aldose reductase inhibitors and the complications of diabetes mellitus. Diabet Med 10: 214-230.
  776. Chandra D, Jackson EB, Ramana KV, Kelley R, Srivastava SK, et al. (2002) Nitric oxide prevents aldose reductase activation and sorbitol accumulation during diabetes. Diabetes 51: 3095-3101.
  777. Lim SS, Jung SH, Ji J, Shin KH, Keum SR (2001) Synthesis of flavonoids and their effects on aldose reductase and sorbitol accumulation in streptozotocin-induced diabetic rat tissues. J Pharm Pharmacol 53: 653-668.
  778. Canedo-Solares I, Calzada-Ruiz M, Ortiz-Alegria LB, Ortiz Muniz AR, Luna-Pasten H, et al. (2012) Invasion kinetics of human endothelial cells by Toxoplasma gondii RH and ME49 strains. Int J Infect Dis 165: e2-e157.
  779. Lachenmaier SM, Deli MA, Meissner M, Liesenfeld O (2011) Intracellular transport of Toxoplasma gondii through the blood-brain barrier. J Neuroimmunol 232: 119-130.
  780. Knight BC, Brunton CL, Modi NC, Wallace GR, Stanford MR (2005) The effect of Toxoplasma gondii infection on expression of chemokines by rat retinal vascular endothelial cells. J Neuroimmunol 160: 41-47.
  781. Taubert A, Krüll M, Zahner H, Hermosilla C (2006) Toxoplasma gondii and Neospora caninum infections of bovine endothelial cells induce endothelial adhesion molecule gene transcription and subsequent PMN adhesion. Vet Immunol Immunopathol 112: 272-283.
  782. Taubert A, Hermosilla C, Behrendt J, Zahner H (2006) Reaction of bovine endothelial cells in vitro to coccidia (Eimeria bovis, Toxoplasma gondii, Neospora caninum) infections as the expression of a non-adaptive immune response. Berl Munch Tierarztl Wochenschr 119: 274-281.
  783. Furtado JM, Bharadwaj AS, Ashander LM, Olivas A, Smith JR (2012) Migration of toxoplasma gondii-infected dendritic cells across human retinal vascular endothelium. Invest Ophthalmol Vis Sci 53: 6856-6862.
  784. Zamora DO, Rosenbaum JT, Smith JR (2008) Invasion of human retinal vascular endothelial cells by Toxoplasma gondii tachyzoites. Br J Ophthalmol 92: 852-855.
  785. Smith JR, Franc DT, Carter NS, Zamora D, Planck SR, et al. (2004) Susceptibility of retinal vascular endothelium to infection with Toxoplasma gondii tachyzoites. Invest Ophthalmol Vis Sci 45: 1157-1161.
  786. Carruthers VB, Håkansson S, Giddings OK, Sibley LD (2000) Toxoplasma gondii uses sulfated proteoglycans for substrate and host cell attachment. Infect Immun 68: 4005-4011.
  787. Ortega-Barria E, Boothroyd JC (1999) A Toxoplasma lectin-like activity specific for sulfated polysaccharides is involved in host cell infection. J Biol Chem 274: 1267-1276.
  788. Brunton CL, Wallace GR, Graham E, Stanford MR (2000) The effect of cytokines on the replication of T. gondii within rat retinal vascular endothelial cells. J Neuroimmunol 102: 182-188.
  789. Dimier IH, Bout DT (1996) Inhibitory effect of interferon-gamma activated ovine umbilical vein endothelial cells on the intracellular replication of Toxoplasma gondii. Vet Res 27: 527-534.
  790. Ji YS, Sun XM, Liu XY, Suo X (2012) Toxoplasma gondii: Effects of exogenous nitric oxide on egress of tachyzoites from infected macrophages. Exp Parasitol 133: 70-74
  791. Behrendt JH, Taubert A, Zahner H, Hermosilla C (2008) Studies on synchronous egress of coccidian parasites (Neospora caninum, Toxoplasma gondii, Eimeria bovis) from bovine endothelial host cells mediated by calcium ionophore A23187. Vet Res Commun 32: 325-332.
  792. Dimier IH, Bout DT (1993) Co-operation of interleukin-1 beta and tumour necrosis factor-alpha in the activation of human umbilical vein endothelial cells to inhibit Toxoplasma gondii replication. Immunology 79: 336-338.
  793. Schober E, Otto KP, Dost A, Jorch N, Holl R, et al. (2012) Association of epilepsy and type 1 diabetes mellitus in children and adolescents: is there an increased risk for diabetic ketoacidosis? J Pediatr 160: 662-666.
  794. Martinez C, Sullivan T, Hauser WA (2009) Prevalence of acute repetitive seizures (ARS) in the United Kingdom. Epilepsy Res 87: 137-143.
  795. Ramakrishnan R, Appleton R (2012) Study of prevalence of epilepsy in children with type 1 diabetes mellitus. Seizure 21: 292-294.
  796. Caietta E, Halbert C, Lépine A, Khammar A, Cano A, et al. (2012) Association of type 1 diabetes mellitus and epilepsy in children. A cohort of 10 cases. Arch Pediatr 19: 9-16.
  797. Carter RE, Lackland DT, Cleary PA, Yim E, Lopes-Virella MF, et al. (2007) Intensive treatment of diabetes is associated with a reduced rate of peripheral arterial calcification in the diabetes control and complications trial. Diabetes Care 30: 2646-2648.
  798. Wagner VM, Rosenbauer J, Grabert M, Holl RW; German Initiative on Quality Control in Pediatric Diabetology (2008) Severe hypoglycemia, metabolic control and diabetes management in young children with type 1 diabetes using insulin analogs--a follow up report of a large multicenter database. Eur J Ped 167: 241-242.
  799. O'Connell MA, Harvey AS, Mackay MT, Cameron FJ (2008) Does epilepsy occur more frequently in children with Type 1 diabetes? J Paediatr Child Health 44: 586-589.
  800. Jones-Brando L, Torrey EF, Yolken R (2003) Drugs used in the treatment of schizophrenia and bipolar disorder inhibit the replication of Toxoplasma gondii. Schizophr Res 62: 237-244.
  801. Gibbs JP, Adeyeye MC, Yang Z, Shen DD (2004) Valproic acid uptake by bovine brain microvessel endothelial cells: role of efflux transport. Epilepsy Res 58: 53-66.
  802. Glodek-Brzozowska E, Surdej B, Lisowicz L, Czyzyk E, Korczowski B (2011) Coexistence of type I diabetes mellitus and epilepsy in children in south-eastern Poland. Pediatria Pol 86: 596-599.
  803. Stommel EW, Seguin R, Thadani VM, Schwartzman JD, Gilbert K, et al. (2001) Cryptogenic epilepsy: an infectious etiology? Epilepsia 42: 436-438.
  804. Groeper K, McCann ME (2005) Topiramate and metabolic acidosis: a case series and review of literature. Pediatr Anesth 15: 167-170.
  805. Palmer BS (2007) Meta-analysis of three case controlled studies and an ecological study into the link between cryptogenic epilepsy and chronic toxoplasmosis infection. Seizure 16: 657-663.
  806. Szwabowska-Orzeszko E, Jozwiak S, Michalowicz R (1993) Autosomal recessive disease. Acta Neurol Scand 87: 428.
  807. Paiboonpol S (2005) Epilepsia partialis continua as a manifestation of hyperglycemia. J Med Assoc Thai 88: 759-762.
  808. Berg-Beckhoff G, Schüz J, Blettner M, Münster E, Schlaefer K, et al. (2009) History of allergic disease and epilepsy and risk of glioma and meningioma (INTERPHONE study group, Germany). Eur J Epidemiol 24: 433-440.
  809. Prandota J (2009) The importance of toxoplasma gondii infection in diseases presenting with headaches. Headaches and aseptic meningitis may be manifestations of the Jarisch-Herxheimer reaction. Int J Neurosci 119: 2144-2182.
  810. Aamodt AH, Stovner LJ, Midthjell K, Hagen K, Zwart JA (2007) Headache prevalence related to diabetes mellitus. The Head-HUNT study. Eur J Neurol 14: 738-744.
  811. Jousilahti P, Tuomilehto J, Rastenyte D, Vartiainen E (2003) Headache and the risk of stroke: a prospective observational cohort study among 35,056 Finnish men and women. Arch Intern Med 163: 1058-1062.
  812. Split W, Szydlowska M (1997) Headaches in non insulin-dependent diabetes mellitus. Funct Neurol 12: 327-332.
  813. Sillanpää M, Aro H (2000) Headache in teenagers: comorbidity and prognosis. Funct Neurol 15: 116-121.
  814. Tietjen GE, Herial NA, Hardgrove J, Utley C, White L (2007) Migraine comorbidity constellations. Headache 47: 857-865.
  815. Cavestro C, Rosatello A, Micca G, Ravotto M, Marino MP, et al. (2007) Insulin metabolism is altered in migraineurs: a new pathogenic mechanism for migraine? Headache 47: 1436-1442.
  816. Okada H, Araga S, Takeshima T, Nakashima K (1998) Plasma lactic acid and pyruvic acid levels in migraine and tension-type headache. Headache 38: 39-42.
  817. Rainero I, Limone P, Ferrero M, Valfrè W, Pelissetto C, et al. (2005) Insulin sensitivity is impaired in patients with migraine. Cephalalgia 25: 593-597.
  818. Guldiken B, Guldiken S, Demir M, Turgut N, Kabayel L, et al. (2008) Insulin resistance and high sensitivity C-reactive protein in migraine. Can J Neurol 35: 448-451.
  819. Prandota J (2007) 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 14: 63-105.
  820. Koseoglu E, Yazar S, Koc I (2009) Is Toxoplasma gondii a causal agent in migraine? Am J Med Sci 338: 120-122.
  821. Freeman SJ, Roberts W, Daneman D (2005) Type 1 diabetes and autism: is there a link? Diabetes Care 28: 925-926.
  822. Comi AM, Zimmerman AW, Frye VH, Law PA, Peeden JN (1999) Familial clustering of autoimmune disorders and evaluation of medical risk factors in autism. J Child Neurol 14: 388-394.
  823. Menon PS, Vaidyanathan B, Kaur M (2001) Autoimmune thyroid disease in Indian children with type 1 diabetes mellitus. J Pediatr Endocrinol Metab 14: 279-286.
  824. Park YS, Kim TW, Kim WB, Cho BY (2000) Increased prevalence of autoimmune thyroid disease in patients with type 1 diabetes. Korean J Intern Med 15: 202-210.
  825. Prázný M, Skrha J, Límanová Z, Hilgertová J (1999) The evaluation of thyroid and islet autoantibodies in type 1 diabetes mellitus. Sb Lek 100: 205-211.
  826. Harjutsalo V, Tuomilehto J (2006) Type 1 diabetes and autism: is there a link? Diabetes Care 29: 484-485.
  827. Iafusco D, Vanelli M, Songini M, Chiari G, Cardella F, et al. (2006) Type 1 diabetes and autism association seems to be linked to the incidence of diabetes. Diabetes Care 29: 1985-1986.
  828. Prandota J (2012) 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. Nova Science Publishers, New York, 447-638.
  829. Goldszmid RS, Coppens I, Lev A, Caspar P, Mellman I, et al. (2009) Host ER-parasitophorous vacuole interaction provides a route of entry for antigen cross-presentation in Toxoplasma gondii-infected dendritic cells. J Exp Med 206: 399-410.
  830. Delong G (2011) A positive association found between autism prevalence and childhood vaccination uptake across the U.S. population. J Toxicol Environ Health A 74: 903-916.
  831. Sogut S, Zoroglu SS, Ozyurt H, Yilmaz HR, Ozugurlu F, et al. (2003) Changes in nitric oxide levels and antioxidant enzyme activities may have a role in the pathophysiological mechanisms involved in autism. Clin Chim Acta 331: 111-117.
  832. Zoroglu SS, Yurekli M, Meram I, Sogut S, Tutkun H, et al. (2003) Pathophysiological role of nitric oxide and adrenomedullin in autism. Cell Biochem Funct 21: 55-60.
  833. Cohen BI (2006) Ammonia (NH3), nitric oxide (NO) and nitrous oxide (N2O)--the connection with infantile autism. Autism 10: 221-223.
  834. Palmieri L, Persico AM (2010) Mitochondrial dysfunction in autism spectrum disorders: cause or effect? Biochim Biophys Acta 1797: 1130-1137.
  835. Kilbourn RG, Belloni P (1990) Endothelial cell production of nitrogen oxides in response to interferon gamma in combination with tumor necrosis factor, interleukin-1, or endotoxin. J Natl Cancer Inst 82: 772-776.
  836. Nussler AK, Di Silvio M, Billiar TR, Hoffman RA, Geller DA, et al. (1992) Stimulation of the nitric oxide synthase pathway in human hepatocytes by cytokines and endotoxin. J Exp Med 176: 261-264.
  837. Ross R, Glomset JA (1973) Atherosclerosis and the arterial smooth muscle cell: Proliferation of smooth muscle is a key event in the genesis of the lesions of atherosclerosis. Science 180: 1332-1339.
  838. Diamond JR, Karnovsky MJ (1988) Focal and segmental glomerulosclerosis: analogies to atherosclerosis. Kidney Int 33: 917-924.
  839. Camarca ME, Mozzillo E, Nugnes R, Zito E, Falco M, et al. (2012) Celiac disease in type 1 diabetes mellitus. Ital J Pediatr 38: 10.
  840. Franzese A, Iafusco D, Spadaro R, Cavaliere O, Prisco F, et al. (2011) Potential celiac disease in type 1 diabetes: a multicenter study. Diabetes Res Clin Pract 92: 53-56.
  841. Volta U, Tovoli F, Caio G (2011) Clinical and immunological features of celiac disease in patients with Type 1 diabetes mellitus. Expert Rev Gastroenterol Hepatol 5: 479-487.
  842. Virgin HW, Todd JA (2011) Metagenomics and personalized medicine. Cell 147: 44-56.
  843. Cerutti F, Bruno G, Chiarelli F, Lorini R, Meschi F, et al. (2004) Younger age at onset and sex predict celiac disease in children and adolescents with type 1 diabetes: an Italian multicenter study. Diabetes Care 27: 1294-1298.
  844. Valerio G, Maiuri L, Troncone R, Buono P, Lombardi F, et al. (2002) Severe clinical onset of diabetes and increased prevalence of other autoimmune diseases in children with coeliac disease diagnosed before diabetes mellitus. Diabetologia 45: 1719-1722.
  845. Cronin CC, Jackson LM, Feighery C, Shanahan F, Abuzakouk M, et al. (1998) Coeliac disease and epilepsy. QJM 91: 303-308.
  846. Sandberg-Bennich S, Dahlquist G, Källén B (2002) Coeliac disease is associated with intrauterine growth and neonatal infections. Acta Paediatr 91: 30-33.
  847. Rostami Nejad M, Rostami K, Cheraghipour K, Nazemalhosseini Mojarad E, Volta U, et al. (2011) Celiac disease increases the risk of Toxoplasma gondii infection in a large cohort of pregnant women. Am J Gastroenterol 106: 548-549.
  848. Severance EG, Kannan G, Gressitt KL, Xiao J, Alaedini A, et al. (2012) Anti-Gluten Immune Response following Toxoplasma gondii Infection in Mice. PLoS One 7: e50991.
  849. Barragan A, Sibley LD (2002) Transepithelial migration of Toxoplasma gondii is linked to parasite motility and virulence. J Exp Med 195: 1625-1633.
  850. Manar E, Laila Y (2012) The role of breastfeeding and the intake of bovine colostrum in autistic neonatal rats with coeliac disease. Int J Mol Zoology 2: 1-12.
Citation: Prandota J (2013) T. gondii Infection Acquired during Pregnancy and/ or after Birth may be Responsible for Development of both Type 1 and 2 Diabetes Mellitus. J Diabetes Metab 4:241.

Copyright: © 2013 Prandota J. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.