ChapterPDF Available

Rhesus-associated glycoprotein (RhAG) phenotype of the red blood cells modulates T. gondii infection-associated psychomotor performance reaction times and changes in the human personality profile. Impaired function of the CO2, AQP1, and AQP4 gas channels may cause hypoxia and thus enhance neuroinflammation in autistic individuals

Authors:

Abstract

It was reported that in humans the response of the host to latent chronic T. gondii infection was dependent on RhD phenotype. Rh-positive individuals, and RhD-positive heterozygotes in particular, appeared to be protected against latent toxoplasmosis-induced changes of personality, increased frequency of traffic accidents, and weight gain disturbances. One may suggest that these disturbances could be associated with development of brain hypoxia because recent studies showed that the Rhesus-associated glycoprotein (CcEe and D proteins) (RhAG) and water channel aquaporin1 (AQP1) were equally responsible for the normal CO2 permeability of the red blood cell membrane. In addition, AQP4, the predominant water channel expressed primarily in astrocytes and ependymocytes in the brain, also regulated hypoxia through mediation of bicarbonate transport, and a hypoxia inducible factor binding motif has been identified in the promoter region of AQP4 gene. It seems that lack or deficiency of RhAG proteins in the host red blood cell membrane and an impaired function of AQP1 and AQP4 water/gas channels in the central nervous system could be associated with various degrees of brain hypoxia. The proinflammatory changes in brain tissue associated with hypoxia may therefore overlap chronic neuroinflammation characteristic for patients with ASD and some other neurodevelopmental disorders, thus affecting the severity of clinical course and intensity of signs and symptoms present in these individuals. The amelioration of several underlying pathophysiological problems in autistic individuals and improvements in clinical symptoms through the use of hyperbaric oxygen therapy is consistent with this reasoning. It is also suggested that an impaired water movement in brain tissues associated with a decreased water channel AQP4 expression in astrocytes and ependymal cells found in autistic patients may be, at least in part, responsible for development of macrocephaly and increased brain weight reported in some of those individuals.
Chapter
RHESUS-ASSOCIATED GLYCOPROTEIN
(RHAG) PHENOTYPE OF THE RED BLOOD
CELLS MODULATES T. GONDII INFECTION-
ASSOCIATED PSYCHOMOTOR
PERFORMANCE REACTION TIMES AND
CHANGES IN THE HUMAN PERSONALITY
PROFILE. IMPAIRED FUNCTION OF THE
CO2, AQP1, AND AQP4 GAS CHANNELS
MAY CAUSE HYPOXIA AND THUS
ENHANCE NEUROINFLAMMATION IN
AUTISTIC INDIVIDUALS
Joseph Prandota*
* Correspondin Author:
Dr Joseph Prandota,
Department of Social Pediatrics,
Faculty of Health Science, University Medical School,
5 Bartla Street, 51-618, Wroclaw, Poland.
Tel.: +48 71 348 42 10, Fax: +48 71 345 93 24
E-mail: prandota@ak.am.wroc.pl
Joseph Prandota
Dpt of Social Pediatrics, Faculty of Health Sciences, University School of
Medicine, Wroclaw, Poland
ABSTRACT
It was reported that in humans the response of the host to latent
chronic T. gondii infection was dependent on RhD phenotype. Rh-
positive individuals, and RhD-positive heterozygotes in particular,
appeared to be protected against latent toxoplasmosis-induced changes
of personality, increased frequency of traffic accidents, and weight gain
disturbances. One may suggest that these disturbances could be
associated with development of brain hypoxia because recent studies
showed that the Rhesus-associated glycoprotein (CcEe and D proteins)
(RhAG) and water channel aquaporin-1 (AQP1) were equally
responsible for the normal CO2 permeability of the red blood cell
membrane. In addition, AQP4, the predominant water channel
expressed primarily in astrocytes and ependymocytes in the brain, also
regulated hypoxia through mediation of bicarbonate transport, and a
hypoxia inducible factor binding motif has been identified in the
promoter region of AQP4 gene. It seems that lack or deficiency of
RhAG proteins in the host red blood cell membrane and an impaired
function of AQP1 and AQP4 water/gas channels in the central nervous
system could be associated with various degrees of brain hypoxia. The
proinflammatory changes in brain tissue associated with hypoxia may
therefore overlap chronic neuroinflammation characteristic for patients
with ASD and some other neurodevelopmental disorders, thus affecting
the severity of clinical course and intensity of signs and symptoms
present in these individuals. The amelioration of several underlying
pathophysiological problems in autistic individuals and improvements
in clinical symptoms through the use of hyperbaric oxygen therapy is
consistent with this reasoning. It is also suggested that an impaired
water movement in brain tissues associated with a decreased water
channel AQP4 expression in astrocytes and ependymal cells found in
autistic patients may be, at least in part, responsible for development of
macrocephaly and increased brain weight reported in some of those
individuals.
Keywords: T. gondii infection, RhAG phenotype, changes of personality
profile, red blood cell membrane CO2 permeability, aquaporin-1, brain
aquaporin-4, hypoxia, neuroinflammation, autism spectrum disorders, latent
cerebral toxoplasmosis.
2
Brain hypoxia May be Due to Defects in RhAG Phenotype and AQP4
EFFECT OF RH PHENOTYPE ON PERSONALITY PROFILE
IN HUMANS WITH LATENT CHRONIC
T. GONDII INFECTION
Approximately 15-85% of the adult human population is chronically
infected by T. gondii depending on geographical areas, hygenic standards,
eating habits, and exposure to cats [1-3], with the brain preferential
involvement [4]. Until recently, it was believed that in the majority of the
cases, human toxoplasmosis has a benign and asymptomatic course but lead
to lifelong persistence of the parasite [5, 6]. In the USA, 1.5 million
infections are estimated to occur annually, with 15% of them being
asymptomatic [7-9]. Infection with T. gondii results in retinochoroiditis in
6% to 20% of immunocompetent individuals [10], and may be responsible
for neurologic and psychiatric symptoms associated with several diseases
[11].
It was reported that young men diagnosed with chronic T. gondii
infection became more introspective, more jealous, easily bored, showed
reduced psychomotor activity and reaction times, were emotionally unstable,
suspicious, had short temper, low self-esteem, and disregarded social rules
[12]. In addition, they were more prone to guilt and showed group
dependency when compared with young women infected with the parasite,
who showed greater self-esteem, were more inteligent, aware, cordial,
participative, amicable, attentive to others, rigid, loyal and self-sufficient,
respected social rules, were sentimental, socially precise, and affective [7, 13,
14]. Both, infected men and women were significantly more anxious than
noninfected individuals [6].
Recently, few independent studies in humans suggested that the response
of the host to latent T. gondii infection was dependent on RhD phenotype
[15-19]. It was demonstrated that RhD-positive individuals, and RhD-
positive heterozygotes in particular, have been protected against latent
toxoplasmosis-induced impairment of psychomotor performance reaction
times estimated by Cattell’s 16PF and Cloninger’s TCI questionnaires as
compared with Rh-negative individuals [15, 18]. Rh-positive heterozygotes
were permanently while Rh-positive heterozygotes were temporarily
protected against such impairment. RhD-positive and RhD-negative
phenotypes had opposite effects on ego strength, praxemia, ergic tension, and
cooperativeness [17]. RhD-positive individuals were also protected against
the parasite-induced increased risk of traffic accidents [16]. A cohort study
3
Joseph Prandota
performed on a population of military conscripts showed that RhD-negative
Toxoplasma-infected individuals had about three times higher probability of
a traffic accident than RhD-negative the parasite-free persons or RhD-
positive (T. gondii-free or Toxoplasma-infected) individuals [16]. In another
study performed on 980 pregnant women it was found that the RhD-
positivity protected T. gondii-infected women against excessive weight gain
in the first trimester of pregnancy [19].
2. DIMINISHED BASAL CO2 PERMEABILITY OF THE
RED BLOOD CELL MEMBRANE. IMPORTANT ROLE OF
THE RHESUS-ASSOCIATED GLYCOPROTEIN (RHAG) AS
A CO2 AND NH3 GAS CHANNELS
The Rh blood group substance is one of the most abundant proteins in
the erythrocyte membrane and the abnormalities of structure in Rhnull red cells
indicate that the Rh family polypeptide proteins plays a role in maintaining
the flattened (biconcave discoid) shape of the erythrocyte [20-23]. This
cytoskeletal function increases the surface area to volume ratio relative to a
sphere and hence the rate of diffusion of gases, including that of CO2 [23].
Callebaut et al. [24] speculated that either Rh (CcEe or D) protein might be a
candidate for a CO2 channel, but Beckmann et al. [25] proposed that both
RhCcEe and RhD proteins, which constitues a heterooligomer with RhAG,
give rise to the immunological differences between Rh-positive and Rh-
negative people [23, 26, 27] may be particularly important to the human
erythrocyte cytoskeleton role of the Rh blood group substance. Recently,
Endeward et al. [28, 29] confirmed that the high CO2 permeability of the
human red blood cell (RBC) membrane was due to the two blood group
membrane proteins (including CcEe and D proteins) and water channel
aquaporin-1 protein (AQP-1), and each of the RhAG and AQP-1 proteins
was responsible for about half of the normal CO 2 permeability of the RBC
membrane [28, 30] (Table 1).
4
Brain hypoxia May be Due to Defects in RhAG Phenotype and AQP4
Table 1. Time required by red blood cells to complete CO2 release
during passage through the lung capillary by 95% (t95%)
Bioparameter PCO2 (cm/sec) T95% (ms)
No membrane
resistance
50
Normal membrane
permeability 0.15 110
AQP-1null or Rhnull
RBCs
0.07 180
Basal permeability
(AQP-1null + DIDS) 0.01 1000
Values calculated for different CO2 permeabilities of the cell membrane.
Intraerythrocytic carbonic anhydrase acitivity was assumed to be nonlimiting.
DIDS, 4,4’-diisothiocyanato-stilbene-2,2’-disulfonate was used as an inhibitor of
RBC permeability. RBCs, red blood cells.
Endeward et al. [29]; with own modification.
3. RELATIVE CO2 AND NH 3 SELECTIVITIES OF AQP1,
AQP4, AND AQP5
Uehlein et al. [31] showed that an AQP plays a physiological role by
enhancing CO2 uptake by plants. Molecular dynamics simulations suggested
that CO2 can pass through the 4 aquapores of an AQP1 tetramer, and
especially through the central pore between the 4 monomers [32]. AQP1 was
reported to be permeable to CO2 [28], O2 [33], and NO [34]. It was suggested
that the hydrophilic NH3 probably moves exclusively through the monomeric
aquapores of AQP1 and RhAG, and less hydrophilic CO2 could move through
the hydrophilic central pores of all 5 channels, including AQP4 [35]. The low
NH3 permeability of AQP4 could protect the brain from rising blood levels of
NH3, while still allowing CO2 and perhaps NO and O2, to pass [35]. It must
be noted that AQP1 constituted an independent channel for CO2 because it
conducted CO2 in the absence of other proteins of the RBC membrane and
had almost no association with the Rh membrane complex [36, 37]. In
addition, RhAG conducted ammonia, and NH3 competing with CO2 for
entrance and passage of the same gas channel may thus impede access of CO2
because the affinity of the channel is somewhat greater for NH 3 than for CO2
[29] (Table 2). Thus, lack or deficiency of one or two of these proteins in the
5
Joseph Prandota
host RBC membrane may be associated with various degrees of hypoxia,
which can play, at least in part, an important role in development of the
abovementioned changes in personality profile, traffic accidents frequency,
and weight gain disturbances associated with latent T. gondii infection.
Amelioration of several underlying pathophysiological problems in the
patients with ASD and improvements in autistic symptoms through the use of
hyperbaric oxygen therapy [38] is in line with this reasoning.
Table 2. PCO2 of human red blood cells with various blood
group deficiencies
Bioparameters PCO2 ± SD
[cm/s]
n [N] PCO2 ± SD
(10 µm DIDS)
[cm/s]
n [N]
Control 0.15 ± 0.08 85 [10] 0.05b ± 0.015 34 [9]
Fynull 0.16 ± 0.08 17 [2] 0.05b ± 0.02 11 [3]
JKnull 0.17 ± 0.09 11 [2] 0.05b ± 0.03 10 [2]
McLeod 0.17 ± 0.10 10 [1] 0.06 b ± 0.03 6 [1]
Kellnull 0.16 ± 0.04 4 [1] 0.07 b ± 0.02 3 [1]
Rh positive 0.17 ± 0.11 13 [4]
Rh negative 0.16 ± 0.07 20 [5]
Rhnull 0.066a ±
0.022
31 [4] 0.029 b ±
0.004
10 [3]
AQP-1null 0.065a ±
0.029
37 [2] 0.015 b ±
0.003
12 [2]
The only blood group deficiencies exhibiting a reduced P CO2 are Rhnull and AQP-1null
(last two lines). aIn the case of Rhnull and AQP-1null, values in the absence of 4,4’-
diisothiocyanato-stilbene-2,2’-disulfonate (DIDS) indicate a significant
difference to the normal control PCO2 (P < 0.02). bSignificant difference between
a PCO2 value in the presence of DIDS and the corresponding value in the absence
of DIDS (P < 0.02). n is a number of single determinations, and numbers in
square brackets, N, are the numbers of blood samples from different persons
studied. The finding of identical PCO2 in Rh-negative and positive red blood cells
argues against the suggestion of Callebout et al. [24] that either Rh protein (D or
CcEe) might be a candidate for a CO2 channel.
Endeward et al. [29]; with own modification.
6
Brain hypoxia May be Due to Defects in RhAG Phenotype and AQP4
4. IMPORTANT ROLE OF AQP-4 IN MEDIATION OF
BICARBONATE TRANSPORT IN THE CENTRAL NERVOUS
SYSTEM AND REGULATION OF HYPOXIA
Immunological analyses have shown that AQP4 occurs primarily in brain
astrocytes [39, 40], and in endothelial cells [41], ependymocytes [42], and
retinal Müller cells, although at lower levels than in astrocytes [43]. AQP4, a
transmembrane water channel protein [44] is strongly expressed especially at
sites of fluid transport including the pial and ependymal surfaces in contact
with cerebrospinal fluid, in the subarachnoidal space, and the ventricular
system [45] suggesting a role in movement of water between brain and
cerebrospinal fluid compartments [46-48].
Neuroanatomical studies revealed extensive structural brain
abnormalities in individuals with autism. Fatemi et al. [49] found that
patients with autism had significantly decreased AQP4 expression in the
cerebellum. Moreover, antibody to AQP4 was identified as its target antigen
in neuromyelitis optica, and serum AQP4 levels in these patients correlated
with clinical disease activity, therefore suggesting that they are involved in
the pathogenesis of the disease [50, 51]. Decreased AQP4 expression may
suggest that cell structure, cell volume and ionic homeostasis are
compromised, especially that Nicchia et al. [52] showed that cultured
astrocytes from AQP4 knockdown mice had altered morphology and reduced
osmotic permeability. AQP4 in astrocytes was also implicated in etiology of
seizures and epilepsy, since it plays a role for K+ clearance [53]. AQP4 null
mice demonstrated an increased seizure threshold, but also increased seizure
duration [54, 55]. In addition, AQP4 astrocytic processes in brain and spinal
cord surround blood vessels, neuronal perikarya and processes thus providing
covering of the brain microvessels [56, 57]. A role in water homeostasis
control has been proposed also for the extracellular matrix, which in brain
consists mainly of chondroitin sulfate proteoglycans. In spinal white matter
the overlap between AQP4, and glial fibrillary acidic proteins (GFAP) is
almost complete [56]. All these suggestions are consistent with the
abovementioned reasoning that T. gondii infection affects several cells and
regions in the brain, including astrocytes, epithelial cells, and subependyma,
and may play an important role in development of cryptogenic epilepsy [5,
11, 58]. In this context, it is therefore not surprising that several authors
found elevated serum antibodies and autoantibodies to brain proteins, such as
for example GFAP [59], brain epithelial cells and nuclei [60, 61], chondroitin
7
Joseph Prandota
sulfate [62], and maternal anti-fetal brain antibodies. The generation of all
these antibodies may reflect a defense reaction of the host to the complexes
consisting of the immunodominant epitopes of T. gondii secreted/excreted
protein antigens and the host antigen proteins present in various brain cells
and structures. This suggestion may be supported by the finding of the host
cell filaments interaction with cysts, for example, an astrocyte can contain
large numbers of parasites [63], and in vitro studies staining for the astrocyte-
specific structural protein GFAP showed that GFAP filaments actively form
around the early cyst wall development phase eventually encasing the cyst in
a tight sheath, possibly to stabilize its formation [64, 65]. This eventual
complex of the parasite antigen proteins/host GFAP filament proteins may be
a target for the elevated serum antibodies and autoantibodies directed against
neuronal and glial filament proteins in autistic individuals found by Singh et
al. [59], and also may serve as an explanation for the elevated GFAP levels in
various brain regions of autistic patients [66, 67].
Decreased AQP4 expression in astrocytes and ependymal cells found in
ASD individuals may be associated, at least in part, with development of
various degrees of brain hypoxia because Nagelhus et al. [43] reported that
this aquaporin mediates water flux, bicarbonate transport, and activity-
dependent volume changes in the brain [43]. In addition, Gunnarson et al.
[68] identified a HIF binding motif in the promoter region of AQP4 gene. In
astrocyte cultures, hypoxia was shown to down-regulate mRNA and protein
content for AQP4 and AQP9 [69]. In rats exposed to focal cerebral ischemia
and in human brain with cerebral infarction AQP4 immunoreactivity was
found be increased [68, 70, 71]. On the other hand, in an animal model for
autism, Fatemi et al. [72] found that prenatal human influenza viral infection
in mice resulted in a downregulation in AQP4 mRNA expression in
neocortex in the offspring at birth and persisted at adolescence [73]. One
cannot therefore exclude that T. gondii infection of the central nervous
system may exert a similar effect.
Finally, because AQP4 is the most abundant water channel in the central
nervous system, decreased AQP4 expression in the brain must be associated
with the impaired water movements between various compartments. This
may, at least in part, be responsible for the brain enlargement characteristic
for autistic patients, especially cerebral and cerebellar gray and white
matters, by approximately 5-10% [74]. In addition, it seems that T. gondii
invasion of various brain cells and structures may contribute to the generation
of the antibodies against AQP4 found in patients with neuromyelitis optica
[50, 51]. Relapses of this clinical entity preceded by a rise in serum AQP4
8
Brain hypoxia May be Due to Defects in RhAG Phenotype and AQP4
antibody levels strenghtens the case for the parasite possible involvement in
the pathogenesis of this disease.
Recently Rajnarayanan et al. [75] reported that also AQP11 is expressed
in Purkinje cells in the cerebellum, which are known target cells in ASD
individuals. It was found that AQP11 is the only mammalian aquaporin that
has tri-cysteine motif, a high affinity mercury ion binding site [75]. This
property of AQP11 may, at least in part, explain why thimerosal, an
ethylmercury-containing preservative in some vaccines, has a high affinity to
the brain and therefore may accumulate in this tissue, thus possibly
contributing to the risk of autism and other neurodevelopmental disorders
[76]. This reasoning may be supported by the finding that single-dose
exposure to methylmercury in mice with chronic T. gondii infection showed
a synergistic effect, with effects of methylmercury especially on the immune
system and an increase in brain tissue cyst counts [77]. Thus, one cannot
exclude that latent congenital T. gondii infection of the central nervous
system may be a triggering factor responsible for development of ASD and
other neurologic abnormalities in humans.
5. HYPOXIA MAY TRIGGER REACTIVATION OF LATENT
CEREBRAL TOXOPLASMOSIS BECAUSE IT ACTIVATES
HYPOXIA INDUCING FACTOR-1 ASSOCIATED WITH A
PROINFLAMMATORY ACTIVITY
Cerebral toxoplasmosis has special predilection to the caudate nucleus,
subependyma, periaqueductal area, and include perivascular vasculitis [5]. In
humans, neurons, astrocytes, vascular endothelia, and pericytes have been
implicated in the intracellular proliferation of T. gondii in vivo and in vitro
[78-80]. Lüder et al. [58] found that two days post infection with T. gondii
tachyzoites, intracellular parasites were detected within neurons, astrocytes,
and microglial cells of Wistar rat embryos, and 30% of the microglial cells
harbored intracellular parasites. Tachyzoites have been identified in choroid
plexuses causing cerebral toxoplasmosis in AIDS patients [47, 81], and
invaded Purkinje cells in the cerebellum [7], which may indicate that
selective vulnerability of these cells (loss and atrophy) plays an important
role in the etiopathogenesis of autism [66, 82, 83]. Recent reports suggested
that in adults, chronic T. gondii infection with brain cysts could be the cause
of development of headache and cryptogenic epilepsy [84, 85], high
9
Joseph Prandota
frequency of epilepsy in some groups of autistic patients [86], autism
spectrum disorders, obsessive-compulsive disorder [87], and other
neurodevelopmental abnormalities [11, 88-90].
Individuals with RhAGnull red cells phenotype and an impaired function
of AQP-1 and AQP-4 gas channels may experience various degrees of brain
hypoxia. It must be emphasized that T. gondii activates hypoxia-inducible
factor 1 (HIF1) already at physiologically relevant oxygen levels and requires
HIF1 for growth and survival [91]. Hypoxia is associated with an increase in
generation of various pro- and antiinflammatory cytokines and other
biomediators, such as TNF-α, IL-1-β, IL-6, IL-8, chemokines (monocyte
chemoattractant protein-1, CC-chemokine receptor 2, macrophage
inflammatory protein-1α, intercellular adhesion molecule-1), acute phase
proteins gene expression, COX-2 gene transcription, and heme oxygenase-1
(HO-1) [92-101], induction of iNOS [102], and reactive oxygen species
[103]. Moreover, hypoxia markedly decreased T-lymphocyte IL-2 mRNA, a
key cytokine responsible for B-cell proliferation and immunoglobulin
secretion, and ischemic tissues demonstrated intravascular neutrophil
accumulation, vascular damage, and increased vascular wall permeability
[94]. Exposure of primary rat microglial cultures, as well as established
microglial cell line BV-2 to hypoxia induced expression of iNOS and NO
production, indicating that hypoxia could lead also to the inflammatory
activation of microglia involving activation of HIF-1α [104]. HIF-1α is the
master regulator of oxygen homeostasis to meet cell and tissue requirements
in a situation of oxygen deficiency, and NO impairs normoxic degradation of
this biomolecule by inhibition of prolyl hydroxylases [105]. In addition, NO
induces the synthesis of vascular endothelial growth factor by rat vascular
smooth muscle cells, which brings about proliferation of endothelial cells and
increases permeability of the vessel wall [106]. Some biomarkers gene
expression induced by HIF-α are presented in Table 3.
On the other hand, astrocytes induce heme oxygenase-1 (HO-1
expression in microglia, which has protective antioxidant, antiapoptotic, and
anti-inflammatory functions [95], thus providing mechanism for preventing
excessive brain inflammation [100]. Mimickers of HO-1 products, such as
bilirubin, ferrous iron, and a carbon monoxide-releasing molecule reduced
IFN-γ-induced iNOS expression and/or NO release [100]. Transforming
growth factor-β (Table 1) has also been shown to induce HO-1 expression in
microglia [100], and in epithelial cells [107]. The important role of hypoxia
in the pathogenesis of several diseases may be supported by the reports that
10
Brain hypoxia May be Due to Defects in RhAG Phenotype and AQP4
arterial blood oxygen desaturation played a key role in development of SIDS,
refractory hypoxemia was present in patients with pulmonary right-to-left
shunts, subjects with obstructive sleep-apnea syndrome, decompression
illness, and in patients with migraine associated with patent foramen ovale
and atrial septal defects [108-114]. The inflammatory changes in the brain
associated with hypoxia may thus overlap with chronic neuroinflammation
characteristic for ASD individuals [115, 116], thus enhancing severity of the
clinical course, signs and symptoms of ASD.
Table 3. Hypoxia inducible gene expressiona [113]
Erythropoietin
IL-1, IL-6, IL-8
Nitric oxide synthase-2
Heme oxygenase-1
Ornithine decarboxylase; hexokinase 2
Phosphofructokinase L; phosphoglycerate kinase-1
Pyruvate kinase M; glucose transporter-1, -3
Lactate dehydrogenase A
Glyceraldehyde-3-phosphate dehydrogenase
Insulin-like growth factor-2; enolase 1
Aldolase A, C; adenylate kinase 3
Pituitary adenylate cyclase-activating polypeptide
Transforming growth factor β3
Vascular endothelial growth factor
aT. gondii activates hypoxia-inducible factor 1 (HIF1) already at physiologically
relevant oxygen levels and requires HIF1 for growth and survival [91].
6. THE MACROCEPHALY AND INCREASED BRAIN SIZE
CHARACTERISTIC FOR SOME AUTISTIC INDIVIDUALS
MAY BE CAUSED BY A DECREASED AQP4 EXPRESSION
IN THE CENTRAL NERVOUS SYSTEM
Water movement in brain tissues is carefully regulated with
aquaporinAQP1 and AQP4 are the major water channels expressed at fluid-
tissue barriers throughout the brain and play a crucial role in cerebral water
balance. AQP1 is densely packed in choroid plexus cells lining the ventricles
11
Joseph Prandota
and was found to be responsible for cerebrospinal fluid production. AQP4 is
abundant in astrocytes and concentrated in the end-feet structures that
surround capillaries throughout the brain and are present in glia limiting
structures, notably in osmosensory areas, such as the supraoptic nucleus
[117]. It seems therefore that disturbances in these aquaporins function in the
brain may have serious pathophysiological consequences, including a
suggestion that AQP4 may be a target for therapy in brain edema [117]. It is
important therefore that recently, in the cerebellum of autistic patients Fatemi
et al. [73] demonstrated a markedly decreased AQP4 expression. On the
other hand, one must emphasize that morphometric and chemical
neuroimaging studies performed during early childhood in such individuals
showed abnormal brain volume enlargement [74]. At birth, the average head
circumference was almost normal [118], but by 3-4 years of age, brain size
increased normal range by about 10% [119], with a persistent about 5%
difference at older ages [120]. One may suggest therefore that these
abnormalities may be, at least in part, responsible for the impaired water
transport in the brain of some autistic individuals due to the AQP4
irregularities. This recommendation may be supported by the finding that
AQP4-deficient mice had a significantly increased brain extracellular space
volume fraction by about 28% [121]. Moreover, studies on ontogeny of water
transport in rat brain revealed very low levels of AQP4 in the first postnatal
week, a pronounced increase was noted in the second week (from 2% of
adult level at postnatal day 7 to 25% at postnatal day 14) [122]. Finally,
Fujita et al. [69] postulated that the restoration in AQP4 and AQP5
expression levels in cultured rat astrocytes at 32oC from hypoxia-induced
decrease at 37oC is one of the mechanisms by which brain edema is improved
by mild hypothermia under hypoxic conditions. Thus, the above-presented
argumentation is consistent with the changes of head circumference and brain
size during early postnatal developmental age reported in autistic children
[74].
In summary, it seems that RhAGnull red cells phenotype and an impaired
function of AQP-1 and AQP-4 gas channels associated with various degrees
of brain hypoxia may be, at least in part, responsible for the changes in the
personality profiles in some individuals with latent chronic T. gondii
infection. These abnormalities may therefore also affect clinical course and
intensity of signs and symptoms in the patients with ASD and/or other
neurologic disorders. The amelioration of several underlying
pathophysiological disturbances in autistic individuals and improvements in
clinical symptoms through the use of hyperbaric oxygen therapy is consistent
12
Brain hypoxia May be Due to Defects in RhAG Phenotype and AQP4
with this argumentation [38]. Finally, one may also suggest that the
macrocephaly and increased brain size characteristic for some autistic
individuals was caused by the significantly decreased AQP4 expression in
the brain.
REFERENCES
[1] Dubey, J.P., & Beattie, C.P. Toxoplasmosis of animals and man.
Boca Raton, FL: CRC Press, 1988, p. 220.
[2] Tenter, A.M., Heckeroth, A.R., & Weiss, L.M. Toxoplasma gondii:
from animals to humans. International Journal of Parasitology 2000,
30: 1217-1258.
[3] Boyer, K., Marcinak, J., & McLeod, R. Toxoplasma gondii
(Toxoplasmosis). In Principles and Practice of Pediatric Infectious
Diseases 3rd ed. 2007, Edited by: Long, S., Pickering, L.K., Prober,
C.G. New York: Churchill Livingstone.
[4] Carruthers, V.B. & Suzuki, Y. Effects of Toxoplasma gondii infection
on the brain. Schizophrenia Bulletin 2000, 33: 745-751.
[5] McLeod, R., & Remington, J.S. Toxoplasmosis. In R.E. Behrman,
R.M. Kliegman, W.E. Nelson, & V.C. Vaughan (Eds.), Nelson
textbook of pediatrics 1992, (14th ed., pp. 883-892). Philadelphia: WB
Saunders Co.
[6] Flegr, J. Effects of Toxoplasma on human behavior. Schizophrenia
Bulletin 2007, 33: 757-760.
[7] Costa da Silva, R., & Langoni, H. Toxoplasma gondii: host-parasite
interaction and behavior manipulation. Parasitology Research 2009,
105: 893-898.
[8] Vastava, P.B., Pradhan, S., Jha, S., Prasad, K.N., Kumar, S., & Gupta,
R.K. MRI features of toxoplasma encephalitis in the immunocompetent
host: A report of two cases. Neuroradiology 2002, 44: 834-838.
[9] Lescop, J., Brinquin, L., Schill, H., Soulie, D., Sarrazin, J.L., &
Cordoliani, Y.S. Diffuse toxoplasmic encephalitis in a non-
immunosuppressed patient. Journal of Radiology 1995, 76: 21-24.
[10] Vallochi, A.l., Goldberg, A.C., Falcai, A., Ramasawmy, R., Kalil, J., et
al. Molecular markers of susceptibility to ocular toxoplasmosis, host
and guest badly. Clinical Ophthalmology 2008, 2: 837-848.
[11] Prandota, J. The importance of Toxoplasma gondii infection in diseases
presenting with headaches. Headaches and aseptic meningitis may be
13
Joseph Prandota
manifestations of the Jarisch-Herxheimer reaction. International
Journal of Neuroscience 2009, 119: 2144-2182.
[12] Flegr, J., & Havlicek, J. Changes in the personality profile of young
women with latent toxoplasmosis. Folia Parasitologica 1999, 46: 22-
28.
[13] Flegr, J., Kodym, P., & Talarova, V. Correlation of duration of latent
Toxoplasma gondii infection with personality changes in women.
Biological Psychology 2000, 53: 57-68.
[14] Lafferty, K.D. Can the common brain parasite, Toxoplasma gondii,
influence human culture?. Proceedings, Biological Sciences/ The
Royal Society 2006, 273: 2749-2755.
[15] Flegr, J., Novotna, M., Lindova, J., & Havlicek, J. Neurophysiological
effect of the Rh factor. Protective role of the RhD molecule against
Toxoplasma-induced impairment of reaction time in women. Neuro
Endocrinology Letters 2008, 29: 475-481.
[16] Flegr, J., Klose, J., Novotna, M., Berenreitterova, M., & Havlicek, J.
Increased incidence of traffic accidents in Toxoplasma-infected
military drivers and protective effect RhD molecule revealed by a
large-scale prospective cohort study. BMC Infectious Diseases 2009, 9:
e72.
[17] Flegr, J., Novotna, M., Fialova, A., Kolbekova, P., & Gasova, Z. The
influence of RhD phenotype on toxoplasmosis- and age-associated
changes in personality profile of blood donors. Folia Parasitologica
2010, 57: 143-150.
[18] Novotna, M., Havlicek, J., Smith, A.P., Kolbekova, P., Skallova, A.,
Klose, J., et al. Toxoplasma and reaction time: role of toxoplasmosis in
the origin, preservation and geographical distribution of Rh blood
group polymorphism. Parasitology 2008, 135: 1253-1261.
[19] Kankova, S., Sulc, J., & Flegr, J. Increased pregnancy weight gain in
women with latent toxoplasmosis and RhD-positivity protection
against this effect. Parasitology 2010, Jul 6:1-7 [Epub ahead o print].
[20] Ridgwell, K., Tanner, M.J., & Anstee, D.J. The Rhesus (D) polypeptide
is linked to the human erythrocyte cytoskeleton. FEBS Letters 1984,
174: 7-10.
[21] Ridgwell, K., Spurr, N.K., Laguda, B., MacGeoch, C., Avent, N.D., &
Tanner, M.J. Isolation of cDNA clones for a 50 kDa glycoprotein of
the human erythrocyte membrane associated with Rh (rhesus) blood-
group antigen expression. Biochemical Journal 1992, 287: 223-228.
14
Brain hypoxia May be Due to Defects in RhAG Phenotype and AQP4
[22] Van Kim, C.L., Colin, Y., & Cartron, J.P. Rh proteins: key structural
and functional components of the red cell membrane. Blood Reviews
2006, 20: 93-110.
[23] Kustu, S., & Inwood, W. Biological gas channels for NH3 and CO2:
evidence that Rh (Rhesus) proteins are CO2 channels. Transfusion
Clinique et Biologique 2006, 13: 103-110.
[24] Callebaut, I., Dulin, F., Bertrand, O., Ripoche, P., Mouro, I., Colin, Y.,
et al. Hydrophobic cluster analysis and modeling of the human Rh
protein three-dimensional structures. Transfusion Clinique et
Biologique 2006, 13: 70-84.
[25] Beckmann, R., Smythe, J.S., Anstee, D.J., & Tanner, M.J.
Coexpression of band 3 mutants and Rh polypeptides: differential
effects of band 3 on the expression of the Rh complex containing D
polypeptide and the Rh complex containing CcEe polypeptide. Blood
2001, 97: 2496-2505.
[26] Huang, C.H., & Liu, P.Z. New insights into the Rh superfamily of
genes and proteins in erythroid cells and nonerythroid tissues. Blood
Cells and Molecular Diseases 2001, 27: 90-101.
[27] Walensky, L.D., Narla, M., Lux, IV S.E. Disorders of the red cell
membrane. In: Handin RI, Lux IV SE, Stossel TP (eds). Blood:
Principles and Practice of Hematology, 2nd ed. Philadelphia, PA:
Lippincott Williams & Wilkins, 2003, pp. 1709-1858.
[28] Endeward, V., Musa-Aziz, R., Cooper, G.J., Chen, L., Pelletier, M.F.,
Virkki, V., et al. Evidence that aquaporin 1 is the major pathway for
CO2 transport across the human erythrocyte membrane. FASEB
Journal 2006, 20: 1974-1981.
[29] Endeward, V., Cartron, J.-P., Ripoche, P., & Gros, G. RhAG protein of
the Rhesus complex is a CO2 channel in the human red cell membrane.
FASEB Journal 2008, 22: 64-73.
[30] Blank, M.E., & Ehmke, H. Aquaporin-1 and HCO3- -Cl transporter-
mediated transport of CO2 across the human erythrocyte membrane.
Journal of Physiology 2003, 550: 419-429.
[31] Uehlein, N., Lovisolo, C., Seifritz, F., & Kaldenhoff, R. The tobacco
aquaporin NtAQP1 is a membrane CO2 pore with physiological
functions. Nature 2003, 425: 734-737.
[32] Wang, Y., Cohen, J., Boron, W.F., Schulten, K., & Tajkhorshid, E.
Exploring gas permeability of cellular membranes and membrane
15
Joseph Prandota
channels with molecular dynamics. Journal of Structural Biology 2007,
157: 534-544.
[33] Echevarria, M., Munoz-Cabello, A.M., Sanchez-Silva, R., Toledo-Aral,
J.J., & Lopez-Barneo, J. Development of cytosolic hypoxia and
hypoxia-inducible factor stabilization are facilitated by aquaporin-1
expression. Journal of Biological Chemistry 2007, 282: 30207-30215.
[34] Herrera, M., Hong, N.J., & Garvin, J.L. Aquaporin-1 transports NO
across cell membranes. Hypertension 2006, 48: 157-164.
[35] Musa-Aziz, R., Chen, L.-M., Pelletier, M.F., & Boron, W.F. Relative
CO2/NH3 selectivities of AQP1, AQP4, AQP5, AmtB, and RhAg.
Proceedings of the National Academy of Sciences USA 2009, 106:
5406-5411.
[36] Bruce, L.J., Beckmann, R., Ribeiro, M.L., Peters, L.L., Chasis, J.A.,
Delaunay, J., et al. A band 3-based macrocomplex of integral and
peripheral proteins in the RBC membrane. Blood 2003, 101: 4180-
4188.
[37] Cho, M.R., Knowles, D.W., Smith, B.L., Moulds, J.J., Agre, P.,
Mohanda, N., & Golan, D.E. Membrane dynamics of the water
transport protein aquaprotein-1 in intact human red cells. Biophysical
Journal, 1999, 76: 1136-1144.
[38] Rossignol, D.A. Hyperbaric oxygen therapy might improve certain
pathophysiological findings in autism. Medical Hypotheses 2007, 68:
1208-1227.
[39] Nagelhus, E.A., Veruki, M.L., Torp, R., Haug, F.M., Laake, J.H., et al.
Aquaporin-4 water channel protein in the rat retina and optic nerve:
polarized expression in Muller cells and fibrous astrocytes. Journal of
Neuroscience 1998, 18: 2506-2519.
[40] Zelenina, M. Regulation of brain aquaporins. Neurochemistry
International 2010, doi: 10.1016/j.neuint.2010.03.022.
[41] Amiry-Moghaddam M, Frydenlund DS, Ottersen OP. Anchoring of
aquaporin-4 in brain: molecular mechanisms and implications for the
physiology and pathophysiology of water transport. Neuroscience,
2004, 129: 999-1010.
[42] Yang, B., Ma, T., & Verkman, A.S. cDNA cloning, gene organization,
and chromosomal localization of a human mercurial insensitive water
channel. Evidence for distinct transcriptional units. Journal of
Biological Chemistry 1995, 270: 22907-22913.
16
Brain hypoxia May be Due to Defects in RhAG Phenotype and AQP4
[43] Nagelhus, E.A., Mathiisen, T.M., & Ottersen, O.P. Aquaporin-4 in the
central nervous system: cellular and subcellular distribution and
coexpression with KIR4.1. Neuroscience 2004, 129: 905-913.
[44] Badaut, J., Lasbennes, F., Magistretti, P.J., & Regli, L. Aquaporin in
brain: distribution, physiology, and pathophysiology. Cerebral Blood
Flow and Metabolism 2002, 22: 367-378.
[45] Rash, J.E., Yasumura, T., Hudson, C.S., Agre, P., & Nielsen, S. Direct
immunogold labeling of aquaporin-4 in square arrays of astrocyte and
ependymocyte plasma membranes in rat brain and spinal cord.
Proceedings of the National Academy of Sciences USA 1998, 95:
11981-11986.
[46] Verkman, A.S., Binder, D.K., Bloch, O., Auguste, K., & Papadopoulos,
M.C. Three distinct roles of aquaporin-4 in brain function revealed by
knockout mice. Biochimica et Biophysica Acta 2006, 1758: 1085-1093.
[47] Speake, T., Freeman, L.J., & Brown, P.D. Expression of aquaporin 1
and aquaporin 4 water channels in rat choroid plexus. Biochimica and
Biophysica Acta 2003, 1609: 80-86.
[48] Bloch, O., Auguste, K.I., Manley, G.T., & Verkman, A.S. Accelerated
progression of kaolin-induced hydrocephalus in aquaporin 4-deficient
mice. Journal of Cerebral Blood Flow and Metabolism 2007, 26: 1527-
1537.
[49] Fatemi, H.S., Folsom, T.D., Reutiman, T.J., & Lee, S. Expression of
astrocyte markers aquaporin 4 and connexin 43 is altered in brains of
subjects with autism. Synapse 2008, 62: 501-507.
[50] Jarius, S., Aboul-Enein, F., Waters, P., Kuenz, B., Hauser, A., Berger,
T., et al. Antibody to aquaporin-4 in the long-term course of
neuromyelitis optica. Brain 2008, 131: 3072-3080.
[51] Graber, D.J., Levy, M., Kerr, D., & Wade, W.F. Neuromyelitis optica
pathogenesis and aquaporin 4. Journal of Neuroinflammation 2008, 5:
22.
[52] Nicchia, G.P., Srinivas, M., Lei, W., Brosnan, C.F., Frigeri, A., &
Spray, D.C. New possible roles for aquaporin-4 in astrocytes: cell
skeleton and functional relationship with connexin 43. FASEB Journal
2005, 19: 1674-1676.
[53] Amiry-Moghaddam, M., Williamson, A., Palomba, M., Eid, T., de
Lanerolle, N.C., Nagelhus, E.A., et al. Delayed K+ clearance associated
with aquaporin-4 mislocalization: phenotype defects in brains of alpha-
syntrophin-null mice. Proceedings of the National Academy of
Sciences USA 2003, 100: 13615-13620.
17
Joseph Prandota
[54] Binder, D.K., Oshio, K., Ma, T., Verkman, A.S., & Manley, G.T.
Increased seizure threshold in mice lacking aquaporin-4 water channel.
Neuroreport 2004, 15: 419-429.
[55] Binder, D.K., Yao, X., Verkman, A.S., & Manley, G.T. Increased
seizures duration in mice lacking aquaporin-4 water channels. Acta
Neurochirurgica Suppl 2006, 96: 389-392.
[56] Vitellaro-Zuccarello, L., Mazzetti, S., Bosisio, P., Monti, C., & De
Biasi, S. Distribution of aquaporin 4 in rodent spinal cord: relationship
with astrocyte markers and chondroitin sulfate proteoglycans. Glia
2005, 51: 148-159.
[57] Mathiisen, T.M., Lehre, K.P., & Danbolt, N.C. The perivascular
astroglial sheath provides a complete covering of the brain
microvessels: an electron microscopic 3D reconstruction. Glia 2010,
58: 1094-1103.
[58] Lüder, C.G.K., Giraldo-Velasquez, M., Sendtner, M., & Gross, U.
Toxoplasma gondii in primary rat CNS cells: differential contribution
of neurons, astrocytes, and microglial cells for the intracerebral
development and stage differentiation. Experimental Parasitology
1999, 93: 23-32.
[59] Singh, V.K., Warren, R., Averett, R., & Ghaziuddin, M. Circulating
autoantibodies to neuronal and glial filament proteins in autism.
Pediatric Neurology 1997, 17: 88-90.
[60] Singer, H.S., Morris, C.M., Williams, P.N., Yoon, D.Y., Hong, J.J., &
Zimmerman, A.W. Antibrain antibodies in children with autism and
their unaffected siblings. Journal of Neuroimmunology 2006, 178: 149-
155.
[61] Connolly, A.M., Chez, M.G., Pestronk, A., Arnold, S.T., Mehta, S., &
Deuel, R.K. Serum autoantibodies to brain in Landau-Kleffner variant,
autism, and other neurologic disorders. Journal of Pediatrics 1999,
134: 607-613.
[62] Vojdani, A., Campbell, A.W., Anyanwu, E., Kashanian, A., Bock, K.,
& Vojdani, E. Antibodies to neuron-specific antigens in children with
autism: possible cross-reaction with encephalitogenic proteins from
milk, Chlamydia pneumoniae and Streptococcus group A. Journal of
Neuroimmunology 2002, 129: 168-177.
[63] Fagard R, Van Tan H, Creuzet C, & Pelloux H Differential
development of Toxoplasma gondii in neural cells. Parasitology Today
1999, 15: 504-507.
18
Brain hypoxia May be Due to Defects in RhAG Phenotype and AQP4
[64] Halonen, S.K., Weiss, L.M., & Chiu, F.C. Association of host cell
intermediate filaments with Toxoplasma gondii cysts in murine
astrocytes n vitro. International Journal of Parasitology 1998, 28: 815-
823.
[65] Wilson, E.H., & Hunter, C.A. The role of astrocytes in the
immunopathogenesis of toxoplasmic encephalitis. International
Journal of Parasitology 2004, 34: 543-548.
[66] Fatemi, S.H., Laurence, J., Araghi-Niknam, M., Stary, J.M., & Rizvi,
S. Glial fibrillary acidic protein is elevated in superior frontal and
parietal cortices of autistic subjects. International Journal of
Neuropsychopharmacology 2002, 5: 153-154.
[67] Laurence, J.A., & Fatemi, S.H. Glial fibrillary acidic protein is elevated
in superior frontal, parietal and cerebellar cortices of autistic subjects.
Cerebellum 2005, 4: 206-210.
[68] Gunnarson, E., Zelenina, M., & Aperia, A. Regulation of brain
aquaporins. Neuroscience 2004, 129: 947-955.
[69] Fujita, Y., Yamamoto, N., Sobue, K., Inagaki, M., Ito, H., Arima, H., et
al. Effect of mild hypothermia on the expression of aquaporin family in
cultured rat astrocytes under hypoxic condition. Neuroscience
Research 2003, 47: 437-444.
[70] Aoki, K., Uchihara, T., Tsuchiya, K., Nakamura, A., Ikeda, K., &
Wakayama, Y. Enhanced expression of aquaporin 4 in human brain
with infarction. Acta Neuropathologica (Berl) 2003, 106: 121-124.
[71] Taniguchi, M., Yamashita, T., Kumura, E., Tamatani, M., Kobayashi,
A., Yokawa, T., et al. Induction of aquaporin-4 water channel mRNA
after focal cerebral ischemia in rat. Brain Research, Molecular Brain
Research 2000, 78: 131-137.
[72] Fatemi, S.H., Pearce, D.A., Brooks, A.I., & Sidwell, R.W. Prenatal
viral infection in mouse causes differential expression of genes in
brains of mouse progeny: a potential animal model for schizophrenia
and autism. Synapse 2005, 57: 91-99.
[73] Fatemi, S.H., Folsom, T.D., Reutiman, T.J., & Sidwell, R.W. Viral
regulation of aquaporin 4, connexin 43, microcephalin and nucleolin.
Schizophrenia Research 2008, 98, 163-177.
[74] DiCicco-Bloom, E., Lord, C., Zwaigenbaum, L., Courchesne, E.,
Dager, S.R., et al. The developmental neurobiology of autism spectrum
disorder. Journal of Neuroscience 2006, 26: 6897-6906.
[75] Rajnarayanan, R., Varadharajan, S., Isokpehi, R., & Cohly, H. (2009)
Potential role of cation-aquaporin interaction in autism. Biomedical &
19
Joseph Prandota
Engineering Conference, BSEC 2009, First Annual ORNL. Doi:
10.1109/BSEC.2009.5090494.
[76] Waly, M., Olteanu, H., Banerjee, R., Choi, S.-W., Mason, J.B., Parker,
B.S., et al. Activation of methionine synthase by insulin-like growth
factor-1 and dopamine: A target for neurodevelopmental toxins and
thimerosal. Molecular Psychiatry 2004, 9: 358-370.
[77] King, M.D., Lindsay, D.S., Holladay, S., & Ehrich, M. Neurotoxicity
and immunotoxicity assessment in CBA/J mice with chronic
Toxoplasma gondii infection and single-dose exposure to
methylmercury. Int J Toxicol 2003, 22:53-61.
[78] Ghatak, N.R., & Sawyer, D.R. A morphologic study of opportunistic
cerebral toxoplasmosis. Acta Neuropathologica 1978, 42: 217-221.
[79] Peterson, P.K., Gekker, G., Hu, S., & Chao, C.C. Intracellular survival
and multiplication of Toxoplasma gondii in astrocytes. Journal of
Infectious Diseases 1993, 168: 1472-1478.
[80] Halonen, S.K., Lyman, W.D., & Chiu, F.C. Growth and development
of Toxoplasma gondii in human neurons and astrocytes. Journal of
Neuropathology and Experimental Neurology 1996, 55, 154-1150-
1156.
[81] Falangola, M.F., & Petito, C.K. Choroid plexus infection in cerebral
toxoplasmosis in AIDS patients. Neurology 1993, 43: 2035-2040.
[82] Fatemi, S.H., Halt, A.R., Earle, J., Kist, D.A., Realmuto, G.R., et al.
Reduced Purkinje cell size in autistic cerebellum. Biological
Psychiatry 2000, 47: S128.
[83] Ritvo, E.R., Freeman, B.J., Scheibel, A.B., Duong, T., Robinson, H., et
al. Lower Lower Purkinje cell counts in the cerebella of four autistic
subjects: initial findings of the UCLA-NSAC autopsy research report.
American Journal of Psychiatry 1986, 143: 862-866.
[84] Stommel, E.W., Seguin, R., Thadani, V.M., Schwartzman, J.D.,
Gilbert, K., Ryan, K.A., et al. Cryptogenic epilepsy: an infectious
etiology. Epilepsia 2001, 42: 436-438.
[85] Yazar, S., Arman, F., Yalcin, S., Demirtas, F., Yaman, O., & Sahin, I.
Investigation of probable relationship between Toxoplasma gondii and
cryptogenic epilepsy. Seizure 2003, 12: 107-109.
[86] Hrdlicka, M. EEG abnormalities, epilepsy and regression in autism: a
review. Neuro Endocrinology Letters 2008, 29: 405-409.
[87] Brynska, A., Tomaszewicz-Libudzic, C., & Wolańczyk, T. Obsessive-
compulsive disorder and acquired toxoplasmosis. European Child and
Adolescent Psychiatry, 2001, 10: 200-204.
20
Brain hypoxia May be Due to Defects in RhAG Phenotype and AQP4
[88] Prandota, J. Neuropathological changes and clinical features of autism
spectrum disorder participants are similar to that reported in congenital
and chronic cerebral toxoplasmosis in humans and mice. Research in
Autism Spectrum Disorders 2010b, 4: 103-118.
[89] Prandota, J. Autism spectrum disorders may be due to cerebral
toxoplasmosis associated with chronic neuroinflammation causing
persistent hypercytokinemia that resulted in an increased lipid
peroxidation, oxidative stress, and depressed metabolism of
endogenous and exogenous substances. Research in Autism Spectrum
Disorders 2010c, 4: 119-155.
[90] Prandota, J. Metabolic, immune, epigenetic, and endocrine
abnormalities found in individuals with autism spectrum disorders,
Down syndrome and Alzheimer disease may be caused by congenital
and/or acquired chronic cerebral toxoplasmosis. Research in Autism
Spectrum Disorders 2011, 4: 14-59.
[91] Spear, W., Chan, D., Coppens, I., Johnson, R.S., Giaccia, A., & Blader,
U. The host cell transcription factor hypoxia-inducible factor 1 is
required for Toxoplasma gondii growth and survival at physiological
oxygen levels. Cell Microbiology 2006, 8: 339-352.
[92] Wenger, R.H., Rolfs, A., Marti, H.H., Bauer, C. & Gassmann, M.
Hypoxia, a novel inducer of acute phase gene expression in a human
hepatoma cell line. Journal of Biological Chemistry 1995, 270: 27865-
27870.
[93] Yan, S.F., Tritto, I., Pinsky, D., Liao, H., Huang, J., Fuller, G., et al.
Induction of interleukin 6 (IL-6) by hypoxia in vascular cells. Central
role of the binding site for nuclear factor-IL-6. Journal of Biological
Chemistry 1995, 270: 11463-11471.
[94] Scannel, G. Leukocyte responses to hypoxic/ischemia conditions. New
Horizons 1996, 4: 179-183.
[95] Willis, D., Moore, A.R., Frederick, R., & Willoughby, D.A. Heme
oxygenase: a novel target for the modulation of the inflammatory
response. Natural Medicine 1996, 2: 87-90.
[96] Ji, Y.S., Xu, Q. & Schmedtje, J.F. Jr Hypoxia induces high-mobility-
group protein I(Y) and transcription of the cyclooxygenase-2 gene in
human vascular endothelium. Circulation Research 1998, 83: 295-304.
[97] Zhang, W., Smith, C., Howlett, C. & Stanimirovic, D. Inflammatory
activation of human brain endothelial cells by hypoxic astrocytes in
vitro is mediated by IL-1beta. Journal of Cerebral Blood Flow and
Metabolism 2000, 20: 967-978.
21
Joseph Prandota
[98] Stanimirovic, D., Zhang, W., Howlett, C., Lemieux, P. & Smith, C.
Inflammatory gene transcription in human astrocytes exposed to
hypoxia: roles of the nuclear factor-kappaB and autocrine stimulation.
Journal of Neuroimmunology 2001, 119: 365-376.
[99] Haddad, J.J. & Harb, H.L. Cytokines and the regulation of hypoxia-
inducible factor (HIF)-1α. International Immunopharmacology 2005,
5: 461-483.
[100] Min, K.-J., Yang, M., Kim S.-U., Jou, I., & Joe E. Astrocytes induce
hemeoxygenase-1 expression in microglia: a feasible mechanism for
preventing excessive brain inflammation. Journal of Neuroscience
2006, 26: 1880-1887.
[101] Lam, S.Y., Tipoe, G.L., Liong, E.C. & Fung, M.L. Chronic hypoxia
upregulates the expression and function of proinflammatory cytokines
in the rat carotid body. Histochemistry and Cell Biology 2008, 130:
549-559.
[102] Xu, J., He, L., Ahmed, S.H., Chen, S.W., Goldberg, M.P., Beckman,
J.S. & Hsu, C.Y. Oxygen-glucose deprivation induces inducible nitric
oxide synthase and nitrotyrosine expression in cerebral endothelial
cells. Stroke 2000, 31: 1744-1751.
[103] Clanton, T.L. Hypoxia-induced reactive oxygen species formation in
skeletal muscle. Journal of Applied Physiology 2007, 102: 2379-2388.
[104] Lu, D.Y., Liou, H.C., Tang, C.H., & Fu, W.M. Hypoxia-induced iNOS
expression in microglia is regulated by the Pl3 kinase/Akt/mTOR
signalling pathway and activation of hypoxia inducible factor-1alpha.
Biochemical Pharmacology 2006, 72: 992-1000.
[105] Metzen, E., Zhou, J., Jelkmann, W., Fandrey, J., & Brüne, B. Nitric
oxide impairs normoxic degradation of HIF-1α by inhibition of prolyl
hydrolases. Molecular Biology of the Cell 2003, 14: 3470-3481.
[106] Dulak, J., Józkowicz, A., Dembińska-Kieć, A., Guevara, I.,
Zdzieniecka, A., Zmudzińska-Grochot, D., et al. Nitric oxide induces
the synthesis of vascular endothelial growth factor by rat vascular
smooth muscle cells. Arteriosclerosis and Thrombosis Vascular
Biology 2000, 20: 659-666.
[107] Hill-Kapturczak, N., Truong, L., Thamilselvan, V., Visner, G.A., Nick,
H.S., & Agarwal, A. Smad7-dependent regulation of heme oxygenase-
1 by transforming growth factor-beta in human renal epithelial cells.
Journal of Biology and Chemistry, 2000, 275: 40904-40909.
22
Brain hypoxia May be Due to Defects in RhAG Phenotype and AQP4
[108] Wilmshurst, P.T., Byrne, J.C., & Webb-Peploe, M.M. Relation
between interatrial shunts and decompression sickness in divers.
Lancet 1989, 2: 1302-1306.
[109] Wilmshurst, P.T. & Nightingale, S. Relationship between migraine and
cardiac and pulmonary right-to-left shunts. Clinical Science (London)
2001, 100: 215-220.
[110] Wilmshurst, P.T. Nightingale, S., Walsh, K.P., & Morrison, W.L.
Clopidogrel reduces migraine with aura after transcatheter closure of
persistent foramen ovale and atrial septal defects. Heart 2005, 91:
1173-1175.
[111] Shanoudy, H., Soliman, A., Raggi, P., Liu, J.W., Russell, D.C. &
Jarmukli, N.F. Prevalence of patent foramen ovale and its contribution
to hypoxemia in patients with obstructive sleep apnea. Chest 1998,
113: 91-96.
[112] Godart, F., Rey, C., Prat, A., Vincentelli, A., Chmait, A., Francart, C.
& Porte, H. Atrial right-to-left shunting causing severe hypoxaemia
despite normal right-sided pressures. Report of 11 consecutive cases
corrected by percutaneous closure. European Heart Journal 2000, 21:
483-489.
[113] Prandota, J. Possible pathomechanisms of sudden infant death
syndrome. Key role of chronic hypoxia, infection/inflammation states,
cytokine irregularities, and metabolic trauma in genetically predisposed
infants. American Journal of Therapeutics 2004, 11: 517-546.
[114] Prandota, J. Migraine associated with patent foramen ovale may be
caused by reactivation of cerebral toxoplasmosis triggered by arterial
blood oxygen desaturation. International Journal of Neuroscience
2010a, 120: 81-87.
[115] Vargas, D.L., Nascimbene, C., Krishnan, C., Zimmerman, A.W., &
Pardo, C.A. Neuroglial activation and neuroinflammation in the brain
of patients with autism. Annals of Neurology 2005, 57: 67-81.
[116] Pardo, C.A., Vargas, D.L., & Zimmerman, A.W. Immunity, neuroglia
and neuroinflammation in autism. International Review of Psychiatry
2005, 17: 485-495.
[117] Yool, A.J. Aquaporins: multiple roles in the central nervous system.
Neuroscientist 2007, 13: 470-485.
[118] Courchesne, E., & Pierce, K. Brain overgrowth in autism during a
critical time in development: implications for frontal pyramidal neuron
and interneuron development and connectivity. International Journal
of Developmental Neuroscience 2005, 23: 153-170.
23
Joseph Prandota
[119] Redcay, E., & Courchesne, E. When is the brain enlarged in autism? A
meta-analysis of all brain size reports. Biological Psychiatry 2005, 58:
1-9.
[120] Schultz, R.T., Win, L., Jackowski, A., Klin, A., Staib, L.,
Papademetris, X., et al. Brain morphology in autism spectrum
disorders: an MRI study. Paper presented at annual International
Meeting for Autism Research, 2005, Boston, MA, May.
[121] Yao, X, Hrabetova, S., Nicholson, C., & Manley, G.T. Aquaporin-4-
deficient mice have increased extracellular space without tortuosity
change. Journal of Neuroscience 2008, 28: 5460-2464.
[122] Wen, H., Nagelhus E.A., Amiry-Moghaddam, M., Agre, P., Ottersen,
O.P., & Nielsen, S. Ontogeny of water transport in rat brain: postnatal
expression of the aquaporin-4 water channel. European Journal of
Neuroscience 1999, 11: 935-945.
24
... SPSS v. 21. was used for all statistical tests. ...
... It was also suggested that the complex might participate in intracellular pH regulation [20] and consequently also in the regulation of local oxygen tension. It was suggested that RhD-negativity-associated anoxia in certain parts of the nervous system could be responsible for physiological (and also behavioral) effects of the RhD phenotype [21]. The variation of the oxygen tension in various organs and tissues could, of course, influence also other biological functions, including the functions of the immune system. ...
Article
Full-text available
Rhesus-positive and Rhesus-negative persons differ in the presence-absence of highly immunogenic RhD protein on the erythrocyte membrane. The biological function of the RhD molecule is unknown. Its structure suggests that the molecular complex with RhD protein transports NH3 or CO2 molecules across the erythrocyte cell membrane. Some data indicate that RhD positive and RhD negative subjects differ in their tolerance to certain biological factors, including, Toxoplasma infection, aging and fatique. Present cross sectional study performed on 3,130 subjects) showed that Rhesus negative subjects differed in many indices of their health status, including incidences of many disorders. Rhesus negative subjects reported to have more frequent allergic, digestive, heart, hematological, immunity, mental health, and neurological problems. On the population level, a Rhesus-negativity-associated burden could be compensated for, for example, by the heterozygote advantage, but for Rhesus negative subjects this burden represents a serious problem.
... The regression coefficient was medium (B = 4.124; Eta 2 = 0.070; p = 0.137), and the correlation was not significant. A possible association, if it really exists, is difficult to rationalize, however, latent cerebral toxoplasmosis could influence susceptibility to measles because of changes in the immune status of the children (Table 2) [339,340] caused by the parasite or measlesmumps-rubella (MMR) vaccination. ...
... Interestingly, the strongest association of latent toxoplasmosis and traffic accidents was found for RhD negative drivers [372], while the RhD positive subjects, especially RhD positive heterozygotes seem to be relatively protected against impairment of reaction times [373,374] as well as against traffic accidents [372] (the RhD refers to ''Rhesus factor'' with immunogenic D antigen, while RhAG indicates no Rh antigens on red blood cell membranes). The lack or deficiency of RhAG proteins in the host red blood cell membrane and an impaired function of aquaporin P1 and P4 water/gas channels in the central nervous system could be associated with various degrees of brain hypoxia [339], thus affecting usual driving performance possibly in synergy with the effects of toxoplasmosis on reaction times and ability of long-term concentration [3732375]. Since RhD negative individuals are rare in African and Asian populations [376], an association between traffic accidents and prevalence of toxoplasmosis can be expected mainly in countries inhabited by Caucasians. ...
Article
Full-text available
Background Toxoplasmosis is becoming a global health hazard as it infects 30-50% of the world human population. Clinically, the life-long presence of the parasite in tissues of a majority of infected individuals is usually considered asymptomatic. However, a number of studies show that this 'asymptomatic infection' may also lead to development of other human pathologies.Aims of the studyThe purpose of the study was to collect available geoepidemiological data on seroprevalence of toxoplasmosis and search for its relationship with mortality and disability rates in different countries.Methods and findingsPrevalence data published between 1995-2008 for women in child-bearing age were collected for 88 countries (29 European). The association between prevalence of toxoplasmosis and specific disease burden estimated with age-standardized Disability Adjusted Life Year (DALY) or with mortality, was calculated using General Linear Method with Gross Domestic Product per capita (GDP), geolatitude and humidity as covariates, and also using nonparametric partial Kendall correlation test with GDP as a covariate. The prevalence of toxoplasmosis correlated with specific disease burden in particular countries explaining 23% of variability in disease burden in Europe. The analyses revealed that for example, DALY of 23 of 128 analyzed diseases and disease categories on the WHO list showed correlations (18 positive, 5 negative) with prevalence of toxoplasmosis and another 12 diseases showed positive trends (p
... HIF1α stimulated synthesis and release of proinflammatory cytokines IL-1, IL-6, IL-8, NO, and different growth factors (including TGF-β3) by vascular endothelial cells (Table 40), and intensified subclinical inflammatory reactions [1030] . Nb. impaired function of the CO 2 , AQP1, AQP4 gas channels caused by chronic T. gondii infection may also result in hypoxia and thus enhance neroinflammation in autistic individuals [1031] . These abnormalities may at least in part be responsible for various biochemical disturbances and clinical manifestations observed in patients with CHF and some ADs, such as for example inflammatory bowel disease [120,1032] . ...
Article
Full-text available
T. gondii is globally distributed with a high proportion of the world population estimated to be seropositive, and in the U.S. the parasite is responsible for approximately million infections each year. T. gondii tachyzoites infect almost all nucleated cells and their intracellular multiplication and lifelong persistence in the host cells play an important role in triggering and development of autoimmune diseases (ADs). Latent chronic T. gondii infection may be associated with iron, iodine, and folic acid deficiencies that facilitate development and/or progression of ADs. The oral route is the natural portal of entry for the parasite and gastrointestinal manifestations are frequently reported in patients with ADs. Prolactin was found to bind to tachyzoites and this process impairs their adhesion and penetration into the host cells. Hyperprolactinemia (HPRL) demonstrated in patients with different ADs may therefore reflect host defense against T. gondii infection, and several antipsychotic drugs that induce HPRL also have antitoxoplasmatic activity. Leptin and obesity play an important role in triggering and maintenance of inflammation and autoimmunity. T. gondii infection causes a significant increase in leptin levels and there is a significant positive association between the parasite seropositivity and obesity. Nitric oxide (NO) acts as a proapoptotic as well as an antiapoptotic biomodulator, and have a variety effects on autophagy. Overproduction of NO during T. gondii infection causes dysfunction of both these processes and therefore hinders cleaning service of the apoptotic/autophagic cell-derived antigenic remnants, finally leading to triggering and development of ADs. Damage of the olfactory system associated with chronic latent T. gondii infection may affect olfactory bulb volume and various olfactory functions, being therefore at least in part responsible for the smell impairment in ADs. The potent proinflammatory response of macrophages to infection with T. gondii type II may explain the ability of the strain to cause pathology after oral infection. The parasite also triggers the secretion of antiinflammatory cytokines, such as IL-10, TGF-β, and generation of reactive nitrogen intermediates, thus suppressing the development of the TH1 immune responses and deactivating macrophages. Toxoplasma chronic infection-induced cytotoxic T lymphocyte exhaustion leads to development of ADs because of decreased polyfunctionality, cytotoxic capability, cytokine production, proliferative capacity, and metabolic deficiency. The process of CD4+ and CD8+ T-cell immune exhaustion inhibits the immune response, thus facilitating pathogen persistence. Systemic T. gondii infection triggers a rapid and persistent decrease in the size of naïve CD4+ T lymphocyte pooutput due to destruction of the thymic epithelium. Chronic parasite infections characterized by lower pathogen burden usually restricted to tissues, suggest alternative driving forces in the induction of T cell exhaustion, such as parasite encystations. A significantly lower occurrence of antibodies to persistent viral infections reported in patients with some ADs compared with controls may be due to ol, and a long-term thymic atrophy and suppressed (exhausted) function of host B cells. Both T. gondiiand viral-associated inflammatory processes may be mutually overlapping which lead to worsening or improving clinical course of ADs depending on final temporary or stable proinflammatory/ antiinflammatory cytokine constellations. Dual-affinity T cell receptors may partly be responsible for frequently observed coinfections of T.gondii with some viruses and bacteria. Commonly reported comorbidities in ADs may at least in part be explained by liver damage caused by the pathogen. Vitamin D deficiency is often found in patients with ADs and there is vast evidence that the vitamin has an important beneficial impact on both innate and acquired immunity. Moreover, vitamin D exerts toxoplasmacidal effects and therefore should find a firm place in treatment regimens used in ADs.
... It must be admitted, however, that RhD-negative individuals are subject to stronger negative effects of aging and smoking on health estimated on the basis of the self-rated number of common viral and bacterial diseases in the past year, regardless of Toxoplasma infection status, and that RhD positivity probably protects against a broader spectrum of negative factors than just the Toxoplasma infection (Flegr et al. 2010. A possible physiological mechanism of physiological and behavioural effects of RhD phenotype that was based on RhD negativity-associated anoxia in certain parts of nervous system was suggested by Prandota (2012). ...
Article
Full-text available
Latent toxoplasmosis is probably the most common protistan parasitic disease with many indirect negative impacts on human health. One of the important impacts is impaired psychomotor function leading to reduced driving efficiency in Toxoplasma-seropositive subjects. Numerous case-control studies have established a positive relation between the seroprevalence of Toxoplasma gondii (Nicolle et Manceaux, 1908) and probability of traffic accidents in study populations. The prevalence of toxoplasmosis varies between populations according to local geographical conditions, hygienic practices and kitchen habits. Similarly, we see a striking variation in the incidence of traffic accidents across countries. Hence, we compiled the largest ever data set on the seroprevalence of toxoplasmosis and tried to understand its role in traffic accident-related deaths and disabilities across 87 countries. Simple non-parametric analysis showed a positive and strong relation of T. gondii seroprevalence and traffic accident related disabilities. Further, we conducted multivariate analysis to control for confounding factors. After controlling for wealth, geographical latitude, health of population, length of roads and number of vehicles, the correlation disappeared. When the frequency of RhD negativity and its interaction with toxoplasmosis were included into the model, the effects of toxoplasmosis seemingly returned. However, the analysed data suffered from the problem of multicollinearity. When a proper method of analysis, ridge regression, was applied, the effects of toxoplasmosis prevalence and RhD negativity frequency disappeared again. The existence of a strong correlation between the prevalence of toxoplasmosis and health of population in particular countries, which was the probable cause of multicollinearity and possible reason for the negative result of the present study, suggests that 'asymptomatic' latent toxoplasmosis could have a large impact on public health.
Article
Full-text available
The parasite Toxoplasma gondii influences the behaviour of infected animals and probably also personality of infected humans. Subjects with a Rhesus-positive blood group are protected against certain behavioural effects associated with Toxoplasma infection, including the deterioration of reaction times and personality factor shift. Here, we searched for differences in the toxoplasmosis-associated effects between RhD-positive and RhD-negative subjects by testing 502 soldiers with two personality tests and two intelligence tests. The infected subjects expressed lower levels of all potentially pathognomic factors measured with the N-70 questionnaire and in neurasthenia measured with NEO-PI-R. The RhD-positive, Toxoplasma-infected subjects expressed lower while RhD-negative, Toxoplasma-infected subjects expressed higher intelligence than their Toxoplasma-free peers. The observed Toxoplasma-associated differences were always larger in RhD-negative than in RhD-positive subjects. RhD phenotype plays an important role in the strength and direction of association between latent toxoplasmosis and not only psychomotor performance, but also personality and intelligence.
Article
Full-text available
Toxoplasma gondii, a parasite infecting 20-60% of humans in various countries, influences the behaviour of infected animal and human hosts. Infected human subjects have changes in several of Cattell's and Cloninger's personality factors. Recently, three independent studies have shown that Rh-positive subjects are protected against the T. gondii-induced changes of reaction times and increased risk of traffic accidents. Here we searched for evidence of similar effects of RhD phenotype on toxoplasmosis- or aging-associated changes in the personality profile of about 302 blood donors. We found that Rh-positive and Rh-negative subjects responded differently to toxoplasmosis. In addition to the already known effects of toxoplasmosis on novelty seeking, self transcendence, superego strength and protension, we also found effects of RhD phenotype on ego strength, protension, and praxernia, as well as opposite effects of toxoplasmosis on ego strength, praxernia, ergic tension and cooperativeness in Rh-positive and Rh-negative subjects. Moreover, our results indicate that RhD phenotype might influence not only the effect of toxoplasmosis but also the effect of aging on specific personality traits.
Article
Full-text available
Approximately two billion people worldwide are chronically infected with T. gondii and yet with largely unknown consequences. On the other hand, several authors reported an association between migraine and patent foramen ovale (PFO), and different types of headaches, including migraine, may be precipitated by various diseased states or medications associated with marked immune irregularities, which sometimes cause reactivation of latent cerebral toxoplasmosis (CT). Recently, in a group of 104 subjects with migraine, 46 individuals (44.2%) were found to be seropositive for T. gondii. PFO, atrial septal defects, as well as pulmonary right-to-left shunts are usually associated with a various degree of arterial blood oxygen desaturation. Hypoxia is associated with an increase in the generation of several proinflammatory cytokines and other inflammation mediators, such as TNF-alpha, IL-1-beta, IL-6, IL-8, chemokines (monocyte chemoattractant protein-1, CC-chemokine receptor 2, macrophage inflammatory protein-1alpha, intercellular adhesion molecule-1), acute-phase protein gene expressions, COX-2 gene transcription, induction of iNOS, and reactive oxygen species. Moreover, hypoxia markedly decreased T-lymphocyte IL-2 mRNA, a key cytokine responsible for B-cell proliferation and immunoglobulin secretion, and ischemic tissues demonstrated intravascular neutrophil accumulation, vascular damage, and increased vascular wall permeability. Interestingly, T. gondii activates hypoxia-inducible factor 1 already at physiologically relevant oxygen levels and requires HIF1 for growth and survival. These abnormalities may cause imbalance in the host/T. gondii immune system, which finally results in the reactivation of CT. In addition, hypoxia may participate in paradoxical microembolism because arterial oxygen desaturation enhances expression of plasminogen activator inhibitor-1, an important factor which suppresses fibrinolysis, and this effect may be further amplified by a decreased expression of plasminogen activators, finally causing blood hypercoagulability and paradoxical microembolism. In summary, further studies are required to verify the above-presented pathomechanisms probably responsible for the association between PFO and the development of migraine. It is possible that some migraineurs with PFO may benefit from evaluation and treatment of toxoplasmosis in the future once more information is known.
Article
Full-text available
Worldwide, approximately 2 billion people are chronically infected with T. gondii with largely unknown consequences. This review presents clinical symptoms, differential diagnosis, triggering factors, treatment, and pathomechanisms responsible for idiopathic intracranial hypertension, pseudotumor cerebri, and aseptic meningitis. Literature cited in this work illustrates that immune state and other biologic mediator imbalances due to various endogenous and exogenous triggering factors may markedly affect latent central nervous system T. gondii infection/inflammation intensity, and cause reactivation of cerebral toxoplasmosis (CT). Irregularities in pro- and anti-inflammatory processes may markedly disturb the host and/or T. gondii defense mechanisms important for immune control of the parasite thereby manifesting as a wide range of neurologic symptoms and signs observed in some patients with migraine, epilepsy, celiac disease, Henoch-Schönlein purpura, and other brain disorders. This is consistent with reactivation of CT in mice after treatment with dexamethasone associated with depression of type T(H)1 immune response, and development of CT after administration of etanercept or other bioproducts. It seems that various types of headaches, epilepsy, aseptic meningitis, systemic adverse reactions to drugs or other substances represent the Jarisch-Herxheimer reaction due to apoptosis of T. gondii tachyzoites. Also development of some brain tumors, such as ependymoma and glioma may be associated with a chronic course of CT. Thus, all these patients should be tested for T. gondii infection.
Article
Full-text available
The biological function of RhD protein, a major component of the Rh blood group system, is largely unknown. No phenotypic effect of RhD protein, except its role in hemolytic disease of newborns and protective role against Toxoplasma-induced impairment of reaction times in men, has been described. Here we searched for a protective effect of RhD positivity against Toxoplasma-induced prolongation of reaction times in a set of 110 male and 226 female students of the Faculty of Science tested for latent toxoplasmosis and concentration of testosterone in saliva. RhD-positive subjects have been confirmed to be less sensitive to the influence of latent toxoplasmosis on reaction times than Rh-negative subjects. While a protective role of RhD positivity has been demonstrated previously in four populations of men, the present study has shown a similar effect in 226 female students. Our results have also shown that the concentration of testosterone in saliva strongly influences (reduces) reaction times (especially in men) and therefore, this factor should be controlled in future reaction times studies. The observed effects of RhD phenotype could provide not only a clue to the long-standing evolutionary enigma of the origin of RhD polymorphism in humans (the effect of balancing selection), differences in the RhD+ allele frequencies in geographically distinct populations (resulting from geographic variation in the prevalence of Toxoplasma gondii), but might also be the missing piece in the puzzle of the physiological function of the RhD molecule.
Article
Full-text available
The RhD protein which is the RHD gene product and a major component of the Rh blood group system carries the strongest blood group immunogen, the D-antigen. This antigen is absent in a significant minority of the human population (RhD-negatives) due to RHD deletion or alternation. The origin and persistence of this RhD polymorphism is an old evolutionary enigma. Before the advent of modern medicine, the carriers of the rarer allele (e.g. RhD-negative women in the population of RhD-positives or RhD-positive men in the population of RhD-negatives) were at a disadvantage as some of their children (RhD-positive children born to pre-immunized RhD-negative mothers) were at a higher risk of foetal or newborn death or health impairment from haemolytic disease. Therefore, the RhD-polymorphism should be unstable, unless the disadvantage of carriers of the locally less abundant allele is counterbalanced by, for example, higher viability of the heterozygotes. Here we demonstrated for the first time that among Toxoplasma-free subjects the RhD-negative men had faster reaction times than Rh-positive subjects and showed that heterozygous men with both the RhD plus and RhD minus alleles were protected against prolongation of reaction times caused by infection with the common protozoan parasite Toxoplasma gondii. Our results suggest that the balancing selection favouring heterozygotes could explain the origin and stability of the RhD polymorphism. Moreover, an unequal prevalence of toxoplasmosis in different countries could explain pronounced differences in frequencies of RhD-negative phenotype in geographically distinct populations.
Article
Mechanisms of hypoxia-induced activation of nuclear factor-κB (NF-κB) and inflammatory genes were investigated in fetal human astrocytes in culture. Astrocytes were subjected to interleukin-1β (IL-1β; 50–100 u/ml; 4–24 h), or to a 4-h hypoxia (<2% O2) followed by a 4–24-h reoxygenation. NF-κB binding and transcriptional activity increased up to 10-fold in astrocytes exposed to IL-1β, and up to 3-fold in astrocytes subjected to hypoxia followed by reoxygenation. Both IL-1β- mRNAs and proteins hypoxia-induced NF-κB activation were blocked by the proteasome inhibitor, MG-132. MG-132 inhibited IL-1β-induced up-regulation of IL-1β and IL-8 mRNA and protein but increased hypoxia-stimulated expression/release of IL-1β and IL-8. IL-1 receptor antagonist (IL-1Ra) blocked both hypoxic astrocyte-conditioned media-induced NF-κB activation and the expression/release of IL-1β and IL-8. Astrocytes subjected to hypoxia in the presence of IL-1Ra failed to activate NF-κB, but expressed elevated levels of IL-1β and IL-8. The data suggest that hypoxia/reoxygenation-induced up-regulation of IL-1β and IL-8 in human astrocytes has two components, a NF-κB independent up-regulation during hypoxia, followed by amplification through autocrine IL-1β-induced NF-κB activation during reoxygenation.
Article
Worldwide, approximately 2 billion people are chronically infected with Toxoplasma gondii with largely yet unknown consequences. Patients with autism spectrum disorders (ASD) similarly as mice with chronic toxoplasmosis have persistent neuroinflammation, hypercytokinemia with hypermetabolism associated with enhanced lipid peroxidation, and extreme changes in the weight resulting in obesity or wasting. Data presented in this review suggest that environmental triggering factors such as pregnancy, viral/bacterial infections, vaccinations, medications, and other substances caused reactivation of latent cerebral toxoplasmosis because of changes in intensity of latent central nervous system T. gondii infection/inflammation and finally resulted in development of ASD. Examples of such environmental factors together with their respective biomarker abnormalities are: pregnancy (increased NO, IL-1β, TNF-α, IL-6, IL-10, prolactin; decreased IFN-γ, IL-12), neuroborreliosis (increased IL-1β, sIL-1R2, TNF-α, IFN-γ, IL-6, IL-10, IL-12, IL-18, transforming growth factor-β1 (TGF-β1)), viral infections (increased IL-1β, IL-6, IL-8, TNF-α, IFN-γ/α/β, TGF-β1), thimerosal (increased IL-5, IL-13; decreased IFN-γ, TNF-α, IL-6, IL-12p70, NOS), and valproic acid (increased NO, reactive oxygen species; decreased TNF-α, IL-6, IFN-γ). The imbalances in pro- and antiinflammatory processes could markedly hinder host defense mechanisms important for immune control of the parasite, such as the production of NO, cytokines, and reactive oxygen/nitrogen species, tryptophan degradation by indoleamine 2,3-dioxygenase and/or tryptophan 2,3-dioxygenase, limitation of the availability of intracellular iron to T. gondii, and the mechanisms mediated by an IFN-γ responsive gene family. These fluctuations could result in a recurrent profuse multiplication of T. gondii in the brain associated with persistent neuroinflammation, chronic overproduction of pro- and antiinflammatory cytokines, and NO causing increased oxidative stress, and significantly depressed activity of several enzymes including cytochrome P450 monooxygenase family responsible for metabolism of physiological substrates and xenobiotics, such as steroids, fatty acids, prostaglandins, drugs, pollutants, and carcinogens, finally leading to development of ASD. This reasoning may be supported by such abnormal metabolic events as: (1) patients with ASD have significantly decreased melatonin levels caused by marked deficit in acetylserotonin methyltransferase activity, possibly resulting from maternal and/or fetal/postnatal overproduction of NO, characteristic for this clinical entity; (2) thimerosal inhibited both insulin-like growth factor-1- and dopamine-stimulated methylation reactions, and depressed methionine synthase activity, the metabolic events important for promoting normal neurodevelopment; (3) valproic acid, a strong histone deacetylase inhibitor, have potent anti-T. gondii activity. Thus, patients with ASD should be tested for T. gondii infection.
Article
Toxoplasma gondii is a protozoan parasite that infects about a third of human population. It is generally believed that in immunocompetent hosts, the parasite infection takes usually asymptomatic course and induces self-limiting disease, but in immunocompromised individuals may cause significant morbidity and mortality. T. gondii uses sulfated proteoglycans for host cell invasion and sulfated sugars on the surface of host cells may functions as key parasite receptors. Patients with autism spectrum disorders (ASD) have many inborn or acquired abnormalities of metabolism, including impaired sulfation and sulfoxidation. The impaired sulfation of dehydroepiandrosterone (DHEA) to DHEA-S affected normal development of various brain functions because DHEA-S inhibited vascular neuroinflammation in ASD individuals probably caused by cerebral toxoplasmosis (CT). Treatment of endothelial cells with DHEA-S dramatically inhibited the TNF-α-induced activation of NF-κB, an inflammatory transcription factor, and increased protein levels of the NF-κB inhibitor, IκB-α. A significant decrease in sulfation capacity found during pregnancy compared with post partum probably reflect a defense reaction of the host due to increased production of proinflammatory cytokines associated with frequent and widespread infection with this parasite. This suggestion may be supported by the finding that TNF and IL-1 mediated inhibitory effect of lipopolysaccharide on DHEA sulfotransferase mRNA level in Hep3B human hepatoma cells. It seems however that the impaired sulfation and sulfonation may be also beneficial for the host because lack or a markedly diminished anionic charge of the host cells associated with this event did not promote binding to the negatively charged outer leaflet of T. gondii plasma membranes. Phosphorylation of the parasite and/or host proteins is also of great importance in the process of T. gondii–host cell interaction. Furthermore, the increased male to female ratio characteristic for autistic participants most likely resulted from significantly increased testosterone levels associated with congenital T. gondii infection. It was demonstrated that the parasite, aging and dietary restriction have been able to induce DNA breakage, therefore one may suggest that such an epigenetic mechanism play an important role in development of Down syndrome (DS). Several data may support this notion: (a) autism occurs 10 times more often in children with trisomy 21 than in the general population, (b) the parasite can be transmitted by semen and ovum, (c) autistic children exhibit impaired DNA methylation capacity, and (d) T. gondii affect chromatin structure and may cause dysregulation of the host cell cycle. Alzheimer disease (AD) also may be caused by CT because this abnormality is more prevalent in women, characterizes with a skewed capacity for xenobiotic metabolism especially of compounds containing sulfur that manifest as a decreased plasma levels of DHEA-S, and has marked immune irregularies in part due to aging. Moreover, chronic neuroinflammation characteristic for AD and DS individuals is associated with vascular lesions, patients with AD have increased levels of DNA breaks in the cerebral cortex, markedly enhanced production of proinflammatory cytokines, reactive oxygen species, and lipid peroxidation, disturbances in glucose metabolism, and irregularities in hypothalamic–pituitary axis. It must be noted that similar metabolic and endocrine disturbances have been reported also in humans and mice with chronic toxoplasmosis. Overproduction of IFN-γ and other proinflammatory cytokines associated with persistent neuroinflammation resulted in neurodegeneration and induced amyloid-β production also in DS, as well as accounted for cognitive impairment. Because bradyzoites and sporozoites throughout their life cycle accumulate large amounts of crystalline storage polysaccharide granules analogous to amylopectin within the cytoplasm and are able to build more complex macromolecules, they may be at least in part responsible for the production of amyloid-β senile plaques. Moreover, it seems that the accumulation of iron in senile plaques reflect a defense of the host against T. gondii because this transition metallic ion is necessary for proliferation of tachyzoites. Finally, the beneficial effects of ibuprofen in the patients with AD that restored cellular immunity, decreased production of proinflammatory cytokines, NO, amyloid-β, reduced lipid peroxidation and free radical generation, were consistent with the suggestion that congenital and/or acquired chronic latent CT play an important role in development of these types of neurodegeneration.
Article
Anatomic, histopathologic, and MRI/SPET studies of autistic spectrum disorders (ASD) patients’ brains confirm existence of very early developmental deficits. In congenital and chronic murine toxoplasmosis several cerebral anomalies also have been reported, and worldwide, approximately two billion people are chronically infected with T. gondii with largely yet unknown consequences. The aim of the study was therefore to compare brain abnormalities in ASD patients with those found in mice with cerebral toxoplasmosis (CT) because this may help in understanding pathophysiology of ASD. Data from available published studies were analyzed to compare postmortem pathologic changes found in the brains of ASD patients with those of mice developed after intraperitoneal T. gondii infection. Patients with ASD had the following brain abnormalities: active neuroinflammatory process notably in cerebellum, microglial nodules, accumulation of perivascular macrophages, decreased number and size of Purkinje cells in cerebellar nuclei and inferior olive, hypoperfusion of brain. Mice with congenital toxoplasmosis also had persistent neuroinflammation and ventricular enlargement, periventricular edema, meningeal and perivascular inflammation, and focal loss of Purkinje and granule cells. In murine acquired CT, the brain anomalies included: ventricular dilatation probably reflecting loss of brain parenchyma; perivascular inflammation particularly in hippocampus, and periaqueductal/periventricular areas, Purkinje cell layer markedly disfigured with focal loss of cells; perivascular cuffing by mononuclear cells and localized microglial/inflammatory nodules. Infection of mice with different strains of T. gondii resulted in distinctive neuropathological changes and various stadium of maturity of cysts and the parasite itself, which is in line with the diversity of the autistic phenotypes. Also, the abnormalities in behavior and clinical features associated with autism resembled that reported in chronic latent toxoplasmosis in humans and rodents. All these similarities suggest that T. gondii should be regarded as an important infectious factor that may trigger development of ASD and some other neurodegenerative diseases, such as obsessive–compulsive and attention-deficit/hyperactivity disorders, and cryptogenic epilepsy. Thus, all these patients should be tested for T. gondii infection.
Article
Autism is associated with a high frequency of epileptiform EEG abnormalities (prevalence range 10.3-72.4%) and epilepsy (prevalence range 0-44.5%). A significant subgroup of autistic children (20-49%) experience autistic regression. The relationship among EEG abnormalities, epilepsy, and regression in autistic patients is not yet well understood. In this review, the current knowledge of the relationship is summarized. The evidence from clinical studies does not support the view that EEG abnormalities play a role in autistic regression. The majority of studies also failed to find any significant relationship between epilepsy and autistic regression. However, some results indicated that the higher the prevalence of epilepsy in the sample, the greater the probability of there being a significant association between epilepsy and autistic regression. Further research on the topic is needed.