ArticlePDF Available

Intracellular and Extracellular Redox Status and Free Radical Generation in Primary Immune Cells from Children with Autism

Authors:

Abstract and Figures

The modulation of the redox microenvironment is an important regulator of immune cell activation and proliferation. To investigate immune cell redox status in autism we quantified the intracellular glutathione redox couple (GSH/GSSG) in resting peripheral blood mononuclear cells (PBMCs), activated monocytes and CD4 T cells and the extracellular cysteine/cystine redox couple in the plasma from 43 children with autism and 41 age-matched control children. Resting PBMCs and activated monocytes from children with autism exhibited significantly higher oxidized glutathione (GSSG) and percent oxidized glutathione equivalents and decreased glutathione redox status (GSH/GSSG). In activated CD4 T cells from children with autism, the percent oxidized glutathione equivalents were similarly increased, and GSH and GSH/GSSG were decreased. In the plasma, both glutathione and cysteine redox ratios were decreased in autistic compared to control children. Consistent with decreased intracellular and extracellular redox status, generation of free radicals was significantly elevated in lymphocytes from the autistic children. These data indicate primary immune cells from autistic children have a more oxidized intracellular and extracellular microenvironment and a deficit in glutathione-mediated redox/antioxidant capacity compared to control children. These results suggest that the loss of glutathione redox homeostasis and chronic oxidative stress may contribute to immune dysregulation in autism.
Content may be subject to copyright.
Hindawi Publishing Corporation
Autism Research and Treatment
Volume 2012, Article ID 986519, 10 pages
doi:10.1155/2012/986519
Research Article
Intracellular and Extracellular Redox Status and Free Radical
Generation in Primary Immune Cells from Children with Autism
Shannon Rose, Stepan Melnyk, Timothy A. Trusty, Oleksandra Pavliv,
Lisa Seidel, Jingyun Li, Todd Nick, and S. Jill James
Department of Pediatrics, Arkansas Children’s Hospital Research Institute, University of Arkansas for Medical Sciences,
Little Rock, AR 72202, USA
Correspondence should be addressed to S. Jill James, jamesjill@uams.edu
Received 30 July 2011; Revised 12 August 2011; Accepted 12 September 2011
Academic Editor: Antonio M. Persico
Copyright © 2012 Shannon Rose et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
The modulation of the redox microenvironment is an important regulator of immune cell activation and proliferation. To
investigate immune cell redox status in autism we quantified the intracellular glutathione redox couple (GSH/GSSG) in resting
peripheral blood mononuclear cells (PBMCs), activated monocytes and CD4 T cells and the extracellular cysteine/cystine redox
couple in the plasma from 43 children with autism and 41 age-matched control children. Resting PBMCs and activated monocytes
from children with autism exhibited significantly higher oxidized glutathione (GSSG) and percent oxidized glutathione equivalents
and decreased glutathione redox status (GSH/GSSG). In activated CD4 T cells from children with autism, the percent oxidized
glutathione equivalents were similarly increased, and GSH and GSH/GSSG were decreased. In the plasma, both glutathione
and cysteine redox ratios were decreased in autistic compared to control children. Consistent with decreased intracellular and
extracellular redox status, generation of free radicals was significantly elevated in lymphocytes from the autistic children. These
data indicate primary immune cells from autistic children have a more oxidized intracellular and extracellular microenvironment
and a deficit in glutathione-mediated redox/antioxidant capacity compared to control children. These results suggest that the loss
of glutathione redox homeostasis and chronic oxidative stress may contribute to immune dysregulation in autism.
1. Introduction
Autism is a behaviorally defined neurodevelopmental disor-
der that usually presents in early childhood and is charac-
terized by significant impairments in social interaction and
communication and by abnormal repetitive hyper-focused
behaviors. The prevalence of autism spectrum disorders has
increased more than 10-fold in the last two decades, now
affecting one in 110 US children, yet the etiology of these
disorders remains elusive [1]. Glutathione depletion and oxi-
dative stress have been implicated in the pathology of numer-
ous neurobehavioral disorders including schizophrenia [2],
bipolar disorder [3], and Alzheimer’s disease [4]. Accumu-
lating evidence suggests that redox imbalance and oxidative
stress may also contribute to autism pathophysiology. Mul-
tiple biomarkers of oxidative stress have been identified in
blood samples from children with autism [512]. Our group
has reported a decrease in concentrations of glutathione
(GSH) and several of its metabolic precursors, an increase
in oxidized glutathione disulfide (GSSG), and a decrease in
glutathione redox ratio (GSH/GSSG) in case-control evalua-
tions of plasma and lymphoblastoid cell lines derived from
children with autism [1316]. Recently, several interactive
polymorphisms in enzymes regulating glutathione synthesis
were found to be more prevalent in children with autism
suggesting that the glutathione deficit and predisposition to
oxidative stress may be genetically based in some children
[17].
Oxidative stress occurs when cellular antioxidant defense
mechanisms fail to counterbalance endogenous ROS produc-
tion and/or exogenous prooxidant environmental exposures.
Glutathione (γ-L-glutamyl-L-cysteinylglycine) is a tripeptide
that functions as the major intracellular antioxidant and
redox buer against macromolecular oxidative damage. The
glutathione thiol/disulfide redox couple (GSH/GSSG) is the
predominant mechanism for maintaining the intracellular
2 Autism Research and Treatment
microenvironment in a highly reduced state that is essential
for antioxidant/detoxification capacity, redox enzyme regula-
tion, cell cycle progression, and transcription of antioxidant
response elements (ARE) [1823]. Subtle variation in the
relative concentrations of reduced and oxidized glutathione
provides a dynamic redox signaling mechanism that regu-
lates these vital cellular processes [2427]. For example, in
both CNS precursor cells and na¨
ıve immune cells, intracel-
lular glutathione redox status is the primary determinant
modulating the cellular decision to undergo cell cycle arrest,
differentiation, or proliferation [27]. A reducing intracellular
environment is required for proliferation, while a more
oxidized microenvironment favors cell cycle arrest and dif-
ferentiation. A chronic deficit in the GSH/GSSG redox ratio
is considered to be a reliable indicator of oxidative stress and
increased vulnerability to oxidative damage from prooxidant
environmental exposures [28,29].
In the extracellular plasma compartment, the cysteine/
cystine (thiol/disulfide) redox couple independently provides
the ambient redox environment for circulating immune cells.
The ambient extracellular cysteine/cystine redox potential
has been shown to be more oxidized than the intracellular
GSH/GSSG redox potential and is independently regulated
[30]. Dynamic shifts in the plasma cysteine/cystine redox
potential alter the redox status of cysteine moieties in cell
surface proteins to induce conformational changes in protein
structure that can reversibly alter function [31,32]. For ex-
ample, under oxidizing extracellular conditions, redox-sensi-
tive cysteine residues in the catalytic core of protein tyrosine
phosphatases become oxidized and reversibly inactivate
enzyme activity depending on the ambient cysteine/cystine
redox potential [31,33,34]. Extracellular cysteine/cystine
redox status is emerging as an important new signal trans-
duction mechanism that can induce posttranslational alter-
ations in downstream redox-sensitive proteins including a
variety of enzymes, transcription factors, receptors, adhesion
molecules, and membrane signaling proteins resulting in the
dynamic modulation of their activity and function [32,35,
36].
Recent studies have revealed numerous immunologic
abnormalities among children with autism including alter-
ations in immune cell proportions [3740] and shifts in
helper T-cell subpopulations after mitogenic stimulation
[41,42]. Peripheral blood mononuclear cells (PBMCs) from
individuals with autism have been shown to produce higher
levels of proinflammatory cytokines and abnormal levels of
regulatory cytokines compared to control PBMCs at baseline
and upon mitogenic stimulation [4346]. Taken together,
the immunological studies suggest a role for a dysregulated
immune system in autism that potentially could be related to
a deficit in glutathione-mediated antioxidant capacity and an
oxidized microenvironment in immune cells. To investigate
this possibility, we examined whether primary immune cells
(PBMCs) from children with autism exhibit decreased in-
tracellular glutathione redox capacity compared to PBMCs
from age-matched control children and whether a more
oxidized intracellular and extracellular microenvironment
is associated with increased production of oxidizing intra-
cellular free radicals. Because immune cells from children
with autism have been shown to have abnormal responses
to stimulation, we also elected to challenge the PBMCs with
immune activators known to promote oxidative stress and
measure the resulting intracellular glutathione redox status
in activated isolated monocytes and T cells.
2. Subjects and Methods
2.1. Participants. This investigation was conducted on a
subset of children from the autism IMAGE (Integrated
Metabolic and Genomic Endeavor) study at Arkansas Chil-
dren’s Hospital Research Institute (ACHRI) that has recruit-
ed over 162 case and control families to date. The IMAGE
cohort for this study consisted of 43 children diagnosed
with autistic disorder and 41 unaected control children
(16 of which were unaected siblings). The autism case
families were recruited locally after referral to the University
of Arkansas for Medical Sciences (UAMS), Dennis Devel-
opmental Center and diagnosed by trained developmental
pediatricians. Children aged 3 to 10 with a diagnosis of
autistic disorder as defined by the Diagnostic and Statistical
Manual of Mental Disorders, Fourth Edition (DSM-IV 299.0),
the Autism Diagnostic Observation Schedule (ADOS),
and/or the Childhood Autism Rating Scales (CARS >30)
were enrolled. Children diagnosed with other conditions on
the autism spectrum or rare genetic diseases associated with
symptoms of autism were excluded from the study. Children
with chronic seizure disorders, recent infection, and high-
dose vitamin or mineral supplements exceeding the RDA
were also excluded because these conditions are potential
confounders that could aect redox status. Unaected sib-
lings and unrelated, neurotypical children aged 3 to 10 with
no medical history of behavioral or neurologic abnormalities
by parent report made up the comparison group. The
protocol was approved by the Institutional Review Board at
UAMS, and all parents signed informed consent.
2.2. Materials. Culture flasks, plates, and pipettes were ob-
tained from Corning Life Sciences (Lowell, Mass, USA).
RPMI 1640, penicillin/streptomycin, Dulbecco’s phosphate-
buered saline (PBS), fetal bovine serum (FBS), and glu-
tamine were purchased from Life Technologies (Carlsbad,
Calif, USA). Carboxy-H2DCFDA (6-carboxy-2,7-dichlo-
rodihydrofluorescein diacetate, diacetoxymethyl ester) was
obtained from Molecular Probes (Carlsbad, Calif, USA).
Human Monocyte Isolation Kit II and Human CD4 T
cell Isolation Kit II were purchased from Miltenyi Biotec
(Bergisch-Gladbach, Germany). Histopaque-1077 and all
other chemicals were obtained from Sigma-Aldrich (St.
Louis, Mo, USA).
2.3. Isolation of PBMCs and Stimulation of Monocytes and
CD4 T Cells. Fasting blood samples (20 mL) were collected
before 9:00 AM into EDTA-Vacutainer tubes and immedi-
ately chilled on ice before centrifuging at 1300 ×gfor10min
at 4C. Aliquots of plasma were stored at 80C in cryostat
tubes until extraction and HPLC quantification. PBMCs
were isolated by centrifugation over Histopaque-1077. Red
Autism Research and Treatment 3
blood cells were lysed using a brief (15 s) incubation with
1 mL ice-cold water. Approximately, 30 ×106PBMCs were
resuspended in RPMI 1640 medium (supplemented with
10% FBS, 1% penicillin/streptomycin, and 2 mM glutamine)
at a density of 106cells/mL. Note that because we were unable
to obtain 20 mL blood volume from every child, it was not
possible to isolate and analyze monocytes and CD4 T cells
for all participants. For monocyte stimulation, PBMCs were
treated with 0.1 μg/mL lipopolysaccharide (LPS); for T-cell
stimulation, PBMCs were treated with 10 ng/mL phorbol 12-
myristate 13-acetate (PMA) and 1 μg/mL ionomycin. Cells
were placed in a humidified 5% CO2incubator at 37C
for 4 hr. Stimulated monocytes and CD4 T cells were then
isolated by negative selection using magnetic cell labeling as
described by the manufacturer (Miltenyi Biotec, Bergisch-
Gladbach, Germany). Using flow cytometry, we determined
that 75% of isolated monocytes are positive for CD14 and
that 87% of isolated CD4 T cells are positive for CD4. For
HPLC quantification of GSH and GSSG, approximately 2 ×
106unstimulated (resting) PBMCs, stimulated monocytes,
or stimulated CD4 T cells were pelleted, snap frozen on dry
ice,andstoredat80C.
2.4. Cell Extraction and HPLC Quantification of Intracellular
Glutathione and Plasma Cysteine Redox Status. The storage
interval at 80C before extraction was consistently between
1-2 weeks after blood draw and cell isolation to minimize
potential metabolite interconversion. The methodological
details for intracellular and extracellular GSH extraction
and HPLC elution and electrochemical detection have been
described previously [15,16], and metabolite detection does
not require derivatization. Although most GSSG is present
as a mixed disulfide with other thiols including cysteine,
our measurements detect only the free GSSG in plasma.
Glutathione and cysteine concentrations were calculated
from peak areas of standard calibration curves using HPLC
software. Intracellular results are expressed as nanomoles
per milligram of protein using the BCA Protein Assay Kit
(Pierce, Rockford, Ill, USA), and plasma results are expressed
as micromoles per liter.
2.5. Measurement of Intracellular Free Radicals. Carboxy-
H2DCFDA (DCF) is a membrane-permeable ROS/RNS-sen-
sitive probe that remains nonfluorescent until oxidized by
intracellular free radicals. The intensity of DCF fluorescence
is directly proportional to the level of free radical oxidation.
Approximately, 106PBMCs were resuspended in 1 mL RPMI
1640 medium supplemented with 10% FBS, 1% penicil-
lin/streptomycin, and 2 mM glutamine and stained in the
dark for 20 min with 1 μMDCFat37
C. Stained cells were
washed and resuspended in PBS and analyzed immediately
onaPartecCyFlowflowcytometer(G
¨
orlitz, Germany) using
488 nm excitation wavelength with 530/30 nm (FL1) emis-
sion filter. For each analysis, the fluorescence properties of
10000 cells were collected, and the data were analyzed using
the FCS Express software (De Novo Software, Los Angeles,
Calif, USA). Intracellular free radical levels are expressed as
median fluorescence intensity (MFI) of subject sample DCF
Tab le 1: Demographics of study population.
Case children
n=43
Control children
n=41
Age; mean (SD) 5.42 (1.98) 6.16 (2.29)
Male; n(%) 36 (84) 20 (49)
White; n(%) 38 (88.4) 31 (75.6)
Asian; n(%) 2 (4.65) 0 (0)
African American; n(%) 2 (4.65) 8 (19.5)
Hispanic; n(%) 1 (2.3) 2 (4.9)
OTC multivitamin use; n(%) 17 (39.5) 8 (19.5)
fluorescence normalized to DCF fluorescence of a standard
PBMC preparation. As an internal control, the standard
PBMC preparation was isolated from a 100 mL blood sample
from an unaected healthy adult volunteer, aliquoted and
frozen at 180C in 90% FBS/10% DMSO. An aliquot of
the standard PBMC preparation was stained and analyzed
with each subject sample. Evaluation of oxidizing free radical
production was possible only in those case and unrelated
control samples for which sucient (20 mL) blood volume
was obtained.
2.6. Statistical Analysis. Within the control group, 16 of the
41 unaected control children were case siblings. There were
27 additional case children without a sibling and 25 addi-
tional unrelated control children comprising the total case-
control cohort of 84 children. To down-weight the impact of
outliers, three metabolites observations were curtailed at the
extremes of the distributions for PBMC GSH, PBMC GSSG,
and Monocytes GSH/GSSG (see footnote in Table 2). The
sibling data are correlated resulting in a combined sample
of correlated and uncorrelated data; thus, the assumption of
all data being independent is not satisfied for the standard
two-sample t-test. To make use of all data from dependent
and independent observations, we used the corrected Z-test
proposed by Looney and Jones [47]. This statistical approach
provides adequate control of Type 1 errors and has more
power than a standard Student’s t-test. Because the DCF data
compared cases and unrelated controls (without siblings)
the standard Student’s t-test was used with significance set
at 0.05. Nonparametric intercorrelations (Spearman correla-
tion coecients) between age and gender and the 7 outcome
variables, GSH, GSSG, GSH/GSSG, % oxidized glutathione,
cysteine, cystine, and cysteine/cystine were determined with
the significance level set at 0.05. Data was analyzed using SAS
9.2 software (SAS Institute Inc, Cary, NC, USA).
3. Results
3.1. Demographics of Study Population. Ta b l e 1 lists the
demographics of the study population. The only major
dierences between cases and controls are that the control
group was composed of a greater proportion of females and
African Americans, whereas the case group had a greater pro-
portion of Asian subjects. Over-the-counter multivitamin
supplement use was higher among cases (39.5%) compared
4 Autism Research and Treatment
Tab le 2: Intracellular glutathione redox status in resting PBMCs and activated monocytes and CD4 T cells.
Metabolite Case children Control children Corrected Z-test
nMean ±SD nMean ±SD Dierence
(95% CI) Pvalue
Resting PBMCs
GSH (nmol/mg protein) 43 25.45 ±8.16 41 23.35 ±6.38 2.09 (1.09, 5.29) 0.19
GSSG (nmol/mg protein) 43 0.90 ±0.3410.66 ±0.23 0.24 (0.13, 0.35) <0.001
GSH/GSSG 43 29.58 ±9.04 41 37.58 ±10.89 7.99 (12.51, 3.48) <0.001
Oxidized GSH (%) 43 0.07 ±0.02 41 0.05 ±0.01 0.02 (0.0075, 0.024) <0.001
Activated monocytes
GSH (nmol/mg protein) 18 7.73 ±3.16 20 8.55 ±2.50.82 (2.02, 0.38) 0.18
GSSG (nmol/mg protein) 18 0.62 ±0.24 20 0.47 ±0.17 0.14 (0.03, 0.25) 0.01
GSH/GSSG 18 13.31 ±7.26 20 19.30 ±6.35 5.98 (9.99, 1.97) 0.003
Oxidized GSH (%) 18 0.14 ±0.05 20 0.10 ±0.03 0.04 (0.02, 0.07) <0.001
Activated CD4 T cells
GSH (nmol/mg protein) 18 6.82 ±3.01910.16 ±3.74 3.33 (5.24, 1.42) <0.001
GSSG (nmol/mg protein) 18 0.68 ±0.29 19 0.63 ±0.24 0.05 (0.11, 0.22) 0.51
GSH/GSSG 18 10.47 ±4.19 19 17.49 ±6.95 7.02 (10.17, 3.87) <0.001
Oxidized GSH (%) 18 0.17 ±0.05 19 0.11 ±0.05 0.05 (0.03, 0.08) <0.001
GSH: glutathione; GSSG: oxidized glutathione disulfide; oxidized GSH: (%)2GSSG/(GSH+2GSSG); curtailment: PBMC GSH >45 set =45 (n=1); PBMC
GSSG >1.75 set =1.75 (n=1); Monocytes GSH/GSSG >35 set =35 (n=1).
to controls (19.5%); however, the glutathione redox status
was statistically unaected by vitamin use (data not shown).
3.2. Decreased Intracellular Glutathione Redox Status in
Autism. Table 2 presents the relative intracellular concen-
trations of GSH, GSSG, the glutathione redox ratio, and
the percentage of oxidized glutathione equivalents in resting
(unstim-ulated) PBMCs and in isolated stimulated mon-
ocytes and CD4 T cells from children with autism and
age-matched control children. The percent oxidized gluta-
thione is expressed in absolute glutathione equivalents as
2GSSG/(GSH+2GSSG). Relative to controls, the intracellular
concentration of GSSG and the percent oxidized glutathione
were significantly increased (40%), and the GSH/GSSG
ratio decreased (21%) in PBMCs from children with
autism (P<0.001). After stimulation with LPS, mono-
cytes from children with autism also exhibited significantly
decreased GSH/GSSG (31%, P=0.003), increased GSSG
concentration (32%, P=0.01), and 40% higher percent
oxidized glutathione (P<0.001). In mitogen-stimulated
CD4 T cells from children with autism, the intracellular GSH
concentration was 33% lower, the GSH/GSSG was 40%
lower (P<0.001), and the percent oxidized glutathione
was 55% higher than in stimulated CD4 T cells from
control children (<0.001). As expected, activation with LPS
and PMA both resulted in decreased intracellular GSH
levels and GSH/GSSG in isolated monocytes and CD4 T
cells compared to resting (unstimulated) PBMCs. Upon
stimulation, there was a greater decrease in intracellular
GSH and GSH/GSSG in both CD4 T cells and monocytes
from children with autism compared to control children.
Neither age nor gender was significantly correlated with any
of the outcome measures. The protein content per 106cells
did not dier between case and control children (data not
shown).
3.3. Decreased Extracellular Glutathione and Cysteine Redox
Status in Autism. Ta b l e 3 presents the relative concentrations
of GSH, GSSG, GSH/GSSG, % oxidized GSH, cysteine,
cystine, and the cysteine/cystine redox ratio in the extracel-
lular plasma compartment. Children with autism exhibited
a significantly decreased extracellular concentration of GSH
(21%) and GSH/GSSG (54%) and increased concentra-
tion of GSSG and the percent oxidized glutathione (52%
and 82%, resp., P<0.001). Figures 1(a) and 1(b) com-
pare GSH/GSSG and % oxidized glutathione equivalents,
respectively, in plasma, T cells, and monocytes from case and
control children and graphically demonstrates the consistent
decrease in both extracellular and intracellular glutathione
redox status among the case children.
The dynamic balance between the reduced and oxidized
forms of glutathione can also be expressed as the redox
potential or reducing power of the GSH/GSSG redox couple
(Eh) and can be calculated from the Nernst equation, Eh=
E0+RT/nF ln[disulfide]/([thiol 1] [thiol 2]), where E0
is the standard potential for the glutathione redox couple
(264 mV), Ris the gas constant (8.314 J/Kmol), Tis the
absolute temperature of analytical measurement (25C=
298K), nis 2 for the number of electrons transferred,
and Fis Faraday’s constant (96,485 coulomb/mol) [48].
The calculated Ehvalue for the GSH pool in the children
with autism is 116 mV, which is 12 mV more oxidized
than in the control children, with an Ehvalue of 128 mV
(Table 3).
Autism Research and Treatment 5
Tab le 3: Extracellular (plasma) glutathione and cysteine redox status.
Metabolite Case children Control children Corrected Z-test
nMean ±SD nMean ±SD Dierence
(95% CI) Pvalue
Plasma
GSH (μM) 38 1.58 ±0.23 41 1.99 ±0.22 0.41 (0.50, 0.31) <0.001
GSSG (μM) 38 0.20 ±0.06 41 0.13 ±0.04 0.07 (0.05, 0.09) <0.001
GSH/GSSG 38 8.24 ±2.20 41 17.14 ±5.54 8.73 (10.52, 6.94) <0.001
Oxidized GSH (%) 38 0.20 ±0.05 41 0.11 ±0.03 0.09 (0.07, 0.10) <0.001
Ehfor GSH 116 mV 128 mV
Cysteine (μM) 41 21.7±4.88 41 21.43 ±4.08 0.13 (1.88, 2.14) 0.90
Cystine (μM) 41 29.2±10.641 19.26 ±4.89.73 (6.25, 13.2) <0.001
Cysteine/Cystine 41 0.79 ±0.18 41 1.14 ±0.18 0.33 (0.41, 0.26) <0.001
Ehfor Cysteine 106 mV 111 mV
GSH: glutathione; GSSG: oxidized glutathione disulfide; Eh: steady-state redox potential; Ehfor GSH: 264 mV +(30 mV) log([GSSG]/[GSH]2); Ehfor
cysteine: 250 mV + (30 mV) log([CySSCy]/[Cys]2).
0
5
10
15
20
25
30
GSH/GSSG
Control
Case
Plasma Activated monocytes Activated T cells
(a)
Control
Case
Plasma Activated monocytes Activated T cells
0
0.05
0.1
0.15
0.2
0.25
0.3
Oxidized GSH (%)
(b)
Figure 1: Intracellular and extracellular glutathione redox imbalance in autism. (a) presents the GSH/GSSG in plasma, isolated activated
monocytes, and CD4 T cells from case and control children; (b) presents the % oxidized glutathione equivalents. Both extracellular and
intracellular glutathione redox status are consistently significantly decreased among the case children (P<0.01).
The concentration of cystine, the oxidized form of
cysteine, was significantly elevated (52%), while the cys-
teine/cystine redox ratio was significantly decreased (31%)
in plasma from children with autism (P<0.001). The Eh
value for the cysteine pool can also be calculated from the
Nernst equation (see above) where the E0for cysteine is equal
to 250 mV [30]. The calculated Ehvalue for the cysteine
pool in children with autism is 106 mV, or 5 mV more
oxidized than the control Ehvalue of 111 mV (Table 3).
3.4. Elevated Free Radical Production in Autism. The level of
intracellular free radicals was measured in available resting
PBMCs from children with autism (n=15) and unaected
control children (n=16) using DCF, an ROS/RNS-sensitive
fluorescent probe. Monocytes and lymphocytes were gated
based on light scatter properties (size and density) and ana-
lyzed separately. Figure 2 presents the median fluorescence
intensity (MFI) of lymphocytes from children with autism
and unaected control children (normalized to MFI of the
standard PBMC preparation). Gated lymphocytes from chil-
dren with autism exhibited a significantly higher mean level
of intracellular free radicals compared to lymphocytes from
control children (P<0.05). No dierences in free radical
production were observed in gated monocytes from case and
control children. Intracellular free radical production was
not correlated with age or gender in this cohort.
4. Discussion
Oxidative stress is generally defined as an imbalance between
oxidant production and endogenous antioxidant defense
mechanisms and can be clinically defined in humans by a
decrease in the redox status of GSH/GSSG and cysteine/cys-
tine thiol/disulfide redox couples [49].Therelativeequi-
librium between reduced and oxidized sulfhydryl groups
defines the ambient redox state. Low glutathione redox status
6 Autism Research and Treatment
has been associated with the pathophysiology of several neu-
robehavioral disorders including schizophrenia [2,50], bipo-
lar disorder [3], alcoholism [51], HIV [52], and Alzheimer’s
disease [53]. This is the first study to evaluate intracellular
glutathione-mediated antioxidant/redox capacity in primary
cells from children with autism as well as the extracellular
plasma cysteine/cystine redox status. Because these two redox
systems are compartmentalized and independently regulat-
ed, evaluation of both redox couples provides a complete
picture of the primary immune cell microenvironment in
children with autism. Supporting and extending our previ-
ous findings of decreased plasma and lymphoblastoid cell
GSH/GSSG, we now report that both primary immune cell
GSH/GSSG and plasma cysteine/cystine redox couples are
similarly compromised resulting in a more oxidized immune
cell microenvironment in children with autism compared to
control children.
Recent evidence supports the notion that subtle fluctu-
ations in ambient redox status may provide an important
regulatory mechanism that can dynamically modulate im-
mune cell function. Activation and proliferation of T cells
require a reducing intracellular microenvironment, whereas
a more oxidized environment promotes cell cycle arrest and
blunted responsiveness to immune stimulation [5457]. For
example, a mechanism involving extracellular redox mod-
ulation by regulatory T cells (Tregs) was recently elucidated
by Yan et al. [35]. Tregs were shown to inhibit the release of
cysteine into the immune synapse between dendritic cells
and na¨
ıve T cells, which eectively reduces GSH levels in T
cells by eliminating the rate-limiting amino acid for GSH
synthesis. A high ratio of reduced to oxidized glutathione is
required for cell cycle progression from G1 to S phase and
induction of the T-cell proliferative response [55]. Thus, the
more oxidized GSH/GSSG redox state of the intracellular
glutathione pool in PBMCs and in activated CD4 T cells
observed in children with autism (Table 2) would suggest
a hyporesponsive phenotype that is less conducive to T-cell
activation and proliferation. Consistent with this hypothesis,
several recent studies have documented abnormalities in
the adaptive immune response in children with autism
[44,58].
A glutathione deficit in T cells has been shown to nega-
tively aect the adaptive immune response and T-cell pro-
liferation by reducing IL-2 receptor turnover and IL-2-
dependent DNA synthesis [59,60]. In monocytes, an oxi-
dized intracellular environment has been shown to alter the
cytokine profile and skew the Th1 and Th2 balance [61,62].
Studies in mice have demonstrated that the intracellular GSH
content of antigen presenting cells (APCs) reversibly alters
the Th1 and Th2 cytokine response pattern [61]. Specifically,
a GSH deficit reduced Th1-associated IFN-γproduction and
exaggerated Th2-associated IL-4 production. Restoration of
GSH restored the Th1 cytokine response and normalized
the Th2 response. Consistent with these observations, two
independent studies have reported that helper T-cell sub-
populations in PBMCs from children with autism are shifted
towards T helper 2 (Th2) dominance [41,42]. Further, a
decrease in T-cell IL-2 receptor expression has been reported
Intracellular free radicals
(normalized to standard)
Control Case
1.5
1
0.5
0
Figure 2: Intracellular Free Radicals are Elevated in Lymphocytes
from Children with Autism. Intracellular free radicals were mea-
sured in freshly isolated PBMC from children with autism and
unaected control children using 1uM DCF. Presented is median
fluorescent intensity (MFI) of the gated lymphocyte population
from subject samples normalized to MFI of a standard PBMC
preparation also treated with 1 uM DCF and analyzed with each
subject sample. Lymphocytes from children with autism exhibited
a significantly higher mean level of intracellular free radicals than
controls (P=0.04). Control median (95% CI) =0.576 (0.551–
0.640); case median (95% CI) =0.689 (0.561–1.086).
to be associated with decreased proliferative response after
mitogen stimulation in children with autism [58].
The more oxidized GSH/GSSG redox status in plasma
and primary immune cells in children with autism (Figure 1)
may oer a mechanistic explanation for the abnormal adap-
tive immune response previously reported in these children.
When intracellular oxidative stress exceeds glutathione redox
capacity, cells export GSSG into the plasma as a mechanism
to restore internal redox homeostasis [49,63]. The elevated
GSSG concentrations in PBMCs (Table 2) suggest that
the GSSG export mechanism and intracellular antioxidant
capacity were not sucient to maintain intracellular redox
homeostasis and that redox imbalance was chronic in these
children. The association between a more oxidized immune
cell microenvironment and an abnormal adaptive immune
response warrants continued investigation especially in light
of the potential reversibility of immune dysfunction with
targeted treatment to restore redox homeostasis [15].
The calculated Ehvalues for the extracellular GSH and
cysteine pools (Table 3) in our control population dier
somewhat from previously published values. In adults, the
plasma glutathione Ehis more reduced at around 137 mV,
and the plasma cysteine redox couple is more oxidized at
80 mV [30,48]. These discrepancies may reflect method-
ological dierences in sample preparation in that our electro-
chemical detection does not require derivatization for detec-
tion. It is also possible that children (age 3–10 years) may
have less reducing capacity than previously reported in adults
(age 25–35 years) [48]. Nonetheless, our calculated Ehvalues
are consistent with previous reports that plasma cysteine Eh
(111 mV) is more oxidized than that of GSH (128 mv).
Autism Research and Treatment 7
Mean intracellular free radical production was higher in
primary lymphocytes from children with autism relative to
lymphocytes from age-matched control children (Figure 2)
and was driven by a subset of 5 (33.3%) children whose
lymphocytes exhibited especially high levels of free radicals.
Mitochondria are the primary producers and targets of
intracellular free radicals, and mitochondrial dysfunction has
been postulated to be a contributing factor in the pathogen-
esis of autism and numerous other neurological disorders
[6467]. In a lymphoblastoid cell model, we previously dem-
onstrated that the GSH/GSSG redox ratio in mitochondria
was significantly lower in autism compared to control cells
and was associated with a significantly lower mitochondrial
membrane potential after nitrosative stress [16]. It is well
established that mitochondria are highly concentrated in
presynaptic terminals and that loss of redox control can neg-
atively aect the eciency of neurotransmission and synap-
tic plasticity [68,69]. Similarly, mitochondrial localization
and redox signaling at the immunological synapse between
lymphocytes and antigen presenting cells are required for
immune activation, and excessive ROS can interrupt these
signaling pathways [7072]. A recent study of mitochondrial
defects in lymphocytes from children with autism found
decreased complex I activity and overreplication of and de-
letions in mitochondrial DNA compared to control lym-
phocytes [73]. Based on this evidence, it is plausible to hy-
pothesize that mitochondria may be the source of the
increased levels of lymphocyte free radicals observed in the
subset of autistic children presented in Figure 2. Consistent
with this hypothesis, a recent meta-analysis estimated that
mitochondria dysfunction may aect up to 30% of children
with autism [64]. Based on this evidence, further study of
mitochondrial function and redox status in lymphocytes
from children with autism is warranted.
Relevant to our observations, two recent papers have
revealed that an oxidized extracellular cysteine/cystine redox
status can initiate a redox signaling cascade that stimulates
intracellular mitochondrial ROS production as a mechanism
to initiate an inflammatory immune response [74,75]. The
signal transduction from the extracellular to intracellular
compartments occurs through oxidative modification of
redox-reactive cysteines on cell surface proteins. Exposed
cysteine sulfhydryl groups on proteins can be reversibly oxi-
dized to sulfenic acid or disulfide bonds resulting in altered
protein structure and function that initiate downstream
redox signaling cascades [33,76,77]. In an elegant series
of experiments, Imhoand Hansen demonstrated that mi-
tochondrial ROS production was significantly increased in
cells incubated under extracellular oxidized cysteine/cystine
redox conditions [74]. The stimulated intracellular ROS
production resulted in the expression of Nrf-2, the transcrip-
tion factor responsible for initiation of the inflammatory
response. Treatment to block the availability of cell surface
cysteine thiol groups abrogated mitochondrial ROS produc-
tion and Nrf-2 expression. Go et al. confirmed and extended
these observations by demonstrating that treatment to main-
tain mitochondrial redox status abrogated ROS production
in the presence of oxidized extracellular cysteine/cystine
[75]. Although the precise mechanism for the oxidative
cysteine/cystine-dependent signaling for mitochondrial ROS
production is not yet clear; the authors provide evidence of
a possible link to changes in the redox state of cytoskeletal
proteins that could be functionally linked to the mitochon-
drial membrane. Other studies have demonstrated that an
oxidized plasma cysteine/cystine redox potential is associated
with proinflammatory conditions [78,79]andcanbemod-
ulated by diet [80,81]. These observations support the pos-
sibility that the oxidized plasma cysteine/cystine in children
with autism may be functionally related to the increase in
lymphocyte free radical production observed and contribute
to immune cell abnormalities in these children.
In summary, we show for the first time that both the
extracellular and intracellular immune cell compartments
are more oxidized in children with autism compared to age-
matched unaected control children. Randomized clinical
trials will be needed to determine whether treatment to
normalize plasma and intracellular redox status will improve
immune cell function and possibly the health and behavior
in children with autism.
Conflict of Interests
The authors declare no conflict of interests.
Acknowledgments
The authors would like to express their gratitude to the
families in Arkansas aected by autism whose participa-
tion made this study possible. They also acknowledge the
invaluable help of the nurses and clinicians at the Dennis
Developmental Center for referral and evaluation. This
research was supported, in part, with funding from the
National Institute of Child Health and Development (RO1
HD051873; SJJ), the Department of Defense (AS073218P1;
SJJ), and by grants from the Arkansas Children’s Hospital
and Arkansas Biosciences Institute (SJJ).
References
[1] CDC, “Prevalence of autism spectrum disorders—autism and
developmental disabilities monitoring network, United States,
2006,Morbidity and Mortality Weekly Report, vol. 58, no. 10,
pp. 1–20, 2009.
[2] J. K. Yao, S. Leonard, and R. Reddy, “Altered glutathione
redox state in schizophrenia,” Disease Markers,vol.22,no.1-2,
pp. 83–93, 2006.
[3]A.C.Andreazza,M.Kauer-SantAnna,B.N.Freyetal.,
“Oxidative stress markers in bipolar disorder: a meta-analysis,
Journal of Aective Disorders, vol. 111, no. 2-3, pp. 135–144,
2008.
[4] W. R. Markesbery, “Oxidative stress hypothesis in Alzheimer’s
disease,Free Radical Biology and Medicine, vol. 23, no. 1,
pp. 134–147, 1997.
[5] G. A. Mostafa, E. S. El-Hadidi, D. H. Hewedi, and M. M.
Abdou, “Oxidative stress in Egyptian children with autism:
relation to autoimmunity,Journal of Neuroimmunology,
vol. 219, no. 1-2, pp. 114–118, 2010.
[6] N. A. Meguid, A. A. Dardir, E. R. Abdel-Raouf, and A. Hashish,
“Evaluation of oxidative stress in autism: defective antioxidant
8 Autism Research and Treatment
enzymes and increased lipid peroxidation,Biological Trace
Element Research, vol. 143, no. 1, pp. 58–65, 2011.
[7] A. Chauhan, V. Chauhan, W. T. Brown, and I. Cohen,
“Oxidative stress in autism: increased lipid peroxidation and
reduced serum levels of ceruloplasmin and transferrin—the
antioxidant proteins,Life Sciences, vol. 75, no. 21, pp. 2539–
2549, 2004.
[8] S.S.Zoroglu,F.Armutcu,S.Ozenetal.,“Increasedoxidative
stress and altered activities of erythrocyte free radical scaveng-
ing enzymes in autism,European Archives of Psychiatry and
Clinical Neuroscience, vol. 254, no. 3, pp. 143–147, 2004.
[9] S. S¨
oˇ
g¨
ut,S.S.Zoro
ˇ
glu, H. ¨
Ozyurt et al., “Changes in nitric
oxide levels and antioxidant enzyme activities may have a role
in the pathophysiological mechanisms involved in autism,”
Clinica Chimica Acta, vol. 331, no. 1-2, pp. 111–117, 2003.
[10] T. L. Sweeten, D. J. Posey, S. Shankar, and C. J. McDougle,
“High nitric oxide production in autistic disorder: a possible
role for interferon-γ,” Biological Psychiatry,vol.55,no.4,
pp. 434–437, 2004.
[11] M. A. Junaid, D. Kowal, M. Barua, P. S. Pullarkat, S. S. Brooks,
and R. K. Pullarkat, “Proteomic studies identified a single
nucleotide polymorphism in glyoxalase I as autism suscepti-
bility factor,American Journal of Medical Genetics, vol. 131,
no. 1, pp. 11–17, 2004.
[12] M. Boso, E. Emanuele, P. Minoretti et al., “Alterations of
circulating endogenous secretory RAGE and S100A9 levels
indicating dysfunction of the AGE-RAGE axis in autism,Neu-
roscience Letters, vol. 410, no. 3, pp. 169–173, 2006.
[13] S. J. James, P. Cutler, S. Melnyk et al., “Metabolic biomarkers of
increased oxidative stress and impaired methylation capacity
in children with autism,American Journal of Clinical Nutri-
tion, vol. 80, no. 6, pp. 1611–1617, 2004.
[14] S. J. James, S. Melnyk, S. Jernigan et al., “Metabolic endophe-
notype and related genotypes are associated with oxidative
stress in children with autism,American Journal of Medical
Genetics, Part B, vol. 141, no. 8, pp. 947–956, 2006.
[15] S. J. James, S. Melnyk, G. Fuchs et al., “Ecacy of methylcobal-
amin and folinic acid treatment on glutathione redox status in
children with autism,American Journal of Clinical Nutrition,
vol. 89, no. 1, pp. 425–430, 2009.
[16] S. J. James, S. Rose, S. Melnyk et al., “Cellular and mito-
chondrial glutathione redox imbalance in lymphoblastoid cells
derived from children with autism,FASEB Journal, vol. 23,
no. 8, pp. 2374–2383, 2009.
[17] K.Bowers,Q.Li,J.Bressler,D.Avramopoulos,C.Newschaer,
and M. D. Fallin, “Glutathione pathway gene variation and
risk of autism spectrum disorders,Journal of Neurodevelop-
mental Disorders, vol. 3, no. 2, pp. 132–143, 2011.
[18] S. Biswas, A. S. Chida, and I. Rahman, “Redox modifications
of protein-thiols: emerging roles in cell signaling,Biochemical
Pharmacology, vol. 71, no. 5, pp. 551–564, 2006.
[19] G. Filomeni, G. Rotilio, and M. R. Ciriolo, “Cell signalling
and the glutathione redox system,Biochemical Pharmacology,
vol. 64, no. 5-6, pp. 1057–1064, 2002.
[20] M. Fratelli, L. O. Goodwin, U. A. Ørom et al., “Gene expres-
sion profiling reveals a signaling role of glutathione in redox
regulation,Proceedings of the National Academy of Sciences of
the United States of America, vol. 102, no. 39, pp. 13998–14003,
2005.
[21] A. Pastore, G. Federici, E. Bertini, and F. Piemonte, “Analysis of
glutathione: implication in redox and detoxification,Clinica
Chimica Acta, vol. 333, no. 1-2, pp. 19–39, 2003.
[22] Y. W. Kwon, H. Masutani, H. Nakamura, Y. Ishii, and J. Yodoi,
“Redox regulation of cell growth and cell death,Biological
Chemistry, vol. 384, no. 7, pp. 991–996, 2003.
[23] F. Q. Schafer and G. R. Buettner, “Redox environment of the
cell as viewed through the redox state of the glutathione disul-
fide/glutathione couple,Free Radical Biology and Medicine,
vol. 30, no. 11, pp. 1191–1212, 2001.
[24] H. M. Lanˇ
der, A. J. Mllbank, J. M. Tauras et al., “Redox regula-
tion of cell signalling,” Nature, vol. 381, no. 6581, pp. 380–381,
1996.
[25] P. Ghezzi, “Regulation of protein function by glutathionyla-
tion,Free Radical Research, vol. 39, no. 6, pp. 573–580, 2005.
[26] D. A. Dickinson and H. J. Forman, “Glutathione in defense
and signaling: lessons from a small thiol,Annals of the New
York Academy of Sciences, vol. 973, pp. 488–504, 2002.
[27] M. Noble, J. Smith, J. Power, and M. Mayer-Pr¨
oschel, “Redox
state as a central modulator of precursor cell function,Annals
of the New York Academy of Sciences, vol. 991, pp. 251–271,
2003.
[28] D. P. Jones, “Extracellular redox state: refining the definition of
oxidative stress in aging,Rejuvenation Research, vol. 9, no. 2,
pp. 169–181, 2006.
[29] M. Asensi, J. Sastre, F. V. Pallardo et al., “Ratio of reduced to
oxidized glutathione as indicator of oxidative stress status and
DNA damage,Methods in Enzymology, vol. 299, pp. 267–276,
1999.
[30] S. S. Iyer, C. J. Accardi, T. R. Ziegler et al., “Cysteine redox
potential determines pro-inflammatory IL-1βlevels,PLoS
One, vol. 4, no. 3, Article ID e5017, 2009.
[31] D. Barford, “The role of cysteine residues as redox-sensitive
regulatory switches,” Current Opinion in Structural Biology,
vol. 14, no. 6, pp. 679–686, 2004.
[32] D. P. Jones, Y. M. Go, C. L. Anderson, T. R. Ziegler, J. M.
Kinkade Jr., and W. G. Kirlin, “Cysteine/cystine couple is a
newly recognized node in the circuitry for biologic redox sig-
naling and control,FASEB Journal, vol. 18, no. 11, pp. 1246–
1248, 2004.
[33] D. Spadaro, B. W. Yun, S. H. Spoel, C. Chu, Y. Q. Wang, and
G. J. Loake, “The redox switch: dynamic regulation of protein
function by cysteine modifications,Physiologia Plantarum,
vol. 138, no. 4, pp. 360–371, 2010.
[34] J. Den Hertog, A. Groen, and T. van der Wijk, “Redox regu-
lation of protein-tyrosine phosphatases,Archives of Biochem-
istry and Biophysics, vol. 434, no. 1, pp. 11–15, 2005.
[35] Z. Yan, S. K. Garg, J. Kipnis, and R. Banerjee, “Extracellular
redox modulation by regulatory T cells,Nature Chemical
Biology, vol. 5, no. 10, pp. 721–723, 2009.
[36]C.E.Cooper,R.P.Patel,P.S.Brookes,andV.M.Darley-
Usmar, “Nanotransducers in cellular redox signaling: mod-
ification of thiols by reactive oxygen and nitrogen species,”
Trends in Biochemical Sciences, vol. 27, no. 10, pp. 489–492,
2002.
[37] R. P. Warren, L. J. Yonk, R. A. Burger et al., “Deficiency of
suppressor-inducer (CD4+CD45RA+) T cells in autism,Im-
munological Investigations, vol. 19, no. 3, pp. 245–251, 1990.
[38] L. J. Yonk, R. P. Warren, R. A. Burger et al., “CD4+helper T
cell depression in autism,Immunology Letters, vol. 25, no. 4,
pp. 341–346, 1990.
[39] D. R. Denney, B. W. Frei, and G. R. Ganey, “Lymphocyte
subsets and interleukin-2 receptors in autistic children,
Journal of Autism and Developmental Disorders,vol.26,no.1,
pp. 87–97, 1996.
Autism Research and Treatment 9
[40] T. L. Sweeten, D. J. Posey, and C. J. McDougle, “High blood
monocyte counts and neopterin levels in children with autistic
disorder,American Journal of Psychiatry, vol. 160, no. 9,
pp. 1691–1693, 2003.
[41] S. Gupta, S. Aggarwal, B. Rashanravan, and T. Lee, “Th1-
and Th2-like cytokines in CD4+and CD8+Tcellsinautism,
Journal of Neuroimmunology, vol. 85, no. 1, pp. 106–109, 1998.
[42] C.A.Molloy,A.L.Morrow,J.Meinzen-Derretal.,“Elevated
cytokine levels in children with autism spectrum disorder,
Journal of Neuroimmunology, vol. 172, no. 1-2, pp. 198–205,
2006.
[43] P. Ashwood and A. J. Wakefield, “Immune activation of pe-
ripheral blood and mucosal CD3+lymphocyte cytokine pro-
files in children with autism and gastrointestinal symptoms,
Journal of Neuroimmunology, vol. 173, no. 1-2, pp. 126–134,
2006.
[44] H. Jyonouchi, S. Sun, and H. Le, “Proinflammatory and
regulatory cytokine production associated with innate and
adaptive immune responses in children with autism spectrum
disorders and developmental regression,Journal of Neuroim-
munology, vol. 120, no. 1-2, pp. 170–179, 2001.
[45] P. Ashwood, A. Enstrom, P. Krakowiak et al., “Decreased
transforming growth factor beta1 in autism: a potential link
between immune dysregulation and impairment in clinical
behavioral outcomes,Journal of Neuroimmunology, vol. 204,
no. 1-2, pp. 149–153, 2008.
[46] K. Okada, K. Hashimoto, Y. Iwata et al., “Decreased serum
levels of transforming growth factor-β1 in patients with
autism,Progress in Neuro-Psychopharmacology and Biological
Psychiatry, vol. 31, no. 1, pp. 187–190, 2007.
[47] S. W. Looney and P. W. Jones, “A method for comparing
two normal means using combined samples of correlated and
uncorrelated data,Statistics in Medicine,vol.22,no.9,
pp. 1601–1610, 2003.
[48] D. P. Jones, J. L. Carlson, V. C. Mody, J. Cai, M. J. Lynn, and
P. Sternberg, “Redox state of glutathione in human plasma,
Free Radical Biology and Medicine, vol. 28, no. 4, pp. 625–635,
2000.
[49] Y. M. Go and D. P. Jones, “Redox compartmentalization in
eukaryotic cells,Biochimica et Biophysica Acta, vol. 1780,
no. 11, pp. 1273–1290, 2008.
[50] R. Gysin, R. Kraftsik, J. Sandell et al., “Impaired glutathione
synthesis in schizophrenia: convergent genetic and functional
evidence,Proceedings of the National Academy of Sciences of
the United States of America, vol. 104, no. 42, pp. 16621–16626,
2007.
[51] M. Y. Yeh, E. L. Burnham, M. Moss, and L. A. Brown, “Chronic
alcoholism alters systemic and pulmonary glutathione redox
status,American Journal of Respiratory and Critical Care Med-
icine, vol. 176, no. 3, pp. 270–276, 2007.
[52] H. Nakamura, H. Masutani, and J. Yodoi, “Redox imbalance
and its control in HIV infection,Antioxidants and Redox
Signaling, vol. 4, no. 3, pp. 455–464, 2002.
[53] C. Cecchi, S. Latorraca, S. Sorbi et al., “Gluthatione level is
altered in lymphoblasts from patients with familial Alzheim-
er’s disease,Neuroscience Letters, vol. 275, no. 2, pp. 152–154,
1999.
[54] A. Larbi, J. Kempf, and G. Pawelec, “Oxidative stress modula-
tion and T cell activation,Experimental Gerontology, vol. 42,
no. 9, pp. 852–858, 2007.
[55]J.P.MessinaandD.A.Lawrence,“Cellcycleprogressionof
glutathione-depleted human peripheral blood mononuclear
cells is inhibited at S phase,Journal of Immunology, vol. 143,
no. 6, pp. 1974–1981, 1989.
[56] M. Klemke, G. H. Wabnitz, F. Funke, B. Funk, H. Kirchgessner,
and Y. Samstag, “Oxidation of cofilin mediates T cell hypore-
sponsiveness under oxidative stress conditions,” Immunity,
vol. 29, no. 3, pp. 404–413, 2008.
[57] T. Ando, K. Mimura, C. C. Johansson et al., “Transduction
with the antioxidant enzyme catalase protects human T cells
against oxidative stress,Journal of Immunology, vol. 181,
no. 12, pp. 8382–8390, 2008.
[58] P.Ashwood,P.Krakowiak,I.Hertz-Picciotto,R.Hansen,I.
N. Pessah, and J. Van de Water, “Altered T cell responses in
children with autism,Brain, Behavior, and Immunity, vol. 25,
no. 5, pp. 840–849, 2011.
[59] C. M. Liang, N. Lee, D. Cattell, and S. M. Liang, “Glutathione
regulates interleukin-2 activity on cytotoxic T-cells,” Journal of
Biological Chemistry, vol. 264, no. 23, pp. 13519–13523, 1989.
[60] H. Gmunder, S. Roth, H. P. Eck, H. Gallas, S. Mihm, and
W. Droge, “Interleukin-2 mRNA expression, lymphokine pro-
duction and DNA synthesis in glutathione-depleted T cells,
Cellular Immunology, vol. 130, no. 2, pp. 520–528, 1990.
[61]J.D.Peterson,L.A.Herzenberg,K.Vasquez,andC.
Waltenbaugh, “Glutathione levels in antigen-presenting cells
modulate Th1 versus Th2 response patterns,Proceedings of
the National Academy of Sciences of the United States of Ameri-
ca, vol. 95, no. 6, pp. 3071–3076, 1998.
[62] Y. Murata, T. Shimamura, and J. Hamuro, “The polarization of
Th1/Th2 balance is dependent on the intracellular thiol redox
status of macrophages due to the distinctive cytokine produc-
tion,International Immunology, vol. 14, no. 2, pp. 201–212,
2002.
[63] L. Ekl¨
ow,H.Thor,andS.Orrenius,“Formationandeux of
glutathione disulfide studied in isolated rat hepatocytes,FEBS
Letters, vol. 127, no. 1, pp. 125–128, 1981.
[64] D. A. Rossignol and R. E. Frye, “Mitochondrial dysfunction
in autism spectrum disorders: a systematic review and meta-
analysis,Molecular Psychiatry. In press.
[65] A. H. Schapira, J. M. Cooper, D. Dexter, P. Jenner, J. B. Clark,
and C. D. Marsden, “Mitochondrial complex I deficiency in
Parkinson’s disease,” The Lancet, vol. 1, no. 8649, p. 1269, 1989.
[66] A. Maruszak and C. Zekanowski, “Mitochondrial dysfunction
and Alzheimer’s disease,Progress in NeuroPsychopharmacolo-
gy and Biological Psychiatry, vol. 35, no. 2, pp. 320–330, 2011.
[67] H. B. Clay, S. Sillivan, and C. Konradi, “Mitochondrial dys-
function and pathology in bipolar disorder and schizophre-
nia,International Journal of Developmental Neuroscience,
vol. 29, no. 3, pp. 311–324, 2011.
[68] D. J. Keating, “Mitochondrial dysfunction, oxidative stress,
regulation of exocytosis and their relevance to neurodegen-
erative diseases,Journal of Neurochemistry, vol. 104, no. 2,
pp. 298–305, 2008.
[69] Z. Li, K. I. Okamoto, Y. Hayashi, and M. Sheng, “The
importance of dendritic mitochondria in the morphogenesis
and plasticity of spines and synapses,Cell, vol. 119, no. 6,
pp. 873–887, 2004.
[70] A. Quintana, C. Schwindling, A. S. Wenning et al., “T cell ac-
tivation requires mitochondrial translocation to the immuno-
logical synapse,Proceedings of the National Academy of Scien-
ces of the United States of America, vol. 104, no. 36, pp. 14418–
14423, 2007.
[71] G. Pani, R. Colavitti, S. Borrello, and T. Galeotti, “Redox reg-
ulation of lymphocyte signaling,” IUBMB Life,vol.49,no.5,
pp. 381–389, 2000.
[72] T. M. Buttke and P. A. Sandstrom, “Redox regulation of pro-
grammed cell death in lymphocytes,Free Radical Research,
vol. 22, no. 5, pp. 389–397, 1995.
10 Autism Research and Treatment
[73] C. Giulivi, Y. F. Zhang, A. Omanska-Klusek et al., “Mitochon-
drial dysfunction in autism,Journal of the American Medical
Association, vol. 304, no. 21, pp. 2389–2396, 2010.
[74] B. R. Imhoand J. M. Hansen, “Extracellular redox status
regulates Nrf2 activation through mitochondrial reactive oxy-
gen species,Biochemical Journal, vol. 424, no. 3, pp. 491–500,
2009.
[75] Y. M. Go, H. Park, M. Koval et al., “A key role for mitochondria
in endothelial signaling by plasma cysteine/cystine redox
potential,Free Radical Biology and Medicine, vol. 48, no. 2,
pp. 275–283, 2010.
[76] L. I. Leichert and U. Jakob, “Protein thiol modifications
visualized in vivo,PLoS Biology, vol. 2, no. 11, Article ID e333,
2004.
[77] P. Ghezzi, “Oxidoreduction of protein thiols in redox regula-
tion,Biochemical Society Transactions, vol. 33, no. 6, pp. 1378–
1381, 2005.
[78] S.S.Iyer,D.P.Jones,K.L.Brigham,andM.Rojas,“Oxidation
of plasma cysteine/cystine redox state in endotoxin-induced
lung injury,American Journal of Respiratory Cell and Molecu-
lar Biology, vol. 40, no. 1, pp. 90–98, 2009.
[79] S. K. Garg, V. Vitvitsky, R. Albin, and R. Banerjee, “Astrocytic
redox remodeling by amyloid beta peptide,Antioxidants and
Redox Signaling, vol. 14, no. 12, pp. 2385–2397, 2011.
[80] S. E. Moriarty-Craige, J. Adkison, M. Lynn et al., “Antioxidant
supplements prevent oxidation of cysteine/cystine redox in
patients with age-related macular degeneration,American
Journal of Ophthalmology, vol. 140, no. 6, pp. 1020–1026, 2005.
[81] D. P. Jones, Y. Park, N. Gletsu-Miller et al., “Dietary sulfur
amino acid eects on fasting plasma cysteine/cystine redox
potential in humans,Nutrition, vol. 27, no. 2, pp. 199–205,
2011.
... Previous case-control studies have reported that children with ASD show abnormal plasma metabolite levels in the glutathione redox pathway [116][117][118]. A decrease in reduced glutathione (GSH), increase in oxidized glutathione disulfide (GSSG), and decrease in the glutathione redox ratio (GSH/GSSG) have been reported in children with ASD [116]. Oxidative stress in children with ASD has been reported by the mitochondrial dysfunction and increased oxidative stress markers such as decreased antioxidant enzyme activity, increased lipid peroxidation, and accumulation of advanced glycation products in peripheral blood [97,99,100,[104][105][106][107][108] (Figure 2). ...
... Transferrin acts as an antioxidant by reducing the concentration of free ferrous ion, and abnormalities in ceruloplasmin and transferrin levels may lead to abnormal iron and copper metabolism in patients with ASD [100,115]. Previous case-control studies have reported that children with ASD show abnormal plasma metabolite levels in the glutathione redox pathway [116][117][118]. A decrease in reduced glutathione (GSH), increase in oxidized glutathione disulfide (GSSG), and decrease in the glutathione redox ratio (GSH/GSSG) have been reported in children with ASD [116]. ...
... Previous case-control studies have reported that children with ASD show abnormal plasma metabolite levels in the glutathione redox pathway [116][117][118]. A decrease in reduced glutathione (GSH), increase in oxidized glutathione disulfide (GSSG), and decrease in the glutathione redox ratio (GSH/GSSG) have been reported in children with ASD [116]. ...
Article
Full-text available
Autism spectrum disorder (ASD) is a neurodevelopmental disorder (NDD) characterized by impairments in social communication, repetitive behaviors, restricted interests, and hyperesthesia/hypesthesia caused by genetic and/or environmental factors. In recent years, inflammation and oxidative stress have been implicated in the pathogenesis of ASD. In this review, we discuss the inflammation and oxidative stress in the pathophysiology of ASD, particularly focusing on maternal immune activation (MIA). MIA is a one of the common environmental risk factors for the onset of ASD during pregnancy. It induces an immune reaction in the pregnant mother’s body, resulting in further inflammation and oxidative stress in the placenta and fetal brain. These negative factors cause neurodevelopmental impairments in the developing fetal brain and subsequently cause behavioral symptoms in the offspring. In addition, we also discuss the effects of anti-inflammatory drugs and antioxidants in basic studies on animals and clinical studies of ASD. Our review provides the latest findings and new insights into the involvements of inflammation and oxidative stress in the pathogenesis of ASD.
... No reports have been found regarding GSH/GSSG levels in neutrophils. However, previous studies have reported that the GSH/GSSG ratio in the monocytes and T cells of healthy children ranges from 12 to 25 and from 10 to 24, respectively [50]. Our findings show similar values to these previous reports, although it must be considered that our measurements were performed in neutrophils from healthy young adults that were seeded in cell culture media (RPMI 1640) + 2% AS supplemented with a mixture of antioxidants (GSH + NAC and ALL) and stimulated with LPS. ...
Article
Full-text available
Neutrophil extracellular traps (NETs) require reactive oxygen species (ROS) to eliminate pathogens by inducing oxidative stress. However, this process can also cause tissue damage to the host. Neutrophils contain high concentrations of vitamin C (1.5 mM) compared to the bloodstream (0.1 mM), and this antioxidant can interact with vitamin E and glutathione (GSH) inside the cell to maintain the redox balance. Previous studies have investigated the effect of vitamins E or C and N-acetyl cysteine (NAC) on NET formation, but the interactions of these molecules in neutrophils remain unknown. In this study, we investigated the effect of antioxidants alone and two combinations on NET formation and oxidative stress. Neutrophils were pre-loaded with GSH + NAC or vitamin E + vitamin C + GSH + NAC (termed ALL), and LPS-induced NET formation was assessed using fluorometry and immunofluorescence. Antioxidant effects were evaluated by measuring the total antioxidant capacity (TAC), GSH/GSSG ratio, ROS production, nitrite + nitrate levels, and lipid peroxidation. Our results showed that even low doses of antioxidants are capable of decreasing NETs. Furthermore, the combinations augmented TAC and GSH/GSSG ratio and decreased ROS, nitrites + nitrates, and malondialdehyde (MDA) levels in supplemented neutrophils in vitro.
... Finally, 3-nitrotyrosine (3-NT), a measure of oxidative damage to proteins, and 3-chlorotyrosine (3-CT), a measure of immune activity, were also measured. Measurements were performed within 2 weeks of collection [40,41]. ...
Article
Full-text available
Autism Spectrum Disorder (ASD) is associated with many variations in metabolism, but the exact correlates of these metabolic disturbances with behavior and development and their links to other core metabolic disruptions are understudied. In this study, large-scale targeted LC-MS/MS metabolomic analysis was conducted on fasting morning plasma samples from 57 children with ASD (29 with neurodevelopmental regression, NDR) and 37 healthy controls of similar age and gender. Linear model determined the metabolic signatures of ASD with and without NDR, measures of behavior and neurodevelopment, as well as markers of oxidative stress, inflammation, redox, methylation, and mitochondrial metabolism. MetaboAnalyst ver 5.0 (the Wishart Research Group at the University of Alberta, Edmonton, Canada) identified the pathways associated with altered metabolic signatures. Differences in histidine and glutathione metabolism as well as aromatic amino acid (AAA) biosynthesis differentiated ASD from controls. NDR was associated with disruption in nicotinamide and energy metabolism. Sleep and neurodevelopment were associated with energy metabolism while neurodevelopment was also associated with purine metabolism and aminoacyl-tRNA biosynthesis. While behavior was associated with some of the same pathways as neurodevelopment, it was also associated with alternations in neurotransmitter metabolism. Alterations in methylation was associated with aminoacyl-tRNA biosynthesis and branched chain amino acid (BCAA) and nicotinamide metabolism. Alterations in glutathione metabolism was associated with changes in glycine, serine and threonine, BCAA and AAA metabolism. Markers of oxidative stress and inflammation were associated with energy metabolism and aminoacyl-tRNA biosynthe-sis. Alterations in mitochondrial metabolism was associated with alterations in energy metabolism and L-glutamine. Using behavioral and biochemical markers, this study finds convergent disturbances in specific metabolic pathways with ASD, particularly changes in energy, nicotinamide, neu-rotransmitters, and BCAA, as well as aminoacyl-tRNA biosynthesis.
... Redox homeostasis is a balance between the production of oxidants and endogenous antioxidant defense mechanisms and its disturbance leads to oxidative stress that has deteriorating effects on cells [16]. Oxidative stress, clinically defined by a decrease in the GSH/GSSG ratio, has been described in many neurobehavioral disorders, including autism [13,37,39,61]. Indeed, as mentioned above, multiple studies, including ours, reported lower GSH levels and a lower GSH/GSSG ratio in patients with autism in comparison to healthy controls [16,18,23,26], which were elevated during methylcobalamin treatment [31,37,44]. ...
Article
Full-text available
(1) Background: Autism, also known as autism-spectrum disorder, is a pervasive developmental disorder affecting social skills and psychological status in particular. The complex etiopathogenesis of autism limits efficient therapy, which leads to problems with the normal social integration of the individual and causes severe family distress. Injectable methylcobalamin was shown to improve the clinical status of patients via enhanced cell oxidative status and/or methylation capacity. Here we tested the efficiency of a syrup form of methylcobalamin in treating autism. (2) Methods: Methylcobalamin was administered daily at 500 µg dose to autistic children and young adults (n = 25) during a 200-day period. Clinical and psychological status was evaluated by parents and psychologists and plasma levels of reduced and oxidized glutathione, vitamin B12, homocysteine, and cysteine were determined before the treatment, and at day 100 and day 200 of the treatment. (3) Results: Good patient compliance was reported. Methylcobalamin treatment gradually improved the overall clinical and psychological status, with the highest impact in the social domain, followed by the cognitive, behavioral and communication characteristics. Changes in the clinical and psychological status were strongly associated with the changes in the level of reduced glutathione and reduced/oxidized glutathione ratio. (4) Conclusion: A high dose of methylcobalamin administered in syrup form ameliorates the clinical and psychological status of autistic individuals, probably due to the improved oxidative status.
... La evidencia existente apoya el rol de estrés oxidativo en la patogénesis y manifestaciones clínicas en niños con autismo 7 . Además, numerosos estudios han demostrado un elevado daño oxidativo y una reducción en la defensa antioxidante en los niños diagnosticados con TEA 6,23,24,25,26 , aun cuando en otros estudios los datos clínicos fueron inconsistentes 27,28 . ...
Article
Full-text available
Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders, impaired social interaction, language, and communication, which has increased worldwide in recent years. The deterioration of this condition occurs mainly at the brain level, currently it has been postulated that mitochondrial dysfunction, increased oxidative stress and decreased antioxidant defense leads to an imbalance in the ability to counteract the harmful effects of oxidative stress, such as oxidative degradation of lipids, proteins, and DNA that can cause damage to brain tissue, leading to the clinical symptoms and behaviors of ASD. Mitochondrial dysfunction can occur due to abnormalities in the electron transport chain, which induces and increases oxidative stress. On the other hand, the brain is extremely vulnerable to oxidative stress, due to its high oxygen consumption, its limited antioxidant capacity, and higher amounts of fatty acids and iron. This increased susceptibility of the brain to oxidative damage highlights the importance of understanding the role of oxidative stress in the clinical manifestations of ASD. Several studies have observed an increase in oxidative stress markers and a decrease in antioxidant enzymes in autism. Therefore, improving the oxidative state and maintaining the redox balance could improve the clinical manifestations of autism. The present study aims to carry out a narrative review on oxidative stress and mitochondrial dysfunction associated with ASD.
Article
Background Obesity-associated inflammation drives the development of insulin resistance and type 2 diabetes. We sought to identify associations of circulating regulatory T cells (Treg) with the degree of obesity (eg, body mass index Z-score [BMIz]), insulin resistance (homeostatic model of insulin resistance [HOMA-IR]), and glycemic control (HbA1c) in children and adolescents. We further sought to examine associations among bioenergetics of peripheral blood mononuclear cells (PBMCs) and CD4 T cells and BMIz, HOMA-IR, and HbA1c. Methods A total of 65 children and adolescents between the ages 5 and 17 years were studied. HbA1c and fasting levels of plasma glucose and insulin were measured. We quantified circulating Tregs (CD3⁺CD4⁺CD25⁺CD127⁻FoxP3⁺) by flow cytometry, and measured mitochondrial respiration (oxygen consumption rate [OCR]) and glycolysis (extracellular acidification rate [ECAR]) in PBMCs and isolated CD4 T cells by Seahorse extracellular flux analysis. Results Tregs (% CD4) are negatively associated with BMIz but positively associated with HOMA-IR. In PBMCs, OCR/ECAR (a ratio of mitochondrial respiration to glycolysis) is positively associated with BMIz but negatively associated with HbA1c. Conclusions In children, Tregs decrease as body mass index increases; however, the metabolic stress and inflammation associated with insulin resistance may induce a compensatory increase in Tregs. The degree of obesity is also associated with a shift away from glycolysis in PBMCs but as HbA1c declines, metabolism shifts back toward glycolysis. Comprehensive metabolic assessment of the immune system is needed to better understand the implications immune cell metabolic alterations in the progression from a healthy insulin-sensitive state toward glucose intolerance in children. Trial registration This observational study was registered at the ClinicalTrials.gov (NCT03960333, https://clinicaltrials.gov/study/NCT03960333?term=NCT03960333&rank=1).
Article
Background Autism, a neurodevelopment disorder that was regarded as insignificant in the past is thereby now recognized as a major problem with an increasing prevalence over the past 50 years affecting 1 out of every 160 children worldwide. Recent accumulating evidence indicates that autism is a consequence of the fusion of environmental, genetic, and epigenetic components. Due to the lack of effective pharmacotherapy, alternative approaches are being explored for their beneficial effect on autistic symptoms. Methods A literature review was performed identifying previously published clinical studies that were set up as an alternative therapy for alleviating the symptoms of autism. The data were collected from PubMed and Google Scholar databases. Results A total of 13 kinds of interventions including video modelling, play therapy, music therapy, yoga, social skills training, sensory integration (SIT), scalp acupuncture, medical clowning, animal-assisted activity, theatre-based intervention, Tai Chu Chuan training, novel mattress technology, and magnetic resonance imaging were found to be affecting the symptoms associated with autism. Conclusion From sufficient clinical evidence, it was estimated that alternative approaches such as music therapy and play therapy have the most beneficial effect in mitigating the symptoms to an extent.
Article
Full-text available
The coronavirus disease 2019 (COVID-19) pandemic affects all countries and populations worldwide, significantly impacting people with autism with a high risk of morbidity and mortality due to COVID-19. Approximately 25% of children with autism have an asymptomatic or symptomatic immune deficiency or dysfunction. In addition, they frequently have various comorbid conditions that increase the severity of COVID-19. In addition, severe COVID-19 during pregnancy may increase the risk of autism in the offspring. Furthermore, severe acute respiratory syndrome coronavirus 2 could target human nervous system tissues due to its neurotrophic effects. The COVID-19 pandemic intensely impacts many patients and families in the autism community, especially the complex management of autism-associated disorders during the complete lockdown. During the complete lockdown, children with autism had difficulties coping with the change in their routine, lack of access to special education services, limited physical space available, and problems related to food and sleep. Additionally, children with autism or intellectual disabilities are more liable to be abused by others during the pandemic when the standard community supports are no longer functioning to protect them. Early detection and vaccination of children with autism against COVID-19 are highly indicated. They should be prioritized for testing, vaccination, and proper management of COVID-19 and other infectious diseases. In this review, we discuss the various effects of COVID-19 on children with autism, the difficulties they face, the increased risk of infection during pregnancy, how to alleviate the impact of COVID-19, and how to correct the inequalities in children with autism.
Chapter
Ketogenic diets have been used to treat epilepsy for nearly a century. Alongside enduring clinical success with a ketogenic diet, metabolism’s critical role in health and in diseases in the central nervous system and throughout the body is increasingly appreciated. Furthermore, metabolism-based strategies have been proven equal or even superior to pharmacological treatments in specific cases and for specific diseases. Rather than causing unwanted off-target pharmacological side effects, addressing metabolic dysfunction can improve overall health simultaneously. Enduring interest in the ketogenic diet’s proven efficacy in stopping seizures and emerging efficacy in other disorders has fueled renewed efforts to determine key mechanisms and diverse applications of metabolic therapies. In parallel, multiple strategies are being developed to mobilize similar metabolic benefits without reliance on such a strict diet. Research interest in metabolic therapies has spread into laboratories and clinics of every discipline, and could yield entirely new classes of drugs and treatment regimens. This work is the first comprehensive scientific resource on the ketogenic diet, covering the latest research into the mechanisms, established and emerging applications, metabolic alternatives, and implications for health and disease. Experts in clinical and basic research share their research into mechanisms spanning from ion channels to epigenetics, their insights based on decades of experience with the ketogenic diet in epilepsy, and their evidence for emerging applications ranging from autism to Alzheimer’s disease to brain cancer.
Article
Full-text available
Current thinking attributes the balance between T helper 1 (Th1) and Th2 cytokine response patterns in immune responses to the nature of the antigen, the genetic composition of the host, and the cytokines involved in the early interaction between T cells and antigen-presenting cells. Here we introduce glutathione, a tripeptide that regulates intracellular redox and other aspects of cell physiology, as a key regulatory element in this process. By using three different methods to deplete glutathione from T cell receptor transgenic and conventional mice and studying in vivo and/or in vitro responses to three distinct antigens, we show that glutathione levels in antigen-presenting cells determine whether Th1 or Th2 response patterns predominate. These findings present new insights into immune response alterations in HIV and other diseases. Further, they potentially offer an explanation for the well known differences in immune responses in "Th1" and "Th2" mouse strains.
Article
Full-text available
Despite evidence that autism is highly heritable with estimates of 15 or more genes involved, few studies have directly examined associations of multiple gene interactions. Since inability to effectively combat oxidative stress has been suggested as a mechanism of autism, we examined genetic variation 42 genes (308 single-nucleotide polymorphisms (SNPs)) related to glutathione, the most important antioxidant in the brain, for both marginal association and multi-gene interaction among 318 case–parent trios from The Autism Genetic Resource Exchange. Models of multi-SNP interactions were estimated using the trio Logic Regression method. A three-SNP joint effect was observed for genotype combinations of SNPs in glutaredoxin, glutaredoxin 3 (GLRX3), and cystathione gamma lyase (CTH); OR = 3.78, 95% CI: 2.36, 6.04. Marginal associations were observed for four genes including two involved in the three-way interaction: CTH, alcohol dehydrogenase 5, gamma-glutamylcysteine synthetase, catalytic subunit and GLRX3. These results suggest that variation in genes involved in counterbalancing oxidative stress may contribute to autism, though replication is necessary. Electronic supplementary material The online version of this article (doi:10.1007/s11689-011-9077-4) contains supplementary material, which is available to authorized users.
Article
Full-text available
A comprehensive literature search was performed to collate evidence of mitochondrial dysfunction in autism spectrum disorders (ASDs) with two primary objectives. First, features of mitochondrial dysfunction in the general population of children with ASD were identified. Second, characteristics of mitochondrial dysfunction in children with ASD and concomitant mitochondrial disease (MD) were compared with published literature of two general populations: ASD children without MD, and non-ASD children with MD. The prevalence of MD in the general population of ASD was 5.0% (95% confidence interval 3.2, 6.9%), much higher than found in the general population (≈ 0.01%). The prevalence of abnormal biomarker values of mitochondrial dysfunction was high in ASD, much higher than the prevalence of MD. Variances and mean values of many mitochondrial biomarkers (lactate, pyruvate, carnitine and ubiquinone) were significantly different between ASD and controls. Some markers correlated with ASD severity. Neuroimaging, in vitro and post-mortem brain studies were consistent with an elevated prevalence of mitochondrial dysfunction in ASD. Taken together, these findings suggest children with ASD have a spectrum of mitochondrial dysfunction of differing severity. Eighteen publications representing a total of 112 children with ASD and MD (ASD/MD) were identified. The prevalence of developmental regression (52%), seizures (41%), motor delay (51%), gastrointestinal abnormalities (74%), female gender (39%), and elevated lactate (78%) and pyruvate (45%) was significantly higher in ASD/MD compared with the general ASD population. The prevalence of many of these abnormalities was similar to the general population of children with MD, suggesting that ASD/MD represents a distinct subgroup of children with MD. Most ASD/MD cases (79%) were not associated with genetic abnormalities, raising the possibility of secondary mitochondrial dysfunction. Treatment studies for ASD/MD were limited, although improvements were noted in some studies with carnitine, co-enzyme Q10 and B-vitamins. Many studies suffered from limitations, including small sample sizes, referral or publication biases, and variability in protocols for selecting children for MD workup, collecting mitochondrial biomarkers and defining MD. Overall, this evidence supports the notion that mitochondrial dysfunction is associated with ASD. Additional studies are needed to further define the role of mitochondrial dysfunction in ASD.
Article
Full-text available
Astrocytes are critical for neuronal redox homeostasis providing them with cysteine needed for glutathione synthesis. In this study, we demonstrate that the astrocytic redox response signature provoked by amyloid beta (Aβ) is distinct from that of a general oxidant (tertiary-butylhydroperoxide [t-BuOOH]). Acute Aβ treatment increased cystathionine β-synthase (CBS) levels and enhanced transsulfuration flux in contrast to repeated Aβ exposure, which decreased CBS and catalase protein levels. Although t-BuOOH also increased transsulfuration flux, CBS levels were unaffected. The net effect of Aβ treatment was an oxidative shift in the intracellular glutathione/glutathione disulfide redox potential in contrast to a reductive shift in response to peroxide. In the extracellular compartment, Aβ, but not t-BuOOH, enhanced cystine uptake and cysteine accumulation, and resulted in remodeling of the extracellular cysteine/cystine redox potential in the reductive direction. The redox changes elicited by Aβ but not peroxide were associated with enhanced DNA synthesis. CBS activity and protein levels tended to be lower in cerebellum from patients with Alzheimer's disease than in age-matched controls. Our study suggests that the alterations in astrocytic redox status could compromise the neuroprotective potential of astrocytes and may be a potential new target for therapeutic intervention in Alzheimer's disease.
Chapter
Introduction Glutathione and Redox Regulation in Immunity Protein Cysteine Oxidation Mechanisms for PSSG Formation and the Complex Scenario of Protein Glutathionylation Deglutathionylation Identification of Proteins Undergoing Glutathionylation Functional Consequences of Protein Glutathionylation Structural Changes Induced by Protein Glutathionylation Conclusions References
Article
Redox state is a term used widely in the research field of free radicals and oxidative stress. Unfortunately, it is used as a general term referring to relative changes that are not well defined or quantitated. In this review we provide a definition for the redox environment of biological fluids, cell organelles, cells, or tissue. We illustrate how the reduction potential of various redox couples can be estimated with the Nernst equation and show how pH and the concentrations of the species comprising different redox couples influence the reduction potential. We discuss how the redox state of the glutathione disulfide-glutathione couple (GSSG/2GSH) can serve as an important indicator of redox environment. There are many redox couples in a cell that work together to maintain the redox environment; the GSSG/2GSH couple is the most abundant redox couple in a cell. Changes of the half-cell reduction potential (E(hc)) of the GSSG/2GSH couple appear to correlate with the biological status of the cell: proliferation E(hc) approximately -240 mV; differentiation E(hc) approximately -200 mV; or apoptosis E(hc) approximately -170 mV. These estimates can be used to more fully understand the redox biochemistry that results from oxidative stress. These are the first steps toward a new quantitative biology, which hopefully will provide a rationale and understanding of the cellular mechanisms associated with cell growth and development, signaling, and reductive or oxidative stress.
Article
The mechanisms of thiol metabolism and chemistry have particular relevance to both cellular defenses against toxicant exposure and to redox signaling. Here, we will focus on glutathione (GSH), the major endogenous low- molecular-weight nonprotein thiol synthesized de novo in mammalian cells. The major pathways for GSH metabolism in defense of the cell are reduction of hydroperoxides by glutathione peroxidases (GSHPx) and some peroxiredoxins, which yield glutathione disulfide (GSSG), and conjugation reactions catalyzed by glutathione-S-transferases. GSSG can be reduced to GSH by glutathione reductase, but glutathione conjugates are excreted from cells. The exoenzyme γ-glutamyltranspeptidase (GGT) removes the glutamate from extracellular GSH, producing cysteinyl-glycine from which a dipeptidase then generates cysteine, an amino acid often limiting for de novo GSH synthesis. Synthesis of GSH from the constituent amino acids occurs in two regulated, enzymatically catalyzed steps. The signaling pathways leading to activation of the transcription factors that regulate these genes are a current area of intense investigation. The elucidation of the signaling for GSH biosynthesis in human bronchial epithelial cells in response to 4-hydroxynonenal (4HNE), an end product of lipid peroxidation, will be used as an example. GSH also participates in redox signaling through the removal of H2O2, which has the properties of a second messenger, and by reversing the formation of sulfenic acid, a moiety formed by reaction of critical cysteine residues in signaling proteins with H2O2. Disruption of GSH metabolism will therefore have major a impact upon function of cells in terms of both defense and normal physiology.
Article
Diverse functions of eukaryotic cells are optimized by organization of compatible chemistries into distinct compartments defined by the structures of lipid-containing membranes, multiprotein complexes and oligomeric structures of saccharides and nucleic acids. This structural and chemical organization is coordinated, in part, through cysteine residues of proteins which undergo reversible oxidation-reduction and serve as chemical/structural transducing elements. The central thiol/disulfide redox couples, thioredoxin-1, thioredoxin-2, GSH/GSSG and cysteine/cystine (Cys/CySS), are not in equilibrium with each other and are maintained at distinct, non-equilibrium potentials in mitochondria, nuclei, the secretory pathway and the extracellular space. Mitochondria contain the most reducing compartment, have the highest rates of electron transfer and are highly sensitive to oxidation. Nuclei also have more reduced redox potentials but are relatively resistant to oxidation. The secretory pathway contains oxidative systems which introduce disulfides into proteins for export. The cytoplasm contains few metabolic oxidases and this maintains an environment for redox signaling dependent upon NADPH oxidases and NO synthases. Extracellular compartments are maintained at stable oxidizing potentials. Controlled changes in cytoplasmic GSH/GSSG redox potential are associated with functional state, varying with proliferation, differentiation and apoptosis. Variation in extracellular Cys/CySS redox potential is also associated with proliferation, cell adhesion and apoptosis. Thus, cellular redox biology is inseparable from redox compartmentalization. Further elucidation of the redox control networks within compartments will improve the mechanistic understanding of cell functions and their disruption in disease.