ArticlePDF Available

Advances in the development of novel antioxidant therapies as an approach for fetal alcohol syndrome prevention

Authors:

Abstract and Figures

Ethanol is the most common human teratogen, and its consumption during pregnancy can produce a wide range of abnormalities in infants known as fetal alcohol spectrum disorder (FASD). The major characteristics of FASD can be divided into: (i) growth retardation, (ii) craniofacial abnormalities, and (iii) central nervous system (CNS) dysfunction. FASD is the most common cause of nongenetic mental retardation in Western countries. Although the underlying molecular mechanisms of ethanol neurotoxicity are not completely determined, the induction of oxidative stress is believed to be one central process linked to the development of the disease. Currently, there is no known effective strategy for prevention (other than alcohol avoidance) or treatment. In the present review we will provide the state of art in the evidence for the use of antioxidants as a potential therapeutic strategy for the treatment using whole-embryo and culture cells models of FASD. We conclude that the imbalance of the intracellular redox state contributes to the pathogenesis observed in FASD models, and we suggest that antioxidant therapy can be considered a new efficient strategy to mitigate the effects of prenatal ethanol exposure. Birth Defects Research (Part A), 2014. © 2014 Wiley Periodicals, Inc.
Content may be subject to copyright.
Advances in the Development of Novel Antioxidant Therapies
as an Approach for Fetal Alcohol Syndrome Prevention
Xavier Joya
1,2
, Oscar Garcia-Algar*
1,2,3
, Judith Salat-Batlle
1,2
, Cristina Pujades
4
,
and Oriol Vall
1,2,3
Ethanol is the most common human teratogen, and its consumption during
pregnancy can produce a wide range of abnormalities in infants known as
fetal alcohol spectrum disorder (FASD). The major characteristics of FASD
can be divided into: (i) growth retardation, (ii) craniofacial abnormalities, and
(iii) central nervous system (CNS) dysfunction. FASD is the most common
cause of nongenetic mental retardation in Western countries. Although the
underlying molecular mechanisms of ethanol neurotoxicity are not completely
determined, the induction of oxidative stress is believed to be one central
process linked to the development of the disease. Currently, there is no
known effective strategy for prevention (other than alcohol avoidance) or
treatment. In the present review we will provide the state of art in the
evidence for the use of antioxidants as a potential therapeutic strategy for the
treatment using whole-embryo and culture cells models of FASD. We
conclude that the imbalance of the intracellular redox state contributes to the
pathogenesis observed in FASD models, and we suggest that antioxidant
therapy can be considered a new efficient strategy to mitigate the effects of
prenatal ethanol exposure.
Birth Defects Research (Part A) 00:000–000, 2014.
V
C2014 Wiley Periodicals, Inc.
Key words: fetal alcohol syndrome; neuroprotective; treatment; oxidative stress;
teratogenesis
Introduction
Alcohol is known to be a teratogen and its consumption
during pregnancy can produce a wide range of adverse
effects in the developing fetus. The severity of fetal dam-
age due to ethanol exposure depends on several factors
which include the timing, pattern, and dose of consump-
tion (Abel and Hannigan, 1995). Maternal ethanol con-
sumption can develop a spectrum of physical, cognitive,
and behavioral disabilities in newborns known as fetal
alcohol spectrum disorder (FASD). The most severe form,
that includes morphological abnormalities is defined as
fetal alcohol syndrome (FAS) (de Sanctis et al., 2011; Joya
et al., 2012; Memo et al., 2013). The classical dysmorphic
facial features of FAS include microcephaly, a rather flat
midface with short palpebral fissures, low nasal bridge
with short nose and long smooth or flat phylum with a
narrow vermilion of the upper lip (de Sanctis et al., 2011).
This disease is also characterized by failure to thrive, that
starts in the prenatal age and persists postnatally, and by
neurocognitive defects (Memo et al., 2013). Using these
criteria, the diagnosis of FAS missed many individuals
without phenotipical diagnostic clues. The term FASD was
not intended to be used as a clinical diagnosis, but an
umbrella containing diagnoses as FAS, partial FAS and
alcohol related neurodevelopmental disorders (ARND).
FASD includes the range of individuals who have from the
full syndrome to only a few issues about learning and
behavior, and no facial or growth signs (May et al., 2010).
Currently, in Europe there are no systematic data on FAS
and FASD prevalence, nor on prenatal exposure to ethanol.
In Canada, the prevalence of FAS and FASD has been
reported to be 1 to 3 and 9 per 1000 live births, respec-
tively, higher than the FAS prevalence observed in the
United States (0.5–2.0 per 1000 live births (Goh et al.,
2008).
It is well known that in adults, ethanol-induced dam-
age is mediated by induction of oxidative stress and its
plays a major role in different mechanisms such in the
case of liver injury (Dey and Cederbaum, 2006). Similarly,
prenatal ethanol exposure has been shown to cause an
increase in oxidative stress in developing organs, including
the brain (Reyes et al., 1993; Heaton et al., 2003). Even a
brief exposure to ethanol, the fetal brain alters its redox
balance (Dong et al., 2010). On the other hand, it is gener-
ally admitted that antioxidants treatment cause the oppo-
site effect (Busby et al., 2002; Neese et al., 2004).
The brain is the principal target tissue of prenatal
ethanol exposure and it possesses the highest oxygen
1
Unitat de Recerca Inf
ancia i Entorn (URIE), Institut Hospital del Mar d’Inves-
tigacions Me
`diques (IMIM), Barcelona, Spain
2
Red de Salud Materno-Infantil y del Desarrollo (SAMID), Programa RETICS,
Instituto Carlos III, Madrid, Spain
3
Departament de Pediatria, Ginecologia i Obstetricia i de Medicina Preventiva,
Universitat Aut
onoma de Barcelona (UAB), Bellaterra, Spain
4
Department of Experimental and Health Sciences, Universitat Pompeu Fabra
(UPF), Parc de Recerca Biomedica de Barcelona, Barcelona, Spain.
This study was supported by Grants from Fondo de Investigaciones Sanitarias
(FIS) (PI10/02593; PI13/01135), from the Instituto Carlos III (Madrid, Spain),
RecerCaixa (OG085818) and Red de Salud Materno-Infantil y del Desarrollo
(SAMID) (RD12/0026/0003) from the Instituto Carlos III (Spain), intramural
funding of the Neuroscience Program at IMIM (Institut Hospital del Mar
d’Investigacions Me
`diques) and partially supported by Generalitat de Catalu-
nya (Spain) AGAUR (2009SGR1388)
*Correspondence to: Oscar Garcia Algar, Unitat de Recerca Inf
ancia i Entorn
(URIE), Institut Hospital del Mar d’Investigacions Me
`diques (IMIM), Parc de
Recerca Biome
`dica de Barcelona (PRBB), C/ Dr. Aiguader 88, 08003, Barce-
lona, Spain. E-mail: 90458@parcdesalutmar.cat
Published online 0 Month 2014 in Wiley Online Library (wileyonlinelibrary.com).
Doi: 10.1002/bdra.23290
V
C2014 Wiley Periodicals, Inc.
metabolic rate in the body because its cells use 20% of
the total oxygen consumed by the organism (Sokoloff,
1999). For this reason, it presents the highest quantity of
reactive oxygen species (ROS) production during oxidative
metabolism. Moreover, the production of ROS can be
increased by the presence of high content of unsaturated
fatty acids that can be substrates for ROS production. Fur-
thermore, the antioxidant defense system of the brain is
limited with respect to other organs. Particularly, the activ-
ities of antioxidant enzymes such superoxide dismutase
(SOD), catalase, and/or glutathione peroxidase (GPx) are
lower (Floyd and Carney, 1992). For all of these reasons
mentioned above, the neural fetal cells are more vulnera-
ble to neurotoxic effects of oxidative stress than the adult
brain cells, because the levels of antioxidant enzymes and
nonenzymatic endogenous antioxidants in the developing
fetus are lower compared with adults (Bergamini et al.,
2004).
Oxidative stress is a general term used to describe an
imbalance between the systemic manifestation of ROS, and
a biological system’s ability to readily detoxify these reac-
tive intermediates or to repair the resulting damage. The
main intracellular source of ROS is the oxidative phospho-
rylation generated by the mitochondria. Moreover, other
enzymes such xanthine oxidase and NADPH oxidases
(NOX/XOX) can produce ROS in the cytoplasm. NOX is a
multi-subunit enzyme complex that is activated and
induced by inflammatory signals (Infanger et al., 2006).
This inflammatory signals include several external sources
such UV light, chemical reagents, cigarette smoke, drugs
and/or ethanol consumption (Zadak et al., 2009). Activa-
tion of glial cells, especially microglia, that release of pro-
inflammatory factors (TNFa,IL-1b, IL-6, etc.) and ROS
have been implicated in several models of neurodegenera-
tion (Lucas et al., 2006; Block and Hong, 2007). ROS react
with cellular molecules including proteins, lipids and DNA
causing genetic alterations (Finkel and Holbrook, 2000)
and finally culminate in cell death (activation of apoptosis
cascades). In humans, oxidative stress is a pathogenic
mechanism involved in the development of cancer (Halli-
well, 2007) Parkinson’s disease (Valko et al., 2007) fragile
X syndrome (de Diego-Otero et al., 2009) or autism (James
et al., 2004).
The organism presents a variety of defense mecha-
nisms that can be referred to as the endogenous antioxi-
dant system (Halliwell and Gutteridge, 1995). Endogenous
antioxidants can inhibit the ROS formation or promote the
free radicals scavenging. These endogenous antioxidants
can be broadly divided into: nonenzymatic and enzymatic
origin. Endogenous nonenzymatic antioxidants include thi-
ols and glutathione (GSH) (Halliwell, 2006). On the other
hand, enzymatic antioxidants include: (1) SOD, (2) cata-
lase, (3) the glutathione system, which encompasses the
enzymes: (3.1) glutathione reductase (GR) and uses GSH
and NADPH as co-factors. (3.2.) glutathione peroxidase
(GPx) reduces hydrogen peroxide and other organic perox-
ides at the expense of GSH, which is in turn oxidized to
form glutathione disulfide (GSSG) and (3.3.) glutathione
S-transferases (GSTs) that catalyze the conjugation of the
reduced form of GSH to xenobiotic substrates for the pur-
pose of detoxification. In the brain, most GST is located in
glial cells (which are also rich in GSH), helping protect
neuronal populations that have a low content of this co-
factor (Astor et al., 1988; Hayes and Strange, 1995; Salinas
and Wong, 1999). Given its extensive functions list, GSH is
probably the most important endogenous nonenzymatic
antioxidant.
It is well known that alcohol produces high levels of
ROS production through its metabolism (Fig. 1). Ethanol is
metabolized to acetaldehyde by the alcohol dehydrogenase
(ADH) in the liver. Alternatively, ethanol can also be
metabolized by cytochrome P450 2E1 present in the liver
and brain. Of interest, the reaction catalyzed by cyto-
chrome P450 2E1 leads to an increase in the generation of
acetaldehyde and hydroxyl radicals in both tissues. Acetal-
dehyde can then be further oxidized into acetate by the
enzyme acetaldehyde dehydrogenase, these reaction
results in an increase in the activity of respiratory chain
and consequently produces ROS.
An increasing body of evidence has postulated the role
of oxidative stress in FASD evidenced by ROS production
in animals and in vitro models (Heaton et al., 2002, 2003;
Smith et al., 2005; Kane et al., 2008; Dong et al., 2010).
These evidences have been related to the damage on: (1)
lipid peroxidation (Henderson et al., 1995; Chen et al.,
1997; Perez et al., 2006), (2) protein peroxidation (Marino
et al., 2004; Shirpoor et al., 2009), and (3) DNA (Chu
et al., 2007; Dong et al., 2010).
The principal difficulty in this scenario is the heteroge-
neous experimental designs available in the literature.
Parameters such: methodology, ethanol exposure period,
peak of blood alcohol concentration (BAC) reached and
the time of analysis, as well as tissues analyzed and
FIGURE 1. Ethanol metabolism and induction of oxidative stress. Main molecu-
lar mechanisms which ethanol causes oxidative stress. ADH, alcohol dehydro-
genase; ALDH, aldehyde dehydrogenase; NAD, nicotinamide adenine
dinucleotide; NADH, reduced form of NAD; ROS, reactive oxygen species.
2 ANTIOXIDANT THERAPIES FOR FAS PREVENTION
markers evaluated, are not standardized due to make com-
parisons between experiments is impossible. Given this
scenario, the present review is focused on the most recent
findings in novel antioxidant therapeutic approach for the
mitigation of prenatal alcohol exposure effects.
Antioxidant Supplementation as a Therapeutic
Intervention for FASD Prevention
Despite the role of oxidative stress in FASD is also con-
firmed by numerous studies showing the beneficial
impact of antioxidant therapy upon prenatal ethanol
exposure effects, there are a great number of experimen-
tal variables (including the mode and period of ethanol
exposure, the antioxidant selected, the time of adminis-
tration and the age of animals at the time of analysis as
well as the neuropathological parameters evaluated). This
fact complicates direct comparison among the studies
published. Thus, to facilitate the following discussion, the
experimental parameters and the major findings in a
wide range of literature studies are summarized in
Tab l e 1 .
EFFECT OF ANTIOXIDANT SUPPLEMENTATION ON THE AMELIORATION
OF BIRTH DEFECTS
The defects associated with FASD are variable and lie
along a continuum spectrum going from the most severe
form, represented by deficiencies in brain growth (reduced
head circumference and/or structural brain anomaly) to
distinct facial features (microcephaly, short palpebral fis-
sures, thin upper lip and/or smooth philtrum) (Jones and
Smith, 1973). Related with this, some in vivo studies have
indicated that antioxidant treatments can prevent or
reduce growth retardation and/or the occurrence of mal-
formations as a consequence of ethanol exposure during
development. Using Xenopus laevis co-treated with vitamin
C, Peng et al. showed a decrease in microencephaly inci-
dence and growth retardation (Peng et al., 2005). Further-
more, Chen et al. administered ethanol to pregnant mice
dams in combination with EUK-134, a synthetic
manganese-porphyrin complexe similar to SOD and cata-
lase. The co-treatment with EUK-134 reduced the inci-
dence of forelimb malformations in the offspring pups
(Chen et al., 2004). The treatment with vitamin E in preg-
nant mice dams treated with ethanol normalized fetal
development (Wentzel et al., 2006). Similarly, black gin-
seng (Panax ginseng) improved most of the morphological
scores in mice embryos (Lee et al., 2009). Another birth
outcome commonly seen in children exposed prenatally to
ethanol are congenital heart defects (Karunamuni et al.,
2014). Using zebrafish embryos as animal model Reimers
et al. evaluated the effect of lipoic acid, vitamin E and Tro-
lox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic
acid, a powerful free radical scavenger). These antioxi-
dants partially attenuated the pericardial edema incidence
(Reimers et al., 2006).
EFFECT OF ANTIOXIDANT SUPPLEMENTATION ON THE
NEUROANATOMICAL ARCHITECTURE
In several cases, antioxidant treatment was also shown to
have a positive impact at the neuroanatomical level. In
this sense, embryos prenatally exposed to ethanol and
treated with vitamin E attenuated the reduction in the
number of Purkinje cells in the lobule I of the cerebellum
(Heaton et al., 2000a). Similar results were obtained by
Lee et al. using black ginseng as a therapeutic approach.
Embryos co-treated with ethanol and black ginseng
showed similar head length compared with the control
group (including fore-, mid-, and hindbrain) (Lee et al.,
2009). Using a Guinean pig model treated with a combina-
tion of high doses of vitamin C and E, protects against the
loss of hippocampal weight (Nash et al., 2007). In accord-
ance with these results, Marino et al. observed an amino-
ration of hippocampal neuronal loss (Marino et al., 2004).
Finally, co-treatment of ethanol-exposed pregnant dams
with silymarin showed to be useful to prevent the
ethanol-induced impairment in corpus callosum develop-
ment (Moreland et al., 2002). On the other hand, not all
the studies reported beneficial results. U83836E, a new
vitamin E derivate generated by Upjohn Company (Kala-
mazoo, MI), did not attenuate neonatal alcohol-induced
microencephaly or Purkinje cell loss in lobule I (Grisel and
Chen, 2005). In fact, it has been reported that the protec-
tive efficacy of U83836E may be dose related, and high
doses of the drug can be cytotoxic (Mertsch et al., 1998).
Anthocyanins, a large subgroup of flavonoids present in
many vegetables and fruits, are safe and potent antioxi-
dants that can cross the blood–brain barrier and be dis-
tributed in the CNS (Passamonti et al., 2005). An example
of anthocyanins is cyanidin-3-glucoside (C3G) obtained
from blackberries. C3G has been demonstrated that
presents a potent antioxidant and anti-tumor capacity
(Ding et al., 2006). Promising results provided by Chen
et al. showed that C3G can ameliorate ethanol-induced
neuronal death blocking GSK3bactivation (Ke et al.,
2011).
The role of antioxidants treatment in relation to behav-
ioral deficits is inconclusive and not all experimental or
clinical studies find beneficial effects. In some cases, the
neuroprotection conferred by antioxidant therapy was
translated into an improvement of the behavioral deficits
and learning abnormalities associated with perinatal etha-
nol exposure (Busby et al., 2002; Vink et al., 2005; Miller
et al., 2013). For example, Busby et al. co-administered
silymarin and ethanol throughout gestation and detected
that silymarin improve several behavioral deficits in the
rat adult offspring (Busby et al., 2002). The co-treatment
with vitamin C and E in pregnant Guinean pigs mitigated
the ethanol-induced deficit in the task-retention compo-
nent of the water-maze activity. However, other study,
using the same vitamin regimen did not mitigate the
ethanol-induced impairment in hippocampal long-term
BIRTH DEFECTS RESEARCH (PART A) 00:00–00 (2014) 3
TABLE 1. Summary of the principal characteristics of novel targets with antioxidant activity for the prevention of FASD
Natural antioxidants
Drug
Experimental design /
Animals used
Range of drug concen-
trations
used / Administration /
Time
Range of EtOH concen-
trations / Administra-
tion / Time Observations Reference
In vivo experiment.
Guinea pigs.
250 mg/kg / PO / GD2
– GD67
9 g/kg/day / PO / GD2
– GD67
- Reduces lipid peroxidation in the liver.
-Enhances the activities of
glutathione peroxidase and reductase.
-Reduces the activity of GGT.
Suresh et al., 1999
In vivo experiment.
Xenopus laevis
0.1 mM / 2 h before
the EtOH treatment
0 – 2% (v/v) / Stage 13
to 22
- Inhibits ROS production.
- Activates NF-kB pathway.
-Prevents microencephaly.
Peng et al., 2005
Vitamin C In vivo experiment.
Guinea pigs
250 mg/day / PO / GD2
– GD67
4 g/kg/day / PO / GD2
– GD67
- Protects hippocampal weight versus brain
weight.
-Does not mitigate impairment of hippocampal
long-term potentiation.
Nash et al., 2007
In vitro experiment.Primary hippocam-
pal neuronal cells from Sprague–
Dawley rats at GD17.5
1 mM / 24 h 100 mM / 24 h - Decreases expression of Bax, caspase-9, cas-
pase-3, cytochrome-c
- Increases expression of Bcl-2
Naseer et al., 2011
EGCG In vitro experiment.Primary fetal rhom-
boencephalic neurons from Sprague-
Dawley rats at GD14.
0.001 mM / 24 h 75 mM / 24 h - Decreases the number of apoptotic cells Antonio and Druse,
2008
In vivo experiment.C57BL/6J mice 200 - 400 mg/kg/day /
PO / GD7 - 8
0.005 20.02 ml/g /
IP / GD8
- Normalizes head sizes.
-Normalizes Otx1 and Sox2 expression levels.
-Decreases H2O2 and MDA.
Long et al., 2010
Ginkgolide B In vitro experiment.PC12 cells 0.001 – 0.050 mM / 4h
before EtOH
treatment
100 mM / 24 h - Decreases caspase-3.
- Decreases ROS production.
-Does not affect ethanol-induced ADH and
CYP2E1 activities.
Zhang et al., 2011
In vitro experiment. Primary fetal rhom-
boencephalic neurons from Sprague-
Dawley rats at GD14.
0.01 mM / 24 h 75 mM / 24 h - Decreases the number of apoptotic cells Antonio and Druse,
2008
4 ANTIOXIDANT THERAPIES FOR FAS PREVENTION
TABLE 1. Continued
Natural antioxidants
Drug
Experimental design /
Animals used
Range of drug concen-
trations
used / Administration /
Time
Range of EtOH concen-
trations / Administra-
tion / Time Observations Reference
Resveratrol In vitro experiment. Primary CGNs from
Long-Evans rats at PD7
2 – 100 mg/kg / 1
– 24 h before EtOH
treatment
BAC: 80 mM / 5 h - Decreases ROS levels.
-Restores the expression levels of Nrf2 in the
nucleus.
-Retains the expression and activity of NADPH
quinine oxidoreductase 1 and SOD.
Kumar et al., 2011
In vitro experiment. Primary DRG
neurons from Wistar rats at GD15.
0.0001 – 0.030 mM /
24 h
325.6 mM / 24 h - Increases the number of extended nerve fibers
and neurons that migrated from the DRG
explants.
-Inhibits EtOH-induced apoptosis.
-Recovers SOD and GSH expression.
Yuan et al., 2013b
In vitro experiment.Primary SCs from
Wistar rats at PD3.
30 mM / 96 h 1500 mg/dl / 96 h - Recovers cell viability.
-Increases the BDNF and GDNF expression.
Yuan et al., 2013a
Curcumin In vitro experiment.Primary fetal
rhomboencephalic neurons from
Sprague–Dawley rats at GD14.
0.001 mM / 24 h 75 mM / 24 h - Decreases the number of apoptotic cells. Antonio and Druse,
2008
In vivo experiment.Wistar rats PD5 30 – 60 mg/kg / PO /
PD6 – 28
5 g/kg / PO / PD7 – 9 - Ameliorates neuroinflamation (oxidative nitro-
sative stress, TNF-a, IL-1b, and TGF-b1).
-Decreases neuronal apoptosis (NF-jb and cas-
pase 3) in both cerebral cortex and
hippocampus.
Tiwari and Chopra,
2012
Melatonin In vivo experiment.Sprague-Dawley rats 20 mg/kg/day / PD4 – 9 6 g/kg/day / PD4 – 9 - Does not decrease the apoptotic Purkinje cell
number.
Grisel and Chen,
2005
In vitro experiment.Primary
rhomboencephalic neurons from
Sprague–Dawley rats at GD14.
0.001 mM / 24 h 75 mM / 24 h - Decreases the number of apoptotic cells. Antonio and Druse,
2008
Thymoquinone In vitro experiment.Primary cortical neu-
rons from Sprague-Dawley rats at
GD17.5
0.01 – 0.035 mM /
12 h
100 mM / 12 h - Inhibits apoptotic events (increasing Bcl-2
expression)- Reduces the cleavage of PARP-1.
Ullah et al., 2012
BIRTH DEFECTS RESEARCH (PART A) 00:00–00 (2014) 5
TABLE 1. Continued
Natural antioxidants
Drug
Experimental design /
Animals used
Range of drug concen-
trations
used / Administration /
Time
Range of EtOH concen-
trations / Administra-
tion / Time Observations Reference
Sulforaphane In vitro experiment.NCC (JoMa1.3 cells) 2.5310
24
–4310
23
mM/24h–72h
50 – 200 mM /
24 – 72 h
- Increases Nrf2 activation and activates the
downstream expression of endogenous
antioxidants.
Chen et al., 2013
Capsaicin In vitro experiment.Whole embryo cul-
ture of Sprague–Dawley rats at
GD8.5
10
28
–10
27
lg/ml / 17 h 1 lg/ml / 17 h - Recovers SOD and GSH activity. Kim et al., 2008
Black ginseng In vitro experiment.Culture ICR rat
embryo from GD8.5
0 - 100 mg/ml / 48 h 0.017 mM / 48 h - Normalizes morphological scores (including
head length, fore-, mid- and hindbrain).
Lee et al., 2009
In vitro experiment.Primary embryonic
hippocampus from Long-Evans rats
at GD18
0.05 mM / 2 h or 16 h 0 – 346.6 mM /
2 h or 16 h
- Protects neuronal viability of embryonic hippo-
campal cultures against ethanol.
Mitchell et al.,
1999a,b
In vivo experiment.Long-Evans rats. 30 - 60 IU/100 ml /
IG / PD4 – 5
12% / IG / PD4 – 5 - Prevents the loss of Purkinje cells. Heaton et al.,
2000a,b
In vitro experiment.Primary CGN from
Long-Evans rats at PD8.
0.050 mM / 24 h 86.7 – 346.6 mM / 24 h - Restores the expression of NTFs (BDNF and
neurotrophin-3).
-Diminishes the cellular disturbances in oxida-
tive processes.
Heaton et al., 2004
In vivo experiment.Long-Evans rats 2000 g/kg / PD6 5.25 g/kg / PD7 – 9 - Alleviates the increase in protein carbonyls.
-Does not improve spatial learning in the
ethanol-exposed animals.
Marino et al., 2004
In vivo experiment.Long-Evans rats. 12.26 mg/kg/day /
PD4 – 9
2.625 g/kg/day /
PD4 – 9
- Fails to protect against reduction of cerebellar
Purkinje cells.
Tran et al., 2005
In vitro experiment.Primary CGN from
Long-Evans rats at PD9.
0.050 mM / 24 h 86.7 – 346.6 mM /
24 h
- Amplifies the g-GCS and total GSH protein
expression levels.
Siler-Marsiglio et al.,
2005
Vitamin E In vivo experiment. Zebrafish embryos. Data not shown /
3 – 24hpf
200 mM / 3 – 24 hpf - Attenuates the incidence of pericardial edema.
-Does not provide protection against cell death.
Reimers et al., 2006
6 ANTIOXIDANT THERAPIES FOR FAS PREVENTION
TABLE 1. Continued
Natural antioxidants
Drug
Experimental design /
Animals used
Range of drug concen-
trations
used / Administration /
Time
Range of EtOH concen-
trations / Administra-
tion / Time Observations Reference
In vivo experiment. Sprague–Dawley
rats
5% / GD1 – 20 20% / GD1 – 20 - Does not affect BAC.
-Normalizes fetal development.
-Normalizes fetal hepatic levels of 8-iso-
PGF2a.
Wentzel et al., 2006
In vivo experiment. Guinea pigs. 100 – 250 mg/day / PO
/ GD2 – 67
4 g/kg/day / PO / GD2
–67
- Protects hippocampal weight relative to brain
weight.
-Does not mitigate the EtOH-induced impair-
ment of hippocampal long-term potentiation.
Nash et al., 2007
In vivo experiment. Wistar rats. 300 mg/day / SC / GD7
– PD21
4.5 g/kg/day / SC / GD7
– PD21
- Decreases DNA damage.
-Restores the elevated level of Hcy to control
levels.
Shirpoor et al., 2009
In vitro experiment. Primary CGNs from
rats at PD8.
0.050 mM / 24 h 86.6 mM / 24 h - Reduces Bax translocation.
-Decreases ROS production.
Heaton et al., 2011
In vitro experiment. Chicken embryos 50 mM 15 – 50% - Diminished mortality and growth retardation. Satiroglu-Tufan and
Tufan, 2004
Silymarin In vivo experiment. Fisher/344 rats 400 mg/kg / OA BAC: 6.7% / OA - Amelioration of the effects upon the develop-
ing fetal rat brain.
Moreland et al., 2002
C3G In vitro experiment. Neuro2a cells 5mM 87 mM - Restores the neurite outgrowt h Chen et al., 2009
In vivo experiment. C57BL/6 mice 10 – 30 mg/kg / IP 2.5 g/kg / SCPD7 - Reduces EtOH-meditated caspase-3 activation
in the cerebral cortex blocking GSK3b
Ke et al., 2011
Synthetic antioxidants
U83836E In vivo experiment. Sprague-Dawley rats 20 mg/kg/day / PD4 – 9 6 g/kg/day / PD4 – 9 - Melatonin does not decrease the Purkinje cell
number.
-Does not change BAC measured on PD 6.
Grisel and Chen,
2005
Trolox In vivo experiment. Zebrafish embryos. Data not shown / 3 –
24hpf
200 mM / 3 – 24hpf - Attenuated the incidence of pericardial
edema.
-Does not provided protection against cell
death.
Reimers et al., 2006
BIRTH DEFECTS RESEARCH (PART A) 00:00–00 (2014) 7
TABLE 1. Continued
Natural antioxidants
Drug
Experimental design /
Animals used
Range of drug concen-
trations
used / Administration /
Time
Range of EtOH concen-
trations / Administra-
tion / Time Observations Reference
In vivo experiment. Zebrafish embryos. 0.1 mM / 3 – 24hpf 200 mM / 3 – 24 hpf - Attenuates the incidence of pericardial edema.
-Does not provided protection against cell
death.
Reimers et al., 2006
Alpha lipoic acid In vitro experiment. Primary rhomboen-
cephalic neurons from Sprague–
Dawley rats at GD14
0.01 mM / 24 h 75 mM / 24 h - Decreases the number of apoptotic cells. Antonio and Druse,
2008
In vivo experiment. Wistar rats. 100 mg/kg / IP / GD7 –
PD21
4.5 g/kg / SC / GD7 –
PD21
- Decreases DNA damage.
-Restores the elevated protein carbonyl and
lipid hydroperoxide levels.
Shirpoor et al., 2008
Pycnogenol In vitro experiment. Primary CGNs from
Long-Evans rats at PD9
25 - 100 mg/ml / 5s –
24 h
86.7 – 346.6 mM / 5s
–24h
- Decreases cell death and reduces the activa-
tion of caspase-3.
Siler-Marsiglio et al.,
2004
tBHQ In vitro experiment. Primary NCC from
C57BL/6J mice from GD10.5
0.010 mM / 16h before
EtOH treatment.
100 mM / 24 h - tBHQ alone increases the protein expression
of Nrf2 and its downstream antioxidants.
-tBHQ-mediated antioxidant response pre-
vents oxidative stress andapoptosis.
Yan et al., 2010
Diphenylene iodo-
nium (DPI)
In vivo experiment. C57BL/6J mice 4 mg/kg / IP / GD9 2.9 g/kg / IPA / GD9 - DPI prevented ethanol-induced increases NOX
enzyme activity, ROS generation and oxida-
tive DNA damage.
-DPI reduces caspase-3 activation and
diminished prevalence of apoptosis.
Dong et al., 2010
D3T In vivo experiment. C57BL/6J mice 5 mg/kg / IP / GD8 2.9 g/kg / IP / GD8 - D3T increases Nrf2 protein levels and Nrf2-
ARE binding, and strongly induces the
mRNA expression of Nrf2 downstream target
genes.
-D3T decreases the levels of ROS.
Dong et al., 2008
In vitro experiment. PC12 cells. 0.05 mM / 16h before
EtOH
200 mM / 24 h - D3T treatment reduces ethanol-induced apo-
ptosis stabilizing Nrf2.
Dong et al., 2011
8 ANTIOXIDANT THERAPIES FOR FAS PREVENTION
potentiation (Nash et al., 2007). Overall, the results of
these studies indicate that maternal administration of
high-dose vitamins C plus E throughout gestation has lim-
ited efficacy and potential adverse effects (such low birth
weight) as a therapeutic intervention (Poston et al., 2006).
Furthermore, a recent prospective observational study
conducted by a Canadian work team in pregnant women
supplementing with mega-doses of vitamin E, detected an
apparent decrease in mean birth weight that could not be
explained by other variables including maternal age, gesta-
tional (Boskovic et al., 2005). In light of the results, the
EViCE (Effectiveness of Vitamin C and E in alcohol exposed
pregnancies) study was suspended (Goh et al., 2007).
EFFECT OF ANTIOXIDANT THERAPY ON THE ENDOGENOUS OXIDATIVE
STRESS LEVELS
The use of compounds with antioxidant properties has
also been consistently shown to reduce oxidative stress
levels and/or to increase the endogenous antioxidant
capacity in the rodent brains of different models of FASD.
The antioxidant vitamin C inhibited ROS production in
Xenopus laevis embryos exposed to ethanol (Peng et al.,
2005). Vitamin E is the natural antioxidant most com-
monly used and several studies have also shown its bene-
ficial effects in decreasing oxidative stress in different
models of FASD. Recently, in offspring rat pups exposed
prenatally to ethanol, vitamin E reversed the levels of pro-
tein and lipid oxidation in both hippocampus and cerebel-
lum (Shirpoor et al., 2009). In the same tissue, using rat
pups vitamin E, alleviated oxidative stress (Marino et al.,
2004). Similarly, maternal vitamin E treatment restores
the fetal hepatic isoprostanes (Wentzel et al., 2006). (–)-
Epigallocatechin-3-gallate (EGCG) is another powerful anti-
oxidant and is believed to be responsible for most of the
health benefits attributed to green tea consumption (Nagle
et al., 2006). Long et al., using a FASD murine model found
that EGCG provided significant protection against ethanol-
associated embryonic developmental retardation. This pro-
tection seems to be mediated by its antioxidative proper-
ties (Long et al., 2010). Resveratrol (3,5,40-trihydroxy-
trans-stilbene) has been shown to be a promising natural
compound with antiapoptotic, free radical-scavenging, and
antilipoprotein peroxidation properties (Shakibaei et al.,
2009). Using a mice model of FASD, the treatment with
resveratrol, before ethanol exposure, restores nuclear fac-
tor (erythroid-derived 2)-like 2 (Nrf2) transcription factor
levels in cerebellum granule neurons (CGNs) and in the
same tissue, and this fact promotes the survival of these
cells (Kumar et al., 2011). Nrf2 has been demonstrated to
be a critical transcription factor that regulates the induc-
tion of phase 2 antioxidant enzymes detoxifying and anti-
oxidant genes (Zhang, 2006; Nguyen et al., 2009).
Thymoqinone (TQ), the active component of Nigella sativa
seeds, has broad and versatile pharmacological effects.
These effects include strong antioxidant activity against
free radical-generating agents (Houghton et al., 1995). TQ
stimulates resistance to oxidative stress decreasing the
elevated levels of malondialdehyde (MDA), and stimulating
catalase and SOD expression (Al-Majed et al., 2006). Sul-
foraphane (SFN) is a natural isothiocyanate, found abun-
dantly in broccoli sprouts. Compelling evidence indicates
that SFN-rich broccoli sprouts and other SFN food sources
trigger the induction of phase 2 detoxifying genes and
antioxidant enzymes, through activation of Nrf2 signaling,
and can aid in preventing cancer and other diseases (Din-
kova-Kostova, 2002). Chen et al. (2013) showed the Nrf2-
mediated antioxidant response on neural crest cells
(NCCs) exposed to ethanol. Capsaicin (8-methyl-N-vanillyl-
6-nonemide) is the major pungent principle of hot peppers
of the plant genus Capsicum. Kim et al. treated with capsa-
icin embryos exposed prenatally to ethanol. These animals
recovered their SOD activity and GPx and GPx mRNAs
expression (Kim et al., 2008). Lipoic acid and its reduced
form, dihydrolipoic acid (DHLA) eliminate hydroxyl radi-
cals and hypochlorous acid with a potency comparable to
GSH (Biewenga and Bast, 1995). Tert-butylhydroquinone
(tBHQ) increases Nrf2 protein stability through inhibition
of the Keap1-mediated ubiquitination. An in vitro model
that used NCCs co-exposed to ethanol and tBHQ showed
less oxidative stress and apoptosis (Yan et al., 2010).
Diphenylene Iodonium (DPI) is a NOX inhibitor. NOX
enzymes can catalyze NADPH-dependent reduction of oxy-
gen to generate superoxide anion (Banfi et al., 2003) and
interestingly, ethanol activates NOX and the subsequent
ROS generation (Wang et al., 2012). Dong et al. (2010)
examined the effect of co-administration of DPI with etha-
nol on pregnant mouse. The results support the hypothe-
sis that DPI is a promising molecular target for blocking
NOX, a critical source of ROS in ethanol-exposed embryos.
3H-1,2 dithiole-3-thione (D3T) is a potent cancer
chemopreventive agent that prevents mutation and pro-
vides protection against neoplasia initiation (Otieno et al.,
2000). In addition, activation of the Nrf2 pathway, by oral
administration of D3T, has recently been reported to con-
fer partial protection against 1-methyl-4-phenyl-1,2,3,6-
tetrahydropyridine (MPTP)-induced neurotoxicity (Burton
et al., 2006). The protective effects of D3T in animals have
been associated with induction of the detoxifying and anti-
oxidant enzymes SOD, catalase and c-glutamylcysteine syn-
thetase (c-GCS) (Otieno et al., 2000; Munday and Munday,
2004; Cao et al., 2006). Dong et al. (2008) exposed mice
embryos to D3T decreasing ROS generation.
PROTECTIVE EFFECT OF ANTIOXIDANTS USING IN VITRO MODELS
The beneficial role of antioxidants have been corroborated
by several in vitro studies (Mitchell et al., 1999a ; Heaton
et al., 2000a; Lee et al., 2009). A prospective apoptotic
effect has been described for vitamin C. Using primary-
cultured neuronal cells co-treated with Vitamin C and
ethanol the expression of Bax, caspase-9, caspase-3 and
BIRTH DEFECTS RESEARCH (PART A) 00:00–00 (2014) 9
cytochrome-c decreased while the expression of antiapop-
totic Bcl-2 protein increased significantly (Naseer et al.,
2011). Furthermore, vitamin E has been extensively dem-
onstrated that prevents alcohol-induced cell loss using in
vitro models (Mitchell et al., 1999a; Heaton et al., 2000a;
Siler-Marsiglio et al., 2004, 2005). For example, neuronal
viability was significantly higher in cell cultures previously
treated with ethanol and maintained on vitamin E or
b-carotene (Mitchell et al., 1999b). A recent study has
demonstrated that EGCG and resveratrol could protect
fetal rhomboencephalic neurons from ethanol-induced
apoptosis (Antonio and Druse, 2008). In agreement with
that, resveratrol prevents ethanol-induced apoptosis dur-
ing mouse blastocyst development (Huang et al., 2007).
Ginkgolide B (GB), originally extracted from Ginkgo biloba
leaves, is one of the major components of traditional Chi-
nese medicine (Maclennan et al., 2002). It has been shown
that GB can scavenge free radicals and inhibit seryl and
aspartyl proteases (Brunetti et al., 2006), protecting
against neural damage. Preliminary in vitro results have
demonstrated the powerful antioxidant characteristics of
GB inhibiting ethanol-induced cell apoptosis (Zhang et al.,
2011). Another antioxidant with natural origin is curcu-
min, the principal curcuminoid found in turmeric. Curcu-
min has potent antiamyloid (Wang et al., 2010) anti-
ischemic (Shukla et al., 2008) and anti-inflammatory prop-
erties (Basnet and Skalko-Basnet, 2011). All of these char-
acteristics seem to be mediated by its pharmacological
actions with respect to its antioxidant effect. As a conse-
quence, curcumin presents protective effects against
ethanol-induced apoptosis. This was initially observed
using primary fetal rhomboencephalic neurons (Antonio
and Druse, 2008) and postnatal pup rats (Tiwari and Cho-
pra, 2013). Recently, it has been reported that TQ, by
means of direct reduction of intracellular ROS, protects
against cell death induced by serum/glucose deprivation
in PC12 cells by means of a direct reduction in intracellu-
lar ROS (Mousavi et al., 2010). Moreover, TQ maintained
normal physiological mitochondrial transmembrane poten-
tial. These findings suggest that TQ is a potential protec-
tive agent against ethanol-induced neuronal apoptosis. All
of these results strongly support the idea that an increase
in oxidative stress is one of the mechanisms by which
ethanol induces apoptotic cell death in fetal neurons.
Finally, with respect to the action of anthocyanins, Chen
et al. demonstrated that C3G can recover the reduction of
neurite outgrowth caused by ethanol treatment. Moreover,
this process is mediated by glycogen synthase kinase 3b
(GSK3b) (Chen et al., 2009).
The alterations observed in these systems occur as a
consequence of oxidative stress but the intermediate
mechanisms are already unknown and depend on the
magnitude, pattern and timing of the exposure (as certain
as the genetic susceptibilities to ethanol also exert an
influence on the dose and period of exposure). Thus, it is
possible that oxidative stress only represents a single
molecular process involved in ethanol-induced damage.
Consequently, treatment with antioxidants might not be
enough to counter act the effects of perinatal ethanol
exposure.
Nevertheless, antioxidants (alone or in combination
with other therapeutic agents) might still be good candi-
dates for the mitigation of some of the deficits observed in
individuals with FASD. Further studies in animal models
are warranted to identify the optimal cocktail of antioxi-
dant compounds in addition to test therapeutic strategies
that use antioxidants in combination with other pharmaco-
logical drugs.
CONCLUSIONS
FASD is a major public health problem, being the leading
cause of preventable mental retardation and birth defects
in the Western countries (May et al., 2009). The simplest
method for the prevention of FASD is avoiding any alcohol
intake during pregnancy. However, a widespread and appa-
rently increasing incidence of FASD has been observed
recently (Fig. 2) (Abel, 2006; Riley et al., 2011). Whereas a
great effort should be made to avoid ethanol consumption,
several pharmacological approaches for the prevention of
FASD are currently under active research and some of
them have already generated patents (Martinez and Egea,
2007). Identification of effective interventions and treat-
ments for FASD is, therefore, critical. Ideally, one would
intervene at the time of alcohol exposure, thereby directly
preventing or reducing the amount of alcohol-related dam-
age. Based on the mechanisms involved in the ethanol-
induced damage include neurotrophic agents (Heaton
et al., 2000b), neuroactive peptides (Vink et al., 2005) and
antioxidants. Nutritional supplementation may also miti-
gate alcohol’s teratogenic effects. Nutritional supplements
may compensate for changes in the bioavailability of
FIGURE 2. Current strategies for the prevention and treatment of FASD.
Extracted from Martinez and Egea (2007).
10 ANTIOXIDANT THERAPIES FOR FAS PREVENTION
nutrients due to alcohol metabolism (Lieber, 2000). Chol-
ine supplementation during early postnatal development
reduces the severity of some ethanol-induced neurobeha-
vioral alterations (Thomas et al., 2000, 2009, 2010). Simi-
larly, folic acid (FA) supplementation in young women can
prevent intrauterine growth restriction, neural tube
defects and other congenital anomalies (Eskes, 1997;
Scholl and Johnson, 2000). FA can also ameliorate toxicity
induced by ethanol (Gutierrez et al., 2007; Yanaguita et al.,
2008).
Nevertheless, it is well-known that oxidative stress
plays a pivotal role in the development of the disease.
This increase in the levels of ROS production has direct
consequences on the ethanol metabolism due to its actions
on mitochondrial bioenergetics and in the antioxidant sys-
tem. Future research is warranted to test these
hypotheses.
Different animal models have been used for the study
of FASD but the knowledge about the cellular and molecu-
lar processes are not completely understood. Given the
use of different modes of ethanol administration and dif-
ferent exposure periods, to make comparisons among
studies is a challenging issue and drawing clear conclu-
sions may be difficult to understand though the compari-
son between studies. For this reason, it will be necessary
uniform methodologies (including the same model and
controlling external confusing variables such as dose and
administration procedure, BAC peak achieved, time and/or
duration of exposure) to analyze the effect of prenatal
ethanol exposure on different indicators of oxidative stress
in a systematic manner.
Furthermore, it would be interesting to explore
whether the use of antioxidants later on in life would also
have beneficial effects in FASD models. To date, there is
only one clinical study, showing no significant differences
in the urine levels of lipid peroxidation products in
women who drunk during pregnancy compared with non-
drinkers pregnant women (Signore et al., 2008). It is
important to mention that this study did not evaluate the
oxidative stress levels in the newborns. This is particularly
important because, to date, most studies have only ana-
lyzed the effects of antioxidant compounds in models of
FASD when these are administered concurrently with etha-
nol. The majority of these publications evidence that anti-
oxidant treatment can be beneficial in the amelioration of
some characteristics of FASD (Table 1). However, this strat-
egy has not been explored on humans. Nevertheless,
remains to be clear if the antioxidant therapy can be bene-
ficial in the amelioration of some biochemical and behav-
ioral characteristics in children with FASD. However, for
other neurodevelopmental disorders such as autism (Akins
et al., 2010), attention deficit/hyperactivity disorder
(ADHD) (Chovanova et al., 2006) or fragile-X syndrome
(de Diego-Otero et al., 2009), have been explored the
administration of antioxidants showing beneficial effects in
the mitigation of these disease effects. Within this sce-
nario, antioxidants (either alone or in combination with
other therapies) are strong candidates for clinical trials
design in FASD-affected children to prevent or to revert
deleterious effects of ethanol during neurodevelopment.
Acknowledgment
The authors thank J. Klein for excellent language editing
service.
References
Abel EL. 2006. Fetal alcohol syndrome: a cautionary note. Curr
Pharm Des 12:1521–1529.
Abel EL, Hannigan JH. 1995. Maternal risk factors in fetal alcohol
syndrome: provocative and permissive influences. Neurotoxicol
Teratol 17:445–462.
Akins RS, Angkustsiri K, Hansen RL. 2010. Complementary and
alternative medicine in autism: an evidence-based approach to
negotiating safe and efficacious interventions with families. Neu-
rotherapeutics 7:307–319.
Al-Majed AA, Al-Omar FA, Nagi MN. 2006. Neuroprotective effects
of thymoquinone against transient forebrain ischemia in the rat
hippocampus. Eur J Pharmacol 543:40–47.
Antonio AM, Druse MJ. 2008. Antioxidants prevent ethanol-
associated apoptosis in fetal rhombencephalic neurons. Brain
Res 1204:16–23.
Astor MB, Anderson ME, Meister A. 1988. Relationship between
intracellular GSH levels and hypoxic cell radiosensitivity. Pharma-
col Ther 39:115–121.
Banfi B, Clark RA, Steger K, Krause KH. 2003. Two novel proteins
activate superoxide generation by the NADPH oxidase NOX1.
J Biol Chem 278:3510–3513.
Basnet P, Skalko-Basnet N. 2011. Curcumin: an anti-inflammatory
molecule from a curry spice on the path to cancer treatment.
Molecules 16:4567–4598.
Bergamini CM, Gambetti S, Dondi A, Cervellati C. 2004. Oxygen,
reactive oxygen species and tissue damage. Curr Pharm Des 10:
1611–1626.
Biewenga GP, Bast A. 1995. Reaction of lipoic acid with ebselen
and hypochlorous acid. Methods Enzymol 251:303–314.
Block ML, Hong JS. 2007. Chronic microglial activation and pro-
gressive dopaminergic neurotoxicity. Biochem Soc Trans 35(Pt
5):1127–1132.
Boskovic R, Gargaun L, Oren D, et al. 2005. Pregnancy outcome
following high doses of Vitamin E supplementation. Reprod Toxi-
col 20:85–88.
Brunetti L, Orlando G, Menghini L, et al. 2006. Ginkgo biloba leaf
extract reverses amyloid beta-peptide-induced isoprostane pro-
duction in rat brain in vitro. Planta Med 72:1296–1299.
BIRTH DEFECTS RESEARCH (PART A) 00:00–00 (2014) 11
Burton NC, Kensler TW, Guilarte TR. 2006. In vivo modulation of
the Parkinsonian phenotype by Nrf2. Neurotoxicology 27:1094–
1100.
Busby A, La Grange L, Edwards J, King J. 2002. The use of a sily-
marin/phospholipid compound as a fetoprotectant from ethanol-
induced behavioral deficits. J Herb Pharmacother 2:39–47.
Cao Z, Zhu H, Zhang L, et al. 2006. Antioxidants and phase 2
enzymes in cardiomyocytes: chemical inducibility and chemopro-
tection against oxidant and simulated ischemia-reperfusion
injury. Exp Biol Med (Maywood) 231:1353–1364.
Chen G, Bower KA, Xu M, et al. 2009. Cyanidin-3-glucoside
reverses ethanol-induced inhibition of neurite outgrowth: role of
glycogen synthase kinase 3 Beta. Neurotox Res 15:321–331.
Chen JJ, Schenker S, Henderson GI. 1997. 4-hydroxynonenal lev-
els are enhanced in fetal liver mitochondria by in utero ethanol
exposure. Hepatology 25:142–147.
Chen SY, Dehart DB, Sulik KK. 2004. Protection from ethanol-
induced limb malformations by the superoxide dismutase/cata-
lase mimetic, EUK-134. FASEB J 18:1234–1236.
Chen X, Liu J, Chen SY. 2013. Sulforaphane protects against
ethanol-induced oxidative stress and apoptosis in neural crest
cells by the induction of Nrf2-mediated antioxidant response. Br
J Pharmacol 169:437–448.
Chovanova Z, Muchova J, Sivonova M, et al. 2006. Effect of poly-
phenolic extract, Pycnogenol, on the level of 8-oxoguanine in
children suffering from attention deficit/hyperactivity disorder.
Free Radic Res 40:1003–1010.
Chu J, Tong M, de la Monte SM. 2007. Chronic ethanol exposure
causes mitochondrial dysfunction and oxidative stress in imma-
ture central nervous system neurons. Acta Neuropathol 113:
659–673.
de Diego-Otero Y, Romero-Zerbo Y, el Bekay R, et al. 2009.
Alpha-tocopherol protects against oxidative stress in the fragile X
knockout mouse: an experimental therapeutic approach for the
Fmr1 deficiency. Neuropsychopharmacology 34:1011–1026.
de Sanctis L, Memo L, Pichini S, et al. 2011. Fetal alcohol syn-
drome: new perspectives for an ancient and underestimated
problem. J Matern Fetal Neonatal Med 24(Suppl 1):34–37.
Dey A, Cederbaum AI. 2006. Alcohol and oxidative liver injury.
Hepatology 43(Suppl 1):S63–S74.
Ding M, Feng R, Wang SY, et al. 2006. Cyanidin-3-glucoside, a
natural product derived from blackberry, exhibits chemopreven-
tive and chemotherapeutic activity. J Biol Chem 281:17359–
17368.
Dinkova-Kostova AT. 2002. Protection against cancer by plant
phenylpropenoids: induction of mammalian anticarcinogenic
enzymes. Mini Rev Med Chem 2:595–610.
Dong J, Sulik KK, Chen SY. 2008. Nrf2-mediated transcriptional
induction of antioxidant response in mouse embryos exposed to
ethanol in vivo: implications for the prevention of fetal alcohol
spectrum disorders. Antioxid Redox Signal 10:2023–2033.
Dong J, Sulik KK, Chen SY. 2010. The role of NOX enzymes in
ethanol-induced oxidative stress and apoptosis in mouse
embryos. Toxicol Lett 193:94–100.
Dong J, Yan D, Chen SY. 2011. Stabilization of Nrf2 protein by
D3T provides protection against ethanol-induced apoptosis in
PC12 cells. PLoS One 6:e16845.
Eskes TK. 1997. Folates and the fetus. Eur J Obstet Gynecol
Reprod Biol 71:105–111.
Finkel T, Holbrook NJ. 2000. Oxidants, oxidative stress and the
biology of ageing. Nature 408:239–247.
Floyd RA, Carney JM. 1992. Free radical damage to protein and
DNA: mechanisms involved and relevant observations on brain
undergoing oxidative stress. Ann Neurol 32(Suppl):S22–S27.
Goh YI, Chudley AE, Clarren SK, et al. 2008. Development of
Canadian screening tools for fetal alcohol spectrum disorder. Can
J Clin Pharmacol 15:e344–e366.
Goh YI, Ungar W, Rovet J, Koren G. 2007. Mega-dose vitamin C
and E in preventing FASD: the decision to terminate the study
prematurely. J FAS Int 5:1–3.
Grisel JJ, Chen WJ. 2005. Antioxidant pretreatment does not
ameliorate alcohol-induced Purkinje cell loss in the developing
rat cerebellum. Alcohol Clin Exp Res 29:1223–1229.
Gutierrez CM, Ribeiro CN, de Lima GA, et al. 2007. An experimental
study on the effects of ethanol and folic acid deficiency, alone or in
combination, on pregnant Swiss mice. Pathology 39:495–503.
Halliwell B. 2006. Reactive species and antioxidants. Redox biol-
ogy is a fundamental theme of aerobic life. Plant Physiol 141:
312–322.
Halliwell B. 2007. Oxidative stress and cancer: have we moved
forward? Biochem J 401:1–11.
Halliwell B, Gutteridge JM. 1995. The definition and measure-
ment of antioxidants in biological systems. Free Radic Biol Med
18:125–126.
Hayes JD, Strange RC. 1995. Potential contribution of the gluta-
thione S-transferase supergene family to resistance to oxidative
stress. Free Radic Res 22:193–207.
Heaton MB, Madorsky I, Paiva M, Siler-Marsiglio KI. 2004. Vita-
min E amelioration of ethanol neurotoxicity involves modulation
of apoptosis-related protein levels in neonatal rat cerebellar
granule cells. Brain Res Dev Brain Res 150:117–124.
Heaton MB, Mitchell JJ, Paiva M. 2000a. Amelioration of ethanol-
induced neurotoxicity in the neonatal rat central nervous system
by antioxidant therapy. Alcohol Clin Exp Res 24:512–518.
Heaton MB, Mitchell JJ, Paiva M, Walker DW. 2000b. Ethanol-
induced alterations in the expression of neurotrophic factors in
12 ANTIOXIDANT THERAPIES FOR FAS PREVENTION
the developing rat central nervous system. Brain Res Dev Brain
Res 121:97–107.
Heaton MB, Paiva M, Madorsky I, et al. 2003. Effects of ethanol
on neurotrophic factors, apoptosis-related proteins, endogenous
antioxidants, and reactive oxygen species in neonatal striatum:
relationship to periods of vulnerability. Brain Res Dev Brain Res
140:237–252.
Heaton MB, Paiva M, Mayer J, Miller R. 2002. Ethanol-mediated
generation of reactive oxygen species in developing rat cerebel-
lum. Neurosci Lett 334:83–86.
Henderson GI, Devi BG, Perez A, Schenker S. 1995. In utero etha-
nol exposure elicits oxidative stress in the rat fetus. Alcohol Clin
Exp Res 19:714–720.
Houghton PJ, Zarka R, de las Heras B, Hoult JR. 1995. Fixed oil
of Nigella sativa and derived thymoquinone inhibit eicosanoid
generation in leukocytes and membrane lipid peroxidation.
Planta Med 61:33–36.
Huang LH, Shiao NH, Hsuuw YD, Chan WH. 2007. Protective
effects of resveratrol on ethanol-induced apoptosis in embryonic
stem cells and disruption of embryonic development in mouse
blastocysts. Toxicology 242:109–122.
Infanger DW, Sharma RV, Davisson RL. 2006. NADPH oxidases of
the brain: distribution, regulation, and function. Antioxid Redox
Signal 8:1583–1596.
James SJ, Cutler P, Melnyk S, et al. 2004. Metabolic biomarkers of
increased oxidative stress and impaired methylation capacity in
children with autism. Am J Clin Nutr 80:1611–1617.
Jones KL, Smith DW. 1973. Recognition of the fetal alcohol syn-
drome in early infancy. Lancet 302:999–1001.
Joya X, Friguls B, Ortigosa S, et al. 2012. Determination of
maternal-fetal biomarkers of prenatal exposure to ethanol: a
review. J Pharm Biomed Anal 69:209–222.
Kane CJ, Chang JY, Roberson PK, et al. 2008. Ethanol exposure of
neonatal rats does not increase biomarkers of oxidative stress in
isolated cerebellar granule neurons. Alcohol 42:29–36.
Karunamuni G, Gu S, Doughman YQ, et al. 2014. Ethanol expo-
sure alters early cardiac function in the looping heart: a mecha-
nism for congenital heart defects? Am J Physiol Heart Circ
Physiol 306:H414–H421.
Ke Z, Liu Y, Wang X, et al. 2011. Cyanidin-3-glucoside ameliorates
ethanol neurotoxicity in the developing brain. J Neurosci Res 89:
1676–1684.
Kim MR, Lee KN, Yon JM, et al. 2008. Capsaicin prevents ethanol-
induced teratogenicity in cultured mouse whole embryos. Reprod
Toxicol 26:292–297.
Kumar A, Singh CK, Lavoie HA, et al. 2011. Resveratrol restores
Nrf2 level and prevents ethanol-induced toxic effects in the cere-
bellum of a rodent model of fetal alcohol spectrum disorders.
Mol Pharmacol 80:446–457.
Lee SR, Kim MR, Yon JM, et al. 2009. Black ginseng inhibits
ethanol-induced teratogenesis in cultured mouse embryos
through its effects on antioxidant activity. Toxicol In Vitro 23:47–
52.
Lieber CS. 2000. ALCOHOL: its metabolism and interaction with
nutrients. Annu Rev Nutr 20:395–430.
Long L, Li Y, Wang YD, et al. 2010. The preventive effect of oral
EGCG in a fetal alcohol spectrum disorder mouse model. Alcohol
Clin Exp Res 34:1929–1936.
Lucas SM, Rothwell NJ, Gibson RM. 2006. The role of inflamma-
tion in CNS injury and disease. Br J Pharmacol 147(Suppl 1):
S232–S240.
Maclennan KM, Darlington CL, Smith PF. 2002. The CNS effects
of Ginkgo biloba extracts and ginkgolide B. Prog Neurobiol 67:
235–257.
Marino MD, Aksenov MY, Kelly SJ. 2004. Vitamin E protects
against alcohol-induced cell loss and oxidative stress in the neo-
natal rat hippocampus. Int J Dev Neurosci 22:363–377.
Martinez SE, Egea G. 2007. Novel molecular targets for the pre-
vention of fetal alcohol syndrome. Recent Pat CNS Drug Discov
2:23–35.
May PA, Gossage JP, Kalberg WO, et al. 2009. Prevalence and epi-
demiologic characteristics of FASD from various research meth-
ods with an emphasis on recent in-school studies. Dev Disabil
Res Rev 15:176–192.
May PA, Gossage JP, Smith M, et al. 2010. Population differences
in dysmorphic features among children with fetal alcohol spec-
trum disorders. J Dev Behav Pediatr 31:304–316.
Memo L, Gnoato E, Caminiti S, et al. 2013. Fetal alcohol spectrum
disorders and fetal alcohol syndrome: the state of the art and
new diagnostic tools. Early Hum Dev 89(Suppl 1):S40–S43.
Mertsch K, Grune T, Kunstmann S, et al. 1998. Protective effects
of the thiophosphate amifostine (WR 2721) and a lazaroid
(U83836E) on lipid peroxidation in endothelial cells during
hypoxia/reoxygenation. Biochem Pharmacol 56:945–954.
Miller L, Shapiro AM, Cheng J, Wells PG. 2013. The free radical
spin trapping agent phenylbutylnitrone reduces fetal brain DNA
oxidation and postnatal cognitive deficits caused by in utero
exposure to a non-structurally teratogenic dose of ethanol: a role
for oxidative stress. Free Radic Biol Med 60:223–232.
Mitchell JJ, Paiva M, Heaton MB. 1999a. The antioxidants vitamin
E and beta-carotene protect against ethanol-induced neurotoxic-
ity in embryonic rat hippocampal cultures. Alcohol 17:163–168.
Mitchell JJ, Paiva M, Heaton MB. 1999b. Vitamin E and beta-
carotene protect against ethanol combined with ischemia in an
embryonic rat hippocampal culture model of fetal alcohol syn-
drome. Neurosci Lett 263:189–192.
Moreland N, La Grange L, Montoya R. 2002. Impact of in utero
exposure to EtOH on corpus callosum development and paw
BIRTH DEFECTS RESEARCH (PART A) 00:00–00 (2014) 13
preference in rats: protective effects of silymarin. BMC Comple-
ment Altern Med 2:10.
Mousavi SH, Tayarani-Najaran Z, Asghari M, Sadeghnia HR. 2010.
Protective effect of Nigella sativa extract and thymoquinone on
serum/glucose deprivation-induced PC12 cells death. Cell Mol
Neurobiol 30:591–598.
Munday R, Munday CM. 2004. Induction of phase II enzymes by
3H-1,2-dithiole-3-thione: dose-response study in rats. Carcino-
genesis 25:1721–1725.
Nagle DG, Ferreira D, Zhou YD. 2006. Epigallocatechin-3-gallate
(EGCG): chemical and biomedical perspectives. Phytochemistry
67:1849–1855.
Naseer MI, Ullah N, Ullah I, et al. 2011. Vitamin C protects
against ethanol and PTZ-induced apoptotic neurodegeneration in
prenatal rat hippocampal neurons. Synapse 65:562–571.
Nash CM, Ibram F, Dringenberg HC, et al. 2007. Effects of mater-
nal administration of vitamins C and E on ethanol neurobehavio-
ral teratogenicity in the guinea pig. Alcohol 41:577–586.
Neese S, La Grange L, Trujillo E, Romero D. 2004. The effects of
ethanol and silymarin treatment during gestation on spatial
working memory. BMC Complement Altern Med 4:4.
Nguyen T, Nioi P, Pickett CB. 2009. The Nrf2-antioxidant
response element signaling pathway and its activation by oxida-
tive stress. J Biol Chem 284:13291–13295.
Otieno MA, Kensler TW, Guyton KZ. 2000. Chemoprotective 3H-
1,2-dithiole-3-thione induces antioxidant genes in vivo. Free
Radic Biol Med 28:944–952.
Passamonti S, Vrhovsek U, Vanzo A, Mattivi F. 2005. Fast access
of some grape pigments to the brain. J Agric Food Chem 53:
7029–7034.
Peng Y, Kwok KH, Yang PH, et al. 2005. Ascorbic acid inhibits
ROS production, NF-kappa B activation and prevents ethanol-
induced growth retardation and microencephaly. Neuropharma-
cology 48:426–434.
Perez MJ, Velasco E, Monte MJ, et al. 2006. Maternal ethanol con-
sumption during pregnancy enhances bile acid-induced oxidative
stress and apoptosis in fetal rat liver. Toxicology 225:183–194.
Poston L, Briley AL, Seed PT, et al. 2006. Vitamin C and vitamin
E in pregnant women at risk for pre-eclampsia (VIP trial): rando-
mised placebo-controlled trial. Lancet 367:1145–1154.
Reimers MJ, La Du JK, Periera CB, et al. 2006. Ethanol-dependent
toxicity in zebrafish is partially attenuated by antioxidants. Neu-
rotoxicol Teratol 28:497–508.
Reyes E, Ott S, Robinson B. 1993. Effects of in utero administra-
tion of alcohol on glutathione levels in brain and liver. Alcohol
Clin Exp Res 17:877–881.
Riley EP, Infante MA, Warren KR. 2011. Fetal alcohol spectrum
disorders: an overview. Neuropsychol Rev 21:73–80.
Salinas AE, Wong MG. 1999. Glutathione S-transferases–a review.
Curr Med Chem 6:279–309.
Satiroglu-Tufan NL, Tufan AC. 2004. Amelioration of ethanol-
induced growth retardation by all-trans-retinoic acid and alpha-
tocopherol in shell-less culture of the chick embryo. Reprod Toxi-
col 18:407–412.
Scholl TO, Johnson WG. 2000. Folic acid: influence on the out-
come of pregnancy. Am J Clin Nutr 71(Suppl):1295S–1303S.
Shakibaei M, Harikumar KB, Aggarwal BB. 2009. Resveratrol
addiction: to die or not to die. Mol Nutr Food Res 53:115–128.
Shirpoor A, Minassian S, Salami S, Khadem-Ansari MH,
Yeghiazaryan M. 2008. Alpha–lipoic acid decreases DNA damage
and oxidative stress induced by alcohol in the developing hippo-
campus and cerebellum of rat. Cell Physiol Biochem 22:769–776.
Shirpoor A, Salami S, Khadem-Ansari MH, et al. 2009. Protective
effect of vitamin E against ethanol-induced hyperhomocysteine-
mia, DNA damage, and atrophy in the developing male rat brain.
Alcohol Clin Exp Res 33:1181–1186.
Shukla PK, Khanna VK, Ali MM, et al. 2008. Anti-ischemic effect
of curcumin in rat brain. Neurochem Res 33:1036–1043.
Signore C, Aros S, Morrow JD, et al. 2008. Markers of oxidative stress
andsystemicvasoconstrictioninpregnantwomendrinking>or
548 g of alcohol per day. Alcohol Clin Exp Res 32:1893–1898.
Siler-Marsiglio KI, Pan Q, Paiva M, et al. 2005. Mitochondrially
targeted vitamin E and vitamin E mitigate ethanol-mediated
effects on cerebellar granule cell antioxidant defense systems.
Brain Res 1052:202–211.
Siler-Marsiglio KI, Shaw G, Heaton MB. 2004. Pycnogenol and
vitamin E inhibit ethanol-induced apoptosis in rat cerebellar
granule cells. J Neurobiol 59:261–271.
Smith AM, Zeve DR, Grisel JJ, Chen WJ. 2005. Neonatal alcohol
exposure increases malondialdehyde (MDA) and glutathione
(GSH) levels in the developing cerebellum. Brain Res Dev Brain
Res 160:231–238.
Sokoloff L. 1999. Energetics of functional activation in neural tis-
sues. Neurochem Res 24:321–329.
Suresh MV, Sreeranjit Kumar CV, Lal JJ, Indira M. 1999. Impact of
massive ascorbic acid supplementation on alcohol induced oxida-
tive stress in guinea pigs. Toxicol Lett 104:221–229.
Thomas JD, Abou EJ, Dominguez HD. 2009. Prenatal choline supple-
mentation mitigates the adverse effects of prenatal alcohol expo-
sure on development in rats. Neurotoxicol Teratol 31:303–311.
Thomas JD, Idrus NM, Monk BR, Dominguez HD. 2010. Prenatal
choline supplementation mitigates behavioral alterations associ-
ated with prenatal alcohol exposure in rats. Birth Defects Res A
Clin Mol Teratol 88:827–837.
Thomas JD, La Fiette MH, Quinn VR, Riley EP. 2000. Neonatal
choline supplementation ameliorates the effects of prenatal
14 ANTIOXIDANT THERAPIES FOR FAS PREVENTION
alcohol exposure on a discrimination learning task in rats. Neu-
rotoxicol Teratol 22:703–711.
Tiwari V, Chopra K. 2013. Protective effect of curcumin against
chronic alcohol-induced cognitive deficits and neuroinflammation
in the adult rat brain. Neuroscience 244:147–158.
Tran TD, Jackson HD, Horn KH, Goodlett CR. 2005. Vitamin E
does not protect against neonatal ethanol-induced cerebellar
damage or deficits in eyeblink classical conditioning in rats. Alco-
hol Clin Exp Res 29:117–129.
Ullah I, Ullah N, Naseer MI, Lee HY, Kim MO. 2012. Neuroprotec-
tion with metformin and thymoquinone against ethanol-induced
apoptotic neurodegeneration in prenatal rat cortical neurons.
BMC Neurosci 13:11.
Valko M, Leibfritz D, Moncol J, et al. 2007. Free radicals and anti-
oxidants in normal physiological functions and human disease.
Int J Biochem Cell Biol 39:44–84.
Vink J, Auth J, Abebe DT, et al. 2005. Novel peptides prevent
alcohol-induced spatial learning deficits and proinflammatory
cytokine release in a mouse model of fetal alcohol syndrome. Am
J Obstet Gynecol 193(Pt 1):825–829.
Wang HM, Zhao YX, Zhang S, et al. 2010. PPARgamma agonist
curcumin reduces the amyloid-beta-stimulated inflammatory
responses in primary astrocytes. J Alzheimers Dis 20:1189–1199.
Wang X, Ke Z, Chen G, et al. 2012. Cdc42-dependent activation of
NADPH oxidase is involved in ethanol-induced neuronal oxida-
tive stress. PLoS One 7:e38075.
Wentzel P, Rydberg U, Eriksson UJ. 2006. Antioxidative treatment
diminishes ethanol-induced congenital malformations in the rat.
Alcohol Clin Exp Res 30:1752–1760.
Yan D, Dong J, Sulik KK, Chen SY. 2010. Induction of the Nrf2-
driven antioxidant response by tert-butylhydroquinone prevents
ethanol-induced apoptosis in cranial neural crest cells. Biochem
Pharmacol 80:144–149.
Yanaguita MY, Gutierrez CM, Ribeiro CN, et al. 2008. Pregnancy
outcome in ethanol-treated mice with folic acid supplementation
in saccharose. Childs Nerv Syst 24:99–104.
Yuan H, Zhang J, Liu H, Li Z. 2013a. The protective effects of
resveratrol on Schwann cells with toxicity induced by ethanol in
vitro. Neurochem Int 63:146–153.
Yuan H, Zhang W, Li H, Chen C, Liu H, Li Z. 2013b. Neuroprotec-
tive effects of resveratrol on embryonic dorsal root ganglion neu-
rons with neurotoxicity induced by ethanol. Food Chem Toxicol
55:192–201.
Zadak Z, Hyspler R, Ticha A, et al. 2009. Antioxidants and
vitamins in clinical conditions. Physiol Res 58(Suppl 1):S13–
S17.
Zhang C, Tian X, Luo Y, Meng X. 2011. Ginkgolide B attenuates
ethanol-induced neurotoxicity through regulating NADPH oxi-
dases. Toxicology 287:124–130.
Zhang DD. 2006. Mechanistic studies of the Nrf2-Keap1 signaling
pathway. Drug Metab Rev 38:769–789.
BIRTH DEFECTS RESEARCH (PART A) 00:00–00 (2014) 15
... In addition to all this, it must be considered that alcohol consumption during pregnancy can be associated with the consumption of other drugs or teratogenic substances, as when a mother consumes alcohol during pregnancy, the consumption of other drugs is more frequent [18]. During exposure to teratogenic substances, the baby is also influenced by the home environment or the social environment in which the mother lives during this period because an environment in which teratogenic or toxic substances are consumed by the mother or even the consumption of such substances by other people can affect pregnancy. ...
... All these factors make the brain tissue especially susceptible to ROS reactions produced by alcohol. In addition, it must be considered that fetal cells are more exposed to ethanol because they have fewer ethanol-degrading enzymes and because ethanol remains longer in fetal blood [18,76]. ...
Article
Full-text available
Alcohol, a widely consumed drug, exerts significant toxic effects on the human organism. This review focuses on its impact during fetal development, when it leads to a spectrum of disorders collectively termed Fetal Alcohol Spectrum Disorders (FASD). Children afflicted by FASD exhibit distinct clinical manifestations, including facial dysmorphism, delayed growth, and neurological and behavioral disorders. These behavioral issues encompass diminished intellectual capacity, memory impairment, and heightened impulsiveness. While the precise mechanisms underlying alcohol-induced fetal damage remain incompletely understood, research indicates a pivotal role for reactive oxygen species (ROS) that are released during alcohol metabolism, inciting inflammation at the cerebral level. Ethanol metabolism amplifies the generation of oxidant molecules, inducing through alterations in enzymatic and non-enzymatic systems responsible for cellular homeostasis. Alcohol consumption disrupts endogenous enzyme activity and fosters lipid peroxidation in consumers, potentially affecting the developing fetus. Addressing this concern, administration of metformin during the prenatal period, corresponding to the third trimester of human pregnancy, emerges as a potential therapeutic intervention for mitigating FASD. This proposed approach holds promise for ameliorating the adverse effects of alcohol exposure on fetal development and warrants further investigation.
... The supplementation with natural compounds with antioxidant properties, such as the polyphenols extracted from vegetables, might, of course, counteract the toxic prooxidant effect of alcohol abuse during pregnancy [82][83][84][85][86][87]. Furthermore, a healthy diet during pregnancy containing proper amounts of fresh vegetables containing polyphenols, as evidenced by the Mediterranean diet, might reduce the oxidative stress induced by gestational alcohol drinking [88][89][90][91][92][93][94][95]. ...
Article
Full-text available
Prenatal alcohol exposure is responsible for increasing chronic disease risk in later life, including obesity and metabolic syndrome. Alcohol drinking may compromise endogenous antioxidant capacity, causing an increase in free radicals and reactive oxygen species in the newborn. Excessive reactive oxygen species could attack the cellular proteins, lipids, and nucleic acids, leading to cellular dysfunction. Moreover, oxidative stress could play a crucial role in the altered synthesis and release of neurotrophins and progressive mitochondrial modifications with uncontrolled apoptosis. This narrative review aims to underline the important role of alcohol abuse in oxidative stress events and consequent metabolic and neurocognitive impairments in children exposed to alcohol during gestational life.
... In Europe, one in four women consume alcohol during pregnancy (World Health Organization [WHO], 2018). FAS is the most severe manifestation of FASD, and it is associated with several clinical signs, such as craniofacial dysmorphism, growth retardation and neurodevelopmental deficits (Joya et al., 2015). Based on these criteria, a perinatal diagnosis of FAS is possible. ...
Article
Full-text available
Recent data showed that prenatal alcohol exposure (PAE) impairs the “placenta–brain” axis controlling fetal brain angiogenesis in human and preclinical models. Placental growth factor (PlGF) has been identified as a proangiogenic messenger between these two organs. CD146, a partner of the VEGFR-1/2 signalosome, is involved in placental angiogenesis and exists as a soluble circulating form. The aim of the present study was to investigate whether placental CD146 may contribute to brain vascular defects described in fetal alcohol spectrum disorder. At a physiological level, quantitative reverse transcription polymerase chain reaction experiments performed in human placenta showed that CD146 is expressed in developing villi and that membrane and soluble forms of CD146 are differentially expressed from the first trimester to term. In the mouse placenta, a similar expression pattern of CD146 was found. CD146 immunoreactivity was detected in the labyrinth zone and colocalized with CD31-positive endothelial cells. Significant amounts of soluble CD146 were quantified by ELISA in fetal blood, and the levels decreased after birth. In the fetal brain, the membrane form of CD146 was the majority and colocalized with microvessels. At a pathophysiological level, PAE induced marked dysregulation of CD146 expression. The soluble form of CD146 decreased in both placenta and fetal blood, whereas it increased in the fetal brain. Similarly, the expression of several members of the CD146 signalosome, such as VEGFR2 and PSEN, was differentially impaired between the two organs by PAE. At a functional level, targeted repression of placental CD146 by in utero electroporation (IUE) of CRISPR/Cas9 lentiviral plasmids resulted in (i) a decrease in cortical vessel density, (ii) a loss of radial vascular organization, and (iii) a reduced density of oligodendrocytes. Statistical analysis showed that the more the vasculature was impaired, the more the cortical oligodendrocyte density was reduced. Altogether, these data support that placental CD146 contributes to the proangiogenic “placenta–brain” axis and that placental CD146 dysfunction contributes to the cortical oligo-vascular development. Soluble CD146 would represent a promising placental biomarker candidate representative of alcohol-induced neurovascular defects in neonates, as recently suggested by PlGF (patents WO2016207253 and WO2018100143).
... Women who consume large amounts of alcohol often have iron deficiency, which increases the risk of FASD, and iron supplementation may be valuable 297 . Although novel in utero therapies with potential to prevent harm from PAE have been explored in preclinical models, none have been proven safe or effective in human RCTs [298][299][300][301][302][303][304][305][306][307] . Candidate therapies include agents that reduce ethanol-induced oxidative stress, cerebral neuronal apoptosis, growth deficits and structural anomalies caused by PAE 308 . ...
Article
Alcohol readily crosses the placenta and may disrupt fetal development. Harm from prenatal alcohol exposure (PAE) is determined by the dose, pattern, timing and duration of exposure, fetal and maternal genetics, maternal nutrition, concurrent substance use, and epigenetic responses. A safe dose of alcohol use during pregnancy has not been established. PAE can cause fetal alcohol spectrum disorders (FASD), which are characterized by neurodevelopmental impairment with or without facial dysmorphology, congenital anomalies and poor growth. FASD are a leading preventable cause of birth defects and developmental disability. The prevalence of FASD in 76 countries is >1% and is high in individuals living in out-of-home care or engaged in justice and mental health systems. The social and economic effects of FASD are profound, but the diagnosis is often missed or delayed and receives little public recognition. Future research should be informed by people living with FASD and be guided by cultural context, seek consensus on diagnostic criteria and evidence-based treatments, and describe the pathophysiology and lifelong effects of FASD. Imperatives include reducing stigma, equitable access to services, improved quality of life for people with FASD and FASD prevention in future generations. Fetal alcohol spectrum disorders (FASD) are characterized by neurodevelopmental impairment with or without facial dysmorphology, congenital anomalies and poor growth. This Primer discusses the epidemiology, pathophysiology, diagnosis and treatment of FASD.
... FASD referred to the group of disabilities as the result of prenatal exposure to alcohol, marked by neurobehavioral dysfunction, dysmorphology of the face, and stunted growth [173,174]. To date, FASD and other alcohol-related changes have no treatment, and the traits remain throughout life; however, early intervention, including developmental therapy or behavioral interventions, may resist certain disorders' and disabilities' development and progression [175,176]. The potential of catechins to permeate various organs and their anti-oxidative effects bring them to the table for FASD treatment options [177]. ...
Article
Full-text available
Neurodegenerative diseases exert an overwhelming socioeconomic burden all around the globe. They are mainly characterized by modified protein accumulation that might trigger various biological responses, including oxidative stress, inflammation, regulation of signaling pathways, and excitotoxicity. These disorders have been widely studied during the last decade in the hopes of developing symptom-oriented therapeutics. However, no definitive cure has yet been discovered. Tea is one of the world’s most popular beverages. The same plant, Camellia Sinensis (L.).O. Kuntze, is used to make green, black, and oolong teas. Green tea has been most thoroughly studied because of its anti-cancer, anti-obesity, antidiabetic, anti-inflammatory, and neuroprotective properties. The beneficial effect of consumption of tea on neurodegenerative disorders has been reported in several human interventional and observational studies. The polyphenolic compounds found in green tea, known as catechins, have been demonstrated to have many therapeutic effects. They can help in preventing and, somehow, treating neurodegenerative diseases. Catechins show anti-inflammatory as well as antioxidant effects via blocking cytokines’ excessive production and inflammatory pathways, as well as chelating metal ions and free radical scavenging. They may inhibit tau protein phosphorylation, amyloid beta aggregation, and release of apoptotic proteins. They can also lower alpha-synuclein levels and boost dopamine levels. All these factors have the potential to affect neurodegenerative disorders. This review will examine catechins’ neuroprotective effects by highlighting their biological, pharmacological, antioxidant, and metal chelation abilities, with a focus on their ability to activate diverse cellular pathways in the brain. This review also points out the mechanisms of catechins in various neurodegenerative and cognitive diseases, including Alzheimer’s, Parkinson’s, multiple sclerosis, and cognitive deficit.
... More alcohol consumption during pregnancy can be common cause of teratogenecity on fetus who born from heavy drunker, and FASD is the most common cause of intelligent quotient disability in the fetus who born from pregnant women consume alcohol in high amount and also correlated with the advancement of behavioral disturbances and physical abnormality. [20][21][22][23][24][25][26] Prenatal alcohol exposure can be cause neurological and behavioural challenges which are the class of FASDs. In the world the prevalence of FASD in the children population have been estimated at 7.7% cases per thousand, with the greatest rates 19.8 in thousand observed in the European region. ...
Article
Full-text available
Miscarriage, premature birth, stillbirth, low birth weight, and a range of lifelong disorders can caused by excessive alcohol consumption during pregnancy is called fetal alcohol spectrum disorders. Alcohol is the most hazardous teratogens which are substances that can detriment an advancing fetus. Every time a pregnant woman has a drink; her unborn child has only, too. Alcohol, like carbon monoxide from cigarettes, passes easily through the placenta from the mother's bloodstream into her baby's blood and puts her fetus at risk of having a fetal alcohol spectrum disorder. Drinking alcohol while pregnant can result in cognitive, social, and motor inadequacies and distinctive lifelong challenges. Usage of alcoholic beverages by pregnant women perhaps results in abortion, fetal mortality and prematurity. The intentions of this review is to teach the women about risk of alcohol consumption during pregnancy on their child and also on themselves and warn them to cease alcohol during pregnancy totally to safe their child from various defects and themselves from unexpected health challenges.
... This ameliorative effect of Ginger may be attributed to its antioxidant effect [28] . As, related reports have showed that treatment with antioxidant can reduce malformations and growth retardation [29] . In line with that, it was reported that co-administration of vitamin E with valproic acid and phenytoin significantly improved the fetal weight, length and malformations [30] . ...
... Maternal alcohol consumption can lead to the irreversible condition of fetal alcohol syndrome (FAS), the most severe form of the alcohol spectrum disorders (FASDs) (Guerri et al., 2009;de Sanctis et al., 2011;May and Gossage, 2011;Memo et al., 2013;Joya et al., 2015;Van Heertum and Rossi, 2017). FASD is characterized by fetal and birth anomalies, intrauterine growth retardation (IUGR), numerous physical, cognitive, and behavioral defects in newborns and children (Hoyme et al., 2016;Roozen et al., 2016). ...
Article
Full-text available
Adequate placentation, placental tissue remodeling and vascularization is essential for the success of gestation and optimal fetal growth. Recently, it was suggested that abnormal placenta induced by maternal alcohol consumption may participate in fetal growth restriction and relevant clinical manifestations of the Fetal Alcohol Spectrum Disorders (FASD). Particularly, periconceptional alcohol consumption up to early gestation can alter placentation and angiogenesis that persists in pregnancy beyond the exposure period. Experimental evidence suggests that abnormal placenta following maternal alcohol intake is associated with insufficient vascularization and defective trophoblast development, growth and function in early gestation. Accumulated data indicate that impaired vascular endothelial growth factor (VEGF) system, including their downstream effectors, the nitric oxide (NO) and metalloproteinases (MMPs), is a pivotal spatio-temporal altered mechanism underlying the early placental vascular alterations induced by maternal alcohol consumption. In this review we propose that the periconceptional alcohol intake up to early organogenesis (first trimester) alters the VEGF-NO-MMPs system in trophoblastic-decidual tissues, generating imbalances in the trophoblastic proliferation/apoptosis, insufficient trophoblastic development, differentiation and migration, deficient labyrinthine vascularization, and uncompleted remodelation and transformation of decidual spiral arterioles. Consequently, abnormal placenta with insufficiency blood perfusion, vasoconstriction and reduced labyrinthine blood exchange can be generated. Herein, we review emerging knowledge of abnormal placenta linked to pregnancy complications and FASD produced by gestational alcohol ingestion and provide evidence of the early abnormal placental angiogenesis-vascularization and growth associated to decidual-trophoblastic dysregulation of VEGF system after periconceptional alcohol consumption up to mid-gestation, in a mouse model.
Article
Objective: To assess the degree of influence of intrauterine alcoholization on the formation of various structural components of the brain of human embryos. Material and methods: Twenty-six samples of embryonic material from 8 to 11 weeks of intrauterine development were studied. The material was divided into four subgroups in accordance with the gestational age (Control 1 - 8-9 weeks of gestation and Control 2 - 10-11 weeks of gestation) and the history of the mother (presence or absence of the diagnosis «Alcoholism stage I-II» in the anamnesis). Morphometry was subjected to semi-thin sections stained by Nissl. The diameter and area of each individual tissue element (neuroblasts, glioblasts, vessels of the microvasculature, as well as the determination of the specific area (the ratio of the total area of the studied structure to the area of the entire section) and the calculation of the average number of these structures per unit area of the section, were determined. The AxioVision 4.8 program (Carl Zeiss, Germany) was used for analysis, and the Mann-Whitney test was used for statistical analysis of differences between the samples (significant differences, p<0.05). Results: An insufficient increase in the area of vessels of the microvasculature was revealed in combination with a compensatory increase in their number per unit area of the section in the Alcohol groups compared with intact groups (48.5 μm2 vs 83.3 μm2, p<0.05). When comparing the sizes of glioblasts in the Control and Alcohol subgroups at different stages of development, a lag in the sizes of cellular structures in the Alcohol groups at the initial stages was revealed (average area 21.3 μm2 vs 32.1 μm2; 12.9 μm2 vs 13.3 μm2). When comparing data on later periods, no significant differences were found, only an increase in the specific number of cells in subgroup Alcohol 2 was noted (p<0.05). In neuroblasts, there was also a decrease in cell size with an increase in gestational age both among the Control and among the Alcohol subgroups. However, the cell sizes in Alcohol 2 exceeded those in Control 2 and their number was smaller (p<0.05). Conclusion: Alcohol leads to changes in the size and number of neuroblasts, glioblasts and vessels of the microvasculature and, as a result, to a disproportionate development of the entire brain tissue. The changes progress with an increase in the development period.
Article
Full-text available
The Glutathione S-transferases (GSTs) form a group of multi-gene isoenzymes involved in the cellular detoxification of both xenobiotic and endobiotic compounds. GSTs have been divided into a number of subclasses, alpha, mu, pi, and theta. The classification was made on the basis of sequence similarity and immunological cross-reactivity. GSTs show a high level of specificity toward GSH but the electrophilic second substrate can vary significantly both between and within the classes in spite of their sequence similarity. X-ray crystallography and site-directed mutagenesis studies have together elucidated the structure and mechanism of GSTs. Catalysis occurs by conjugation with glutathione (GSH) and the less toxic and more hydrophilic products can then be partially metabolised and excreted. This invaluable service is however disadvantageous during chemotherapy where GSTs have been associated with multi-drug resistance of tumour cells. Levels of expression of different isoforms of GSTs are tissue specific. The variations in expression between normal and tumour cells are of interest and in most cases the levels of GSTs are increased, especially p-GST. Understanding the complex role that GSTs play in drug resistance begins with determining the pattern of isoform expression and the substrate specificities of each isoform. The use of isozyme-specific, GSH analogues as inhibitors to modulate GST activity during chemotherapy is a promising strategy in the battle against cancer. This review attempts to provide a detailed overview of the literature concerning the different classes of GSTs, their function and mechanism and the use of GSTs as therapeutic targets for disease as current at the time of submission
Article
Full-text available
Alcohol-induced congenital heart defects (CHDs) are frequently among the most life-threatening and require surgical correction in newborns. The etiology of these defects, collectively known as Fetal Alcohol Syndrome (FAS), has been the focus of much study, particularly involving cellular and molecular mechanisms. Few studies have addressed the influential role of altered cardiac function in early embryogenesis, due to a lack of tools with the capability to assay tiny beating hearts. To overcome this gap in our understanding, we used optical coherence tomography (OCT), a non-destructive imaging modality capable of micrometer-scale resolution imaging, to rapidly and accurately map cardiovascular structure and hemodynamics in real time under physiological conditions. In this study, we exposed avian embryos to a single dose of alcohol/ethanol at gastrulation when the embryo is sensitive to the induction of birth defects. Late-stage hearts were analyzed using standard histological analysis with a focus on the atrio-ventricular valves. Early cardiac function was assayed using Doppler OCT, and structural analysis of the cardiac cushions was performed using OCT imaging. Our results indicated that ethanol-exposed embryos developed late-stage valvuloseptal defects. At early stages, they exhibited increased regurgitant flow and developed smaller atrio-ventricular cardiac cushions, in comparison to controls (uninjected and saline-injected embryos). The embryos also exhibited abnormal flexion/torsion of the body. Our evidence suggests that ethanol-induced alterations in early cardiac function have the potential to contribute to late-stage valve and septal defects, thus demonstrating that functional parameters may serve as early and sensitive gauges of cardiac normalcy and abnormalities.
Article
Full-text available
Since 2004 we have been conducting a randomized control double blind trial on the favourable effect of mega-vitamin C and E in pregnant women with heavy alcohol exposure. The study was terminated in the summer of 2006 due to new evidence showing intrauterine growth restriction caused by such treatment in women with pre-eclampsia.
Article
Full-text available
The glutathione S-transferase (GST) supergene family comprises gene families that encode isoenzymes that are widely expressed in mammalian tissue cytosols and membranes. Both cytosolic (particularly the isoenzymes encoded by the alpha, mu and theta gene families) and microsomal GST catalyse the conjugation of reduced glutathione (GSH) with a wide variety of electrophiles which include known carcinogens as well as various compounds that are products of oxidative stress including oxidised DNA and lipid. Indeed, several lines of evidence suggest certain of these isoenzymes play a pivotal role in protecting cells from the consequences of such stress. An assessment of the importance of these GST in humans is presently difficult however, because the number of alpha and theta class genes is not known and, the catalytic preferences of even identified isoforms is not always clear.
Article
Background: Rodent studies have shown that heavy binge-like ethanol (EtOH) exposure during the brain growth spurt [postnatal days (PD) 4-9] causes cerebellar neuronal loss and deficits in cerebellar-mediated eyeblink classical conditioning (ECC). Oxidative stress has been implicated in EtOH-mediated brain damage, and studies using vitamin E have reported amelioration of EtOH-induced tissue damage, including protection in rats against EtOH-induced cerebellar Purkinje cell (PC) loss on PD 4 to 5. The purpose of this study was to determine whether dietary supplementation with vitamin E concurrent with binge EtOH exposure on PD 4 to 9 in rats would attenuate the cerebellar cell death and ECC deficits. Methods: Rat pups were given one of five different neonatal treatments: (1) intubation with EtOH in milk formula (twice daily, total dose 5.25 g/kg/day), (2) intubation with EtOH in milk formula supplemented with vitamin E (12.26 mg/kg/feeding), (3) intubation with milk formula that contained vitamin E only, (4) sham intubations, or (5) normally reared unintubated controls. Between PD 26 and 33, subjects received short-delay ECC for 3 consecutive days. Unbiased stereological cell counts were performed on cerebellar PCs of left cerebellar lobules I to VI and neurons of the interpositus nucleus. In a separate study with PD 4 pups, the effects of vitamin E on EtOH-induced expression of caspase-3 active subunits were assessed using Western blot analysis. Results: EtOH-treated groups showed significant deficits in acquisition of conditioned eyeblink responses and reductions in cerebellar PCs and interpositus nucleus neurons compared with controls. Vitamin E supplementation failed to protect against these deficits. Vitamin E also failed to protect against increases in caspase-3 active subunit expression induced by acute binge EtOH exposure on PD 4. Conclusions: In contrast to the previously reported neuroprotective potential of antioxidants on EtOH-mediated cerebellar damage, vitamin E supplementation did not diminish EtOH-induced structural and functional damage to the cerebellum in this model of binge EtOH exposure during the brain growth spurt in rats.
Article
Lipid peroxidation has been implicated in ethanol-induced liver injury and observed in fetal liver and brain after maternal ethanol consumption with mitochondria being the target organelles. This process generates a highly reactive and toxic product, 4-hydroxynonenal (HNE). In the present study, HNE levels and metabolism were assessed in mitochondria of fetal and maternal liver after in vivo ethanol exposure. Female Sprague-Dawley rats received five doses of ethanol (4 g/kg orally at 12-hour intervals) and were killed on day 19 of gestation. The results showed that HNE levels were enhanced in hepatic mitochondria of fetal rats exposed to ethanol, far in excess of that in adult liver mitochondria. Measurement of HNE metabolism showed that fetal mitochondria had a lower capacity for HNE catabolism than adult mitochondria. In adult mitochondria, HNE could be metabolized by nicotine adenine dinucleotide-dependent oxidation, reduced glutathione conjugation, and reduced nicotine adenine dinucleotide-dependent reduction, whereas in fetal liver only the former two pathways were active, but to a lesser degree than in adult mitochondria. On the other hand, mitochondria from fetal liver showed a higher production of HNE when oxidative stress was induced with t-butyl hydroperoxide. Prior in vivo ethanol exposure further potentiated HNE formation in t-butyl hydroperoxide-stimulated fetal liver mitochondria, but not in adult mitochondria. These findings indicate that increased levels of HNE in fetal liver mitochondria after maternal ethanol consumption reflect a higher susceptibility to HNE formation in addition to a lesser capacity to metabolize it. The enhanced accumulation of this toxic aldehyde may contribute to oxidative damage observed in fetal tissues after in utero ethanol exposure.(Hepatology 1997 Jan;25(1):142-7)
Article
This chapter discusses the reaction of lipoic acid with ebselen and hypochlorous acid. Lipoic acid plays a pivotal role in energy metabolism. Its antioxidant activity, however, is also recognized in normal and pathological conditions. In vivo, lipoic acid shuffles between its reduced [dihydrolipoic acid or 6,8-dimercaptooctanoic acid, L(SH)2] and oxidized state (lipoic acid or 1,2-dithiolane-3-pentanoic acid). Dihydrolipoic acid is characterized by two thiol groups per molecule; these thiol groups provide the compound with its good antioxidant efficacy. In this regard, its role in the glutathione (GSH) peroxidase-like activity of the small molecule ebselen is interesting. Ebselen is an organoselenium compound with antiinflammatory activity. It catalyzes the reduction of peroxides by GSH. Replacing GSH by L(SH)2 improves the peroxidase-like activity of ebselen. In the oxidized state of lipoic acid, the two sulfur atoms are attached to each other as part of a five-membered 1,2-dithiolane ring. The strain in this ring is responsible for a unique reactivity. An example of this reactivity is the ability of this molecule to scavenge hypochlorous acid (HOCl). By scavenging the neutrophil oxidant HOCl, the elastase inhibitor αl-antiproteinase (API) is protected from oxidation. The reaction scheme for the catalysis of the GSH peroxidase reaction by ebselen is analogous to that of the enzyme.
Article
Although at least one small-scale study has suggested that supplemental vitamins C and E may lower the risk of preeclampsia, others have yielded negative results. The present randomized, placebo-controlled trial evaluated these antioxidants in 2410 women, seen at 25 centers, who were viewed as being at increased risk of preeclampsia because of such factors as preeclampsia in the preceding pregnancy, HELLP syndrome (hemolysis, elevated liver enzymes, low platelets), eclampsia in any past pregnancy at any gestational stage, essential hypertension requiring medication, and type 1 or type 2 diabetes. Participants were assigned to receive either 1000 mg vitamin C plus 400 IU vitamin E or a matched placebo each day from the second trimester of pregnancy to delivery. Preeclampsia developed in 15% of study participants. Vitamin supplementation did not lower the risk regardless of the level of risk at entry to the trial. Risk rates ranged from 11% in primiparous obese women and those with multiple pregnancies to 32% in women having chronic renal disease. The risk for women with multiple risk factors was 26%. There were no group differences in either severe preeclampsia (with marked hypertension) or early-onset preeclampsia (delivery for preeclampsia before 34 weeks gestation). More women in the supplemented group were treated for gestational hypertension. Infants whose mothers received vitamin supplements were significantly likelier to have low birth weight, but there was no difference in small-for-gestational-age infants. Preterm births were comparably frequent in the supplemented and control groups, but infants of supplemented mothers more often had a cord arterial blood pH less than 7. The risk of perinatal death was similar in the 2 groups. These findings fail to support routine supplementation with vitamins C and E for pregnant women at increased risk of developing preeclampsia. Supplementation may have adverse effects such as low birth weight and low arterial cord blood pH and, for this reason, cannot be recommended, at least in the doses used in this trial.