Vous êtes sur la page 1sur 6

Available online at www.sciencedirect.

com Current Opinion in

ScienceDirect Toxicology

Oxidative stress during development:


Chemical-induced teratogenesis
Jason M. Hansen, Benjamin R. Jacob and Ted B. Piorczynski

Abstract through unknown etiologies [1]. Chemicals that produce


Oxidative stress has been shown to be an important contrib- birth defects, known as teratogens, contribute only a small
utor to birth defects, including structural malformation and amount to the total known causes of birth defects in
neurobehavioral deficits. At high levels, oxidative stress pro- humans, but this may be an underestimate of environ-
motes apoptosis but at lower levels that do not cause cell mental influences in poor developmental outcomes. Of
death, redox-sensitive pathways can be disrupted. Chemicals the known human teratogens, many are known to produce
that cause birth defects usually provide a very distinct devel- oxidative stress [1], suggesting an important mechanistic
opmental outcome, dependent upon gestational periods of link between oxidative stress, redox signaling and devel-
exposure and level of exposure, but these observations also opment. Here, in this review, we outline the role of
suggest that there are specific pathways involved in the oxidative stress in developmental toxicity and chemical-
manifestation of a given defect. Here, we review overarching induced embryopathy.
themes of oxidative stress and their effects during develop-
ment and provide specific examples of some of the most 2. Oxidative stress and redox signaling
notorious human teratogens that produce oxidative stress as Oxidative stress has been classically defined as an
part of their teratogenic mechanism. Moving forward, future imbalance between oxidizing and reducing equivalents,
studies should center on providing a more profound under- where the former predominates [2]. In severely pro-
standing of redox-sensitive elements during development to oxidizing environments, cellular macromolecules are
develop potential preventative interventions. damaged promoting apoptosis or necrosis [2]. With less
Addresses toxic levels of oxidative stress, redox states are simply
Department of Physiology and Developmental Biology, College of Life shifted but can disrupt or delay normal cell function. In
Sciences, Brigham Young University, Provo, UT, 84602, USA some cases, these redox-sensitive pathways are linked to
Corresponding author: Hansen, Jason M (jason_hansen@byu.edu)
systems that support the restoration of functional cellular
Email address: ben.richard.jacob@gmail.com (B.R. Jacob), redox states, prior to the oxidation-induced promotion of
ted.piorczynski@gmail.com (T.B. Piorczynski) cell death (Fig. 1). However, there are other redox-
sensitive pathways that are likely more closely tied to
developmental pathways, where untimely shifts in
Current Opinion in Toxicology 2018, 7:110–115
cellular redox states could result in aberrant signaling of
This review comes from a themed issue on Oxidative Toxicology: redox-sensitive pathways, support untimely cellular
Role of ROS
function and ultimately promote dysmorphogenesis.
Available online 7 November 2017
For a complete overview see the Issue and the Editorial Development is a mixture of different cellular actions,
https://doi.org/10.1016/j.cotox.2017.11.003 such as proliferation and differentiation, which occur in
2468-2020/© 2017 Elsevier B.V. All rights reserved. a well-coordinated manner, where disruption of these
events can result in poor developmental outcomes.
Proliferative, differentiative and apoptotic activities are,
Keywords
Glutathione, Birth defects, Oxidative stress, Redox, Signaling,
in part, regulated by cellular redox states [3] and as
Apoptosis. such, can be altered during periods of oxidative stress
Abbreviations and redox imbalance. Perhaps the most susceptible pe-
ASK1, Apoptosis Signaling Kinase 1; Cys, Cysteine; D3T, 3H-1,2- riods in the embryo are periods defined by transition
dithiole-3-thione; Eh, Redox Potential; FASD, Fetal Alcohol Spectrum from an important developmental event to another (e.g.
Disorder; Grx, Glutaredoxin; GSH, Glutathione; GSSG, Glutathione
Disulfide; H2O2, Hydrogen Peroxide; MAP, Mitogen-activated Protein;
proliferation/differentiation) in developing target
NAC, N-Acetylcysteine; NF-kB, Nuclear Factor Kappa B; Nrf2, NF- organs and systems (Fig. 2). Chemical-induced oxidative
E2p45-related factor 2; ROS, Reactive Oxygen Species; -SOH, Sulfenic stress can result in prolonged periods of redox imbalance
Acid; -SSG, S-Glutathionylation; Trx1, Thioredoxin-1. that alter developmental programming from regular
schedule events.
1. Introduction
Alarmingly, one in every 33 births present with a birth 3. Redox regulation during development
defect, ranging from relatively minor, non-life-threatening The intracellular redox state has been shown to be an
effects to severe, lethal developmental outcomes. Un- integral part of many essential developmental processes.
fortunately, upwards of 70% of all birth defects occur The influence that redox states have on development is

Current Opinion in Toxicology 2018, 7:110–115 www.sciencedirect.com


Oxidative stress and birth defects Hansen et al. 111

Fig. 1 and the bioactivation of chemicals to more reactive,


oxidizing metabolites (e.g. ethanol is metabolized to
reactive acetaldehyde via alcohol dehydrogenase). Loss
of ROS regulation can be damaging during development
and thus, a tightly controlled redox state is optimal for
normal embryogenesis [5]. While there are many cellular
processes that rely on redox-dependent mechanisms, we
have highlighted only three in this review that are most
relevant to developmental toxicity-related processes.

3.1. Cellular signaling


Cellular signaling has been widely shown to incorporate
ROS, like H2O2, as a signaling intermediate and sec-
Comparison of the activation of redox-sensitive pathways (green line) with ondary messenger. For example, epidermal growth factor
pro-cell death (red line) signaling. Under normal, usually pro-reducing, signaling produces H2O2 to oxidize proteins in tyrosine
intracellular redox states, compensatory redox signaling and cell death is
kinase cascades and promote DNA synthesis and
low. As redox states shift to a more oxidizing environment, as would occur
with chemical-induced oxidative stress, redox-sensitive elements and cellular proliferation [6]. Similar results were found with
pathways would be activated earlier in the redox spectrum and at more platelet-derived growth factors when their tyrosine re-
moderate redox shifts. As redox states progress to more pro-oxidizing ceptor signaling systems were blocked by the inhibition
potentials, various redox-sensitive signaling pathways could be deacti- of ROS production, indicating the use of redox shifts in
vated dependent on their redox sensitivity. As these corrective pathways
are either shut off or deactivated, apoptotic signaling will predominate to
cellular signaling [7]. Furthermore, H2O2 is a necessary
promote cell death. intermediate for glucose-induced insulin secretion [8].
During development, the relative value of oxygen in
metabolism fluctuates as the preferential source for
Fig. 2 cellular energy shifts from glycolysis to oxidative phos-
phorylation. This change shifts the redox state of the
cell when the embryo uses mitochondrial production of
ROS during metabolic switching [9,10].

Additionally, H2O2 has been shown to activate the


transcription factor NF-E2p45-related factor 2 (Nrf2)
during times of electrophilic and oxidative stress. Nrf2
then signals the increased gene expression of antioxi-
dant and phase II detoxification enzymes [11]. The
Nrf2-mediated response is an important element of the
antioxidant defense system in developing embryos. In
some models, embryos show distinct, inherent changes
to redox states during development, where cellular
Redox switches involved in cellular function. Under highly reducing con- redox states during periods of differentiation are
ditions, proliferation-related pathways are switched on (green line), but as generally more oxidizing than those observed during
redox states transition to a more oxidizing condition, pro-proliferation
switches are deactivated and pro-differentiation switches (blue line) are
proliferation [12]. These changes may be an influencing
activated. Excessive shifts to extremely pro-oxidizing redox states switch factor in how Nrf2 is regulated during development after
off pro-differentiation pathways and promote apoptosis (red line) and oxidant exposure [13]. There are a number of tran-
necrosis (black line). During development, transitional periods (outlined in scription factors, like Nrf2, that are controlled by redox
yellow) may be the most susceptible periods to chemical-induced redox states, many of which are involved in developmental
signaling disruption. Untimely switching may prevent normal develop-
mental processes and cause poor developmental outcomes (structural
events, and thus, support redox regulation involvement
malformations, neurobehavioral deficits, etc.). in development [14,15]. The specific mechanisms for
the redox-sensitive signaling methods are currently
under investigation, but may be directly or indirectly
complex and far reaching, affecting a variety of cellular tethered to intracellular redox states. While S-thiolation
systems. Because cellular processes affect the redox state has been suggested as a redox-control mechanism, S-
of the cell, the redox regulation of these processes can glutathionylation (-SSG) is seen as a direct control
involve feedback control [4]. Reactive oxygen species regulator of redox-dependent responses [16]. Indirectly,
(ROS) are understood to have beneficial as well as loss of reducing power may promote the increased
deleterious effects on embryo development. However, availability of ROS to cause protein oxidation (devel-
aberrant production of ROS to toxic levels can occur opment of sulfenic acids, -SOH). As such, through di-
through a variety of mechanisms including changes to sulfide exchange mechanisms, both indirect and direct
cellular metabolism, alterations to mitochondrial kinetics redox control of proteins can overlap [17].

www.sciencedirect.com Current Opinion in Toxicology 2018, 7:110–115


112 Oxidative Toxicology: Role of ROS

3.2. Differentiation 4. Disruption of redox control during


Cellular differentiation relies on some of the cellular development through chemical exposures
signaling mechanisms previously described and subse- Alterations in redox status influence multiple cellular
quently is also influenced by the redox potential of the processes that may negatively affect embryonic devel-
cell. Human stem cell differentiation into adipocytes opment. Consequently, embryonic cells are sensitive to
and osteocytes has been shown to be directly related to the direct action of certain drugs or to the environ-
the glutathione (GSH) redox potential (Eh); in both mental changes produced by drugs that induce oxidative
processes, the overall GSH Eh shifted to a more stress [29]. While multiple teratogens have been shown
oxidizing state, albeit at different rates during the dif- to increase ROS production and induce oxidative stress,
ferentiation period [18] and may highlight specific redox we will focus on only a few of the most prevalent ones for
regulation into certain phenotypes. Although redox this review.
states were not the primary measure, studies show that
mitochondrial ROS seem to be involved in CD4þ and 4.1. Thalidomide
CD8þ memory T cell differentiation [19]. Reactive In the 1950s, this non-addictive sedative was widely
oxygen species have also been implicated as a signal of prescribed to pregnant women to ameliorate nausea
adipocyte differentiation [20,21] and osteocyte differ- associated with morning sickness. Shortly thereafter,
entiation [22]. Reducing ROS levels in hematopoietic thalidomide exposure during the first trimester of
stem cells promotes a more reducing environment pregnancy was shown to be responsible for a number of
which correlated with a restriction in the cell’s ability to deformities in newborns, including phocomelia and
differentiate [23]. These findings suggest a more amelia [30,31]. Recently, interest in thalidomide has
nuanced role for shifts in intracellular redox potentials resurfaced as it has received FDA approval as an effec-
and indicate that, at different redox potentials and at tive therapy for multiple myeloma and erythema nodo-
different stages of development, an embryo can be more sum leprosum [32]. Multiple sources have shown that
or less prone to differentiate. thalidomide increases ROS production, induces oxida-
tive stress and disrupts the function of the redox-
3.3. Apoptosis sensitive transcription factor, Nuclear Factor Kappa B
Redox state is also linked with the activation and (NF-kB), an important mediator of limb bud outgrowth,
stimulation of apoptosis [24,25]. Reduced thioredoxin, a and appear to contribute to limb reduction defects [33e
protein oxidoreductase, can bind to and inhibit 36]. Pretreatment with various antioxidants reduces the
apoptosis signaling kinase 1 (ASK1), a member of the associated thalidomide-induced terata in animal models
MAP3 kinase family [26]. However, upon oxidation, and restores redox states to preserve normal cellular
Trx1 becomes unbound and allows ASK1 to become fully signaling and function [37,38], further suggesting the
active promoting apoptosis [27]. As such, changes in role of oxidative stress, ROS generation and redox
cellular redox environments specific to Trx1 Eh can imbalance in thalidomide teratogenesis.
directly influence ASK1 activity and stimulate related
downstream apoptotic activities. 4.2. Phenytoin
Phenytoin is a widely used anticonvulsant drug that
As GSH becomes oxidized to glutathione disulfide induces oxidative stress [39] and is associated with an
(GSSG), protein targets can become S-glutathionylated increased risk for isolated cleft palate in children
and as a post-translational modification, protein function following maternal exposure during the first trimester of
can be changed. In relation to apoptosis, studies have pregnancy [40]. In addition, phenytoin results in
shown that in Fas-mediated death pathways, Fas can be oxidative DNA damage and dysmorphogenesis, which
glutathionylated on a redox-sensitive cysteine residue can be reduced by the antioxidant superoxide dismutase
(Cys294) further promoting its downstream pro- [41]. Phenytoin-induced defects in animal models may
apoptotic signaling [28]. Fas-SSG increase FasL bind- be reduced through pretreatment with N-acetylcysteine
ing and accentuated apoptosis. Removal or addition of (NAC), a precursor to GSH synthesis. Conversely, pre-
GSH moieties to proteins can occur through the action treatment with antioxidant depleting agents increased
of glutaredoxins (Grx), but mostly de-glutathionylate the occurrence of defects [39]. Curcumin, a pigment
proteins under normal conditions. For de- from plants known to have antioxidant properties, is
glutathionylation reactions, Grxs will reduce proteins effective in preventing phenytoin-induced cognitive
and become glutathionylated itself (Grx-SSG). Over- impairment and oxidative stress in rats [42,43]. In other
expression of Grx prevented the formation of Fas-SSG studies, GSH levels in fetal organs were significantly
and reduced apoptosis. As such, the role of GSH Eh depleted when phenytoin was given from gestational
requires further study to better understand the pro- day 7e18, yet interestingly, antioxidant supplementa-
posed mechanisms here, especially in the context of tion (vitamin E), in this case, failed to rescue changes to
development. GSH concentrations [44].

Current Opinion in Toxicology 2018, 7:110–115 www.sciencedirect.com


Oxidative stress and birth defects Hansen et al. 113

4.3. Ethanol 4.4. Mercury


Ethanol consumption during pregnancy can result in a Mercury is a naturally occurring element found in the
wide range of adverse effects in the developing fetus. earth’s crust that has been released into the biosphere as
Fetal Alcohol Spectrum Disorder (FASD) comprises the a consequence of natural processes including volcanic
vast range of conditions that can occur when alcohol is activity, fires and erosion. Recently, anthropogenic
consumed during various periods of pregnancy. Fetal sources have greatly contributed to the environmental
Alcohol Syndrome (FAS), the most severe condition, is distribution of mercury via coal mining and chemical
characterized by facial abnormalities, growth retarda- syntheses. While various types of mercury occur in
tion, and central nervous system impairment [45]. Evi- nature, perhaps the most detrimental, both develop-
dence from in vitro and in vivo models of prenatal ethanol mentally and otherwise, is methylmercury, the most
exposure suggest that oxidative stress contributes to abundant organic mercury. Methylmercury has been
deficits seen in FASD, and that antioxidants are po- shown to bioaccumulate in organisms causing severe
tential candidates for the treatment of alcohol-induced health issues including irreversible brain damage and
developmental disorders [46]. In In vitro culture, em- even death [1]. Fetal methylmercury poisoning has been
bryos demonstrated a decrease in GSH concentrations well documented in Japan, where mothers consumed
with ethanol exposure and similar GSH depletion was fish contaminated by methylmercury resulting in
observed with in utero exposure [47]. In the same study, numerous children being born with neurobehavioral
embryonic cysteine was also significantly decreased but deficits [52]. In mice, various in utero effects were
to an even greater extent than GSH with both in vitro observed with methylmercury exposure, including cleft
and in utero exposures. Ethanol caused an increase in palate, exencephaly, missing limbs and facial deformities
H2O2 and superoxide production in cultured fetal he- [53]. Microglial cells exposed to methylmercury showed
patocytes, as well as an increase in malondialdehyde, a a concentration- and time-dependent increase in ROS
marker of lipid oxidative damage [48,49]. The Nrf2- generation and a concomitant decrease in the ratio of
inducer, D-L-sulforaphane, protects neural crest cells, GSH and GSSG [54]. In a mouse model, administration
an ethanol-sensitive cell population implicated in of NAC prevented methylmercury-induced malforma-
FASD, against ethanol-induced oxidative stress and tions [55]. In mouse fetuses, methylmercury caused a
apoptosis [50]. Furthermore, in utero, administration of significant increase in GSSG and decrease in GSH,
the Nrf2 inducer, 3H-1,2-dithiole-3-thione (D3T), supporting an environmental redox shift correlation with
improved antioxidant enzyme expression and activities its developmental toxicity [56]. Interestingly, mercury
and prevented cranial apoptosis in the embryonic neural exposure did not increase GSH synthesis pathways in
folds [51]. the fetus as the GSH synthesis enzyme activity was

Fig. 3

Overview of oxidative stress in developing tissues. Following chemical exposure, changes to cellular metabolism, mitochondrial kinetics or the bio-
activation of xenobiotics into reactive metabolites can promote an increase in ROS production increases and shift intracellular redox states. Excessively
high levels of ROS generation and/or highly oxidized redox states can promote apoptosis (black arrows), but under more moderate conditions, shifts in
redox state can promote disruption to redox-sensitive cell signaling (red arrows), including important developmental pathways. Outcomes can include
structural malformations and neurobehavioral deficits. However, redox shifts can also activate compensatory pathways that are designed to restore redox
imbalance and increase ROS detoxification (green arrows). Rapid and timely restoration of redox states may serve as a means to prevent prolonged
periods of signal disruption and support normal developmental patterning.

www.sciencedirect.com Current Opinion in Toxicology 2018, 7:110–115


114 Oxidative Toxicology: Role of ROS

unchanged, but increases in activity were observed in 10. Murphy MP: How mitochondria produce reactive oxygen
species. Biochem J 2009, 417:1–13.
the visceral yolk sac, which may suggest poor oxidant
pathway responses in the embryo/fetus during these 11. Dinkova-Kostova AT, Abramov AY: The emerging role of Nrf2 in
mitochondrial function. Free Radic Biol Med 2015, 88:179–188.
stages of development.
12. Timme-Laragy AR, Goldstone JV, Imhoff BR, Stegeman JJ,
Hahn ME, Hansen JM: Glutathione redox dynamics and
5. Conclusion expression of glutathione-related genes in the developing
embryo. Free Radic Biol Med 2013, 65:89–101.
During development, cells are in flux, as demonstrated
13. Sant KE, Hansen JM, Williams LM, Tran NL, Goldstone JV,
by specified periods of proliferation, differentiation and Stegeman JJ, Hahn ME, Timme-Laragy A: The role of Nrf1 and
apoptosis. Oxidant exposure can increase ROS levels Nrf2 in the regulation of glutathione and redox dynamics in
resulting in variable effects. Dependent upon the the developing zebrafish embryo. Redox Biol 2017, 13:
207–218.
degree of ROS generation, cellular macromolecules can
14. Arrigo AP: Gene expression and the thiol redox state. Free
be severely or irreversibly damaged to the point where Radic Biol Med 1999, 27:936–944.
cells will undergo apoptosis, whereas under lower ROS
15. Marinho HS, Real C, Cyrne L, Soares H, Antunes F: Hydrogen
conditions, redox states can be altered to modify redox- peroxide sensing, signaling and regulation of transcription
sensitive signaling pathways (Fig. 3). With the latter, factors. Redox Biol 2014, 2:535–562.
there are pathways that favor the correction and resto- 16. Kuksal N, Chalker J, Mailloux RJ: Progress in understanding
ration of redox states to reverse redox imbalanced the molecular oxygen paradox – function of mitochondrial
reactive oxygen species in cell signaling. Biol Chem 2017,
cellular environments. However, during periods of redox 298:1209–1227.
imbalance, developmental stage specific redox-sensitive
17. Gilbert HF: Thiol/disulfide exchange equilibria and disulfide
pathways can still be perturbed, resulting in failed bond stability. Methods Enzym 1995, 251:8–28.
developmental programming and poor developmental 18. Imhoff BR, Hansen JM: Differential redox potential profiles
outcomes such as structural malformations and/or during adipogenesis and osteogenesis. Cell Mol Biol Lett
neurobehavioral deficits. Clearly, moving forward to 2011, 16:149–161.
better understand environmental influences, both 19. Weinberg SE, Sena LA, Chandel NS: Mitochondria in the
physical and chemical, that act to disrupt timely regulation of innate and adaptive immunity. Immunity 2015,
42:406–417.
developmental signaling through dysregulation of redox
20. Carriere A, Carmona MC, Fernandez Y, Rigoulet M, Wenger RH,
states is an area of great human health interest and Penicaud L, Casteilla L: Mitochondrial reactive oxygen species
should be further studied to develop intervention and control the transcription factor CHOP-10/GADD153 and
prevention strategies. adipocyte differentiation: a mechanism for hypoxia-
dependent effect. J Biol Chem 2004, 279:40462–40469.
21. Kim JH, Kim SH, Song SY, Kim WS, Song SU, Yi T, Jeon MS,
Conflict of interest Chung HM, Xia Y, Sung JH: Hypoxia induces adipocyte dif-
ferentiation of adipose-derived stem cells by triggering
The authors declare no conflict of interest. reactive oxygen species generation. Cell Biol Int 2014, 38:
32–40.
References 22. Lee NK, Choi YG, Baik JY, Han SY, Jeong DW, Bae YS, Kim N,
1. Hansen JM, Harris C: Redox control of teratogenesis. Reprod Lee SY: A crucial role for reactive oxygen species in RANKL-
Toxicol 2013, 35:165–179. induced osteoclast differentiation. Blood 2005, 106:852–859.
2. Sies H: Oxidative stress. London, UK: Academic Press; 1985. 23. Juntilla MM, Patil VD, Calamito M, Joshi RP, Birnbaum MJ,
Koretzky GA: AKT1 and AKT2 maintain hematopoietic stem
3. Schafer FQ, Buettner GR: Redox environment of the cell as cell function by regulating reactive oxygen species. Blood
viewed through the redox state of the glutathione disulfide/ 2010, 115:4030–4038.
glutathione couple. Free Radic Biol Med 2001, 30:1191–1212.
24. Wu CC, Bratton SB: Regulation of the intrinsic apoptosis
4. Covarrubias L, Hernandez-Garcia D, Schnabel D, Salas-Vidal E, pathway by reactive oxygen species. Antioxidants Redox
Castro-Obregon S: Function of reactive oxygen species during Signal 2013, 19:546–558.
animal development: passive or active? Dev Biol 2008, 320:
1–11. 25. Kupsco A, Schlenk D: Oxidative stress, unfolded protein
response, and apoptosis in developmental toxicity. Int Rev
5. Castagne V, Lefevre K, Natero R, Clarke PG, Bedker DA: An Cell Mol Biol 2015, 317:1–66.
optimal redox status for the survival of axotomized gan-
glion cells in the developing retina. Neuroscience 1999, 93: 26. Ichijo H, Nishida E, Irie K, ten Dijke P, Saitoh M, Moriguchi T,
313–320. Takagi M, Matsumoto K, Miyazono K, Gotoh Y: Induction of
apoptosis by ASK1, a mammalian MAPKKK that activates
6. Paulsen CE, Truong TH, Garcia FJ, Homann A, Gupta V, SAPK/JNK and p38 signaling pathways. Science 1997, 275:
Leonard SE, Carroll KS: Peroxide-dependent sulfenylation of 90–94.
the EGFR catalytic site enhances kinase activity. Nat Chem
Biol 2011, 8:57–64. 27. Katagiri K, Matsuzawa A, Ichijo H: Regulation of apoptosis
signal-regulating kinase 1 in redox signaling. Methods Enzym
7. Truong TH, Carroll KS: Redox regulation of protein kinases. 2010, 474:277–288.
Crit Rev Biochem Mol Biol 2013, 48:332–356.
28. Anathy V, Aesif SW, Guala AS, Havermans M, Reynaert NL, Ho YS,
8. Leloup C, Tourrel-Cuzin C, Magnan C, Karaca M, Castel J, Budd RC, Janssen-Heininger YM: Redox amplification of
Carneiro L, Colombani AL, Ktorza A, Casteilla L, Penicaud L: Mito- apoptosis by caspase-dependent cleavage of glutaredoxin 1
chondrial reactive oxygen species are obligatory signals for and S-glutathionylation of Fas. J Cell Biol 2009, 184:241–252.
glucose-induced insulin secretion. Diabetes 2009, 58:673–681.
29. Dennery PA: Effects of oxidative stress on embryonic devel-
9. Dumollard R, Carroll J, Duchen MR, Campbell K, Swann K: opment. Birth Defects Res C, Embryo Today Rev 2007, 81:
Mitochondrial function and redox state in mammalian em- 155–162.
bryos. Semin Cell Dev Biol 2009, 20:346–353.

Current Opinion in Toxicology 2018, 7:110–115 www.sciencedirect.com


Oxidative stress and birth defects Hansen et al. 115

30. Karnofsky DA: Drugs as teratogens in animals and man. Annu 44. Navarova J, Ujhazy E, Dubovicky M, Mach M: Phenytoin
Rev Pharmacol 1965, 5:447–472. induced oxidative stress in pre- and postnatal rat develop-
ment – effect of vitamin E on selective biochemical variables.
31. Franks ME, Macpherson GR, Figg WD: Thalidomide. Lancet Biomed Papers Med Fac Univ Palacky Olomouc Czechoslov
2004, 363:1802–1811. 2005, 149:325–328.
32. Beedie SL, Peer CJ, Pisle S, Gardner ER, Mahony C, 45. Jones KL, Smith DW: Recognition of the fetal alcohol syn-
Barnett S, Ambrozak A, Gutschow M, Chau CH, Vargesson N, drome in early infancy. Lancet 1973, 302:999–1001.
Figg WD: Anticancer properties of a novel class of tetra-
fluorinated thalidomide analogues. Mol Cancer Ther 2015, 46. Brocardo PS, Gil-Mohapel J, Christie BR: The role of oxidative
14:2228–2237. stress in fetal alcohol spectrum disorders. Brain Res Rev
2011, 67:209–225.
33. Hansen JM, Carney EW, Harris C: Differential alteration by
thalidomide of the glutathione content of rat vs. rabbit con- 47. Akella SS, Beck MJ, Philbert MA, Harris C: Comparison of
ceptuses in vitro. Reprod Toxicol 1999, 13:547–554. in vitro and in utero ethanol exposure on indices of oxidative
stress. Vitro Mol Toxicol 2000, 13:281–296.
34. Hansen JM, Gong SG, Philbert M, Harris C: Misregulation of
gene expression in the redox-sensitive NF-kappab-depen- 48. Dreosti IE, Manuel SJ, Buckley RA: Superoxide dismutase (EC
dent limb outgrowth pathway by thalidomide. Dev Dyn Official 1.15.1.1), manganese and the effect of ethanol in adult and
Publ Am Assoc Anatomists 2002, 225:186–194. foetal rats. Br J Nutr 1982, 48:205–210.
35. Bushdid PB, Brantley DM, Yull FE, Blaeuer GL, Hoffman LH, 49. Dreosti IE, Partick EJ: Zinc, ethanol, and lipid peroxidation in
Niswander L, Kerr LD: Inhibition of NF-kappaB activity results adult and fetal rats. Biol Trace Elem Res 1987, 14:179–191.
in disruption of the apical ectodermal ridge and aberrant limb
morphogenesis. Nature 1998, 392:615–618. 50. Chen X, Liu J, Chen SY: Sulforaphane protects against
ethanol-induced oxidative stress and apoptosis in neural
36. Kanegae Y, Tavares AT, Izpisua Belmonte JC, Verma IM: Role of crest cells by the induction of Nrf2-mediated antioxidant
Rel/NF-kappaB transcription factors during the outgrowth of response. Br J Pharmacol 2013, 169:437–448.
the vertebrate limb. Nature 1998, 392:611–614.
51. Dong J, Sulik KK, Chen SY: Nrf2-mediated transcriptional in-
37. Hansen JM, Harris C: A novel hypothesis for thalidomide- duction of antioxidant response in mouse embryos exposed
induced limb teratogenesis: redox misregulation of the NF- to ethanol in vivo: implications for the prevention of fetal
kappaB pathway. Antioxid Redox Signal 2004, 6:1–14. alcohol spectrum disorders. Antioxid Redox Signal 2008, 10:
2023–2033.
38. Parman T, Wiley MJ, Wells PG: Free radical-mediated oxidative
DNA damage in the mechanism of thalidomide teratogenicity. 52. Harada M: Minamata disease: methylmercury poisoning in
Nat Med 1999, 5:582–585. Japan caused by environmental pollution. Crit Rev Toxicol
1995, 25:1–24.
39. Wong M, Wells PG: Effects of N-acetylcysteine on fetal
development and on phenytoin teratogenicity in mice. Tera- 53. Su MQ, Okita GT: Embryocidal and teratogenic effects of
tog Carcinog Mutagen 1988, 8:65–79. methylmercury in mice. Toxicol Appl Pharmacol 1976, 38:
207–216.
40. Puho EH, Szunyogh M, Metneki J, Czeizel AE: Drug treatment
during pregnancy and isolated orofacial clefts in Hungary. 54. Ni M, Li X, Yin Z, Jiang H, Sidoryk-Wegrzynowicz M, Milatovic D,
Cleft palate Craniofacial J Official Publ Am Cleft Palate Cranio- Cai J, Aschner M: Methylmercury induces acute oxidative
facial Assoc 2007, 44:194–202. stress, altering Nrf2 protein level in primary microglial cells.
Toxicol Sci Official J Soc Toxicol 2010, 116:590–603.
41. Winn LM, Wells PG: Phenytoin-initiated DNA oxidation in
murine embryo culture, and embryo protection by the anti- 55. Ornaghi F, Ferrini S, Prati M, Giavini E: The protective effects of
oxidative enzymes superoxide dismutase and catalase: evi- N-acetyl-L-cysteine against methyl mercury embryotoxicity
dence for reactive oxygen species-mediated DNA oxidation in mice. Fundam Appl Toxicol Official J Soc Toxicol 1993, 20:
in the molecular mechanism of phenytoin teratogenicity. Mol 437–445.
Pharmacol 1995, 48:112–120.
56. Thompson SA, White CC, Krejsa CM, Eaton DL, Kavanagh TJ:
42. Menon VP, Sudheer AR: Antioxidant and anti-inflammatory Modulation of glutathione and glutamate-L-cysteine ligase by
properties of curcumin. Adv Exp Med Biol 2007, 595:105–125. methylmercury during mouse development. Toxicol Sci Offi-
cial J Soc Toxicol 2000, 57:141–146.
43. Reeta KH, Mehla J, Gupta YK: Curcumin is protective against
phenytoin-induced cognitive impairment and oxidative stress
in rats. Brain Res 2009, 1301:52–60.

www.sciencedirect.com Current Opinion in Toxicology 2018, 7:110–115

Vous aimerez peut-être aussi