Vous êtes sur la page 1sur 12

Available online at www.sciencedirect.

com

NeuroToxicology 29 (2008) 190201

Review

How environmental and genetic factors combine to cause autism: A redox/methylation hypothesis
Richard Deth *, Christina Muratore, Jorge Benzecry, Verna-Ann Power-Charnitsky, Mostafa Waly
Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02468, United States Received 31 January 2007; accepted 27 September 2007 Available online 13 October 2007

Abstract Recently higher rates of autism diagnosis suggest involvement of environmental factors in causing this developmental disorder, in concert with genetic risk factors. Autistic children exhibit evidence of oxidative stress and impaired methylation, which may reect effects of toxic exposure on sulfur metabolism. We review the metabolic relationship between oxidative stress and methylation, with particular emphasis on adaptive responses that limit activity of cobalamin and folate-dependent methionine synthase. Methionine synthase activity is required for dopamine-stimulated phospholipid methylation, a unique membrane-delimited signaling process mediated by the D4 dopamine receptor that promotes neuronal synchronization and attention, and synchrony is impaired in autism. Genetic polymorphisms adversely affecting sulfur metabolism, methylation, detoxication, dopamine signaling and the formation of neuronal networks occur more frequently in autistic subjects. On the basis of these observations, a redox/methylation hypothesis of autism is described, in which oxidative stress, initiated by environment factors in genetically vulnerable individuals, leads to impaired methylation and neurological decits secondary to reductions in the capacity for synchronizing neural networks. # 2007 Elsevier Inc. All rights reserved.
Keywords: Arsenic; Attention; Attention-decit hyperactivity disorder (ADHD); D4 dopamine receptor; Folic acid; Heavy metal; Lead; Mercury; Oxidative stress; Neuronal synchronization; Pesticide; Phospholipid methylation; Thimerosal; Vitamin B12; Xenobiotic

Contents 1. 2. 3. 4. 5. 6. Sulfur metabolism and oxidative stress . . . . . . . . . . D4 dopamine receptor-mediated PLM . . . . . . . . . . Heavy metals, xenobiotics, redox and methylation . . Oxidative stress in autism . . . . . . . . . . . . . . . . . . . Redox/methylation-related genetic factors in autism. A redox/methylation hypothesis of autism . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191 193 194 195 196 197 198 198

During the past several decades the prevalence of autism and related pervasive developmental disorders in the U.S. has dramatically escalated to epidemic levels, affecting 3 in 10,000

* Corresponding author at: Northeastern University, 312 Mugar, 360 Huntington Avenue, Boston, MA 02115, United States. Tel.: +1 617 373 4064; fax: +1 617 373 8886. E-mail address: r.deth@neu.edu (R. Deth). 0161-813X/$ see front matter # 2007 Elsevier Inc. All rights reserved. doi:10.1016/j.neuro.2007.09.010

children in 1970, but 66 in 10,000 in 2002 (Rice et al., 2007). The possible origins of this increase have been the subject of considerable public debate (Blaxill, 2004), and advances in detection and broadening of the diagnostic criteria for autism have been suggested to play a role (Fombonne et al., 2006), while genetic factors are clearly important, as indicated by high concordance rates among twins and siblings (Smalley et al., 1988). However, genetic factors alone cannot account for an epidemic that developed in the relatively short period of 1020

R. Deth et al. / NeuroToxicology 29 (2008) 190201

191

years (Herbert et al., 2006). Thus environmental factors are very likely to account for the major portion of the increased prevalence of autism. Exposure to xenobiotics is an inevitable feature of contemporary life, driven by an ever increasing number of threatening chemicals found in air, water and food supplies and other materials we come in contact with during our daily routines. Heavy metals, such arsenic, lead and mercury, listed as the three highest priority hazardous substances by the U.S. Department of Health and Human Services (http:// www.atsdr.cdc.gov/cercla/05list.html), are of particularly high concern, since even low levels are associated with neurological impairments, including attention-decit hyperactivity disorder (ADHD) and lower IQ (Lanphear et al., 2005; Trasande et al., 2005; Wright et al., 2006). Other heavy metals (cadmium, antimony, manganese, nickel, etc.) exert similar effects. It has been proposed that rising rates of autism are linked to increases in vaccine-derived doses of the ethylmercury derivative thimerosal, although this remains a controversial proposal (Bernard et al., 2002). Heavy metal and xenobiotic exposure may also contribute to neurodegenerative disorders including Parkinsons and Alzheimers diseases (Domingo, 2006; Mellick, 2006; Zintzaras and Hadjigeorgiou, 2004), indicating that the human brain is an especially sensitive target. While individual xenobiotics and heavy metals each produce a unique constellation of pathological insults reecting their individual chemical reactivity, almost all such agents directly or indirectly impact cellular redox status and associated pathways of sulfur metabolism (Valko et al., 2005). Indeed, sulfur metabolism can be considered a nal common pathway of toxicity, reecting the summed inuence of diverse environmental insults. This role is no great surprise, since sulfur metabolism has evolved as a primary defense system against stressful insults, orchestrating a large number of processes to maintain normal cellular function and survival (Winyard et al., 2005). Recent studies of sulfur metabolism in children with autism reveal a pattern of abnormalities indicative of the presence of oxidative stress and impaired methylation (James et al., 2004, 2006). We previously described the shared ability of a number of neurodevelopmental toxins, including lead, mercury, thimerosal and alcohol, to potently inhibit activity of methionine synthase (MS), the ubiquitous vitamin B12 and folate-dependent enzyme that converts homocysteine (HCY) to methionine (Waly et al., 2004). As described below, MS activity is highly sensitive to oxidative stress. MS activity is also required for dopamine-stimulated phospholipid methylation (PLM), a unique signaling activity of the D4 subtype dopamine receptor, that appears to be critical for synchronization of brain activity during attention (Demiralp et al., 2007; Deth, 2003). Impaired synchronization is a feature of autism, and a large body of literature links D4 dopamine receptors to ADHD (Faraone and Khan, 2006; LaHoste et al., 1996), suggesting that impaired methylation activity of MS could limit dopaminestimulated PLM in autism and ADHD. Based upon the above, a redox/methylation hypothesis of autism is advanced, proposing that environmental insults initiate autism in genetically sensitive individuals by promoting

cellular oxidative stress and initiating adaptive responses that include reduced methylation activity. Impaired methylation in turn leads to developmental delay and decits in attention and neuronal synchronization that are hallmarks of autism. 1. Sulfur metabolism and oxidative stress All cellular functions are affected by the prevailing redox status, and sulfur metabolism plays a central role in maintaining a redox potential that is favorable for homeostasis. The cysteine-containing tripeptide glutathione (GSH) serves as the primary intracellular antioxidant, and is maintained at a remarkably high concentration (e.g. 525 mM), providing a reservoir of metabolic reducing equivalents (Akerboom et al., 1982). The ratio of reduced to oxidized forms of GSH (GSH/ GSSG) can be as high as 100, but when the rate of GSH oxidation exceeds the rate of its formation, this ratio can be dramatically reduced, creating a state of oxidative stress (Grifth, 1999). The ratio of reduced and oxidized forms of other thiols, such as cysteine and homocysteine (HCY), also contribute to cellular redox status and can equilibrate with GSH/GSSG, but they are present at much lower concentrations and consequently are less inuential. The status of protein thiols and disuldes is closely inuenced by redox status, and oxidative stress causes metabolic alterations that can disrupt normal cellular function and can lead to cell death. Some metabolic consequences of oxidative stress serve to counteract the condition by increasing the GSH to GSSG ratio. For example, activity of NADPHdependent glutathione reductase can be increased (Hamburg et al., 1994) and/or GSSG can be exported from the cell in order to restore redox balance (Suzuki and Sugiyama, 1998). However, de novo GSH synthesis is critical to maintain adequate levels of GSH and to sustain cellular redox balance. As outlined in Fig. 1A, intracellular levels of the thiol amino acid cysteine are rate limiting for GSH synthesis, thus augmenting cysteine availability is a crucial mechanism by which cells increase GSH to meet demand. There are three sources of cysteine: (1) uptake from outside of the cell; (2) protein catabolism; (3) transsulfuration of HCY. Uptake of extracellular cysteine is accomplished by specic transport proteins, and in neurons the primary protein is excitatory amino acid transporter-3 (EAAT3), so named because it also transports glutamic acid (glutamate) (Himi et al., 2003; Shashidharan et al., 1997). Recent studies show that EAAT3 protects neurons against oxidative stress by providing cysteine uptake (Aoyama et al., 2006), and evidence indicates that EAAT3 activity is increased by activation of the tyrosine kinase-signaling pathway (Fournier et al., 2004), implying that neuronal growth factors can promote neuronal survival by increasing cysteine uptake and GSH synthesis. Catabolism of proteins is increased in response to stress, and the released cysteine and methionine can be utilized for GSH synthesis. The proteasome is the primary source of intracellular protease activity, cleaving ubiquitin-tagged proteins to release their amino acids. Ubiquitin ligase activity is regulated by modications to active site cysteine residues (Obin et al., 1998), providing redox

192

R. Deth et al. / NeuroToxicology 29 (2008) 190201

Fig. 1. Adaptations of sulfur metabolism to oxidative stress. (Panel A) Methylation and redox buffering activities are equally supported by the methionine cycle and transsulfuration during normal redox conditions. (Panel B) During oxidative stress multiple adaptive mechanisms shift the ux of sulfur resources toward GSH synthesis, including reduced activity of methionine synthase, increased activity of cystathionine-b-synthase (CBS) and decreased activity of cysteine dioxygenase (CDO). Lower methionine synthase activity reduces methylation, including dopamine-stimulated phospholipid methylation and its role in attention.

regulation of proteolysis. However, since cysteine is a rarely utilized amino acid, increased protein catabolism must be considered an option of last resort for augmenting GSH synthesis. Cysteine is synthesized via transsulfuration (Fig. 1A) and its availability for GSH synthesis can be increased by diverting more HCY out of the methionine cycle to transsulfuration. Control of this metabolic branchpoint is a fundamental adaptive response for regulating cellular redox status. Moreover, relative activities of methionine synthase (MS) and cystathionine-bsynthase (CBS) determine the balance between methylation and redox buffering, and both enzymes are responsive to cellular oxidative status (Banerjee et al., 2003; Deplancke and Gaskins, 2002; Ludwig and Matthews, 1997; Persa et al., 2004). Cysteine dioxygenase (CDO) utilizes molecular oxygen to convert cysteine to cysteine sulnate, which is further metabolized to sulfate and taurine (Fig. 1A), competing with GSH synthesis for available cysteine. In response to lower cysteine levels and oxidative stress, CDO degradation by the

ubiquitin/proteasome system is accelerated (Stipanuk et al., 2004), increasing cysteine availability for GSH synthesis (Fig. 1B), another adaptive response of sulfur metabolism to oxidative stress. CBS is a vitamin B6 (pyridoxal)-dependent enzyme catalyzing ligation of HCY and serine to form cystathionine, which is subsequently hydrolyzed to cysteine and a-ketobutyrate by cystathionine-g-lyase (CGL). CBS contains a heme group that monitors redox status, and its oxidation to the ferric state is associated with increased activity (Banerjee et al., 2003). CBS activity is negatively regulated by its carboxy-terminal domain, but binding of the methyl donor S-adenosylmethionine (SAM) relieves this inhibition, such that transsulfuration is normally restricted unless adequate SAM levels are achieved. In response to oxidative stress, the SAM-binding regulatory domain is cleaved by a ubiquitin/proteasome-dependent mechanism, increasing CBS activity and rendering it SAM-independent. Thus oxidative stress augments transsulfuration to increase de novo GSH synthesis, and methylation capacity is diminished as a result. Testosterone decreases CBS activity, lowers GSH levels and increases susceptibility to oxidative stress (Prudova et al., 2007), which may account for the higher prevalence of autism in males. Restricting MS activity promotes HCY diversion toward GSH synthesis, and acute oxidative stress simultaneously decreases MS activity and increases CBS activity (Persa et al., 2004). During evolution different strategies for MS inhibition have been utilized. In plants (e.g. Arabidopsis) and E. coli, MS inhibition is accomplished by thiolation, wherein accumulating GSSG reacts with an active-site cysteine to provide inactivation (Dixon et al., 2005; Hondorp and Matthews, 2004). In higher species, including man, oxidative stress rapidly inhibits MS by promoting oxidation of its cobalamin (vitamin B12) cofactor (Ludwig and Matthews, 1997). Indeed, the biosynthetic pathway for cobalamin appears to have evolved as a metabolic adaptation to an increasingly oxidative environment (Raymond and Segre, 2006). The cobalt atom of cobalamins corrin ring, which provides the essence of MS activity, exists in different oxidation states during the enzymatic cycle (Fig. 2B). In its Cbl(I) state it abstracts a methyl group from methylfolate to form methylcobalamin (MeCbl) with cobalamin in its Cbl(III) state (Evans et al., 2004). Cbl(I) is regarded as a super-nucleophile and can readily oxidize to inactive Cbl(II), depending upon whether or not it encounters an oxidizing molecule (e.g. reactive oxygen species (ROS) or electrophilic xenobiotic metabolites) in its local environment (Liptak and Brunold, 2006). As such, Cbl(I) serves as an exquisitely sensitive redox sensor for the intracellular environment, and when it oxidizes, MS activity is temporarily halted, leading to increased GSH synthesis. The sensitivity of Cbl(I) to oxidation is restricted by a cap domain that hovers above the vulnerable cobalt atom while it awaits the next methyl group from methyl methylfolate (Bandarian et al., 2002). Cobalamin inactivation is thus another adaptive mechanism to maintain cellular redox balance. Notably, the probability of Cbl(I) oxidation increases when methylfolate levels are low, since it must wait longer to be

R. Deth et al. / NeuroToxicology 29 (2008) 190201

193

Fig. 2. Structure and function of methionine synthase. (Panel A) Methionine synthase contains ve domains and a cobalamin cofactor. Composite molecular model based upon structures from Bandarian et al. (2002) and Evans et al. (2004). Methylfolate and homocysteine domains alternate in transferring a methyl group to and from cobalamin, while the cap domain partially protects cobalamin from oxidation while it awaits methylation. The SAM-binding domain provides a methyl group to oxidized cobalamin, reactivating the enzyme. (Panel B) During primary turnover Cbl(I) is vulnerable to oxidation, depending upon prevailing levels of reactive oxygen species (ROS) and electrophilic xenobiotic metabolites. Formation of methylcobalamin, either via the SAM-binding domain and methionine synthase reductase or via replacement of oxidized Cbl(II), allows enzyme reactivation.

SAH accumulate, and SAH is a powerful inhibitor of methylation reactions (Fig. 1B). Thus oxidative stress leads not only to inhibition of HCY methylation by MS, but also to a general inhibition of cellular methylation reactions, including DNA methylation and phospholipid methylation as important examples. Decreased DNA methylation, such as that induced by oxidative stress, increases expression of genes under the negative inuence of methylation, including genes that promote GSH synthesis and/or alleviate oxidative stress, or otherwise participate in the inammatory response (Chen and Kunsch, 2004; Fratelli et al., 2005). While adaptive epigenetic responses may be critical for cell survival, particularly in the short-term, they also interrupt normal cellular function, depending upon the intensity and duration of the oxidative challenge. Transient exposure to oxidative stressors normally allows sulfur metabolism and epigenetic patterns to return to normal, reversing adaptive responses as GSH levels return to homeostatic values. However, prolonged exposure to heavy metals and xenobiotics can cause long-lasting adaptive epigenetic responses with pathologic consequences, and the particular pathological manifestation (i.e. the particular oxidative stress-induced disease) may reect an individuals genetic background, reected in his/her pattern of single nucleoside polymorphisms (SNPs). Risk-associated SNPs may alter amino acids in the protein product (e.g. enzyme), inuence transcription efciency or otherwise affect the role of the gene, but are distinct from de novo mutations in that they occur in 1% or more of the population, and contribute to normal diversity. Thus increased exposure to environmental stressors places an entire population at risk, but genetically vulnerable subpopulations are most likely to manifest a particular disorder, such as autism. In this regard, increased oxidative stress can be viewed as a condition where certain genetic variations prove useful or harmful. 2. D4 dopamine receptor-mediated PLM Dopamine plays a key role in attention. Among ve dopamine receptor subtypes, the D4 receptor has the unique ability to transfer folate-derived methyl groups to the plasma membrane phospholipid phosphatidylethanolamine (PE), a process known as dopamine-stimulated phospholipid methylation or PLM (Sharma et al., 1999; Zhao et al., 2001). Levels of PE in erythrocytes of autistic children are signicantly reduced (Chauhan and Chauhan, 2006). The molecular basis for dopamine-stimulated PLM lies in a methionine residue (MET313), unique to the D4 receptor, participating in a methylation cycle paralleling the methionine cycle (Fig. 1A). However, while the methionine cycle utilizes methionine as a source of methyl groups, dopamine-stimulated PLM is absolutely dependent upon methylfolate and MS activity. Consequently, reductions in MS activity, such as those brought about by oxidative insults, will directly reduce dopaminestimulated PLM (Fig. 1B). When PE is methylated in response to dopamine, membrane uidity in the D4 receptor microenvironment is increased since methylated PE occupies more space and lipid-packing density

methylated, implying that lower activity of methylenetetrahydrofolate reductase (MTHFR) will promote MS inhibition and increased GSH synthesis. Reactivation of MS after Cbl(I) oxidation is accomplished by converting Cbl(II) to MeCbl. In the classical mechanism Cbl(II) is reduced to Cbl(I) by methionine synthase reductase (MTRR), followed by addition of a SAM-derived methyl group provided by the SAM-binding domain (Bandarian et al., 2002). In an alternative mechanism that requires Cbl(II) dissociation, Cbl(II) is converted to hydroxocobalamin, which reacts with GSH to form glutathionylcobalamin that is further converted to MeCbl in a SAM-dependent reaction (Pezacka et al., 1990). Since it requires GSH, the latter mechanism is highly attuned to redox status, assuring that MS will only be reactivated when GSH levels are adequate. When MS activity is inhibited by oxidative stress, it not only reduces methylation of HCY, but also inhibits all methylation reactions, exerting a broad and powerful inuence. HCY formation from S-adenosylhomocysteine (SAH) hydrolysis during the methionine cycle is a reversible reaction, and SAH synthesis from adenosine and HCY is thermodynamically favored (Ueland, 1982). When MS is inactivated, both HCYand

194

R. Deth et al. / NeuroToxicology 29 (2008) 190201

is decreased. Dopamine-stimulated PLM is estimated to reach a turnover rate of 2050 methylations/sec/receptor (Deth, 2003), allowing dopamine to rapidly alter local membrane properties. This biophysical effect serves as a membrane-delimited signaling mechanism initiated by the D4 receptor that can inuence the activity of nearby membrane proteins. We proposed that this unique mechanism allows D4 receptors to modulate the resonance frequency of neural networks during dopamine-induced attention (Deth, 2003; Deth et al., 2004; Kuznetsova and Deth, 2007). Consistent with this proposal, D4 receptor activation promotes a shift of neuronal network ring to gamma frequency during attention (Demiralp et al., 2007), while D4 receptor blocking drugs reduce gamma frequency synchronization during attention (Ahveninen et al., 2000), and interfere with synchronization-dependent learning (Laviolette et al., 2005). While a role for D4 receptors in attention and neuronal synchronization is well supported in the literature, involvement of dopamine-stimulated PLM in these events has not yet been demonstrated, and the sequence of events outlined above therefore remains speculative. The human D4 receptor displays a distinctive repeat motif found only in primates, and there is a remarkable degree of individual variability in the number and composition of repeats. A 48 bp sequence in the D4 receptor gene is present as a 211fold repeat and 35 different versions of the sequence have been identied, making it one of the most variable human genes (Wang et al., 2004). The repeat sequence codes for proline-rich segments in the receptor that serve as attachment sites for SH3 domain-containing signaling and scaffold proteins (Oldenhof et al., 1998). Thus the D4 receptor serves as a center for multiple forms of signal generation, involving not only classical G protein pathways, but also tyrosine kinase, MAP kinase, and NF-kB pathways (Oak et al., 2001; Zhen et al., 2001). PLMinduced changes in membrane uidity can modulate the energy barrier for conformational motions of integral membrane proteins, including ion channels or transporters, enzymes and receptors, and this modulation can alter resonance properties of neurons and neuronal assemblies, shifting attended information to gamma frequency (Deth et al., 2004). Analysis of a large, worldwide sample showed that the fourrepeat D4 receptor allele is most common ($65%), followed by the seven-repeat ($25%) and two-repeat forms ($5%), although there are large differences between ethnic groups (Chang et al., 1996). There is evidence that the seven-repeat allele arose by a relatively recent mutational event about 50,000 years ago, and that it exhibits positive selection (Wang et al., 2004). The sevenrepeat allele is associated with increased novelty-seeking behaviors (Benjamin et al., 1996; Ebstein et al., 1996), and the level of attention-associated gamma synchrony is greater in subjects with the seven-repeat allele, as compared to two or four repeats (Demiralp et al., 2007). However, presence of the sevenrepeat allele is also associated with a three- to vefold higher risk of ADHD (LaHoste et al., 1996; Faraone and Khan, 2006), and contributes to lower IQ in the ADHD cohort, in conjunction with a SNP in the dopamine transporter (Mill et al., 2006). We found that dopamine-stimulated PLM is lower for the seven-repeat form vs. two- or four-repeat, but the potency of dopamine is

greater and dopamine activation of methionine synthase is greater for the seven-repeat form of the receptor (Deth et al., 2004). These differences may be relevant to the increased ADHD risk associated with the seven-repeat receptor, but the frequency of the seven-repeat allele is not increased in autism (Grady et al., 2005). Similar to autism, the prevalence of ADHD has markedly increased during the past several decades, and the 4:1 predominance of males in ADHD is similar to autism. Since ADHD is associated with elevated plasma levels of lead and mercury (Braun et al., 2006; Cheuk and Wong, 2006), oxidative stress and lower MS activity might contribute to its pathogenesis. Froehlich et al. (2007) examined the interaction between D4 receptor repeat alleles and the severity of leadinduced neurological impairment. Performance on an attentionrelated task decreased in proportion to documented blood lead levels, and the level of impairment was signicantly greater at any level of lead for boys lacking the seven-repeat allele, but not for girls, and not in boys carrying the seven-repeat allele. Thus the seven-repeat allele of the D4 receptor appears to confer protection against lead-induced cognitive impairments, at least in boys, representing an example of a gene-environment interaction. However, the seven-repeat allele was associated with signicantly poorer performance on a working memory task for both boys and girls. Additional studies are needed to clarify what appears to be a complex relationship between D4 receptor genotype, heavy metal sensitivity and gender. 3. Heavy metals, xenobiotics, redox and methylation The ability of heavy metals to bind to thiol groups and to disrupt pathways of sulfur metabolism is well established. Indeed, the traditional name for thiols is mercaptans, recognizing their afnity for mercury. Sulfur metabolism is important for the excretion of xenobiotics (e.g. sulfation and formation of mercapturic acid derivatives) and their oxidized metabolites contribute to oxidative stress. Since many pesticides and preservatives function by disrupting redox events, it is not surprising they should exert similar effects in humans. Cysteine residues play critical roles in most proteins, so it is difcult, if not impossible, to identify a specic protein as the critical target for heavy metal toxicity. Cysteine residues are common participants in catalysis and transfer reactions, since the sulfur bond allows adducts to form and subsequently be released. Heavy metals such as mercury bind tightly to the thiolate anion, and in its divalent state the inorganic mercuric cation can simultaneously bind two thiolates, increasing its retention to almost irreversible levels. Cysteine residues are commonly viewed as simple reduced thiols (SH); however, under physiological conditions they also exist as a mixture of modied forms, including mixed disuldes with glutathione, cysteine and homocysteine, oxided forms including sulfenic acid (SOH), sulnic acid (SO2H), and sulfonic acid (SO3H), or S-nitrosocysteine (SNO). These modications play a central role in orchestrating cellular metabolism, especially during oxidative stress, and binding of heavy metals to thiol groups disrupts this orchestration.

R. Deth et al. / NeuroToxicology 29 (2008) 190201

195

While almost all proteins can be inhibited by heavy metals at sufcient concentrations, environmental exposures will preferentially affect the most sensitive targets. Analysis of neurological decits as a function of plasma lead concentrations failed to identify a threshold level that could be considered as safe, with cognitive decits still observed at concentrations below 10 mg/dl (0.5 mM) (Lanphear et al., 2005); In addition, ADHD has been reported to associated with elevated plasma mercury levels (0.4 mg/dl or 20 nM) (Cheuk and Wong, 2006). Candidate targets for heavy metal-induced neurological toxicity should therefore be inhibited at these concentrations or below. MS-dependent PLM activity in human neuronal cells is exceptionally sensitive to heavy metals, with IC50 values of 0.5 and 0.1 mM for lead and mercury, and 0.05, 0.04, 0.2 and 0.1 nM for arsenic, cadmium, antimony and thimerosal, respectively (Waly et al., 2004; Waly and Deth, unpublished data). Thus neuronal MS activity can be considered a candidate target for causing heavy metal-associated ADHD, and may also be a candidate for causing autism. Cellular levels of GSH are signicantly lowered by mercury and other heavy metals, although the precise cause remains unclear (Agrawal et al., 2006; Sakurai et al., 2005; Shenker et al., 1993). However, the decrease in GSH is not associated with a large increase of GSSG, and therefore cannot be attributed to a simple shift in redox status, but rather to a reduction in the total GSH/GSSG pool. This could reect decreased GSH synthesis, increased extrusion of GSH or increased GSH metabolism. Consistent with the latter mechanism, it has been proposed that binding of divalent mercury to GSH facilitates cleavage of its gamma-glutamyl residue (Rubino et al., 2006). As reviewed by Schafer and Buettner (2001), GSH/GSSG redox status exerts a broad inuence on cellular activities, including proliferation, differentiation and survival. Conjugation of xenobiotics to GSH, either directly or glutathione-S-transferase catalyzed, is a common mechanism for their metabolism and eventual clearance from the body. Increased exposure therefore stresses sulfur metabolism and competes with redox buffering for available GSH. Conversely, clearance of xenobiotics, as well as heavy metals, is delayed under oxidative stress conditions, prolonging their residence in the body and increasing their opportunity to exert toxic effects. Xenobiotics are substrates for cytochrome P-450 enzymes, yielding oxidized products including hydroxides, quinones or epoxides. The latter electrophilic products readily react with the supernucleophile Cob(I) state of cobalamin, leading to formation of inactive alkylcobalamin adducts (Watson et al., 2004). However, GSH-dependent conversion of Cbl(I) to gluthionylcobalamin protects against alkylation, which may be important for conserving MS activity in the presence of xenobiotics. Depleted GSH levels would therefore increase MS sensitivity to xenobiotics. Some heavy metals, such as mercury, arsenic and antimony, are methylated in a biological environment, and their organoderivatives exhibit distinctly different distribution and toxicity proles. Methylmercury readily crosses the blood brain barrier and is one of the most potent neurotoxicants known (Sanfeliu

et al., 2003). In the brain methylmercury is de-methylated to inorganic mercury, which has a very slow clearance rate (i.e. years). A comparative study in primates showed that ethylmercury derived from the vaccine preservation thimerosal releases more inorganic mercury in the brain than is released by methylmercury (Burbacher et al., 2005). Arsenic is mono- or di-methylated via a SAM-dependent methyltransferase (Thomas et al., 2007), while antimony is methylated using methionine as the methyl donor (Dopp et al., 2004). Recent reports of high arsenic levels in chicken [arsenicals are administered to increase growth rates], raises concern about its possible adverse effects on methylation-regulated processes (Lasky et al., 2004). The high sensitivity of neuronal tissue to heavy metalinduced oxidative stress and resultant inhibition of methylation may reect lower transsulfuration activity in neurons. Initially it was reported that neurons lacked cystathionase activity (Finkelstein, 1990), consistent with very high levels of cystathionine (Tallan et al., 1958). Neurons are therefore highly dependent upon cystine and cysteine uptake for GSH synthesis, and are more vulnerable to heavy metal-induced oxidative stress. However, functional transsulfuration was recently demonstrated in cultured neurons and in fetal brain, including a signicant decrease in GSH levels upon inhibition of cystathionase (Vitvitsky et al., 2006). While additional studies are required, transsulfuration does appear to occur in neurons, although cysthathionase activity is limited compared to other tissues. 4. Oxidative stress in autism As previously reviewed (Chauhan and Chauhan, 2006; Kern and Jones, 2006; McGinnis, 2004) there is mounting evidence of oxidative stress and inammation in autism. Plasma levels of methionine cycle and transsulfuration metabolites are reported to be abnormal in autistic individuals (Geier and Geier, 2006; James et al., 2004, 2006). Adenosine and SAH levels are increased while HCY, methionine and SAM levels are low, consistent with reduced MS activity and increased CBS activity, while the SAM/SAH ratio is signicantly reduced, indicating impaired methylation capacity. Cystathionine, cysteine and GSH levels are each reduced along with the GSH/GSSG ratio, reecting increased oxidative stress. Elevated HCY levels have also been reported in autism (Pasca et al., 2006). Supplementation with a combination of betaine (trimethylglycine) and folinic acid (5-formylTHF) normalized methionine cycle metabolites, but transsulfuration metabolites remained abnormal (James et al., 2004). Upon further addition of methylcobalamin, levels of all metabolites, as well as SAM/ SAH and GSH/GSSG ratios returned to normal. If these abnormal metabolic proles are conrmed by others, they will represent a critically important clue to the origins of autism. Oxidative stress in autism is associated with increased plasma levels of malonyldialdehyde, urinary levels of fatty acid and lipid peroxidation biomarkers (Chauhan et al., 2004; Ming et al., 2005; Yao et al., 2006; Zoroglu et al., 2004). Elevated levels of inammatory cytokines and evidence of microglial

196

R. Deth et al. / NeuroToxicology 29 (2008) 190201

activation microglial activation was observed in post-mortem brain sections indicating the presence of neuroinammation (Vargas et al., 2005). Microglia monitor the local environment and provide a macrophage-like function in the brain, releasing pro-inammatory substances upon activation. In addition, microglia take-up organic mercury and convert it to the more toxic inorganic mercury (Charleston et al., 1995), and in primate cortex, chronic methylmercury exposure leads to a large increase in activated microglia (Charleston et al., 1994). Heavy metals can therefore cause oxidative stress in neurons not only by their direct inuence on sulfur metabolism, but also by promoting microglia-based neuroinammation. 5. Redox/methylation-related genetic factors in autism As noted above, genetic risk factors play a critical role in autism, particularly as they combine with environmental exposures (for a review see Persico and Bourgeron, 2006) and a number of mutations and SNPs have been identied that have special relevance for oxidative stress and impaired methylation. A rare purely genetic form of autism is caused by mutations affecting the enzyme adenylosuccinate lysase (ASL) (Stone et al., 1992). ASL is required for de novo purine synthesis, a pathway associated with a number of inborn errors of metabolism causing developmental disorders. ASL mutations divert an extraordinary proportion of folate-derived carbon atoms toward purine synthesis in an effort to offset impaired enzyme activity, reducing the availability of methylfolate for MS. Autism is a prominent feature of Rett syndrome, commonly caused by mutations in the MeCP2 gene, which encodes a protein that binds to methylated DNA and promotes gene silencing (Amir et al., 1999). Fragile-X syndrome, which can include autism, is caused by expansion of CpG methylation sites in the FMR-1 gene (McConkie-Rosell et al., 1993), and folate deciency increases fragility at the FMR-1 locus (Hagerman et al., 1983). Dendritic spine density is reduced in Fragile-X (Irwin et al., 2000), which may weaken the ability to modulate neural networks. Several studies have found an association between autism and chromosomal defects involving 15q1113, a region subject to methylation-dependent genomic imprinting containing genes for a type 3 ubiquitin ligase (UBE3A) (Baker et al., 1994; Bolton et al., 2004; Bundey et al., 1994). This region also codes for a translocase (ATP10C) responsible for maintaining high levels of the phospholipid PE at the inner membrane surface where it serves as substrate for D4 receptor-mediated PLM (Herzing et al., 2001). Mutations in 15q1113 are linked to Angelman, Prader-Willi and Rett syndromes in addition to autism (Thatcher et al., 2005), and knockout of the Rettassociated MeCP2 gene also results in reduced levels of UBE3A (Samaco et al., 2005), indicating broad involvement of this locus in developmental disorders. Decreased plasma adenosine deaminase (ADA) activity was rst reported in autistic subjects, by Stubbs et al. (1982). Several studies subsequently reported a higher frequency of a lesser active ADA allele among autistic subjects from an Italian kindred (Lucarelli et al., 2002; Persico et al., 2000). Lower

ADA activity leads to adenosine accumulation, increased SAH levels, decreased HCY levels, and reduced transsulfuration, a pattern found in autism (James et al., 2004, 2006). Methylfolate, the primary circulating form of folate, is transported into cells by the reduced folate carrier (RFC), which can exhibit a SNP (A80G) associated with elevated levels of HCY (Chango et al., 2000), whose frequency is increased in autism (James et al., 2006). Methylfolate is synthesized by methyltetrahydrofolate reductase (MTHFR) and the MTHFR gene exhibits two common polymorphisms, C677T and A1298C. Homozygosity for C677T reduces enzyme activity and elevates HCY levels, particularly when folate levels are low (Molloy et al., 1997), while A1298C reduces MTHFR activity, but without elevating HCY (Friedman et al., 1999). Boris et al. (2004) found a higher frequency of homozygous and heterozygous C677T genotypes among autistic subjects (23% and 56%) vs. controls (11% and 41%), and compound heterozygotes were also more common among autistic subjects (25%) than controls (15%). James et al. (2006) did not nd a signicant association of C677T or A1298C with autism when each was evaluated individually, but they contributed to an increased risk when combined with other SNPs. Transcobalamin II (TCN2) facilitates cellular uptake of cobalamin, and a C776G SNP in TCN2, lowers its afnity for cobalamin (Miller et al., 2002). Homozygosity for C776G is associated with lower plasma levels of the transcobalamin::cobalamin complex and increased HCY levels, and homozygosity for C776G is more common in autistic children (26%) vs. controls (16%) (James et al., 2006). Thus intracellular cobalamin levels are likely to be lower in autism, placing methionine synthase activity at risk. Glutathione-S-transferase M1 (GSTM1), which conjugates GSH to toxic electrophiles, is reduced or absent in individuals carrying the GSTM1*0 (null) allele, increasing their sensitivity to xenobiotics (Hung et al., 2004). Two studies have reported an association between the null allele and autism (Buyske et al., 2006; James et al., 2006), suggesting that GST*M1 contributes to the risk of oxidative stress and autism. Paraoxonase 1 (PON1) detoxies organophosphate pesticides, and its activity is lower in serum of autistic subjects, in association with elevated levels of HCY and lower levels of cobalamin (Pasca et al., 2006). SNPs in the PON1 gene that lower its activity are more common in autistic subjects in the U.S., but not in Italian subjects, which corresponds with a much higher use of organophosphates in the U.S. (DAmelio et al., 2005). PON1 also is responsible for hydrolysis of a reactive cyclic form of homocysteine, homocysteine thiolactone, which decreases insulin release and insulin responsiveness in a redox-dependent manner (Najib and Sanchez-Margalet, 2001; Patterson et al., 2007). Catechol-O-methyltransferase (COMT) inactivates dopamine and other catecholamine neurotransmitters and exhibits a polymorphism (G472A) yielding a V158M substitution in the protein that lowers enzyme activity three- to fourfold (Lachman et al., 1996). Homozygosity for G472A is higher in autistics (26%) vs. controls (16%) (James et al., 2006), although the A allele is usually associated with increased cognitive abilities

R. Deth et al. / NeuroToxicology 29 (2008) 190201

197

(Malhotra et al., 2002). An autism-associated decrease in methylation capacity could synergize with lower activity of the V158M enzyme to produce a large increase in dopamine levels, and impaired MS activity may not sustain an adequate supply of methyl groups to the D4 receptor under these circumstances. Reelin, a product of the RELN gene, is an extracellular protease participating in the migration of cortical neurons, particularly parvalbumin-expressing GABAergic interneurons, during development and also modulates neuronal ring activity and long-term potentiation (Beffert et al., 2006; Fatemi, 2005). Reelin expression is subject to epigenetic regulation by methylation (Chen et al., 2002), and lower brain levels are found in autism (Fatemi et al., 2001), suggesting hypermethylation of the RELN locus. Consistent with this relationship, variants of RELN involving repeat sequences in the 50 -UTR are associated with autism (Persico et al., 2001). D4 dopamine receptors are abundant in the parvalbumin-expressing GABAergic interneurons that produce reelin (Mrzljak et al., 1996), and networks containing these interneurons are important in generating gamma frequency oscillations during attention (Bartos et al., 2007). Development of parvalbuminexpressing interneurons requires hepatocyte growth factor/ scatter factor and its tyrosine kinase-linked receptor MET, and a recent study found a higher frequency of a SNP that lowers MET transcription in autistic subjects (Campbell et al., 2006). Synchronized gamma activity is reduced in autism (Wilson et al., 2006), which may reect impaired dopamine-stimulated PLM in the context of SNPs affecting reelin, MET and other determinants of interneuron networks. Autism-associated mutations in neuroligin (NLGN3 and NLGN4) (Laumonnier et al., 2004), which stabilizes synapses, may also affect synchronization of neuronal networks. 6. A redox/methylation hypothesis of autism The preceding observations support a redox/methylation hypothesis of autism. As summarized in Fig. 3, genetic and environmental factors both play fundamental roles in dening the risk of autism, although their relative contribution can vary greatly. Genetic factors are sufcient for mutations of ASL, Rett and Angelman/Prader-Willi syndromes, while the occurrence of autism in Fragile-X syndrome and other intermediate examples (e.g. tuberous sclerosis) depends upon additional genetic or environmental factors. However, most autism cases arising during the past two decades undoubtedly reect a major role for environmental factors, including, but not limited to, heavy metal and xenobiotic exposure. In these cases, genetic factors still dene the at-risk population, but instead of frank mutations, risk arises from combinations of polymorphisms (SNPs) carried by signicant proportions of the human population. In a particular individual the likelihood and severity of oxidative stress in response to a potentially toxic environmental exposure depends upon the presence or absence of SNPs directly or indirectly affecting sulfur metabolism and/ or other metabolic systems that respond to such exposures (e.g. PON1, GSTM1*0). The level of MS inhibition and impaired methylation depends upon the extent of oxidative stress, but

Fig. 3. A redox/methylation hypothesis of autism. Environmental factors (e.g. heavy metals and xenobiotics) can precipitate oxidative stress in a vulnerable subpopulation possessing risk genes (shown in italics), initiating multiple adaptive responses involving sulfur metabolism. Inhibition of methionine synthase broadly reduces methylation activity, with DNA methylation and dopamine-stimulated phospholipid methylation being important examples. Reduced DNA methylation interferes with epigenetic events that are fundamental to normal development. Impairment of dopamine-stimulated phospholipid methylation limits frequency-dependent synchronization of neuronal networks, reected as decits in attention and cognition. While all cell types are subject to similar effects, which may be manifested as autism-associated symptoms, neuronal cells exhibit higher sensitivity to oxidative stress.

also on SNPs affecting cobalamin and folate status, as well as SNPs affecting enzymes and metabolites of the methionine cycle (e.g. MTHFR, RFC, TCN2). A lower SAM/SAH ratio reduces the probability of DNA methylation, with consequences for epigenetic regulation of gene expression and its pivotal role in developmental trajectory, and SNPs impacting any of the multiple steps leading to gene silencing or imprinting will inuence the severity of disruption. Since oxidative stress is a systemic feature of autism (James et al., 2004, 2006), consequences of impaired methylation and epigenetic disruption will also be expressed in non-neuronal tissues, giving rise to diverse symptoms such immune or GI dysfunction, which are commonly seen in autism. Since D4 receptor-mediate dopamine-stimulated PLM is absolutely dependent upon MS activity, SNPs promoting oxidative stress and impaired methylation confer risk to its role in synchronizing neural networks, synergizing with SNPs affecting dopaminergic function (e.g. COMT) and/or the neuronal substrates participating in synchronization (e.g. RELN, MET or NGLN3/4). The risk of autism can theoretically be inuenced by SNPs acting at any level in metabolic and neuroanatomic pathways supporting neuronal synchronization, which is essential for complex abilities that are a hallmark of the human brain. These SNPs have presumably been retained because they can, in certain circumstances, contribute in a positive manner to attentive and cognitive abilities. However, in a more challenging environment, such as increased exposure to heavy metals and xenobiotics, these same features provide a source of risk.

198

R. Deth et al. / NeuroToxicology 29 (2008) 190201 Bernard S, Enayati A, Roger H, Binstock T, Redwood L. The role of mercury in the pathogenesis of autism. Mol Psychiatry 2002;7(Suppl 2):S423. Blaxill MF. Whats going on? The question of time trends in autism Public Health Rep 2004;119:53651. Bolton PF, Veltman MW, Weisblatt E, Holmes JR, Thomas NS, Youings SA, et al. Chromosome 15q1113 abnormalities and other medical conditions in individuals with autism spectrum disorders. Psychiatr Genet 2004;14:1317. Boris M, Goldblatt A, Galanko J, James SJ. Association of MTHFR gene variants with autism. J Am Phys Surg 2004;9:1068. Braun JM, Kahn RS, Froehlich T, Auinger P, Lanphear BP. Exposures to environmental toxicants and attention decit hyperactivity disorder in U.S. children. Environ Health Perspect 2006;114:19049. Bundey S, Hardy C, Vickers S, Kilpatrick MW, Corbett JA. Duplication of the 15q1113 region in a patient with autism, epilepsy and ataxia. Dev Med Child Neurol 1994;36:73642. Burbacher TM, Shen DD, Liberato N, Grant KS, Cernichiari E, Clarkson T. Comparison of blood and brain mercury levels in infant monkeys exposed to methylmercury or vaccines containing thimerosal. Environ Health Perspect 2005;113:101521. Buyske S, Williams TA, Mars AE, Stenroos ES, Ming SX, Wang R, et al. Analysis of case-parent trios at a locus with a deletion allele: association of GSTM1 with autism. BMC Genet 2006;7:8 [Epub ahead of print]. Campbell DB, Sutcliffe JS, Ebert PJ, Militerni R, Bravaccio C, Trillo S, et al. A genetic variant that disrupts MET transcription is associated with autism. Proc Natl Acad Sci USA 2006;103:168349. Chang FM, Kidd JR, Livak KJ, Pakstis AJ, Kidd KK. The world-wide distribution of allele frequencies at the human dopamine D4 receptor locus. Hum Genet 1996;98:91101. Chango A, Emery-Fillon N, de Courcy GP, Lambert D, Pster M, Rosenblatt DS, et al. A polymorphism (80G ! A) in the reduced folate carrier gene and its associations with folate status and homocysteinemia. Mol Genet Metab 2000;70:3105. Charleston JS, Bolender RP, Mottet NK, Body RL, Vahter ME, Burbacher TM. Increases in the number of reactive glia in the visual cortex of Macaca fascicularis following subclinical long-term methyl mercury exposure. Toxicol Appl Pharmacol 1994;129:196206. Charleston JS, Body RL, Mottet NK, Vahter ME, Burbacher TM. Autometallographic determination of inorganic mercury distribution in the cortex of the calcarine sulcus of the monkey Macaca fascicularis following long-term subclinical exposure to ethylmercury and mercuric chloride. Toxicol Appl Pharmacol 1995;132:32533. Chauhan A, Chauhan V. Oxidative stress in autism. Pathophysiology 2006;13:17181. Chauhan A, Chauhan V, Brown WT, Cohen I. Oxidative stress in autism: increased lipid peroxidation and reduced serum levels of ceruloplasmin and transferringthe antioxidant proteins. Life Sci 2004;75:253949. Chen Y, Sharma RP, Costa RH, Costa E, Grayson DR. On the epigenetic regulation of the human reelin promoter. Nucl Acids Res 2002;30:29309. Chen XL, Kunsch C. Induction of cytoprotective genes through Nrf2/antioxidant response element pathway: a new therapeutic approach for the treatment of inammatory diseases. Curr Pharm Des 2004;10:87991. Cheuk DK, Wong V. Attention-decit hyperactivity disorder and blood mercury level: a case-control study in Chinese children. Neuropediatrics 2006;37: 23440. DAmelio M, Ricci I, Sacco R, Liu X, DAgruma L, Muscarella LA, et al. Paraoxonase gene variants are associated with autism in North America, but not in Italy: possible regional specicity in geneenvironment interactions. Mol Psychiatry 2005;10:100616. Demiralp T, Herrmann CS, Erdal ME, Ergenoglu T, Keskin YH, Ergen M, et al. DRD4 and DAT1 polymorphisms modulate human gamma band responses. Cereb Cortex 2007;17:100719. Deplancke B, Gaskins HR. Redox control of the transsulfuration and glutathione biosynthesis pathways. Curr Opin Clin Nutr Metab Care 2002;5:8592. Deth RC. Molecular origins of attention: the dopamine-folate connection. Amsterdam: Kluwer Academic Publishers; 2003.

We hope that our redox/methylation hypothesis promotes improved understanding of the molecular origins of autism. The validity of any hypothesis requires that it account for relevant and previously disparate observations. Our redox/methylation hypothesis does integrate ndings across genetic, biochemical, and neurological domains, but does not explicitly account for all autism observations (e.g. abnormalities in brain size, myelination patterns or serotonin levels). However, it may serve as a useful starting point that can be critically tested and accordingly revised or even discarded.A useful hypothesis for autism should not only specify causative factors, but also identify strategies for treatment. The ability of a regimen of folinic acid, betaine and methylcobalamin to normalize plasma levels of sulfur metabolites (James et al., 2004) indicates that methylation support and antioxidant strategies are likely to be useful in treating autism. Further clinical assessment of these and other therapeutic approaches is needed in order to validate their utility. It is reasonable to project that other conditions in which oxidative stress play a role may also benet from these treatments. Acknowledgements The authors wish to acknowledge research support to RD provided by SafeMinds, Autism Research Institute, and Cure Autism Now. References
Agrawal A, Kaushal P, Agrawal S, Gollapudi S, Gupta S. Thimerosal induces TH2 responses via inuencing cytokine secretion by human dendritic cells. J Leukoc Biol; November 1, 2006 [Epub ahead of print]. Ahveninen J, Kahkonen S, Tiitinen H, Pekkonen E, Huttunen J, Kaakkola S, et al. Suppression of transient 40-Hz auditory response by haloperidol suggests modulation of human selective attention by dopamine D2 receptors. Neurosci Lett 2000;292:2932. Akerboom TP, Bilzer M, Sies H. The relationship of biliary glutathione disulde efux and intracellular glutathione disulde content in perfused rat liver. J Biol Chem 1982;257:424852. Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett syndrome is caused by mutations in X-linked MeCP2, encoding methylCpG-binding protein 2. Nat Genet 1999;23:1858. Aoyama K, Suh SW, Hamby AM, Liu J, Chan WY, Chen Y, et al. Neuronal glutathione deciency and age-dependent neurodegeneration in the EAAC1 decient mouse. Nat Neurosci 2006;9:11926. Baker P, Piven J, Schwartz S, Patil S. Brief report: duplication of chromosome 15q1113 in two individuals with autistic disorder. J Autism Dev Disord 1994;24:52935. Bandarian V, Pattridge KA, Lennon BW, Huddler DP, Matthews RG, Ludwig ML. Domain alternation switches B(12)-dependent methionine synthase to the activation conformation. Nat Struct Biol 2002;9:536. Banerjee R, Evande R, Kabil O, Ojha S, Taoka S. Reaction mechanism and regulation of cystathionine beta-synthase. Biochim Biophys Acta 2003;1647:305. Bartos M, Vida I, Jonas P. Synaptic mechanisms of synchronized gamma oscillations in inhibitory interneuron networks. Nat Rev Neurosci 2007;8:4556. Beffert U, Durudas A, Weeber EJ, Stolt PC, Giehl KM, Sweatt JD, et al. Functional dissection of Reelin signaling by site-directed disruption of Disabled-1 adaptor binding to apolipoprotein E receptor 2: distinct roles in development and synaptic plasticity. J Neurosci 2006;26:204152. Benjamin J, Li L, Patterson C, Greenberg BD, Murphy DL, Hamer DH. Population and familial association between the D4 dopamine receptor gene and measures of Novelty Seeking. Nat Genet 1996;12:814.

R. Deth et al. / NeuroToxicology 29 (2008) 190201 Deth RC, Kuznetsova A, Waly M. Attention-related signaling activities of the D4 dopamine receptor. In: Michael Posner, editor. Cognitive neuroscience of attention. New York: Guilford Publications Inc.; 2004. p. 26982. Dixon DP, Skipsey M, Grundy NM, Edwards R. Stress-induced protein Sglutathionylation in Arabidopsis. Plant Physiol 2005;138:223344. Domingo JL. Aluminum and other metals in Alzheimers disease: a review of potential therapy with chelating agents. J Alzheimers Dis 2006;10:33141. Dopp E, Hartmann LM, Florea AM, Rettenmeier AW, Hirner AV. Environmental distribution, analysis, and toxicity of organometal(loid) compounds. Crit Rev Toxicol 2004;34:30133. Ebstein RP, Novick O, Umansky R, Priel B, Osher Y, Blaine D, et al. Dopamine D4 receptor (D4DR) exon III polymorphism associated with the human personality trait of Novelty Seeking. Nat Genet 1996;12:7880. Evans JC, Huddler DP, Hilgers MT, Romanchuk G, Matthews RG, Ludwig ML. Structures of the N-terminal modules imply large domain motions during catalysis by methionine synthase. Proc Natl Acad Sci USA 2004;101:3729 36. Faraone SV, Khan SA. Candidate gene studies of attention-decit/hyperactivity disorder. J Clin Psychiatry 2006;67(Suppl 8):1320. Fatemi SH, Stary JM, Halt AR, Realmuto GR. Dysregulation of Reelin and Bcl2 proteins in autistic cerebellum. J Autism Dev Disord 2001;31:52935. Fatemi SH. Reelin glycoprotein: structure, biology and roles in health and disease. Mol Psychiatry 2005;10:2517. Finkelstein JD. Methionine metabolism in mammals. J Nutr Biochem 1990;1:22837. Fombonne E, Zakarian R, Bennett A, Meng L, McLean-Heywood D. Pervasive developmental disorders in Montreal, Quebec Canada: prevalence and links with immunizations. Pediatrics 2006;118:e13950. Fournier KM, Gonzalez MI, Robinson MB. Rapid trafcking of the neuronal glutamate transporter EAAC1: evidence for distinct trafcking pathways differentially regulated by protein kinase C and platelet-derived growth factor. J Biol Chem 2004;279:3450513. Fratelli M, Goodwin LO, Orom UA, Lombardi S, Tonelli R, Mengozzi M, et al. Gene expression proling reveals a signaling role of glutathione in redox regulation. Proc Natl Acad Sci USA 2005;102:139984003. Friedman G, Goldschmidt N, Friedlander Y, Ben-Yehuda A, Selhub J, Babaey S, et al. A common mutation A1298C in human methylenetetrahydrofolate reductase gene: association with plasma total homocysteine and folate concentrations. J Nutr 1999;129:165661. Froehlich TE, Lanphear BP, Dietrich KN, Cory-Slechta DA, Wang N, Kahn RS. Interactive effects of a DRD4 polymorphism, lead, and sex on executive functions in children. Biol Psychiatry 2007;62:2439. Geier DA, Geier MR. A clinical and laboratory evaluation of methionine cycletranssulfuration and androgen pathway markers in children with autistic disorders. Horm Res 2006;66:1828. Grady DL, Harxhi A, Smith M, Flodman P, Spence MA, Swanson JM, et al. Sequence variants of the DRD4 gene in autism: further evidence that rare DRD4 7R haplotypes are ADHD specic. Am J Med Genet B: Neuropsychiatr Genet 2005;136:335. Grifth OW. Biologic and pharmacologic regulation of mammalian glutathione synthesis. Free Radic Biol Med 1999;27:92235. Hagerman RJ, McBogg P, Hagerman PJ. The fragile X syndrome: history, diagnosis, and treatment. J Dev Behav Pediatr 1983;4:12230. Hamburg DC, Tonoki H, Welty SE, Geske RS, Montgomery CA, Hansen TN. Endotoxin induces glutathione reductase activity in lungs of mice. Pediatr Res 1994;35:3115. Herbert MR, Russo JP, Yang S, Roohi J, Blaxill M, Kahler SG, et al. Autism and environmental genomics. Neurotoxicology 2006;27:67184. Herzing LB, Kim SJ, Cook EH Jr, Ledbetter DH. The human aminophospholipid-transporting ATPase gene ATP10C maps adjacent to UBE3A and exhibits similar imprinted expression. Am J Hum Genet 2001;68:15015. Himi T, Ikeda M, Yasuhara T, Nishida M, Morita I. Role of neuronal glutamate transporter in the cysteine uptake and intracellular glutathione levels in cultured cortical neurons. J Neural Transm 2003;110:133748. Hondorp ER, Matthews RG. Oxidative stress inactivates cobalamin-independent methionine synthase (MetE) in Escherichia coli. PLoS Biol 2004;2:e336.

199

Hung RJ, Boffetta P, Brennan P, Malaveille C, Hautefeuille A, Donato F, et al. GST, NAT, SULT1A1, CYP1B1 genetic polymorphisms, interactions with environmental exposures and bladder cancer risk in a high-risk population. Int J Cancer 2004;110:4598604. Irwin SA, Galvez R, Greenough WT. Dendritic spine structural anomalies in fragile-X mental retardation syndrome. Cereb Cortex 2000;10:1038 44. James SJ, Cutler P, Melnyk S, Jernigan S, Janak L, Gaylor DW, et al. Metabolic biomarkers of increased oxidative stress and impaired methylation capacity in children with autism. Am J Clin Nutr 2004;80:16117. James SJ, Melnyk S, Jernigan S, Cleves MA, Halsted CH, Wong DH, et al. Metabolic endophenotype and related genotypes are associated with oxidative stress in children with autism. Am J Med Genet B: Neuropsychiatr Genet 2006;141:94756. Kern JK, Jones AM. Evidence of toxicity, oxidative stress, and neuronal insult in autism. J Toxicol Environ Health B: Crit Rev 2006;9:48599. Kuznetsova AY, Deth RC. A model for modulation of neuronal synchronization by D4 dopamine receptor-mediated phospholipid methylation. J Comput Neurosci 2007 [Epub ahead of print]. Lachman HM, Papolos DF, Saito T, Yu YM, Szumlanski CL, Weinshilboum RM. Human catechol-O-methyltransferase pharmacogenetics: description of a functional polymorphism and its potential application to neuropsychiatric disorders. Pharmacogenetics 1996;6:24350. LaHoste GJ, Swanson JM, Wigal SB, Glabe C, Wigal T, King N, et al. Dopamine D4 receptor gene polymorphism is associated with attention decit hyperactivity disorder. Mol Psychiatry 1996;1:1214. Lanphear BP, Hornung R, Khoury J, Yolton K, Baghurst P, Bellinger DC, et al. Low-level environmental lead exposure and childrens intellectual function: an international pooled analysis. Environ Health Perspect 2005;113:8949. Lasky T, Sun W, Kadry A, Hoffman MK. Mean total arsenic concentrations in chicken 19892000 and estimated exposures for consumers of chicken. Environ Health Perspect 2004;112:1821. Laumonnier F, Bonnet-Brilhault F, Gomot M, Blanc R, David A, Moizard MP, et al. X-linked mental retardation and autism are associated with a mutation in the NLGN4 gene, a member of the neuroligin family. Am J Hum Genet 2004;74:5527. Laviolette SR, Lipski WJ, Grace AA. A subpopulation of neurons in the medial prefrontal cortex encodes emotional learning with burst and frequency codes through a dopamine D4 receptor-dependent basolateral amygdala input. J Neurosci 2005;25:606675. Liptak MD, Brunold TC. Related spectroscopic and computational studies of Co1 + cobalamin: spectral and electronic properties of the superreduced B12 cofactor. J Am Chem Soc 2006;128:914456. Lucarelli P, Saccucci P, Bottini N, De Luca D, Fiumara A, Elia M, et al. Twoloci ADA haplotypes in autistic disorder. Am J Med Genet 2002;108:339 40. Ludwig ML, Matthews RG. Structure-based perspectives on B12-dependent enzymes. Annu Rev Biochem 1997;66:269313. Malhotra AK, Kestler LJ, Mazzanti C, Bates JA, Goldberg T, Goldman D. A functional polymorphism in the COMT gene and performance on a test of prefrontal cognition. Am J Psychiatry 2002;159:6524. McConkie-Rosell A, Lachiewicz AM, Spiridigliozzi GA, Tarleton J, Schoenwald S, Phelan MC, et al. Evidence that methylation of the FMR-I locus is responsible for variable phenotypic expression of the fragile X syndrome. Am J Hum Genet 1993;53:8009. McGinnis WR. Oxidative stress in autism. Altern Ther Health Med 2004;10:22 36. Mellick GD. CYP450, genetics and Parkinsons disease: gene environment interactions hold the key. J Neural Transm Suppl 2006;70:15965. Mill J, Caspi A, Williams BS, Craig I, Taylor A, Polo-Tomas M, et al. Prediction of heterogeneity in intelligence and adult prognosis by genetic polymorphisms in the dopamine system among children with attentiondecit/hyperactivity disorder: evidence from 2 birth cohorts. Arch Gen Psychiatry 2006;63:4629. Miller JW, Ramos MI, Garrod MG, Flynn MA, Green R. Transcobalamin II 775G > C polymorphism and indices of vitamin B12 status in healthy older adults. Blood 2002;100:71820.

200

R. Deth et al. / NeuroToxicology 29 (2008) 190201 Sharma A, Kramer ML, Wick PF, Liu D, Chari S, Shim S, et al. D4 dopamine receptor-mediated phospholipid methylation and its implications for mental illnesses such as schizophrenia. Mol Psychiatry 1999;4:23546. Shashidharan P, Huntley GW, Murray JM, Buku A, Moran T, Walsh MJ, et al. Immunohistochemical localization of the neuron-specic glutamate transporter EAAC1 (EAAT3) in rat brain and spinal cord revealed by a novel monoclonal antibody. Brain Res 1997;773:13948. Shenker BJ, Mayro JS, Rooney C, Vitale L, Shapiro IM. Immunotoxic effects of mercuric compounds on human lymphocytes and monocytes IV. Alterations in cellular glutathione content. Immunopharmacol Immunotoxicol 1993;15:27390. Smalley SL, Asarnow RF, Spence MA, Autism genetics. A decade of research. Arch Gen Psychiatry 1988;45:95361. Stone RL, Aimi J, Barshop BA, Jaeken J, Van den Berghe G, Zalkin H, et al. A mutation in adenylosuccinate lyase associated with mental retardation and autistic features. Nat Genet 1992;1:5963. Stipanuk MH, Hirschberger LL, Londono MP, Cresenzi CL, Yu AF. The ubiquitin-proteasome system is responsible for cysteine-responsive regulation of cysteine dioxygenase concentration in liver. Am J Physiol Endocrinol Metab 2004;286:E43948. Stubbs G, Litt M, Lis E, Jackson R, Voth W, Lindberg A, et al. Adenosine deaminase activity decreased in autism. J Am Acad Child Psychiatry 1982;21:714. Suzuki H, Sugiyama Y. Excretion of GSSG and glutathione conjugates mediated by MRP1 and cMOAT/MRP2. Semin Liver Dis 1998;18:35976. Tallan HH, Moore S, Stein WH. L-Cystathionine in human brain. J Biol Chem 1958;230:70716. Thatcher KN, Peddada S, Yasui DH, Lasalle JM. Homologous pairing of 15q1113 imprinted domains in brain is developmentally regulated but decient in Rett and autism samples. Hum Mol Genet 2005;14:78597. Thomas DJ, Li J, Waters SB, Xing W, Adair BM, Drobna Z, et al. Arsenic (+3 oxidation state) methyltransferase and the methylation of arsenicals. Exp Biol Med (Maywood) 2007;232:313. Trasande L, Landrigan PJ, Schechter C. Public health and economic consequences of methyl mercury toxicity to the developing brain. Environ Health Perspect 2005;113:5906. Ueland PM. Pharmacological and biochemical aspects of S-adenosylhomocysteine and S-adenosylhomocysteine hydrolase. Pharmacol Rev 1982;34:223 53. Valko M, Morris H, Cronin MT. Metals, toxicity and oxidative stress. Curr Med Chem 2005;12:1161208. Vargas DL, Nascimbene C, Krishnan C, Zimmerman AW, Pardo CA. Neuroglial activation and neuroinammation in the brain of patients with autism. Ann Neurol 2005;57:6781. Vitvitsky V, Thomas M, Ghorpade A, Gendelman HE, Banerjee R. A functional transsulfuration pathway in the brain links to glutathione homeostasis. J Biol Chem 2006;281:3578593. Wang E, Ding YC, Flodman P, Kidd JR, Kidd KK, Grady DL, et al. The genetic architecture of selection at the human dopamine receptor D4 (DRD4) gene locus. Am J Hum Genet 2004;74:93144. Waly M, Olteanu H, Banerjee R, Choi SW, Mason JB, Parker BS, et al. Activation of methionine synthase by insulin-like growth factor-1 and dopamine: a target for neurodevelopmental toxins and thimerosal. Mol Psychiatry 2004;9:35870. Watson WP, Munter T, Golding BT. A new role for glutathione: protection of vitamin B12 from depletion by xenobiotics. Chem Res Toxicol 2004;17:15627. Wilson TW, Rojas DC, Reite ML, Teale PD, Rogers SJ. Children and adolescents with autism exhibit reduced MEG steady-state gamma responses. Biol Psychiatry; September 1, 2006 [Epub ahead of print]. Winyard PG, Moody CJ, Jacob C. Oxidative activation of antioxidant defence. Trends Biochem Sci 2005;30:45361. Wright RO, Amarasiriwardena C, Woolf AD, Jim R, Bellinger DC. Neuropsychological correlates of hair arsenic, manganese, and cadmium levels in school-age children residing near a hazardous waste site. Neurotoxicology 2006;27:2106. Yao Y, Walsh WJ, McGinnis WR, Pratico D. Altered vascular phenotype in autism: correlation with oxidative stress. Arch Neurol 2006;63:11614.

Ming X, Stein TP, Brimacombe M, Johnson WG, Lambert GH, Wagner GC. Increased excretion of a lipid peroxidation biomarker in autism. Prostaglandins Leukot Essent Fatty Acids 2005;73:37984. Molloy AM, Daly S, Mills JL, Kirke PN, Whitehead AS, Ramsbottom D, et al. Thermolabile variant of 5,10-methylenetetrahydrofolate reductase associated with low red-cell folates: implications for folate intake recommendations. Lancet 1997;349:15913. Mrzljak L, Bergson C, Pappy M, Huff R, Levenson R, Goldman-Rakic PS. Localization of dopamine D4 receptors in GABAergic neurons of the primate brain. Nature 1996;381:2458. Najib S, Sanchez-Margalet V. Homocysteine thiolactone inhibits insulin signaling, and glutathione has a protective effect. J Mol Endocrinol 2001;27:8591. Oak JN, Lavine N, Van Tol HH. Dopamine D(4) and D(2L) receptor stimulation of the mitogen-activated protein kinase pathway is dependent on transactivation of the platelet-derived growth factor receptor. Mol Pharmacol 2001;60:92103. Obin M, Shang F, Gong X, Handelman G, Blumberg J, Taylor A. Redox regulation of ubiquitin-conjugating enzymes: mechanistic insights using the thiol-specic oxidant diamide. FASEB J 1998;12:5619. Oldenhof J, Vickery R, Ana M, Oak J, Ray A, Schoots O, et al. SH3 binding domains in the dopamine D4 receptor. Biochemistry 1998;37:1572636. Pasca SP, Nemes B, Vlase L, Gagyi CE, Dronca E, Miu AC, et al. High levels of homocysteine and low serum paraoxonase 1 arylesterase activity in children with autism. Life Sci 2006;78:22448. Patterson S, Flatt PR, McClenaghan NH. Major metabolic homocysteinederivative, homocysteine thiolactone, exerts changes in pancreatic betacell glucose-sensing, cellular signal transduction and integrity. Arch Biochem Biophys 2007;461:28793. Persa C, Pierce A, Ma Z, Kabil O, Lou MF. The presence of a transsulfuration pathway in the lens: a new oxidative stress defense system. Exp Eye Res 2004;79:87586. Persico AM, Militerni R, Bravaccio C, Schneider C, Melmed R, Trillo S, et al. Adenosine deaminase alleles and autistic disorder: case-control and familybased association studies. Am J Med Genet 2000;96:78490. Persico AM, DAgruma L, Maiorano N, Totaro A, Militerni R, Bravaccio C, et al. Collaborative Linkage Study of Autism. Reelin gene alleles and haplotypes as a factor predisposing to autistic disorder. Mol Psychiatry 2001;6:1509. Persico AM, Bourgeron T. Searching for ways out of the autism maze: genetic, epigenetic and environmental clues. Trends Neurosci 2006;29:34958. Pezacka E, Green R, Jacobsen DW. Glutathionylcobalamin as an intermediate in the formation of cobalamin coenzymes. Biochem Biophys Res Commun 1990;169:44350. Prudova A, Albin M, Bauman Z, Lin A, Vitvitsky V, Banerjee R. Testosterone regulation of homocysteine metabolism modulates redox status in human prostate cancer cells. Antioxid Redox Signal 2007;9:187582. Raymond J, Segre D. The effect of oxygen on biochemical networks and the evolution of complex life. Science 2006;311:17647. Rice C, et al. Prevalence of autism spectrum disordersautism and developmental disabilities monitoring network. CDC Surv Rep 2007;56(SS01):12 28. Rubino FM, Pitton M, Brambilla G, Colombi A. A study of the glutathione metaboloma peptides by energy-resolved mass spectrometry as a tool to investigate into the interference of toxic heavy metals with their metabolic processes. J Mass Spectrom 2006;41:157893. Sakurai T, Ochiai M, Kojima C, Ohta T, Sakurai MH, Takada NO, et al. Preventive mechanism of cellular glutathione in monomethylarsonic acidinduced cytolethality. Toxicol Appl Pharmacol 2005;206:5465. Samaco RC, Hogart A, LaSalle JM. Epigenetic overlap in autismspectrum neurodevelopmental disorders: MeCP2 deciency causes reduced expression of UBE3A and GABRB3. Hum Mol Genet 2005;14:48392. Sanfeliu C, Sebastia J, Cristofol R, Rodriguez-Farre E. Neurotoxicity of organomercurial compounds. Neurotox Res 2003;5:283305. Schafer FQ, Buettner GR. Redox environment of the cell as viewed through the redox state of the glutathione disulde/glutathione couple. Free Radic Biol Med 2001;30:1191212.

R. Deth et al. / NeuroToxicology 29 (2008) 190201 Zhao R, Chen Y, Tan W, Waly M, Sharma A, Stover P, et al. Relationship between dopamine-stimulated phospholipid methylation and the singlecarbon folate pathway. J Neurochem 2001;78:78896. Zhen X, Zhang J, Johnson GP, Friedman E. D(4) dopamine receptor differentially regulates Akt/nuclear factor-kappa b and extracellular signal-regulated kinase pathways in D(4)MN9D cells. Mol Pharmacol 2001;60:85764.

201

Zintzaras E, Hadjigeorgiou GM. Association of paraoxonase 1 gene polymorphisms with risk of Parkinsons disease: a meta-analysis. J Hum Genet 2004;49:47481. Zoroglu SS, Armutcu F, Ozen S, Gurel A, Sivasli E, Yetkin O, et al. Increased oxidative stress and altered activities of erythrocyte free radical scavenging enzymes in autism. Eur Arch Psychiatry Clin Neurosci 2004;254:1437.

Vous aimerez peut-être aussi