Protein Adduct Formation as a Molecular Mechanism in Neurotoxicity

Richard M. LoPachin*,1 and Anthony P. DeCaprio{dagger}

* Department of Anesthesiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York 10467-2490, and {dagger} Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts at Amherst, Amherst, Massachusetts 01003

1 To whom correspondence should be addressed at Montefiore Medical Center, Moses Research Tower – 7, 111 E. 2210th St., Bronx, NY 10467-2490. Fax: (718) 515-4903. E-mail: lopachin{at}aecom.yu.edu.

Received March 24, 2005; accepted April 28, 2005


    ABSTRACT
 TOP
 ABSTRACT
 INTRODUCTION
 REFERENCES
 
Chemicals that cause nerve injury and neurological deficits are a structurally diverse group. For the majority, the corresponding molecular mechanisms of neurotoxicity are poorly understood. Many toxicants (e.g., hepatotoxicants) of other organ systems and/or their oxidative metabolites have been identified as electrophiles and will react with cellular proteins by covalently binding nucleophilic amino acid residues. Cellular toxicity occurs when adduct formation disrupts protein structure and/or function, which secondarily causes damage to submembrane organelles, metabolic pathways, or cytological processes. Since many neurotoxicants are also electrophiles, the corresponding pathophysiological mechanism might involve protein adduction. In this review, we will summarize the principles of covalent bond formation that govern reactions between xenobiotic electrophiles and biological nucleophiles. Because a neurotoxicant can form adducts with multiple nucleophilic residues on proteins, the challenge is to identify the mechanistically important adduct. In this regard, it is now recognized that despite widespread chemical adduction of tissue proteins, neurotoxicity can be mediated through binding of specific target nucleophiles in key neuronal proteins. Acrylamide and 2,5-hexanedione are prototypical neurotoxicants that presumably act through the formation of protein adducts. To illustrate both the promise and the difficulty of adduct research, these electrophilic chemicals will be discussed with respect to covalent bond formation, suspected protein sites of adduction, and proposed mechanisms of neurotoxicity. The goals of future investigations are to identify and quantify specific protein adducts that play a causal role in the generation of neurotoxicity induced by electrophilic neurotoxicants. This is a challenging but critical objective that will be facilitated by recent advances in proteomic methodologies.

Key Words: toxic neuropathy; axonopathy; protein adduct; electrophilic chemicals; acrylamide; 2,5-hexanedione.


    INTRODUCTION
 TOP
 ABSTRACT
 INTRODUCTION
 REFERENCES
 
Exposure to numerous structurally diverse chemicals can cause significant morphological and functional damage to nerve cells in the CNS and PNS. In pursuing the corresponding mechanisms of this neurotoxicity, the majority of previous studies have focused on the impact of these chemicals on neuronal structure (e.g., axons), organelles (e.g., mitochondria), or metabolic processes (e.g., fast anterograde transport, ion regulation). Despite significant research, the pathophysiological and molecular mechanisms for many of these neurotoxicants remains poorly defined. However, a common mechanistic theme in the toxicological sciences has been formation of chemical adducts with macromolecules. It is now recognized that many organ system toxicants (e.g., acetaminophen, chloramphenicol, bromobenzene) and/or their active metabolites are electrophiles (electron deficient) that form irreversible covalent bonds (adducts) with nucleophilic centers (unshared electrons) on proteins, DNA, and RNA. Presumably cell toxicity occurs when adduct formation disrupts the structure and/or function of macromolecules (Coles, 1985Go; Hinson and Roberts, 1992Go; Nelson and Pearson, 1990Go). Since many neurotoxicants and their metabolites are electrophiles (Table 1), it is possible that adduct formation is also a critical step in the neuropathogenic processes initiated by exposure to these chemicals. Indeed, protein adduction has been considered as a possible mechanism of neurotoxic action for the organophosphate insecticides, carbon disulfide, and the hexacarbon solvents (reviewed in Graham et al., 1995Go; LoPachin and DeCaprio, 2004Go; Lotti, 2000Go). Nonetheless, despite some promising research activity, adduct formation has not been adequately explored as a more global consequence of neurotoxicant exposure. Therefore, the purpose of this review is to generate further interest in protein adduction as the initiating pathophysiological process in many types of chemical-induced nerve cell injury. Since electrophile/nucleophile theory has received limited attention in the neurotoxicology literature, we will start with a discussion of the physiochemical principles that govern covalent interactions between biological macromolecules and reactive neurotoxicants. This presentation will be limited to protein adduction since research currently suggests that proteins, and not nucleic acids, are the most likely sites of adduction involved in the pathogenesis of toxic nerve cell injury. We will then discuss how adduction might cause protein dysfunction and how such an effect could mediate toxicity at the cellular level. To provide neurotoxicological context, the general principles and concepts of adduct formation will be related to 2,5-hexanedione (HD) neuropathy, for which the results from two decades of research support a mechanism involving adduction of neuronal proteins. In addition, we will present new evidence that acrylamide (ACR) neurotoxicity is mediated by adduct formation with nerve terminal proteins.


View this table:
[in this window]
[in a new window]
 
TABLE 1 Examples of Electrophilic Neurotoxicants (Metabolites)

 
Physiochemical Principles of Protein Adduct Formation
Numerous neurotoxic chemicals and/or their active metabolites are electrophiles that will react with nucleophilic centers that have high electron density. The reaction of an electrophilic species with a nucleophilic molecule generally occurs via a substitution or addition mechanism and involves donation of an electron pair by the nucleophile and subsequent formation of a covalent bond. Many atomic and molecular determinants of electrophilicity and nucleophilicity have been postulated and investigated (see reviews by Chermette, 1999Go; Parthasarathi et al., 2004Go). Currently, the most accepted concept classifies electrophiles and nucleophiles according to a "hard acid/soft base" (HSAB) model (Loechler, 1994Go; Pearson and Songstad, 1967Go; Swain and Scott, 1953Go). Thus, hard electrophiles have either a high positive charge density or a formal positive charge at the electrophilic center. Conversely, soft electrophiles have a lower positive charge density. Because of their high charge density, hard electrophiles have valence electron shells that are not easily polarized (i.e., deformed), whereas the opposite is true for soft electrophiles. It is the relative tendency of these outer shells to polarize that is a major determinant of the selective reactivity of electrophiles with their nucleophilic targets (see ahead). Hard nucleophiles have high electronegativity and low polarization of valence electrons, whereas soft nucleophiles have low electronegativity and are more polarizable. The softest biological nucleophilic sites are cysteine thiol groups on proteins and glutathione (GSH; Table 2). Of moderate hardness are primary and secondary amino groups (lysine and histidine, respectively) on proteins, whereas the hardest nucleophiles are the oxygen atoms of purines and pyrimidines (Table 2).


View this table:
[in this window]
[in a new window]
 
TABLE 2 List of Hard and Soft Electrophiles and Hard and Soft Nucleophiles

 
Based on the HSAB theory, the reaction rates and selectivities of electrophiles and nucleophiles are dependent upon comparable states of "hardness." Specifically, a soft electrophile such as acrylamide will react predominantly with a soft nucleophile such as the thiol group of cysteine. A hard electrophile such as the methyl carbonium ion formed from dimethyl nitrosamine will react with hard nucleophiles such as the oxygen atoms of DNA. This preferential reactivity is due primarily to the high-energy transition state that acts as a barrier to the reaction of, for example, a hard electrophile with a soft nucleophile (Coles, 1985Go; Pearson and Songstad, 1967Go). Electrophilic xenobiotics adduct biological nucleophilic sites through a number of covalent reaction mechanisms. For example, the polarized double bonds of acrylamide, acrolein, and other {alpha},ß-unsaturated chemicals react by Michael addition of the nucleophile across the vinyl group (Fig. 3). Nucleophilic attack by electrophilic epoxide chemicals (e.g., glycidamide, the oxidized metabolite of acrylamide) occurs through opening of the strained ring (for a more detailed discussion of covalent binding during adduct formation see Coles, 1985Go; Friedman, 1973Go; Harding, 1985Go; Kemp and Vellaccio, 1980Go).



View larger version (20K):
[in this window]
[in a new window]
 
FIG. 3. This figure illustrates the formation of protein adducts by HD and ACR. As a {gamma}-diketone electrophile, HD reacts covalently with nucleophilic lysine {varepsilon}-amine groups to form 2,5-dimethylpyrrole adducts on proteins (A). ACR is an {alpha},ß-unsaturated aldehyde that forms adducts with cysteine sulfhydryl groups via a Michael carbonyl condensation reaction (B).

 
Relative hardness and softness are clearly important characteristics for covalent bond formation in biological systems. However, protein adduction is dependent not only upon the physiochemical nature of the electrophile but also upon the microenvironment of the nucleophilic center, which can vary significantly even among centers of the same elemental type (e.g., sulfur or amino groups). Thus, nucleophilic reactivity among free sulfhydryl groups on polythiol proteins can be diverse and, consequently, soft electrophilic chemicals will adduct the more reactive thiol groups on a given protein (Vogel and Nivard, 1994Go). This diversity in nucleophilic reactivity is a function of both steric and electronic factors mediated primarily by protein tertiary structure. For example, it is now recognized that vicinal acidic and basic amino acids significantly influence reactivity of the target nucleophilic group. In hemoglobin, Cysß93 is involved in nitric oxide (NO) signaling and is arranged in a three-dimensional motif with the basic amino acid histidine (Hisß146) and the acidic residue aspartate (Aspß94; Stamler et al., 1997Go). In the oxygenated state (relaxed conformation), the sulfur of Cysß93 is adjacent to the imidazolium group (pKa = 6.0) of histidine, which facilitates deprotonation of the sulfur residue. When compared to other cysteine residues that do not exist in a comparable motif, deprotonation significantly increases the relative nucleophilicity of Cysß93 (by lowering the pKa) and promotes nitrosylation of the sulfhydryl group. In the deoxygenated state (tense conformation), the Cysß93 of hemoglobin is in proximity to the carboxyl group of aspartate. This results in protonation of the sulfur and a reduction in both the nucleophilicity and the rate of nitrosylation of this residue. This example demonstrates how reactivity among common nucleophilic sites can vary within a protein and provides a basis for understanding the selectivity and toxicological specificity of protein adduct formation. The example also shows how acid-base catalysis can alternatively regulate the nitrosylation and denitrosylation of a protein nucleophilic center. In turn, this illustrates how the redox-state of a single but functionally critical nucleophile can modulate an important physiological process such as NO signaling (see Hess et al., 2001Go; Jaffrey et al., 2001Go; Stamler et al., 1997Go).

The Influence of Xenobiotic Biotransformation on Sites of Adduct Formation
The biotransformation of a chemical to an active electrophilic derivative appears to be a common mechanism in the toxicological sciences (Fig. 1; Ketterer, 1980Go; Miller and Miller, 1981Go). As discussed above, depending upon the physiochemical nature of the electrophile, the resulting metabolite can produce toxicity by reacting with: (1) soft nucleophilic sites on proteins and free thiols such as GSH or (2) harder nucleophilic centers on DNA and RNA. Alternatively, both the parent and metabolite can produce adduct-based toxicity (Fig. 1). Frequently, the parent chemical is a soft electrophile that is metabolically converted to a harder toxic metabolite. For example, acrylonitrile (ACN; cyanoethylene) has been shown to have both carcinogenic and neurotoxic actions (Willhite, 1982Go; Woutersen, 1998Go). The vinyl group of ACN is a soft electrophilic center that reacts with free sulfhydryl groups on GSH and protein cysteines (Fig. 1). However, metabolic epoxidation of the double bond produces the relatively hard electrophilic metabolite, cyanoethylene oxide (Fig. 1; Sumner et al., 1999Go). This epoxide will form adducts primarily with nucleophilic sites on DNA (Guengerich et al., 1981Go; Oesch et al., 1971Go). Similarly, acrylamide (ACR) is an {alpha},ß-unsaturated carbonyl and is a well-recognized human neurotoxicant and possible carcinogen (Fig. 1; reviewed in LoPachin et al., 2003aGo; LoPachin, 2004Go; Dearfield et al., 1995Go). As a soft electrophile, ACR will adduct thiol groups on proteins and GSH (LoPachin et al., 2003bGo, 2004aGo; Tong et al., 2004Go). Cytochrome P450 2E1-mediated metabolism of ACR yields the epoxide derivative, glycidamide (Fig. 1; Calleman et al., 1990Go; Sumner et al., 1992Go). This harder electrophilic metabolite will react with nucleophilic centers on adenine and guanine of DNA (Gamboa da Costa et al., 2003Go). These examples show that both the parent chemical and oxidative metabolite can exhibit distinct electrophilic characteristics. This difference in electrophilicity predicts correspondingly different nucleophilic targets for the parent (i.e., protein thiols) and harder metabolite (i.e., nucleic acids). That these divergent macromolecular targets mediate different adduct-based toxicities is suggested by the observation that most genotoxic chemicals are hard electrophiles that adduct hard nucleophilic sites on DNA. In contrast, chemicals that produce noncarcinogenic cytotoxicity (e.g., neurotoxicity, hepatotoxicity) are soft electrophiles such as the {alpha},ß-unsaturated aldehydes, ketones or related derivatives that bind cysteine sulfhydryl groups on proteins (reviewed in Hinson and Roberts, 1992Go; Coles, 1985Go).



View larger version (20K):
[in this window]
[in a new window]
 
FIG. 1. This figure shows examples of an unreactive parent chemical (n-hexane) and its neurotoxic reactive metabolite (2,5-hexanedione). Also illustrated are several examples of neurotoxicants for which both the parent molecule and metabolite exhibit electrophilic reactivity.

 
The Toxicity Spectrum of Soft Electrophiles
In general, the different preferential toxicities of hard electrophiles (i.e., genotoxicity) and their soft counterparts (i.e., cytotoxicity) are understandable based upon their presumed macromolecular targets; i.e., DNA vs. proteins, respectively. However, soft electrophiles can cause a wide spectrum of cytotoxicity, which is counterintuitive since these chemicals theoretically adduct common nucleophilic sites on proteins. For example, {alpha},ß-unsaturated carbonyl compounds are a chemically broad group of soft electrophiles that form cysteine adducts on proteins via Michael additions (Esterbauer et al., 1991Go; see also Fig. 3). This chemical class includes compounds that produce a variety of organ-specific toxicities; e.g., acrolein and acrylamide (neurotoxicity), allylamine (cardiotoxicity), 2-methylfuran (renal toxicity), and the reactive metabolite of acetaminophen, N-acetyl-p-benzoquinone imine (hepatotoxicity). Because these electrophilic toxicants interact with similar nucleophilic residues that have a wide distribution among diverse tissue proteins, a common, more generalized toxicity might be anticipated. Yet, the observed selective toxicities suggest that individual chemicals of this group differ with respect to their tendency to form protein adducts in specific target organs (Cohen et al., 1997Go; Tornqvist et al., 2002Go). The mechanism underlying this apparent selectivity is complex and not completely understood. However, the selective toxicities exhibited by this class of chemicals could be, at least in part, due to characteristic toxicokinetic parameters that determine the tissue distribution and target concentrations of a given electrophile. Also likely involved are the respective physiochemical attributes of both the electrophile (hard vs. soft) and its nucleophilic site (steric and electronic factors) that ultimately determine the probability of adduct formation in different tissues and individual proteins. Thus, although soft electrophilic toxicants react with nucleophilic sites that are common to many proteins across tissues, selective toxicity is possible based on inherent toxicokinetic and physiochemical characteristics that can limit the reaction of an electrophile to a specific set of nucleophiles in a target tissue. Clearly, the selectivity of macromolecular adduct formation is an area of adduct toxicity that requires continued research.

Putative Mechanisms of Adduct-Based Neurotoxicity
In this section, we will discuss mechanisms by which protein adduction might induce neurotoxicity at the molecular and cellular levels. The principles of adduct-based mechanisms are presented schematically in Figure 2. Although substantial evidence supports a role for adduct formation in toxic cell injury, for many chemicals a direct causal relationship has not been established. This is not an indictment of adduct-based mechanisms, but rather reflects the in vivo complexity of chemical-protein adduction and the limited analytical methodologies previously available. Recent advances in proteomic techniques should accelerate progress in the detection and quantification of low in vivo adduct levels (reviewed in Harder et al., 2003Go; Liebler, 2002Go; LoPachin et al., 2003cGo; Tornqvist et al., 2002Go). It should also be noted that mechanisms indirectly involving adduct formation, most notably oxidative stress, can also play a role in the production of toxicity. Some electrophilic chemicals lead either directly or indirectly to generation of reactive oxygen species (e.g., superoxide anion, hydroxyl radicals) that subsequently cause oxidative damage. In addition, soft electrophiles that adduct thiol groups can significantly decrease cellular reducing equivalents (e.g., NADPH, glutathione, vitamin E) and thereby shift the redox balance of the cell toward oxidation. Regardless of the oxidative mechanism, lipid peroxidation, protein thiol oxidation and other oxidative changes can, in addition to direct covalent protein modifications, lead to cytotoxicity.



View larger version (18K):
[in this window]
[in a new window]
 
FIG. 2. This figure shows, in schematic form, the salient features of adduct-based neurotoxicity.

 
We have proposed several possible reasons why, despite adduction of common nucleophilic sites on proteins, soft electrophilic chemicals could produce tissue-specific. Even within the target tissue (e.g., PNS or liver), however, these nucleophilic centers are ubiquitous and diverse cellular pathways and processes exist that might be affected by adduction of constituent proteins. Given this lack of site specificity within the target tissue it is not clear how adduct formation could produce selective toxicity. Accumulating evidence suggests that electrophilic chemicals act by adducting specific amino acids that are critical to tertiary structure and/or protein functions such as enzymatic activity, ion translocation, or protein-protein interactions (reviewed in Friedman, 1973Go; Harding, 1985Go; Hinson and Roberts, 1992Go; Nelson and Pearson, 1990Go). For example, the role of the skeletal muscle Ca2+ release channel/ryanodine receptor (RyR1) in excitation-contraction coupling is highly sensitive to disruption by the sulfhydryl alkylating agent, N-ethylmaleimide (NEM; Moore et al., 1999Go). There are approximately 400 cysteines within the tetrameric RYR1 channel complex that represent potential sites of alkylation (Eu et al., 2000Go). Nonetheless, several lines of evidence indicate that NEM inhibition is mediated by selective adduction of the sulfhydryl group on Cys 3635 (Eu et al., 2000Go; Sun et al., 2001Go; Yamaguchi et al., 2001Go). Thus, although other cysteine residues are adducted, NEM alkylation of a specific cysteine can lead to impaired RyR1 channel function and thereby promote muscle toxicity. It can also be deduced from this example that a large proportion of the adducts formed on this and other proteins are toxicologically irrelevant since the corresponding amino acids do not play a direct role in either protein structure or function.

Even when adduction of a critical amino acid residue and subsequent changes in protein structure and/or function can be demonstrated, it cannot be assumed a priori that this effect necessarily produces cellular injury. This caveat is particularly germane to the chemical adduction of proteins that are members of metabolic pathways (e.g., glycolysis) or are constituents of complexes (e.g., SNARE core complexes) that mediate neuronal processes (e.g., neurotransmitter release, energy production). Here, the function of the pathway or process is more likely to be affected if the adduct-inhibited protein plays a key regulatory role or is otherwise operationally critical. This concept is best illustrated by the research surrounding the "glycolysis" hypothesis of toxic axonopathies. It was proposed that acrylamide inhibited the activities of glycolytic enzymes resulting in an axonal energy deficit and subsequent degeneration of distal fibers (Spencer et al., 1979Go). Supporting evidence suggested that acrylamide intoxication of rats decreased the activities of neuron specific enolase (NSE) and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) in brain homogenates (Howland, 1981Go; Howland et al., 1980Go). Additional results implied that this effect was due to adduction of cysteine residues in the active sites of these enzymes (Orstan and Gafni, 1990Go). However, other research showed that acrylamide did not affect lactate production or other indices of brain glycolytic pathway function (Brimijoin and Hammond, 1985Go; LoPachin et al., 1984Go; Matsuoka and Igisu, 1992Go). The absence of an effect on glycolytic flux is consistent with additional data showing that acrylamide did not inhibit the activity of phosphofructokinase (PFK), the rate-limiting glycolytic enzyme (Howland, 1981Go; Sakamoto and Hashimoto, 1985Go). Thus, despite adduction of functionally important amino acid residues (cysteines) and subsequent glycolytic enzyme inhibition (e.g., GAPDH and NSE activities), acrylamide did not decrease overall glucose flux. Protein adduct formation is, therefore, not necessarily a prelude to toxicity. Instead, toxicity occurs only when the activities of key regulatory proteins (e.g., PFK) are inhibited by adduct formation.

In the preceding sections, we have defined the physiochemical attributes of electrophilic chemicals and their protein nucleophilic sites of adduction. In addition, we have discussed the molecular mechanisms that might mediate the production of selective cellular toxicity by electrophilic xenobiotics. To place these concepts within a neurotoxicological perspective, in the following section we will provide a brief overview of the substantial evidence supporting a role for protein adduct formation in {gamma}-diketone neuropathy. Also presented will be results from recent studies, which suggest that ACR produces neurotoxicity through adduction of cysteine residues on certain nerve terminal proteins that regulate membrane fusion processes such as neurotransmission.

The Role of Adduct Formation in 2,5-Hexanedione Neurotoxicity
HD is the active {gamma}-diketone metabolite of the neurotoxic hexacarbon solvents n-hexane and methyl n-butyl ketone (Couri and Milks, 1982Go; Krasavage et al., 1980Go). HD intoxication of laboratory animals produces loss of body weight and changes in several neurological parameters, including gait abnormalities (ataxia) and reductions in hindlimb skeletal muscle strength (LoPachin et al., 2002Go; Spencer and Schaumburg, 1977aGo). Quantitative morphometric and electrophysiologic studies conducted over the past 20 years have shown that axon atrophy in the PNS and CNS is the morphological hallmark of {gamma}-diketone neuropathy (reviewed in LoPachin and Lehning, 1997Go; LoPachin et al., 2000Go; LoPachin and DeCaprio, 2004Go). Previous investigations have revealed that, regardless of exposure rate (100–400 mg/kg/day), axon atrophy was an early consequence of HD neurotoxicity. HD-induced atrophy developed in conjunction with the onsets of neurological deficits and decreases in nerve conduction velocity (Lehning et al., 2000Go; Yagi, 1994Go). Since it is well documented that loss of caliber produces changes in axonal cable properties and nerve conduction (Sakaguchi et al., 1993Go), the atrophy induced by {gamma}-diketone intoxication could be causally related to the observed nerve dysfunction. Together, these attributes suggest that the development of axon atrophy is a necessary event in the pathophysiological process that leads to {gamma}-diketone neurological toxicity.

The mechanism by which HD produces axon atrophy is not known, but likely involves either direct or indirect disruption of neurophysiological processes that maintain axon caliber. Based on this premise, the neurofilament (NF) subunits, NF-light (NF-L), NF-medium (NF-M) and NF-heavy (NF-H) are primary target candidates (DeCaprio and O'Neill, 1985Go), since these proteins play an important role in determining the size of mature axons (reviewed in Muma and Hoffman, 1993Go). Indeed, we have found that the axon atrophy induced by HD is correlated with a loss of these cytoskeletal proteins in PNS and CNS tissues (Chiu et al., 2000Go; LoPachin et al., 2004bGo, 2005Go). Whereas the reason for this protein loss is not understood, we have hypothesized that HD adduction of NF proteins interferes with the turnover and maintenance of the axonal cytoskeleton (see below). HD is a diketone electrophile that reacts covalently with nucleophilic lysine {varepsilon}-amine groups (Fig. 3) to form 2,5-dimethylpyrrole adducts on NFs and other proteins (DeCaprio et al., 1982Go, 1983Go; DeCaprio and O'Neil, 1985Go; Graham et al., 1982Go). Pyrrole formation is unique to {gamma}-diketones, which have a two carbon spacing between the carbonyl functions and is the basis of the structure-activity relationships that have been established for {gamma}-diketone neuropathy (see Anthony et al., 1983Go; DeCaprio et al., 1988Go; Genter et al., 1988Go; Monaco et al., 1985Go). Graham and colleagues (1991Go, 1995Go) suggested that once formed, pyrrole adducts undergo oxidative reactions that yield crosslinked NF proteins. However, the pathophysiological relevance of this secondary step has not been established (reviewed in DeCaprio, 2000Go; LoPachin and DeCaprio, 2004Go; LoPachin and Lehning, 1997Go).

Although evidence has shown that the formation of pyrrole adducts on lysine residues is a critical and required event in the production of HD neuropathy, the low levels of NF adducts formed during {gamma}-diketone exposure and the lack of protein specificity appear to argue against a causative role. Specifically, during in vivo {gamma}-diketone exposure non-neuronal proteins (e.g., hemoglobin, serum albumin) that are unlikely to be involved in neurotoxic mechanisms exhibit pyrrole adduct levels that are quantitatively similar to those of cytoskeletal proteins (DeCaprio and O'Neill, 1985Go; Genter St. Clair et al., 1988Go; Pyle et al., 1992Go). This observation suggests a wide range of targets and is consistent with the fact that virtually all proteins contain one or more lysine {varepsilon}-amine sidechains that are potential sites of adduction for HD. Moreover, in vivo and in vitro studies showed that only a small fraction (e.g., <5%) of total available lysyl {varepsilon}-amine groups on NF proteins were converted to pyrrole adducts by HD exposure (DeCaprio and O'Neill, 1985Go; DeCaprio and Fowke, 1992Go). If NFs are the primary neuropathogenic target, then one might expect that substantial adduct formation in NF proteins should occur. However, in support of a specific involvement of pyrrole formation, in vitro experiments demonstrated that the adducted lysine residues were primarily located within KSP repeats on the C-terminal ("tail") regions of NF-M and NF-H subunit proteins (DeCaprio and Fowke, 1992Go; DeCaprio et al., 1997Go). These regions are critically involved in the interactions of NF subunits with the cytoskeletal polymer (see below). Based on this selectivity, it has been proposed that, despite a broad range of adducted proteins, {gamma}-diketone neuropathy is mediated by adduction of a limited number of critical lysine amine groups on NF subunits (DeCaprio and Fowke, 1992Go; DeCaprio et al., 1997Go).

How does pyrrole formation cause axonal NF protein loss and subsequent atrophy? NF interactions with the cytoskeletal polymer could be modified directly by HD adduction of critical C-terminal lysine groups. Conversion of lysine amine groups to pyrrole adducts will cause a loss of net positive charge and formation of a hydrophobic moiety at the reaction site. This is likely to be followed by changes in NF physiological characteristics, which might include solubility, electrostatic potential, and three-dimensional structure (reviewed in DeCaprio, 1985Go, 1987Go; Sayre et al., 1985Go). If these chemically modified NF proteins cannot interact appropriately with the cytoskeletal network, axon atrophy could develop as accelerated transport and degradation of adducted, unincorporated triplet protein deplete regional subunit levels (for details see LoPachin and DeCaprio, 2004Go). The elegant proteomic studies involving HD illustrate the important concept that although neurotoxicant exposure might be associated with seemingly indiscriminant adduct formation, adduction of certain key nucleophilic protein residues (i.e., lysine residues within KSP repeats on NF tail regions) might mediate selective compromise of a critical neuronal character (i.e., axon caliber).

The Role of Adduct Formation in Acrylamide Neurotoxicity
Acrylamide (ACR) is a water-soluble, vinyl monomer (Figs. 1 and 3) that has multiple applications in the chemical and manufacturing industries; e.g., ore processing, soil grouting and dye synthesis. In addition, ACR is used extensively in molecular laboratories for separation of macromolecules by gel chromatography and is present in certain foods that have been prepared at very high temperatures (Tareke et al., 2000Go). Long-term, low-level exposure to monomeric ACR produces ataxia and skeletal muscle weakness in humans and experimental laboratory animal models (LeQuesne, 1985Go; LoPachin et al., 2002Go; Spencer and Schaumburg, 1974aGo). Early morphological studies suggested that the neurological defects associated with ACR intoxication were mediated by degeneration of distal axon regions in the PNS and CNS (reviewed in Spencer and Schaumburg, 1974bGo, 1977aGo,bGo, 1980aGo). However, substantial evidence from other morphological, electrophysiological, and neurochemical studies now implicate nerve terminals and cerebellar Purkinje neurons as neurotoxicologically relevant sites of ACR action (reviewed in LoPachin et al., 2002Go, 2003; LoPachin, 2004Go). Whereas ACR is a soft electrophile, the parent chemical can be metabolized to the reactive epoxide, glycidamide (Calleman et al., 1990Go). Therefore, at issue is the role of this metabolite in the production of neurotoxicity. A qualitative morphological study has suggested that glycidamide is involved in the mechanism of distal axon degeneration and accompanying neurological deficits in ACR-exposed rats (Abou-Donia et al., 1993Go). However, other research failed to find evidence for the induction of neurotoxicity by this metabolite (Barber et al., 2001Go; Brat and Brimijion, 1993Go; Costa et al., 1992Go, 1995Go). Thus, although glycidamide adduction of nucleic acid residues might mediate the genotoxicity associated with ACR intoxication of rodents (see above), neurotoxicity appears to be a product of the parent compound.

The molecular mechanism by which ACR produces nerve terminal and Purkinje cell damage has not been fully delineated. Nonetheless, understanding the structure and chemical properties of ACR can provide insight into the molecular mechanism of neurotoxicity. As indicated previously, ACR is an {alpha},ß-unsaturated aldehyde with electrophilic reactivity at the carbonyl carbon atom. As a soft electrophile, ACR could conceivably adduct amines, imidazoles and sulfhydryl groups on proteins via the Michael carbonyl condensation reaction (Fig. 3; Friedman, 1973Go; Kemp and Vellaccio, 1980Go). However, the reactivity of free thiols is greater than that of other soft nucleophilic centers and, consequently, the preferential in vivo target of ACR is sulfhydryl groups on protein cysteine residues and glutathione (reviewed in Calleman, 1996Go). Early chemical measurements (Bergmark et al., 1991Go; Cavins and Friedman, 1968Go; Dixit et al., 1986Go; Kemplay and Cavanagh, 1984aGo,bGo; Sega et al., 1989Go) and more recent mass spectroscopy studies (Barber and LoPachin, 2004Go; Bordini et al., 1999Go, 2000Go; Hall et al., 1993Go) have shown that ACR selectively reacts with cysteine residues of proteins to form S-(2-carbamoylethyl)-cysteine adducts. That cysteine adduct formation in nervous tissue might have mechanistic significance is suggested by the finding that in vitro exposure of brain synaptosomes to graded concentrations of ACR (0.001–1.0 M) produced decreases in evoked neurotransmitter release and parallel increases in adduct levels (Barber and LoPachin, 2004Go). Furthermore, in synaptosomes isolated from brains of ACR-intoxicated rats, cysteine adduct levels increased in concert with the temporal development of neurological deficits (Barber and LoPachin, 2004Go). These data indicate that the molecular mechanism of ACR neurotoxicity likely involves the formation of thiol adducts on neuronal proteins. Nonetheless, results from other studies do not support this conclusion (Hashimoto and Aldridge, 1970Go; Lapin et al., 1982Go; Martenson et al., 1995aGo,bGo). For example, Martenson et al. (1995aGo,bGo) showed that relatively high in vitro ACR concentrations (10–100 mM) caused morphological disruptions of dorsal root ganglion growth cones that were similar to changes induced by much lower concentrations of other sulfhydryl reagents; e.g., iodoacetic acid (IAA; 6.75–27 µM) or ethacrynic acid (ECA; 33.5–100 nM). The authors concluded that, since it is unlikely that mM concentrations of ACR are achieved during whole animal intoxication, the mechanism of in vivo nerve damage could not involve sulfhydryl alkylation. However, these findings are not necessarily contrary to an involvement of thiol adduction. It is important to recognize that all three chemicals produced similar in vitro neurotoxicity, although the potency of ACR for this effect was significantly lower than the relative potencies of either IAA or ECA. In a recent study of in vitro synaptosomal neurotransmitter release, LoPachin et al. (2004a)Go reported a similar dispersion of potencies among ACR, IAA and N-ethylmaleimide (NEM) with respect to inhibition of exocytosis. Because ACR is a relatively weak electrophile (Barber and LoPachin, 2004Go; Cavins and Freidman, 1968Go), high in vitro concentrations are likely required to generate, on an acute basis, intracellular cysteine adduct levels that exceed toxic thresholds. Furthermore, the relatively low potency for adduct formation is consistent with the cumulative in vivo neurotoxicity caused by ACR, where relatively long exposure durations or high daily dose-rates are necessary for the induction of neurological deficits (see detailed discussion in Barber and LoPachin, 2004Go; LoPachin et al., 2004aGo).

If adduction of protein cysteine residues is the basis of ACR neurotoxicity, it is not clear how such a generalized reaction leads to a specific effect on neurotransmitter release. One possibility is that ACR produces a relatively selective neurotoxic response by reacting with cysteine groups on proteins that regulate membrane fusion processes in nerve terminals and cell bodies. Our most recent research has focused on presynaptic mechanisms and has shown that ACR can form cysteine adducts with many nerve terminal proteins (Barber and LoPachin, 2004Go; LoPachin et al., 2004aGo). Among the adducted proteins that have been identified, several play a critical role in synaptic vesicle-membrane fusion and neurotransmitter release; i.e., N-ethylmaleimide-sensitive fusion (NSF) protein and synaptosomal associated protein of 25 kDa (SNAP-25; Barber and LoPachin, 2004Go). In particular, ACR adduction of NSF has significant mechanistic relevance. The fusion of synaptic vesicles with the presynaptic membrane, which is necessary for transmitter release, is mediated by the formation of 7S cis-SNARE complexes. NSF is an ATPase that dissociates the SNARE complexes into the corresponding trans-protein components; i.e., SNAP-25, synaptobrevin, syntaxin 1. Disassembly of presynaptic SNARE complexes is accomplished by ATP hydrolysis and allows continuous cycling of vesicle-membrane fusion (reviewed in Whiteheart et al., 2001Go). Recent tandem mass spectrometric analyses of NSF revealed that ACR formed adducts with Cys 264 located within domain I (#255-266) of the nucleotide-binding consensus sequence (Barber and LoPachin, 2004Go). This residue is critically involved in ATP hydrolysis and, therefore, determines the function of NSF (Matsushita et al., 2003Go; Tagaya et al., 1993Go; Whiteheart et al., 1994Go). Previous studies have shown that sulfhydryl alkylation by NEM inhibits NSF activity, reduces synaptosomal neurotransmitter release, and increases the levels of 7S SNARE complex in exposed synaptosomes (Lonart and Sudhof, 2000Go; LoPachin et al., 2004aGo; Nedvetsky et al., 2000Go; Tagaya et al., 1993Go; Whiteheart et al., 1994Go). Similarly, in vitro exposure to ACR also produced concentration-dependent decreases in synaptosomal release (LoPachin et al., 2004aGo) and increased the content of 7S SNARE complexes (Barber and LoPachin, 2004Go). These findings suggest that ACR does not affect the protein-protein interactions that mediate SNARE core assembly. Rather, the observed accumulation of synaptosomal 7S complexes is consistent with ACR inhibition of NSF ATPase activity, presumably through adduction of Cys 264.

Growing evidence now indicates that the neurological deficits associated with ACR intoxication are mediated by disruption of membrane fusion processes. Whereas the molecular mechanism of this disruption is not known, recent experiments suggest that ACR forms adducts with functionally important cysteine residues (Cys 264) on proteins (NSF) that play a regulatory role in membrane fusion. Whereas our proteomic studies have dealt primarily with presynaptic events, it is possible that Purkinje cell injury in the cerebellum also involves inhibition of critical membrane fusion processes (reviewed in LoPachin et al., 2003bGo; LoPachin, 2004Go). In general, it is becoming increasingly obvious that many cellular pathways and processes are regulated by cysteine sulfhydryl groups that act as redox sites for posttranslational modification of protein function by nitric oxide and other presumed signal mechanisms (Broillet, 1999Go; Forman et al., 2002Go; Jaffrey et al., 2001Go; Stamler et al., 2001Go). This suggests that the diverse toxicities induced by other soft electrophiles (see above) involve adduction of regulatory sulfhydryl groups that are subject to redox-based modulation.

Conclusions and Future Research Directions
In this review we have considered the possible mechanistic roles of protein adduct formation in toxic neuropathies. For other organ system toxicants and/or their active metabolites, covalent interaction with proteins has been a frequently proposed mechanism of cellular toxicity. However, although the formation of adducts by certain neurotoxicants has been exploited as a biomarker of exposure (DeCaprio, 1997Go; Tornqvist et al., 2002Go), protein adducts as a neuropathogenic hallmark have received limited attention. Nonetheless, many neurotoxic chemicals have electrophilic centers that could react with nucleophilic amino acid residues to form aducts on neuronal proteins (Table 1). Adduct formation could negatively impact the tertiary structure and/or function of these proteins and thereby interfere with, for example, energy metabolism, axonal transport or presynaptic neurotransmitter release. In contrast, the lack of specificity and low levels of adducts formed with key proteins seems counterintuitive to an involvement in primary pathophysiological processes. Regardless of this apparent enigma, it is possible for electrophilic neurotoxicants to produce specific effects by reacting with functionally critical nucleophilic centers on proteins that regulate cellular pathways or processes. Whereas the molecular mechanism of many neurotoxicants remains poorly defined, for others a greater understanding of neuropathogenesis has been achieved by investigating the role of protein adduct formation. Thus, for neurotoxicants such as carbon disulfide, isoniazid, dithiobiuret, sodium pyridinethione or p-bromophenylacetylurea the electrophilic nature and nucleophilic targets should continue to be explored as potential mechanistic components. This adduct-based approach could implement new research directions and lead to a better understanding of neurotoxic processes. Finally, understanding the chemistry and pathophysiological consequences of adduct formation could not only benefit neurotoxicology, but might also provide insight into mechanisms of human neurodegenerative diseases. For example, the neurodegeneration associated with Alzheimer's disease (AD) appears to involve oxidative damage characterized by protein oxidation (Hensley et al., 1995Go), increased expression of antioxidant enzymes (Pappolla et al., 1998Go), and elevated lipid peroxidation (Sayre et al., 1997Go). Lipid peroxidation produces several {alpha},ß-unsaturated aldehydes (e.g., acrolein, malondialdehyde, 4-hydroxy-2-nonenal) among which, acrolein has the highest nucleophilic reactivity (Picklo et al., 2002Go; Uchida et al., 1998Go). Acrolein-protein adducts have been detected in brains of AD patients (Calingasan et al., 1999Go) and are considered to be a biomarker of the accompanying oxidative stress (Uchida et al., 1998Go). In addition, it has been hypothesized that acrolein adduct formation with lysine residues on tau proteins might play a role in the development of neurofibrillary tangles that are a pathological hallmark of AD (Calingasan et al., 1999Go; Picklo et al., 2002Go). It is interesting to note that acrolein is a ubiquitous environmental pollutant (Ghilarducci and Tjeerdema, 1995Go). Thus, the pathophysiology of AD could involve protein adduct formation mediated by both endogenous and exogenous neurotoxic components.

The goals for future research should be to identify and quantitate neurotoxicologically important protein adducts and to establish their causal role in the production of nerve cell injury. This latter requirement is perhaps the most challenging, although recent advances in proteomic technology will facilitate such investigations. Current neurotoxicant classification schemes are based on presumed neuronal sites of action (e.g., axon, neuron, nerve terminal) as suggested by previous morphological characterizations (e.g., Spencer and Schaumburg, 1980aGo). However, morphological changes are often terminal endpoint events and, as such, are insensitive indices of neurotoxicity and of limited value (see LoPachin et al., 2000Go; LoPachin, 2004Go; LoPachin and DeCaprio, 2004Go). As we have discussed, the chemical structure of a neurotoxicant, the resulting toxicokinetics and strength of the corresponding electrophilic center are likely to determine target nucleophilic residues on specific proteins. This in turn could dictate the site of neuronal action and possible neuropathogenic mechanism. Therefore, development of future nosological schemes should incorporate the chemical nature of the toxicant and corresponding potential for protein adduct formation.


    ACKNOWLEDGMENTS
 
The authors acknowledge the helpful comments and criticisms of Dr. David Barber (University of Florida), Dr. Joseph Ross (Ross Toxicological Services), and Dr. Lisa Opanashuk (University of Rochester). Some of the research described in this manuscript was supported by NIH Grants from NIEHS to R.M.L. (ESO7912-07, ESO3830-18) and to A.P.D. (ESO5172). Conflict of interest: none declared.


    REFERENCES
 TOP
 ABSTRACT
 INTRODUCTION
 REFERENCES
 
Abou-Donia, M. B., Ibrahim, S. M., Corcoran, J. J., Lack, L., Friedman, M. A., and Lapadula, D. M. (1993). Neurotoxicity of glycidamide, an acrylamide metabolite, following intraperitoneal injections in rats. J. Toxicol. Environ. Health 39, 447–464.[ISI][Medline]

Anthony, D. C., Boekelheide, K., and Graham, D. G. (1983). The effect of 3,4-dimethyl substitution on the neurotoxicity of 2,5-hexanedione. I. Accelerated clinical neuropathy is accompanied by more proximal axonal swellings. Tox. Appl. Pharmacol. 71, 362–371.[CrossRef][ISI][Medline]

Barber, D., Hunt, J. R., Ehrich, M., Lehning, E. J., and LoPachin, R. M. (2001). Metabolism, toxicokinetics and hemoglobin adduct formation in rats following subacute and subchronic acrylamide dosing. NeuroToxicology 22, 341–353.[CrossRef][ISI][Medline]

Barber, D. S., and LoPachin, R. M. (2004). Proteomic analysis of acrylamide-protein adduct formation in rat brain synaptosomes. Toxicol. Appl. Pharmacol. 201, 120–136.[CrossRef][ISI][Medline]

Bergmark, E., Calleman, C. J., and Costa, L. G. (1991). Formation of hemoglobin adducts of acrylamide and its epoxide metabolite glycidamide in the rat. Toxicol. Appl. Pharmacol. 111, 352–363.[CrossRef][ISI][Medline]

Bordini, E., Hamdan, M., and Righetti, P. G. (1999). Probing the reactivity of S-S bridges to acrylamide in some proteins under high pH conditions by matrix-assisted laser desorption/ionisation. Rapid Comm. Mass Spec. 13, 1818–1827.[CrossRef][ISI][Medline]

Bordini, E., Hamdan, M., and Righetti, P. G. (2000). Probing acrylamide alkylation sites in cysteine-free proteins by matrix-assisted laser desorption/ionisation time-of-flight. Rapid Comm. Mass Spec. 14, 840–848.[CrossRef][ISI][Medline]

Brat, D. J., and Brimijion, S. (1993). Acrylamide and glycidamide impair neurite outgrowth in differentiating N1E.115 neuroblastoma without disturbing rapid bi-directional transport of organelles observed by video microscopy. J. Neurochem. 60, 2145–2152.[ISI][Medline]

Brimijoin, W. S., and Hammond, P. I. (1985). Acrylamide neuropathy in the rat: Effects of energy metabolism in sciatic nerve. Proc. Mayo Clinic 60, 3–8.[ISI][Medline]

Broillet, M.-C. (1999). S-Nitrosylation of proteins. Cell. Mol. Life Sci. 55, 1036–1042.[CrossRef][ISI][Medline]

Calingasan, N. Y., Uchida, K., and Gibson, G. E. (1999). Protein-bound acrolein: A novel marker of oxidative stress in Alzheimer's disease. J. Neurochem. 72, 751–756.[CrossRef][ISI][Medline]

Calleman, C. J. (1996). The metabolism and pharmacokinetics of acrylamide: Implications for mechanisms of toxicity and human risk estimation. Drug Metab. Rev. 28, 527–590.[ISI][Medline]

Calleman, C. J., Bergmark, E., and Costa, L. G. (1990). Acrylamide is metabolized to glycidamide in the rat: evidence from hemoglobin adduct formation. Chem. Res. Toxicol. 3, 406–412.[CrossRef][ISI][Medline]

Cavins, J. F., and Friedman, M. (1968). Specific modification of protein sulfhydryl groups with {alpha},ß-unsaturated compounds. J. Biol. Chem. 243, 3357–3360.[Abstract/Free Full Text]

Chermette, H. (1999). Chemicals reactivity indexes in density functional theory. J. Computational Chem. 20, 129–154.[CrossRef][ISI]

Chiu, F. C., Opanashuk, L. A., He, D. K., Lehning, E. J., and LoPachin, R. M. (2000). {gamma}-Diketone peripheral neuropathy. II. Neurofilament subunit content. Toxicol. Appl. Pharmacol. 165, 141–147.[CrossRef][ISI][Medline]

Cohen, S. D., Pumford, N. R., Khairallah, E. A., Boekelheide, K., Pohl, L. R., Amouzadeh, H. R., and Hinson, J. A. (1997). Selective protein covalent binding and target organ toxicity. Tox. Appl. Pharmacol. 143, 1–12.[CrossRef][ISI][Medline]

Coles, B. (1984–1985). Effects of modifying structure on electrophilic reactions with biological nucleophiles. Drug Metab. Rev. 15, 1307–1334.

Costa, L. G., Deng, H., Gregotti, C., Manzo, L., Faustman, E. M., Bergmark, E., and Calleman, C. J. (1992). Comparative studies on the neuro- and reproductive toxicity of acrylamide and its epoxide metabolite glycidamide in the rat. NeuroToxicology 13, 219–224.[ISI][Medline]

Costa, L. G., Deng, H., Calleman, C. J., and Bergmark, E. (1995). Evaluation of the neurotoxicity of glycidamide, an epoxide metabolite of acrylamide: Behavioral, neurochemical and morphological studies. Toxicology 98, 151–161.[CrossRef][ISI][Medline]

Couri, D., and Milks, M. (1982). Toxicity and the metabolism of the neurotoxic hexacarbons n-hexane, 2-hexanone and 2,5-hexanedione. Ann. Rev. Pharmacol. Toxicol. 22, 145–166.[CrossRef][ISI][Medline]

Dearfield, K. L., Douglas, G. R., Ehling, U. H., Moore, M. M., Sega, G. A., and Bruskick, D. J. (1995). Acrylamide: A review of its genotoxicity and an assessment of heritable genetic risk. Mut. Res. 330, 71–99.[ISI][Medline]

DeCaprio, A. P. (1985). Molecular mechanisms of diketone neurotoxicity. Chem.-Biol. Interactions 54, 257–270.[ISI][Medline]

DeCaprio, A. P. (1987). n-Hexane neurotoxicity. A mechanism involving pyrrole adduct formation in axonal cytoskeletal protein. NeuroToxicology 8, 199–210.[ISI][Medline]

DeCaprio, A. P. (1997). Biomarkers: Coming of age for environmental health and risk assessment. Enrion. Sci. Tech. 31, 1837–1848.[CrossRef]

DeCaprio, A. P. (2000). n-Hexane, metabolites and derivative. In Experimental and Clinical Neurotoxicology, 2nd ed. (P. S. Spencer and H. H. Schaumburg, Eds.), pp. 633–648. Oxford University Press, New York.

DeCaprio, A. P., and O'Neill, E. A. (1985). Alterations in rat axonal cytoskeletal proteins induced by in vitro and in vivo 2,5-hexanedione exposure. Toxicol. Appl. Pharmacol. 78, 235–247.[CrossRef][ISI][Medline]

DeCaprio, A. P., and Fowke, J. H. (1992). Limited and selective adduction of carboxyl-terminal lysines in the high molecular weight neurofilament proteins by 2,5-hexanedione in vitro. Brain Res. 586, 219–228.[CrossRef][ISI][Medline]

DeCaprio, A. P., Olajos, E. J., and Weber, P. (1982). Covalent binding of a neurotoxic n-hexane metabolite: Conversion of primary amines to substituted pyrrole adducts by 2,5-hexanedione. Toxicol. Appl. Pharmacol. 65, 440–450.[CrossRef][ISI][Medline]

DeCaprio, A. P., Strominger, N. L., and Weber, P. (1983). Neurotoxicity and protein binding of 2,5-hexanedione in the hen. Toxicol. Appl. Pharmacol. 68, 297–307.[CrossRef][ISI][Medline]

DeCaprio, A. P., Briggs, R. G., Jackowski, S. J., and Kim, J. C. S. (1988). Comparative neurotoxicity and pyrrole-forming potential of 2,5-hexanedione and perdeuterio-2,5-hexanedione in the rat. Toxicol. Appl. Pharmacol. 92, 75–85.[CrossRef][ISI][Medline]

DeCaprio, A. P., Kinney, E. A., and Fowke, J. H. (1997). Regioselective binding of 2,5-hexanedione to high-molecular-weight rat neurofilament proteins in vitro. Toxicol. Appl. Pharmacol. 145, 211–217.[CrossRef][ISI][Medline]

Dixit, R., Das, M., Seth, P. K., and Mukhtar, H. (1986). Interaction of acrylamide with bovine serum albumin. Environ. Res. 40, 365–371.[CrossRef][ISI][Medline]

Esterbauer, H., Schaur, R. J., and Zollner, J. (1991). Chemistry and biochemistry of 4-hydroxynonenal, malonaldehye and related aldehydes. Free Rad. Biol. Med. 11, 81–128.[CrossRef][ISI][Medline]

Eu, J. P., Sun, J., Xu, L., Stamler, J. S., and Meissner, G. (2000). The skeletal muscle calcium release channel: Coupled O2 sensor and NO signaling functions. Cell 102, 499–509.[CrossRef][ISI][Medline]

Forman, H. J., Torres, M., and Fukuto, J. (2002). Redox signaling. Mol. Cell. Biochem. 234/235, 49–62.[CrossRef]

Friedman, M. (1973). Reactive and unreactive sulfhydryl groups. In The Chemistry and Biochemistry of the Sulfhydryl Group in Amino Acids, Peptides and Proteins, pp. 311–348. Pergamon Press, New York.

Gamboa da Costa, G., Churchwell, M. I., Hamilton, P., Von Tungeln, L. S., Beland, F. A., Marques, M. M., and Doerge, D. R. (2003). DNA adduct formation from acrylamide via conversion to glycidamide in adult and neonatal mice. Chem. Res. Toxicol. 16, 1328–1337.[CrossRef][ISI][Medline]

Genter St. Clair, M. B., Amarnath, V., Moody, M. A., Anthony, D. C., Anderson, C. W., and Graham, D. G. (1988). Pyrrole oxidation and protein cross-linking as necessary steps in the development of {gamma}-diketone neuropathy. Chem. Res. Toxicol. 1, 179–185.[CrossRef][ISI][Medline]

Ghilarducci, D. P., and Tjeerdema, R. S. (1995). Fate and effects of acrolein. Rev. Environ. Contam. Toxicol. 144, 95–146.[ISI][Medline]

Graham, D. G., Anthony, D. C., Boekelheide, K., Maschmann, N. A., Richards, R. G., Wolfram, J. W., and Shaw, B. R. (1982). Studies of the molecular pathogenesis of hexane neuropathy. II. Evidence that pyrrole derivitization of lysyl residues leads to protein crosslinking. Tox. Appl. Pharmacol. 64, 415–422.[CrossRef][ISI][Medline]

Graham, D. G., Genter St. Clair, M. B., Amarnath, V., and Anthony, D. C. (1991). Molecular mechanisms of {gamma}-diketone neuropathy. Adv. Exp. Med. Biol. 283, 427–431.[Medline]

Graham, D. G., Amarnath, V., Valentine, W. M., Pyle, S. J., and Anthony D. C. (1995). Pathogenetic studies of hexane and carbon disulfide neurotoxicity. Crit. Rev. Toxicol. 25, 91–112.[ISI][Medline]

Guengerich, F. P., Geiger, L. E., Hogy, L. L., and Wright, P. L. (1981). In vitro metabolism of acrylonitrile to 2-cyanoethylene oxide, reaction with glutathione, and irreversible biding to proteins and nucleic acids. Cancer Res. 41, 4925–4931.[ISI][Medline]

Hall, S. C., Smith, D. M., Masiarz, F. R., Soo, V. W., Tran, H. M., Epstein, L. B., and Burlingame, A. L. (1993). Mass spectrometric and Edman sequencing of lipocortin I isolated by two-dimensional SDS/PAGE of human melanoma lysates. Proc. Natl. Acad. Sci. 90, 1927–1931.[Abstract/Free Full Text]

Harder, A., Escher, B. I., and Schwarzenbach, R. P. (2003). Applicability and limitation of QSARs for the toxicity of electrophilic chemicals. Environ. Sci. Technol. 37, 4955–4961.[CrossRef][ISI][Medline]

Harding, J. J. (1985). Nonenzymatic covalent posttranslational modification of proteins in vivo. In Advances in Protein Chemistry (C. B. Anfinsen, J. T. Edsall, and F. M. Richards, Eds.), 37, 247–334.

Hashimoto, K., and Aldridge, W. N. (1970). Biochemical studies of acrylamide, a neurotoxic agent. Biochem. Pharmacol. 19, 2591–2604.[CrossRef][ISI][Medline]

Hensley, K., Carney, J. M., Mattson, M. P., Aksenova, M., Aksenova, M., Gabbita, S. P., Wu, J. F., Carney, J. M., Lovell, M., Markesbery, W. R., and Butterfield, D. A. (1995). Brain regional correspondence between Alzheimer's disease histopathology and biomarkers of protein oxidation. J. Neurochem. 65, 2146–2156.[ISI][Medline]

Hess, D. T., Matsumoto, A., Nudeman, R., and Stamler, J. S. (2001). S-Nitrosylation: spectrum and specificity. Nature Cell Biol. 3, E46–E49.[CrossRef][ISI][Medline]

Hinson, J. A., and Roberts, D. W. (1992). Role of covalent and noncovalent interactions in cell toxicity: effects on proteins. Annu Rev Pharmacol Toxicol. 32, 471–510.[CrossRef][ISI][Medline]

Howland, R. D. (1981). The etiology of acrylamide neuropathy: Enolase, phosphofructokinase, and glyceraldehyde-3-phosphate dehydrogenase activities in peripheral nerve, spinal cord, brain and skeletal muscle of acrylamide-intoxicated cats. Tox. Appl. Pharmacol. 60, 324–333.[CrossRef][ISI][Medline]

Howland, R. D., Vyas, I. L., and Lowndes, H. E. (1980). The etiology of acrylamide neuropathy: Possible involvement of neuron-specific enolase. Brain Res. 190, 529–535.[CrossRef][ISI][Medline]

Jaffrey, S. R., Erdjument-Bromage, H., Ferris, C. D., Tempst, P., and Snyder, S. H. (2001). Protein S-nitrosylation: A physiological signal for neuronal nitric acid. Nature Cell Biol. 3, 193–197.[CrossRef][ISI][Medline]

Kemp, D. S., and Vellaccio, F. (1980). Conjugated alkenes. In Organic Chemistry, pp. 650–684. Worth Publishers, New York.

Kemplay, S., and Cavanagh J. B. (1984a). Effects of acrylamide and other sulfhydryl compound in vivo and in vitro on staining of motor nerve terminals by the zinc iodide-osmium technique. Musc. Nerve 7, 94–100.[CrossRef][ISI]

Kemplay, S., and Cavanagh J. B. (1984b). Effects of acrylamide and some other sulfhydryl reagents on spontaneous and pathologically induced terminal sprouting from motor end-plates. Musc. Nerve 7, 101–109.[CrossRef][ISI]

Ketterer, B. (1980). Interactions between carcinogens and proteins. Brit. Med. Bull. 36, 71–78.[ISI][Medline]

Krasavage, W. J., O'Donoghue, J. L., DiVincenzo, G. D., and Terhaar, C. J. (1980). The relative neurotoxicity of methyl n-butyl ketone, n-hexane and their metabolites. Toxicol. Appl. Pharmacol. 52, 433–441.[CrossRef][ISI][Medline]

Lapin, E. P., Weissbarth, S., Maker, H. S., and Lehrer, G. M. (1982). The sensitivities of creatine and adenylate kinases to the neurotoxins acrylamide and methyl n-butyl ketone. Environ. Res. 28, 21–31.[CrossRef][ISI][Medline]

Lehning, E. J., Jortner, B. S., Fox, J. H., Arezzo, J. C., Kitano, T., and LoPachin, R. M. (2000). {gamma}-Diketone peripheral neuropathy. I. Quantitative morphometric analyses of axonal atrophy and swelling. Toxicol. Appl. Pharmacol. 165, 127–140.[CrossRef][ISI][Medline]

LeQuesne, P. M. (1985). Clinical and morphological findings in acrylamide toxicity. NeuroToxicology 6, 17–24.

Liebler, D. C. (2002). Proteomic approaches to characterize protein modifications: New tools to study the effects of environmental exposures. Environ. Health Persp. 110, 3–9.[ISI][Medline]

Loechler, E. L. (1994). A violation of the Swain-Scott principle, and not SN1 versus SN2 reaction mechanisms, explains why carcinogenic alkylating agents can form different proportions of adducts at oxygen versus nitrogen in DNA. Chem. Res. Toxicol. 7, 277–280.[CrossRef][ISI][Medline]

Lonart, G., and Sudhof, T. C. (2000). Assembly of SNARE core complexes prior to neurotransmitter release sets the readily releasable pool of synaptic vesicles. J. Biol. Chem. 275, 27703–27707.[Abstract/Free Full Text]

LoPachin, R. M., Moore, R. W., Menahan, L. A., and Peterson, R. E. (1984). Glucose-dependent lactate production by homogenates of neuronal tissues prepared from rats treated with 2,4-dithiobiuret, acrylamide, p-bromophenylacetylurea and 2,5-hexanedione. NeuroToxicology 5, 25–36.[ISI][Medline]

LoPachin, R. M., and Lehning, E. J. (1997). The relevance of axonal swelling and atrophy to {gamma}-diketone neurotoxicity. NeuroToxicology 18, 7–22.[ISI][Medline]

LoPachin, R. M., Lehning, E. J., Opanashuk, L. A., and Jortner, B. S. (2000). Rate of neurotoxicant exposure determines morphological manifestations. Toxicol. Appl. Pharmacol. 167, 75–86.[CrossRef][ISI][Medline]

LoPachin, R. M., Ross, J. F., Reid, M. L., Das, S., Mansukhani, S., and Lehning, E. J. (2002). Neurological evaluation of toxic axonopathies in rats: acrylamide and 2,5-hexanedione. NeuroToxicology 23, 95–110.[CrossRef][ISI][Medline]

LoPachin, R. M., Jones, R. C., Patterson, T. A., Slikker, W., and Barber, D. S. (2003a). Application of proteomics to the study of molecular mechanisms in neurotoxicology. NeuroToxicology 24, 761–775.[CrossRef][ISI][Medline]

LoPachin, R. M., Balaban, C. D., and Ross, J. F. (2003b). Acrylamide axonopathy revisited. Toxicol. Appl. Pharmacol. 188, 135–153.[CrossRef][ISI][Medline]

LoPachin, R. M., Jones, R. C., Patterson, T. A., Slikker, W., and Barber, D. S. (2003c). Application of proteomics to the study of molecular mechanisms in neurotoxicology. NeuroToxicology 24, 761–775.[CrossRef][ISI][Medline]

LoPachin, R. M. (2004). The changing view of acrylamide neurotoxicity. NeuroToxicology 25, 617–630.[CrossRef][ISI][Medline]

LoPachin, R. M., and DeCaprio, A. P. (2004). {gamma}-Diketone neuropathy: Axon atrophy and the role of cytoskeletal protein adduction. Toxicol. Appl. Pharmacol. 199, 20–34.[CrossRef][ISI][Medline]

LoPachin, R. M., Schwarcz, A. I., Gaughan, C. L., Mansukhani, S., and Das, S. (2004a). In vivo and in vitro effects of acrylamide on synaptosomal neurotransmitter uptake and release. NeuroToxicology 25, 349–363.[CrossRef][ISI][Medline]

LoPachin, R. M., He, D., Reid, M. L., and Opanashuk, L. A. (2004b). 2,5-Hexanedione-induced changes in the monomeric neurofilament protein content of rat spinal cord fractions. Tox. Appl. Pharmacol. 198, 61–73.[CrossRef][ISI][Medline]

LoPachin, R. M., He, D., and Reid, M. L. (2005). 2,5-Hexanedione-induced changes in the neurofilament protein subunit pools of rat peripheral nerve. NeuroToxicology. 26, 229–240.[CrossRef][ISI][Medline]

Lotti, M. (2000). Organophosphorous compounds. In Experimental and Clinical Neurotoxicology, pp. 897–925. Oxford University Press, New York.

Martenson, C. H., Sheetz, M., and Graham, D. G. (1995a). In vitro acrylamide exposure alters growth cone morphology. Toxicol. Appl. Pharmacol. 131, 119–129.[CrossRef][ISI][Medline]

Martenson, C. H., Odom, A., Sheetz, M. P., and Graham, D. G. (1995b). The effect of acrylamide and other sulfhydryl alkylators on the ability of dynein and kinesin to translocate microtubules in vitro. Toxicol. Appl. Pharmacol. 133, 73–81.[CrossRef][ISI][Medline]

Matsuoka, M., and Igisu, H. (1992). Brain energy metabolites in mice intoxicated with acrylamide: effects of ischemia. Tox. Letters 62, 39–43.[CrossRef][ISI]

Matsushita, K., Morrell, C. N., Cambien, B., Yang, S.-X., Yamakuchi, M., Bao C., Hara M. R., Quick, R.A., Cao, W., O'Rourke, B., Lowenstein, J. M., Pevsner, J., Wagner, D. D., and Lowenstein, C. J. (2003). Nitric oxide regulates exocytosis by S-nitrosylation of N-ethylmaleimide-sensitive factor. Cell 115, 139–150.[CrossRef][ISI][Medline]

Miller, E. C., and Miller, J. A. (1981). Searches for the ultimate chemical carcinogens and their reactions with cellular macromolecules. Cancer 47, 2327–2345.[ISI][Medline]

Monaco, S., Autilio-Gambetti, L., Zabel, D., and Gambetti, P. (1985). Giant axonal neuropathy. Acceleration of neurofilament transport in optic axons. Proc. Natl. Acad. Sci. 82, 920–924.[Abstract/Free Full Text]

Moore, C. P., Rodney, G., Zhang, J.-Z., Santacruz-Toloza, L., Strasburg, G., and Hamilton, S. L. (1999). Apocalmodulin and Ca2+ calmodulin bind to the same region on the skeletal muscle Ca2+ release channel. Biochemistry 38, 8532–8537.[CrossRef][ISI][Medline]

Muma, N. A., and Hoffman, P. N. (1993). Neurofilaments are intrinsic determinants of axonal caliber. Micron 24, 677–683.[CrossRef][ISI]

Nedvetsky, P. I., Konev, S. V., Rakovich, A. A., Petrenko, S. V., and Mongin, A. A. (2000). Effects of nitric oxide donors on Ca2+-dependent [14C]GABA release from brain synaptosomes: The role of –SH groups. Biochemistry 65, 1027–1035.[Medline]

Nelson, S. D., and Pearson, P. G. (1990). Covalent and noncovalent interactions in acute lethal cell injury caused by chemicals. Annu. Rev. Pharmacol. Toxicol. 30, 169–195.[CrossRef][ISI][Medline]

Oesch, R., Kaubisch, N., Jerina, D. M., and Daly, J. W. (1971). Hepatic epoxide hydrase. Structure-activity relationships for substrates and inhibitors. Biochemistry 10, 4858–4864.[CrossRef][ISI][Medline]

Orstan, A., and Gafni, A. (1990). The interaction of acrylamide with glyceraldehyde-3-phosphate dehydrogenase. Structural modifications in the enzyme studied by fluorescence techniques. Photochem. Photobiol. 51, 725–731.[ISI][Medline]

Pappolla, M. A., Chyan, Y. J., Kim, K. S., and Bozner, P. (1998). Evidence of oxidative stress and in vivo neurotoxicity of beta-amyloid in a transgenic mouse model of Alzheimer's disease: A chronic oxidative paradigm for testing antioxidant therapies in vivo. Am. J. Pathol. 152, 871–877.[Abstract]

Parthasarathi, R., Subramanian, V., Roy, D. R., and Chattaraj, P. K. (2004). Electrophilicity index as a possible descriptor of biological activity. Bioorg. Medicinal Chem. 12, 5533–5543.[CrossRef][ISI]

Pearson, R. G., and Songstad, J. (1967). Application of the principle of hard and soft acids and bases to organic chemistry. J. Am. Chem. Soc. 89, 1827–1836.[CrossRef][ISI]

Picklo, M. J., Montine, T. J., Amarnath, V., and Neely, M. D. (2002). Carbonyl toxicology and Alzheimer's disease. Toxicol. Appl. Pharmacol. 184, 187–197.[CrossRef][ISI][Medline]

Pyle, S. J., Amarnath, V., Graham, D. G., and Anthony, D. C. (1992). Alterations in neurofilament transport induced by 2,5-hexanedione and 3-acetyl-2,5-hexanedione. J. Neuropathol. Exp. Neurol. 51, 451–458.[ISI][Medline]

Sakaguchi, T., Okada, M., Kitamura, T., and Kawasaki, K. (1993). Reduced diameter and conduction velocity of myelinated fibers in the sciatic nerve of a neurofilament-deficient mutant quail. Neurosci. Lett. 153, 65–68.[CrossRef][ISI][Medline]

Sega, G. A., Valdivia, A. R. P., Tancongco, C. P., and Brimer, P. A. (1989). Acrylamide binding to the DNA and protamine of spermiogenic stages in the mouse and its relationship to genetic damage. Mutat. Res. 216, 221–230.[CrossRef][ISI][Medline]

Sakamoto, J., and Hashimoto, K. (1985). Effects of acrylamide and related compounds on glycolytic enzymes in mouse brain in vitro. Arch. Toxicol. 57, 276–281.[CrossRef][ISI][Medline]

Sayre, L. M., Autilio-Gambetti, L., and Gambetti, P. (1985). Pathogenesis of experimental giant neurofilamentous axonopathies: A unified hypothesis based on chemical modification of neurofilaments. Brain Res. Rev. 10, 69–83.[CrossRef][ISI]

Sayre, L. M., Zelasko, D. A., Harris, P. L. R., Perry, G., Salomon, R. G., and Smith, M. A. (1997). 4-Hydroxynonenal-derived advanced lipid peroxidation end products are increased in Alzheimer's disease. J. Neurochem. 68, 2092–2097.[ISI][Medline]

Spencer, P. S., and Schaumburg H. H. (1974a). A review of acrylamide neurotoxicity. Part I. Properties, uses and human exposure. Can. J. Neurol. Sci. 1, 151–169.

Spencer, P. S., and Schaumburg H. H. (1974b). A review of acrylamide neurotoxicity. Part II. Experimental animal neurotoxicity and pathologic mechanisms. Can. J. Neurol. Sci. 1, 170–192.[Medline]

Spencer, P. S., and Schaumburg H. H. (1976). Central-peripheral distal axonopathy- The pathology of dying-back polyneuropathies. In Progress in Neuropathology (H. Zimmerman, Ed.), pp. 253–276. Grune & Stratton, New York.

Spencer, P. S., and Schaumburg, H. H. (1977a). Ultrastructural studies of the dying-back process. III. The evolution of experimental peripheral giant axonal degeneration. J. Neuropath. Exp. Neurol. 36, 276–299.[ISI][Medline]

Spencer, P. S., and Schaumburg, H. H. (1977b). Ultrastructural studies of the dying-back process. IV. Differential vulnerability of PNS and CNS fibers in experimental central-peripheral distal axonopathy. J. Neuropath. Exp. Neurol. 36, 300–320.[ISI][Medline]

Spencer, P. S., Sabri, M. I., Schaumburg, H. H., and Moore, C. L. (1979). Does a defect of energy metabolism in the nerve fiber underlie axonal degeneration in polyneuropathies? Annals Neurol. 5, 501–507.[CrossRef][ISI][Medline]

Spencer, P. S., and Schaumburg, H. H. (1980a). Classification of neurotoxic disease: A morphological approach. In Experimental and Clinical Neurotoxicology (P. S. Spencer and H. H. Schaumburg, Eds.), pp. 92–99. Williams and Wilkins, Baltimore.

Spencer, P. S., Couri, D., and Schaumburg, H. H. (1980b). n-Hexane and methyl n-butyl ketone. In Experimental and Clinical Neurotoxicology (P. S. Spencer and H. H. Schaumburg, Eds.), pp. 456–475. Williams and Wilkins, Baltimore.

Stamler, J. S., Toone, E. J., Lipton, S. A., and Sucher, N. J. (1997). (S)NO signals: translocation, regulation and a consensus motif. Neuron 18, 691–696.[CrossRef][ISI][Medline]

Stamler, J. S., Lamas, S., and Fang, F. C. (2001). Nitrosylation: The prototypic redox-based signaling mechanism. Cell 106, 675–683.[CrossRef][ISI][Medline]

Sumner, S. C. J., MacNeela, J. P., and Fennell, T. R. (1992). Characterization and quantitation of urinary metabolites of [1,2,3–13C]acrylamide in rats and mice using 13C nuclear magnetic resonance spectroscopy. Chem. Res. Toxicol. 5, 81–89.[CrossRef][ISI][Medline]

Sumner, S. C. J., Fennell, T. R., Moore, T. A., Chanas, B., Gonzalez, F., and Ghanayem, B.I. (1999). Role of cytochrome P450 2E1 in the metabolism of acrylamide and acrylonitrile in mice. Chem. Res. Toxicol. 12, 1110–1116.[CrossRef][ISI][Medline]

Sun, J., Xin, C., Eu, J. P., Stamler, J. S., and Meissner, G. (2001). Cysteine-3635 is responsible for skeletal muscle ryanodine receptor modulation by NO. Proc. Natl. Acad. Sci. 98, 11158–11162.[Abstract/Free Full Text]

Swain, C. G., and Scott, C. B. (1953). Quantitative correlation of relative rates. Comparison of hydroxide ion with other nucleophilic reagents toward alkyl halides, esters, epoxides and acyl halides. J. Am. Chem. Soc. 75, 141–147.[CrossRef][ISI]

Tagaya, M., Wilson, D. W., Brunner, M., Arango, N., and Rothman, J. E. (1993). Domain structure of an N-ethylmaleimide-sensitive fusion protein involved in vesicular transport. J. Biol. Chem. 268, 2662–2666.[Abstract/Free Full Text]

Tareke, E., Rydberg, P., Karlsson, P., Eriksson, S., and Tornqvist, M. (2000). Acrylamide: A cooking carcinogen? Chem. Res. Toxicol. 13, 517–522.[CrossRef][ISI][Medline]

Tong, G. C., Cornwell, W. K., and Means, G. E. (2004). Reactions of acrylamide with glutathione and serum albumin. Toxicol. Lett. 147, 127–131.[CrossRef][ISI][Medline]

Tornqvist, M., Fred, C., Haglund, J., Helleberg, H., Paulsson, B., and Rydberg, P. (2002). Protein adducts: Quantitative and qualitative aspects of their formation, analysis and applications. J. Chromatog. B 778, 279–308.[CrossRef][ISI]

Uchida, K., Kanematsu, M., Sakai, K., Matsuda, T., Hattori, N., Mizuno, Y., Suzuki, D., Miyata, T., Noguchi, N., Niki, E., and Osawa, T. (1998). Protein-bound acrolein: Potential markers for oxidative stress. Proc. Nat. Acad. Sci. 95, 4882–4887.[Abstract/Free Full Text]

Vogel, E. W., and Nivard, M. J. M. (1994). The subtlety of alkylating agents in reactions with biological macromolecules. Mutat. Res. 305, 13–32.[CrossRef][ISI][Medline]

Whiteheart, S. W., Rossnagel, K., Buhrow, S. A., Brunner, M., Jaenicke, R., and Rothman, J. E. (1994). N-Ethylmaleimide-sensitive fusion protein: A trimeric ATPase whose hydrolysis of ATP is required for membrane fusion. J. Cell Biol. 126, 945–954.[Abstract]

Whiteheart, S. W., Schraw, T., and Matveeva, E. A. (2001). N-Ethylmaleimide sensitive factor (NSF) structure and function. Int. Rev. Cytol. 207, 71–112.[ISI][Medline]

Willhite, C. C. (1982). Toxicology update: acrylonitrile. J. Appl. Toxicol. 2, 54–63.

Woutersen, R. A. (1998). Toxicological profile of acrylonitrile. Scand. J. Work Environ. Health 24, 5–9.[ISI][Medline]

Yagi, Y. (1994) Effects of 2,5-hexanedione on rat spinal pyramidal tract. Jpn. J. Ind. Health 36, 173–180.

Yamaguchi, N., Xin, C., and Meissner, G. (2001). Identification of apocalcodulin and Ca2+-calmodulin regulatory domain in skeletal muscle Ca2+ release channel, ryanodine receptors. J. Biol. Chem. 276, 22579–22585.[Abstract/Free Full Text]