Elsevier

NeuroToxicology

Volume 24, Issue 1, January 2003, Pages 125-136
NeuroToxicology

Acrylamide Neuropathy: III. Spatiotemporal Characteristics of Nerve Cell Damage in Forebrain

https://doi.org/10.1016/S0161-813X(02)00155-9Get rights and content

Abstract

Previous studies of acrylamide (ACR) neuropathy in rat PNS [Toxicol. Appl. Pharmacol. (1998) 151: 211–221] and in spinal cord, brainstem and cerebellum [NeuroToxicology (2002a) 23: 397–414; NeuroToxicology (2002b) 23: 415–429] have suggested that axon degeneration was not a primary effect and was, therefore, of unclear neurotoxicological significance. To conclude our studies of neurodegeneration in rat CNS during ACR neurotoxicity, a cupric silver stain method was used to define spatiotemporal characteristics of nerve cell body, dendrite, axon and terminal argyrophilia in forebrain regions and nuclei. Rats were exposed to ACR at a dose-rate of either 50 mg/kg per day (i.p.) or 21 mg/kg per day (p.o.) and at selected times brains were removed and processed for silver staining. Results show that intoxication at either ACR dose-rate produced a terminalopathy, i.e. nerve terminal degeneration and swelling were present in the absence of significant argyrophilic changes in neuronal cell bodies, dendrites or axons. Exposure to the higher ACR dose-rate caused early onset (day 5), widespread nerve terminal degeneration in most of the major forebrain areas, e.g. cerebral cortex, thalamus, hypothalamus and basal ganglia. At the lower dose-rate, nerve terminal degeneration in the forebrain developed early (day 7) but exhibited a relatively limited spatial distribution, i.e. anteroventral thalamic nucleus and the pars reticulata of the substantia nigra. Several hippocampal regions were affected at a later time point (day 28), i.e. CA1 field and subicular complex. At both dose-rates, argyrophilic changes in forebrain nerve terminals developed prior to the onset of significant gait abnormalities. Thus, in forebrain, ACR intoxication produced a pure terminalopathy that developed prior to the onset of significant neurological changes and progressed as a function of exposure. Neither dose-rate used in this study was associated with axon degeneration in any forebrain region. Our findings indicate that nerve terminals were selectively affected in forebrain areas and, therefore, might be primary sites of ACR action.

Section snippets

INTRODUCTION

Acrylamide (ACR) is a well-recognized neurotoxicant that is used extensively in several chemical industries (US Environmental Protection Agency, 1985, US Environmental Protection Agency, 1988; Spencer and Schaumburg, 1974a, Spencer and Schaumburg, 1974b; Tilson, 1981). Exposure of humans and laboratory animals to monomeric ACR causes ataxia and hindlimb skeletal muscle weakness presumably mediated by axon damage classified as a central-peripheral distal axonopathy (see reviews by Gold and

Treatment and Neurological Evaluation of Animals

All aspects of this study were in accordance with the NIH Guide for Care and Use of Laboratory Animals and were approved by the Montefiore Medical Center Animal Care Committee. Adult male rats (Sprague–Dawley, 250–275 g; Taconic Farms, Germantown, NY) were used in this study. Rats were housed individually in polycarbonate boxes, and drinking water and Purina Rodent Laboratory Chow (Purina Mills Inc., St. Louis, MO) were available ad libitum. The animal room was maintained at approximately 22 °C

Neurological Evaluation

Rats intoxicated at the 50 mg/kg per day dose-rate exposure group had a mean (±S.E.M.) starting body weight of 267±3 g, which declined steadily to 250±5 g at endpoint (11 days; Fig. 1A). This represents a 6% decrease relative to initial weight. Rats in the age-matched control group had a similar starting body weight (270±4 g) and, during the 11 day experimental period, gained approximately 27% of original body weight (333±7 g). Thus, when compared to age-matched controls, the mean body weight of

DISCUSSION

We have suggested that axon degeneration was a secondary effect related to duration of ACR exposure (see reviews by LoPachin et al., 2000, LoPachin et al., 2002a). However, supporting evidence was primarily derived from studies in PNS tissues (e.g. sciatic, tibial and sural nerve; Lehning et al., 1998). Based on results from previous investigations (O’Shaughnessy and Losos, 1986, Burek et al., 1980, Yoshimura et al., 1992), the possibility existed that at higher dose-rates axonopathy was

SUMMARY

The present study has shown that, regardless of dose-rate, nerve terminal degeneration in rat forebrain was an early consequence of ACR intoxication. This effect was selective since neither dendrites, cell bodies nor axons exhibited argyrophilic changes at any dose-rate. In PNS, brainstem and spinal cord, ACR intoxication at the 50 mg/kg per day dose-rate also produced selective terminalopathy, whereas at the 21 mg/kg per day dose-rate, initial nerve terminal argyrophilia was followed by axon

Acknowledgements

Research presented in this manuscript was supported by a grant (to R.M.L.) from the National Institute of Environmental Health Sciences (RO1 ES03830-16) and by funds provided by the Procter and Gamble Co., Cincinnati, OH.

References (64)

  • E.J Lehning et al.

    Acrylamide neuropathy. I. Spatiotemporal characteristics of nerve cell damage in rat cerebellum

    NeuroToxicology

    (2002)
  • E.J Lehning et al.

    Acrylamide neuropathy. II. Spatiotemporal characteristics of nerve cell damage in rat brainstem and spinal cord

    NeuroToxicology

    (2002)
  • R.M LoPachin

    The role of fast axonal transport in acrylamide pathophysiology: mechanism or epiphenomenon?

    NeuroToxicology

    (2002)
  • R.M LoPachin et al.

    Rate of neurotoxicant exposure determines morphologic manifestations of distal axonopathy

    Toxicol. Appl. Pharmacol.

    (2000)
  • R.M LoPachin et al.

    Nerve terminals as the primary site of acrylamide action: a hypothesis

    NeuroToxicology

    (2002)
  • R.M LoPachin et al.

    Neurological evaluation of chemically-induced neuropathies: acrylamide and 2,5-hexanedione

    NeuroToxicology

    (2002)
  • S.A Morris et al.

    Synaptic vesicle recycling: the Ferrari of endocytosis?

    Curr. Biol.

    (1995)
  • T.C Sudhof

    The synaptic vesicle cycle revisited

    Neuron

    (2000)
  • Balaban CD. The use of selective silver degeneration stains in neurotoxicology. In: Isaacson RL, Jensen KF, editors....
  • F Benfenati et al.

    Protein–protein interactions and protein modules in the control of neurotransmitter release

    Philos. Trans. R. Soc. Lond.

    (1999)
  • J.D Burek et al.

    Subchronic toxicity of acrylamide administered to rats in drinking water followed by up to 144 days of recovery

    J. Environ. Pathol. Toxicol.

    (1980)
  • C.J Calleman

    The metabolism and pharmacokinetics of acrylamide: implicationsfor mechanisms of toxicity and human risk estimation

    Drug Met. Rev.

    (1996)
  • J.B Cavanagh

    The significance of the dying-back process in experimental and human neurological disease

    Int. Rev. Exp. Pathol.

    (1964)
  • J.B Cavanagh

    The dying back process

    Arch. Pathol. Lab. Med.

    (1979)
  • J.B Cavanagh

    The pathokinetics of acrylamide intoxication: a reassessment of the problem

    Neuropathol. Appl. Neurobiol.

    (1982)
  • Cavanagh JB. Mechanisms of axon degeneration in three toxic neuropathies: organophosphorus, acrylamide and hexacarbon...
  • J.B Cavanagh et al.

    Ultrastructural features of the Purkinje cell damage caused by acrylamide in the rat: a new phenomenon in cellular neuropathology

    J. Neurocytol.

    (1983)
  • J.B Cavanagh et al.

    Selective loss of Purkinje cells from the rat cerebellum caused by acrylamide and the responses of β-glucuronidase and β-galactosidase

    Acta Neuropathol.

    (1982)
  • R.L DeGrandchamp et al.

    Early degeneration and sprouting at the rat neuromuscular junction following acrylamide administration

    Neuropathol. Appl. Neurobiol.

    (1990)
  • de Olmos JS, Ebbesson SOE, Heimer L. Silver methods for the impregnation of degenerating axoplasm. In: Heimer L,...
  • A.S Fix et al.

    Integrated evaluation of central nervous system lesions: stains for neurons, astrocytes and microglia reveal the spatial and temporal features of MK-80-induced neuronal necrosis in the rat cerebral cortex

    Toxicol. Pathol.

    (1996)
  • A.S Fix et al.

    MK-801 neurotoxicity in cupric silver-stained sections: lesion reconstruction by three-dimentional computer image analysis

    Toxicol. Pathol.

    (2000)
  • Cited by (49)

    • Subchronic exposure to acrylamide leads to pancreatic islet remodeling determined by alpha cell expansion and beta cell mass reduction in adult rats

      2018, Acta Histochemica
      Citation Excerpt :

      The main route of acrylamide formation in food is Maillard chemical reaction between amino acids, especially asparagine, particularly present in potatoes and grains, and reducing sugars such as glucose and fructose (Mottram et al., 2002; Stadler et al., 2002; Yaylayan et al., 2003). So far studies have already shown adverse health effects of acrylamide including neurotoxicity (Burek et al., 1980; Tyl et al., 2000; LoPachin et al., 2002; Lehning et al., 2003), carcinogenicity (Carere, 2006; Hogervorst et al., 2010), reproductive and developmental toxicity (Dearfield et al., 1988; Tyl and Friedman, 2003; Wang et al., 2010) and endocrine disruption (Sakamoto and Hashimoto, 1986; Khan et al., 1999; Yang et al., 2005; Mannaa et al., 2006; Hamdy et al., 2012). Ability of acrylamide to induce cancer in experimental animals places it in a probable human carcinogen class of chemicals by both International Agency for Research on Cancer – IARC (Group 2A) and US Environmental Protection Agency – EPA (Group B2).

    • Acrylamide alters glycogen content and enzyme activities in the liver of juvenile rat

      2015, Acta Histochemica
      Citation Excerpt :

      Acrylamide (AA) (CASR No. 79-06-1) is highly reactive, water-soluble monomer which is considered as toxic and potentially cancer causing chemical to humans. Adverse health effects regarding AA and its more reactive metabolite glycidamide (GA) were detected in experimental animals, and included neurotoxicity (Burek et al., 1980; Tyl et al., 2000; Lehning et al., 2003; LoPachin et al., 2003), genotoxicity (Paulsson et al., 2003; Maniere et al., 2005; Yang et al., 2005), and carcinogenicity (Carere, 2006; Hogervorst et al., 2010). Well-documented human epidemiological studies claim that AA has neurotoxic effects (Garland and Patterson, 1967; He et al., 1989) and it is probably carcinogenic to humans (IARC, 1994).

    • Structural and ultrastructural evidence of neurotoxic effects of fried potato chips on rat postnatal development

      2011, Nutrition
      Citation Excerpt :

      Studies on experimental models have shown that ACR exposure results in skeletal muscle weakness, particularly in the gastrocnemius muscle [20,22–24]. A recent study has shown that ACR levels of potato chips prepared by frying increase with frying temperature (19.6 ng/g at 170°C, 39 ng/g at 180°C, and 95 ng/g at 190°C) [25]. Furthermore, humans are continuously exposed to ACR at low levels through the diet (and potentially smoking), which raises the possibility of long-term neurologic degeneration caused by ACR during the life span [26].

    View all citing articles on Scopus
    View full text