Elsevier

Brain Research

Volume 1237, 27 October 2008, Pages 91-100
Brain Research

Research Report
Choline supplementation attenuates learning deficits associated with neonatal alcohol exposure in the rat: Effects of varying the timing of choline administration

https://doi.org/10.1016/j.brainres.2008.08.048Get rights and content

Abstract

Despite the harmful effects of fetal alcohol exposure, some pregnant women continue to drink alcohol. Thus, it is imperative to pursue safe, effective treatments for children with fetal alcohol spectrum disorders. Using an animal model, our laboratory has demonstrated that choline, an essential nutrient, effectively reduces the severity of some fetal alcohol effects, even when administered after the ethanol insult is complete. The present study investigated whether there is a critical developmental period when choline is most effective in attenuating ethanol's teratogenic effects. Sprague–Dawley rats were exposed to 5.25 g/kg/day ethanol during the third trimester equivalent brain growth spurt (postnatal days (PD) 4–9) via intubation. A non-intubation control group and a sham intubation control group were included. Following ethanol exposure, pups received subcutaneous injections of saline vehicle or choline chloride (100 mg/kg/day) from PD 11–20, PD 21–30, or PD 11–30. Beginning on PD 45, subjects were tested on a Morris water maze spatial learning task. Performance of both the ethanol-exposed group that did not receive choline and the ethanol-exposed group treated with choline from PD 21–30 was significantly impaired compared to controls during acquisition of the Morris water maze task. Performance of ethanol-exposed groups treated with choline from PD 11–20 or PD 11–30 was intermediate, not differing significantly from any other groups. However, during the probe trial, ethanol exposure produced significant deficits in spatial memory which were mitigated by all choline treatments, regardless of the timing of administration. These findings suggest that choline's therapeutic window may be very large, or spans across the two developmental periods examined in this study. Importantly, these findings indicate that choline supplementation may effectively reduce some alcohol-related learning impairments, even when administered in later childhood.

Introduction

Fetal alcohol spectrum disorders (FASD) is a term used to describe the wide range of physical, neurological, and behavioral alterations associated with prenatal exposure to ethanol (NIAAA, 2000, Riley and McGee, 2005, Sokol et al., 2003). Alcohol-related disruptions in central nervous system (CNS) development constitute the most devastating consequences (see Spadoni et al., 2007 for review), with damage to areas such as the cerebellum (O'Hare et al., 2005, Sowell et al., 1996), corpus callosum (Bookstein et al., 2007, Riley et al., 1995, Sowell et al., 2008a, Sowell et al., 2001), caudate (Mattson et al., 1996), cortex (Sowell et al., 2008b) and hippocampus (Autti-Ramo et al., 2002, Riikonen et al., 1999). Ethanol-induced neuropathology leads to a variety of behavioral problems, including hyperactivity, attention deficits, motor dysfunction, impairments in language and social skills, and learning deficits (Coles et al., 1997, Kelly et al., 2000, Mattson et al., 2001, Riley and McGee, 2005, Roebuck et al., 1999). Although FASDs are preventable, many pregnant women still drink alcohol and FASDs continue to constitute a serious health concern throughout the world (Warren et al., 2001). In the U.S. alone, it is estimated that 1 in 100 live births exhibit at least some adverse effects of prenatal alcohol exposure (Sampson et al., 1997). Thus, it is imperative that potential interventions/treatments for FASD be identified.

Numerous animal studies have shown that pharmacological manipulations may effectively block ethanol's teratogenic effects (Bonthius et al., 2003, Chen et al., 2005, Endres et al., 2005, Ieraci and Herrera, 2006, Thomas et al., 2001, Wilkemeyer et al., 2003, Wilkemeyer et al., 2002). However, to be effective, such experimental therapeutics would have to administered during ethanol exposure and there is concern that such treatments could exert undesirable effects on nontargeted developmental processes. Moreover, the opportunity for treatment may not be available until a child is already born with FASD. Fortunately, both human and animal studies have shown that postnatal environmental manipulations can reduce the severity of some fetal alcohol effects. Animal studies have shown that enriched environment (Hannigan and Berman, 2000, Hannigan et al., 1993), exercise (Christie et al., 2005, Thomas et al., in press), and acrobatic motor training (Klintsova et al., 2000, Klintsova et al., 2002) can reduce alcohol's behavioral teratogenic effects. Similarly, clinical studies have shown that specialized training can improve social skills (O'Connor et al., 2006), behavioral problems (Kable et al., 2007) and math skills (Kable et al., 2007). These data suggest that the CNS can respond to environmental interventions even after a developmental alcohol insult.

We have shown that choline supplementation may serve as an effective dietary intervention to reduce the severity of alcohol's adverse effects on behavioral development. Choline is an essential nutrient that is critical for brain development and function (Zeisel, 2006a). A growing literature illustrates that pre- and/or early postnatal choline supplementation in otherwise typically developing rats leads to long-lasting morphological (Li et al., 2004, Loy et al., 1991, Williams et al., 1998), electrophysiological (Jones et al., 1999, Pyapali et al., 1998), and neurochemical (Blusztajn et al., 1998, Cermak et al., 1999, Cermak et al., 1998, Coutcher et al., 1992, Meck et al., 1989, Montoya et al., 2000) changes in the CNS that contribute to long-lasting cognitive enhancements (McCann et al., 2006, Meck and Williams, 1997, Meck et al., 1988, Meck and Williams, 2003). Perinatal choline supplementation can also enhance cognitive improvements to later environmental manipulations, such as enriched environment (Tees, 1999). Using a rat model of fetal alcohol exposure, we found that choline supplementation not only reduces the severity of prenatal alcohol effects when administered concurrently with the alcohol (Thomas et al., submitted for publication), but is also effective when administered postnatally. For example, we first reported that choline supplementation during postnatal days (PD) 2–21 reduced the severity of working memory deficits in adult rats following prenatal alcohol exposure (Thomas et al., 2000). Importantly, these findings illustrated that choline is effective even when administered after ethanol exposure is complete. More recently, it's been shown that choline supplementation from either PD 4–30 or 10–30 reduces overactivity in the open field (Thomas et al., 2007, Thomas et al., 2004), spatial reversal learning deficits (Thomas et al., 2004), trace eyeblink conditioning deficits (Tran and Thomas, 2007), trace fear conditioning deficits (Wagner and Hunt, 2006), and spatial learning deficits (Thomas et al., 2007) associated with alcohol exposure during the 3rd trimester equivalent (PD 4–9). In all of these studies, behavioral testing occurred after choline treatment had ceased, indicating that early postnatal choline supplementation leads to relatively long-lasting changes in behavior.

One important question is whether there are critical periods during which choline is most effective in mitigating ethanol's adverse effects on behavioral development. Indeed, the effects of choline supplementation among rats not exposed to alcohol have been shown to depend on the timing of administration. Choline supplementation during gestation produces more robust long-lasting improvements in visuospatial memory compared to postnatal choline supplementation (from PD 1–24), although the combination of pre- and postnatal choline supplementation is more effective than either period alone (Meck et al., 1989). Meck et al. (2008) further investigated critical periods of choline sensitivity among otherwise typically developing rats, administering choline for various periods throughout gestation and up to PD 75. They found that choline led to long-lasting improvements in spatial memory, as well as increased hippocampal dendritic spine densities, only when administered from gestational days (GD) 12–17 or PD 16–30. However, to date, no studies have examined the effects of varying developmental timing of choline administration among subjects exposed to alcohol during development.

In the present experiment, rat pups were exposed to ethanol during a period of brain development equivalent to the third trimester brain growth spurt. This period of brain development occurs postnatally in rats, from PD 4–9 (Dobbing and Sands, 1979). Subjects were subsequently treated with choline (choline chloride 100 mg/kg/day via s.c. injection) during one of two developmental phases following alcohol exposure: PD 11–20 (EtOH c/s) or PD 21–30 (EtOH s/c), or a combination of both periods (PD 11–30) (EtOH c/c). Ethanol-treated subjects that did not receive choline (EtOH s/s), sham intubated (SHAM s/s), and non-intubated controls (NC s/s) were included and injected with saline from PD 11–30. From PD 45–50, subjects were tested on a standard spatial learning version of the Morris maze, where the location of a hidden escape platform within a tank of opaque water remained constant throughout training for each subject and had to be found using extra-maze spatial cues. Twenty-four hours after training, subjects were tested on a probe trial to measure spatial memory. Finally, performance on a visible platform was measured to determine if other variables (i.e. motivation, motor, sensory) influenced spatial learning performance.

Section snippets

Body growth

Body weight data are shown in Fig. 1. Male and female data from the ethanol exposure period, PD 4–9, were analyzed separately (panels A and C). There were no significant differences in body weight among groups at PD 4. Beginning on PD 5, ethanol-exposed subjects lagged in growth, producing a significant main effect of treatment [F(5,105) = 11.5, p < 0.001], as well as an interaction of day by treatment [F(25,525) = 15.4, p < 0.001]. In addition, body weights of the male subjects increased in comparison

Discussion

This study is the first demonstration that as few as 10 days of postnatal choline supplementation can reduce the severity of developmental alcohol effects. Alcohol exposure during the third trimester equivalent produced significant deficits in both the acquisition and probe phases of the Morris water maze spatial learning task, consistent with previous reports (Berman and Hannigan, 2000, Goodlett and Johnson, 1997, Kelly et al., 1988). Although choline did not significantly reduce

Subjects

Subjects were 114 Sprague–Dawley rats (7–11 subjects/treatment /sex), offspring of animals bred at the San Diego State University Animal Care Facility. Multiparous dams were housed with males overnight, and the presence of a seminal plug in the morning marked gestational day (GD) 0. Pregnant females were then singly housed with food and water ad libitum on a 12:12-hr light/dark schedule with lights on at 0600. On postnatal day (PD) 1 (GD 23), litters were pseudorandomly culled to 10 pups (5

Acknowledgment

This project was funded by NIAAA grant AA012446.

References (73)

  • KellyS.J. et al.

    Effects of prenatal alcohol exposure on social behavior in humans and other species

    Neurotoxicol. Teratol.

    (2000)
  • KellyS.J. et al.

    Impaired spatial navigation in adult female but not adult male rats exposed to alcohol during the brain growth spurt

    Behav. Brain Res.

    (1988)
  • KlintsovaA.Y. et al.

    Therapeutic motor training ameliorates cerebellar effects of postnatal binge alcohol

    Neurotoxicol. Teratol.

    (2000)
  • KlintsovaA.Y. et al.

    Therapeutic effects of complex motor training on motor performance deficits induced by neonatal binge-like alcohol exposure in rats: II. A quantitative stereological study of synaptic plasticity in female rat cerebellum

    Brain Res.

    (2002)
  • KovachevaV.P. et al.

    Gestational choline deficiency causes global and Igf2 gene DNA hypermethylation by up-regulation of Dnmt1 expression

    J. Biol. Chem.

    (2007)
  • MatthewsD.A. et al.

    Development of cholinergic innervation in the hippocampal formation of the rat. I. Histochemical demonstration of acetylcholinesterase activity

    Dev. Biol.

    (1974)
  • McCannJ.C. et al.

    An overview of evidence for a causal relationship between dietary availability of choline during development and cognitive function in offspring

    Neurosci. Biobehav. Rev.

    (2006)
  • MeckW.H. et al.

    Metabolic imprinting of choline by its availability during gestation: implications for memory and attentional processing across the lifespan

    Neurosci. Biobehav. Rev.

    (2003)
  • MontoyaD.A. et al.

    Prenatal choline exposure alters hippocampal responsiveness to cholinergic stimulation in adulthood

    Brain Res. Dev. Brain Res.

    (2000)
  • NadlerJ.V. et al.

    Development of cholinergic innervation in the hippocampal formation of the rat. II. Quantitative changes in choline acetyltransferase and acetylcholinesterase activities

    Dev. Biol.

    (1974)
  • SpadoniA.D. et al.

    Neuroimaging and fetal alcohol spectrum disorders

    Neurosci. Biobehav. Rev.

    (2007)
  • TeesR.C.

    The influences of rearing environment and neonatal choline dietary supplementation on spatial learning and memory in adult rats

    Behav. Brain Res.

    (1999)
  • ThomasJ.D. et al.

    Perinatal choline supplementation attenuates behavioral alterations associated with neonatal alcohol exposure in rats

    Neurotoxicol. Teratol.

    (2004)
  • ThomasJ.D. et al.

    Neonatal choline supplementation ameliorates the effects of prenatal alcohol exposure on a discrimination learning task in rats

    Neurotoxicol. Teratol.

    (2000)
  • WilliamsC.L. et al.

    Hypertrophy of basal forebrain neurons and enhanced visuospatial memory in perinatally choline-supplemented rats

    Brain Res.

    (1998)
  • Autti-RamoI. et al.

    MRI findings in children with school problems who had been exposed prenatally to alcohol

    Dev. Med. Child. Neurol.

    (2002)
  • AznavourN. et al.

    Postnatal development of the cholinergic innervation in the dorsal hippocampus of rat: quantitative light and electron microscopic immunocytochemical study

    J. Comp. Neurol.

    (2005)
  • BermanR.F. et al.

    Effects of prenatal alcohol exposure on the hippocampus: spatial behavior, electrophysiology, and neuroanatomy

    Hippocampus

    (2000)
  • BooksteinF.L. et al.

    Many infants prenatally exposed to high levels of alcohol show one particular anomaly of the corpus callosum

    Alcohol Clin. Exp. Res.

    (2007)
  • CermakJ.M. et al.

    Prenatal availability of choline modifies development of the hippocampal cholinergic system

    Faseb J.

    (1998)
  • CermakJ.M. et al.

    Prenatal availability of choline alters the development of acetylcholinesterase in the rat hippocampus

    Dev. Neurosci.

    (1999)
  • ChenS.Y. et al.

    Peptide-mediated protection from ethanol-induced neural tube defects

    Dev. Neurosci.

    (2005)
  • ChristieB.R. et al.

    Voluntary exercise rescues deficits in spatial memory and long-term potentiation in prenatal ethanol-exposed male rats

    Eur. J. Neurosci.

    (2005)
  • ColesC.D. et al.

    A comparison of children affected by prenatal alcohol exposure and attention deficit, hyperactivity disorder

    Alcohol Clin. Exp. Res.

    (1997)
  • CoutcherJ.B. et al.

    Dietary choline supplementation increases the density of nicotine binding sites in rat brain

    J. Pharmacol. Exp. Ther.

    (1992)
  • IeraciA. et al.

    Nicotinamide protects against ethanol-induced apoptotic neurodegeneration in the developing mouse brain

    PLoS Med.

    (2006)
  • Cited by (0)

    View full text