Behavioral and neurotoxicological effects of subchronic manganese exposure in rats

https://doi.org/10.1016/j.etap.2004.12.046Get rights and content

Abstract

In male Wistar rats, behavioral and electrophysiological investigations, and blood and brain manganese level determinations, were performed; during 10 weeks treatment with low-dose manganese chloride and a 12 weeks post-treatment period. Three groups of 16 animals each received daily doses of 14.84 and 59.36 mg/kg b.w. MnCl2 (control: distilled water) via gavage. During treatment period, Mn accumulation was seen first in the blood, then in the brain samples of the high-dose animals. Short- and long-term spatial memory performance of the treated animals decreased, spontaneous open field activity (OF) was reduced. The number of acoustic startle responses (ASR), and the pre-pulse inhibition (PPI) of these, diminished. In the cortical and hippocampal spontaneous activity, power spectrum was shifted to higher frequencies. The latency of the sensory evoked potentials increased, and their duration, decreased. By the end of the post-treatment period, Mn levels returned to the control in all samples. The impairment of long-term spatial memory remained, as did the number of acoustic startle responses. Pre-pulse inhibition, however, returned to the pre-treatment levels. The changes of the open field activity disappeared but a residual effect could be revealed by administration of d-amphetamine. The electrophysiological effects were partially reversed.

By applying a complex set of methods, it was possible to obtain new data for a better-based relationship between the known effects of Mn at neuronal level and the behavioral and electrophysiological outcomes of Mn exposure.

Introduction

Manganese (Mn) is, in small amounts, an essential micronutrient (WHO, 1981), being cofactor in metallo-enzymes. In prolonged exposure to higher doses it is, however, a potential environmental neurotoxicant. In the general population, excess dietary intake (beyond the estimated safe and adequate daily amount) is more typical, as in case of babies fed by cow milk or soybean-based formulas (Marlowe and Bliss, 1993), or in the population having environmental exposure via the drinking water. In Greece, 50 mg/l Mn in the drinking water was associated with neurological effects (Kondakis et al., 1989). Mn as man-made environmental contaminant originates from Mn-containing waste (e.g. batteries), methylcyclopentadienyl manganese tricarbonyl (MMT) used as anti-knock petrol additive, and organo-Mn agricultural fungicides (ATSDR, 2000).

Following ingestion, inorganic Mn is absorbed in the intestine mostly in trivalent form (Cotzias et al., 1971) in a homeostatic self-limiting process. The main sites of deposition are the mitochondria-rich tissues, e.g. liver, pancreas, pituitary gland and the muscles (Barceloux, 1999). Brain is among the primary target organs in chronic Mn exposure (Roels et al., 1987), and the turnover of Mn there is much slower than in other parts of the body (Feldman, 1992).

In subchronic dosage, Mn appears in the cerebral and cerebellar cortex (Dorman et al., 2000). In case of chronic intake, Mn is found in motor control centers such as the basal ganglia, particularly the globus pallidus, striatum and substantia nigra where it causes degeneration (Yamada et al., 1986). Mn accumulation was also described in the pons and medulla (Chan et al., 1992), and in the hippocampus (Takeda et al., 1998).

The high sensitivity of different behavior types and the integration of different (motor, sensory, attentional, motivational) behavioral functions is especially important in assessment neurotoxic risk due, e.g. to Mn. Excess Mn was shown to cause several neurotoxic effects such as childhood hyperactivity disorder (Barlow, 1983), manganese psychosis, extrapyramidal dysfunction (Quaghebeur et al., 1996), motor deficit (above 7.5 μg/l blood Mn; Mergler et al., 1999); as well as altered childhood psychomotor development (Takser et al., 2003) and impairments of several memory process (list learning, visual recognition, digit span). Lucchini et al. (1999) found irritability and motor functional damage in exposed workers having ca. 10 μg/l blood Mn level. A child developed severe epilepsy following exposure to welding fumes resulting in 15–20 μg/l blood Mn (Hernandez et al., 2003).

Similar deficits were also induced by Mn in animals. Öner and Sentürk (1992) saw impaired T-maze learning in rats receiving 375 μg/kg b.w. Mn for 30 days. Ingersoll et al. (1995) elicited spontaneous hypoactivity by intrathecal administration of MnCl2. Dorman et al. (2000) described altered acoustic startle responses (ASR) in neonatal rats receiving 25 and 50 mg/kg b.w. MnCl2.

Significant relationship between neurotoxic alterations, neurotransmitter or modulator metabolism (first of all catecholamines), and Mn exposure have been described (Neff et al., 1969). Mn, in case of prolonged low level exposure, accumulates in the nigrostrial dopaminergic pathway (Subhash and Padmashree, 1991). The resulting dopaminergic–glutamatergic interactions are involved in the Mn effects on extrapyramidal motor functions and, indirectly, in the sensorimotor integration (Calabresi et al., 1997).

Direct toxicity of Mn to dopaminergic neurons was described by Parenti et al. (1987). The involvement of other transmitters can result from dopaminergic control upon these (Takeda et al., 2002) or by direct effect of Mn. The latter is know to exist in the GABAergic (Trepper et al., 1998) and glutamatergic system. Reduced glial glutamate uptake (Aschner et al., 1999) leads, as a direct result of impaired astrocytic-neuronal interactions (Hazell, 2002), to increased extracellular glutamate concentration, the excitotoxic effect of which could play a key role in manganese-induced neuronal cell death. Marked reactive astrocytosis, with significant hypertrophy of glial fibrillary acid protein-immunoreactive—GFAP-IR—astrocytes, was seen on Mn exposure in the globus pallidus (Baeck et al., 2003). In the hippocampal dentate gyrus, Mn exposure caused a decrease of GFAP-IR area in juvenile, but an increase in adult, rats (Pappas et al., 1997). This, together with the numerous effects of Mn exposure on the behavior, raised the possibility that these functional and morphological changes induced by Mn develop in parallel and are in detectable causal relationship.

There has been ample literature on the negative effects of MnCl2 on behavioral outcomes in rats. Several mechanisms have been purposed, such as influence on transmitter systems (Tran et al., 2002, Gwiazda et al., 2002), or directly on structural elements like hippocampal astrocytes (Aschner, 1996) and the midbrain and basal ganglia (Erikson and Aschner, 2002). The dentate gyrus (primarily the granule cells) plays an important role in the acquisition of new information (Ogura et al., 2002) and is possibly neural substrate of spatial reference and working memory, and of synaptic plasticity (Ikegaya et al., 1995). Agents acting on the dentate gyrus possibly affect learning and memory performance, locomotor activity, radial maze performance and spontaneous motor activity, and psychomotor performance.

In the works cited above, however, behavioral effects were not supported by histochemical or electrophysiological findings, and no follow-up in the after-exposure period was included. The electrophysiological effect of Mn exposure is in itself an open question: some authors found disturbances of spontaneous or stimulus-evoked cortical activity in workers exposed to comparable airborne Mn levels (Sinczuk-Walczak et al., 2001) while others did not (Deschamps et al., 2001).

Hence, our experiment involved a complex behavioral test battery (8-arm radial maze, open field activity—OF, acoustic startle response—ASR, and pre-pulse inhibition—PPI). Our behavioral investigations were supplemented by cortical electrophysiology (electrocorticogram—ECoG, evoked potentials—EP), immunhistochemistry (GFAP in various parts of the hippocampus), and by Mn level determination in blood, cortex and hippocampus. All investigations were performed both during and after the period of Mn administration. It was also attempted to prove the involvement of the dopamine (DA) system by applying a dopaminergic agonist in the elimination period.

Section snippets

Animals, housing, treatment

Young adult male Wistar rats (220–250 g body weight at start), obtained at the University's Breeding Center, were used. The animals were housed under controlled conditions of temperature (22–24 °C) and photoperiod (12-h light:12-h dark cycle with light starting at 06:00 h), with free access to drinking water. Three weeks before starting the treatment, the animals (up to 4 rats per cage) were put in cages of 28 cm × 40 cm × 20 cm. The memory test used required that during the 10 weeks of treatment the

Manganese levels

Levels of Mn in various tissues, at the end of the 5th and 10th week of MnCl2 application and the 12th post-treatment week, are given in Table 2. Compared to the 0th week (pre-administration) value of 0.0181 ± 0.003 μg/g, blood Mn levels increased in a dose- and time-dependent way. In the high dose group, the increase was significant both in the 5th treatment week (F2,6 = 12.16; high dose versus control, p < 0.01; high versus low dose, p < 0.01) and in the 10th treatment week (F2,12 = 9.51; high dose

Discussion

Subchronic administration of MnCl2 to the rats in this study resulted in marked internal exposure (shown by the Mn levels in the cortex and hippocampus; Table 2). Mn levels in the control rats’ brains, originating from the background Mn present in standard food and drinking water, were similar to that described earlier by Rehnberg et al. (1982).

From the gastrointestinal tract, absorption of ionic Mn is low (<10%) and self-limited in the rats (Davis et al., 1992). Once absorbed, Mn is bound to

Conclusion

The main points of conclusion drawn from the above results are as given below.

Oral MnCl2 treatment, 14.84 and 59.36 mg/kg b.w., for 10 weeks brought about, primarily in the high-dose group, an increase in first the blood, then cortex and hippocampus Mn levels.

During the treatment period, altered PPI and cortical spontaneous activity in the treated rats suggested the involvement of brainstem cholinergic systems (pre-pulse inhibition pathway and ascending cortical activation) due to Mn-induced

References (62)

  • B.A. Pappas et al.

    Perinatal manganese exposure: behavioral, neurochemical and histopathological effects in rat

    Neurotox. Teratol.

    (1997)
  • M.N. Subhash et al.

    Effect of manganese on biogenic amine metabolism in regions of the rat brain

    Food Chem. Toxicol.

    (1991)
  • A. Takeda et al.

    Manganese concentration in rat brain: manganese transport from the peripheral tissues

    Neurosci. Lett.

    (1998)
  • A. Takeda et al.

    Manganese influences the levels of neurotransmitters in synapses in rat brain

    Neuroscience

    (2002)
  • L. Takser et al.

    Manganese, monoamine metabolite levels at birth, and childhood psychomotor development

    Neurotoxicology

    (2003)
  • M. Tomas-Camardiel et al.

    Differential regulation of glutamic acid decarboxylase mRNA and tyrosine hydroxylase mRNA expression in the aged manganese-treated rats

    Mol. Brain Res.

    (2002)
  • T.T. Tran et al.

    Effects of neonatal dietary manganese exposure on brain dopamine levels and neurocognitive functions

    Neurotoxicology

    (2002)
  • M. Aschner et al.

    Manganese uptake and distribution in the central nervous system (CNS)

    Neurotoxicology

    (1999)
  • M. Aschner

    The functional significance of brain metallothioneins

    FASEB J.

    (1996)
  • ATSDR, 2000. Toxicological Profile for Manganese. US Department of Health and Human Services, Atlanta, GA, USA, pp....
  • S.Y. Baeck et al.

    Effect of manganese exposure on MPTP neurotoxicities

    Neurotoxicology

    (2003)
  • D.G. Barceloux

    Manganese

    Clin. Toxicol.

    (1999)
  • W.W. Beatty et al.

    Spatial memory in rats: time course of working memory and effect of anesthetics

    Behav. Neural Biol.

    (1982)
  • W.C. Bowman et al.

    Textbook of Pharmacology

    (1980)
  • P. Calabresi et al.

    The neostriatum beyond the motor function: experimental and clinical evidence

    Neuroscience

    (1997)
  • A.W.K. Chan et al.

    Changes in brain regional manganese and magnesium levels during postnatal development: modulations by chronic manganese administration

    Metab. Brain Dis.

    (1992)
  • P.B. Clarkson et al.

    Anatomical analysis of the involvement of mesolimbocortical dopamine in the locomotor stimulant actions of d-amphetamine and apomorphine

    Psychopharmacology

    (1988)
  • G.C. Cotzias et al.

    Chronic manganese poisoning: clearance of tissue manganese concentrations with persistence of the neurological picture

    Neurology

    (1968)
  • G.C. Cotzias et al.

    Metabolic modification of Parkinson's disease and of chronic manganese poisoning

    Annu. Rev. Med.

    (1971)
  • F.J. Deschamps et al.

    Neurological effects in workers exposed to manganese

    J. Occup. Environ. Med.

    (2001)
  • D.C. Dorman et al.

    Neurotoxicity of manganese chloride in neonatal and adult CD rats following subchronic (21-day) high-dose oral exposure

    J. Appl. Toxicol.

    (2000)
  • Cited by (32)

    • YAC128 mouse model of Huntington disease is protected against subtle chronic manganese (Mn)-induced behavioral and neuropathological changes

      2021, NeuroToxicology
      Citation Excerpt :

      In Study 1, WT mice exposed to the highest Mn dose [50 mg/kg MnCl2 4(H2O)] exhibited behavioral changes by spending more time in the open zone of the EZM and were overall less active in the open field but neither behavior was affected in YAC128 mice. Decreased locomotor activity in WT was an expected outcome of Mn-induced toxicity (Bouabid et al., 2014; Cordova et al., 2012; Vezér et al., 2005; Asser et al., 2019) though transient increases in locomotor activity have also been documented following Mn exposure (Nachtman et al., 1986; Avila-Costa et al., 2018). Reported effects of Mn on exploratory behavior or time spent in the open zone of the EZM are inconsistent (Moreno et al., 2009; Ye and Kim, 2015).

    • Could male reproductive system be the main target of subchronic exposure to manganese in adult animals?

      2018, Toxicology
      Citation Excerpt :

      Postnatal exposures to Mn have already been reported to exert no effects on anxiety behaviors (Pappas et al., 1997, Kern et al., 2010) while prenatal exposures exert great adverse effects (Molina et al., 2011; Lazrishvili et al., 2011). The absence of differences in the Pre-Pulse Inhibition test (PPI) among experimental groups are evidence of preservation of a pre-attention filtering system protecting the brain from sensory overload (Geyer et al., 2001) while the impaired locomotor activity observed in the current study had already been described by other authors using similar doses of Mn (e.g., Amos-Kroohs et al., 2017; Ávila et al., 2010; Santos et al., 2012; Andrade et al., 2013; Vezér et al., 2005). Direct comparisons with previous studies using behavioral tests are difficult, since variations in the experimental model often substantially alter the pattern of behavioral test results.

    • Urinary delta-ALA: A potential biomarker of exposure and neurotoxic effect in rats co-treated with a mixture of lead, arsenic and manganese

      2013, NeuroToxicology
      Citation Excerpt :

      Treatments were carried out according to the experimental design shown in Table 1. The administrated doses were chosen based on previous reports, showing behavioral alterations in rodents (Reddy et al., 2003; Rodríguez et al., 2010; Vezér et al., 2005; Marreilha dos Santos et al., 2011). Behavior assays and collection of 24-h urine in metabolic cages were performed prior (pre-dosing – PD) and at the end of the experimental period.

    View all citing articles on Scopus
    View full text