DOI QR코드

DOI QR Code

Influence of Lead on Repetitive Behavior and Dopamine Metabolism in a Mouse Model of Iron Overload

  • Chang, JuOae (Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University) ;
  • Kueon, Chojin (Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University) ;
  • Kim, Jonghan (Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University)
  • Received : 2014.09.04
  • Accepted : 2014.10.06
  • Published : 2014.12.31

Abstract

Exposures to lead (Pb) are associated with neurological problems including psychiatric disorders and impaired learning and memory. Pb can be absorbed by iron transporters, which are up-regulated in hereditary hemochromatosis, an iron overload disorder in which increased iron deposition in various parenchymal organs promote metal-induced oxidative damage. While dysfunction in HFE (High Fe) gene is the major cause of hemochromatosis, the transport and toxicity of Pb in Hfe-related hemochromatosis are largely unknown. To elucidate the relationship between HFE gene dysfunction and Pb absorption, H67D knock-in Hfe-mutant and wild-type mice were given drinking water containing Pb 1.6 mg/ml ad libitum for 6 weeks and examined for behavioral phenotypes using the nestlet-shredding and marble-burying tests. Latency to nestlet-shredding in Pb-treated wild-type mice was prolonged compared with non-exposed wild-types (p < 0.001), whereas Pb exposure did not alter shredding latency in Hfe-mutant mice. In the marble-burying test, Hfe-mutant mice showed an increased number of marbles buried compared with wild-type mice (p = 0.002), indicating more repetitive behavior upon Hfe mutation. Importantly, Pb-exposed wild-type mice buried more marbles than non-exposed wild-types, whereas the number of marbles buried by Hfe-mutant mice did not change whether or not exposed to Pb. These results suggest that Hfe mutation could normalize Pb-induced behavioral alteration. To explore the mechanism of repetitive behavior caused by Pb, western blot analysis was conducted for proteins involved in brain dopamine metabolism. The levels of tyrosine hydroxylase and dopamine transporter increased upon Pb exposure in both genotypes, whereas Hfe-mutant mice displayed down-regulation of the dopamine transporter and dopamine D1 receptor with D2 receptor elevated. Taken together, our data support the idea that both Pb exposure and Hfe mutation increase repetitive behavior in mice and further suggest that these behavioral changes could be associated with altered dopaminergic neurotransmission, providing a therapeutic basis for psychiatric disorders caused by Pb toxicity.

Keywords

References

  1. Toscano, C.D. and Guilarte, T.R. (2005) Lead neurotoxicity: from exposure to molecular effects. Brain Res. Rev., 49, 529-554. https://doi.org/10.1016/j.brainresrev.2005.02.004
  2. Kim, S., Arora, M., Fernandez, C., Landero, J., Caruso, J. and Chen, A. (2013) Lead, mercury, and cadmium exposure and attention deficit hyperactivity disorder in children. Environ. Res., 126, 105-110. https://doi.org/10.1016/j.envres.2013.08.008
  3. Moreira, E.G., Vassilieff, I. and Vassilieff, V.S. (2001) Developmental lead exposure: behavioral alterations in the short and long term. Neurotoxicol. Teratol., 23, 489-495. https://doi.org/10.1016/S0892-0362(01)00159-3
  4. Levin, R., Grown, M.J., Kashtock, M.E., Jacobs, D.E., Whelan, E.A., Rodman, J., Schock, M.R., Padilla, A. and Sinks, T. (2008) Lead exposures in U.S. Children, 2008: implications for prevention. Environ. Health Perspect., 116, 1285-1293. https://doi.org/10.1289/ehp.11241
  5. Hu, H., Shine, J. and Wright, R.O. (2007) The challenge posed to children's health by mixtures of toxic waste: the Tar Creek superfund site as a case-study. Pediatr. Clin. North Am., 54, 155-175. https://doi.org/10.1016/j.pcl.2006.11.009
  6. Garrick, M.D., Dolan, K.G., Horbinski, C., Ghio, A.J., Higgins, D., Porubcin, M., Moore, E.G., Hainsworth, L.N., Umbreit, J.N., Conrad, M.E., Feng, L., Lis, A., Roth, J.A., Singleton, S. and Garrick, L.M. (2003) DMT1: a mammalian transporter for multiple metals. Biometals, 16, 41-54. https://doi.org/10.1023/A:1020702213099
  7. Hashmi, N.S., Kachru, D.N., Khandelwal, S. and Tandon, S.K. (1989) Interrelationship between iron deficiency and lead intoxication (Part 2). Biol. Trace Elem. Res., 22, 299-307. https://doi.org/10.1007/BF02916618
  8. Pietrangelo, A. (2004) Hereditary hemochromatosis-a new look at an old disease. N. Engl. J. Med., 350, 2383-2397. https://doi.org/10.1056/NEJMra031573
  9. Zhang, A., Park, S.K., Wright, R.O., Weisskopf, M.G., Mukherjee, B., Nie, H., Sparrow, D. and Hu, H. (2010) HFE H63D polymorphism as a modifier of the effect of cumulative lead exposure on pulse pressure: the Normative Aging Study. Environ. Health Perspect., 118, 1261-1266. https://doi.org/10.1289/ehp.1002251
  10. Lee, S.H., Kim, J.W., Shin, S.H., Kang, K.P., Choi, H.C., Park, K.U., Kim, H.Y., Kang, W. and Jeong, S.H. (2009) HFE gene mutations, serum ferritin level, transferrin saturation, and their clinical correlates in a Korean population. Dig. Dis. Sci., 54, 879-886. https://doi.org/10.1007/s10620-008-0432-6
  11. Nandar, W. and Connor, J.R. (2011) HFE gene variants affect iron in the brain. J. Nutr., 141, 729S-739S. https://doi.org/10.3945/jn.110.130351
  12. Gebril, O.H. and Meguid, N.A. (2011) HFE gene polymorphisms and the risk for autism in Egyptian children and impact on the effect of oxidative stress. Dis. Markers, 31, 289-294. https://doi.org/10.1155/2011/605620
  13. Liu, Y., Lee, S.Y., Neely, E., Nandar, W., Moyo, M., Simmons, Z. and Connor, J.R. (2011) Mutant HFE H63D protein is associated with prolonged endoplasmic reticulum stress and increased neuronal vulnerability. J. Biol. Chem., 286, 13161-13170. https://doi.org/10.1074/jbc.M110.170944
  14. Nandar, W., Neely, E.B., Unger, E. and Connor, J.R. (2013) A mutation in the HFE gene is associated with altered brain iron profiles and increased oxidative stress in mice. Biochim. Biophys. Acta, 1832, 729-741. https://doi.org/10.1016/j.bbadis.2013.02.009
  15. Berglund, M., Lindberg, A.L., Rahman, M., Yunus, M., Grander, M., Lonnerdal, B. and Vahter, M. (2011) Gender and age differences in mixed metal exposure and urinary excretion. Environ. Res., 111, 1271-1279. https://doi.org/10.1016/j.envres.2011.09.002
  16. Molina, R.M., Phattanarudee, S., Kim, J., Thompson, K., Wessling-Resnick, M., Maher, T.J. and Brain, J.D. (2011) Ingestion of Mn and Pb by rats during and after pregnancy alters iron metabolism and behavior in offspring. Neurotoxicology, 32, 413-422. https://doi.org/10.1016/j.neuro.2011.03.010
  17. Morgan, E.H. and Oates, P.S. (2002) Mechanisms and regulation of intestinal iron absorption. Blood Cells Mol. Dis., 29, 384-399. https://doi.org/10.1006/bcmd.2002.0578
  18. Joshi, J.G. and Zimmeman, A. (1988) Ferritin: an expanded role in metabolic regulation. Toxicology, 48, 21-29. https://doi.org/10.1016/0300-483X(88)90055-8
  19. Thomas, A., Burant, A., Bui, N., Graham, D., Yuva-Paylor, L.A. and Paylor, R. (2009) Marble burying reflects a repetitive and perseverative behavior more than novelty-induced anxiety. Psychopharmacology (Berl), 204, 361-373. https://doi.org/10.1007/s00213-009-1466-y
  20. Angoa-Perez, M., Kane, M.J., Briggs, D.I., Francescutti, D.M. and Kuhn, D.M. (2013) Marble burying and nestlet shredding as tests of repetitive, compulsive-like behaviors in mice. J. Visualized Exp., 24, 50978.
  21. Jirkof, P., Fleischmann, T., Cesarovic, N., Rettich, A., Vogel, J. and Arras, M. (2013) Assessment of postsurgical distress and pain in laboratory mice by nest complexity scoring. Lab. Anim., 47, 153-161. https://doi.org/10.1177/0023677213475603
  22. Silveman, J.L., Yang, M., Lord, C. and Crawley, J.N. (2010) Behavioural phenotyping assays for mouse models of autism. Nat. Rev. Neurosci., 11, 490-502. https://doi.org/10.1038/nrn2851
  23. Fortune, T. and Lurie, D.I. (2009) Chronic low-level lead exposure affects the monoaminergic system in the mouse superior olivary complex. J. Comp. Neurol., 513, 542-558. https://doi.org/10.1002/cne.21978
  24. Pokora, M.J., Richfield, E.K. and Cory-Slechta, D.A. (1996) Preferential vulnerability of nucleus accumbens dopamine binding sites to low-level lead exposure: time course of effects and interactions with chronic dopamine agonist treatments. J. Neurochem., 67, 1540-1550.
  25. Bianco, L.E., Wiesinger, J., Earley, C.J., Jones, B.C. and Beard, J.L. (2008) Iron deficiency alters dopamine uptake and response to L-DOPA injection in Sprague-Dawley rats. J. Neurochem., 106, 205-215. https://doi.org/10.1111/j.1471-4159.2008.05358.x
  26. Taylor, J.L., Rajbhandari, A.K., Berridge, K.C. and Aldridge, J.W. (2010) Dopamine receptor modulation of repetitive grooming actions in the rat: potential relevance for Tourette syndrome. Brain Res., 1322, 92-101. https://doi.org/10.1016/j.brainres.2010.01.052
  27. Kordas, K., Ettinger, A.S., Bellinger, D.C., Schnaas, L., Tellez Rojo, M.M., Hernandez-Avila, M., Hu, H. and Wright, R.O. (2011) A dopamine receptor (DRD2) but not dopamine transporter (DAT1) gene polymorphism is associated with neurocognitive development of Mexican preschool children with lead exposure. J. Pediatr., 159, 638-643. https://doi.org/10.1016/j.jpeds.2011.03.043
  28. Lakshmi Priya, M.D. and Geetha, A. (2011) Level of trace elements (copper, zinc, magnesium and selenium) and toxic elements (lead and mercury) in the hair and nail of children with autism. Biol. Trace Elem. Res., 142, 148-158. https://doi.org/10.1007/s12011-010-8766-2
  29. Latif, A., Heinz, P. and Cook, R. (2002) Iron deficiency in autism and Asperger syndrome. Autism, 6, 103-114. https://doi.org/10.1177/1362361302006001008

Cited by

  1. Loss of Hfe Function Reverses Impaired Recognition Memory Caused by Olfactory Manganese Exposure in Mice vol.31, pp.1, 2015, https://doi.org/10.5487/TR.2015.31.1.017
  2. Effect of Hfe Deficiency on Memory Capacity and Motor Coordination after Manganese Exposure by Drinking Water in Mice vol.31, pp.4, 2015, https://doi.org/10.5487/TR.2015.31.4.347
  3. Loss of divalent metal transporter 1 function promotes brain copper accumulation and increases impulsivity vol.138, pp.6, 2016, https://doi.org/10.1111/jnc.13717
  4. Blood lead concentrations and attention deficit hyperactivity disorder in Korean children: a hospital-based case control study vol.16, pp.1, 2016, https://doi.org/10.1186/s12887-016-0696-5
  5. Short-term manganese inhalation decreases brain dopamine transporter levels without disrupting motor skills in rats vol.41, pp.3, 2016, https://doi.org/10.2131/jts.41.391
  6. Role of fatty acid composites in the toxicity of titanium dioxide nanoparticles used in cosmetic products vol.41, pp.4, 2016, https://doi.org/10.2131/jts.41.533
  7. The sarin-like organophosphorus agent bis(isopropyl methyl)phosphonate induces ER stress in human astrocytoma cells vol.41, pp.5, 2016, https://doi.org/10.2131/jts.41.617
  8. Mutation in HFE gene decreases manganese accumulation and oxidative stress in the brain after olfactory manganese exposure vol.8, pp.6, 2016, https://doi.org/10.1039/C6MT00080K
  9. Iron overload exacerbates age-associated cardiac hypertrophy in a mouse model of hemochromatosis vol.7, pp.1, 2017, https://doi.org/10.1038/s41598-017-05810-2
  10. Lead Transiently Promotes Granulocyte-Macrophage Progenitor Differentiation and Subsequently Suppresses Common Myeloid Progenitor Differentiation pp.1096-0929, 2017, https://doi.org/10.1093/toxsci/kfx176
  11. Oral nucleic acid therapy using multicompartmental delivery systems pp.19395116, 2017, https://doi.org/10.1002/wnan.1478
  12. Evaluation of Whole Blood Trace Element Levels in Chinese Children with Autism Spectrum Disorder pp.1559-0720, 2019, https://doi.org/10.1007/s12011-018-1615-4
  13. Brain iron loading impairs DNA methylation and alters GABAergic function in mice vol.33, pp.2, 2019, https://doi.org/10.1096/fj.201801116RR