Skip to main content

Advertisement

Log in

Early and sustained exposure to high-sucrose diet triggers hippocampal ER stress in young rats

  • Original Article
  • Published:
Metabolic Brain Disease Aims and scope Submit manuscript

Abstract

Early-life environmental insults have been shown to promote long-term development of chronic non-communicable diseases, including metabolic disturbances and mental illnesses. As such, premature consumption of high-sugar foods has been associated to early onset of detrimental outcomes, whereas underlying mechanisms are still poorly understood. In the present study, we sought to investigate whether early and sustained exposure to high-sucrose diet promotes metabolic disturbances that ultimately might anticipate neurological injuries. At postnatal day 21, weaned male rats started to be fed a standard chow (10 % sucrose, CTR) or a high-sucrose diet (25 % sucrose, HSD) for 9 weeks prior to euthanasia at postnatal day 90. HSD did not alter weight gain and feed efficiency between groups, but increased visceral, non-visceral and brown adipose tissue accumulation. HSD rats demonstrated elevated blood glucose levels in both fasting and fed states, which were associated to impaired glucose tolerance. Peripheral insulin sensitivity did not change, whereas hepatic insulin resistance was supported by increased serum triglyceride levels, as well as higher TyG index values. Assessment of hippocampal gene expression showed endoplasmic reticulum (ER) stress pathways were activated in HSD rats, as compared to CTR. HSD rats had overexpression of unfolded protein response sensors, PERK and ATF6; ER chaperone, PDIA2 and apoptosis-related genes, CHOP and Caspase 3; but decreased expression of chaperone GRP78. Finally, HSD rats demonstrated impaired neuromuscular function and anxious behavior, but preserved cognitive parameters. In conclusion, our data indicate that early exposure to HSD promote metabolic disturbances, which disrupt hippocampus homeostasis and might precociously affect its neurobehavioral functions.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  • Alberti K et al. (2009) Harmonizing the metabolic syndrome a joint interim statement of the international diabetes federation task force on epidemiology and prevention; National Heart, Lung, and Blood Institute; American Heart Association; world heart federation. International Atherosclerosis Society; and International Association for the Study of Obesity Circulation 120:1640–1645

    CAS  PubMed  Google Scholar 

  • Albouy G et al. (2008) Both the hippocampus and striatum are involved in consolidation of motor sequence memory. Neuron 58:261–272. doi:10.1016/j.neuron.2008.02.008

    Article  CAS  PubMed  Google Scholar 

  • Arruda AP et al. (2011a) Low-grade hypothalamic inflammation leads to defective thermogenesis, insulin resistance, and impaired insulin secretion. Endocrinology 152:1314–1326. doi:10.1210/en.2010-0659

    Article  CAS  PubMed  Google Scholar 

  • Arruda AP, Milanski M, Velloso LA (2011b) Hypothalamic inflammation and thermogenesis: the brown adipose tissue connection. J Bioenerg Biomembr 43:53–58. doi:10.1007/s10863-011-9325-z

    Article  CAS  PubMed  Google Scholar 

  • Bast T, Wilson IA, Witter MP, Morris RG (2009) From rapid place learning to behavioral performance: a key role for the intermediate hippocampus. PLoS Biol 7:e1000089. doi:10.1371/journal.pbio.1000089

    Article  PubMed  PubMed Central  Google Scholar 

  • Bernardis L, Patterson B (1968) Correlation between'Lee index'and carcass fat content in weanling and adult female rats with hypothalamic lesions. J Endocrinol 40:527–528

    Article  CAS  PubMed  Google Scholar 

  • Bizeau ME, Pagliassotti MJ (2005) Hepatic adaptations to sucrose and fructose. Metabolism 54:1189–1201. doi:10.1016/j.metabol.2005.04.004

    Article  CAS  PubMed  Google Scholar 

  • Bray GA (1991) Obesity, a disorder of nutrient partitioning: the MONA LISA hypothesis. J Nutr 121:1146–1162

    CAS  PubMed  Google Scholar 

  • Bray GA (2010) Fructose: pure, white, and deadly? Fructose, by any other name, is a health hazard. J Diabetes Sci Technol 4:1003–1007

    Article  PubMed  PubMed Central  Google Scholar 

  • Bray GA, York DA (1998) The MONA LISA hypothesis in the time of leptin Recent Prog Horm Res 53:95–117; discussion 117-118

  • Bray GA, Nielsen SJ, Popkin BM (2004) Consumption of high-fructose corn syrup in beverages may play a role in the epidemic of obesity. Am J Clin Nutr 79:537–543

    CAS  PubMed  Google Scholar 

  • Calvo-Ochoa E, Arias C (2015) Cellular and metabolic alterations in the hippocampus caused by insulin signalling dysfunction and its association with cognitive impairment during aging and Alzheimer's disease: studies in animal models. Diabetes Metab Res Rev 31:1–13. doi:10.1002/dmrr.2531

    Article  CAS  PubMed  Google Scholar 

  • Calvo-Ochoa E, Hernandez-Ortega K, Ferrera P, Morimoto S, Arias C (2014) Short-term high-fat-and-fructose feeding produces insulin signaling alterations accompanied by neurite and synaptic reduction and astroglial activation in the rat hippocampus. J Cereb Blood Flow Metab 34:1001–1008. doi:10.1038/jcbfm.2014.48

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Cenquizca LA, Swanson LW (2007) Spatial organization of direct hippocampal field CA1 axonal projections to the rest of the cerebral cortex. Brain Res Rev 56:1–26

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Chan KA, Tsoulis MW, Sloboda DM (2015) Early-life nutritional effects on the female reproductive system. J Endocrinol 224:R45–R62. doi:10.1530/JOE-14-0469

    Article  CAS  PubMed  Google Scholar 

  • Christoffel KK, Wang X, Binns HJ (2012) Early origins of child obesity: bridging disciplines and phases of development – September 30–October 1, 2010, Int J Environ Res Public Health 9:1227–1262 doi:10.3390/ijerph9041227

  • Cnop M, Foufelle F, Velloso LA (2012) Endoplasmic reticulum stress, obesity and diabetes. Trends Mol Med 18:59–68. doi:10.1016/j.molmed.2011.07.010

    Article  CAS  PubMed  Google Scholar 

  • Cohen JC, Schall R (1988) Reassessing the effects of simple carbohydrates on the serum triglyceride responses to fat meals. Am J Clin Nutr 48:1031–1034

    CAS  PubMed  Google Scholar 

  • Cooper C (2013) David barker (1938-2013). Nature 502:304. doi:10.1038/502304a

    Article  PubMed  Google Scholar 

  • Davidson TL, Chan K, Jarrard LE, Kanoski SE, Clegg DJ, Benoit SC (2009) Contributions of the hippocampus and medial prefrontal cortex to energy and body weight regulation. Hippocampus 19:235–252. doi:10.1002/hipo.20499

    Article  PubMed  PubMed Central  Google Scholar 

  • de Lima DC, Silveira SA, Haibara AS, Coimbra CC (2008) The enhanced hyperglycemic response to hemorrhage hypotension in obese rats is related to an impaired baroreflex. Metab Brain Dis 23:361–373

    Article  CAS  PubMed  Google Scholar 

  • de Queiroz KB, Rodovalho GV, Guimarães JB, de Lima DC, Coimbra CC, Evangelista EA, Guerra-Sá R (2012) Endurance training blocks uncoupling protein 1 up-regulation in brown adipose tissue while increasing uncoupling protein 3 in the muscle tissue of rats fed with a high-sugar diet. Nutr Res 32:709–717

    Article  PubMed  Google Scholar 

  • Du T, Yuan G, Zhang M, Zhou X, Sun X, Yu X (2014) Clinical usefulness of lipid ratios, visceral adiposity indicators, and the triglycerides and glucose index as risk markers of insulin resistance. Cardiovasc Diabetol 13:146

    Article  PubMed  PubMed Central  Google Scholar 

  • Gibson S, Gunn P, Wittekind A, Cottrell R (2013) The effects of sucrose on metabolic health: a systematic review of human intervention studies in healthy adults. Crit Rev Food Sci Nutr 53:591–614

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Gluckman PD, Hanson MA, Beedle AS (2007) Non-genomic transgenerational inheritance of disease risk. BioEssays 29:145–154. doi:10.1002/bies.20522

    Article  CAS  PubMed  Google Scholar 

  • Gray JA, McNaughton N (1983) Comparison between the behavioural effects of septal and hippocampal lesions: a review. Neurosci Biobehav Rev 7:119–188

    Article  CAS  PubMed  Google Scholar 

  • Guerrero-Romero F et al. (2010) The product of triglycerides and glucose, a simple measure of insulin sensitivity Comparison with the euglycemic-hyperinsulinemic clamp. J Clin Endocrinol Metabol 95:3347–3351

    Article  CAS  Google Scholar 

  • Hall C, Ballachey EL (1932) A study of the rat's behavior in a field. A contribution to method in comparative psychology. University of California Publications in Psychology, Berkeley, University of California Press

    Google Scholar 

  • Hetz C (2012) The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol 13:89–102

    CAS  PubMed  Google Scholar 

  • Hetz Mollereau B (2014) Disturbance of endoplasmic reticulum proteostasis in neurodegenerative diseases. Nat Rev Neurosci 15:233–249. doi:10.1038/nrn3689

    Article  PubMed  Google Scholar 

  • Inadera H (2013) Developmental origins of obesity and type 2 diabetes: molecular aspects and role of chemicals. Environ Health Prev Med 18:185–197. doi:10.1007/s12199-013-0328-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Jocken JW, Blaak EE (2008) Catecholamine-induced lipolysis in adipose tissue and skeletal muscle in obesity. Physiol Behav 94:219–230

    Article  CAS  PubMed  Google Scholar 

  • Kanarek RB, Orthen-Gambill N (1982) ) Differential effects of sucrose, fructose and glucose on carbohydrate-induced obesity in rats. J Nutr 112:1546–1554

    CAS  PubMed  Google Scholar 

  • Kanazawa M et al. (2003) Effects of a high-sucrose diet on body weight, plasma triglycerides, and stress tolerance. Nutr Rev 61:S27–S33

    Article  PubMed  Google Scholar 

  • Kanoski SE, Davidson TL (2011) Western diet consumption and cognitive impairment: links to hippocampal dysfunction and obesity. Physiol Behav 103:59–68. doi:10.1016/j.physbeh.2010.12.003

    Article  CAS  PubMed  Google Scholar 

  • Kuwabara T et al. (2011) Insulin biosynthesis in neuronal progenitors derived from adult hippocampus and the olfactory bulb. EMBO Mol Med 3:742–754. doi:10.1002/emmm.201100177

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Lin JH, Li H, Zhang Y, Ron D, Walter P (2009) Divergent effects of PERK and IRE1 signaling on cell viability. PLoS One 4:e4170. doi:10.1371/journal.pone.0004170

    Article  PubMed  PubMed Central  Google Scholar 

  • Mainardi M, Fusco S, Grassi C (2015) Modulation of hippocampal neural plasticity by glucose-related signaling. Neural Plast 2015:657928. doi:10.1155/2015/657928

    PubMed  PubMed Central  Google Scholar 

  • Morris R (1984) Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods 11:47–60

    Article  CAS  PubMed  Google Scholar 

  • Ozcan L, Tabas I (2012) Role of endoplasmic reticulum stress in metabolic disease and other disorders. Annu Rev Med 63:317

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Pawlak DB, Kushner JA, Ludwig DS (2004) Effects of dietary glycaemic index on adiposity, glucose homoeostasis, and plasma lipids in animals. Lancet 364:778–785

    Article  CAS  PubMed  Google Scholar 

  • Pyndt Jorgensen B et al. (2014) A possible link between food and mood: dietary impact on gut microbiota and behavior in BALB/c mice. PLoS One 9:e103398. doi:10.1371/journal.pone.0103398

    Article  PubMed  PubMed Central  Google Scholar 

  • Rippe JM, Angelopoulos TJ (2013) Sucrose, high-fructose corn syrup, and fructose, their metabolism and potential health effects: what do we really know? Adv Nutr 4:236–245. doi:10.3945/an.112.002824

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Rizkalla SW, Luo J, Guilhem I, Boillot J, Bruzzo F, Chevalier A, Slama G (1992) Comparative effects of 6 week fructose, dextrose and starch feeding on fat-cell lipolysis in normal rats: effects of isoproterenol, theophylline and insulin. Mol Cell Biochem 109:127–132

    Article  CAS  PubMed  Google Scholar 

  • Roussel BD, Kruppa AJ, Miranda E, Crowther DC, Lomas DA, Marciniak SJ (2013) Endoplasmic reticulum dysfunction in neurological disease. Lancet Neurol 12:105–118. doi:10.1016/S1474-4422(12)70238-7

    Article  CAS  PubMed  Google Scholar 

  • Santos CX, Tanaka LY, Wosniak J Jr, Laurindo FR (2009) Mechanisms and implications of reactive oxygen species generation during the unfolded protein response: roles of endoplasmic reticulum oxidoreductases, mitochondrial electron transport, and NADPH oxidase. Antioxid Redox Signal 11:2409–2427

    Article  CAS  PubMed  Google Scholar 

  • Simental-Mendia LE, Rodriguez-Moran M, Guerrero-Romero F (2008) The product of fasting glucose and triglycerides as surrogate for identifying insulin resistance in apparently healthy subjects. Metab Syndr Relat Disord 6:299–304. doi:10.1089/met.2008.0034

    Article  CAS  PubMed  Google Scholar 

  • Srodulski S et al. (2014) Neuroinflammation and neurologic deficits in diabetes linked to brain accumulation of amylin. Mol Neurodegener 9:30

    Article  PubMed  PubMed Central  Google Scholar 

  • Stephens DN (1980) Does the lee obesity index measure general obesity? Physiol Behav 25:313–315

    Article  CAS  PubMed  Google Scholar 

  • Tappy L, Le KA, Tran C, Paquot N (2010) Fructose and metabolic diseases: new findings, new questions. Nutrition 26:1044–1049. doi:10.1016/j.nut.2010.02.014

    Article  CAS  PubMed  Google Scholar 

  • Thresher JS, Podolin DA, Wei Y, Mazzeo RS, Pagliassotti MJ (2000) Comparison of the effects of sucrose and fructose on insulin action and glucose tolerance. Am J Phys Regul Integr Comp Phys 279:R1334–R1340

    CAS  Google Scholar 

  • Toida S, Takahashi M, Shimizu H, Sato N, Shimomura Y, Kobayashi I (1996) Effect of high sucrose feeding on fat accumulation in the male. Wistar Rat Obesity Research 4:561–568

    Article  CAS  PubMed  Google Scholar 

  • van der Harg JM et al. (2014) The unfolded protein response mediates reversible tau phosphorylation induced by metabolic stress. Cell Death Dis 5:e1393. doi:10.1038/cddis.2014.354

    Article  PubMed  PubMed Central  Google Scholar 

  • Vaughan M (1962) The production and release of glycerol by adipose tissue incubated in vitro. J Biol Chem 237:3354–3358

    CAS  PubMed  Google Scholar 

  • Velloso LA, Schwartz MW (2011) Altered hypothalamic function in diet-induced obesity. Int J Obes 35:1455–1465. doi:10.1038/ijo.2011.56

    Article  CAS  Google Scholar 

  • Yudkin J (1972) Sugar and disease. Nature 239:197–199

    Article  CAS  PubMed  Google Scholar 

  • Yudkin J (1983) Sugar and obesity. Lancet 2:794

    Article  CAS  PubMed  Google Scholar 

  • Zhang WN, Bast T, Xu Y, Feldon J (2014) Temporary inhibition of dorsal or ventral hippocampus by muscimol: distinct effects on measures of innate anxiety on the elevated plus maze, but similar disruption of contextual fear conditioning. Behav Brain Res 262:47–56. doi:10.1016/j.bbr.2013.10.044

    Article  CAS  PubMed  Google Scholar 

  • Zhang Y, Fischer KE, Soto V, Liu Y, Sosnowska D, Richardson A, Salmon AB (2015) Obesity-induced oxidative stress, accelerated functional decline with age and increased mortality in mice. Arch Biochem Biophys 576:39–48. doi:10.1016/j.abb.2014.12.018

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhao Y, Yan Y, Zhao Z, Li S, Yin J (2015) The dynamic changes of endoplasmic reticulum stress pathway markers GRP78 and CHOP in the hippocampus of diabetic mice. Brain Res Bull 111:27–35. doi:10.1016/j.brainresbull.2014.12.006

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

Authors are thankful to the LeFisio’s staff for all the technical support during experimental procedures, especially to Caroline Vale, Danylo Noleto and Pâmela Santos. This work was funded by Fundação de Amparo à Pesquisa e ao Desenvolvimento Científico e Tecnológico do Maranhão – FAPEMA through the grants PAEDT-00380/14, UNIVERSAL-00523/14, and UNIVERSAL-00792/14. TMM received fellowship from Conselho Nacional de Desenvolvimento Científico e Tecnológico – CNPq.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Antonio Marcus de Andrade Paes.

Ethics declarations

Conflict of interest

Authors declare the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pinto, B.A.S., Melo, T.M., Flister, K.F.T. et al. Early and sustained exposure to high-sucrose diet triggers hippocampal ER stress in young rats. Metab Brain Dis 31, 917–927 (2016). https://doi.org/10.1007/s11011-016-9830-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11011-016-9830-1

Keywords

Navigation