Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2008 Dec 1.
Published in final edited form as: Curr Opin Pharmacol. 2007 Nov 26;7(6):613–616. doi: 10.1016/j.coph.2007.10.008

A Potential Role for the Hippocampus in Energy Intake and Body Weight Regulation

T L Davidson 1, Scott E Kanoski 1, Lindsey A Schier 1, Deborah J Clegg 2, Stephen C Benoit 2
PMCID: PMC2223183  NIHMSID: NIHMS36003  PMID: 18032108

Abstract

Recent research and theory point to the possibility that hippocampal-dependent learning and memory mechanisms translate neurohormonal signals of energy balance into adaptive behavioral outcomes involved with the inhibition of food intake. The present paper summarizes these findings and ideas and considers the hypothesis that excessive caloric intake and obesity may be produced by dietary and other factors that are known to alter hippocampal functioning.

Keywords: energy homeostasis, memory, associative learning, obesity, rat


Much research has been devoted to identifying and understanding the role of inhibitory gut–brain signals in the control of energy intake and body weight [1]. A widely-held view is that the arrival of nutrients in the gut gives rise to relatively short-term hormonal (e.g., cholecystokinin (CCK)) meal-termination or “satiety” signals [2]. The effectiveness of these signals is thought to be modulated by circulating adiposity hormones (e.g., leptin and insulin) which provide information about the longer-term, as opposed to meal-related, status of bodily energy stores [3]. In addition, ghrelin has been identified as a gastric peptide that functions as a physiological meal initiation or “hunger” cue [4] that is elicited not only as a result of a change in an animal’s nutrient status but also as a learned anticipatory response to environmental cues associated with food [5]. Peripherally administered CCK and leptin appear to have interoceptive sensory consequences similar to those produced by a low level (e.g., 1 hr) of food deprivation [6], whereas the cue properties of peripherally or centrally administered ghrelin are similar to higher (e.g., 23-hr) levels of food deprivation [7,8]. All of these signals are transmitted to the brain where they are thought to be detected primarily by hypothalamic and hindbrain nuclei [2, 9].

While the identification of physiological meal-related and adiposity signals has contributed much to our understanding of the control of food intake and body weight regulation, relatively little is known about how the information provided by these cues is translated by the brain into adaptive behavioral outcomes. Although the hypothalamus and hindbrain have been identified as brain substrates that are involved with the detection of satiety, adiposity, and hunger cues, the ultimate decision to eat or to refrain from eating may depend on the processing of these signals at other brain sites involved with the higher-order control of behavior.

The hippocampus, a medial temporal lobe structure long regarded as an important substrate for learning and memory [10], has received increasing attention for its potential role in energy regulation. While some have suggested that hippocampal participation in energy homeostasis might not rely exclusively on learning and memory [11, 12], findings and theoretical developments encourage the hypothesis that hippocampal-dependent learning and memory mechanisms might contribute directly to the higher-order control of food intake. The purpose of this paper is to summarize and integrate some of these findings and ideas.

What does the hippocampus do?

Historically, [10] the hippocampus has been identified most closely with (a) encoding and retrieval of spatial relations among objects in the environment (i.e., spatial memory]) and (b) the formation and recall of memories about events and facts (i.e., declarative memory). While these conceptualizations are based primarily on studies of human amnesia, other more recent views of hippocampal function are derived from modern associative learning theories. Morris [13] noted that several of these newer accounts converge on the idea that the hippocampus is needed to resolve “predictable ambiguities” that exist when a single stimulus consistently signals different outcomes dependent on the presence or absence of other cues.

Negative occasion setting is a hippocampal-dependent process involved with learning to resolve predictable ambiguities. Rats with hippocampal lesions exhibit impaired negative occasion setting when they readily learn to respond to a conditioned stimulus (CS) that is consistently followed by delivery of a food pellet unconditioned stimulus (US) but are impaired at learning that a different cue (i.e., a negative occasion setter) signals when the CS will not be followed by the US. In this problem, although the ambiguity involved with predicting when the CS will be followed by reinforcement or nonreinforcement is resolved by the presentation of the negative occasion setting cue, rats with hippocampal lesions tend to respond to the CS as much on nonreinforced trials when the negative occasion setter is present as on reinforced trials when the CS occurs alone [14].

Removing the hippocampus also interferes with the ability of rats to use context cues (e.g., background, apparatus, temporal cues) to signal that a previously trained CS will no longer be followed by its unconditioned stimulus (US). According to Holland and Bouton [15], negative occasion setting by context cues may be a specialized function of the hippocampus. A recent imaging study [16] supports a similar conclusion. Human subjects were trained in one context with a visual CS that signaled delivery of a mild shock. The CS was subsequently extinguished in a different context. Functional magnetic resonance imagery (fMRI) showed that after extinction, the ventromedial prefrontal cortex (VMPFC) and the hippocampus were activated when the CS was presented in the extinction context, but not when it occurred in the training context. Consistent with a negative occasion setting interpretation, this finding suggests that retrieval of the memory depends on hippocampal-dependent gating of CS outputs to the VMPFC by the extinction context.

To eat or not to eat-- A case of predictable ambiguity?

When food and stimuli associated with food are encountered, these cues may evoke vigorous appetitive and consummatory responding on some occasions and little or no responding at other times. A common interpretation of this pattern of behavior is that animals engage in appetitive and eating behavior until they become satiated and then refrain from making these responses until satiety wanes [9]. How does satiety inhibit, and the absence of satiety promote, appetitive responding? The answer to this question may depend on an animal’s ability to resolve a predictable ambiguity by learning that satiety signals predict when food cues will not be followed by an appetitive postingestive US. In other words, just as experimentally programmed negative occasion setters resolve ambiguity by predicting when a CS will not be followed by its US, interoceptive satiety signals may resolve ambiguity by predicting when food cues will not be followed by appetitive postingestive outcomes [17].

The ability of regulatory hormones to modulate the strength of appetitive behavior may also depend on their effects on hippocampal-dependent learning and memory processes. The hippocampus is densely populated with both leptin and insulin receptors [12] and administration of each of these peptides has been shown to enhance both hippocampal-dependent spatial memory and hippocampal long-term potentiation (LTP) [18-20], a reported cellular basis for learning and memory [21]. Furthermore, mutant rats lacking CCK receptors not only become obese but also exhibit impaired hippocampal-dependent learning [22, 23]. Recent neuroanatomical findings also directly link the hippocampal CA1 cell field to hypothalamic nuclei and other brain circuits thought to underlie energy regulation [24]. Other data point more directly to the hippocampus as a processor of satiety information. Using fMRI, Wang et al [25] reported that, in obese people, the hippocampus is the site of greatest activation following gastric stimulation known to have effects on intake, stomach distention, hormonal and vagal activity similar to those produced by eating a large meal. In addition, fMRI showed that consuming a liquid meal to satiation decreased hippocampal blood flow for people who were obese or were formerly obese, but not for people who had never been obese ([26], also see [27]). In sum, these results indicate that (a) the hippocampus is sensitive to satiety signals; (b) at least some these signals induce changes in hippocampal activity that are thought to facilitate learning and memory; (c) the hippocampus is part of a neural circuit whereby the information provided by satiety signals could be transmitted from the gut to the hippocampus and from the hippocampus to forebrain circuits involved with energy regulation; (d) sensitivity of the hippocampus to these signals may be altered in people who have a history of energy dysregulation.

There is also evidence that the inhibitory control of food intake and appetitive behavior depends on the structural integrity of the hippocampus. For example, after eating a full meal densely amnesic human patients with hippocampal damage will eat a full second meal that is offered only minutes later [28, 29]. Higgs [30] demonstrated that for neurologically intact humans, memories of a prior meal help to inhibit subsequent intake. Densely amnesic patients may be less able to inhibit intake because their access to these memories is very limited. The results also suggest that hippocampal damage might interfere with satiety signaling by both interoceptive and exteroceptive cues.

Food sated rats with highly selective neurotoxic lesions confined to the hippocampus show increased appetitive behavior (e.g., food cup approach, bar pressing) relative to intact controls [31-33] and are impaired in using interoceptive cues arising from low (e.g., 1-hr) and high (e.g., 23-hr) levels of food deprivation as discriminative stimuli ([32,34] also see [35]). Consistent with a role for the hippocampus in negative occasion setting, in these latter problems, lesioned rats were impaired at using deprivation state cues to inhibit their behavior on nonreinforced trials. Furthermore, when intake suppression during recovery from surgery is accounted for, hippocampal-lesioned rats also show increased food intake and body weight gain [17]. These results suggest that the behavioral inhibition by energy state signals depends on the hippocampus.

Obesity—a hippocampal-dependent phenomenon?

Recent findings indicate dietary factors that promote excessive food intake and weight gain can also interfere with hippocampal functioning. For example, epidemiological data associate intake of diets high in saturated fat with weight gain [36][45] and memory deficits [37]. It may be that cognitive deficits are secondary to effects of high-fat diets on the development of insulin resistance. Rats and humans with diabetes mellitus show age-related performance impairments on memory tasks [38], and recent findings from rats indicate that these effects may be accompanied by changes in hippocampal insulin sensitivity [39].

The detrimental effects of high-fat diet on learning and memory may also be related to decreased expression of hippocampal brain-derived neurotrophic factor (BDNF), which is known to play an important role in activity-dependent synaptic plasticity in the adult brain [40,41]. In rodents, administration of exogenous BDNF decreases food intake, whereas genetic models with deficient BDNF signaling exhibit hyperphagia and obesity related primarily to marked increases in meal frequency, but not meal-size or duration [40, 42]. This meal pattern suggests that, like rats with selective hippocampal lesions, BDNF-deficient mice may be impaired at inhibiting responding to pre-oral and oral food cues that evoke learned appetitive responses [31, 32]. Impaired performance on hippocampal-dependent learning and memory tasks and reduced hippocampal BDNF is also found in rats that have been maintained on a high-fat diet [43, 44]. In humans, obese children and adolescents exhibit reduced serum BDNF levels relative to their normal weight counterparts, when variability due to age, gender, race, pubertal status, and platelet count is accounted for ([45] but see [46]). Furthermore, in what appears to be the only study of its kind with humans, an 8-year old female with haploinsufficiency for BDNF exhibited hyperphagia, severe obesity, and cognitive impairments [47].

Current research and theory has tended to treat the effects of BDNF on energy regulation and on cognitive functioning as largely independent phenomena, which involve distinct (e.g., hypothalamic and hippocampal, respectively) neural substrates [40, 48]. A question of interest is whether changes in BDNF or other physiological signals (e.g., leptin, insulin) could contribute to energy dysregulation and obesity as a primary consequence of impairing hippocampal functioning. For example, if intake of high-fat (or other) diets disrupts hippocampal functioning, and if one hippocampal function is to inhibit the ability cues associated with those diets to evoke appetitive and consummatory behaviors, this weakening of inhibitory control could promote obesity as part of a “vicious circle” of increasing fat intake, more severe disruption of hippocampal functioning, and further weakening of the inhibitory control [17]. Although direct tests are needed, much of the data presented in this brief review seem consistent with this general type of working hypothesis.

Conclusions

Researchers have identified a number of contact points between physiological signals and circuits involved with energy regulation and the hippocampus, a brain structure involved with learning and memory. Based on these findings, now may be the time to begin connecting these points within a more integrative conceptual framework. In addition to providing a more complete account of how animals maintain energy balance, this framework may lead to new therapeutic approaches the problems of obesity and cognitive decline.

Acknowledgments

The authors thank Leonard E. Jarrard for helpful comments and suggestions during the preparation of this manuscript. The authors also thank Andrea Tracy, Elwood Walls, and Larry Swanson for discussions that helped to develop and refine many of the ideas that are presented in this paper. Funding in support of this work was provided by Grants R01 HD44179 and R01 HD29792 from the National Institutes of Health to TLD.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Schwartz GJ. Biology of eating behavior in obesity. Obesity Research. 2004;12(Suppl 2):102S–106S. doi: 10.1038/oby.2004.274. [DOI] [PubMed] [Google Scholar]
  • 2.Moran TH. Gut peptide signaling in the controls of food intake. Obesity. 2006;14(Suppl 5):250S–253S. doi: 10.1038/oby.2006.318. [DOI] [PubMed] [Google Scholar]
  • 3.Benoit SC, Clegg DJ, Seeley RJ, Woods SC. Insulin and leptin as adiposity signals. Recent Progress in Hormone Research. 2004;59:267–285. doi: 10.1210/rp.59.1.267. [DOI] [PubMed] [Google Scholar]
  • 4.Cummings DE, Overduin J. Gastrointestinal regulation of food intake. Journal of Clinical Investigation. 2007;117:13–23. doi: 10.1172/JCI30227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Drazen DL, Vahl TP, D’Alessio DA, Seeley RJ, Woods SC. Effects of a fixed meal pattern on ghrelin secretion: evidence for a learned response independent of nutrient status. Endocrinology. 2006;147:23–30. doi: 10.1210/en.2005-0973. see comment. [DOI] [PubMed] [Google Scholar]
  • 6.Kanoski SE, Walls EK, Davidson TL. Interoceptive “satiety” signals produced by leptin and CCK. Peptides. 2007;28:988–1002. doi: 10.1016/j.peptides.2007.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Davidson TL, Kanoski SE, Tracy AL, Walls EK, Clegg D, Benoit SC. The interoceptive cue properties of ghrelin generalize to cues produced by food deprivation. Peptides. 2005;26:1602–1610. doi: 10.1016/j.peptides.2005.02.014. [DOI] [PubMed] [Google Scholar]
  • 8.Jewett DC, Lefever TW, Flashinski DP, Koffarnus MN, Cameron CR, Hehli DJ, Grace MK, Levine AS. Intraparaventricular neuropeptide Y and ghrelin induce learned behaviors that report food deprivation in rats. Neuroreport. 2006;17:733–737. doi: 10.1097/01.wnr.0000215767.94528.fb. [DOI] [PubMed] [Google Scholar]
  • 9.Woods SC. Gastrointestinal satiety signals I. An overview of gastrointestinal signals that influence food intake. American Journal of Physiology - Gastrointestinal & Liver Physiology. 2004;286:G7–13. doi: 10.1152/ajpgi.00448.2003. [DOI] [PubMed] [Google Scholar]
  • 10.Squire LR. Memory systems of the brain: a brief history and current perspective. Neurobiol Learn Mem. 2004;82:171–177. doi: 10.1016/j.nlm.2004.06.005. [DOI] [PubMed] [Google Scholar]
  • 11.Fehm HL, Kern W, Peters A. The selfish brain: competition for energy resources. Progress in Brain Research. 2006;153:129–140. doi: 10.1016/S0079-6123(06)53007-9. [DOI] [PubMed] [Google Scholar]
  • 12.Lathe R. Hormones and the hippocampus. J Endocrinol. 2001;169:205–231. doi: 10.1677/joe.0.1690205. [DOI] [PubMed] [Google Scholar]
  • 13.Morrris RGM. Theories of hippocampal function. In: Andersen P, Morris R, Amaral D, Bliss T, O’Keefe J, editors. The Hippocampus Book. Oxford University Press; 2006. pp. 581–713. [Google Scholar]
  • 14.Holland PC, Lamoureux JA, Han JS, Gallagher M. Hippocampal lesions interfere with Pavlovian negative occasion setting. Hippocampus. 1999;9:143–157. doi: 10.1002/(SICI)1098-1063(1999)9:2<143::AID-HIPO6>3.0.CO;2-Z. [DOI] [PubMed] [Google Scholar]
  • 15.Holland PC, Bouton ME. Hippocampus and context in classical conditioning. Current Opinion in Neurobiology. 1999;9:195–202. doi: 10.1016/s0959-4388(99)80027-0. [DOI] [PubMed] [Google Scholar]
  • 16.Kalisch R, Korenfeld E, Stephan KE, Weiskopf N, Seymour B, Dolan RJ. Context-dependent human extinction memory is mediated by a ventromedial prefrontal and hippocampal network. J Neurosci. 2006;26:9503–9511. doi: 10.1523/JNEUROSCI.2021-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Davidson TL, Kanoski SE, Walls EK, Jarrard LE. Memory inhibition and energy regulation. Physiology & Behavior. 2005;86:731–746. doi: 10.1016/j.physbeh.2005.09.004. [DOI] [PubMed] [Google Scholar]
  • 18.Farr SA, Banks WA, Morley JE. Effects of leptin on memory processing. Peptides. 2006;27:1420–1425. doi: 10.1016/j.peptides.2005.10.006. [DOI] [PubMed] [Google Scholar]
  • 19.Harvey J. Leptin: a diverse regulator of neuronal function. Journal of Neurochemistry. 2007;100:307–313. doi: 10.1111/j.1471-4159.2006.04205.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Zhao WQ, Chen H, Quon MJ, Alkon DL. Insulin and the insulin receptor in experimental models of learning and memory. European Journal of Pharmacology. 2004;490:71–81. doi: 10.1016/j.ejphar.2004.02.045. [DOI] [PubMed] [Google Scholar]
  • 21.Lynch MA. Long-term potentiation and memory. Physiological Reviews. 2004;84:87–136. doi: 10.1152/physrev.00014.2003. [DOI] [PubMed] [Google Scholar]
  • 22.Matsushita H, Akiyoshi J, Kai K, Ishii N, Kodama K, Tsutsumi T, Isogawa K, Nagayama H. Spatial memory impairment in OLETF rats without cholecystokinin - a receptor. Neuropeptides. 2003;37:271–276. doi: 10.1016/s0143-4179(03)00083-0. [DOI] [PubMed] [Google Scholar]
  • 23.Moran TH, Katz LF, Plata-Salaman CR, Schwartz GJ. Disordered food intake and obesity in rats lacking cholecystokinin A receptors. Am J Physiol. 1998;274:R618–625. doi: 10.1152/ajpregu.1998.274.3.R618. [DOI] [PubMed] [Google Scholar]
  • 24.Cenquizca LA, Swanson LW. Analysis of direct hippocampal cortical field CA1 axonal projections to diencephalon in the rat. Journal of Comparative Neurology. 2006;497:101–114. doi: 10.1002/cne.20985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wang GJ, Yang J, Volkow ND, Telang F, Ma Y, Zhu W, Wong CT, Tomasi D, Thanos PK, Fowler JS. Gastric stimulation in obese subjects activates the hippocampus and other regions involved in brain reward circuitry. Proc Natl Acad Sci U S A. 2006;103:15641–15645. doi: 10.1073/pnas.0601977103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.DelParigi A, Chen K, Salbe AD, Hill JO, Wing RR, Reiman EM, Tataranni PA. Persistence of abnormal neural responses to a meal in postobese individuals. Int J Obes Relat Metab Disord. 2004;28:370–377. doi: 10.1038/sj.ijo.0802558. [DOI] [PubMed] [Google Scholar]
  • 27.Gautier JF, Del Parigi A, Chen K, Salbe AD, Bandy D, Pratley RE, Ravussin E, Reiman EM, Tataranni PA. Effect of satiation on brain activity in obese and lean women. Obesity Research. 2001;9:676–684. doi: 10.1038/oby.2001.92. see comment. [DOI] [PubMed] [Google Scholar]
  • 28.Hebben N, Corkin S, Eichenbaum H, Shedlack K. Diminished ability to interpret and report internal states after bilateral medial temporal resection: case H.M. Behavioral Neuroscience. 1985;99:1031–1039. doi: 10.1037//0735-7044.99.6.1031. [DOI] [PubMed] [Google Scholar]
  • 29.Rozin P, Dow S, Moscovitch M, Rajaram S. What causes humans to begin and end a meal? A role for memory for what has been eaten, as evidenced by a study of multiple meal eating in amnesic patients. Psychological Science. 1998;9:392–396. [Google Scholar]
  • 30.Higgs S. Memory and its role in appetite regulation. Physiology & Behavior. 2005;85:67–72. doi: 10.1016/j.physbeh.2005.04.003. [DOI] [PubMed] [Google Scholar]
  • 31.Clifton PG, Vickers SP, Somerville EM. Little and often: Ingestive behavior patterns following hippocampal lesions in rats. Behavioral Neuroscience. 1998;112:502–511. doi: 10.1037//0735-7044.112.3.502. [DOI] [PubMed] [Google Scholar]
  • 32.Davidson TL, Jarrard LE. A role for hippocampus in the utilization of hunger signals. Behav Neural Biol. 1993;59:167–171. doi: 10.1016/0163-1047(93)90925-8. [DOI] [PubMed] [Google Scholar]
  • 33.Schmelzeis MC, Mittleman G. The hippocampus and reward: effects of hippocampal lesions on progressive-ratio responding. Behavioral Neuroscience. 1996;110:1049–1066. doi: 10.1037//0735-7044.110.5.1049. [DOI] [PubMed] [Google Scholar]
  • 34.Hock BJ, Jr, Bunsey MD. Differential effects of dorsal and ventral hippocampal lesions. Journal of Neuroscience. 1998;18:7027–7032. doi: 10.1523/JNEUROSCI.18-17-07027.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kennedy PJ, Shapiro ML. Retrieving memories via internal context requires the hippocampus. Journal of Neuroscience. 2004;24:6979–6985. doi: 10.1523/JNEUROSCI.1388-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tremblay A. Dietary fat and body weight set point. Nutrition Reviews. 2004;62:S75–77. doi: 10.1111/j.1753-4887.2004.tb00092.x. [DOI] [PubMed] [Google Scholar]
  • 37.Haan MN, Wallace R. Can dementia be prevented? Brain aging in a population-based context. Annual Review of Public Health. 2004;25:1–24. doi: 10.1146/annurev.publhealth.25.101802.122951. [DOI] [PubMed] [Google Scholar]
  • 38.Biessels GJ, Gispen WH. The impact of diabetes on cognition: what can be learned from rodent models? Neurobiology of Aging. 2005;26(Suppl 1):36–41. doi: 10.1016/j.neurobiolaging.2005.08.015. [DOI] [PubMed] [Google Scholar]
  • 39.Winocur G, Greenwood CE, Piroli GG, Grillo CA, Reznikov LR, Reagan LP, McEwen BS. Memory impairment in obese Zucker rats: an investigation of cognitive function in an animal model of insulin resistance and obesity. Behavioral Neuroscience. 2005;119:1389–1395. doi: 10.1037/0735-7044.119.5.1389. [DOI] [PubMed] [Google Scholar]
  • 40.Lebrun B, Bariohay B, Moyse E, Jean A. Brain-derived neurotrophic factor (BDNF) and food intake regulation: a minireview. Autonomic Neuroscience-Basic & Clinical. 2006;126-127:30–38. doi: 10.1016/j.autneu.2006.02.027. [DOI] [PubMed] [Google Scholar]
  • 41.Yamada K, Nabeshima T. Brain-derived neurotrophic factor/TrkB signaling in memory processes. Journal of Pharmacological Sciences. 2003;91:267–270. doi: 10.1254/jphs.91.267. [DOI] [PubMed] [Google Scholar]
  • 42.Fox EA, Byerly MS. A mechanism underlying mature-onset obesity: evidence from the hyperphagic phenotype of brain-derived neurotrophic factor mutants. American Journal of Physiology - Regulatory Integrative & Comparative Physiology. 2004;286:R994–1004. doi: 10.1152/ajpregu.00727.2003. [DOI] [PubMed] [Google Scholar]
  • 43.Kanoski SE, Meisel RL, Mullins AJ, Davidson TL. The effects of energy-rich diets on discrimination reversal learning and on BDNF in the hippocampus and prefrontal cortex of the rat. Behav Brain Res. 2007;182:57–66. doi: 10.1016/j.bbr.2007.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Molteni R, Barnard RJ, Ying Z, Roberts CK, Gomez-Pinilla F. A high-fat, refined sugar diet reduces hippocampal brain-derived neurotrophic factor, neuronal plasticity, and learning. Neuroscience. 2002;112:803–814. doi: 10.1016/s0306-4522(02)00123-9. [DOI] [PubMed] [Google Scholar]
  • 45.El-Gharbawy AH, Adler-Wailes DC, Mirch MC, Theim KR, Ranzenhofer L, Tanofsky-Kraff M, Yanovski JA. Serum brain-derived neurotrophic factor concentrations in lean and overweight children and adolescents. Journal of Clinical Endocrinology & Metabolism. 2006;91:3548–3552. doi: 10.1210/jc.2006-0658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Monteleone P, Tortorella A, Martiadis V, Serritella C, Fuschino A, Maj M. Opposite changes in the serum brain-derived neurotrophic factor in anorexia nervosa and obesity. Psychosomatic Medicine. 2004;66:744–748. doi: 10.1097/01.psy.0000138119.12956.99. [DOI] [PubMed] [Google Scholar]
  • 47.Gray J, Yeo GS, Cox JJ, Morton J, Adlam AL, Keogh JM, Yanovski JA, El Gharbawy A, Han JC, Tung YC, et al. Hyperphagia, severe obesity, impaired cognitive function, and hyperactivity associated with functional loss of one copy of the brain-derived neurotrophic factor (BDNF) gene. Diabetes. 2006;55:3366–3371. doi: 10.2337/db06-0550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Monteggia LM, Barrot M, Powell CM, Berton O, Galanis V, Gemelli T, Meuth S, Nagy A, Greene RW, Nestler EJ. Essential role of brain-derived neurotrophic factor in adult hippocampal function. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:10827–10832. doi: 10.1073/pnas.0402141101. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES