Skip to main content

OPINION article

Front. Endocrinol., 18 July 2014
Sec. Neuroendocrine Science

The Gut–Brain-Axis as a Target to Treat Stress-Induced Obesity

  • 1School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
  • 2Department of Neuroscience, Carleton University, Ottawa, ON, Canada

The emergence of obesity as a pandemic has led to increased efforts to determine the causes for this disorder and potential new treatments to prevent and/or treat those affected by it. The mechanisms underlying the ontogeny of obesity are complex. They involve an interaction between a genetic predisposition to this disorder and environmental conditions that catalyze the development of an obese phenotype (1). It has become evident that stress may be a strong environmental factor leading to metabolic changes that lead to obesity. Stress is a concept coined to describe the state that is generated when physiological or psychological wellbeing is challenged. It is associated with physiological and behavioral responses considered adaptive and conducive to reduce or to cope with the challenges posed by the stressor (2, 3). Continuous stressful events, however, result in pathological states including some of the same conditions associated with obesity (4, 5). In particular, continuous social stressors result in increased body weight and abdominal fat deposition, insulin resistance, and cardiovascular disease (2, 4). The underlying mechanisms and relevant potential treatments are less well documented. Nevertheless, there is increasing evidence to suggest that psychological stressors represent a homeostatic challenge, and as such have a strong impact on the brain systems associated with homeostatic control (3).

It is clear that the brain plays a critical role in the regulation of energy balance, and as such represents a target for therapeutic intervention. For instance, cells groups within the hypothalamic arcuate nucleus (ARC) are important for regulating food intake and energy balance, whereas a number of regions across the mesolimbic dopaminergic system and ascending noradrenergic inputs stemming from the brain stem regulate hedonic and short term feeding responses (6). In spite of this, few viable therapeutic options have emerged from these advances particularly given the fact that many drugs targeting these systems have substantial side effects. Here, we propose that the periphery, and in particular the gut, may represent an alternate target for treatments that can reduce obesity, particularly in the face of stress.

Regulation of Food Intake and Energy Balance by Gut Hormones

Recent evidence has brought greater attention to the gut as a key contributor to the regulation of food intake and energy balance. The gut serves both as a sensory organ for nutrients and can regulate the activity of brain centers associated with the regulation of food intake and energy balance. One indication of the importance of the gut–brain-axis is that gastrointestinal cells serve as nutrient sensors and produce hormonal and neural responses to nutrients that target the brain to modulate food intake and energy balance (7). A number of experiments have demonstrated that animals can detect and bar press for intragastric infusions of solutions containing sucrose or fat infused directly into the gut (7). The presence of lipids in the gut decreases hepatic glucose production, linking the gut–brain-axis with liver function (8). Vagotomy, inhibition of the N-methyl-d-aspartate receptor in the nucleus of the solitary tract, sympathetic denervation, and blockade of β2-adrenoceptor abolished the effects of lipid on the regulation of glucose homeostasis (9). Notably, the gut–brain–liver-axis is disturbed by chronic exposure to a high fat diet (10). In addition to stimulating the ascending vagus nerve, cells in the gut signal the brain through a number of endocrine signals. These include peptide YY (PYY), neuropeptide Y (NPY), cholecystokinin (CCK), oxyntomodulin (OXM), glucagon-like peptide-1 (GLP1), and ghrelin, all of which control appetite and glucose homeostasis (1114). While PYY, CCK, OXM, and GLP1 are anorectic and some increase energy expenditure, ghrelin is a potent orexigenic hormone that also influences metabolic rate by favoring the utilization of carbohydrates instead of lipids as a source of energy, resulting in increased adiposity and body weight (15). Given that stressors and the physiological responses elicited to cope with them can generate a substantial energy drain, it is not surprising that these gut signals are altered during the stress response, and hence could represent a novel target to control stress-induced obesity.

Stress and the Gut–Brain-Axis

The effects of stress on the gastrointestinal system have been known for a long time, in particular the effects of stress on gastric motility and on gastric acid secretion. Continuous stress has been associated with gastric ulceration and other gastrointestinal disorders like irritable bowel syndrome. One would presume that, if the gut plays an important role in the regulation of energy balance, and if the function of the gut is altered during stress, then stress could alter the function of the gut to promote obesity. The clearest evidence for this is the effect of stress on ghrelin secretion. Ghrelin, a 28 amino acid peptide secreted by oxyntic cells in the stomach and upper intestine, is the only gastrointestinal peptide known to stimulate food intake and alter energy expenditure (15, 16). Plasma ghrelin rises following an acute fast or during periods of caloric restriction, where the daily intake of accessible food is lower than the daily ad libitum access (17). Interestingly, ghrelin is secreted concomitantly with glucocorticoids following acute and chronic stress (18, 19) and plasma active ghrelin concentrations remain significantly elevated in the late phase of a stress session (18).

Acute stressors elevate plasma ghrelin through the activation of the sympathetic and enteric nervous system, but recent data suggest that stress-induced ghrelin secretion may be the result of stimulation of corticotropin releasing hormone (CRH) receptors in the gut, CRH and CRH-related peptides such as urocortin-1 and 2 (2022). For instance, central stimulation of CRH1 and CRH2 receptors produces stress like effect in gastrointestinal motility, gastric emptying, and colonic propulsion, whereas blockade of CRH1 and CRH2 receptors prevents some of these effects (22). Ghrelin secretion in response to stress may also be related to the effects of locally released urocortin-1 acting on CRH2 receptors in the gut (21). This process may be important acutely, given that urocortin reduces pain in the gastrointestinal tract (23), and given that ghrelin protects the stomach against gastric ulceration induced by repeated stressors (2426). Problems, however, may arise when the stressor is chronic. For instance, in mice, chronic social defeat stress regimen that lasts 10–21 days, increases ghrelin concentrations in concert with increases in caloric intake and weight gain (27, 28). This stress paradigm also increases hypothalamic expression of orexigenic peptides such as NPY and Agouti-related peptide, and plasma biomarkers indicative of obesity, an effect that persisted for at least 2 weeks after the stress paradigm was terminated (27). In contrast, GHSR KO mice or mice receiving chronic intracerebroventricular infusions of a ghrelin receptor antagonist do not increase their caloric intake or weight gain in response to the same stressor (27). Thus, it is clear that prolonged periods of social stress can lead to high ghrelin concentrations that promote higher caloric intake and alterations in energy expenditure that lead to weight gain and adipose tissue accumulation.

Another mechanism by which stress-induced ghrelin secretion is the stimulation of the mesolimbic dopaminergic system that is critical for the regulation of reward seeking behaviors. Ghrelin receptors are found in dopamine neurons within the midbrain ventral tegmental area (VTA), and here ghrelin can stimulate dopamine release and food intake and motivation to obtain palatable foods, and ghrelin receptor antagonism prevents this (1, 29, 30). Similarly, mice with genetic deletion of the GHSR show less preference for high calorie foods. Selectively restoring ghrelin receptors in dopamine producing cells can enhance their preference for these foods (31, 32). During stress, ghrelin may act in the VTA to increase appetite, but prolonged exposure to stressors may ultimately prevent ghrelin from increasing appetite in this region and ultimately lead to anhedonia (28, 31). Given these data, ghrelin, urocortin-1, and their respective receptors represent promising potential peripheral targets to reduce stress-induced weight gain and appetite.

Besides ghrelin, other gut peptides are also secreted and may have an influence in the stress response, although less is known about how prolonged periods of stress affect the secretion of these peptides. Acute stressors like restraint cause increases in the peripheral and central release of NPY, GLP1, CCK, OXM, and motilin (3335). Of these, NPY has received special attention for a number of reasons. NPY neurons in the ARC are important in the integration of peripheral signals regulating energy balance including those coming from the gut, and project to hypothalamic and extrahypothalamic brain region to stimulate feeding and to alter behavior including those associated with mood (36, 37). Sympathetic nervous system terminals also release NPY. Following chronic stress, increased sympathetic release of NPY leads to inflammatory responses, fat angiogenesis, and adipocyte enlargement and proliferation ultimately leading to obesity, and these effects are mediated by Y2 receptors localized in adipocytes (38). It is not known if gut derived NPY is over-secreted following chronic stress, or if it has similar direct effects on adipocytes as NPY secreted by sympathetic terminals, but it is not unlikely that this would contribute to an obesogenic state.

A hormone that could counter the NPY effects is GLP1. This peptide is released by L-cells in the gut and has emerged as an important player in the regulation of appetite and glucose homeostasis (39). In addition, GLP1 is released centrally and acts both in the hypothalamus and midbrain VTA dopamine cells to reduce appetite, increase energy expenditure, and decrease motivated behaviors (4043). Interestingly, GLP1 KO mice have abnormal hormonal responses to acute stressors (44). Within the periphery, GLP1 can act locally to protect the gut from stress-induced gastric acid secretion, and is important for altering gastric motility (45, 46). More importantly, GLP1 protects a number of tissues affected by chronic stress including pancreatic β-cells, cardiomyocytes, and kidney cells, while reducing cytokine induced inflammation (4750). Whether chronic stress results in altered secretion of either of these peptides is not known, and it may be critical to determine if this is the case in order to fully determine the usefulness of these peptides as potential treatments for stress-induced pathology.

Potential Future Directions

One of the problems that exist with trying to counter pathological conditions associated with stress is that either the stressor is difficult to remove or the stressor leaves symptoms that persist in spite of the stressor being removed. In this sense, pharmacological interventions derived from gut peptides and aimed at reducing metabolic alterations caused by stress may not represent a “magic bullet” that can reverse metabolic changes to an optimal state. These, however, may become interventions that can help to deal with these problems in the short term while a patient finds a way to deal or remove herself from the stressor. For example, drugs that act to decrease ghrelin signaling (i.e., ghrelin receptor antagonists or inverse agonists) could be used to decrease stress-induced caloric intake and weight gain as well as stress-induced gastric alterations (51, 52). Nevertheless, enthusiasm for these types of drugs has been hampered by evidence suggesting that stress-induced ghrelin secretion is necessary not only to maintain metabolic homeostasis but also to prevent stress-induced depressive like behaviors and reduce anxiety (28, 53, 54). These data remain unclear, however, as other studies show that ghrelin is actually anxiogenic and increases the formation of fearful memories (5559). Clearly, an in depth analysis of these data is required to explain these paradoxical results, but at the very least, they suggest that drugs blocking the ghrelin system could have a negative impact on mood. Furthermore, the use of ghrelin receptor antagonists or inverse agonists may cause undesired side effects given the ligand independent interaction between ghrelin receptors and other G-coupled protein receptors in the central nervous system (60).

Perhaps a better alternative would be to use drugs that decrease acyl-ghrelin levels without depleting the system from ghrelin, or altering GHSR signaling thereby maintain ghrelin’s protective effects. One potential target for this is ghrelin-O-acyltransferase (GOAT, also known as MBOAT4), an enzyme that is required for the esterification process that links n-octanoic acid to the ghrelin molecule (61). The GOAT enzyme is produced by the same cells that secrete ghrelin (61), and drugs that reduce the activity of this enzyme not only reduce plasma active (acylated) ghrelin concentrations, but they also cause a decrease in weight gain and adiposity in mice (62). Whether GOAT inhibitors improve metabolic changes caused by stressors remains to be determined. Alternatively, des-acyl ghrelin may also be useful given that, like GOAT inhibitors, des-acyl ghrelin and its analogs decrease acyl-ghrelin concentrations, decrease high fat diet intake, weight gain, and adiposity, improve glycemic index, and are protective in cardiomyocytes in a GHSR independent manner (6365). Finally, CRH2 receptor antagonists could be used to prevent stress-induced release of ghrelin to prevent the over-secretion of this peptide.

Nevertheless, GLP1 may be the most viable target at the moment since a number of analogs for this peptide are already FDA approved and currently used in the control of type II diabetes. Thus, drugs that mimic GLP1 or that decrease the activity of dipeptidyl-peptidase IV, an enzyme that cleaves GLP1 into an inactive byproduct, may be useful in increasing incretin tone and reducing the effects of stress on metabolism by doing so. GLP1 treatments may, however, be most useful when acting peripherally and not centrally, as GLP1 and its analogs can exacerbate stress responses and decrease motivated behaviors acting in the brain (66, 67). This, however, may not be the case as a GLP1 analog that crosses the blood brain barrier did not have an anxiogenic effect, and increased hippocampal neurogenesis (68).

In conclusion, it is only through identifying and understanding the mechanisms responsible for stress-induced obesity that effective therapeutics can be generated. Gut peptides associated with hunger and satiety may represent important players in these mechanisms, as they are also modulated by the responses to stressors. More importantly, they may also represent a potential therapeutic avenue for acute pharmacological intervention given that these are produced peripherally, and also influence the central nervous system. Nevertheless, relatively speaking, little is known about how these peptides are regulated in the face of stress, particularly chronic stressors. This knowledge is critically needed to determine if these peptides and their receptors will be useful for the treatment of stress-induced pathological conditions including obesity and metabolic syndrome.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

This work was funded by a Canadian Institutes for Health Research operating grant (Alfonso Abizaid) and research funds from the Monash University Malaysia (Chooi Yeng Lee).

References

1. Abizaid A, Gao Q, Horvath TL. Thoughts for food: brain mechanisms and peripheral energy balance. Neuron (2006) 51:691–702. doi:10.1016/j.neuron.2006.08.025

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

2. Anisman H, Merali Z, Hayley S. Neurotransmitter, peptide and cytokine processes in relation to depressive disorder: comorbidity between depression and neurodegenerative disorders. Prog Neurobiol (2008) 85:1–74. doi:10.1016/j.pneurobio.2008.01.004

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

3. McEwen BS. Physiology and neurobiology of stress and adaptation: central role of the brain. Physiol Rev (2007) 87:873–904. doi:10.1152/physrev.00041.2006

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

4. Black PH. The inflammatory consequences of psychologic stress: relationship to insulin resistance, obesity, atherosclerosis and diabetes mellitus, type II. Med Hypotheses (2006) 67:879–91. doi:10.1016/j.mehy.2006.04.008

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

5. Coccurello R, D’Amato FR, Moles A. Chronic social stress, hedonism and vulnerability to obesity: lessons from rodents. Neurosci Biobehav Rev (2009) 33:537–50. doi:10.1016/j.neubiorev.2008.05.018

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

6. Abizaid A, Horvath TL. Brain circuits regulating energy homeostasis. Regul Pept (2008) 149:3–10. doi:10.1016/j.regpep.2007.10.006

CrossRef Full Text

7. Sclafani A, Ackroff K. Role of gut nutrient sensing in stimulating appetite and conditioning food preferences. Am J Physiol Regul Integr Comp Physiol (2012) 302:R1119–33. doi:10.1152/ajpregu.00038.2012

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

8. Rasmussen BA, Breen DM, Lam TK. Lipid sensing in the gut, brain and liver. Trends Endocrinol Metab (2012) 23:49–55. doi:10.1016/j.tem.2011.11.001

CrossRef Full Text

9. Schwartz GJ. Gut fat sensing in the negative feedback control of energy balance – recent advances. Physiol Behav (2011) 104:621–3. doi:10.1016/j.physbeh.2011.05.003

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

10. Lee CY. The effect of high fat-diet-induced pathophysiological changes in the gut on obesity: what should be the ideal treatment? Clin Transl Gastroenterol (2013) 4:e39. doi:10.1038/ctg.2013.11

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

11. Sam AH, Troke RC, Tan TM, Bewick GA. The role of the gut/brain axis in modulating food intake. Neuropharmacology (2012) 63:46–56. doi:10.1016/j.neuropharm.2011.10.008

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

12. Dham S, Banerji MA. The brain-gut axis in regulation of appetite and obesity. Pediatr Endocrinol Rev (2006) 3(Suppl 4):544–54.

13. Konturek SJ, Konturek JW, Pawlik T, Brzozowski T. Brain-gut axis and its role in the control of food intake. J Physiol Pharmacol (2004) 55:137–54.

Pubmed Abstract | Pubmed Full Text

14. Janssen S, Depoortere I. Nutrient sensing in the gut: new roads to therapeutics? Trends Endocrinol Metab (2013) 24:92–100. doi:10.1016/j.tem.2012.11.006

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

15. Tschop M, Smiley DL, Heiman ML. Ghrelin induces adiposity in rodents. Nature (2000) 407:908–13. doi:10.1038/35038090

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

16. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature (1999) 402:656–60. doi:10.1038/45230

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

17. Toshinai K, Mondal MS, Nakazato M, Date Y, Murakami N, Kojima M, et al. Upregulation of Ghrelin expression in the stomach upon fasting, insulin-induced hypoglycemia, and leptin administration. Biochem Biophys Res Commun (2001) 281:1220–5. doi:10.1006/bbrc.2001.4518

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

18. Kristenssson E, Sundqvist M, Astin M, Kjerling M, Mattsson H, Dornonville de la Cour C, et al. Acute psychological stress raises plasma ghrelin in the rat. Regul Pept (2006) 134:114–7. doi:10.1016/j.regpep.2006.02.003

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

19. Ochi M, Tominaga K, Tanaka F, Tanigawa T, Shiba M, Watanabe T, et al. Effect of chronic stress on gastric emptying and plasma ghrelin levels in rats. Life Sci (2008) 82:862–8. doi:10.1016/j.lfs.2008.01.020

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

20. Mundinger TO, Cummings DE, Taborsky GJ Jr. Direct stimulation of ghrelin secretion by sympathetic nerves. Endocrinology (2006) 147:2893–901. doi:10.1210/en.2005-1182

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

21. Wang L, Stengel A, Goebel-Stengel M, Shaikh A, Yuan PQ, Tache Y. Intravenous injection of urocortin 1 induces a CRF2 mediated increase in circulating ghrelin and glucose levels through distinct mechanisms in rats. Peptides (2013) 39:164–70. doi:10.1016/j.peptides.2012.11.009

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

22. Tache Y, Million M, Nelson AG, Lamy C, Wang L. Role of corticotropin-releasing factor pathways in stress-related alterations of colonic motor function and viscerosensibility in female rodents. Gend Med (2005) 2:146–54. doi:10.1016/S1550-8579(05)80043-9

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

23. Million M, Maillot C, Saunders P, Rivier J, Vale W, Tache Y. Human urocortin II, a new CRF-related peptide, displays selective CRF(2)-mediated action on gastric transit in rats. Am J Physiol Gastrointest Liver Physiol (2002) 282:G34–40. doi:10.1152/ajpgi.00283.2001

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

24. Brzozowski T, Konturek PC, Konturek SJ, Kwiecien S, Drozdowicz D, Bielanski W, et al. Exogenous and endogenous ghrelin in gastroprotection against stress-induced gastric damage. Regul Pept (2004) 120:39–51. doi:10.1016/j.regpep.2004.02.010

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

25. Brzozowski T, Konturek PC, Sliwowski Z, Drozdowicz D, Kwiecien S, Pawlik M, et al. Neural aspects of ghrelin-induced gastroprotection against mucosal injury induced by noxious agents. J Physiol Pharmacol (2006) 57(Suppl 6):63–76.

Pubmed Abstract | Pubmed Full Text

26. Sibilia V, Rindi G, Pagani F, Rapetti D, Locatelli V, Torsello A, et al. Ghrelin protects against ethanol-induced gastric ulcers in rats: studies on the mechanisms of action. Endocrinology (2003) 144:353–9. doi:10.1210/en.2002-220756

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

27. Patterson ZR, Khazall R, Mackay H, Anisman H, Abizaid A. Central ghrelin signaling mediates the metabolic response of C57BL/6 male mice to chronic social defeat stress. Endocrinology (2013) 154:1080–91. doi:10.1210/en.2012-1834

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

28. Lutter M, Sakata I, Osborne-Lawrence S, Rovinsky SA, Anderson JG, Jung S, et al. The orexigenic hormone ghrelin defends against depressive symptoms of chronic stress. Nat Neurosci (2008) 11:752–3. doi:10.1038/nn.2139

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

29. Jerlhag E, Egecioglu E, Dickson SL, Andersson M, Svensson L, Engel JA. Ghrelin stimulates locomotor activity and accumbal dopamine-overflow via central cholinergic systems in mice: implications for its involvement in brain reward. Addict Biol (2006) 11:45–54. doi:10.1111/j.1369-1600.2006.00002.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

30. King SJ, Isaacs AM, O’Farrell E, Abizaid A. Motivation to obtain preferred foods is enhanced by ghrelin in the ventral tegmental area. Horm Behav (2011) 60:572–80. doi:10.1016/j.yhbeh.2011.08.006

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

31. Chuang JC, Perello M, Sakata I, Osborne-Lawrence S, Savitt JM, Lutter M, et al. Ghrelin mediates stress-induced food-reward behavior in mice. J Clin Invest (2011) 121:2684–92. doi:10.1172/JCI57660

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

32. Perello M, Sakata I, Birnbaum S, Chuang JC, Osborne-Lawrence S, Rovinsky SA, et al. Ghrelin increases the rewarding value of high-fat diet in an orexin-dependent manner. Biol Psychiatry (2010) 67:880–6. doi:10.1016/j.biopsych.2009.10.030

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

33. Liang C, Luo H, Liu Y, Cao J, Xia H. Plasma hormones facilitated the hypermotility of the colon in a chronic stress rat model. PLoS One (2012) 7:e31774. doi:10.1371/journal.pone.0031774

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

34. Fukudo S, Suzuki J. Colonic motility, autonomic function, and gastrointestinal hormones under psychological stress on irritable bowel syndrome. Tohoku J Exp Med (1987) 151:373–85. doi:10.1620/tjem.151.373

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

35. Jonsson BH, Hellstrom PM. Motilin- and neuropeptide Y-like immunoreactivity in a psychophysiological stress experiment on patients with functional dyspepsia. Integr Physiol Behav Sci (2000) 35:256–65. doi:10.1007/BF02688788

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

36. Gray TS, Morley JE. Neuropeptide Y: anatomical distribution and possible function in mammalian nervous system. Life Sci (1986) 38:389–401. doi:10.1016/0024-3205(86)90061-5

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

37. Elias CF, Saper CB, Maratos-Flier E, Tritos NA, Lee C, Kelly J, et al. Chemically defined projections linking the mediobasal hypothalamus and the lateral hypothalamic area. J Comp Neurol (1998) 402:442–59. doi:10.1002/(SICI)1096-9861(19981228)402:4<442::AID-CNE2>3.3.CO;2-I

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

38. Kuo LE, Kitlinska JB, Tilan JU, Li L, Baker SB, Johnson MD, et al. Neuropeptide Y acts directly in the periphery on fat tissue and mediates stress-induced obesity and metabolic syndrome. Nat Med (2007) 13:803–11. doi:10.1038/nm0907-1120

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

39. van Dijk G, Thiele TE. Glucagon-like peptide-1 (7-36) amide: a central regulator of satiety and interoceptive stress. Neuropeptides (1999) 33:406–14. doi:10.1054/npep.1999.0053

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

40. O’Shea D, Gunn I, Chen X, Bloom S, Herbert J. A role for central glucagon-like peptide-1 in temperature regulation. Neuroreport (1996) 7:830–2. doi:10.1097/00001756-199602290-00035

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

41. Turton MD, O’Shea D, Gunn I, Beak SA, Edwards CM, Meeran K, et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature (1996) 379:69–72. doi:10.1038/379069a0

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

42. Alhadeff AL, Rupprecht LE, Hayes MR. GLP-1 neurons in the nucleus of the solitary tract project directly to the ventral tegmental area and nucleus accumbens to control for food intake. Endocrinology (2012) 153:647–58. doi:10.1210/en.2011-1443

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

43. Shirazi RH, Dickson SL, Skibicka KP. Gut peptide GLP-1 and its analogue, exendin-4, decrease alcohol intake and reward. PLoS One (2013) 8:e61965. doi:10.1371/journal.pone.0061965

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

44. MacLusky NJ, Cook S, Scrocchi L, Shin J, Kim J, Vaccarino F, et al. Neuroendocrine function and response to stress in mice with complete disruption of glucagon-like peptide-1 receptor signaling. Endocrinology (2000) 141:752–62. doi:10.1210/en.141.2.752

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

45. Schirra J, Katschinski M, Weidmann C, Schafer T, Wank U, Arnold R, et al. Gastric emptying and release of incretin hormones after glucose ingestion in humans. J Clin Invest (1996) 97:92–103. doi:10.1172/JCI118411

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

46. Schirra J, Kuwert P, Wank U, Leicht P, Arnold R, Goke B, et al. Differential effects of subcutaneous GLP-1 on gastric emptying, antroduodenal motility, and pancreatic function in men. Proc Assoc Am Physicians (1997) 109:84–97.

Pubmed Abstract | Pubmed Full Text

47. Younce CW, Burmeister MA, Ayala JE. Exendin-4 attenuates high glucose-induced cardiomyocyte apoptosis via inhibition of endoplasmic reticulum stress and activation of SERCA2a. Am J Physiol Cell Physiol (2013) 304:C508–18. doi:10.1152/ajpcell.00248.2012

CrossRef Full Text

48. Wang D, Luo P, Wang Y, Li W, Wang C, Sun D, et al. Glucagon-like peptide-1 protects against cardiac microvascular injury in diabetes via a cAMP/PKA/Rho-dependent mechanism. Diabetes (2013) 62:1697–708. doi:10.2337/db12-1025

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

49. Tsunekawa S, Yamamoto N, Tsukamoto K, Itoh Y, Kaneko Y, Kimura T, et al. Protection of pancreatic beta-cells by exendin-4 may involve the reduction of endoplasmic reticulum stress; in vivo and in vitro studies. J Endocrinol (2007) 193:65–74. doi:10.1677/JOE-06-0148

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

50. Fujita H, Morii T, Fujishima H, Sato T, Shimizu T, Hosoba M, et al. The protective roles of GLP-1R signaling in diabetic nephropathy: possible mechanism and therapeutic potential. Kidney Int (2014) 85:579–89. doi:10.1038/ki.2013.427

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

51. Nahata M, Muto S, Oridate N, Ohnishi S, Nakagawa K, Sadakane C, et al. Impaired ghrelin signaling is associated with gastrointestinal dysmotility in rats with gastroesophageal reflux disease. Am J Physiol Gastrointest Liver Physiol (2012) 303:G42–53. doi:10.1152/ajpgi.00462.2011

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

52. Nahata M, Saegusa Y, Sadakane C, Yamada C, Nakagawa K, Okubo N, et al. Administration of exogenous acylated ghrelin or rikkunshito, an endogenous ghrelin enhancer, improves the decrease in postprandial gastric motility in an acute restraint stress mouse model. Neurogastroenterol Motil (2014) 26:821–31. doi:10.1111/nmo.12336

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

53. Carlini VP, Machado DG, Buteler F, Ghersi M, Ponzio MF, Martini AC, et al. Acute ghrelin administration reverses depressive-like behavior induced by bilateral olfactory bulbectomy in mice. Peptides (2012) 35:160–5. doi:10.1016/j.peptides.2012.03.031

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

54. Spencer SJ, Xu L, Clarke MA, Lemus M, Reichenbach A, Geenen B, et al. Ghrelin regulates the hypothalamic-pituitary-adrenal axis and restricts anxiety after acute stress. Biol Psychiatry (2012) 72:457–65. doi:10.1016/j.biopsych.2012.03.010

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

55. Asakawa A, Inui A, Kaga T, Yuzuriha H, Nagata T, Fujimiya M, et al. A role of ghrelin in neuroendocrine and behavioral responses to stress in mice. Neuroendocrinology (2001) 74:143–7. doi:10.1159/000054680

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

56. Carlini VP, Monzon ME, Varas MM, Cragnolini AB, Schioth HB, Scimonelli TN, et al. Ghrelin increases anxiety-like behavior and memory retention in rats. Biochem Biophys Res Commun (2002) 299:739–43. doi:10.1016/S0006-291X(02)02740-7

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

57. Currie PJ, Khelemsky R, Rigsbee EM, Dono LM, Coiro CD, Chapman CD, et al. Ghrelin is an orexigenic peptide and elicits anxiety-like behaviors following administration into discrete regions of the hypothalamus. Behav Brain Res (2012) 226:96–105. doi:10.1016/j.bbr.2011.08.037

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

58. Hansson C, Haage D, Taube M, Egecioglu E, Salome N, Dickson SL. Central administration of ghrelin alters emotional responses in rats: behavioural, electrophysiological and molecular evidence. Neuroscience (2011) 180:201–11. doi:10.1016/j.neuroscience.2011.02.002

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

59. Meyer RM, Burgos-Robles A, Liu E, Correia SS, Goosens KA. A ghrelin-growth hormone axis drives stress-induced vulnerability to enhanced fear. Mol Psychiatry (2013):1–11. doi:10.1038/mp.2013.135

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

60. Kern A, Albarran-Zeckler R, Walsh HE, Smith RG. Apo-ghrelin receptor forms heteromers with DRD2 in hypothalamic neurons and is essential for anorexigenic effects of DRD2 agonism. Neuron (2012) 73:317–32. doi:10.1016/j.neuron.2011.10.038

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

61. Yang J, Brown MS, Liang G, Grishin NV, Goldstein JL. Identification of the acyltransferase that octanoylates ghrelin, an appetite-stimulating peptide hormone. Cell (2008) 132:387–96. doi:10.1016/j.cell.2008.01.017

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

62. Barnett BP, Hwang Y, Taylor MS, Kirchner H, Pfluger PT, Bernard V, et al. Glucose and weight control in mice with a designed ghrelin O-acyltransferase inhibitor. Science (2010) 330:1689–92. doi:10.1126/science.1196154

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

63. Delhanty PJ, Huisman M, Baldeon-Rojas LY, van den Berge I, Grefhorst A, Abribat T, et al. Des-acyl ghrelin analogs prevent high-fat-diet-induced dysregulation of glucose homeostasis. FASEB J (2013) 27:1690–700. doi:10.1096/fj.12-221143

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

64. Zhang W, Chai B, Li JY, Wang H, Mulholland MW. Effect of des-acyl ghrelin on adiposity and glucose metabolism. Endocrinology (2008) 149:4710–6. doi:10.1210/en.2008-0263

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

65. Baldanzi G, Filigheddu N, Cutrupi S, Catapano F, Bonissoni S, Fubini A, et al. Ghrelin and des-acyl ghrelin inhibit cell death in cardiomyocytes and endothelial cells through ERK1/2 and PI 3-kinase/AKT. J Cell Biol (2002) 159:1029–37. doi:10.1083/jcb.200207165

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

66. Skibicka KP, Dickson SL. Enteroendocrine hormones – central effects on behavior. Curr Opin Pharmacol (2013) 13:977–82. doi:10.1016/j.coph.2013.09.004

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

67. Ghosal S, Myers B, Herman JP. Role of central glucagon-like peptide-1 in stress regulation. Physiol Behav (2013) 122:201–7. doi:10.1016/j.physbeh.2013.04.003

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

68. McGovern SF, Hunter K, Holscher C. Effects of the glucagon-like polypeptide-1 analogue (Val8) GLP-1 on learning, progenitor cell proliferation and neurogenesis in the C57B/16 mouse brain. Brain Res (2012) 1473:204–13. doi:10.1016/j.brainres.2012.07.029

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Keywords: gut, obesity, brain, stress, gut peptides

Citation: Lee CY and Abizaid A (2014) The gut–brain-axis as a target to treat stress-induced obesity. Front. Endocrinol. 5:117. doi: 10.3389/fendo.2014.00117

Received: 29 April 2014; Accepted: 03 July 2014;
Published online: 18 July 2014.

Edited by:

Quentin Pittman, University of Calgary, Canada

Reviewed by:

Andreas Stengel, University of California Los Angeles, USA
Zane B. Andrews, Monash University, Australia

Copyright: © 2014 Lee and Abizaid. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: chooi.yeng.lee@monash.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.