Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Ghrelin: An orexigenic and somatotrophic signal from the stomach

Abstract

Ghrelin, an endogenous ligand for the growth hormone secretagogue receptor, is synthesized principally in the stomach and is released in response to fasting. Ghrelin is structurally related to motilin and, together, they represent a novel family of gut–brain regulatory peptides. In addition to having a powerful effect on the secretion of growth hormone, ghrelin stimulates energy production and signals directly to the hypothalamic regulatory nuclei that control energy homeostasis. The study of ghrelin has extended our understanding of how growth is controlled, and has shown that the stomach is an important component of this system.

Key Points

  • The identification of growth hormone secretagogues led to the discovery of a new receptor with homology to the motilin receptor. Subsequent studies led to the identification of its endogenous ligand — ghrelin. Ghrelin can indeed stimulate the release of growth hormone from the pituitary in a way that is independent of the action of growth-hormone-releasing hormone.

  • In addition to its effect on the release of growth hormone, ghrelin is an important regulator of food intake. It is released from the stomach in response to fasting, and increases feeding behaviour by acting on the arcuate nucleus of the hypothalamus.

  • The action of ghrelin is opposite to that of another important regulator of food intake — leptin. Leptin is released from adipose tissue, and its plasma levels decrease in response to fasting. This molecule also acts on the arcuate nucleus of the hypothalamus, where it has an anorexigenic effect.

  • The discovery of ghrelin has several clinical implications. It can be used to stimulate the release of growth hormone in cases of human deficiency, by acting on the endogenous oscillators that control pulsatile hormone release. It can also be used for the regulation of body weight by stimulating food intake, particularly in conditions accompanied by cachexia, such as cancer and AIDS.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Growth hormone secretagogues.
Figure 2: A new regulatory peptide system — the motilin–ghrelin family.
Figure 3: A simplified model of the action of ghrelin and leptin on feeding-regulatory circuitry.

Similar content being viewed by others

References

  1. Elmquist, J. K., Maratos-Flier, E., Saper, C. B. & Flier, J. S. Unraveling the central nervous system pathways underlying responses to leptin. Nature Neurosci. 1, 445–450 (1998).

    Article  CAS  PubMed  Google Scholar 

  2. Flier, J. S. & Maratos-Flier, E. Obesity and the hypothalamus: novel peptides for new pathways. Cell 92, 437–440 (1998).

    Article  CAS  PubMed  Google Scholar 

  3. Woods, S. C., Seeley, R. J., Porte, D. Jr & Schwartz, M. W. Signals that regulate food intake and energy homeostasis. Science 280, 1378–1383 (1998).

    Article  CAS  PubMed  Google Scholar 

  4. Bray, G. A. & York, D. A. The MONA LISA hypothesis in the time of leptin. Recent Prog. Horm. Res. 53, 95–117 (1998).

    CAS  PubMed  Google Scholar 

  5. Friedman, J. M. & Halaas, J. L. Leptin and the regulation of body weight in mammals. Nature 395, 763–770 (1998).

    Article  CAS  PubMed  Google Scholar 

  6. Inui, A. Feeding and body-weight regulation by hypothalamic neuropeptides — mediation of the actions of leptin. Trends Neurosci. 22, 62–67 (1999).

    Article  CAS  PubMed  Google Scholar 

  7. Kalra, S. P. et al. Interacting appetite-regulating pathways in the hypothalamic regulation of body weight. Endocr. Rev. 20, 68–100 (1999).

    CAS  PubMed  Google Scholar 

  8. Schwartz, M. W., Woods, S. C., Porte, D., Seeley, R. J. & Baskin, D. G. Central nervous system control of food intake. Nature 404, 661–671 (2000).A recent review that provides a summary of hypothalamic effector molecules and neuronal circuits that control food intake and are regulated by leptin and insulin.

    Article  CAS  PubMed  Google Scholar 

  9. Inui, A. Transgenic approach to the study of body weight regulation. Pharmacol. Rev. 52, 35–61 (2000).

    CAS  PubMed  Google Scholar 

  10. Inui, A. Transgenic study of energy homeostasis equation: implications and confounding influences. FASEB J. 14, 2158–2170 (2000).

    Article  CAS  PubMed  Google Scholar 

  11. Kojima, M. et al. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402, 656–660 (1999).Describes the isolation of ghrelin, an endogenous ligand of the growth hormone secretagogue (GHS) receptor, from stomach extracts.

    Article  CAS  PubMed  Google Scholar 

  12. Kojima, M., Hosoda, H., Matsuo, H. & Kangawa, K. Ghrelin: discovery of the natural endogenous ligand for the growth hormone secretagogue receptor. Trends Endocrinol. Metab. 12, 118–122 (2001).

    Article  CAS  PubMed  Google Scholar 

  13. Frohman, L. A. & Jansson, J. O. Growth hormone-releasing hormone. Endocr. Rev. 7, 223–253 (1986).

    Article  CAS  PubMed  Google Scholar 

  14. Frohman, L. A., Downs, T. R. & Chomczynski, P. Regulation of growth hormone secretion. Front. Neuroendocrinol. 13, 344–405 (1992).

    CAS  PubMed  Google Scholar 

  15. Smith, R. G. et al. Modulation of pulsatile GH release through a novel receptor in hypothalamus and pituitary gland. Recent Prog. Horm. Res. 51, 261–285 (1996).

    CAS  PubMed  Google Scholar 

  16. Frohman, L. A. et al. Secretagogues and the somatotrope: signaling and proliferation. Recent Prog. Horm. Res. 55, 269–291 (2000).

    CAS  PubMed  Google Scholar 

  17. Momany, F. A. et al. Design, synthesis, and biological activity of peptides which release growth hormone in vitro. Endocrinology 108, 31–39 (1981).

    Article  CAS  PubMed  Google Scholar 

  18. Bowers, C. Y. et al. Structure–activity relationships of a synthetic pentapeptide that specifically releases growth hormone in vitro. Endocrinology 106, 663–667 (1980).

    Article  CAS  PubMed  Google Scholar 

  19. Bowers, C. Y., Momany, F. A., Reynolds, G. A. & Hong, A. On the in vitro and in vivo activity of a new synthetic hexapeptide that acts on the pituitary to specifically release growth hormone. Endocrinology 114, 1537–1545 (1984).Reports the development of potent GHSs, including growth-hormone-releasing peptide 6 (GHRP-6).

    Article  CAS  PubMed  Google Scholar 

  20. Bowers, C. Y., Sartor, A. A., Reynolds, D. G. & Badger, T. A. M. On the actions of the growth hormone-releasing hexapeptide, GHRP-6. Endocrinology 128, 2027–2035 (1991).

    Article  CAS  PubMed  Google Scholar 

  21. Korbonits, M. & Grossman, A. Growth hormone-releasing peptide and its analogues: novel stimuli to growth hormone release. Trends Endocrinol. Metab. 6, 43–49 (1995).

    Article  CAS  PubMed  Google Scholar 

  22. Smith, R. G. et al. Peptidomimetic regulation of growth hormone secretion. Endocr. Rev. 18, 621–645 (1997).This review summarizes the GHS–GHS receptor system, its control of GH secretion and other physiological functions, as well as its clinical implications.

    Article  CAS  PubMed  Google Scholar 

  23. Casanueva, F. F. & Dieguez, C. Growth hormone secretagogues: physiological role and clinical utility. Trends Endocrinol. Metab. 10, 30–38 (1999).

    Article  CAS  PubMed  Google Scholar 

  24. Guillemin, R. et al. Growth hormone-releasing factor from a human pancreatic tumor that caused acromegaly. Science 218, 585–587 (1982).

    Article  CAS  PubMed  Google Scholar 

  25. Dieguez, C. & Casanueva, F. F. Ghrelin: a step forward in the understanding of somatotroph cell function and growth regulation. Eur. J. Endocrinol. 142, 413–417 (2000).

    Article  CAS  PubMed  Google Scholar 

  26. Patchett, A. A. et al. The design and biological activities of L-163,191 (MK-0677): a potent, orally active growth hormone secretagogue. Proc. Natl Acad. Sci. USA 92, 7001–7005 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Howard, A. D. et al. A receptor in pituitary and hypothalamus that functions in growth hormone release. Science 273, 974–977 (1996).The first demonstration of a complementary DNA that encodes the GHS receptor by the use of an expression cloning technique.

    Article  CAS  PubMed  Google Scholar 

  28. Dean, D. C. et al. Development of a high specific activity sulfur-35-labeled sulfonamide radioligand that allowed the identification of a new growth hormone secretagogue receptor. J. Med. Chem. 39, 1767–1770 (1996).

    Article  CAS  PubMed  Google Scholar 

  29. Pong, S. S. et al. Identification of a new G-protein-linked receptor for growth hormone secretagogues. Mol. Endocrinol. 10, 57–61 (1996).

    CAS  PubMed  Google Scholar 

  30. McKee, K. K. et al. Molecular analysis of rat pituitary and hypothalamic growth hormone secretagogue receptors. Mol. Endocrinol. 11, 415–423 (1997).

    Article  CAS  PubMed  Google Scholar 

  31. Guan, X. M. et al. Distribution of mRNA encoding the growth hormone secretagogue receptor in brain and peripheral tissues. Brain Res. Mol. Brain Res. 48, 23–29 (1997).

    Article  CAS  PubMed  Google Scholar 

  32. Chen, C., Wu, D. & Clarke, I. J. Signal transduction systems employed by synthetic GH-releasing peptides in somatotrophs. J. Endocrinol. 148, 381–386 (1996).

    Article  CAS  PubMed  Google Scholar 

  33. Wren, A. M. et al. The novel hypothalamic peptide ghrelin stimulates food intake and growth hormone secretion. Endocrinology 141, 4325–4328 (2000).

    Article  CAS  PubMed  Google Scholar 

  34. Takaya, K. et al. Ghrelin strongly stimulates growth hormone release in humans. J. Clin. Endocrinol. Metab. 85, 4908–4911 (2000).

    Article  CAS  PubMed  Google Scholar 

  35. Date, Y. et al. Central effects of a novel acylated peptide, ghrelin, on growth hormone release in rats. Biochem. Biophys. Res. Commun. 275, 477–480 (2000).

    Article  CAS  PubMed  Google Scholar 

  36. Seoane, L. M. Ghrelin elicites a marked stimulatory effect on GH secretion in freely-moving rats. Eur. J. Endocrinol. 143, R7–9 (2000).

    Article  CAS  PubMed  Google Scholar 

  37. Peino, R. et al. Ghrelin-induced growth hormone secretion in humans. Eur. J. Endocrinol. 143, R11–14 (2000).

    Article  CAS  PubMed  Google Scholar 

  38. Bednarek, M. A. et al. Structure–function studies on the new growth hormone-releasing peptide, ghrelin: minimal sequence of ghrelin necessary for activation of growth hormone secretagogue receptor 1a. J. Med. Chem. 43, 4370–4376 (2000).

    Article  CAS  PubMed  Google Scholar 

  39. Rehfeld, J. F. The new biology of gastrointestinal hormones. Physiol. Rev. 78, 1087–1108 (1998).

    Article  CAS  PubMed  Google Scholar 

  40. Hosoda, H., Kojima, M., Matsuo, H. & Kangawa, K. Purification and characterization of rat des-Gln14-ghrelin, a second endogenous ligand for the growth hormone secretagogue receptor. J. Biol. Chem. 275, 21995–22000 (2000).

    Article  CAS  PubMed  Google Scholar 

  41. Gualillo, O. et al. Ghrelin, a novel placental-derived hormone. Endocrinology 142, 788–794 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. Tullin, S. et al. Adenosine is an agonist of the growth hormone secretagogue receptor. Endocrinology 141, 3397–3402 (2000).

    Article  CAS  PubMed  Google Scholar 

  43. Smith, R. G. et al. Adenosine: a partial agonist of the growth hormone secretagogue receptor. Biochem. Biophys. Res. Commun. 276, 1306–1313 (2000).

    Article  CAS  PubMed  Google Scholar 

  44. Tomasetto, C. et al. Identification and characterization of a novel gastric peptide hormone: the motilin-related peptide. Gastroenterology 119, 395–405 (2000).Identifies a novel cDNA from the stomach that encodes a protein named prepromotilin-related peptide (on the basis of its sequence similarity to prepromotilin).

    Article  CAS  PubMed  Google Scholar 

  45. Itoh, Z. Motilin and clinical application. Peptides 18, 593–608 (1997).

    Article  CAS  PubMed  Google Scholar 

  46. Peeters, T. L. Erythromycin and other macrolides as prokinetic agents. Gastroenterology 105, 1886–1899 (1993).

    Article  CAS  PubMed  Google Scholar 

  47. Asakawa, A. et al. Ghrelin is an appetite-stimulatory signal from stomach with structual resemblance to motilin. Gastroenterology 120, 337–345 (2001).Ghrelin is the first appetite-stimulatory signal to be isolated from the stomach, and is the motilin-related peptide described in reference 44.

    Article  CAS  PubMed  Google Scholar 

  48. Del Rincon, J. P., Thorner, M. O. & Gaylinn, B. D. Motilin-related peptide and ghrelin: lessons from molecular techniques, peptide chemistry, and receptor biology. Gastroenterology 120, 587–588 (2001).

    Article  CAS  PubMed  Google Scholar 

  49. Coulie, B. J. & Miller, L. J. Identification of motilin-related peptide. Gastroenterology 120, 588–589 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Feighner, S. D. et al. Receptor for motilin identified in the human gastrointestinal system. Science 284, 2184–2188 (1999).

    Article  CAS  PubMed  Google Scholar 

  51. Thielemans, L., Depoortere, I., Assche, G. V., Bender, E. & Peeters, T. L. Demonstration of a functional motilin receptor in TE671 cells from human cerebellum. Brain Res. 895, 119–128 (2001).

    Article  CAS  PubMed  Google Scholar 

  52. Samson, W. K., Lumpkin, M. D., Nilaver, G. & McCann, S. M. Motilin: a novel growth hormone releasing agent. Brain Res. Bull. 12, 57–62 (1984).

    Article  CAS  PubMed  Google Scholar 

  53. Date, Y. et al. Ghrelin, a novel growth hormone-releasing acylated peptide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats and humans. Endocrinology 141, 4255–4261 (2000).

    Article  CAS  PubMed  Google Scholar 

  54. Hosoda, H., Kojima, M., Matsuo, H. & Kangawa, K. Ghrelin and des-acyl ghrelin: two major forms of rat ghrelin peptide in gastrointestinal tissue. Biochem. Biophys. Res. Commun. 279, 909–913 (2001).

    Article  CAS  Google Scholar 

  55. Depoortere, I., De Clercq, P., Svoboda, M., Bare, L. & Peeters, T. L. Identification of motilin mRNA in the brain of man and rabbit. Conservation of polymorphism of the motilin gene across species. Peptides 10, 1497–1503 (1997).

    Article  Google Scholar 

  56. Okada, K. et al. Intracerebroventricular administration of the growth hormone-releasing peptide KP-102 increases food intake in free-feeding rats. Endocrinology 137, 5155–5158 (1996).

    Article  CAS  PubMed  Google Scholar 

  57. Torsello, A. et al. Novel hexarelin analogs stimulate feeding in the rat through a mechanism not involving growth hormone release. Eur. J. Pharmacol. 360, 123–129 (1998).

    Article  CAS  PubMed  Google Scholar 

  58. Locke, W., Kirgis, H. D., Bowers, C. Y. & Abdoh, A. A. Intracerebroventricular growth-hormone-releasing peptide-6 stimulates eating without affecting plasma growth hormone responses in rats. Life Sci. 56, 1347–1352 (1995).

    Article  CAS  PubMed  Google Scholar 

  59. Ghigo, E. et al. Endocrine and non-endocrine activities of growth hormone secretagogues in humans. Horm. Res. 51, 9–15 (1999).

    CAS  PubMed  Google Scholar 

  60. Tschöp, M., Smiley, D. L. & Heiman, M. L. Ghrelin induces adiposity in rodents. Nature 407, 908–913 (2000).Indicates an involvement of ghrelin in the regulation of energy balance by showing that ghrelin causes weight gain by reducing fat use.

    Article  PubMed  Google Scholar 

  61. Nakazato, M. et al. A role for ghrelin in the central regulation of feeding. Nature 409, 194–198 (2001).Ghrelin is a physiological mediator of feeding and acts through neuropeptide Y and agouti-related protein in the hypothalamus.

    Article  CAS  PubMed  Google Scholar 

  62. Shintani, M. et al. Ghrelin, an endogenous growth hormone secretagogue, is a novel orexigenic peptide that antagonizes leptin action through the activation of hypothalamic neuropeptide Y/Y1 receptor pathway. Diabetes 50, 227–232 (2001).

    Article  CAS  PubMed  Google Scholar 

  63. Hewson, A. K. & Dickson, S. L. Systemic administration of ghrelin induces Fos and Egr-1 proteins in the hypothalamic arcuate nucleus of fasted and fed rats. J. Neuroendocrinol. 12, 1047–1049 (2000).

    Article  CAS  PubMed  Google Scholar 

  64. Hirosue, Y. et al. Cholecystokinin octapeptide analogues suppress food intake via central CCK-A receptors in mice. Am. J. Physiol. 265, R481–486 (1993).

    CAS  PubMed  Google Scholar 

  65. Smith, G. P. & Gibbs, J. Satiating effect of cholecystokinin. Ann. NY Acad. Sci. 713, 236–241 (1994).

    Article  CAS  PubMed  Google Scholar 

  66. Moran, T. H. Cholecystokinin and satiety: current perspectives. Nutrition 16, 858–865 (2000).

    Article  CAS  PubMed  Google Scholar 

  67. Schwartz, G. J. The role of gastrointestinal vagal afferents in the control of food intake: current prospects. Nutrition 16, 866–873 (2000).

    Article  CAS  PubMed  Google Scholar 

  68. Yoshida-Yoneda, E., O-Lee, T. J., Wei, J. Y., Vigna, S. R. & Tache, Y. Peripheral bombesin induces gastric vagal afferent activation in rats. Am. J. Physiol. 271, R1584–1593 (1996).

    CAS  PubMed  Google Scholar 

  69. Kurosawa, M., Uvnas, M. K., Miyasaka, K. & Lundeberg, T. Interleukin-1 increases activity of the gastric vagal afferent nerve partly via stimulation of type A CCK receptor in anesthetized rats. J. Auton. Nerv. Syst. 12, 72–78 (1997).

    Article  Google Scholar 

  70. Wang, Y. H., Tache, Y., Sheibel, A. B., Go, V. L. & Wei, J. Y. Two types of leptin-responsive gastric vagal afferent terminals: an in vitro single-unit study in rats. Am. J. Physiol. 273, R833–837 (1997).

    CAS  PubMed  Google Scholar 

  71. Masuda, Y. et al. Ghrelin stimulates gastric acid secretion and motility in rats. Biochem. Biophys. Res. Commun. 276, 905–908 (2000).

    Article  CAS  PubMed  Google Scholar 

  72. Morley, J. E. Neuropeptide regulation of appetite and weight. Endocr. Rev. 8, 256–287 (1987).

    Article  CAS  PubMed  Google Scholar 

  73. Zhang, Y. et al. Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425–432 (1994).

    Article  CAS  PubMed  Google Scholar 

  74. Baskin, D. G., Breininger, J. F. & Schwartz, M. W. Leptin receptor mRNA identifies a subpopulation of neuropeptide Y neurons activated by fasting in rat hypothalamus. Diabetes 48, 828–833 (1999).

    Article  CAS  PubMed  Google Scholar 

  75. Cheung, C. C., Clifton, D. K. & Steiner, R. A. Proopiomelanocortin neurons are direct targets for leptin in the hypothalamus. Endocrinology 138, 4489–4492 (1997).

    Article  CAS  PubMed  Google Scholar 

  76. Campfield, L. A., Smith, F. J., Guisez, Y., Devos, R. & Burn, P. Recombinant mouse OB protein: evidence for a peripheral signal linking adiposity and central neural networks. Science 269, 546–549 (1995).

    Article  CAS  PubMed  Google Scholar 

  77. Schwartz, M. W., Figlewicz, D. P., Baskin, D. G., Woods, S. C. & Porte, D. Jr Insulin in the brain: a hormonal regulator of energy balance. Endocr. Rev. 13, 387–414 (1992).

    CAS  PubMed  Google Scholar 

  78. Ahima, R. S. & Flier, J. S. Leptin. Annu. Rev. Physiol. 62, 413–437 (2000).

    Article  CAS  PubMed  Google Scholar 

  79. Sawchenko, P. E. Toward a new neurobiology of energy balance, appetite, and obesity: the anatomists weigh in. J. Comp. Neurol. 402, 435–441 (1998).

    Article  CAS  PubMed  Google Scholar 

  80. Elmquist, J. K., Elias, C. F. & Saper, C. B. From lesions to leptin: hypothalamic control of food intake and body weight. Neuron 22, 221–232 (1999).

    Article  CAS  PubMed  Google Scholar 

  81. Satoh, N. et al. The arcuate nucleus as a primary site of satiety effect of leptin in rats. Neurosci. Lett. 224, 149–152 (1997).

    Article  CAS  PubMed  Google Scholar 

  82. Tang-Christensen, M., Holst, J. J., Hartmann, B. & Vrang, N. The arcuate nucleus is pivotal in mediating the anorectic effects of centrally administered leptin. Neuroreport 10, 1183–1187 (1999).

    Article  CAS  PubMed  Google Scholar 

  83. Dawson, R., Pelleymounter, M. A., Millard, W. J., Liu, S. & Eppler, B. Attenuation of leptin-mediated effects by monosodium glutamate-induced arcuate nucleus damage. Am. J. Physiol. 273, E202–206 (1997).

    CAS  PubMed  Google Scholar 

  84. Tartaglia, L. A. et al. Identification and expression cloning of a leptin receptor, OB-R. Cell 83, 1263–1271 (1995).

    Article  CAS  PubMed  Google Scholar 

  85. Tartaglia, L. A. The leptin receptor. J. Biol. Chem. 272, 6093–6096 (1997).

    Article  CAS  PubMed  Google Scholar 

  86. Hahn, T. M., Breininger, J. F., Baskin, D. G. & Schwartz, M. W. Coexpression of Agrp and NPY in fasting-activated hypothalamic neurons. Nature Neurosci. 1, 271–272 (1998).

    Article  CAS  PubMed  Google Scholar 

  87. Broberger, C., Johansen, J., Johansson, C., Schalling, M. & Hökfelt, T. The neuropeptide Y/agouti gene-related protein (AGRP) brain circuitry in normal, anorectic, and monosodium glutamate-treated mice. Proc. Natl Acad. Sci. USA 95, 15043–15048 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Elias, C. F. et al. Leptin activates hypothalamic CART neurons projecting to the spinal cord. Neuron 21, 1375–1385 (1998).

    Article  CAS  PubMed  Google Scholar 

  89. Ollmann, M. M. et al. Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science 278, 135–138 (1997).

    Article  CAS  PubMed  Google Scholar 

  90. Shutter, J. R. et al. Hypothalamic expression of ART, a novel gene related to agouti, is up-regulated in obese and diabetic mutant mice. Genes Dev. 11, 593–602 (1997).

    Article  CAS  PubMed  Google Scholar 

  91. Fan, W., Boston, B. A., Kesterson, R. A., Hruby, V. J. & Cone, R. D. Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature 385, 165–168 (1997).

    Article  CAS  PubMed  Google Scholar 

  92. Stephens, T. W. et al. The role of neuropeptide Y in the antiobesity action of the obese gene product. Nature 377, 530–532 (1995).

    Article  CAS  PubMed  Google Scholar 

  93. Kristensen, P. et al. Hypothalamic CART is a new anorectic peptide regulated by leptin. Nature 393, 72–76 (1998).

    Article  CAS  PubMed  Google Scholar 

  94. Schwartz, M. W. et al. Leptin increases hypothalamic pro-opiomelanocortin mRNA expression in the rostral arcuate nucleus. Diabetes 46, 2119–2123 (1997).

    Article  CAS  PubMed  Google Scholar 

  95. Thornton, J. E., Cheung, C. C., Clifton, D. K., & Steiner, R. A. Regulation of hypothalamic proopiomelanocortin mRNA by leptin in ob/ob mice. Endocrinology 138, 5063–5066 (1997).

    Article  CAS  PubMed  Google Scholar 

  96. Cowley, M. A. et al. Integration of NPY, AGRP, and melanocortin signals in the hypothalamic paraventricular nucleus: evidence of a cellular basis for the adipostat. Neuron 24, 155–163 (1999).

    Article  CAS  PubMed  Google Scholar 

  97. Kalra, S. P., Dube, M. G., Sahu, A., Phelps, C. P. & Kalra, P. S. Neuropeptide Y secretion increases in the paraventricular nucleus in association with increased appetite for food. Proc. Natl Acad. Sci. USA 88, 10931–10935 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Dickson, S. L., Leng, G. & Robinson, I. C. Systemic administration of growth hormone-releasing peptide activates hypothalamic arcuate neurons. Neuroscience 53, 303–306 (1993).

    Article  CAS  PubMed  Google Scholar 

  99. Kamegai, J., Hasegawa, O., Minami, S., Sugihara, H. & Wakabayashi, I. The growth hormone-releasing peptide KP-102 induces c-fos expression in the arcuate nucleus. Brain Res. Mol. Brain Res. 39, 153–159 (1996).

    Article  CAS  PubMed  Google Scholar 

  100. Willesen, M. G., Kristensen, P. & Romer, J. Co-localization of growth hormone secretagogue receptor and NPY mRNA in the arcuate nucleus of the rat. Neuroendocrinology 70, 306–316 (1999).

    Article  CAS  PubMed  Google Scholar 

  101. Kamegai, J. et al. Central effect of ghrelin, an endogenous growth hormone secretagogue, on hypothalamic peptide gene expression. Endocrinology 141, 4797–4800 (2000).

    Article  CAS  PubMed  Google Scholar 

  102. Torsello, A. et al. Differential orexigenic effects of hexarelin and its analogs in the rat hypothalamus: indication for multiple growth hormone secretagogue receptor subtypes. Neuroendocrinology 72, 327–332 (2000).

    Article  CAS  PubMed  Google Scholar 

  103. Inui, A. Neuropeptide Y feeding receptors: are multiple subtypes involved? Trends Pharmacol. Sci. 20, 43–46 (1999).

    Article  CAS  PubMed  Google Scholar 

  104. Herzog, H. Y4 receptor deletion improves fertility in ob/ob mice without affecting the obese phenotype. Proc. VIth Int. Neuropeptide Y Conf. 06 (2001).

  105. Naveilhan, P. et al. Normal feeding behavior, body weight and leptin response require the neuropeptide Y Y2 receptor. Nature Med. 5, 1188–1193 (1999).

    Article  CAS  PubMed  Google Scholar 

  106. Sainsbury, A., Couzens, M., Lui, M. & Herzog, H. Critical role of Y2 receptors in body weight regulation revealed by adult-induced arcuate-specific gene deletion. Proc. VIth Int. Neuropeptide Y Conf. 014 (2001).

  107. Hagan, M. M. et al. Long-term orexigenic effects of AGRP-(83-132) involve mechanisms other than melanocortin receptor blockade. Am. J. Physiol. 279, R47–52 (2000).

    CAS  Google Scholar 

  108. Hagan, M. M., Rushing, P. A., Benoit, S. C., Woods, S. C. & Seeley, R. J. Opioid receptor involvement in the effect of AgRP- (83-132) on food intake and food selection. Am. J. Physiol. 280, R814–821 (2001).

    Article  CAS  Google Scholar 

  109. Erickson, J. C., Clegg, K. E. & Palmiter, R. D. Sensitivity to leptin and susceptibility to seizures of mice lacking neuropeptide Y. Nature 381, 415–421 (1996).

    Article  CAS  PubMed  Google Scholar 

  110. Kohno, D. & Yada, T. Ghrelin increases cytosolic Ca2+ in the hypothalamic arcuate nucleus neurons. Jpn. J. Physiol. 51 (in the press).

  111. Cowley, M. A. et al. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 411, 480–484 (2001).

    Article  CAS  PubMed  Google Scholar 

  112. Toshinai, K. et al. Upregulation of ghrelin expression in the stomach upon fasting, insulin-induced hypoglycemia, and leptin administration. Biochem. Biophys. Res. Commun. 281, 1220–1225 (2001).

    Article  CAS  PubMed  Google Scholar 

  113. Bado, A. et al. The stomach is a source of leptin. Nature 394, 790–793 (1998).Reports the presence of leptin in the stomach; this hormone is probably involved in CCK-mediated effects, including satiety.

    Article  CAS  PubMed  Google Scholar 

  114. Barrachina, M. D., Martinez, V., Wang, L., Wei, J. Y. & Tache, Y. Synergistic interaction between leptin and cholecystokinin to reduce short-term food intake in lean mice. Proc. Natl Acad. Sci. USA 94, 10455–10460 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Matson, C. A. & Ritter, R. C. Long-term CCK–leptin synergy suggests a role for CCK in the regulation of body weight. Am. J. Physiol. 276, R1038–1045 (1999).

    Article  CAS  PubMed  Google Scholar 

  116. Tolle, V. et al. In vivo and in vitro effects of ghrelin/motilin-related peptide on growth hormone secretion in the rat. Neuroendocrinology 73, 54–61 (2001).

    Article  CAS  PubMed  Google Scholar 

  117. Minami, S., Kamegai, J., Sugihara, H., Suzuki, N. & Wakabayashi, I. Growth hormone inhibits its own secretion by acting on the hypothalamus through its receptors on neuropeptide Y neurons in the arcuate nucleus and somatostatin neurons in the periventricular nucleus. Endocr. J. 45, S19–26 (1998).

    Article  CAS  PubMed  Google Scholar 

  118. Bowers, C. Y. Unnatural growth hormone-releasing peptide begets natural ghrelin. J. Clin. Endocrinol. Metab. 86, 1464–1469 (2001).

    Article  CAS  PubMed  Google Scholar 

  119. Korbonits, M. et al. The expression of the growth hormone secretagogue receptor ligand ghrelin in normal and abnormal human pituitary and other neuroendocrine tumors. J. Clin. Endocrinol. Metab. 86, 881–887 (2001).

    CAS  PubMed  Google Scholar 

  120. Chapman, I. M. et al. Stimulation of the growth hormone (GH)-insulin-like growth factor-I axis by daily oral administration of a GH secretagogue (MK-0677) in healthy elderly subjects. J. Clin. Endocrinol. Metab. 81, 4249–4257 (1996).

    CAS  PubMed  Google Scholar 

  121. Ghigo, E. et al. Biologic activities of growth hormone secretagogues in humans. Endocrine 14, 87–93 (2001).

    Article  CAS  PubMed  Google Scholar 

  122. Dieguez, C. & Casanueva, F. F. Influence of metabolic substrates and obesity on growth hormone secretion. Trends Endocrinol. Metab. 6, 55–59 (1995).

    Article  CAS  PubMed  Google Scholar 

  123. Tschöp, M. et al. Circulating ghrelin levels are decreased in human obesity. Diabetes 50, 707–709 (2001).

    Article  PubMed  Google Scholar 

  124. Levin, B. E. & Keesey, R. E. Defense of differing body weight set points in diet-induced obese and resistant rats. Am. J. Physiol. 274, R412–419 (1998).

    CAS  PubMed  Google Scholar 

  125. Levin, B. E. Arcuate NPY neurons and energy homeostasis in diet-induced obese and resistant rats. Am. J. Physiol. 276, R382–387 (1999).

    CAS  PubMed  Google Scholar 

  126. Kaga, T. et al. Modest overexpression of NPY in the brain leads to obesity after high sucrose feeding. Diabetes 50, 1206–1210 (2001).

    Article  CAS  PubMed  Google Scholar 

  127. Ziotopoulou, M., Mantzoros, C. S., Hileman, S. M. & Flier, J. S. Differental expression of hypothalamic neuropeptides in the early phase of diet-induced obesity in mice. Am. J. Physiol. 279, E838–845 (2000).

    CAS  Google Scholar 

  128. Flier, J. S. Clinical review 94: what's in a name? In search of leptin's physiologic role. J. Clin. Endocrinol. Metab. 83, 1407–1413 (1998).

    CAS  PubMed  Google Scholar 

  129. Ahima, R. S. et al. Role of leptin in the neuroendocrine response to fasting. Nature 382, 250–252 (1996).

    Article  CAS  PubMed  Google Scholar 

  130. Bray, G. A. & York, D. A. Hypothalamic and genetic obesity in experimental animals: an autonomic and endocrine hypothesis. Physiol. Rev. 59, 719–790 (1979).

    Article  CAS  PubMed  Google Scholar 

  131. Schwartz, M. W., Dallman, M. F. & Woods, S. C. Hypothalamic response to starvation: implications for the study of wasting disorders. Am. J. Physiol. 269, R949–957 (1995).

    CAS  PubMed  Google Scholar 

  132. Inui, A. Cancer anorexia–cachexia syndrome: are neuropeptides the key? Cancer Res. 59, 4493–4501 (1999).

    CAS  PubMed  Google Scholar 

  133. Plata-Salaman, C. R. Central nervous system mechanisms contributing to the cachexia–anorexia syndrome. Nutrition 16, 1009–1012 (2000).

    Article  CAS  PubMed  Google Scholar 

  134. Inui, A. Cytokine and sickness behavior: implications from knockout animal models. Trends Immunol. (in the press).

  135. Gayle, D., Ilyin, S. E. & Plata-Salaman, C. R. Central nervous system IL-1 beta system and neuropeptide Y mRNAs during IL-1 beta-induced anorexia in rats. Brain Res. Bull. 44, 311–317 (1997).

    Article  CAS  PubMed  Google Scholar 

  136. Welle, S. Growth hormone and insulin-like growth factor-I as anabolic agents. Curr. Opin. Clin. Nutr. Metab. Care 1, 257–262 (1998).

    Article  CAS  PubMed  Google Scholar 

  137. Van den Berghe, G. Novel insights into the neuroendocrinology of critical illness. Eur. J. Endocrinol. 143, 1–13 (2000).

    Article  CAS  PubMed  Google Scholar 

  138. Murphy, M. G. et al. MK-677, an orally active growth hormone secretagogue, reverses diet-induced catabolism. J. Clin. Endocrinol. Metab. 83, 320–325 (1998).

    CAS  PubMed  Google Scholar 

  139. Van den Berghe, G. et al. Neuroendocrinology of prolonged critical illness: effects of exogenous thyrotropin-releasing hormone and its combination with growth hormone secretagogues. J. Clin. Endocrinol. Metab. 83, 309–319 (1998).

    CAS  PubMed  Google Scholar 

  140. Hansen, B. S. et al. Pharmacological characterisation of a new oral GH secretagogue, NN703. Eur. J. Endocrinol. 141, 180–189 (1999).

    Article  CAS  PubMed  Google Scholar 

  141. Leibowitz, S. F. Brain peptides and obesity: pharmacologic treatment. Obes. Res. 3, 573S–589S (1995).

    Article  CAS  PubMed  Google Scholar 

  142. Levine, A. S. & Billington, C. J. Obesity: progress through genetic manipulation. Curr. Biol. 8, R251–252 (1998).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

I sincerely thank M. Kasuga and S. Baba of Kobe University, and M. Nakazato of Miyazaki Medical College, for stimulating discussions. The work was supported by grants from the Japanese Ministry of Education, Science, Sports, and Culture.

Author information

Authors and Affiliations

Authors

Related links

Related links

DATABASE LINKS

leptin

insulin

ghrelin

GHS receptor

GH

GHRH

somatostatin

SSTR2

SSTR5

phospholipase C

gastrin

CCK

IGF1

motilin

motilin receptor

NPY

AGRP

MCH

galanin

Fos

Egr1

IL1β

GRP

POMC

CART

MC4 receptor

orexin/hypocretin

Y1

Y5

Y4

PP

Y2

Glossary

SOMATOTROPHS

Cells of the anterior pituitary that are responsible for the production of growth hormone or somatotropin. Somatotroph adenomas cause acromegaly.

HYPOTHALAMIC–HYPOPHYSEAL PORTAL SYSTEM

A system of blood vessels (portal veins) that links the capillaries of the hypothalamus with those of the pituitary.

MOTILIN

A 22-amino-acid peptide produced in the mucosa of the small intestine, which stimulates contractions of the stomach and the release of pepsin.

OXYNTIC MUCOSA

The acid-secreting parietal cells of the gastric mucosa.

ENTEROCHROMAFFIN CELLS

Endocrine cells in the gastric mucosa, which synthesize and secrete histamine in response to stimulation by the hormone gastrin.

PEPTIDE YY

A protein found in the small intestine, which inhibits molecule secretion from the exocrine pancreas.

CIONIN

A molecule from the protochordate Ciona intestinalis that represents the oldest true member of the cholecystokinin/gastrin family.

CERULEIN

A member of the cholecystokinin/gastrin family isolated from the frog Hyla caerulea. It stimulates the secretion of gastric acid in the small intestine of several amphibians.

RESPIRATORY QUOTIENT

The ratio of the volume of CO2 released to the volume of O2 consumed by a body tissue or an organism. The oxidation of carbohydrate results in a respiratory quotient of 1.0, whereas the oxidation of fat results in a quotient of 0.7. So, an increase in the respiratory quotient is sometimes related to increased use of carbohydrate and reduced use of fat to meet specific energy requirements.

AGOUTI-RELATED PEPTIDE

An antagonist of melanocortin receptors that has an important role in the control of food intake. Administration and overexpression of this peptide result in hyperphagia and weight gain.

OREXINS

Peptides associated with feeding behaviour that have also been associated with disturbances of sleep such as narcolepsy.

BOMBESIN

A strong releaser of gastrin and cholecystokinin, which is found in the gut and the brain. It also has a mitogenic action in several cell types.

PANCREATIC POLYPEPTIDE

A molecule, first detected as an impurity present in insulin preparations, which is released after food intake and seems to act as a regulator of pancreatic and gastrointestinal functions.

ACROMEGALY

A disease characterized by the gradual enlargement of the bones of hands, feet, head and chest, and thickening of the skin, lips and vocal chords. It is caused by increased sensitivity to or increased production of growth hormone.

SOMATOPAUSE

A phase in the human lifespan during which the levels of growth hormone are reduced, resulting in the dehydration of cells and organs, and a reduction in their size and function. Somatopause usually begins in people in their mid-forties, but can start earlier.

CACHEXIA

A condition caused by chronic diseases such as cancer, which is characterized by wasting, emaciation, feebleness and inanition.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Inui, A. Ghrelin: An orexigenic and somatotrophic signal from the stomach. Nat Rev Neurosci 2, 551–560 (2001). https://doi.org/10.1038/35086018

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/35086018

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing