Top-Rated Free Essay
Preview

Energy Policy

Powerful Essays
7518 Words
Grammar
Grammar
Plagiarism
Plagiarism
Writing
Writing
Score
Score
Energy Policy
0013-7227/01/$03.00/0 Printed in U.S.A.

Endocrinology 142(10):4163– 4169 Copyright © 2001 by The Endocrine Society

Minireview: Ghrelin and the Regulation of Energy Balance—A Hypothalamic Perspective
TAMAS L. HORVATH, SABRINA DIANO, PETER SOTONYI, MARK HEIMAN, ¨ MATTHIAS TSCHOP
AND

Reproductive Neuroscience Unit (T.L.H., S.D.), Department of Obstetrics and Gynecology and Department of Neurobiology (T.L.H.), Yale Medical School, New Haven, Connecticut 06520; Department of Anatomy and Histology (P.S.), Faculty of Veterinary Science, Szent Istvan University, Budapest, Hungary 1400; and Lilly Research Laboratories (M.H., M.T.), Eli Lilly & Co., Indianapolis, Indiana 46285
The recently discovered hormone, ghrelin, has been recognized as an important regulator of GH secretion and energy homeostasis. Orexigenic and adipogenic ghrelin is produced by the stomach, intestine, placenta, pituitary, and possibly in the hypothalamus. The concentration of circulating ghrelin, principally derived from the stomach, is influenced by acute and chronic changes in nutritional state. To date, most studies focused on the role of ghrelin in GH secretion or its function in complementing leptin action to prevent energy deficits. The potential significance of ghrelin in the etiology of obesity and cachexia as well as in the regulation of growth processes is the subject of ongoing discussions. A large quantity of information based on clinical trials and experimental studies with ghrelin and previously available synthetic ghrelin receptor agonists (GH secretagogues) must now be integrated with a rapidly increasing amount of data on the central regulation of metabolism and appetite. In this overview, we summarize recent findings and strategies on the integration of ghrelin into neuroendocrine networks that regulate energy homeostasis. (Endocrinology 142: 4163– 4169, 2001)

O

BESITY AND RELATED disorders are among the leading causes of illness and mortality in the developed world (1). To better understand the pathophysiological mechanisms that underlie metabolic disorders, increasing attention has been paid to central regulatory elements in energy homeostasis, including food intake and energy expenditure (2–5). The past two decades have provided overwhelming evidence of the critical role that hypothalamic peptidergic systems play in the central regulation of appetite and metabolism (6, 7). The discovery of ghrelin (8 –11) and its influence on appetite, fuel utilization, body weight, and body composition that is complementary to ghrelin’s GHreleasing effect (12) adds yet another component to the complexity in the central regulation of energy balance.
Discovery of Ghrelin

Reverse pharmacology may be an appropriate term to describe the road to ghrelin’s discovery. First, synthetic agonists with ghrelin-like activity [GH-releasing peptides (GHRPs) and GH secretagogues (GHSs)] were discovered by Bowers and co-workers in the late seventies (9, 13–15), followed by the cloning of ghrelin-GHS-receptor (GHS-R) in 1996 by Smith and co-workers (16 –18). Subsequently, the elegant studies by Kojima and co-workers led to the identification of an acylated 28 residue peptide as an endogenous bioactive ligand for the GHS-R (8 –11). It was called ghrelin, a term that contains “ghre-” as the etymological root for “growth” in many languages. “GH” and “relin,” a suffix for releasing substances in generic names according to the USP
Abbreviations: AGRP, Agouti-related protein; GHRP, GH-releasing peptide; GHS, GH-secretagogue; GHS-R, GHS receptor.

Dictionary of USAN and International Drug Names, also represents an abbreviation for “growth-hormone-release,” a characteristic effect of ghrelin (8, 11). The Ser3-acylation that seems to be responsible for bioactivity of ghrelin is a modification that has been observed for the first time in mammalian physiology. There are no data to support the tempting speculation that the purpose of this modification is to increase ghrelin’s lipophilic properties to facilitate transport across the blood brain barrier. However, the octanoyl side chain is essential for binding and activation of the GHS-R subtype-1a in vitro (19). Ghrelin might also bind to different GHS-R subtypes or receptor families where the octanoyl side chain is not needed. Detection and purification of the gastric enzyme responsible for the acylation of ghrelin may shed light on this fascinating question and will possibly even reveal the existence of other putative hormones carrying this modification.
Sources of Ghrelin

Ghrelin is predominantly produced by the stomach (8, 20 –22), whereas substantially lower amounts are derived from bowel (21, 22), pituitary (23), kidney (24), placenta (25), and hypothalamus (8, 22). Although the majority of circulating ghrelin is produced in the stomach, other sources may increase ghrelin secretion in a compensatory manner. After gastrectomy, for example, plasma ghrelin level is surprisingly reduced only by 65% (26). One of the most urgent and debatable hypotheses is whether ghrelin is produced in physiologically relevant amounts in the hypothalamus. This unresolved issue is the focus of several research groups. Data published or presented at recent meetings, in accordance with our own on-

4163

4164

Endocrinology, October 2001, 142(10):4163– 4169

Horvath et al. • Ghrelin and Regulation of Energy Balance

going investigations, have shown ghrelin to be present in several regions of the hypothalamus through the use of immunohistochemical detection methods (8, 22). Depending on the ghrelin epitope recognized by the antibody in use, ghrelin-positive cells have been identified in varying hypothalamic areas, whereas all of these antisera have been successfully used in RIAs. Detection of hypothalamic ghrelin mRNA by use of PCR has been accomplished (8); however, this issue needs to be further investigated by the detection and regional distribution of ghrelin mRNA by in situ hybridization. At this point, it is not clear if the detection of ghrelin in the hypothalamus using immunohistochemistry reflects ghrelin peptide that is produced by hypothalamic neurons or ghrelin peptide that is derived from the stomach. It seems logical that gastric ghrelin reflects an acute nutritional state. However, even minimal ghrelin expression in the hypothalamus (8, 12) or circulating placental ghrelin during pregnancy (25) may significantly influence food intake, nutrition partitioning, and fat utilization. Regardless of the source, in the end it is most likely the modulation of hypothalamic circuits by ghrelin that mediates changes in energy homeostasis.
Ghrelin and GH Secretion

Based on rodent experiments (27–29) and clinical studies (30 –33), it is evident that ghrelin is indeed a potent GHreleasing agent. However, no significant correlation seems to exist between plasma ghrelin concentrations and circulating levels of GH or IGF-I (unpublished data, Tschop et al.) even ¨ though both ghrelin and GH increase during fasting (34, 35). Very recent data indicate that most of the ghrelin-induced GH secretion is not only directly opposed by somatostatin action, but also involves mediation through GHRH (33, 36, 37). However, ghrelin also releases GH in vitro from primary rat pituitary cells (8, 12), and GHRP-2, a potent ghrelin receptor agonist, releases GH in vivo in patients with GHRH receptor mutations (38). This indicates the existence of GHRH-independent effects of ghrelin on GH secretion mediated by hypophyseal GHS-Rs, which were originally cloned from the pituitary (16). Alternatively, ghrelin may stimulate an unidentified hypothalamic agent (U-factor) that, in turn, stimulates GH release (39).
Ghrelin and Energy Balance

The first published evidence for the involvement of ghrelin in the regulation of appetite was provided by Ghigo and co-workers (30). They described that 3 out of 4 healthy volunteers spontaneously reported hunger following ghrelin administration as a “side effect” in a clinical study analyzing GH release (30). This hunger-inducing effect of ghrelin has now been confirmed in two more studies, where, again, 3 out of 7 (33) and 9 out of 11 individuals report hunger as the only sensation after ghrelin injection (40). A large number of animal studies added strength to the argument that ghrelin is involved in the regulation of energy balance. For example, exogenous ghrelin induces adiposity in rodents by stimulating an acute increase in food intake, as well as a reduction in fat utilization (12, 41– 46). Adipogenic as well as orexigenic effects of ghrelin are independent from its ability to stimulate

GH secretion (12, 46) and are most likely mediated by a specific central network of neurons that is also modulated by leptin (2–7, 9, 12, 41– 46). Regulation of ghrelin secretion, as well as its biological effects, appear to be opposite those of leptin. However, from a teleological point of view, ghrelin and leptin might really be complementary players of one regulatory system that has developed to inform the central nervous system about the current status of acute and chronic energy balance (12, 38 – 49). In addition, a specific role for ghrelin might be to ensure the provision of calories that GH requires for growth and repair (41). In humans, circulating ghrelin levels are decreased in chronic (obesity) (48) and acute (caloric intake) (26, 34, 47) states of positive energy balance, whereas plasma levels of ghrelin are increased by fasting (12, 34) and in cachectic patients with anorexia nervosa (26). Of course, it has yet to be proven that the rather modest changes in circulating ghrelin, in the 100 fmol range, have physiological relevance for hypothalamic receptor sites. One plausible explanation is that if ghrelin is indeed a hormone signaling the need to conserve energy (12), ghrelin secretion is triggered to counter further deficit of energy storage and to prevent starvation or cachexia. A very recent study shows a pre-meal rise of human plasma ghrelin, suggesting a possible role of ghrelin as a hunger signal triggering meal initiation (34). In rodents, fasting and hypoglycemia increase ghrelin levels, whereas intake of food, especially carbohydrates (dextrose), decreases ghrelin secretion (12, 41, 50). We speculate that this obvious connection between glucose levels, ghrelin secretion and GH secretion is likely to be involved in the physiological mechanism of diagnostic procedures such as oral glucose tolerance testing (for acromegaly) and insulin tolerance testing (for GH deficiency). Differential effects of ghrelin might be mediated by separate ghrelin (GHS-R) subtypes as recently suggested by Thorner and co-workers (51). Based on a series of elaborate studies using GHS-R antagonists ([d-Lys3]GHRP-6 and BMS-265711, also an NPY-antagonist) and an NPY-Y1-R antagonist ([d-Trp32]NPY), they showed that the orexigenic effect of ghrelin can be dissociated from its GH releasing effects, suggesting distinct GHS-R-subtypes. Based on the observation of differential orexigenic effects of hexarelin and its analogs and GH secretagogue actions at the pituitary gland (52, 53), the existence of additional subtypes of the GHS-R (16 –18) had previously been hypothesized. The putative adipogenic effects of ghrelin in humans remains to be shown because it is possible that ghrelin has different effects on energy balance in humans and rodents. In addition, ghrelin-induced adiposity could be only a transient effect and the therapeutic potential of ghrelin in cachectic humans might therefore turn out to be as disappointing as the efficacy of leptin for the therapy of human obesity (5, 54). Carefully conducted clinical studies focusing on body composition as well as long-term studies on ghrelin treatment in rodents are necessary to further address this question.
Ghrelin and Brain Centers of Energy Balance

Our current understanding of the involvement of different hypothalamic systems in metabolic regulation arises from early degeneration studies in rats. Destruction of distinct

Horvath et al. • Ghrelin and Regulation of Energy Balance

Endocrinology, October 2001, 142(10):4163– 4169 4165

hypothalamic regions, particularly the ventromedial nucleus but also the areas of the paraventricular and dorsomedial nuclei, induced hyperphagia (55– 60). In contrast, discrete lesions placed in the lateral hypothalamus (61, 62) reduced food intake. During the last two decades, a substantial amount of research demonstrated that NPY, administered into the cerebral ventricles (63) or other specific hypothalamic sites (64), induced food intake. However, in addition to NPY, several other hypothalamic peptides were found to affect appetite and feeding behavior (for details see Refs. 2–7). Appetite- stimulating neuropeptides include melanin concentrating hormone, hypocretins/orexins (produced in a distinct subset of neurons of the lateral hypothalamus perifornical region) (65– 68) and agouti-related protein (AGRP, coproduced with NPY in the same arcuate nucleus neurons) (69 –71). Appetite-suppressing neuropeptides include the POMC derivate, -MSH (6, 7, 72) that is produced in arcuate nucleus perikarya (73). An important milestone to link the central regulation of metabolism with peripheral levels of energy storage was the discovery of the adipose hormone, leptin. Genetic mouse or rat mutants, including db/db and ob/ob mice and fa/fa rats become strikingly obese. Molecular analysis has shown that the primary genetic defect in these animals relates to either abolished leptin production (ob/ob mice) or impaired leptin receptors (db/db mice; fa/fa rats; leptin-R) (5, 74 –77). Similar examples of obesity in humans have been found and are associated with a mutation of leptin or the leptin-receptor (78 – 80). Leptin is released by adipose tissue and has been suggested to be the key-signal reflecting adipose stores. Leptin receptors are found in the hypothalamus, particularly in the arcuate nucleus where leptin is thought to exert its primary feedback signaling (81– 87). Recent experiments in rodents and primates have been attempting to tie together the diverse hypothalamic peptidergic systems with hormone receptors, including leptin receptors, to decipher the hypothalamic signaling modality underlying the regulation of daily energy homeostasis (81–92). A schematic illustration of some of these interactions and the way ghrelin signaling may be integrated into these circuits is shown on Fig. 1. Peripheral ghrelin is mainly produced in the gastrointestinal tract (8, 10, 22–24). It reaches ghrelin-receptors in the anterior pituitary and potentially in the mediobasal and mediolateral hypothalamus through the general circulation to stimulate GH release and to regulate energy homeostasis (12). It remains to be determined whether circulating ghrelin can reach brain areas outside of the blood brain barrier only, such as the ventromedial arcuate nucleus (93), or it has the ability to target areas protected by the blood brain barrier. Areas protected by the blood brain barrier include most hypothalamic nuclei and the rest of the brain (93). Ghrelincontaining cells are also present in the mediobasal hypothalamus, where GHRH cells and the neuronal network that regulates energy balance are located (8, 22). Detailed phenotypes and macroscopic connectivity of different hypothalamic networks regulating metabolism have been described by numerous recent outstanding reviews (2–7). Among hypothalamic peptidergic circuits, particular significance is attributed to the arcuate nucleus opiate neurons that produce -MSH, a main anorexigen and energy expenditure enhancer

(72), and to its interrelationship with another group of arcuate nucleus neurons that produce both NPY and an endogenous antagonist of -MSH, AGRP (69). The interaction between these two distinct populations of cells is currently considered as a primum movens in the regulation of energy homeostasis. However, there are other peptidergic circuits within the hypothalamus, including the lateral hypothalamic orexin/hypocretin- and melanin concentrating hormoneproducing cells, that appear to respond to peripheral metabolic signals and alter food intake as well as energy expenditure (65– 68). In light of the aforementioned excellent reviews (2–7), we will avoid an in-depth description of these peptidergic systems here but will attempt to emphasize a better appreciation of the neuronal doctrine for the integration of emerging experimental data on ghrelin. In the brain, receptors for ghrelin were detected in multiple hypothalamic nuclei as well as in the hippocampus, substantia nigra, ventral tegmental area, and dorsal and median raphe nuclei (8, 94 –98). In a series of experiments, Dickson and co-workers, first using synthetic GHS-R agonist, and then ghrelin, provided evidence that this novel metabolic hormone, in fact, interacts with the aforementioned hypothalamic peptidergic systems in the central regulation of metabolism (99 –103). For example, they found that following central ghrelin administration, c-fos, an early proto-oncogen that reflects cellular activity, is induced in the medial arcuate nucleus where NPY/AGRP cells are located (103). It was also shown that Y1-receptor antagonists as well as melanocortin agonists and antisera to both NPY and AGRP may interfere with ghrelin’s feeding-inducing effect (42, 43, 46). However, absence of NPY in genetically engineered NPY-ko mice does not diminish ghrelin-induced feeding or adiposity suggesting a key-role for AGRP in the mediation of ghrelin’s effects on energy balance (12). The effect of ghrelin on metabolism seems to be the exact opposite to that of leptin (2–7, 9, 10, 12). In obesity, when plasma leptin levels are elevated, ghrelin plasma levels are decreased indicating physiological adaptations to the positive energy balance rather than an involvement in the etiology of obesity (48, 49). Of course, it is important to note that, while ghrelin is regulated acutely like a satiety factor, leptin levels are not regulated by meals, but rather by actual increase in adipose stores.
Ghrelin’s Hypothalamic Signaling Requires Synapses

Figure 1 depicts a highly complex interaction between a variety of hypothalamic peptidergic systems, including the putative ghrelin network, in the central regulation of energy balance. It has to be noted, however, that this drawing is not all-inclusive and represents only the “tip of the iceberg.” There are many more hypothalamic and extra-hypothalamic neurotransmitters and neuropeptides that act via the aforementioned circuits (for example coexistence of GABA with NPY; 91) or in separate pathways [for example, ciliary neurotrophic growth factor (CNTF), 104, 105], and are interconnected with the illustrated systems (for further review see Refs. 2, 6, 7). In addition, receptors for the different neuropeptides as well as for peripheral hormones that affect metabolism, including insulin, thyroid hormones, gonadal steroids and glucocorticoids, are also present in these re-

4166

Endocrinology, October 2001, 142(10):4163– 4169

Horvath et al. • Ghrelin and Regulation of Energy Balance

FIG. 1. Schematic representation of the interaction between key hypothalamic peptidergic systems in the central regulation of daily energy homeostasis and their relationship to peripheral and putative hypothalamic ghrelin. Ghrelin, a hunger signal, is released from the stomach into the circulation and may be produced (?) in a subset of hypothalamic neurons (red). Leptin, a satiety signal, is released from white adipose tissue (WAT) into the circulatory system. Ghrelin (red arrows) and leptin (orange arrows) directly target the hypothalamus and brain stem areas. While brain stem areas on this drawing are illustrated as efferent targets of hypothalamic circuits, critical pathways exists from the brain stem to the hypothalamus, as well, that can mediate ascending ghrelin and leptin signaling. AGRP is produced in NPY cells (yellow) and acts to block the inhibitory action of the POMC derivate, -MSH (green), on feeding. Both AGRP/NPY and POMC cells are apparent targets of direct ghrelin action via GHS-R (*). The NPY neurons that receive lateral hypothalamic input, including HCRT (brown) and melanin concentrating hormone (MCH) (blue) innervation, project to a number of regions of the brain, particularly those implicated in feeding mechanisms, including the paraventricular nucleus (PVN), lateral hypothalamus, LH, ventromedial nucleus (VMH), perifornical region (PF), and dorsomedial nucleus (DMH). The same regions also receive direct lateral hypothalamic input as well as innervation from -MSH cells. These regions, in turn, project (large black arrow) widely throughout the brain to loci including the medial thalamic nuclei (MT), central gray (cg), dorsal motor nucleus of the vagus (DMV), cortex, nucleus of the solitary tract (NTS), locus coeruleus (LC), spinal cord, and amygdala. Ghrelin-targeted arcuate nucleus neurons may also affect neuroendocrine cells that are responsible for the regulation of pituitary hormone secretions, including gonadotrophs (LH/FSH), TSH, ACTH, and GH. It is yet to be determined what role central vs. peripheral ghrelin plays in the regulation of this circuitry and at what sites and subcellular levels ghrelin signaling is interacting with that of leptin.

gions. During food deprivation when leptin levels rapidly decline (106) and NPY/AGRP production is elevated, but POMC neurons are suppressed (106 –110), circulating ghrelin levels increase (12, 33, 41, 48) suggesting that leptin and ghrelin coregulate hypothalamic peptidergic systems in opposite ways. These observations further support the hypothesis that ghrelin, as a “hunger signal,” is the counterpart of leptin aiming to prevent further energy deficit. However, considering the extreme complexity of hypothalamic interactions of different peptidergic circuits and peripheral hormone receptors, it is necessary to determine the hierarchy and direction of signaling flow within these systems to understand ghrelin’s central effect on metabolic regulation. For that, a multidisciplinary approach is mandatory. The hypothalamus is composed of a complicated set of regulatory neurons that in most cases cannot be identified by

traditional means of cell segregation, i.e. location, soma size, or dendritic arbor. Therefore, to identify specific types of neurons, cytochemistry must be used. In addition, as in all other brain areas, the primary mode of communication between hypothalamic peptidergic circuits is via synapses. The only reliable way for assessing synapses is by the use of conventional electron microscopy and electrophysiology because proximity of different cells assessed by light microscopy is not a convincing indicator of neuronal interaction. Thus, determination of the qualitative and quantitative synaptological relationship between GHS-Rs, ghrelin-producing neurons, and other key hypothalamic peptidergic systems and their receptors will be an important step for gaining insight into the hypothalamic signaling modality of ghrelin. Of course, the anatomical experiments alone will not be sufficient to determine the actual involvement of the pre-

Horvath et al. • Ghrelin and Regulation of Energy Balance

Endocrinology, October 2001, 142(10):4163– 4169 4167

synaptic ghrelin system in the regulation of the postsynaptic circuit, but provides an invaluable map that is necessary for the correct interpretation of data gathered with other tools. In fact, anatomical studies need to be complemented by parallel electrophysiological analyses. An elegant example of such an approach is the recent work by Cowley and colleagues (111), in which leptin’s effect was analyzed on genetically tagged arcuate nucleus -MSH cells and the qualitative synaptology of these cells was simultaneously assessed. That approach not only eliminated the pitfalls of the individual experimental techniques but immediately provided a more comprehensive view on a given hypothalamic neuronal system (111). The significance in determining the spatial relationship between different afferents using anatomical and electrophysiological tools may further be appreciated when one considers that a synapse is more potently able to affect postsynaptic cells when located proximally either on the cell body or postsynaptic dendrite than when it is located more distally. In addition, both previous electrophysiological (111–113) and morphological observations (92, 111) indicate that an extensive interaction exists between presynaptic terminals to affect hypothalamic cells both in the arcuate nucleus where NPY/AGRP and -MSH cell bodies are located and in a model efferent target, the parvicellular paraventricular nucleus. The impact of ghrelin on arcuate and parvicellular paraventricular nucleus neurons will be readily dependent on their synaptic organization on the postsynaptic cells and their interaction with other systems presynaptically. One of the best examples to illustrate this synaptologic interaction is the relationship between the NPY/AGRP and -MSH systems. Electrophysiological and anatomical observations pointed to both the arcuate and paraventricular nuclei as primary sites for the interplay between AGRP and -MSH systems (111, 114). Because ghrelin’s action appears to be mediated by the NPY/AGRP system, it is not unlikely that ghrelin will act in the arcuate nucleus as well as in the paraventricular nucleus to modulate the interaction between NPY/AGRP and -MSH. It may be that peripheral and central ghrelin contribute equally to the regulation of both of these hypothalamic areas, but it is also conceivable that stomach-derived ghrelin affects the arcuate nucleus where the blood-brain barrier is less effective, whereas hypothalamic ghrelin is more involved in the modulation of hypothalamic sites within the blood-brain barrier, such as the paraventricular nucleus. An alternative and equally feasible pathway for ghrelin signaling from the stomach is via an ascending neural network through the vagus nerve and brain stem nuclei that ultimately reaches the hypothalamus (43). When electrophysiological and anatomical techniques are combined with conventional physiological and molecular biological approaches, as well as with the very recently developed revolutionary tracing technique of DeFalco et al. [(115) which allows tracing of inputs of chemically identified subpopulations of neurons], it is reasonable to expect that not only a thorough understanding of ghrelin’s action will be achieved at a faster pace, but great advances will be made toward the general understanding of the hypothalamic machinery in metabolism regulation.

Acknowledgments
We are thankful to Michael Statnick, Paul Burn, Jose Caro, and Marya Shanabrough for critical review. Received June 8, 2001. Accepted July 27, 2001. Address all correspondence and requests for reprints to: Tamas Horvath, Department of Obstetrics/Gynecology, Yale Medical School, 333 Cedar Street, FMB 339, New Haven, Connecticut 06520. E-mail: tamas.horvath@yale.edu. This work was supported by NIH Grants MH-59847 and RR-14451.

References
1. Mokdad AH, Serdula MK, Dietz WH, Bowman BA, Marks JS, Koplan JP 2000 The continuing epidemic of obesity in the United States. JAMA 284: 1650 –1651 2. Kalra SP, Dube MG, Pu S, Xu B, Horvath TL, Kalra PS 1999 Interacting appetite-regulating pathways in the hypothalamic regulation of body weight. Endocr Rev 20:68 –100 3. Schwartz MW, Woods SC, Porte Jr D, Seeley RJ, Baskin DG 2000 Central nervous system control of food intake. Nature 404:661– 671 4. Elmquist JK, Maratos-Flier E, Saper CB, Flier JS 1998 Unraveling the central nervous system pathways underlying responses to leptin. Nature Neuroscience 6:445– 450 5. Friedman JM, Halaas JL 1998 Leptin and the regulation of body weight in mammals. Nature 395:763–770 6. Ahima RS, Osei SY 2001 Molecular regulation of eating behavior: new insights and prospects for therapeutic strategies. Trends Mol Med 7:205–213 7. Spiegelman BM, Flier JS 2001 Obesity and the regulation of energy balance. Cell 104:531–543 8. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K 1999 Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402:656 – 660 9. Bowers CY 2001 Unnatural growth hormone-releasing peptide begets natural ghrelin. J Clin Endocrinol Metab 86:1464 –1469 10. Kojima M, Hosoda H, Matsuo H, Kangawa K 2001 Ghrelin: discovery of the natural endogenous ligand for the growth hormone secretagogue receptor. Trends Endocrinol Metab 12:118 –122 11. Hosoda H, Kojima M, Matsuo H, Kangawa K 2000 Purification and characterization of rat des-Gln14-Ghrelin, a second endogenous ligand for the growth hormone secretagogue receptor. J Biol Chem 275:21995–2000 12. Tschop M, Smiley D, Heiman ML 2000 Ghrelin induces adiposity in rodents. ¨ Nature 407:908 –913 13. Momany FA, Bowers CY, Reynolds GA, Chang D, Hong A, Newlander K 1981 Design, synthesis, and biological activity of peptides which release growth hormone in vitro. Endocrinology 108:31–39 14. Bowers CY, Momany F, Reynolds GA, Chang D, Hong A, Chang K 1980 Structure-activity relationships of a synthetic pentapeptide that specifically releases growth hormone in vitro. Endocrinology 106:663– 667 15. Bowers CY 1998 Growth hormone-releasing peptide (GHRP). Cell Mol Life Sci 54:1316 –1329 16. Howard AD, Feighner SD, Cully DF, et al. 1996 A receptor in pituitary and hypothalamus that functions in growth hormone release. Science 273:974 –977 17. Smith RG, Pong SS, Hickey G, et al. 1996 Modulation of pulsatile GH release through a novel receptor in hypothalamus and pituitary gland. Rec Prog Horm Res 51:261–286 18. Smith RG, Van der Ploeg LH, Howard AD, et al. 1997 Peptidomimetic regulation of growth hormone secretion. Endocr Rev 18:621– 645 19. Bednarek MA, Feighner SD, Pong SS, et al. 2000 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 20. Dornonville de la Cour C, Bjorkqvist M, Sandvik AK, et al. 2001 A-like cells in the rat stomach contain ghrelin and do not operate under gastrin control. Regul Pept 99:141–150 21. Date Y, Kojima M, Hosoda H, et al. 2000 Ghrelin, a novel growth hormonereleasing acylated peptide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats and humans. Endocrinology 141:4255– 4261 22. Kagotani Y, Sakata I, Yamazaki M, Nakamura K, Hayashi Y, Kangawa K 2001 Localization of ghrelin-immunopositive cells in the rat hypothalamus and intestinal tract. Proceedings of the 83rd Annual Meeting of The Endocrine Society, Denver, CO, 2001, p. 337 23. Korbonits M, Kojima M, Kangawa K, Grossman AB 2001 Presence of ghrelin in normal and adenomatous human pituitary. Endocrine 14:101–104 24. Mori K, Yoshimoto A, Takaya K, et al. 2000 Kidney produces a novel acylated peptide, ghrelin. FEBS Lett 486:213–216 25. Gualillo O, Caminos J, Blanco M, et al. 2001 Ghrelin, a novel placentalderived hormone. Endocrinology 142:788 –794 26. Ariyasu H, Takaya K, Tagami T, et al. 2001 Plasma ghrelin levels are in-

4168

Endocrinology, October 2001, 142(10):4163– 4169

Horvath et al. • Ghrelin and Regulation of Energy Balance

27. 28. 29. 30. 31. 32. 33.

34. 35. 36.

37. 38.

39. 40. 41. 42.

43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53.

fluenced by acute and chronic feeding states in humans. Proceedings of the 83rd Annual Meeting of The Endocrine Society, Denver, CO, 2001, p 336 Date Y, Murakami N, Kojima M, et al. 2000 Central effects of a novel acylated peptide, ghrelin, on growth hormone release in rats. Biochem Biophys Res Commun 275:477– 480 Seoane LM, Tovar S, Baldelli R, et al. 2000 Ghrelin elicits a marked stimulatory effect on GH secretion in freely-moving rats. Eur J Endocrinol 143: R7–R9 Tolle V, Zizzari P, Tomasetto C, Rio MC, Epelbaum J, Bluet-Pajot MT 2001 In vivo and in vitro effects of ghrelin/motilin-related peptide on growth hormone secretion in the rat. Neuroendocrinology 73:54 – 61 Arvat E, Di Vito L, Broglio F, et al. 2000 Preliminary evidence that Ghrelin, the natural GH secretagogue (GHS)-receptor ligand, strongly stimulates GH secretion in humans. J Endocrinol Invest 23:493– 495 Peino R, Baldelli R, Rodriguez-Garcia J, et al. 2000 Ghrelin-induced growth hormone secretion in humans. Eur J Endocrinol 143:R11–R14 Takaya K, Ariyasu H, Kanamoto N, et al. 2000 Ghrelin strongly stimulates growth hormone release in humans. J Clin Endocrinol Metab 85:4908 – 4911 Arvat E, Maccario M, Di Vito L, et al. 2001 Endocrine activities of ghrelin, a natural growth hormone secretagogue (GHS), in humans: comparison and interactions with hexarelin, a nonnatural peptidyl GHS, and GH-releasing hormone. J Clin Endocrinol Metab 86:1169 –1174 Cummings E, Purnell JQ, Frayo SR, Schmidova K, Wisse BE, Weigle DS 2001 A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 50:1714 –1719 Maccario M, Aimaretti G, Corneli G, et al. 2000 Short-term fasting abolishes the sex-related difference in GH and leptin secretion in humans. Am J Physiol Endocrinol Metab 279:E411–E416 Gurd W, Parent G, Eniojukan R, Bowers CY, Tannenbaum GS 2001 Interrelationship between the novel peptide ghrelin and somatostatin/GHRH inregulation of pulsatile GH secretion. Proceedings of the 83rd Annual Meeting of The Endocrine Society, Denver, CO, 2001, p 72 Tannenbaum GS, Bowers CY 2001 Interactions of growth hormone secretagogues and growth hormone-releasing hormone/somatostatin. Endocrine 14:21–27 Gondo RG, Aguiar-Oliveira MH, Hayashida CY, et al. 2001 Growth hormone-releasing peptide-2 stimulates GH secretion in GH-deficient patients with mutated GH-releasing hormone receptor. J Clin Endocrinol Metab 86: 3279 –3283 Bowers CY 1998 Synergistic release of growth hormone by GHRP and GHRH: Scope and implication. In: Bercu BB, Walker BF, eds. Growth hormone secretagogues in clinical practice. Ed. 1. New York: Marcel Dekker Inc.; 1–27 Broglio F, Arvat E, Benso A, et al., Ghrelin, a natural GH secretagogue produced by the stomach, induces hyperglycemia and reduces insulin secretion in humans. J Clin Endocrinol Metab, in press Heiman ML, Tschop M 2001 Ghrelin acts to provide the calories that growth ¨ hormone requires for growth and repair. Proceedings of the 83rd Annual Meeting of The Endocrine Society, Denver, CO, 2001, p 33 Shintani M, Ogawa Y, Ebihara K, et al. 2001 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 Asakawa A, Inui A, Kaga T, et al. 2001 Ghrelin is an appetite-stimulatory signal from stomach with structural resemblance to motilin. Gastroenterology 120:337–345 Kamegai J, Tamura H, Shimizu T, Ishii S, Sugihara H, Wakabayashi I 2000 Central effect of ghrelin, an endogenous growth hormone secretagogue, on hypothalamic peptide gene expression. Endocrinology 141:4797– 4800 Wren AM, Small CJ, Ward HL, et al. 2000 The novel hypothalamic peptide ghrelin stimulates food intake and growth hormone secretion. Endocrinology 141:4325– 4328 Nakazato M, Murakami N, Date Y, et al. 2001 A role for ghrelin in the central regulation of feeding. Nature 409:194 –198 Tschop M, Wawarta R, Riepl RL, et al. 2001 Post-prandial decrease of ¨ circulating human ghrelin levels. J Endocrinol Invest 24:RC19-RC21 Tschop M, Weyer C, Tataranni PA, Devanarayan V, Ravussin E, Heiman ¨ ML 2001 Circulating ghrelin levels are decreased in human obesity. Diabetes 50:707–709 Ravussin E, Tschop M, Heiman ML, Bouchard C 2001 Plasma ghrelin con¨ centration and energy balance: overfeeding and negative energy balance studies in twins. J Clin Endocrinol Metab 86:4547– 4551 Toshinai K, Mondal MS, Nakazato M, et al. 2001 Upregulation of Ghrelin expression in the stomach upon fasting, insulin-induced hypoglycemia, and leptin administration. Biochem Biophys Res Commun 281:1220 –1225 Toth DW, Hellmann PH, Gordon DA, et al. 2001 Evidence that the orexigenic effect of ghrelin is mediated by a distinct GHS-R subtype. Proceedings of the 83rd Annual Meeting of The Endocrine Society, Denver, CO, 2001, p 169 Chen C 2000 Growth hormone secretagogue actions on the pituitary gland: multiple receptors for multiple ligands? Clin Exp Pharmacol Physiol 27: 323–329 Torsello A, Locatelli V, Melis MR, et al. 2000 Differential orexigenic effects of hexarelin and its analogs in the rat hypothalamus: indication for multiple

54. 55. 56.

57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73.

74. 75. 76. 77. 78. 79. 80. 81. 82.

growth hormone secretagogue receptor subtypes. Neuroendocrinology 72: 327–332 Heymsfield SB, Greenberg AS, Fujioka K, et al. 1999 Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA 282:1568 –75 Brobeck JR 1946 Mechanism of the development of obesity in animals with hypothalamic lesions. Physiol Rev 26:541–559 Powley TL, Opsahl CH, Cox JE, Weingarten HP 1980 The role of the hypothalamus in energy homeostasis. In: Morgane PJ, Panskepp J, eds. Handbook of the hypothalamus. Part A: Behavioral studies of the hypothalamus. New York: Marcel Dekker, Inc.; pp 211–298 Bernadis LL, Berlinger LL 1987 The dorsomedial hypothalamic nucleus revisited. Brain Res 434:321–381 Aravich PF, Scalfani A 1983 Paraventricular hypothalamic lesions and medial hypothalamic cuts produce similar hyperphagia syndromes. Behav Neurosci 97:970 –983 Weingarten HP, Chang P, McDonald TJ 1985 Comparisons of the metabolic and behavioral disturbances following paraventricular and ventromedial hypothalamic lesions. Brain Res Bull 14:1551–1559 Tokunaga K, Fukushima M, Kemnitz JW, Bray GA 1986 Comparison of ventromedial and paraventricular lesions in rats that become obese. Am J Physiol 251:R1221–R1227 Powley TL, Keesey RE 1970 Relationship of body weight to the lateral hypothalamic feeding syndrome. J Comp Physiol Psychol 70:25–36 van den Pol AN 1982 Lateral hypothalamic damage and body weight regulation: role of gender, diet, and lesion placement. Am J Physiol 243:R265– R274 Clark JT, Kalra PS, Crowley WR, Kalra SP 1984 Neuropeptide Y and human pancreatic polypeptide stimulate feeding behavior in rats. Endocrinology 115:427– 429 Stanley BG, Chin AS, Leibovitz SF 1985 Feeding and drinking elicited by central injection of neuropeptide Y: evidence for a hypothalamic site(s) of action. Brain Res Bull 14:521–524 de Lecea L, Kilduff TS, Peyron C, et al. 1998 The hypocretins: two hypothalamic peptides with neuroexcitatory activity. Proc Natl Acad Sci USA 95:322–327 Sakurai T, Amemiya A, Ishii M, et al. 1998 Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell 92:573–585 Bittencourt JC, Presse F, Arias C, et al. 1992 The melanin-concentrating hormone system of the rat brain: an immuno-and hybridization histochemical characterization. J Comp Neurol 319:218 –245 Qu D, Ludwig DS, Gammeltoft S, et al. 1996 A role for melanin-concentrating hormone in the central regulation of feeding behavior. Nature 380: 243–247 Lu D, Willard D, Patel IR, et al. 1994 Agouti protein is an antagonist of the melanocyte-stimulating-hormone receptor. Nature 371:799 – 802 Ollmann MM, Wilson BD, Yang YK, et al. 1997 Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science 278:135–138 Fan W, Boston BA, Kesterson RA, Hruby VJ, Cone RD 1997 Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature 385:165–168 Hahn TM, Breininger JF, Baskin DG, Schwartz MW 1998 Coexpression of Agrp and NPY in fasting-activated hypothalamic neurons. Nature Neurosci 1:271–272 Mezey E, Kiss JZ, Mueller GP, Eskay R, Odonohue TL, Palkovits M 1985 Distribution of pro-opiomelanocortin derived peptides, adrenocorticotrope hormone, -melanocyte-stimulating hormone an -endorphin (ACTH, -MSH, -END) in the rat hypothalamus. Brain Res 328:341–347 Halaas JL, Gajiwala KS, Maffei SL, et al. 1995 Weight-reducing effects of the plasma protein encoded by the obese gene. Science 269:543–546 Pelleymounter MA, Cullen MJ, Baker MB, et al. 1995 Effects of the obese gene product on body weight regulation in ob/ob mice. Science 269:540 –543 Campfield LA, Smith FJ, Guisez Y, Devos R, Burn P 1995 Recombinant mouse OB protein: evidence for a peripheral signal linking adiposity and central neural networks. Science 269:546 –549 Leibel RL, Chung WK, Chua Jr SC 1997 The molecular genetics of rodent single gene obesities. J Biol Chem 272:31937–31940 Strobel A, Issad T, Camoin L, Ozata M, Strosberg AD 1998 A leptin missense mutation associated with hypogonadism and morbid obesity. Nat Genet 18:213–215 Montague CT, Farooqi IS, Whitehead JP, et al. 1997 Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387:903–908 Clement K, Vaisse C, Lahlou N, et al. 1998 A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 392:398 – 401 Mercer JG, Hoggard N, Williams LM, et al. 1996 Localization of leptin receptor mRNA and the long splice variant (O-Rb) in mouse hypothalamus and adjacent brain regions by in situ hybridization. FEBS Lett 387:113–116 Diano S, Kalra SP, Horvath TL 1998 Leptin receptor immunoreactivity is

Horvath et al. • Ghrelin and Regulation of Energy Balance

Endocrinology, October 2001, 142(10):4163– 4169 4169

83. 84. 85. 86. 87. 88.

89. 90. 91. 92.

93. 94. 95.

96.

97. 98. 99.

associated with the Golgi apparatus of hypothalamic cells. J Neuroendocrinol 9:647– 650 Elmquist JK, Elias CF, Saper CB 1999 From lesions to leptin: hypothalamic control of food intake and body weight. Neuron 2:221–232 Elmquist JK, Bjorbaek C, Ahima RS, Flier JS, Saper CB 1998 Distributions of leptin receptor mRNA isoforms in the rat brain. J Comp Neurol 395:535–547 Hakansson ML, Brown H, Ghilardi N, Skoda RC, Meister B 1998 Leptin receptor immunoreactivity in chemically defined target neurons of the hypothalamus. J Neurosci 18:559 –572 Yarnell DO, Knight DS, Hamilton K, Tulp O, Tso P 1998 Localization of leptin receptor immunoreactivity in the lean and obese zucker rat brain. Brain Res 785:80 –90 Schwartz MW, Seeley RJ, Campfield LA, Burn P, Baskin DG 1996 Identification of leptin action in rat hypothalamus. J Clin Invest 98:1101–1106 Broberger C, de Lecea L, Sutcliffe JG, Hokfelt T 1998 Hypocretin/orexinand melanin-concentrating hormone-expressing cells from distinct populations in the rodent lateral hypothalamus: relationship to the neuropeptide Y and agouti gene-related protein systems. J Comp Neurol 402:460 – 474 Elias CF, Saper CB, Maratos-Flier E, et al. 1998 Chemically defined projections linking the mediobasal hypothalamus and the lateral hypothalamus area. J Comp Neurol 402:442– 459 Horvath TL, Kalra SP, Naftolin F, Leranth C 1995 Morphological evidence for a galanin-opiate interaction in the rat mediobasal hypothalamus. J Neuroendocrinol 7:579 –588 Horvath TL, Bechmann I, Kalra SP, Naftolin F, Leranth C 1997 Heterogeneity in the neuropeptide Y-containing neurons of the rat arcuate nucleus: GABAergic and non-GABAergic subpopulations. Brain Res 756:283–286 Horvath TL, Diano S, van den Pol AN 1999 Synaptic interaction between hypocretin (orexin) and neuropeptide Y cells in the rodent and primate hypothalamus: a novel circuit implicated in metabolic and endocrine regulations. J Neurosci 19:1072–1087 Merchenthaler I 1991 Neurons with access to the general circulation in the central nervous system of the rat: a retrograde tracing study with fluoro-gold. Neuroscience 44:655– 662 Guan XM, Yu H, Palyha OC, et al. 1997 Distribution of mRNA encoding the growth hormone secretagogue receptor in brain and peripheral tissues. Brain Res Mol Brain Res 48:23–29 Tannenbaum GS, Lapointe M, Beaudet A, Howard AD 1998 Expression of growth hormone secretagogue-receptors by growth hormone-releasing hormone neurons in the mediobasal hypothalamus. Endocrinology 139:4420 – 4423 Shuto Y, Shibasaki T, Wada K, et al. 2001 Generation of polyclonal antiserum against the growth hormone secretagogue receptor (GHS-R): evidence that the GHS-R exists in the hypothalamus, pituitary and stomach of rats. Life Sci 68:991–996 Willesen MG, Kristensen P, Romer J 1999 Co-localization of growth hormone secretagogue receptor and NPY mRNA in the arcuate nucleus of the rat. Neuroendocrinology 70:306 –316 Smith RG, Leonard R, Bailey AR, et al. 2001 Growth hormone secretagogue receptor family members and ligands. Endocrine 14:9 –14 Luckman SM, Rosenzweig I, Dickson SL 1999 Activation of arcuate nucleus

100. 101.

102. 103. 104.

105. 106. 107. 108. 109.

110. 111. 112.

113. 114.

115.

neurons by systemic administration of leptin and growth hormone-releasing peptide-6 in normal and fasted rats. Neuroendocrinology 70:93–100 Dickson SL, Leng G, Robinson IC 1993 Systemic administration of growth hormone-releasing peptide activates hypothalamic arcuate neurons. Neuroscience 52:303–306 Dickson SL, Luckman SM 1997 Induction of c-fos messenger ribonucleic acid in neuropeptide Y and growth hormone (GH)-releasing factor neurons in the rat arcuate nucleus following systemic injection of the GH secretagogue, GH-releasing peptide-6. Endocrinology 138:771–777 Bailey AR, Von Englehardt N, Leng G, Smith RG, Dickson SL 2000 Growth hormone secretagogue activation of the arcuate nucleus and brainstem occurs via a non-noradrenergic pathway. J Neuroendocrinol 12:191–197 Hewson AK, Dickson SL 2000 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 Lambert PD, Anderson KD, Sleeman MW, et al. 2001 Ciliary neurotrophic factor activates leptin-like pathways and reduces body fat, without cachexia or rebound weight gain, even in leptin-resistant obesity. Proc Natl Acad Sci USA 98:4652– 4657 Kalra SP 2001 Circumventing leptin resistance for weight control. Proc Natl Acad Sci USA 98:4279 – 4281 Saladin R, De Vos P, Guerre-Millo M, et al. 1995 Transient increase in obese gene expression after food intake or insulin administration. Nature 377: 527–529 Sahu A, Kalra PS, Kalra SP 1988 Food deprivation and ingestion induced reciprocal changes in neuropeptide Y concentrations in the paraventricular nucleus. Peptides 9:83– 86 Sahu A, White JD, Kalra PS, Kalra SP 1992 Hypothalamic neuropeptide Y gene expression in rats on scheduled feeding regimen. Mol Brain Res 15:15–18 Sahu A 1998 Evidence suggesting that galanin (GAL), melanin-concentrating hormone (MCH), neurotensin (NT), proopiomelanocortin (POMC) and neuropeptide Y (NPY) are targets of leptin signaling in the hypothalamus. Endocrinology 139:795–798 Schwartz MW, Seeley RJ, Woods SC, et al. 1997 Leptin increases hypothalamic pro-opiomelanocortin mRNA expression in the rostral arcuate nucleus. Diabetes 46:2119 –2123 Cowley MA, Smart JL, Rubinstein M, et al. 2001 Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 411:480 – 484 Cowley MA, Pronchuk N, Fan W, Dinulescu DM, Colmers WF, Cone RD 1999 Integration of NPY, AGRP, and melanocortin signals in the hypothalamic paraventricular nucleus: evidence of a cellular basis for the adipostat. Neuron 24:155–163 van den Pol AN, Gao XB, Obrietan K, Kilduff T, Belousov A 1998 Pre- and postsynaptic actions and modulation of neuroendocrine neurons by a new hypothalamic peptide, hypocretin/orexin. J Neurosci 18:7962–7971 Broberger C, Johansen J, Johansson C, Schalling M, Hokfelt T 1998 The neuropeptide Y/agouti gene-related protein (AGRP) brain circuitry in normal, anorectic, and monosodium glutamate-treated mice. Proc Natl Acad Sci USA 95:15043–10048 DeFalco J, Tomishima M, Liu H, et al. 2001 Virus-assisted mapping of neural inputs to a feeding center in the hypothalamus. Science 291:2608 –2613

You May Also Find These Documents Helpful

  • Satisfactory Essays

    Hunger and the Fat Rat

    • 436 Words
    • 2 Pages

    Upon stimulation of the lateral hypothalamus a 5-10 gram increase in daily food consumption and a weight gain of almost 175 grams was noted over a 90 day period.…

    • 436 Words
    • 2 Pages
    Satisfactory Essays
  • Better Essays

    Biochemistry Quiz

    • 396 Words
    • 2 Pages

    1. Receptors for touch, heat, pressure, vision, and hearing are classified as ___ because they sense stimuli that arise external to the body.…

    • 396 Words
    • 2 Pages
    Better Essays
  • Good Essays

    Physio Ex4 Activity1

    • 2297 Words
    • 10 Pages

    Exercise 4: Endocrine System Physiology: Activity 1: Metabolism and Thyroid Hormone Lab Report Pre-lab Quiz Results You scored 100% by answering 6 out of 6 questions correctly. 1. Which of the following statements about metabolism is false? You correctly answered: d. All of the energy from metabolism is ultimately stored in the chemical bonds of ATP. 2. Thyroxine is You correctly answered: c. the most important hormone for maintaining the metabolic rate and body temperature. 3. Thyroid-stimulating hormone (TSH) is You correctly answered: b. produced in the pituitary gland. 4. An injection of TSH to an otherwise normal animal will cause which of the following? You correctly answered: d. goiter development 5. Thyrotropin-releasing hormone (TRH) is You correctly answered: a. secreted by the hypothalamus. 6. Which of the following statements is true? You correctly answered: b. The hypothalamus primarily secretes tropic hormones that stimulate the secretion of other hormones.…

    • 2297 Words
    • 10 Pages
    Good Essays
  • Satisfactory Essays

    PsychSim 5

    • 312 Words
    • 2 Pages

    • What did you learn from this experiment about these two regions of the hypothalamus?…

    • 312 Words
    • 2 Pages
    Satisfactory Essays
  • Better Essays

    Summary Of Forks Over Knives

    • 3397 Words
    • 14 Pages

    Neuroimaging and obesity: current knowledge and future directions. Obesity Reviews, 13(1), 44-56. doi: 0.1111/j.1467-789X.2011.00927.x 8. Chuang, J., Perello, M., Sakata, I., Osborne-Lawrence, S., Savitt, J., Lutter, M., & Zigman, J. (2011). Ghrelin mediates stress-induced food-reward behavior in mice.…

    • 3397 Words
    • 14 Pages
    Better Essays
  • Satisfactory Essays

    Research into homeostasis can be applied to everyday life. For example, Doctors or Pharmaceuticals can develop medication such as special pills which specifically target centres within the homeostasis model – lateral hypothalamus and the ventromedial hypothalamus. Medication can be developed for those who are either obese or underweight. This is because levels of blood glucose have been found to affect an individuals eating behaviours and therefore their weight too. This medication which can treat obesity can help increase glucose levels which in turn activate the ventromedial hypothalamus on. This then reduces the amount the individual eats. Those who are underweight can take medication containing the hormone neuropeptide Y which will activate the lateral hypothalamus. This information is useful because obesity is such a…

    • 347 Words
    • 2 Pages
    Satisfactory Essays
  • Better Essays

    Muller, Timo D., Diego P. Tilve, Jenny Tong, Paul T. Pfluger, and Matthias H. Tschop. "Ghrelin and Its Potential in the Treatment of Eating/wasting Disorders and Cachexia." J Cachex Sarcopenia Muscle 1.2 (2010): 159-67. Print.…

    • 1340 Words
    • 6 Pages
    Better Essays
  • Powerful Essays

    Hindbrain Function

    • 2339 Words
    • 10 Pages

    With obesity being a cultural syndrome in the US, there is a necessity to find solutions for it. Previous work has emphasized the importance of neuropeptides (protein neurotransmitters) in controlling obesity triggering factors such as ingestion and energy expenditure. The orexins (orexin A and orexin B) are a family of neuropeptides important in promoting physical activity, and have also recently been shown to increase energy expenditure through effects on thermogenesis. The research looks at the hindbrain signalling orexin A helps in controlling energy expenditure via thermogenesis. (Lee,…

    • 2339 Words
    • 10 Pages
    Powerful Essays
  • Good Essays

    Essay On Hypothalamus

    • 573 Words
    • 3 Pages

    In vertebrates, the hypothalamus, located at the base if the brain, plays a central role in integrating the endocrine and nervous systems. The hypothalamus receives information from nerves throughout the body and the brain. (Mousa and Mousa, 2003) In response, the hypothalamus initiates endocrine signalling appropriate to environmental conditions. In many vertebrates’ nerve signals from the brain pass information to the hypothalamus about seasonal changes, therefore the hypothalamus regulates the release of reproductive hormones, which are required during breeding seasons. (Campbell and Reece, 2011) The hypothalamus signals travel to the pituitary gland, which is located at the base of the hypothalamus with posterior and anterior parts. The anterior pituitary is an endocrine gland that synthesises and…

    • 573 Words
    • 3 Pages
    Good Essays
  • Powerful Essays

    Gonadotropin-releasing hormone (GnRH) is a peptide hormone that regulates the sexual maturation of mammals from the anterior pituitary gland. GnRH originates in the nasal placode (Funabashi et al., 2002) and migrates to the central nervous system during embryogenesis (Eisthen et al., 2000). There are several prominent locations of GnRH neurons in and out of the central nervous system including the rostral preoptic area (rPOA) which is the area of interest for this study. GnRH neurons specifically in the rPOA regulate the secretion of the gonadotropic hormones luteinising hormone (LH) and follicle stimulating hormone (FSH) (Eisthen et al., 2000; Suter et al., 2000).…

    • 1994 Words
    • 7 Pages
    Powerful Essays
  • Good Essays

    Modern conveniences have shaped our lives. Energy and its many uses help to keep humans safe, feed, and mobile. These modern abilities have also created a strain on our environment and the natural resources that make this existence possible. Currently electric for our community is generated by coal and our water is processes by electric. Our fossil fuels are on the brink of extinction, making the actions we take now more vital than ever. Creating an implemented plan that changes our current consumption of fossil fuels, will perpetuate a cleaner community, stabilize eco-systems, and provide a future for the generations to come. Harnessing renewable natural resources will allow the earth to heal because renewable resources are cleaner, efficient, and readily available.…

    • 629 Words
    • 3 Pages
    Good Essays
  • Satisfactory Essays

    Energy Plan

    • 353 Words
    • 1 Page

    It is important for everyone to do their part to conserve energy. Our resources, such as water, natural gas, oil, coal and the water species are limited. We have to save some for the future. Another reason is that some of these resources pollute the environment. We have to be careful since it causes damage to the environment.…

    • 353 Words
    • 1 Page
    Satisfactory Essays
  • Best Essays

    America's Energy Policy

    • 3119 Words
    • 13 Pages

    The invention of electricity, to many, can be considered one of the greatest inventions by humanity, behind writing and the wheel. From its first discovery in the 1600s till present day, electricity has fascinated the minds of scientist everywhere. Over time society has been established with a very high dependency on this commodity. Imagine our world today without electricity or any of the inventions derived from it. Modern societies would have taken a much different path. Last year the International Energy Agency conducted a survey on the energy statistics for over 30 of the more developed countries. Around 62 percent of the energy produced worldwide was created from combustible fuel source (Monthly Electricity Statistics). When combustible fuel is burned, carbon and other harmful byproducts are released into the atmosphere. This brings about environmental injustice since every living organism in contact with the atmosphere now suffers the consequences which the few benefit from. While I believe this is the more grave concern regarding electricity, the majority of the modernized world might not have an equivalent conclusion.…

    • 3119 Words
    • 13 Pages
    Best Essays
  • Good Essays

    Video Games V. Obesity

    • 1665 Words
    • 7 Pages

    Some people can actually be diagnosed with a condition called: obesity. There are some medical professionals out there that say that Obesity qualifies as a disease. Whether obesity is a condition or disease, Obesity is an abnormal high proportion/percent of body fat. A doctor can assess the percentage of body fat simply and easily by measuring body weight in proportion to height, which is the usual way of determining if an individual is overweight, obese, or at normal, healthy weight. A physician/doctor can make a determination if a patient is truly obese by calculating the BMI, which stands for body mass index. The BMI is calculated by dividing the body weight in kilograms by height in meters squared. BMI can be simply calculated using a weight and height table. The main problem with obesity is that it is a sure indicator of possible medical conditions for example diabetes and heart problems. Also, arthritis has been linked to obesity, along with pulmonary conditions that make breathing a problem, which prevents a person from doing active activities. The purpose of the following essay is to look at two articles that pertain first to the causes of obesity, and second pertain to treatment. Both articles deal with the causal links between other diseases and obesity. When it comes to article #2, since it deals with treatment, and not etiology, that article is more linked to OT issues than the first article that addresses causes.…

    • 1665 Words
    • 7 Pages
    Good Essays
  • Satisfactory Essays

    Energy is very important, especially for the people of the United States. It has become an integral part of our lives and we have grown dependent on it. Therein lies the problem, however, because the more we use it, the less there is. We have to find alternative forms of energy before it is gone and we are left high and dry. Not only is our sources decreasing, it is also polluting our beautiful world. Reducing the use of energy and finding alternatives is our only choice when considering the future of America. Cheap gas may sound great right now but once it is gone, it is gone for good. We cannot be selfish and only think of the now, we must look to the future and preserve our resources so that our grandkids will have a good life. We have made…

    • 258 Words
    • 2 Pages
    Satisfactory Essays

Related Topics