Review article

J. SOBOCKI1 ,G. KROLCZYK1, R.M. HERMAN2, A. MATYJA2, P.J. THOR1


INFLUENCE OF VAGAL NERVE STIMULATION ON FOOD INTAKE AND BODY
WEIGHT - RESULTS OF EXPERIMENTAL STUDIES



3rd Department of General Surgery and Department of Pathophysiology1,1st Department of General and GI Surgery2,
Jagiellonian University, Medical College, Cracow, Poland


  The paper reviews recent advances in vagal nerve stimulation for the control of food intake and body weight. The vagal nerves are the predominant pathway in the "brain-gut axis" responsible for short term regulation of food intake. Stimulation of afferent vagal traffic attenuates food intake by vagal projections to nucleus tractus solitarius, arcuate nucleus and its convergence’s to thalamic center of satiety. A few studies have been published in this field so far. All of them are consistent and show significant decrease in body mass during vagal stimulation. Due to promising results of experimental studies, clinical trials are expected in the near future.

Key words: food intake, vagal stimulation, vagal physiology.


Abbreviations: vagal nerve stimulation (VNS), cholecystokinin (CCK), gamma amino butyric acid (GABA), gastrointestinal (GI), peptide YY (PYY), nitric oxide (NO), positron emission tomography (PET).



INTRODUCTION

The vagus nerves contain major sensory fibers innervating the organs of thoracic and abdominal cavities. Afferent fibers of this nerve are integral part of the brain-gut axis, which take part in feedback loop controlling food intake induced by presence of food in gastrointestinal tract (1). The vagal afferent system detects gastrointestinal events in the periphery and generates appropriate autonomic, endocrine, metabolic and behavioral response. Electrophysiological recording studies identified mechano-, chemo-, osmo- and temperature-receptors. Possibility of food intake control by neuromodulation of the vagus nerve activity is actually considered and seems to be promising (2-4). The vagal nerve endings in upper GI tract are partially responsible for visceral sensation and are activated by different physical and chemical stimuli (5, 6). Duodenal receptors are sensitive predominantly to chemical stimuli and spontaneously inactive and react mostly in "on-of" system (6). The nerve itself is merely a cable receiving and conducting information in the form of electrical impulses from peripheral interface to its central representation (interface) in the brain (7, 8). The presence of food in duodenum elicits specific postprandial motility and secretion modifying interdigestive vagal discharge. The chemical content of the food is most likely differentiated by duodenal vagal chemoreceptors, whereas the size of the meal and induced motility changes stimulate mechanoreceptors of all upper GI tract (7). Actual studies pointed out mostly superiority of meal size (pressure factor) over its chemical content in inducing satiety feeling, however, this action was not definitely proved (9, 10). It seems to be possible that not only the size, but also meal composition influence food intake (11). Clear and different afferent discharge in the vagus nerve resulted in the presence of chemically distinct food in the duodenum (6). The importance of hepatic glucoreception in food intake regulation is not yet clear and needs further studies. One study showed that the alimentary behavior of rats after surgical liver denervation remains unchanged (12). Others suggest important role of hepatic vagal branch dysfunction in development of obesity in diabetics (13).

The presence of unique chemoreceptors in the stomach was not proved. These units are mostly polymodal in the rat and other species (14-16). Products of triglyceride digestion given to the rat stomach inhibit food intake via release of cholecystokinin (CCK), which in turn acts on vagus nerve sensory endings (17-19). Expression of bitter taste receptors of the T2R family in the gastrointestinal tract not only in duodenum, but also in the rat stomach has been reported (20). It is thought that presence of protein G (alfa-gustducin) in taste cells is associated with induction of taste signal, which in turn affects food intake inhibition. Ligand for taste receptor, which is simply food, triggers binding GTP to alfa-gustducin and subsequently stimulates production of second messenger. In the inner layer of stomach epithelium and gut this protein expression was reported. It may indicate potential role also gastric chemoreception in food intake (21). CCK and leptin potentiate subsequent responses to distending loads causing receptor sensitization and increasing afferent vagal traffic more than either load alone or peptide stimulations alone (22).

Satiety hormones are secreted by digestive tract in low concentrations and partially uptaken by the liver. Vagal nerve terminations have receptors for some of these hormones, as PYY (23), CCK (24) and leptin (25). This hormone-nerves transduced vagal satiety information can be effectively transmitted by afferents to control food intake via the brain-gut axis. Activation of feedback loop of this axis seems to be done by food stimuli in the following sequence: size of meal in upper GI tract, its chemical content in duodenum (transduction) and activation of hepatic glucoreceptors. Sensory information collected form peripheral interface is most likely coded in quantitative and qualitative pattern and sequence of afferent vagus nerves discharge. Integrated patterns of prandial and postprandial impulsation remain to be decoded.

RAT STUDIES

Direct electrical stimulation of the ventromedial hypothalamic nuclei enhances both fat utilization and metabolic rate, which has been shown to accompany the inhibition of feeding behavior in laboratory animals (26). Vagal nerve stimulation (VNS) is much easier for safe access and for indirect than direct stimulation of satiety center. Vagal nerves contain approximately 95% of afferent and only 5 % of efferent fibers (27). Afferent signals travel in vagal nerve fibers from mechanoreceptors and chemoreceptors, which are activated by the presence of nutrients in the stomach and in the proximal small intestine that are involved in meal termination. Nutrients arriving via the portal vein may also trigger vagal afferent signals from the liver (28). All of these signals are projected to the nucleus tractus solitarius (NTS), which remains in close relationship with the arcuate nucleus (ARC). ARC has been implicated in the control of feeding behavior due to activation of neurons releasing proopiomelanocortin (POMC) and alpha-melanocyte stimulating hormone (alpha-MSH) in the satiety center in ventromedial hypothalamus. Induction of satiety is accompanied by changes in metabolism. The metabolic and visceral sensory stimuli activate the satiety center and inhibit the hunger center.

Krolczyk et al showed a significant decrease in body weight and food intake induced by VNS in rats compared to controls (2). The same authors published interesting study on pathomechanism of VNS. Authors suggest that VNS effects are partially attenuated by baclofen and are related to GABA(B) receptors (29). Baclofen alone did not significantly change neither food intake nor diurnal body weight. In turn baclofen significantly reduced effect of vagal neuromodulation. This study showed also significant decrease in leptin and glucose levels due to VNS.


PIG AND DOG STUDIES

Gastric pacing was proposed for the treatment of obesity (30, 31). The idea of gastric stimulation for treating obesity was based on the artificial creation of anti-peristaltic motility in the porcine stomach (32). From a physiological point of view, the generation of anti-peristaltic motility induces rather nausea-like condition than satiety. It should be also noted that satiety signals are generated mainly in proximal stomach (33), which plays a role of temporary reservuar for ingested food. Moreover, satiety signals generated by gastric mechanoreceptors (mucosal "touch" receptors, muscular tension receptors, and serosal receptors) are not the unique source of physiological information provided by the digestive tract. Duodenal chemoreceptors and hepatic glucoreceptors are of the same importance as the others (34). Time, diversity and sequence of afferent information is important for the completing of satiety.

According to the physiology of food intake, activation of vagal afferents seems to be logic consequence for induction of satiety. Two approaches to electrode implantation have been proposed. Sobocki et al implanted electrodes by laparotomy access (35), and Roslin et al performed thoracotomy for electrode implantation (36).

Matyja et al used autonomic microchip for vagal stimulation, implanted by laparoscopy. Stimulated animals demonstrated continuous decrease in body weight gain during 8 weeks of experiment, compared to controls. Electrogastrogaphic recording showed decrease in percent of normogastria mostly at cost of tachygastria (37). Reduced percent of normogastria suggests delayed gastric emptying. Despite of this, food intake was not significantly affected, but body weight in stimulated pigs decreased. This discrepancy suggests predominant importance of central rather than peripheral effects of vagal stimulation and lacks the side effects. Tachygastria is often associated with nausea, vomiting and early satiety (38). In this study, however, no vomiting or symptoms related to nausea were noted.

VNS modifies not only centropetal afferent input, but also peripheral output of the both vagal nerves. Studies on recording of mass vagal nerve activity showed two kinds of activity, high amplitude low frequency and high frequency low amplitude signals (39).

Peripheral effects of VNS includes effects on all gastrointestinal functions. Krolczyk et al have shown decreased NO dependent gastric emptying and secretion in rats (2). Similar results were published by Ouyang and coworkers (40). They found that electrical field gastric stimulation (EFS) in dogs with long pulsed exerts protracted effects on gastric emptying, gastric contractility and vagal activity. Long term gastric stimulation in dogs decreased significantly food intake and body weight.

Some interesting data have been published recently on the VNS regarding its pathomechanism of action, influence of hormonal activity, metabolic rate, body composition, and incidence of adverse effects (41). In this study VNS attenuated body weight gain, reduced subcutaneous fat gain and intramuscular fat gain. VNS resulted in lower fat/fat free body mass ratio. No significant change in metabolic rate was observed. In this study laparoscopic technique of electrode implantation was described.

The study of Laskiewicz et al (39), have shown that bilateral VNS is more effective than unilateral stimulation, and suggested that anterior vagal stimulation with posterior vagotomy was as effective as bilateral stimulation. The anterior vagal trunk is suggested for stimulation for at last two reasons: firstly it activates hepatic branch (which play an important role in sensation of satiety), secondly VNS induces weight loss in humans, where electrode is habitually implanted on the left side (42). Bilateral electrode implantation is preferred by Roslin et al (36).


CLINICAL PERSPECTIVE

VNS can control body mass not only in experimental animals but also in humans. Burneo et al published that VNS is capable to decrease body mass in humans, as a side effect of treatment of epilepsy within 6 and 12 months after implantation (42). The pathway of this action involves hypothalamic centers. It was shown by PET-imaging that the application of VNS for treating epilepsy induces hypometabolism of the hypothalamus (43). Consequently, VNS may involve the brain center of satiety and induce a cascade of metabolic and behavioral changes. So far only one report of few implanted cases has been published by Roslin et al. Results of this pilot study were mixed. Significant lost of body weight was achieved only in two of 6 implanted patients.

Due to promising results of experimental studies, clinical trials are expected in the near future.


REFERENCES
  1. Schwartz GJ. The Role of Gastrointestinal Vagal Afferents in the Control of Food Intake: Current Prospects. Nutrition 2000; 16: 866-873.
  2. Krolczyk G, Zurowski D, Sobocki J, et al. Effects of continuous microchip (MC) vagal neuromodulation on gastrointestinal function in rats. J Physiol Pharmacol 2001; 42(4pt1): 705-715.
  3. Sobocki J, Thor PJ, Krolczyk G, Uson J, Diaz-Guemes I, Lipinski M. The cybergut. An experimental study on permanent microchip neuromodulation for control of gut function. Acta Chir Belg 2002; 102: 68-70.
  4. Krolczyk G, Zurowski D, Dobrek L, Laskiewicz J, Thor JP. The role of vagal efferents in regulation of gastric emptying and motility in rats. Folia Med Cracov 2001; 42(3): 141-148.
  5. Davison JS, Clarke GD. Mechanical properties and sensitivity to CCK of vagal gastric slowly adapting mechanoreceptors. Am J Physiol 1988; 255(1 Pt 1): G55-61.
  6. Mei N. Intestinal chemosensitivity. In: Advences in the Innervation of the Gastrointestinal Tract, edited by G.E.Holle and J.D.Wood. Amsterdam, The Netherlands: Elsevier 1992: pp 271-281
  7. Schwartz GJ, Moran TH. Duodenal nutrient exposure elicits nutrient-specific gut motility and vagal afferent signals in rat. Am J Physiol 1998; 274(5 Pt 2): R1236-1242.
  8. Yox DP, Stokesberry H, Ritter RC. Vagotomy attenuates suppression of sham feeding induced by intestinal nutrients. Am J Physiol 1991; 260: R503-R508.
  9. Phillips RJ, Powley TL. Gastric volume rather than nutrient content inhibits food intake. Am J Physiol 1996; 271: R766.
  10. Mathis C, Morgan TH, Schwartz GJ. Load-sensitive rat gastric vagal afferents encode volume but not gastric nutrients. Am J Physiol 1998; 274: R280-R286.
  11. Deutsch JA, Tabuena JA. Learning of gastrointestinal satiety signals. Behav Neural Biol 1986; 45(3): 292-299.
  12. Bellinger LL, Mendel VE, Williams FE, Castonguay TW. The effect of liver denervation on meal patterns, body weight and body composition of rats. Physiol Behav 1984; 33(4): 661-677.
  13. La Fleur SE, Ji H, Manalo SL, Friedman M, Dallman MF. The Hepatic Vagus Mediates Fat-Induced Inhibition of Diabetic Hyperphagia. Diabetes 2003; 52(9): 2321-2330.
  14. Berthoud HR, Neuhuber WL. Functional and chemical anatomy of the afferent vagal system. Auton Neurosci 2000; 85: 1-17.
  15. Berthoud HR, Lynn PA, Blackshaw LA. Vagal and spinal mechanosensors in the rat stomach and colon have multiple receptive fields. Am J Physiol Regul Integr Comp Physiol 2001; 280(5): R1371-1381.
  16. Clarke GD, Davison JS. Mucosal receptors in the gastric antrum and small intestine of the rat with afferent fibbers in the cervical vagus. J Physiol 1978; 284: 55-67.
  17. Gonzalez MF, Deutsch JA. Intragastric injections of partially digested triglyceride suppress feeding in the rat. Physiol Behav 1985; 35(6): 861-865.
  18. Garlicki J, Konturek PK, Majka J, Kwiecieñ N, Konturek SJ. Cholecystokinin receptors and vagal nerves in control of food intake in rats. Am J Physiol 1990; 258 (Endocrinol Metab 21): E40-E45.
  19. Richards W, Hillsley K, Estwood C, Grundy D. Sensitivity of vagal mucosal afferents to cholecystokinin and its role in afferent signal transduction in the rat. J Physiol 1996; 497.2: 473-481.
  20. Wu SV, Rozengurt N, Yang M, Young SH, Sinnett-Smith J, Rozengurt E. Expression of bitter taste receptors of the T2R family in the gastrointestinal tract and enteroendocrine STC-1 cells. Proc Natl Acad Sci USA 2002; 99(4): 2392-2397.
  21. Hofer D, Puschel B, Drenckhahn D. Taste receptor-like cells in the rat gut identified by expression of alpha-gustducin. Proc Natl Acad Sci USA 1996; 93(13): 6631-6634.
  22. Barrachira MD, Martinez W, Wang L, Hei J, Tache J. Synergistic interaction between leptin and cholecystokinin to reduce short term food intake in lean mice. Proc Natl Acad Sci USA 1997; 94: 10455-10460.
  23. Dockray G. Gut endocrine secretions and their relevance to satiety. Curr Opin Pharmacol 2004; 4: 557-560.
  24. Blackshaw LA, Grundy D. Effects of cholecystokinin (CCK-8) on two classes of gastroduodenal vagal afferent fibres. J Auton Nerv Syst 1990; 31: 191-202.
  25. Buyse M, Ovesjo ML, Goiot H, et al. Expression and regulation of leptin receptor proteins in afferent and efferent neurons of the vagus nerve. Eur J Neurosc 2000; 14(1): 64-72.
  26. Ruffin M, Nicolaidis S. Electrical stimulation of the ventromedial hypothalamus enhances both fat utilization and metabolic rate that precede and parallel the inhibition of feeding behavior. Brain Res 1999; 846(1): 23-29.
  27. Prechtl JC, Powley TL. The fiber composition of the abdominal vagus of the rat. Anat Embryol 1990; 181: 101-103.
  28. Havel PJ. Peripheral signals conveying metabolic information to the brain: short-term and long-term regulation of food intake and energy homeostasis. Exp Biol Med 2001; 226(11): 963-977.
  29. Krolczyk G, Laskiewicz J, Sobocki J, Matyja A, Kolasinska-Kloch W, Thor PJ. Effects of backlofen on feeding behavior and body weight of vagally stimulated rats. J Physiol Pharmacol 2005; 56(1): 121-131.
  30. Dargent J. Gastric electrical stimulation as therapy of morbid obesity: preliminary results from the french study. Obes Surg 2002; 12: 21S-25S.
  31. Greenstein RJ, Belachew M. Implantable gastric stimulation (IGS) as therapy for human morbid obesity: report from the 2001 IFSO symposium in Crete. Obes Surg 2002; 12 Suppl 1: 3S-5S.
  32. Cigaina VV, Pinato G, Rigo VV, et al. Gastric peristalsis control by mono situ electrical stimulation: a preliminary study. Obes Surg 1996; 6: 247-249.
  33. Ozaki N, Sengupta N, Gebhart GF. Mechanosensitve properties of gastric vagal afferent fibers in the rat. J Neurophysiol 1999; 82: 2210-2220.
  34. Krolczyk G, Zurowski D, Sobocki J, Laskiewicz J, Thor PJ. Encoding meal in vagal afferent discharge. J Physiol Pharmacol 2004; 55(1): 99-106.
  35. Sobocki J, Thor PJ, Uson J, et al. Microchip vagal pacing reduces food intake and body mass. Hepatogastroenterol 2001; 48: 1783-1787.
  36. Roslin M, Kurian M. The use of electrical stimulation of the vagus nerve to treat morbid obesity. Epilepsy Behav 2001; 2: S11-S16.
  37. A. Matyja, PJ Thor, J. Sobocki, et al. Effects of vagal pacing on food intake and body mass in pigs. Folia Med Cracov 2004; 45(3-4): 55-62.
  38. Geldof H, van der Schee EJ, van Blankenstein M, Grashius IL. Electrogastrographic study of gastric myoelectrical activity in-patients with unexplained nausea and vomiting. Gut 1986; 26: 799-808.
  39. Laskiewicz J, Krolczyk G, Zurowski D, Sobocki J. Matyja A, Thor PJ. Effect of vagal neuromodulation and vagotomy on control of food intake and body weight. J Physiol Pharmacol 2003; 54: 603-610.
  40. Ouyang H, Yin J, Zhu H, Xu X, Chen JD. Effects of gastric electrical field stimulation with long pulses on gastric emptying in dogs. Neurogastroenterol Motil 2003; 15(4): 409-416.
  41. Sobocki J, Fourtanier G, Estany J, Muscari F, Otal Ph. Traitement de l'obesite par stimulation vagale. Etude experimantale d'une nouvelle technique laparoscopique de chirurgie bariatrique. Ann Chir 2004; 129(9): 544.
  42. Burneo JG, Faught E, Knowlton R, Morawetz R, Kuzniecky R. Weight loss associated with vagus nerve stimulation. Neurology 2002; 59(3): 463-464.
  43. Petrucci M, Hoh C, Alksne JF. Thalamic hypometabolism in a patient undergoing vagal nerve stimulation seen on F-18FDGPET imaging. Clin Nucl Med 2003; 28(29): 784-785.

Author’s address: Jacek Sobocki, MD, Ph D 3rd Department of General Surgery Jagiellonian University, Cracow, Poland, ul. Pradnicka 37, 31-302 Krakow.
e-mail: jsobocki@cm-uj.krakow.pl