Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters










Publication year range
1.
Int J Obes (Lond) ; 40(6): 921-8, 2016 06.
Article in English | MEDLINE | ID: mdl-26984418

ABSTRACT

BACKGROUND/OBJECTIVE: Obesity and metabolic diseases are at an alarming level globally and increasingly affect children and adolescents. Gastric bypass and other bariatric surgeries have proven remarkably successful and are increasingly performed worldwide. Reduced desire to eat and changes in eating behavior and food choice account for most of the initial weight loss and diabetes remission after surgery, but the underlying mechanisms of altered gut-brain communication are unknown. SUBJECTS/METHODS: To explore the potential involvement of a powerful brainstem anorexia pathway centered around the lateral parabrachial nucleus (lPBN), we measured meal-induced neuronal activation by means of c-Fos immunohistochemistry in a new high-fat diet-induced obese mouse model of Roux-en-Y gastric bypass (RYGB) at 10 and 40 days after RYGB or sham surgery. RESULTS: Voluntary ingestion of a meal 10 days after RYGB, but not after sham surgery, strongly and selectively activates calcitonin gene-related peptide neurons in the external lPBN as well as neurons in the nucleus tractus solitarius, area postrema and medial amygdala. At 40 days after surgery, meal-induced activation in all these areas was greatly diminished and did not reach statistical significance. CONCLUSIONS: The neural activation pattern and dynamics suggest a role of the brainstem anorexia pathway in the early effects of RYGB on meal size and food intake that may lead to adaptive neural and behavioral changes involved in the control of food intake and body weight at a lower level. However, selective inhibition of this pathway will be required for a more causal implication.


Subject(s)
Anorexia/physiopathology , Disease Models, Animal , Eating , Feeding Behavior , Gastric Bypass , Parabrachial Nucleus/physiopathology , Animals , Diet, High-Fat , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Obesity/pathology , Obesity/surgery , Satiety Response
3.
Int J Obes Suppl ; 6(Suppl 1): S37-S43, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28685029

ABSTRACT

The relatively stable body weight during adulthood is attributed to a homeostatic regulatory mechanism residing in the brain which uses feedback from the body to control energy intake and expenditure. This mechanism guarantees that if perturbed up or down by design, body weight will return to pre-perturbation levels, defined as the defended level or set point. The fact that weight re-gain is common after dieting suggests that obese subjects defend a higher level of body weight. Thus, the set point for body weight is flexible and likely determined by the complex interaction of genetic, epigenetic and environmental factors. Unlike dieting, bariatric surgery does a much better job in producing sustained suppression of food intake and body weight, and an intensive search for the underlying mechanisms has started. Although one explanation for this lasting effect of particularly Roux-en-Y gastric bypass surgery (RYGB) is simple physical restriction due to the invasive surgery, a more exciting explanation is that the surgery physiologically reprograms the body weight defense mechanism. In this non-systematic review, we present behavioral evidence from our own and other studies that defended body weight is lowered after RYGB and sleeve gastrectomy. After these surgeries, rodents return to their preferred lower body weight if over- or underfed for a period of time, and the ability to drastically increase food intake during the anabolic phase strongly argues against the physical restriction hypothesis. However, the underlying mechanisms remain obscure. Although the mechanism involves central leptin and melanocortin signaling pathways, other peripheral signals such as gut hormones and their neural effector pathways likely contribute. Future research using both targeted and non-targeted 'omics' techniques in both humans and rodents as well as modern, genetically targeted, neuronal manipulation techniques in rodents will be necessary.

4.
Obes Rev ; 16 Suppl 1: 77-90, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25614206

ABSTRACT

Bariatric surgery continues to be remarkably efficient in treating obesity and type 2 diabetes mellitus and a debate has started whether it should remain the last resort only or also be used for the prevention of metabolic diseases. Intense research efforts in humans and rodent models are underway to identify the critical mechanisms underlying the beneficial effects with a view towards non-surgical treatment options. This non-systematic review summarizes and interprets some of this literature, with an emphasis on changes in the controls of appetite. Contrary to earlier views, surgery-induced reduction of energy intake and subsequent weight loss appear to be the main drivers for rapid improvements of glycaemic control. The mechanisms responsible for suppression of appetite, particularly in the face of the large weight loss, are not well understood. Although a number of changes in food choice, taste functions, hedonic evaluation, motivation and self-control have been documented in both humans and rodents after surgery, their importance and relative contribution to diminished appetite has not yet been demonstrated. Furthermore, none of the major candidate mechanisms postulated in mediating surgery-induced changes from the gut and other organs to the brain, such as gut hormones and sensory neuronal pathways, have been confirmed yet. Future research efforts should focus on interventional rather than descriptive approaches in both humans and rodent models.


Subject(s)
Appetite Regulation/physiology , Bariatric Surgery , Feeding Behavior , Gastrointestinal Hormones/metabolism , Obesity/surgery , Weight Loss , Animals , Disease Models, Animal , Energy Intake , Energy Metabolism , Humans , Mice , Rats
5.
Int J Obes (Lond) ; 39(5): 798-805, 2015 May.
Article in English | MEDLINE | ID: mdl-25349056

ABSTRACT

OBJECTIVE: Weight regain contributes to the therapeutic failure in 15-20% of type 2 diabetic patients after Roux-en-Y gastric bypass surgery (RYGB), and the mechanism remains largely unknown. This study was conducted to explore the mechanism of weight regain. RESEARCH DESIGN: Wild-type (WT) diet-induced obese (DIO) mice were used to mimic human obesity, and ob/ob mice were used for leptin deficiency-induced obesity. Two groups of mice were compared in weight regain for 10 months after RYGB. Weight loss, food intake, fecal energy loss and energy expenditure were monitored in the study of weight regain. Fasting insulin, insulin tolerance and homeostatic model assessment-insulin resistance were tested for insulin sensitivity under the weight regain. Weight loss from RYGB and calorie restriction was compared for the impact in insulin sensitivity. RESULTS: In WT mice, RYGB induced a sustained weight loss and insulin sensitization over the sham operation in this 10-month study. However, RYGB failed to generate the same effects in leptin-deficient ob/ob mice, which suffered a weight regain over the pre-surgery level. In ob/ob mice, body weight was reduced by RYGB transiently in the first week, recovered in the second week and increased over the baseline thereafter. Weight loss was induced by RYGB relative to that of sham mice, but the loss was not sufficient to keep body weight below the pre-surgery levels. In addition, insulin sensitivity was not improved by the weight loss. The response to RYGB was improved in ob/ob mice by 2 weeks of leptin treatment. Weight loss from calorie restriction had an equivalent effect on insulin sensitization compared with that of RYGB. CONCLUSION: Those data demonstrate that ob/ob mice and DIO mice responded differently to RYGB surgery, suggesting that leptin may be one of the factors required for RYGB to prevent weight regain and diabetes recurrence.


Subject(s)
Gastric Bypass , Leptin/metabolism , Obesity/metabolism , Obesity/surgery , Weight Gain , Weight Loss , Animals , Diet, High-Fat , Disease Models, Animal , Gastric Bypass/methods , Insulin Resistance , Leptin/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Reproducibility of Results
6.
Neurogastroenterol Motil ; 25(1): e70-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23095091

ABSTRACT

BACKGROUND: Roux-en-Y gastric bypass (RYGB) surgery is very effective in reducing excess body weight and improving glucose homeostasis in obese subjects. Changes in the pattern of gut hormone secretion are thought to play a major role, but the mechanisms leading to both changed hormone secretion and beneficial effects remain unclear. Specifically, it is not clear whether changes in the number of hormone-secreting enteroendocrine cells, or changes in the releasing stimuli, or both, are important. METHODS: We estimated numbers of enteroendocrine cells after immunohistochemical staining in fixed tissue samples from rats at 10-11 months after RYGB. KEY RESULTS: Numbers of glucagon-like peptide-1 (GLP-1) (L-cells, co-expressing peptide YY (PYY)), cholecystokinin (CCK), neurotensin, and 5-HT-immunoreactive cells were significantly increased in the Roux and common limbs, but not the biliopancreatic limb in RYGB rats compared with sham-operated, obese rats fed high-fat diet, and chow-fed controls. This increase was mostly accounted for by general hyperplasia of all intestinal wall layers of the nutrient-perfused Roux and common limbs, and less to increased density of expression. The number of ghrelin cells in the bypassed stomach was not different among the three groups. CONCLUSIONS & INFERENCES: The findings suggest that the number of enteroendocrine cells increases passively as the gut adapts, and that the increased total number of L- and I-cells is likely to contribute to the higher circulating levels of GLP-1, PYY, and CCK, potentially leading to suppression of food intake and stimulation of insulin secretion. Whether changes in releasing stimuli also contribute to altered circulating levels will have to be determined in future studies.


Subject(s)
Cholecystokinin/biosynthesis , Enteroendocrine Cells/cytology , Gastric Bypass , Glucagon-Like Peptide 1/biosynthesis , Neurotensin/biosynthesis , Serotonin/biosynthesis , Animals , Enteroendocrine Cells/metabolism , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley
7.
Int J Obes (Lond) ; 35 Suppl 3: S40-4, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21912387

ABSTRACT

Roux-en-Y gastric bypass surgery (RYGB) remains to be the most effective long-term treatment for obesity and its associated comorbidities, but the specific mechanisms involved remain elusive. Because RYGB patients appear to no longer be preoccupied with thoughts about food and are satisfied with much smaller meals and calorically dilute foods, brain reward mechanisms could be involved. Just as obesity can produce maladaptive alterations in reward functions, reversal of obesity by RYGB could normalize these changes or even further reset the food reward system through changes in gut hormone secretion, aversive conditioning and/or secondary effects of weight loss. Future studies with longitudinal assessments of reward behaviors and their underlying neural circuits before and after surgery will be necessary to uncover the specific mechanisms involved. Such new insights could be the base for future 'knifeless' pharmacological and behavioral approaches to obesity.


Subject(s)
Gastric Bypass , Hypothalamus/physiopathology , Neural Pathways/physiopathology , Obesity, Morbid/physiopathology , Satiety Response , Weight Loss , Animals , Food Preferences , Gastrointestinal Hormones/metabolism , Humans , Hypothalamus/metabolism , Neural Pathways/metabolism , Obesity, Morbid/metabolism , Obesity, Morbid/surgery , Rats , Reward
8.
Obes Rev ; 12(11): 984-94, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21729236

ABSTRACT

Bariatric surgery is the most effective method for promoting dramatic and durable weight loss in morbidly obese subjects. Furthermore, type 2 diabetes is resolved in over 80% of patients. The mechanisms behind the amelioration in metabolic abnormalities are largely unknown but may be due to changes in energy metabolism, gut peptides and food preference. The goal of this meeting was to review the latest research to better understand the mechanisms behind the 'magic' of bariatric surgery. Replication of these effects in a non-surgical manner remains one of the ultimate challenges for the treatment of obesity and diabetes. Promising data on energy metabolism, gastrointestinal physiology, hedonic response and food intake were reviewed and discussed.


Subject(s)
Bariatric Surgery/methods , Energy Metabolism/physiology , Obesity, Morbid/surgery , Weight Loss , Ghrelin/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Obesity, Morbid/metabolism , Peptide YY/metabolism , Weight Loss/physiology
9.
Int J Obes (Lond) ; 35(5): 642-51, 2011 May.
Article in English | MEDLINE | ID: mdl-20805826

ABSTRACT

CONTEXT: Roux-en-Y gastric bypass surgery (RYGB) is currently the most effective treatment for morbid obesity, and clinical studies suggest that RYGB patients change food preferences and the desire to eat. OBJECTIVE: To examine hedonic reactions to palatable foods and food choice behavior in an established rat model of RYGB. METHODS AND DESIGN: Male Sprague-Dawley (SD) rats and selected line obesity-prone rats that were rendered obese on a high-fat diet underwent RYGB or sham surgery and were tested for 'liking' and 'wanting' of palatable foods at different caloric densities 4-6 months after surgery. RESULTS: Compared with sham-operated (obese) and age-matched lean control rats, RYGB rats of both models exhibited more positive orofacial responses to low concentrations of sucrose but fewer to high concentrations. These changes in 'liking' by RYGB rats were translated into a shift of the concentration-response curve in the brief access test, with more vigorous licking of low concentrations of sucrose and corn oil, but less licking of the highest concentrations. The changes in hedonic evaluation also resulted in lower long-term preference/acceptance of high-fat diets compared with sham-operated (obese) rats. Furthermore, the reduced 'wanting' of a palatable reward in the incentive runway seen in sham-operated obese SD rats was fully restored after RYGB to the level found in lean control rats. CONCLUSIONS: The results suggest that RYGB leads to a shift in hedonic evaluation, favoring low over high calorie foods and restores obesity-induced alterations in 'liking' and 'wanting'. It remains to be determined whether these effects are simply due to weight loss or specific changes in gut-brain communication. Given the emerging evidence for modulation of cortico-limbic brain structures involved in reward mechanisms by gut hormones, RYGB-induced changes in the secretion of these hormones could potentially be mediating these effects.


Subject(s)
Body Weight/physiology , Food Preferences/physiology , Gastric Bypass , Obesity, Morbid/surgery , Weight Loss/physiology , Animals , Gastric Bypass/methods , Male , Obesity, Morbid/physiopathology , Rats , Rats, Sprague-Dawley , Reward
10.
Neuroscience ; 170(2): 580-8, 2010 Oct 13.
Article in English | MEDLINE | ID: mdl-20654704

ABSTRACT

Overindulgence in easily available energy-dense palatable foods is thought to be an important factor in the current obesity epidemic but the underlying neural mechanisms are not well understood. Here we demonstrate that mu-opioid receptor signaling in the nucleus accumbens may be important. Protracted suppression of endogenous mu-opioid receptor signaling focused on the nucleus accumbens shell for several days by means of microinjected beta-funaltrexamine (BFNA) diminished both "liking" of sucrose, as indicated by fewer positive hedonic orofacial responses, and the incentive reinforcement value ("wanting") of a food reward, as indicated by lower completion speed and increased time being distracted in the incentive runway. BFNA-treatment also decreased responding to sucrose and corn oil in the brief access lick paradigm, a test measuring a combination of mainly taste-guided "liking" and low-effort "wanting", as well as 4 h intake of sucrose solution. These effects were not due to nonspecific permanent neuronal changes, as they were fully reversible. We conclude that endogenous mu-opioid signaling in the nucleus accumbens is necessary for the full display of palatable food-induced hyperphagia through mechanisms including hedonic, motivational, and reinforcement processes. Development of obesity could be the result of predisposing innate differences in these mechanisms or overstimulation of these mechanisms by external factors.


Subject(s)
Appetitive Behavior/physiology , Naltrexone/analogs & derivatives , Nucleus Accumbens/physiology , Receptors, Opioid, mu/physiology , Animals , Male , Microinjections , Motivation/physiology , Naltrexone/administration & dosage , Naltrexone/pharmacology , Nucleus Accumbens/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/antagonists & inhibitors , Reinforcement, Psychology , Reward , Sucrose
11.
Int J Obes (Lond) ; 34(6): 1001-10, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20065959

ABSTRACT

OBJECTIVE: To test the hypothesis that micro-opioid receptor signaling in the nucleus accumbens contributes to hedonic (over)eating and obesity. To investigate the effects of chronic micro-opioid antagonism in the nucleus accumbens core or shell on intake of a palatable diet, and the development of diet-induced obesity in rats. METHODS AND DESIGN: Chronic blockade of micro-opioid receptor signaling in the nucleus accumbens core or shell was achieved by means of repeated injections (every 4-5 days) of the irreversible receptor antagonist beta-funaltrexamine (BFNA) over 3-5 weeks. The diet consisted of either a choice of high-fat chow, chocolate-flavored Ensure and regular chow (each nutritionally complete) or regular chow only. Intake of each food item, body weight and body fat mass were monitored throughout the study. RESULTS: The BFNA injections aimed at either the core or shell of the nucleus accumbens resulted in significantly attenuated intake of palatable diet, body weight gain and fat accretion, compared with vehicle control injections. The injection of BFNA in the core did not significantly change these parameters in chow-fed control rats. The injection of BFNA in the core and shell differentially affected intake of the two palatable food items: in the core, BFNA significantly reduced the intake of high-fat, but not of Ensure, whereas in the shell, it significantly reduced the intake of Ensure, but not of high-fat, compared with vehicle treatment. CONCLUSIONS: Endogenous micro-opioid receptor signaling in the nucleus accumbens core and shell is necessary for palatable diet-induced hyperphagia and obesity to fully develop in rats. Sweet and non-sweet fatty foods may be differentially processed in subcomponents of the ventral striatum.


Subject(s)
Body Weight/drug effects , Food Preferences/drug effects , Nucleus Accumbens/drug effects , Obesity/prevention & control , Receptors, Opioid, mu/antagonists & inhibitors , Animals , Body Weight/physiology , Dietary Fats/administration & dosage , Dietary Sucrose/administration & dosage , Food Preferences/psychology , Food, Formulated , Male , Naltrexone/administration & dosage , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/physiology
12.
Int J Obes (Lond) ; 33 Suppl 2: S8-13, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19528982

ABSTRACT

Powerful biological mechanisms evolved to defend adequate nutrient supply and optimal levels of body weight/adiposity. Low levels of leptin indicating food deprivation and depleted fat stores have been identified as the strongest signals to induce adaptive biological actions such as increased energy intake and reduced energy expenditure. In concert with other signals from the gut and metabolically active tissues, low leptin levels trigger powerful activation of multiple peripheral and brain systems to restore energy balance. It is not just neurons in the arcuate nucleus, but many other brain systems involved in finding potential food sources, smelling and tasting food, and learning to maximize rewarding effects of foods, that are affected by low leptin. Food restriction and fat depletion thus lead to a 'hungry' brain, preoccupied with food. By contrast, because of less (adaptive thrifty fuel efficiency) or lost (lack of predators) evolutionary pressure, the upper limits of body weight/adiposity are not as strongly defended by high levels of leptin and other signals. The modern environment is characterized by the increased availability of large amounts of energy-dense foods and increased presence of powerful food cues, together with minimal physical procurement costs and a sedentary lifestyle. Much of these environmental influences affect cortico-limbic brain areas concerned with learning and memory, reward, mood and emotion. Common obesity results when individual predisposition to deal with a restrictive environment, as engraved by genetics, epigenetics and/or early life experience, is confronted with an environment of plenty. Therefore, increased adiposity in prone individuals should be seen as a normal physiological response to a changed environment, not in the pathology of the regulatory system. The first line of defense should ideally lie in modifications to the environment and lifestyle. However, as such modifications will be slow and incomplete, it is equally important to gain better insight into how the brain deals with environmental stimuli and to develop behavioral strategies to better cope with them. Clearly, alternative therapeutic strategies such as drugs and bariatric surgery should also be considered to prevent or treat this debilitating disease. It will be crucial to understand the functional crosstalk between neural systems responding to metabolic and environmental stimuli, i.e. crosstalk between hypothalamic and cortico-limbic circuitry.


Subject(s)
Appetite Regulation/physiology , Hypothalamus/metabolism , Leptin/physiology , Neural Pathways/physiology , Obesity/physiopathology , Satiety Response/physiology , Adiposity/physiology , Energy Intake/physiology , Energy Metabolism/physiology , Feeding Behavior , Homeostasis/physiology , Humans , Limbic System/physiology , Obesity/psychology
13.
Neurogastroenterol Motil ; 20 Suppl 1: 64-72, 2008 May.
Article in English | MEDLINE | ID: mdl-18402643

ABSTRACT

Studying communication between the gut and the brain is as relevant and exciting as it has been since Pavlov's discoveries a century ago. Although the efferent limb of this communication has witnessed significant advances, it is the afferent, or sensory, limb that has recently made for exciting news. It is now clear that signals from the gut are crucial for the control of appetite and the regulation of energy balance, glucose homeostasis, and more. Ghrelin, discovered just a few years ago, is the first gut hormone that increases appetite, and it may be involved in eating disorders. The stable analogue of glucagon-like peptide-1 has rapidly advanced to one of the most promising treatment options for type-2 diabetes. Changes in the signalling patterns of these and other gut hormones best explain the remarkable capacity of gastric bypass surgery to lower food intake and excess body weight. Given the enormous societal implications of the obesity epidemic, these are no small feats. Together with the older gut hormone cholecystokinin and abundant vagal mechanosensors, the gut continuously sends information to the brain regarding the quality and quantity of ingested nutrients, not only important for satiation and meal termination, but also for the appetitive phase of ingestive behaviour and the patterning of meals within given environmental constraints. By acting not only on brainstem and hypothalamus, this stream of sensory information from the gut to the brain is in a position to generate a feeling of satisfaction and happiness as observed after a satiating meal and exploited in vagal afferent stimulation for depression.


Subject(s)
Brain/physiology , Gastrointestinal Hormones/physiology , Gastrointestinal Tract/physiology , Satiation/physiology , Vagus Nerve/physiology , Animals , Appetite Regulation/physiology , Humans , Neural Pathways/physiology , Personal Satisfaction
14.
Neuroscience ; 135(2): 611-25, 2005.
Article in English | MEDLINE | ID: mdl-16111819

ABSTRACT

Neural signaling by melanin-concentrating hormone and its receptor (SLC-1) has been implicated in the control of energy balance, but due to the wide distribution of melanin-concentrating hormone-containing fibers throughout the neuraxis, its critical sites of action for a particular effect have not been identified. The present study aimed to anatomically and functionally characterize melanin-concentrating hormone innervation of the rat caudal brainstem, as this brain area plays an important role in the neural control of ingestive behavior and autonomic outflow. Using retrograde tracing we demonstrate that a significant proportion (5-15%) of primarily perifornical and far-lateral hypothalamic melanin-concentrating hormone neurons projects to the dorsal vagal complex. In the caudal brainstem, melanin-concentrating hormone-ir axon profiles are distributed densely in most areas including the nucleus of the solitary tract, dorsal motor nucleus of the vagus, and sympathetic premotor areas in the ventral medulla. Close anatomical appositions can be demonstrated between melanin-concentrating hormone-ir axon profiles and tyrosine hydroxylase, GABA, GLP-1, NOS-expressing, and nucleus of the solitary tract neurons activated by gastric nutrient infusion. In medulla slice preparations, bath application of melanin-concentrating hormone inhibited in a concentration-dependent manner the amplitude of excitatory postsynaptic currents evoked by solitary tract stimulation via a pre-synaptic mechanism. Fourth ventricular administration of melanin-concentrating hormone (10 microg) in freely moving rats decreased core body temperature but did not change locomotor activity and food and water intake. We conclude that the rich hypothalamo-medullary melanin-concentrating hormone projections in the rat are mainly inhibitory to nucleus of the solitary tract neurons, but are not involved in the control of food intake. Projections to ventral medullary sites may play a role in the inhibitory effect of melanin-concentrating hormone on energy expenditure.


Subject(s)
Brain Stem/cytology , Energy Metabolism , Feeding Behavior/physiology , Hypothalamic Hormones/metabolism , Melanins/metabolism , Neural Pathways/metabolism , Neurons/metabolism , Pituitary Hormones/metabolism , Animals , Autonomic Nervous System/physiology , Axons/metabolism , Behavior, Animal , Body Temperature/drug effects , Body Temperature/physiology , Brain Stem/metabolism , Cell Count/methods , Cholera Toxin/metabolism , Dose-Response Relationship, Drug , Eating/drug effects , Eating/physiology , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Feeding Behavior/drug effects , Glucagon-Like Peptide 1/metabolism , Hypothalamic Hormones/pharmacology , Immunohistochemistry/methods , In Vitro Techniques , Male , Melanins/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Motor Activity/drug effects , Motor Activity/physiology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neural Inhibition/radiation effects , Neurons/cytology , Neurons/parasitology , Nitric Oxide Synthase/metabolism , Patch-Clamp Techniques/methods , Pituitary Hormones/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors , Tyrosine 3-Monooxygenase/metabolism , gamma-Aminobutyric Acid/metabolism
15.
Neurogastroenterol Motil ; 16 Suppl 1: 28-33, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15066001

ABSTRACT

Here we discuss the neuroanatomy of extrinsic gastrointestinal (GI) afferent neurones, the relationship between structure and function and the role of afferents in disease. Three pathways connect the gut to the central nervous system: vagal afferents signal mainly from upper GI regions, pelvic afferents mainly from the colorectal region and splanchnic afferents from throughout. Vagal afferents mediate reflex regulation of gut function and behaviour, operating mainly at physiological levels. There are two major functional classes - tension receptors, responding to muscular contraction and distension, and mucosal receptors. The function of vagal endings correlates well with their anatomy: tracing studies show intramuscular arrays (IMAs) and intraganglionic laminar endings (IGLEs); IGLEs are now known to respond to tension. Functional mucosal receptors correlate with endings traced to the lamina propria. Pelvic afferents serve similar functions to vagal afferents, and additionally mediate both innocuous and noxious sensations. Splanchnic afferents comprise mucosal and stretch-sensitive afferents with low thresholds in addition to high-threshold serosal/mesenteric afferents suggesting diverse roles. IGLEs, probably of pelvic origin, have been identified recently in the rectum and respond similarly to gastric vagal IGLEs. Gastrointestinal afferents may be sensitized or inhibited by chemical mediators released from several cell types. Whether functional changes have anatomical correlates is not known, but it is likely that they underlie diseases involving visceral hypersensitivity.


Subject(s)
Digestive System/innervation , Enteric Nervous System/anatomy & histology , Nerve Endings/anatomy & histology , Visceral Afferents/anatomy & histology , Animals , Enteric Nervous System/physiology , Humans , Nerve Endings/physiology , Neuronal Plasticity/physiology , Visceral Afferents/physiopathology
16.
Neurogastroenterol Motil ; 14(6): 677-88, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12464090

ABSTRACT

Our aim was to evaluate topographically specific gastric motility changes induced by graded vagal activation. A recently developed method of constructing spatio-temporal maps of motility from video movies was adapted to the in vitro perfused guinea-pig stomach with an intact vagal nerve supply. In the unstimulated preparation, spontaneous activity was low or absent. Bilateral vagal stimulation with frequencies as low as 0.2 Hz triggered weak anally, and in some cases orally, propagating antral contractions at rates of about 5-6 min-1. Upon stimulation with higher frequencies, antral contractions increased significantly in length (starting more proximally) and amplitude, and produced large pressure peaks of up to 25 hPa, with maximal effects at 2-4 Hz. In contrast, the speed of propagation and the interval between peristaltic waves did not change with vagal stimulation at any frequency. Vagal stimulation also produced a significant and frequency-dependent enlargement of the fundus with a maximal effect at 4 Hz. It is concluded that a very low tonic vagal activity is apparently necessary and sufficient to express basic antral motility, while more sustained vagal activity is necessary for high-amplitude gastric contractions and significant sustained fundic relaxation. The constant interval between propagating contractions supports the concept that vagal input impinges on intrinsic enteric neural circuits that have a modulatory role in the myogenic mechanism underlying slow-wave peristalsis, rather than directly on gastric musculature.


Subject(s)
Gastrointestinal Motility/physiology , Muscle Contraction/physiology , Stomach/innervation , Video Recording , Animals , Electric Stimulation , Female , Guinea Pigs , Image Processing, Computer-Assisted , Male , Muscle, Smooth/physiology , Organ Culture Techniques , Vagus Nerve/physiology , Video Recording/methods
17.
Brain Res ; 915(2): 143-54, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11595203

ABSTRACT

Vagal mechano- and chemosensors in the gastrointestinal tract and the portal-hepatic axis signaling the arrival of nutrients are major determinants of the satiation process. Although glutamate and its various receptor subtypes have been shown to transmit gustatory and cardiovascular sensory information at the level of the solitary nucleus (nucleus tractus solitarius; NTS), their involvement in the transmission of gastrointestinal satiety signals is not clear. Gastrointestinal sensors were stimulated by gastric balloon distension or by intraduodenal infusion of either linoleic acid or glucose in chronically catheterized, non-anesthetized rats, leading to activation of second order neurons in the NTS as detected by c-Fos immunohistochemistry. Subsequent (double)-immunohistochemistry for either NMDA or AMPA glutamate receptors was used to determine the percentage of activated neurons expressing a particular receptor subtype. Gastric distension and duodenal nutrient stimuli produced slightly, but significantly different patterns of c-Fos induction in the dorsal vagal complex. Expression of NMDA receptors, as detected by a NR2ab subunit-specific antibody, was abundant throughout the dorsal medulla. The percentage of neurons in the NTS activated by gastric distension (63.9+/-2.9%), linoleic acid (62.8+/-1.4%), and glucose (64.1+/-1.4%), expressing NMDA receptor was similar. Expression of AMPA receptors, as detected by a GLUR2/3 subunit-specific antibody, was equally abundant throughout the dorsal medulla. Again, the percentage of activated neurons expressing GLUR2/3 was similar for the gastric distension (59.8-65.6%) and duodenal linoleic acid (60.6-67.0%) stimuli, and for the various subnuclei of the NTS. Finally, GLUR1-specific immunoreactivity was much less abundant, with only a small percentage of distension-activated (4.4+/-0.4%) and linoleic acid-activated (5.1+/-0.4%) neurons expressing this receptor subunit. The results suggest a widespread, general involvement of both NMDA and AMPA receptors in primary afferent signal transmission at the level of the NTS, with no differential recruitment of the examined receptor subtypes by the different gastrointestinal sensory stimuli. This may indicate a high degree of convergence among sensory signals, or alternatively, the presence of other transmission systems such as peptides referring sensory specificity to second order neurons.


Subject(s)
Appetite Regulation/physiology , Brain Stem/metabolism , Digestive System Physiological Phenomena , Neurons/metabolism , Receptors, AMPA/biosynthesis , Receptors, N-Methyl-D-Aspartate/biosynthesis , Animals , Appetite Regulation/drug effects , Brain Stem/drug effects , Digestive System/drug effects , Glucose/pharmacology , Linoleic Acid/pharmacology , Male , Neurons/drug effects , Neurons/physiology , Rats , Rats, Sprague-Dawley , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/physiology
18.
Cell Tissue Res ; 305(1): 11-23, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11512662

ABSTRACT

One of the physiological functions of circulating cholecystokinin (CCK) is in the control of the pyloric sphincter and the subsequent delivery of nutrients to the small intestine. In order to identify the site(s) of action of CCK in the gastropyloric region, we performed immunohistochemistry using an antibody directed to the C-terminal region of the cholecystokinin A receptor (CCKAR). In the rat, cells that display strong CCKAR immunoreactivity and fit the morphological description of interstitial cells of Cajal (ICC) were found in the distal sphincter muscle and in the circular muscle of the proximal duodenum. Double labeling showed that these cells coexpressed vimentin, but that not all vimentin-positive cells expressed CCKAR. Confirmation that the CCKAR-expressing cells were ICC also came from kit double-labeling experiments in mice. In addition to ICC, circular smooth muscle cells at the tip of the comma-shaped sphincter muscle, but not elsewhere, also exhibited strong, membrane-bound CCKAR immunoreactivity. With higher antibody concentrations, the entire circular muscle displayed moderate CCKAR immunoreactivity, suggesting that circular smooth muscle cells express low levels of CCKAR. Select neurons in the myenteric ganglia near the sphincter muscle proper, the distal antrum, and proximal duodenum, as well as a few single neurons in the submucosa, also expressed strong CCKAR immunoreactivity. Finally, CCKAR-immunoreactive ICC and neurons were not specifically related to vagal afferent intramuscular and intraganglionic endings, and vagal afferents themselves did not exhibit any CCKAR immunoreactivity. These results suggest a role for ICC and enteric neurons in the mediation of CCK effects on pyloric sphincter pressure in addition to direct effects of the hormone on circular smooth muscle.


Subject(s)
Enteric Nervous System/chemistry , Muscle, Smooth/chemistry , Neurons/chemistry , Pylorus/chemistry , Receptors, Cholecystokinin/analysis , Afferent Pathways , Animals , Antibodies , Biological Clocks , Enteric Nervous System/cytology , Gastric Emptying/physiology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/chemistry , Muscle, Smooth/cytology , Muscle, Smooth/innervation , Nerve Fibers/chemistry , Neurons/ultrastructure , Proto-Oncogene Proteins c-kit/analysis , Pylorus/cytology , Pylorus/innervation , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A , Receptors, Cholecystokinin/immunology , Vagus Nerve/cytology , Vimentin/analysis
19.
Am J Physiol Regul Integr Comp Physiol ; 280(5): R1371-81, 2001 May.
Article in English | MEDLINE | ID: mdl-11294756

ABSTRACT

Mechano- and chemosensitive extrinsic primary afferents innervating the gastrointestinal tract convey important information regarding the state of ingested nutrients and specific motor patterns to the central nervous system via splanchnic and vagal nerves. Little is known about the organization of peripheral receptive sites of afferents and their correspondence to morphologically identified terminal structures. Mechano- and chemosensory characteristics and receptive fields of single vagal fibers innervating the stomach as well as lumbar splanchnic nerves innervating the distal colon were identified using an in vitro perifusion system. Twenty-three (17%) of one-hundred thirty-six vagal units identified were found to have multiple, punctate receptive fields, up to 35 mm apart, and were distributed throughout the stomach. Evidence was based on similarity of generated spike forms, occlusion, and latency determinations. Most responded with brief bursts of activity to mucosal stroking with von Frey hairs (10-200 mg) but not to stretch, and 32% responded to capsaicin (10(-5) M). They were classified as rapidly adapting mucosal receptors. Four (8%) of fifty-three single units recorded from the lumbar splanchnic nerve had more than one, punctate receptive field in the distal colon, up to 40 mm apart. They responded to blunt probing, particularly from the serosal side, and variously to chemical stimulation with 5-hydroxytryptamine and capsaicin. We conclude that a proportion of gastrointestinal mechanosensors has multiple receptive fields and suggest that they integrate mechanical and chemical information from an entire organ, constituting the generalists in visceral sensation.


Subject(s)
Colon/innervation , Gastric Mucosa/innervation , Intestinal Mucosa/innervation , Mechanoreceptors/physiology , Spinal Cord/physiology , Stomach/innervation , Vagus Nerve/physiology , Afferent Pathways/physiology , Animals , Axons/physiology , Capsaicin/pharmacology , Chemoreceptor Cells/physiology , Electric Stimulation , Evoked Potentials , Female , In Vitro Techniques , Muscle Contraction , Muscle, Smooth/innervation , Nerve Fibers/physiology , Perfusion , Rats , Rats, Sprague-Dawley , Reaction Time
20.
Brain Res ; 896(1-2): 153-6, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11277984

ABSTRACT

Expression of CART (cocaine-amphetamine-regulated transcript) in the rat hypothalamus is modulated by nutritional status, and injection of synthetic CART peptide into the forebrain ventricular system suppresses food intake, indicating a possible role in hypothalamic control of energy homeostasis. Its recent identification in cell bodies and central terminals of vagal afferent neurons additionally suggests a role in brainstem mechanisms of meal termination and satiety. We demonstrate here that CART[55-102] (0.2 nmol) suppresses short-term sucrose intake and overnight chow intake in non-food-deprived rats even more when delivered into the fourth ventricle as compared to the lateral ventricle. At the threshold dose (0.02-0.08 nmol) no readily noticeable motor impairments were observed. The results are consistent, but do not prove a site of action within the brainstem, possibly in mediating vagal satiety signals at the level of the NTS.


Subject(s)
Dietary Sucrose/pharmacology , Eating/drug effects , Nerve Tissue Proteins/pharmacology , Afferent Pathways/physiology , Animals , Brain Stem/drug effects , Brain Stem/physiology , Fourth Ventricle , Injections, Intraventricular , Male , Rats , Rats, Sprague-Dawley , Satiation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...