Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Front Neurol ; 9: 202, 2018.
Article in English | MEDLINE | ID: mdl-29643832

ABSTRACT

[This corrects the article on p. 32 in vol. 4, PMID: 23565108.].

2.
Nutr J ; 12: 114, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23924506

ABSTRACT

The incidence of obesity has increased dramatically over the past several years, and in parallel, so has the prevalence of type 2 diabetes (T2D). Numerous studies have demonstrated that both obesity and T2D are associated with lower cognitive performance, cognitive decline, and dementia. Intake of dietary fructose has also increased. In fact, high-fructose corn syrup (HFCS) accounts for as much as 40% of caloric sweeteners used in the United States. Given the increase in the incidence of Alzheimer's disease (AD), characterized by an age-related decline in memory and cognitive functioning, in this report we review the effects of obesity on cognitive performance and the impact of high fructose intake in promoting cognitive decline. The paper then considers the effects of omega-3 fatty acids (FAs), which have been linked to promising results in cognitive function including ameliorating the impact of a high-fructose diet.


Subject(s)
Cognition Disorders/physiopathology , Diet , Fructose/adverse effects , Obesity/physiopathology , Cognition/physiology , Cognition Disorders/complications , Cognition Disorders/etiology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/physiopathology , Fatty Acids, Omega-3/administration & dosage , Humans , Obesity/etiology , Sucrose/adverse effects , Sweetening Agents/adverse effects , United States
3.
Front Neurol ; 4: 32, 2013.
Article in English | MEDLINE | ID: mdl-23565108

ABSTRACT

Ischemic stroke continues to be one of the most challenging diseases in translational neurology. Tissue plasminogen activator (tPA) remains the only approved treatment for acute ischemic stroke, but its use is limited to the first hours after stroke onset due to an increased risk of hemorrhagic transformation over time resulting in enhanced brain injury. In this review we discuss the role of matrix metalloproteinases (MMPs) in blood-brain barrier (BBB) disruption as a consequence of ischemic stroke. MMP-9 in particular appears to play an important role in tPA-associated hemorrhagic complications. Reactive oxygen species can enhance the effects of tPA on MMP activation through the loss of caveolin-1 (cav-1), a protein encoded in the cav-1 gene that serves as a critical determinant of BBB permeability. This review provides an overview of MMPs' role in BBB breakdown during acute ischemic stroke. The possible role of MMPs in combination treatment of acute ischemic stroke is also examined.

4.
Brain Behav ; 2(5): 661-77, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23139911

ABSTRACT

Prescription stimulants are often used to treat attention deficit hyperactivity disorder (ADHD). Drugs like methylphenidate (Ritalin, Concerta), dextroamphetamine (Dexedrine), and dextroamphetamine-amphetamine (Adderall) help people with ADHD feel more focused. However, misuse of stimulants by ADHD and nonaffected individuals has dramatically increased over recent years based on students' misconceptions or simple lack of knowledge of associated risks. In this review, we discuss recent advances in the use and increasing misuse of prescription stimulants among high school and college students and athletes. Given the widespread belief that stimulants enhance performance, there are in fact only a few studies reporting the cognitive enhancing effects of stimulants in ADHD and nonaffected individuals. Student athletes should be apprised of the very serious consequences that can emerge when stimulants are used to improve sports performance. Moreover, misuse of stimulants is associated with dangers including psychosis, myocardial infarction, cardiomyopathy, and even sudden death. As ADHD medications are prescribed for long-term treatment, there is a need for long-term safety studies and education on the health risks associated with misuse is imperative.

5.
Springerplus ; 1: 2, 2012.
Article in English | MEDLINE | ID: mdl-23984220

ABSTRACT

Chronic traumatic encephalopathy (CTE) is a form of neurodegeneration that results from repetitive brain trauma. Not surprisingly, CTE has been linked to participation in contact sports such as boxing, hockey and American football. In American football getting "dinged" equates to moments of dizziness, confusion, or grogginess that can follow a blow to the head. There are approximately 100,000 to 300,000 concussive episodes occurring in the game of American football alone each year. It is believed that repetitive brain trauma, with or possibly without symptomatic concussion, sets off a cascade of events that result in neurodegenerative changes highlighted by accumulations of hyperphosphorylated tau and neuronal TAR DNA-binding protein-43 (TDP-43). Symptoms of CTE may begin years or decades later and include a progressive decline of memory, as well as depression, poor impulse control, suicidal behavior, and, eventually, dementia similar to Alzheimer's disease. In some individuals, CTE is also associated with motor neuron disease similar to amyotrophic lateral sclerosis. Given the millions of athletes participating in contact sports that involve repetitive brain trauma, CTE represents an important public health issue. In this review, we discuss recent advances in understanding the etiology of CTE. It is now known that those instances of mild concussion or "dings" that we may have previously not noticed could very well be causing progressive neurodegenerative damage to a player's brain. In the future, focused and intensive study of the risk factors could potentially uncover methods to prevent and treat this disease.

6.
J Transl Med ; 9: 202, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-22115311

ABSTRACT

Obesity is a chronic disease characterized by persistent low-grade inflammation with alterations in gut motility. Motor abnormalities suggest that obesity has effects on the enteric nervous system (ENS), which controls virtually all gut functions. Recent studies have revealed that the gut microbiota can affect obesity and increase inflammatory tone by modulating mucosal barrier function. Furthermore, the observation that inflammatory conditions influence the excitability of enteric neurons may add to the gut dysfunction in obesity. In this article, we discuss recent advances in understanding the role of gut microbiota and inflammation in the pathogenesis of obesity and obesity-related gastrointestinal dysfunction. The potential contribution of sirtuins in protecting or regulating the circuitry of the ENS under inflamed states is also considered.


Subject(s)
Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiopathology , Metagenome/physiology , Obesity/pathology , Obesity/physiopathology , Sirtuins/metabolism , Animals , Enteric Nervous System/pathology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/pathology , Humans , Inflammation/complications , Inflammation/microbiology , Inflammation/pathology , Inflammation/physiopathology , Obesity/complications , Obesity/microbiology
7.
J Transl Med ; 9: 129, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21810260

ABSTRACT

Cigarette smoke is a major risk factor for a number of diseases including lung cancer and respiratory infections. Paradoxically, it also contains nicotine, an anti-inflammatory alkaloid. There is increasing evidence that smokers have a lower incidence of some inflammatory diseases, including ulcerative colitis, and the protective effect involves the activation of a cholinergic anti-inflammatory pathway that requires the α7 nicotinic acetylcholine receptor (α7nAChR) on immune cells. Obesity is characterized by chronic low-grade inflammation, which contributes to insulin resistance. Nicotine significantly improves glucose homeostasis and insulin sensitivity in genetically obese and diet-induced obese mice, which is associated with suppressed adipose tissue inflammation. Inflammation that results in disruption of the epithelial barrier is a hallmark of inflammatory bowel disease, and nicotine is protective in ulcerative colitis. This article summarizes current evidence for the anti-inflammatory effects of nicotine in obesity and ulcerative colitis. Selective agonists for the α7nAChR could represent a promising pharmacological strategy for the treatment of inflammation in obesity and ulcerative colitis. Nevertheless, we should keep in mind that the anti-inflammatory effects of nicotine could be mediated via the expression of several nAChRs on a particular target cell.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis, Ulcerative/drug therapy , Nicotine/therapeutic use , Obesity/drug therapy , Animals , Choline/metabolism , Colitis, Ulcerative/pathology , Humans , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/metabolism , alpha7 Nicotinic Acetylcholine Receptor
8.
Nutr Metab (Lond) ; 7: 79, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20939923

ABSTRACT

Chronic fatigue syndrome (CFS) is a debilitating disease characterized by unexplained disabling fatigue and a combination of accompanying symptoms the pathology of which is incompletely understood. Many CFS patients complain of gut dysfunction. In fact, patients with CFS are more likely to report a previous diagnosis of irritable bowel syndrome (IBS), a common functional disorder of the gut, and experience IBS-related symptoms. Recently, evidence for interactions between the intestinal microbiota, mucosal barrier function, and the immune system have been shown to play a role in the disorder's pathogenesis.Studies examining the microecology of the gastrointestinal (GI) tract have identified specific microorganisms whose presence appears related to disease; in CFS, a role for altered intestinal microbiota in the pathogenesis of the disease has recently been suggested. Mucosal barrier dysfunction promoting bacterial translocation has also been observed. Finally, an altered mucosal immune system has been associated with the disease. In this article, we discuss the interplay between these factors in CFS and how they could play a significant role in GI dysfunction by modulating the activity of the enteric nervous system, the intrinsic innervation of the gut.If an altered intestinal microbiota, mucosal barrier dysfunction, and aberrant intestinal immunity contribute to the pathogenesis of CFS, therapeutic efforts to modify gut microbiota could be a means to modulate the development and/or progression of this disorder. For example, the administration of probiotics could alter the gut microbiota, improve mucosal barrier function, decrease pro-inflammatory cytokines, and have the potential to positively influence mood in patients where both emotional symptoms and inflammatory immune signals are elevated. Probiotics also have the potential to improve gut motility, which is dysfunctional in many CFS patients.

9.
J Neuroinflammation ; 7: 37, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20615234

ABSTRACT

Inflammatory bowel disease is a chronic intestinal inflammatory condition, the pathology of which is incompletely understood. Gut inflammation causes significant changes in neurally controlled gut functions including cramping, abdominal pain, fecal urgency, and explosive diarrhea. These symptoms are caused, at least in part, by prolonged hyperexcitability of enteric neurons that can occur following the resolution of colitis. Mast, enterochromaffin and other immune cells are increased in the colonic mucosa in inflammatory bowel disease and signal the presence of inflammation to the enteric nervous system. Inflammatory mediators include 5-hydroxytryptamine and cytokines, as well as reactive oxygen species and the production of oxidative stress. This review will discuss the effects of inflammation on enteric neural activity and potential therapeutic strategies that target neuroinflammation in the enteric nervous system.


Subject(s)
Enteric Nervous System , Inflammation/pathology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Animals , Cell Death/physiology , Enteric Nervous System/immunology , Enteric Nervous System/pathology , Enteric Nervous System/physiopathology , Humans , Inflammation/immunology , Inflammatory Bowel Diseases/therapy , Intestines/innervation , Intestines/pathology , Intestines/physiology , Intestines/physiopathology , Oxidative Stress , Receptors, Neurotransmitter/metabolism , Signal Transduction/physiology
10.
J Transl Med ; 7: 97, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19919699

ABSTRACT

Acute ischemic stroke is the third leading cause of death in industrialized countries and the most frequent cause of permanent disability in adults worldwide. Despite advances in the understanding of the pathophysiology of cerebral ischemia, therapeutic options remain limited. Only recombinant tissue-plasminogen activator (rt-PA) for thrombolysis is currently approved for use in the treatment of this devastating disease. However, its use is limited by its short therapeutic window (three hours), complications derived essentially from the risk of hemorrhage, and the potential damage from reperfusion/ischemic injury. Two important pathophysiological mechanisms involved during ischemic stroke are oxidative stress and inflammation. Brain tissue is not well equipped with antioxidant defenses, so reactive oxygen species and other free radicals/oxidants, released by inflammatory cells, threaten tissue viability in the vicinity of the ischemic core. This review will discuss the molecular aspects of oxidative stress and inflammation in ischemic stroke and potential therapeutic strategies that target neuroinflammation and the innate immune system. Currently, little is known about endogenous counterregulatory immune mechanisms. However, recent studies showing that regulatory T cells are major cerebroprotective immunomodulators after stroke suggest that targeting the endogenous adaptive immune response may offer novel promising neuroprotectant therapies.


Subject(s)
Inflammation , Stroke , Animals , Chemokines/immunology , Cytokines/immunology , Humans , Inflammation/pathology , Inflammation/physiopathology , Inflammation/therapy , Matrix Metalloproteinases/metabolism , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/therapeutic use , Oxidative Stress , Recovery of Function , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Stroke/pathology , Stroke/physiopathology , Stroke/therapy , T-Lymphocytes/immunology
11.
Regul Pept ; 132(1-3): 9-16, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16125803

ABSTRACT

Orexin A (OXA) increases food intake and inhibits fasting small bowel motility in rats. The aim of this study was to examine the effect of exogenous OXA and endogenous OXA on gastric emptying, acid secretion, glucose metabolism and distribution of orexin immunoreactivity in the stomach. Rats equipped with a gastric fistula were subjected to intravenous (IV) infusion of OXA or the selective orexin-1 receptor (OX1R) antagonist SB-334867-A during saline or pentagastrin infusion. Gastric emptying was studied with a liquid non-nutrient or nutrient, using 51Cr as radioactive marker. Gastric retention was measured after a 20-min infusion of OXA or SB-334867-A. Plasma concentrations of OXA, insulin, glucagon, glucose and gastrin were studied. Immunohistochemistry against OXA, OX1R and gastrin in gastric tissue was performed. OXA alone had no effect on either acid secretion or gastric emptying. SB-334867-A inhibited both basal and pentagastrin-induced gastric acid secretion and increased gastric retention of the liquid nutrient, but not PEG 4000. Plasma gastrin levels were unchanged by IV OXA or SB-334867-A. Plasma OXA levels decreased after intake of the nutrient meal and infusion of the OX1R antagonist. Only weak effects were seen on plasma glucose and insulin by OXA. Immunoreactivity to OXA and OX1R were found in the mucosa, myenteric cells bodies and varicose nerve fibers in ganglia and circular muscle of the stomach. In conclusion, endogenous OXA influences gastric emptying of a nutrient liquid and gastric acid secretion independent of gastrin. This indicates a role for endogenous OXA, not only in metabolic homeostasis, but also in the pre-absorptive processing of nutrients in the gut.


Subject(s)
Gastric Acid/metabolism , Gastric Emptying/physiology , Intracellular Signaling Peptides and Proteins/physiology , Neuropeptides/physiology , Nitric Oxide Synthase Type I/metabolism , Animals , Blood Glucose/metabolism , Gastrins/blood , Glucagon/blood , Immunohistochemistry , Insulin/blood , Intracellular Signaling Peptides and Proteins/blood , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Neuropeptides/blood , Neuropeptides/pharmacology , Orexins , Rats , Rats, Sprague-Dawley
12.
Regul Pept ; 131(1-3): 59-65, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16040140

ABSTRACT

Ghrelin is a gut peptide that is secreted from the stomach and stimulates food intake. There are ghrelin receptors throughout the gut and intracerebroventricular ghrelin has been shown to increase gastric acid secretion. The aim of the present study was to examine the effects of peripherally administered ghrelin on gastric emptying of a non-nutrient and nutrient liquid, as well as, basal and pentagastrin-stimulated gastric acid secretion in awake rats. In addition, gastric contractility was studied in vitro. Rats equipped with a gastric fistula were subjected to an intravenous infusion of ghrelin (10-500 pmol kg(-1) min(-1)) during saline or pentagastrin (90 pmol kg(-1) min(-1)) infusion. After administration of polyethylene glycol (PEG) 4000 with 51Cr as radioactive marker, or a liquid nutrient with (51)Cr, gastric retention was measured after a 20-min infusion of ghrelin (500 pmol kg(-1) min(-1)). In vitro isometric contractions of segments of rat gastric fundus were studied (10(-9) to 10(-6) M). Ghrelin had no effect on basal acid secretion, but at 500 pmol kg(-1) min(-1) ghrelin significantly decreased pentagastrin-stimulated acid secretion. Ghrelin had no effect on gastric emptying of the nutrient liquid, but significantly increased gastric emptying of the non-nutrient liquid. Ghrelin contracted fundus muscle strips dose-dependently (pD2 of 6.93+/-0.7). Ghrelin IV decreased plasma orexin A concentrations and increased plasma somatostatin concentrations. Plasma gastrin concentrations were unchanged during ghrelin infusion. Thus, ghrelin seems to not only effect food intake but also gastric motor and secretory function indicating a multifunctional role for ghrelin in energy homeostasis.


Subject(s)
Gastric Acid/metabolism , Gastric Emptying/drug effects , Peptide Hormones/metabolism , Peptide Hormones/pharmacology , Animals , Gastrins/blood , Ghrelin , Glucagon/blood , Glucose/metabolism , In Vitro Techniques , Insulin/blood , Intracellular Signaling Peptides and Proteins/blood , Male , Muscle Contraction/drug effects , Muscle Contraction/physiology , Neuropeptides/blood , Orexins , Pentagastrin/metabolism , Pentagastrin/pharmacology , Peptide Hormones/administration & dosage , Polyethylene Glycols/administration & dosage , Rats , Rats, Sprague-Dawley , Somatostatin/blood
13.
Am J Physiol Gastrointest Liver Physiol ; 285(5): G992-G1003, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12829438

ABSTRACT

The enteric nervous system (ENS) contains glutamatergic neurons, transporters, and functional ionotropic and groups I and II metabotropic glutamate receptors (mGluRs). The aim of this study was to determine whether the ENS contains functional group III mGluRs. RT-PCR demonstrated the expression of mGluR7 and mGluR8 mRNA in rat myenteric ganglia. Western blot analysis confirmed the presence of mGluR8 protein. Immunocytochemistry, in conjunction with confocal microscopy, demonstrated mGluR8 immunoreactivity in the ENS of several species, including humans. mGluR8 immunoreactivity was localized to the membrane of nerve cell bodies that received glutamatergic input. Significant receptor internalization of mGluR8 was observed on activation, and localization to membrane was observed on blocking with the mGluR III antagonist (RS)-cyclopropyl-4-phosphonophenylglycine (CPPG). mGluR8-positive myenteric neurons contained glutamate or nitric oxide synthase (NOS), a marker of inhibitory motorneurons. Enteric group III mGluRs are functional because mGluR8 agonists inhibited forskolin-induced accumulation of cAMP in isolated myenteric ganglia, and CPPG reduced this effect. In addition, an accelerating effect on guinea pig colonic motility was observed after the application of mGluR8 agonists. Increase in motility was specific, because CPPG inhibited it. Moreover, in the presence of hexamethonium or Nomega-nitro-l-arginine methyl ester, an inhibitor of NOS, responses caused by mGluR8 agonists were abolished. mGluR8 agonists also increased longitudinal muscle contractions. These findings suggest that mGluR8 agonists increase motility by inhibiting nitrergic relaxation and possibly by facilitating cholinergic contractions.


Subject(s)
Enteric Nervous System/physiology , Receptors, Metabotropic Glutamate/physiology , Animals , Blotting, Western , Colforsin/pharmacology , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/biosynthesis , Electric Stimulation , Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Female , Ganglia/metabolism , Gastrointestinal Motility/physiology , Immunohistochemistry , Microscopy, Confocal , Muscle, Smooth/physiology , Myenteric Plexus/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
14.
Diabetes ; 52(1): 111-7, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12502500

ABSTRACT

Orexins (hypocretins) are novel neuropeptides that appear to play a role in the regulation of energy balances. Orexin-A (OXA) increases food intake in rodents, and fasting activates OXA neurons in both the lateral hypothalamic area and gut. OXA is also found in the endocrine pancreas; however, little is known about its release or functional significance. In this study, we show that depolarizing stimuli evoke the release of OXA from rat pancreatic islets in a calcium-dependent manner. Moreover, OXA release is stimulated by low glucose (2.8 mmol/l), similar to glucagon secretion, and inhibited by high glucose (16.7 mmol/l). Fasting increases plasma OXA, supporting the idea that orexin is released in response to hypoglycemia. Cells that secrete glucagon and insulin contain OXA and both cell types express orexin receptors. OXA increases glucagon secretion and decreases glucose-stimulated insulin release from isolated islets. OXA infusion increases plasma glucagon and glucose levels and decreases plasma insulin in fasted rats. We conclude that orexin-containing islet cells, like those in the brain and gut, are glucosensitive and part of a network of glucose "sensing" cells that becomes activated when blood glucose levels fall. OXA may modulate islet hormone secretion to maintain blood glucose levels during fasting.


Subject(s)
Carrier Proteins/metabolism , Glucose/physiology , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/metabolism , Neuropeptides/metabolism , Animals , Blood Glucose/analysis , Carrier Proteins/pharmacology , Culture Techniques , Fasting/blood , Female , Glucagon/blood , Glucagon/metabolism , Glucose/pharmacology , Immunohistochemistry , Insulin/blood , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Male , Neuropeptides/pharmacology , Orexin Receptors , Orexins , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Receptors, Neuropeptide/metabolism , Stimulation, Chemical
15.
Am J Physiol Gastrointest Liver Physiol ; 283(6): G1282-9, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12388194

ABSTRACT

The enteric nervous system (ENS) contains functional ionotropic and group I metabotropic glutamate (mGlu) receptors. In this study, we determined whether enteric neurons express group II mGlu receptors and the effects of mGlu receptor activation on voltage-gated Ca(2+) currents in these cells. (2R,4R)-4-aminopyrrolidine-2,4-dicarboxylate (2R,4R-APDC), a group II mGlu receptor agonist, reversibly suppressed the Ba(2+) current in myenteric neurons isolated from the guinea pig ileum. Significant inhibition was also produced by L-glutamate and the group II mGlu receptor agonists, (2S,2'R,3'R)-2-(2',3'-dicarboxycyclopropyl)glycine (DCG-IV) and (2S,1'S,2'S)-2-(2-carboxycyclopropyl)glycine (L-CCG-I), with a rank order potency of 2R,4R-APDC > DCG-IV > L-glutamate > L-CCG-I, and was reduced by the group II mGlu receptor antagonist LY-341495. Pretreatment of neurons with pertussis toxin (PTX) reduced the action of mGlu receptor agonists, suggesting participation of G(i)/G(o) proteins. Finally, omega-conotoxin GVIA blocked current suppression by DCG-IV, suggesting modulation of N-type calcium channels. mGlu2/3 receptor immunoreactivity was displayed by neurons in culture and in the submucosal and myenteric plexus of the ileum. A subset of these cells displayed a glutamatergic phenotype as shown by the expression of vesicular glutamate transporter 2. These results provide the first evidence for functional group II mGlu receptors in the ENS and show that these receptors are PTX sensitive and negatively coupled to N-type calcium channels. Inhibition of N-type calcium channels produced by activation of group II mGlu receptors may modulate enteric neurotransmission.


Subject(s)
Calcium Channels/physiology , Glycine/analogs & derivatives , Myenteric Plexus/physiology , Neurons/physiology , Proline/analogs & derivatives , Receptors, Metabotropic Glutamate/physiology , Animals , Barium/metabolism , Cadmium/pharmacology , Calcium/pharmacology , Calcium Channel Blockers/pharmacology , Cyclopropanes/pharmacology , Electric Conductivity , GTP-Binding Proteins/physiology , Glutamic Acid/pharmacology , Glycine/pharmacology , Guinea Pigs , Ion Channel Gating , Male , Neurons/chemistry , Nifedipine/pharmacology , Pertussis Toxin/pharmacology , Proline/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/analysis , omega-Conotoxin GVIA/pharmacology
16.
Am J Physiol Endocrinol Metab ; 282(6): E1324-33, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12006363

ABSTRACT

Pancreatic islets contain ionotropic glutamate receptors that can modulate hormone secretion. The purpose of this study was to determine whether islets express functional group III metabotropic glutamate (mGlu) receptors. RT-PCR analysis showed that rat islets express the mGlu8 receptor subtype. mGlu8 receptor immunoreactivity was primarily displayed by glucagon-secreting alpha-cells and intrapancreatic neurons. By demonstrating the immunoreactivities of both glutamate and the vesicular glutamate transporter 2 (VGLUT2) in these cells, we established that alpha-cells express a glutamatergic phenotype. VGLUT2 was concentrated in the secretory granules of islet cells, suggesting that glutamate might play a role in the regulation of glucagon processing. The expression of mGlu8 by glutamatergic cells also suggests that mGlu8 may function as an autoreceptor to regulate glutamate release. Pancreatic group III mGlu receptors are functional because mGlu8 receptor agonists inhibited glucagon release and forskolin-induced accumulation of cAMP in isolated islets, and (R,S)-cyclopropyl-4-phosphonophenylglycine, a group III mGlu receptor antagonist, reduced these effects. Because excess glucagon secretion causes postprandial hyperglycemia in patients with type 2 diabetes, group III mGlu receptor agonists could be of value in the treatment of these patients.


Subject(s)
Glycine/analogs & derivatives , Islets of Langerhans/chemistry , Islets of Langerhans/physiology , Receptors, Metabotropic Glutamate/analysis , Receptors, Metabotropic Glutamate/physiology , Animals , Benzoates/pharmacology , Cell Membrane/chemistry , Colforsin/pharmacology , Cyclic AMP/metabolism , Cytoplasmic Granules/chemistry , DNA Primers , Excitatory Amino Acid Antagonists/pharmacology , Female , Gene Expression , Glucagon/metabolism , Glutamic Acid/analysis , Glycine/pharmacology , Microscopy, Immunoelectron , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/genetics , Reverse Transcriptase Polymerase Chain Reaction , Synaptophysin/analysis , Tissue Distribution
17.
Endocr Rev ; 23(1): 1-15, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11844742

ABSTRACT

Orexins (hypocretins) are a novel pair of neuropeptides implicated in the regulation of energy balances and arousal. Previous reports have indicated that orexins are produced only in the lateral hypothalamic area, although orexin-containing nerve fibers were observed throughout the neuroaxis. Recent evidence shows that orexins and functional orexin receptors are found in the periphery. Vagal and spinal primary afferent neurons, enteric neurons, and endocrine cells in both the gut and pancreas display orexin- and orexin receptor-like immunoreactivity. Orexins excite secretomotor neurons in the guinea pig gut and modulate gastric and intestinal motility and secretion. In addition, orexins modulate hormone release from pancreatic endocrine cells. Moreover, fasting up-regulates the phosphorylated form of cAMP response element binding protein in orexin-immunoreactive enteric neurons, indicating a functional response to food status in these cells. The purpose of this article is to summarize evidence for the existence of a brain-gut network of orexin-containing cells that appears to play a role in the acute regulation of energy homeostasis.


Subject(s)
Brain/physiology , Carrier Proteins/physiology , Intestines/physiology , Intracellular Signaling Peptides and Proteins , Neuropeptides/physiology , Animals , Feeding Behavior/physiology , Humans , Orexins , Pancreas/physiology
SELECTION OF CITATIONS
SEARCH DETAIL