Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Am J Physiol Regul Integr Comp Physiol ; 320(3): R331-R341, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33470183

ABSTRACT

Gastric electrical stimulation (GES) is used clinically to promote proximal GI emptying and motility. In acute experiments, we measured duodenal motor responses elicited by GES applied at 141 randomly chosen electrode sites on the stomach serosal surface. Overnight-fasted (H2O available) anesthetized male rats (n = 81) received intermittent biphasic GES for 5 min (20-s-on/40-s-off cycles; I = 0.3 mA; pw = 0.2 ms; 10 Hz). A strain gauge on the serosal surface of the proximal duodenum of each animal was used to evaluate baseline motor activity and the effect of GES. Using ratios of time blocks compared with a 15-min prestimulation baseline, we evaluated the effects of the 5-min stimulation on concurrent activity, on the 10 min immediately after the stimulation, and on the 15-min period beginning with the onset of stimulation. We mapped the magnitude of the duodenal response (three different motility indices) elicited from the 141 stomach sites. Post hoc electrode site maps associated with duodenal responses suggested three zones similar to the classic regions of forestomach, corpus, and antrum. Maximal excitatory duodenal motor responses were elicited from forestomach sites, whereas inhibitory responses occurred with stimulation of the corpus. Moderate excitatory duodenal responses occurred with stimulation of the antrum. Complex, weak inhibitory/excitatory responses were produced by stimulation at boundaries between stomach regions. Patterns of GES efficacies coincided with distributions of previously mapped vagal afferents, suggesting that excitation of the duodenum is strongest when GES electrodes are situated over stomach concentrations of vagal intramuscular arrays, putative stretch receptors in the muscle wall.


Subject(s)
Duodenum/innervation , Electric Stimulation , Enteric Nervous System/physiology , Gastric Emptying , Gastrointestinal Motility , Stomach/innervation , Animals , Male , Muscle Spindles/physiology , Nerve Fibers, Unmyelinated/physiology , Neural Inhibition , Pressure , Rats, Sprague-Dawley , Reflex , Time Factors , Vagus Nerve/physiology
2.
Int J Food Sci Nutr ; 71(7): 839-844, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32157931

ABSTRACT

We sought to determine whether design of carbohydrate-based microspheres to have different digestion rates, while retaining the same material properties, could modulate gastric emptying through the ileal brake. Microspheres made to have three slow digestion rates and a rapidly digested starch analogue (maltodextrin) were administrated to rats by gavage and starch contents in the stomach, proximal and distal small intestine, and caecum were measured 2 h post-gavage. A stepwise increase in the amount of starch retained in the stomach was found for microspheres with incrementally slower rates of digestion. Postprandial glycaemic and insulinaemic responses were incrementally lower for the different microspheres than for the rapidly digestible control. A second-meal effect was observed for slowly digestible starch (SDS) microspheres compared to glucose. Thus, dietary slowly digestible carbohydrates were designed to elicit incremental significant changes in gastric emptying, glycaemic and insulinaemic responses, and they may be a means to trigger the ileal brake.


Subject(s)
Carbohydrates/chemistry , Carbohydrates/pharmacology , Gastric Emptying/drug effects , Animals , Blood Glucose/drug effects , Drug Design , Gastrointestinal Tract , Insulin/blood , Postprandial Period , Rats , Rats, Sprague-Dawley
3.
Ann N Y Acad Sci ; 1454(1): 14-30, 2019 10.
Article in English | MEDLINE | ID: mdl-31268562

ABSTRACT

Brain-gut neural communications have long been considered limited because of conspicuous numerical mismatches. The vagus, the parasympathetic nerve connecting brain and gut, contains thousands of axons, whereas the gastrointestinal (GI) tract contains millions of intrinsic neurons in local plexuses. The numerical paradox was initially recognized in terms of efferent projections, but the number of afferents, which comprise the majority (≈ 80%) of neurites in the vagus, is also relatively small. The present survey of recent morphological observations suggests that vagal terminals, and more generally autonomic and visceral afferent arbors in the stomach as well as throughout the gut, elaborate arbors that are extensive, regionally specialized, polymorphic, polytopic, and polymodal, commonly with multiplicities of receptors and binding sites-smart terminals. The morphological specializations and dynamic tuning of one-to-many efferent projections and many-to-one convergences of contacts onto afferents create a complex architecture capable of extensive peripheral integration in the brain-gut connectome and offset many of the disparities between axon and target numbers. Appreciating this complex architecture can help in the design of therapies for GI disorders.


Subject(s)
Brain/physiology , Connectome , Stomach/innervation , Vagus Nerve/physiology , Afferent Pathways , Animals , Efferent Pathways , Muscle, Smooth/innervation
4.
Neuroimage ; 197: 200-211, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31029867

ABSTRACT

Functional magnetic resonance imaging (fMRI) is commonly thought to be too slow to capture any neural dynamics faster than 0.1 Hz. However, recent findings demonstrate the feasibility of detecting fMRI activity at higher frequencies beyond 0.2 Hz. The origin, reliability, and generalizability of fast fMRI responses are still under debate and await confirmation through animal experiments with fMRI and invasive electrophysiology. Here, we acquired single-echo and multi-echo fMRI, as well as local field potentials, from anesthetized rat brains given gastric electrical stimulation modulated at 0.2, 0.4 and 0.8 Hz. Such gastric stimuli could drive widespread fMRI responses at corresponding frequencies from the somatosensory and cingulate cortices. Such fast fMRI responses were linearly dependent on echo times and thus indicative of blood oxygenation level dependent nature (BOLD). Local field potentials recorded during the same gastric stimuli revealed transient and phase-locked broadband neural responses, preceding the fMRI responses by as short as 0.5 s. Taken together, these results suggest that gastric stimulation can drive widespread and rapid fMRI responses of BOLD and neural origin, lending support to the feasibility of using fMRI to detect rapid changes in neural activity up to 0.8 Hz under visceral stimulation.


Subject(s)
Brain Mapping/methods , Brain/physiology , Magnetic Resonance Imaging , Stomach/physiology , Animals , Electric Stimulation , Gyrus Cinguli/physiology , Male , Neural Pathways/physiology , Rats, Sprague-Dawley , Somatosensory Cortex/physiology , Stomach/innervation
5.
Mol Nutr Food Res ; 62(5)2018 03.
Article in English | MEDLINE | ID: mdl-29230947

ABSTRACT

SCOPE: Slowly digestible starch (SDS), as a functional carbohydrate providing a slow and sustained glucose release, may be able to modulate food intake through activation of the gut-brain axis. METHODS AND RESULTS: Diet-induced obese rats were used to test the effect on feeding behavior of high-fat (HF) diets containing an SDS, fabricated to digest into the ileum, as compared to rapidly digestible starch (RDS). Ingestion of the HF-SDS diet over an 11-week period reduced daily food intake, through smaller meal size, to the same level as a lean body control group, while the group consuming the HF-RDS diet remained at a high food intake. Expression levels (mRNA) of the hypothalamic orexigenic neuropeptide Y (NPY) and Agouti-related peptide (AgRP) were significantly reduced, and the anorexigenic corticotropin-releasing hormone (CRH) was increased, in the HF-SDS fed group compared to the HF-RDS group, and to the level of the lean control group. CONCLUSION: SDS with digestion into the ileum reduced daily food intake and paralleled suppressed expression of appetite-stimulating neuropeptide genes associated with the gut-brain axis. This novel finding suggests further exploration involving a clinical study and potential development of SDS-based functional foods as an approach to obesity control.


Subject(s)
Brain/metabolism , Dietary Carbohydrates/administration & dosage , Functional Food , Gastrointestinal Tract/metabolism , Obesity/prevention & control , Starch/administration & dosage , Animals , Behavior, Animal , Diet, High-Fat , Energy Intake , Feeding Behavior , Male , Microspheres , Neuropeptide Y/genetics , Rats , Rats, Sprague-Dawley
6.
Acta Neuropathol ; 133(3): 381-393, 2017 03.
Article in English | MEDLINE | ID: mdl-28012041

ABSTRACT

Detection of α-synuclein lesions in peripheral tissues is a feature of human synucleinopathies of likely pathogenetic relevance and bearing important clinical implications. Experiments were carried out to elucidate the relationship between α-synuclein accumulation in the brain and in peripheral organs, and to identify potential pathways involved in long-distance protein transfer. Results of this in vivo study revealed a route-specific transmission of α-synuclein from the rat brain to the stomach. Following targeted midbrain overexpression of human α-synuclein, the exogenous protein was capable of reaching the gastric wall where it was accumulated into preganglionic vagal terminals. This brain-to-stomach connection likely involved intra- and inter-neuronal transfer of non-fibrillar α-synuclein that first reached the medulla oblongata, then gained access into cholinergic neurons of the dorsal motor nucleus of the vagus nerve and finally traveled via efferent fibers of these neurons contained within the vagus nerve. Data also showed a particular propensity of vagal motor neurons and efferents to accrue α-synuclein and deliver it to peripheral tissues; indeed, following its midbrain overexpression, human α-synuclein was detected within gastric nerve endings of visceromotor but not viscerosensory vagal projections. Thus, the dorsal motor nucleus of the vagus nerve represents a key relay center for central-to-peripheral α-synuclein transmission, and efferent vagal fibers may act as unique conduits for protein transfer. The presence of α-synuclein in peripheral tissues could reflect, at least in some synucleinopathy patients, an ongoing pathological process that originates within the brain and, from there, reaches distant organs innervated by motor vagal projections.


Subject(s)
Autonomic Fibers, Preganglionic/metabolism , Brain/metabolism , Gastric Mucosa/metabolism , Vagus Nerve/metabolism , alpha-Synuclein/metabolism , Animals , Brain/cytology , Choline O-Acetyltransferase/metabolism , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Neurons/metabolism , Nodose Ganglion/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Transduction, Genetic , Vagus Nerve/physiology , alpha-Synuclein/genetics
7.
J Comp Neurol ; 524(13): 2577-603, 2016 09 01.
Article in English | MEDLINE | ID: mdl-26850701

ABSTRACT

A full description of the terminal architecture of sympathetic axons innervating the gastrointestinal (GI) tract has not been available. To label sympathetic fibers projecting to the gut muscle wall, dextran biotin was injected into the celiac and superior mesenteric ganglia (CSMG) of rats. Nine days postinjection, animals were euthanized and stomachs and small intestines were processed as whole mounts (submucosa and mucosa removed) to examine CSMG efferent terminals. Myenteric neurons were counterstained with Cuprolinic Blue; catecholaminergic axons were stained immunohistochemically for tyrosine hydroxylase. Essentially all dextran-labeled axons (135 of 136 sampled) were tyrosine hydroxylase-positive. Complete postganglionic arbors (n = 154) in the muscle wall were digitized and analyzed morphometrically. Individual sympathetic axons formed complex arbors of varicose neurites within myenteric ganglia/primary plexus and, concomitantly, long rectilinear arrays of neurites within circular muscle/secondary plexus or longitudinal muscle/tertiary plexus. Very few CSMG neurons projected exclusively (i.e., ∼100% of an arbor's varicose branches) to myenteric plexus (∼2%) or smooth muscle (∼14%). With less stringent inclusion criteria (i.e., ≥85% of an axon's varicose branches), larger minorities of neurons projected predominantly to either myenteric plexus (∼13%) or smooth muscle (∼27%). The majority (i.e., ∼60%) of all individual CSMG postganglionics formed mixed, heterotypic arbors that coinnervated extensively (>15% of their varicose branches per target) both myenteric ganglia and smooth muscle. The fact that ∼87% of all sympathetics projected either extensively or even predominantly to smooth muscle, while simultaneously contacting myenteric plexus, is consistent with the view that these neurons control GI muscle directly, if not exclusively. J. Comp. Neurol. 524:2577-2603, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Ganglia, Sympathetic/physiology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/physiology , Muscle, Smooth/innervation , Muscle, Smooth/physiology , Myenteric Plexus/physiology , Sympathetic Fibers, Postganglionic/physiology , Animals , Ganglia/chemistry , Ganglia/physiology , Ganglia, Sympathetic/chemistry , Gastrointestinal Tract/chemistry , Male , Muscle, Smooth/chemistry , Myenteric Plexus/chemistry , Neurons/chemistry , Neurons/physiology , Rats , Rats, Inbred F344 , Sympathetic Fibers, Postganglionic/chemistry
8.
J Comp Neurol ; 524(4): 713-37, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26355387

ABSTRACT

The fundamental roles that the stomach plays in ingestion and digestion notwithstanding, little morphological information is available on vagal intramuscular arrays (IMAs), the afferents that innervate gastric smooth muscle. To characterize IMAs better, rats were given injections of dextran biotin in the nodose ganglia, and, after tracer transport, stomach whole mounts were collected. Specimens were processed for avidin-biotin permanent labeling, and subsets of the whole mounts were immunohistochemically processed for c-Kit or stained with cuprolinic blue. IMAs (n = 184) were digitized for morphometry and mapping. Throughout the gastric muscle wall, IMAs possessed common phenotypic features. Each IMA was generated by a parent neurite arborizing extensively, forming an array of multiple (mean = 212) branches averaging 193 µm in length. These branches paralleled, and coursed in apposition with, bundles of muscle fibers and interstitial cells of Cajal. Individual arrays averaged 4.3 mm in length and innervated volumes of muscle sheet, presumptive receptive fields, averaging 0.1 mm(3) . Evaluated by region and by muscle sheet, IMAs displayed architectural adaptations to the different loci. A subset (32%) of circular muscle IMAs issued specialized polymorphic collaterals to myenteric ganglia, and a subset (41%) of antral longitudinal muscle IMAs formed specialized net endings associated with the serosal boundary. IMAs were concentrated in regional patterns that correlated with the unique biomechanical adaptations of the stomach, specifically proximal stomach reservoir functions and antral emptying operations. Overall, the structural adaptations and distributions of the IMAs were consonant with the hypothesized stretch receptor roles of the afferents.


Subject(s)
Mechanoreceptors/cytology , Muscle, Smooth/innervation , Stomach/innervation , Vagus Nerve/anatomy & histology , Animals , Immunohistochemistry , Male , Neuroanatomical Tract-Tracing Techniques , Rats, Sprague-Dawley
9.
BMC Pregnancy Childbirth ; 14: 241, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25048443

ABSTRACT

BACKGROUND: Elucidation of the biochemical pathways involved in activation of preterm and term human labour would facilitate the development of effective management and inform judgements regarding the necessity for preterm tocolysis and post-term induction. Prostaglandins act at all stages of human reproduction, and are potentially activators of labour. METHODS: Expression of 15 genes involved in prostaglandin synthesis, transport and degradation was measured by qPCR using tissue samples from human placenta, amnion and choriodecidua at preterm and full-term vaginal and caesarean delivery. Cellular localisation of eight prostaglandin pathway proteins was determined by immunohistochemistry. RESULTS: Expression of prostaglandin pathway genes was differentially affected by factors including gestational age at delivery, and the incidence and duration of labour. Chorioamnionitis/deciduitis was associated with upregulation of PTGS2 (prostaglandin-endoperoxide synthase 2 (prostaglandin G/H synthase and cyclooxygenase)), along with the inflammatory genes IL8 (interleukin 8), S100A8 (S100 calcium binding protein A8) and TLR2 (toll-like receptor 2), in amnion and choriodecidua, and with downregulation of CBR1 (carbonyl reductase 1) and HPGD (hydroxyprostaglandin dehydrogenase 15-(NAD)) in choriodecidua. Protein localisation differed greatly between the various maternal and fetal cell types. CONCLUSIONS: Preterm and term labour are associated with distinct prostaglandin pathway expression profiles; inflammation provokes specific changes, unrelated to the presence of labour; spontaneous and induced term labour are indistinguishable.


Subject(s)
Gene Expression , Labor, Obstetric/genetics , Obstetric Labor, Premature/genetics , Prostaglandins/analysis , Prostaglandins/genetics , Signal Transduction/genetics , 3-Hydroxysteroid Dehydrogenases/analysis , 3-Hydroxysteroid Dehydrogenases/genetics , Adult , Alcohol Oxidoreductases/analysis , Alcohol Oxidoreductases/genetics , Aldehyde Reductase/analysis , Aldehyde Reductase/genetics , Aldo-Keto Reductase Family 1 Member C3 , Amnion/chemistry , Calgranulin A/analysis , Calgranulin A/genetics , Chorioamnionitis/genetics , Chorion/chemistry , Cyclooxygenase 2/analysis , Cyclooxygenase 2/genetics , Cytochrome P-450 Enzyme System/analysis , Cytochrome P-450 Enzyme System/genetics , Decidua/chemistry , Down-Regulation , Female , Gestational Age , Humans , Hydroxyprostaglandin Dehydrogenases/analysis , Hydroxyprostaglandin Dehydrogenases/genetics , Interleukin-1/analysis , Interleukin-1/genetics , Intramolecular Oxidoreductases/analysis , Intramolecular Oxidoreductases/genetics , Labor, Obstetric/metabolism , Multidrug Resistance-Associated Proteins/analysis , Multidrug Resistance-Associated Proteins/genetics , Obstetric Labor, Premature/metabolism , Organic Anion Transporters/analysis , Organic Anion Transporters/genetics , Placenta/chemistry , Pregnancy , Prostaglandin-E Synthases , Prostaglandins/metabolism , Toll-Like Receptor 2/analysis , Toll-Like Receptor 2/genetics , Up-Regulation , Young Adult
10.
Auton Neurosci ; 183: 36-48, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24656895

ABSTRACT

The pylorus is innervated by vagal mechanoreceptors that project to gastrointestinal smooth muscle, but the distributions and specializations of vagal endings in the sphincter have not been fully characterized. To evaluate their organization, the neural tracer dextran biotin was injected into the nodose ganglia of rats. Following tracer transport, animals were perfused, and their pylori and antra were prepared as whole mounts. Specimens were processed to permanently label the tracer, and subsets were counterstained with Cuprolinic blue or immunostained for c-Kit. Intramuscular arrays (IMAs) in the circular muscle comprised the principal vagal afferent innervation of the sphincter. These pyloric ring IMAs were densely distributed and evidenced a variety of structural specializations. Morphometric comparisons between the arbors innervating the pylorus and a corresponding sample of IMAs in the adjacent antral circular muscle highlighted that sphincter IMAs branched profusely, forming more than twice as many branches as did antral IMAs (means of 405 vs. 165, respectively), and condensed their numerous neurites into compact receptive fields (∼48% of the area of antral IMAs) deep in the circular muscle (∼6µm above the submucosa). Separate arbors of IMAs in the sphincter interdigitated and overlapped to form a 360° band of mechanoreceptors encircling the pyloric canal. The annulus of vagal IMA arbors, putative stretch receptors tightly intercalated in the sphincter ring and situated near the lumen of the pyloric canal, creates an architecture with the potential to generate gut reflexes on the basis of pyloric sensory maps of high sensitivity and fine spatial resolution.


Subject(s)
Mechanoreceptors/cytology , Nodose Ganglion/cytology , Pylorus/innervation , Vagus Nerve/cytology , Animals , Immunohistochemistry , Male , Muscle, Smooth/innervation , Neurites , Neuroanatomical Tract-Tracing Techniques , Rats, Sprague-Dawley
11.
Auton Neurosci ; 179(1-2): 108-21, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24104187

ABSTRACT

It is well documented that the intrinsic enteric nervous system of the gastrointestinal (GI) tract sustains neuronal losses and reorganizes as it ages. In contrast, age-related remodeling of the extrinsic sympathetic projections to the wall of the gut is poorly characterized. The present experiment, therefore, surveyed the sympathetic projections to the aged small intestine for axonopathies. Furthermore, the experiment evaluated the specific prediction that catecholaminergic inputs undergo hyperplastic changes. Jejunal tissue was collected from 3-, 8-, 16-, and 24-month-old male Fischer 344 rats, prepared as whole mounts consisting of the muscularis, and processed immunohistochemically for tyrosine hydroxylase, the enzymatic marker for norepinephrine, and either the protein CD163 or the protein MHCII, both phenotypical markers for macrophages. Four distinctive sympathetic axonopathy profiles occurred in the small intestine of the aged rat: (1) swollen and dystrophic terminals, (2) tangled axons, (3) discrete hyperinnervated loci in the smooth muscle wall, including at the bases of Peyer's patches, and (4) ectopic hyperplastic or hyperinnervating axons in the serosa/subserosal layers. In many cases, the axonopathies occurred at localized and limited foci, involving only a few axon terminals, in a pattern consistent with incidences of focal ischemic, vascular, or traumatic insult. The present observations underscore the complexity of the processes of aging on the neural circuitry of the gut, with age-related GI functional impairments likely reflecting a constellation of adjustments that range from selective neuronal losses, through accumulation of cellular debris, to hyperplasias and hyperinnervation of sympathetic inputs.


Subject(s)
Aging/pathology , Autonomic Nervous System Diseases/pathology , Enteric Nervous System/pathology , Muscle, Smooth/innervation , Animals , Immunohistochemistry , Male , Muscle, Smooth/pathology , Rats , Rats, Inbred F344
12.
Auton Neurosci ; 179(1-2): 90-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24044976

ABSTRACT

Little is known about the architecture of the vagal motor units that control esophageal striated muscle, in spite of the fact that these units are necessary, and responsible, for peristalsis. The present experiment was designed to characterize the motor neuron projection fields and terminal arbors forming esophageal motor units. Nucleus ambiguus compact formation neurons of the rat were labeled by bilateral intracranial injections of the anterograde tracer dextran biotin. After tracer transport, thoracic and abdominal esophagi were removed and prepared as whole mounts of muscle wall without mucosa or submucosa. Labeled terminal arbors of individual vagal motor neurons (n=78) in the esophageal wall were inventoried, digitized and analyzed morphometrically. The size of individual vagal motor units innervating striated muscle, throughout thoracic and abdominal esophagus, averaged 52 endplates per motor neuron, a value indicative of fine motor control. A majority (77%) of the motor terminal arbors also issued one or more collateral branches that contacted neurons, including nitric oxide synthase-positive neurons, of local myenteric ganglia. Individual motor neuron terminal arbors co-innervated, or supplied endplates in tandem to, both longitudinal and circular muscle fibers in roughly similar proportions (i.e., two endplates to longitudinal for every three endplates to circular fibers). Both the observation that vagal motor unit collaterals project to myenteric ganglia and the fact that individual motor units co-innervate longitudinal and circular muscle layers are consistent with the hypothesis that elements contributing to peristaltic programming inhere, or are "hardwired," in the peripheral architecture of esophageal motor units.


Subject(s)
Esophagus/innervation , Motor Neurons/cytology , Muscle, Skeletal/innervation , Peristalsis/physiology , Vagus Nerve/cytology , Animals , Esophagus/physiology , Male , Myenteric Plexus/cytology , Rats , Rats, Sprague-Dawley
13.
PLoS One ; 8(4): e59745, 2013.
Article in English | MEDLINE | ID: mdl-23565164

ABSTRACT

For digestion of starch in humans, α-amylase first hydrolyzes starch molecules to produce α-limit dextrins, followed by complete hydrolysis to glucose by the mucosal α-glucosidases in the small intestine. It is known that α-1,6 linkages in starch are hydrolyzed at a lower rate than are α-1,4 linkages. Here, to create designed slowly digestible carbohydrates, the structure of waxy corn starch (WCS) was modified using a known branching enzyme alone (BE) and an in combination with ß-amylase (BA) to increase further the α-1,6 branching ratio. The digestibility of the enzymatically synthesized products was investigated using α-amylase and four recombinant mammalian mucosal α-glucosidases. Enzyme-modified products (BE-WCS and BEBA-WCS) had increased percentage of α-1,6 linkages (WCS: 5.3%, BE-WCS: 7.1%, and BEBA-WCS: 12.9%), decreased weight-average molecular weight (WCS: 1.73×10(8) Da, BE-WCS: 2.76×10(5) Da, and BEBA-WCS 1.62×10(5) Da), and changes in linear chain distributions (WCS: 21.6, BE-WCS: 16.9, BEBA-WCS: 12.2 DPw). Hydrolysis by human pancreatic α-amylase resulted in an increase in the amount of branched α-limit dextrin from 26.8% (WCS) to 56.8% (BEBA-WCS). The α-amylolyzed samples were hydrolyzed by the individual α-glucosidases (100 U) and glucogenesis decreased with all as the branching ratio increased. This is the first report showing that hydrolysis rate of the mammalian mucosal α-glucosidases is limited by the amount of branched α-limit dextrin. When enzyme-treated materials were gavaged to rats, the level of postprandial blood glucose at 60 min from BEBA-WCS was significantly higher than for WCS or BE-WCS. Thus, highly branched glucan structures modified by BE and BA had a comparably slow digesting property both in vitro and in vivo. Such highly branched α-glucans show promise as a food ingredient to control postprandial glucose levels and to attain extended glucose release.


Subject(s)
Glucose/biosynthesis , Glucose/chemistry , Polysaccharides/chemistry , Polysaccharides/metabolism , alpha-Glucosidases/metabolism , Animals , Blood Glucose , Humans , Hydrolysis , Male , Molecular Weight , Mucous Membrane/enzymology , Nuclear Magnetic Resonance, Biomolecular , Rats , Recombinant Proteins/metabolism , Starch/chemistry , Starch/metabolism
14.
Auton Neurosci ; 177(2): 129-42, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23583280

ABSTRACT

To supply a fuller morphological characterization of the vagal afferents innervating the lower esophageal sphincter (LES), specifically to label vagal terminals in the tissues forming the LES in the gastroesophageal junction, the present experiment employed injections of dextran biotin into the nodose ganglia of rats. Four types of vagal afferents innervated the LES. Clasp and sling muscle fibers were directly and prominently innervated by intramuscular arrays (IMAs). Individual IMA terminals subtended about 16° of arc of the esophageal circumference, and, collectively, the terminal fields were distributed within the muscle ring to establish a 360° annulus of mechanoreceptors in the sphincter wall. 3D morphometry of the terminals established that, compared to sling muscle IMAs, clasp muscle IMAs had more extensive arbors and larger receptive fields. In addition, at the cardia, local myenteric ganglia between smooth muscle sheets and striated muscle bundles were innervated by intraganglionic laminar endings (IGLEs), in a pattern similar to the innervation of the myenteric plexus throughout the stomach and esophagus. Finally, as previously described, the principle bundle of sling muscle fibers that links LES sphincter tissue to the antropyloric region of the lesser curvature was innervated by exceptionally long IMAs as well as by unique web ending specializations at the distal attachment of the bundle. Overall, the specialized varieties of densely distributed vagal afferents innervating the LES underscore the conclusion that these sensory projections are critically involved in generating LES reflexes and may be promising targets for managing esophageal dysfunctions.


Subject(s)
Esophageal Sphincter, Lower/innervation , Esophageal Sphincter, Lower/physiology , Vagus Nerve/physiology , Afferent Pathways/physiology , Animals , Male , Rats , Rats, Sprague-Dawley
15.
Anat Rec (Hoboken) ; 296(4): 654-69, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23441091

ABSTRACT

With age, alpha-synuclein (α-SYNC) misfolds and forms insoluble deposits of protein in the myenteric plexus, leading presumably to dystrophy and degeneration in the circuitry controlling gastrointestinal (GI) function. The present experiment examined aggregates of α-SYNC in the aging small intestine and investigated how macrophages in the wall of the GI tract respond to these aberrant deposits. Groups of adult and aged Fisher 344 rats were studied. Whole mounts of duodenal, jejunal, and ileal smooth muscle wall, including the myenteric plexus, were prepared. Double labeling immunohistochemistry was used to stain α-SYNC protein and the phenotypic macrophage antigens CD163 and MHCII. Alpha-synuclein accumulated in dense aggregates in axons of both postganglionic and preganglionic neurons throughout the small intestine. Staining patterns suggested that deposits of protein occur initially in axonal terminals and then spread retrogradely toward the somata. Macrophages that were adjacent to dystrophic terminal processes were swollen and contained vacuoles filled with insoluble α-SYNC, and these macrophages commonly had the phenotype of alternatively activated phagocytes. The present results suggest that macrophages play an active phagocytotic role in removing α-SYNC aggregates that accumulate with age in the neural circuitry of the gut. Our observations further indicate that this housekeeping response does not clear the protein sufficiently to eliminate all synucleinopathies or their precursor aggregates from the healthy aging GI tract. Thus, accumulating deposits of insoluble α-SYNC in the wall of the GI tract may contribute, especially when compounded by disease or inflammation, to the age-associated neuropathies in the gut that compromise GI function.


Subject(s)
Intestine, Small/innervation , Macrophages/metabolism , Muscle, Smooth/innervation , Myenteric Plexus/metabolism , Phagocytosis , alpha-Synuclein/metabolism , Age Factors , Aging , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Autonomic Fibers, Postganglionic/metabolism , Autonomic Fibers, Preganglionic/metabolism , Biomarkers/metabolism , Histocompatibility Antigens Class II/metabolism , Immunohistochemistry , Macrophage Activation , Male , Phenotype , Protein Folding , Rats , Rats, Inbred F344 , Receptors, Cell Surface/metabolism
16.
Auton Neurosci ; 169(1): 12-27, 2012 Jul 02.
Article in English | MEDLINE | ID: mdl-22436622

ABSTRACT

Interactions between macrophages and the autonomic innervation of gastrointestinal (GI) tract smooth muscle have received little experimental attention. To better understand this relationship, immunohistochemistry was performed on GI whole mounts from rats at three ages. The phenotypes, morphologies, and distributions of gut macrophages are consistent with the cells performing extensive housekeeping functions in the smooth muscle layers. Specifically, a dense population of macrophages was located throughout the muscle wall where they were distributed among the muscle fibers and along the vasculature. Macrophages were also associated with ganglia and connectives of the myenteric plexus and with the sympathetic innervation. Additionally, these cells were in tight registration with the dendrites and axons of the myenteric neurons as well as the varicosities along the length of the sympathetic axons, suggestive of a contribution by the macrophages to the homeostasis of both synapses and contacts between the various elements of the enteric circuitry. Similarly, macrophages were involved in the presumed elimination of neuropathies as indicated by their association with dystrophic neurons and neurites which are located throughout the myenteric plexus and smooth muscle wall of aged rats. Importantly, the patterns of macrophage-neuron interactions in the gut paralleled the much more extensively characterized interactions of macrophages (i.e., microglia) and neurons in the CNS. The present observations in the PNS as well as extrapolations from homologous microglia in the CNS suggest that GI macrophages play significant roles in maintaining the nervous system of the gut in the face of wear and tear, disease, and aging.


Subject(s)
Aging , Autonomic Nervous System/physiology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/innervation , Macrophages/immunology , Muscle, Smooth/immunology , Muscle, Smooth/innervation , Animals , Antigens, Differentiation/metabolism , Autonomic Nervous System/cytology , Autonomic Nervous System/metabolism , Cell Aggregation , Central Nervous System/cytology , Central Nervous System/metabolism , Central Nervous System/physiology , Gastrointestinal Tract/cytology , Gastrointestinal Tract/metabolism , Homeostasis , Macrophages/cytology , Macrophages/metabolism , Male , Microglia/cytology , Microglia/immunology , Microglia/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Myenteric Plexus/cytology , Myenteric Plexus/immunology , Myenteric Plexus/metabolism , Neuroanatomical Tract-Tracing Techniques , Neurons/cytology , Neurons/immunology , Neurons/metabolism , Peripheral Nervous System/cytology , Peripheral Nervous System/metabolism , Peripheral Nervous System/physiology , Rats , Rats, Inbred BN , Rats, Inbred F344 , alpha-Synuclein/metabolism
17.
Auton Neurosci ; 153(1-2): 90-8, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-19665435

ABSTRACT

Recent progress in understanding visceral afferents, some of it reviewed in the present issue, serves to underscore how little is known about the aging of the visceral afferents in the gastrointestinal (GI) tract. In spite of the clinical importance of the issue-with age, GI function often becomes severely compromised-only a few initial observations on age-related structural changes of visceral afferents are available. Primary afferent cell bodies in both the nodose ganglia and dorsal root ganglia lose Nissl material and accumulate lipofucsin, inclusions, aggregates, and tangles. Additionally, in changes that we focus on in the present review, vagal visceral afferent terminals in both the muscle wall and the mucosa of the GI tract exhibit age-related structural changes. In aged animals, both of the vagal terminal types examined, namely intraganglionic laminar endings and villus afferents, exhibit dystrophic or regressive morphological changes. These neuropathies are associated with age-related changes in the structural integrity of the target organs of the affected afferents, suggesting that local changes in trophic environment may give rise to the aging of GI innervation. Given the clinical relevance of GI tract aging, a more complete understanding both of how aging alters the innervation of the gut and of how such changes might be mitigated should be made research priorities.


Subject(s)
Aging/physiology , Gastrointestinal Tract/innervation , Vagus Nerve/physiology , Visceral Afferents/physiology , Animals , Humans , Nerve Endings/physiology , Sensory Receptor Cells/metabolism , Vagus Nerve/anatomy & histology , Visceral Afferents/anatomy & histology
18.
Exp Neurol ; 220(1): 109-19, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19664623

ABSTRACT

Dystrophic axons and terminals are common in the myenteric plexus and smooth muscle of the gastrointestinal (GI) tract of aged rats. In young adult rats, alpha-synuclein in its normal state is abundant throughout the myenteric plexus, making this protein-which is prone to fibrillization-a candidate marker for axonopathies in the aged rat. To determine if aggregation of alpha-synuclein is involved in the formation of age-related enteric neuropathies, we sampled the stomach, small intestine and large intestine of adult, middle-aged, and aged virgin male Fischer 344 rats stained for alpha-synuclein in both its normal and pathological states. Alpha-synuclein-positive dystrophic axons and terminals were present throughout the GI tract of middle-aged and aged rats, with immunohistochemical double labeling demonstrating co-localization within nitric oxide synthase-, calretinin-, calbindin-, or tyrosine hydroxylase-positive markedly swollen neurites. However, other dystrophic neurites positive for each of these four markers were not co-reactive for alpha-synuclein. Similarly, a subpopulation of alpha-synuclein inclusions contained deposits immunostained with an anti-tau phospho-specific Ser(262) antibody, but not all of these hyperphosphorylated tau-positive aggregates were co-localized with alpha-synuclein. The presence of heteroplastic and potentially degenerating neural elements and protein aggregates both positive and negative for alpha-synuclein suggests a complex chronological relationship between the onset of degenerative changes and the accumulation of misfolded proteins. Additionally, proteins other than alpha-synuclein appear to be involved in age-related axonopathies. Finally, this study establishes the utility of the aging Fischer 344 rat for the study of synucleopathies and tauopathies in the GI tract.


Subject(s)
Aging/metabolism , Aging/pathology , Autonomic Nervous System Diseases/pathology , Myenteric Plexus/metabolism , Myenteric Plexus/pathology , alpha-Synuclein/metabolism , Animals , Autonomic Nervous System Diseases/physiopathology , Axons/metabolism , Axons/pathology , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Gastrointestinal Tract/innervation , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/physiopathology , Immunohistochemistry , Lewy Bodies/metabolism , Lewy Bodies/pathology , Male , Nitric Oxide Synthase Type I/metabolism , Protein Folding , Rats , Rats, Inbred F344 , Tyrosine 3-Monooxygenase/metabolism , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , Wallerian Degeneration/physiopathology , alpha-Synuclein/analysis , tau Proteins/analysis , tau Proteins/metabolism
19.
J Neurosci Methods ; 178(1): 1-9, 2009 Mar 30.
Article in English | MEDLINE | ID: mdl-19056424

ABSTRACT

None of the anterograde tracers used to label and investigate vagal preganglionic neurons projecting to the viscera has proved optimal for routine and extensive labeling of autonomic terminal fields. To identify an alternative tracer protocol, the present experiment evaluated whether dextran conjugates, which have produced superior results in the CNS, might yield widespread and effective labeling of long, fine-caliber vagal efferents in the peripheral nervous system. The dextran conjugates that were evaluated proved reliable and versatile for labeling the motor neuron pool in its entirety, for single- and multiple-labeling protocols, for both conventional and confocal fluorescence microscopy, and for permanent labeling protocols for brightfield microscopy of the projections to the gastrointestinal (GI) tract. Using a standard ABC kit followed by visualization with DAB as the chromagen, Golgi-like labeling of the vagal efferent terminal fields in the GI wall was achieved with the biotinylated dextrans. The definition of individual terminal varicosities was so sharp and detailed that it was routinely practical to examine the relationship of putative vagal efferent contacts (by the criteria of high magnification light microscopy) with the dendritic and somatic architecture of counterstained neurons in the myenteric plexus. Overall, dextran conjugates provide high-definition labeling of an extensive vagal motor pool in the GI tract, and offer considerable versatility when multiple-staining protocols are needed to elucidate the complexities of the innervation of the gut.


Subject(s)
Dextrans/metabolism , Gastrointestinal Tract/metabolism , Neurons, Afferent/metabolism , Vagus Nerve/metabolism , Animals , Fluorescent Dyes/metabolism , Male , Medulla Oblongata/cytology , Medulla Oblongata/metabolism , Neural Pathways/physiology , Nitric Oxide Synthase/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Silver Staining/methods , alpha-Synuclein/metabolism
20.
Auton Neurosci ; 136(1-2): 1-19, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17537681

ABSTRACT

The gastrointestinal (GI) tract is innervated by intrinsic enteric neurons and by extrinsic projections, including sympathetic and parasympathetic efferents as well as visceral afferents, all of which are compromised by age to different degrees. In the present review, we summarize and illustrate key structural changes in the aging innervation of the gut, and suggest a provisional list of the general patterns of aging of the GI innervation. For example, age-related neuronal losses occur in both the myenteric plexus and submucosal plexus of the intestines. These losses start in adulthood, increase over the rest of the life span, and are specific to cholinergic neurons. Parallel losses of enteric glia also occur. The extent of neuronal and glial loss varies along an oral-to-anal gradient, with the more distal GI tract being more severely affected. Additionally, with aging, dystrophic axonal swellings and markedly dilated varicosities progressively accumulate in the sympathetic, vagal, dorsal root, and enteric nitrergic innervation of the gut. These dramatic and consistent patterns of neuropathy that characterize the aging autonomic nervous system of the GI tract are candidate mechanisms for some of the age-related declines in function evidenced in the elderly.


Subject(s)
Aging/pathology , Autonomic Nervous System Diseases/pathology , Enteric Nervous System/pathology , Gastrointestinal Tract/innervation , Animals , Autonomic Nervous System Diseases/physiopathology , Enteric Nervous System/physiopathology , Gastrointestinal Tract/physiopathology , Humans , Models, Animal , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neuroglia/pathology , Neurons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...