Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 317(2): G210-G221, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31268770

ABSTRACT

The enteric nervous system in the large intestine generates two important patterns relating to motility: 1) propagating rhythmic peristaltic smooth muscle contractions referred to as colonic migrating motor complexes (CMMCs) and 2) tonic inhibition, during which colonic smooth muscle contractions are suppressed. The precise neurobiological substrates underlying each of these patterns are unclear. Using transgenic animals expressing the genetically encoded calcium indicator GCaMP3 to monitor activity or the optogenetic actuator channelrhodopsin (ChR2) to drive activity in defined enteric neuronal subpopulations, we provide evidence that cholinergic and nitrergic neurons play significant roles in mediating CMMCs and tonic inhibition, respectively. Nitrergic neurons [neuronal nitric oxide synthase (nNOS)-positive neurons] expressing GCaMP3 exhibited higher levels of activity during periods of tonic inhibition than during CMMCs. Consistent with these findings, optogenetic activation of ChR2 in nitrergic neurons depressed ongoing CMMCs. Conversely, cholinergic neurons [choline acetyltransferase (ChAT)-positive neurons] expressing GCaMP3 markedly increased their activity during the CMMC. Treatment with the NO synthesis inhibitor Nω-nitro-l-arginine also augmented the activity of ChAT-GCaMP3 neurons, suggesting that the reciprocal patterns of activity exhibited by nitrergic and cholinergic enteric neurons during distinct phases of colonic motility may be related.NEW & NOTEWORTHY Correlating the activity of neuronal populations in the myenteric plexus to distinct periods of gastrointestinal motility is complicated by the difficulty of measuring the activity of specific neuronal subtypes. Here, using mice expressing genetically encoded calcium indicators or the optical actuator channelrhodopsin-2, we provide compelling evidence that cholinergic and nitrergic neurons play important roles in mediating coordinated propagating peristaltic contractions or tonic inhibition, respectively, in the murine colon.


Subject(s)
Cholinergic Neurons , Colon , Nitrergic Neurons , Nitroarginine/pharmacology , Peristalsis , Animals , Animals, Genetically Modified , Cholinergic Neurons/drug effects , Cholinergic Neurons/physiology , Colon/innervation , Colon/physiology , Enteric Nervous System/drug effects , Enteric Nervous System/physiology , Enzyme Inhibitors/pharmacology , Mice , Muscle Contraction/drug effects , Muscle Contraction/physiology , Myoelectric Complex, Migrating/drug effects , Myoelectric Complex, Migrating/physiology , Nitrergic Neurons/drug effects , Nitrergic Neurons/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Optogenetics , Peristalsis/drug effects , Peristalsis/physiology
2.
PLoS One ; 12(2): e0171262, 2017.
Article in English | MEDLINE | ID: mdl-28152551

ABSTRACT

Serum response factor (SRF) transcriptionally regulates expression of contractile genes in smooth muscle cells (SMC). Lack or decrease of SRF is directly linked to a phenotypic change of SMC, leading to hypomotility of smooth muscle in the gastrointestinal (GI) tract. However, the molecular mechanism behind SRF-induced hypomotility in GI smooth muscle is largely unknown. We describe here how SRF plays a functional role in the regulation of the SMC contractility via myotonic dystrophy protein kinase (DMPK) and L-type calcium channel CACNA1C. GI SMC expressed Dmpk and Cacna1c genes into multiple alternative transcriptional isoforms. Deficiency of SRF in SMC of Srf knockout (KO) mice led to reduction of SRF-dependent DMPK, which down-regulated the expression of CACNA1C. Reduction of CACNA1C in KO SMC not only decreased intracellular Ca2+ spikes but also disrupted their coupling between cells resulting in decreased contractility. The role of SRF in the regulation of SMC phenotype and function provides new insight into how SMC lose their contractility leading to hypomotility in pathophysiological conditions within the GI tract.


Subject(s)
Calcium Channels, L-Type/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Myotonin-Protein Kinase/physiology , Serum Response Factor/physiology , Animals , Blotting, Western , Female , Male , Mice , Mice, Knockout , Microscopy, Confocal , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/ultrastructure , Polymerase Chain Reaction , Proteomics , Tamoxifen/pharmacology
3.
Front Cell Neurosci ; 9: 436, 2015.
Article in English | MEDLINE | ID: mdl-26617487

ABSTRACT

Genetically encoded Ca(2+) indicators (GECIs) have been used extensively in many body systems to detect Ca(2+) transients associated with neuronal activity. Their adoption in enteric neurobiology has been slower, although they offer many advantages in terms of selectivity, signal-to-noise and non-invasiveness. Our aims were to utilize a number of cell-specific promoters to express the Ca(2+) indicator GCaMP3 in different classes of neurons and glia to determine their effectiveness in measuring activity in enteric neural networks during colonic motor behaviors. We bred several GCaMP3 mice: (1) Wnt1-GCaMP3, all enteric neurons and glia; (2) GFAP-GCaMP3, enteric glia; (3) nNOS-GaMP3, enteric nitrergic neurons; and (4) ChAT-GCaMP3, enteric cholinergic neurons. These mice allowed us to study the behavior of the enteric neurons in the intact colon maintained at a physiological temperature, especially during the colonic migrating motor complex (CMMC), using low power Ca(2+) imaging. In this preliminary study, we observed neuronal and glial cell Ca(2+) transients in specific cells in both the myenteric and submucous plexus in all of the transgenic mice variants. The number of cells that could be simultaneously imaged at low power (100-1000 active cells) through the undissected gut required advanced motion tracking and analysis routines. The pattern of Ca(2+) transients in myenteric neurons showed significant differences in response to spontaneous, oral or anal stimulation. Brief anal elongation or mucosal stimulation, which evokes a CMMC, were the most effective stimuli and elicited a powerful synchronized and prolonged burst of Ca(2+) transients in many myenteric neurons, especially when compared with the same neurons during a spontaneous CMMC. In contrast, oral elongation, which normally inhibits CMMCs, appeared to suppress Ca(2+) transients in some of the neurons active during a spontaneous or an anally evoked CMMC. The activity in glial networks appeared to follow neural activity but continued long after neural activity had waned. With these new tools an unprecedented level of detail can be recorded from the enteric nervous system (ENS) with minimal manipulation of tissue. These techniques can be extended in order to better understand the roles of particular enteric neurons and glia during normal and disordered motility.

4.
J Physiol ; 593(15): 3225-7, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26228547
5.
J Physiol ; 593(15): 3233, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26228549
6.
J Neurogastroenterol Motil ; 20(4): 423-46, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25273115

ABSTRACT

The colonic migrating motor complex (CMMC) is a critical neurally mediated rhythmic propulsive contraction observed in the large intestine of many mammals. It seems to be equivalent to the high amplitude propagating contractions (HAPCs) in humans. This review focuses on the probable neural mechanisms involved in producing the CMMC or HAPC, their likely de-pendence on mucosal and neuronal serotonin and pacemaker insterstitial cells of Cajal networks and how intrinsic neural re-flexes affect them. Discussed is the possibility that myenteric 5-hydroxytryptamine (5-HT) neurons are not only involved in tonic inhibition of the colon, but are also involved in generating the CMMC and modulation of the entire enteric nervous system, including coupling motility to secretion and blood flow. Mucosal 5-HT appears to be important for the initiation and effective propagation of CMMCs, although this mechanism is a longstanding controversy since the 1950s, which we will address. We argue that the slow apparent propagation of the CMMC/HAPC down the colon is unlikely to result from a slowly conducting wave front of neural activity, but more likely because of an interaction between ascending excitatory and descending (serotonergic) inhibitory neural pathways interacting both within the myenteric plexus and at the level of the muscle. That is, CMMC/HAPC propagation appears to be similar to esophageal peristalsis. The suppression of inhibitory (neuronal nitric oxide synthase) motor neurons and mucosal 5-HT release by an upregulation of prostaglandins has important implications in a num-ber of gastrointestinal disorders, especially slow transit constipation.(J Neurogastroenterol Motil 2014;20:423-446).

7.
J Physiol ; 591(23): 5939-57, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24127620

ABSTRACT

Although there is general agreement that mucosal 5-hydroxytryptamine (5-HT) can initiate peristaltic reflexes in the colon, recent studies have differed as to whether or not the role of mucosal 5-HT is critical. We therefore tested the hypothesis that the secretion of 5-HT from mucosal enterochromaffin (EC) cells is essential for the manifestation of murine colonic peristaltic reflexes. To do so, we analysed the mechanisms underlying faecal pellet propulsion in isolated colons of mice lacking tryptophan hydroxylase 1 (Tph1(-/-) mice), which is the rate-limiting enzyme in the biosynthesis of mucosal but not neuronal 5-HT. We used video analysis of faecal pellet propulsion, tension transducers to record colonic migrating motor complexes (CMMCs) and intracellular microelectrodes to record circular muscle activity occurring spontaneously or following intraluminal distension. When compared with control (Tph1(+/+)) mice, Tph1(-/-) animals exhibited: (1) an elongated colon; (2) larger faecal pellets; (3) orthograde propulsion followed by retropulsion (not observed in Tph1(+/+) colon); (4) slower in vitro propulsion of larger faecal pellets (28% of Tph1(+/+)); (5) CMMCs that infrequently propagated in an oral to anal direction because of impaired descending inhibition; (6) reduced CMMCs and inhibitory responses to intraluminal balloon distension; (7) an absence of reflex activity in response to mucosal stimulation. In addition, (8) thin pellets that propagated along the control colon failed to do so in Tph1(-/-) colon; and (9) the 5-HT3 receptor antagonist ondansetron, which reduced CMMCs and blocked their propagation in Tph1(+/+) mice, failed to alter CMMCs in Tph1(-/-) animals. Our observations suggest that mucosal 5-HT is essential for reflexes driven by mucosal stimulation and is also important for normal propagation of CMMCs and propulsion of pellets in the isolated colon.


Subject(s)
Colon/physiology , Intestinal Mucosa/physiology , Myoelectric Complex, Migrating/physiology , Serotonin/physiology , Tryptophan Hydroxylase/physiology , Animals , Colon/anatomy & histology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peristalsis/physiology , Receptors, Serotonin, 5-HT3/physiology , Reflex
8.
Am J Physiol Gastrointest Liver Physiol ; 303(9): G1004-16, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22961801

ABSTRACT

The mechanisms underlying slow-transit constipation (STC) are unclear. In 50% of patients with STC, some form of outlet obstruction has been reported; also an elongated colon has been linked to patients with STC. Our aims were 1) to develop a murine model of STC induced by partial outlet obstruction and 2) to determine whether this leads to colonic elongation and, consequently, activation of the inhibitory "occult reflex," which may contribute to STC in humans. Using a purse-string suture, we physically reduced the maximal anal sphincter opening in C57BL/6 mice. After 4 days, the mice were euthanized (acutely obstructed), the suture was removed (relieved), or the suture was removed and replaced repeatedly (chronically obstructed, over 24-31 days). In partially obstructed mice, we observed increased cyclooxygenase (COX)-2 levels in muscularis and mucosa, an elongated impacted large bowel, slowed transit, nonpropagating colonic migrating motor complexes (CMMCs), a lack of mucosal reflexes, a depolarized circular muscle with slow-wave activity due to a lack of spontaneous inhibitory junction potentials, muscle hypertrophy, and CMMCs in mucosa-free preparations. Elongation of the empty obstructed colon produced a pronounced occult reflex. Removal of the obstruction or addition of a COX-2 antagonist (in vitro and in vivo) restored membrane potential, spontaneous inhibitory junction potentials, CMMC propagation, and mucosal reflexes. We conclude that partial outlet obstruction increases COX-2 leading to a hyperexcitable colon. This hyperexcitability is largely due to suppression of only descending inhibitory nerve pathways by prostaglandins. The upregulation of motility is suppressed by the occult reflex activated by colonic elongation.


Subject(s)
Constipation , Gastrointestinal Motility , Intestine, Large , Reflex, Stretch/physiology , Animals , Constipation/etiology , Constipation/metabolism , Constipation/physiopathology , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Disease Models, Animal , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Humans , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Intestinal Obstruction/complications , Intestinal Obstruction/metabolism , Intestinal Obstruction/physiopathology , Intestine, Large/metabolism , Intestine, Large/physiopathology , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Muscle, Smooth/innervation , Muscle, Smooth/metabolism , Myenteric Plexus/metabolism , Myoelectric Complex, Migrating/drug effects
9.
Exp Eye Res ; 100: 73-85, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22579493

ABSTRACT

Extraocular muscles (EOMs) have unique calcium handling properties, yet little is known about the dynamics of calcium events underlying ultrafast and tonic contractions in myofibers of intact EOMs. Superior oblique EOMs of juvenile chickens were dissected with their nerve attached, maintained in oxygenated Krebs buffer, and loaded with fluo-4. Spontaneous and nerve stimulation-evoked calcium transients were recorded and, following calcium imaging, some EOMs were double-labeled with rhodamine-conjugated alpha-bungarotoxin (rhBTX) to identify EOM myofiber types. EOMs showed two main types of spontaneous calcium transients, one slow type (calcium waves with 1/2(max) duration of 2-12 s, velocity of 25-50 µm/s) and two fast "flash-like" types (Type 1, 30-90 ms; Type 2, 90-150 ms 1/2(max) duration). Single pulse nerve stimulation evoked fast calcium transients identical to the fast (Type 1) calcium transients. Calcium waves were accompanied by a local myofiber contraction that followed the calcium transient wavefront. The magnitude of calcium-wave induced myofiber contraction far exceeded those of movement induced by nerve stimulation and associated fast calcium transients. Tetrodotoxin eliminated nerve-evoked transients, but not spontaneous transients. Alpha-bungarotoxin eliminated both spontaneous and nerve-evoked fast calcium transients, but not calcium waves, and caffeine increased wave activity. Calcium waves were observed in myofibers lacking spontaneous or evoked fast transients, suggestive of multiply-innervated myofibers, and this was confirmed by double-labeling with rhBTX. We propose that the abundant spontaneous calcium transients and calcium waves with localized contractions that do not depend on innervation may contribute to intrinsic generation of tonic functions of EOMs.


Subject(s)
Calcium/metabolism , Oculomotor Muscles/innervation , Oculomotor Muscles/metabolism , Oculomotor Nerve/metabolism , Aniline Compounds/metabolism , Animals , Animals, Newborn , Bungarotoxins/pharmacology , Calcium Signaling , Chickens , Fluorescent Dyes/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Muscle Contraction/physiology , Rabbits , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Xanthenes/metabolism
10.
J Physiol ; 590(2): 335-50, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22063626

ABSTRACT

Enteric glia cells (EGCs) form a dense network around myenteric neurons in a ganglia and are likely to have not only a supportive role but may also regulate or be regulated by neural activity. Our aims were to determine if EGCs are activated during the colonic migrating motor complex (CMMC) in the isolated murine colon. Strips of longitudinal muscle were removed and Ca(2+) imaging (Fluo-4) used to study activity in EGCs within myenteric ganglia during CMMCs, followed by post hoc S100 staining to reveal EGCs. The cell bodies of EGCs and their processes formed caps and halos, respectively, around some neighbouring myenteric neurons. Some EGCs (36%), which were largely quiescent between CMMCs, exhibited prolonged tetrodotoxin (TTX; 1 µm)-sensitive Ca(2+) transients that peaked ∼39 s following a mucosal stimulus that generated the CMMC, and often outlasted the CMMC (duration ∼23 s). Ca(2+) transients in EGCs often varied in duration within a ganglion; however, the duration of these transients was closely matched by activity in closely apposed nerve varicosities, suggesting EGCs were not only innervated but the effective innervation was localized. Furthermore, all EGCs, even those that were quiescent, responded with robust Ca(2+) transients to KCl, caffeine, nicotine, substance P and GR 64349 (an NK2 agonist), suggesting they were adequately loaded with indicator and that some EGCs may be inhibited by substances released by neighbouring neurons. Intracellular Ca(2+) waves were visualised propagating between closely apposed glia and from glial cell processes to the soma (velocity 12 µm s(-1)) where they produced an accumulative rise in Ca(2+), suggesting that the soma acts as an integrator of Ca(2+) activity. In conclusion, Ca(2+) transients in EGCs occur secondary to nerve activity; their activation is driven by intrinsic excitatory nerve pathways that generate the CMMC.


Subject(s)
Calcium/metabolism , Colon/innervation , Myenteric Plexus/metabolism , Myoelectric Complex, Migrating/physiology , Neuroglia/metabolism , Animals , Caffeine/pharmacology , Mice , Mice, Inbred C57BL , Models, Animal , Myenteric Plexus/cytology , Myenteric Plexus/drug effects , Neuroglia/cytology , Neuroglia/drug effects , Nicotine/pharmacology , Potassium Chloride/pharmacology , Substance P/pharmacology , Tetrodotoxin/pharmacology
11.
J Physiol ; 588(Pt 22): 4453-74, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20876203

ABSTRACT

Colonic migrating motor complexes (CMMCs) are neurally mediated, cyclical contractile and electrical events, which typically propagate along the colon every 2-3 min in the mouse. We examined the interactions between myenteric neurons, interstitial cells of Cajal in the myenteric region (ICC-MY) and smooth muscle cells during CMMCs using Ca(2+) imaging. CMMCs occurred spontaneously or were evoked by stimulating the mucosa locally, or by brushing it at either end of the colon. Between CMMCs, most ICC-MY were often quiescent; their lack of activity was correlated with ongoing Ca(2+) transients in varicosities on the axons of presumably inhibitory motor neurons that were on or surrounded ICC-MY. Ca(2+) transients in other varicosities initiated intracellular Ca(2+) waves in adjacent ICC-MY, which were blocked by atropine, suggesting they were on the axons of excitatory motor neurons. Following TTX (1 µM), or blockade of inhibitory neurotransmission with N(ω)-nitro-L-arginine (L-NA, a NO synthesis inhibitor, 10 µM) and MRS 2500 (a P2Y(1) antagonist, 1 µM), ongoing spark/puff like activity and rhythmic intracellular Ca(2+) waves (38.1 ± 2.9 cycles min(-1)) were observed, yet this activity was uncoupled, even between ICC-MY in close apposition. During spontaneous or evoked CMMCs there was an increase in the frequency (62.9 ± 1.4 cycles min(-1)) and amplitude of Ca(2+) transients in ICC-MY and muscle, which often had synchronized activity. At the same time, activity in varicosites along excitatory and inhibitory motor nerve fibres increased and decreased respectively, leading to an overall excitation of ICC-MY. Atropine (1 µM) reduced the evoked responses in ICC-MY, and subsequent addition of an NK1 antagonist (RP 67580, 500 nM) completely blocked the responses to stimulation, as did applying these drugs in reverse order. An NKII antagonist (MEN 10,376, 500 nM) had no effect on the evoked responses in ICC-MY. Following TTX application, carbachol (1 µM), substance P (1 µM) and an NKI agonist (GR73632, 100 nM) produced the fast oscillations superimposed on a slow increase in Ca(2+) in ICC-MY, whereas SNP (an NO donor, 10 µM) abolished all activity in ICC-MY. In conclusion, ICC-MY, which are under tonic inhibition, are pacemakers whose activity can be synchronized by excitatory nerves to couple the longitudinal and circular muscles during the CMMC. ICC-MY receive excitatory input from motor neurons that release acetylcholine and tachykinins acting on muscarinic and NK1 receptors, respectively.


Subject(s)
Calcium/physiology , Colon/physiology , Interstitial Cells of Cajal/physiology , Intestinal Mucosa/physiology , Myenteric Plexus/physiology , Myoelectric Complex, Migrating/physiology , Animals , Colon/cytology , Interstitial Cells of Cajal/cytology , Intestinal Mucosa/cytology , Intestine, Large/cytology , Intestine, Large/physiology , Mice , Mice, Inbred C57BL , Myenteric Plexus/cytology , Physical Stimulation/methods , Synaptic Transmission/physiology
12.
J Physiol ; 588(Pt 15): 2919-34, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20547675

ABSTRACT

The colonic migrating motor complex (CMMC) is a rhythmically occurring neurally mediated motor pattern. Although the CMMC spontaneously propagates along an empty colon it is responsible for faecal pellet propulsion in the murine large bowel. Unlike the peristaltic reflex, the CMMC is an 'all or none' event that appears to be dependent upon Dogiel Type II/AH neurons for its regenerative slow propagation down the colon. A reduction in the amplitude of CMMCs or an elongated colon have both been thought to underlie slow transit constipation, although whether these phenomena are related has not been considered. In this study we examined the mechanisms by which colonic elongation might affect the CMMC using video imaging of the colon, tension and electrophysiological recordings from the muscle and Ca(2+) imaging of myenteric neurons. As faecal pellets were expelled from the murine colon, it shortened by up to 29%. Elongation of the colon resulted in a linear reduction in the velocity of a faecal pellet and the amplitude of spontaneous CMMCs. Elongation of the oral end of a colonic segment reduced the amplitude of CMMCs, whereas elongation of the anal end of the colon evoked a premature CMMC, and caused the majority of CMMCs to propagate in an anal to oral direction. Dogiel Type II/AH sensory neurons and most other myenteric neurons responded to oral elongation with reduced amplitude and frequency of spontaneous Ca(2+) transients, whereas anal elongation increased their amplitude and frequency in most neurons. The inhibitory effects of colonic elongation were reduced by blocking nitric oxide (NO) production with l-NA (100 mum) and soluble guanylate cyclase with 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ; 10 mum); whereas, l-arginine (1-2 mm) enhanced the inhibitory effects of colonic elongation. In conclusion, polarized neural reflexes can be triggered by longitudinal stretch. The dominant effect of elongation is to reduce CMMCs primarily by inhibiting Dogiel Type II/AH neurons, thus facilitating colonic accommodation and slow transit.


Subject(s)
Gastrointestinal Motility/physiology , Intestine, Large/physiology , Motor Neurons/physiology , Reflex, Stretch/physiology , Animals , Mice , Mice, Inbred C57BL
13.
Am J Physiol Gastrointest Liver Physiol ; 299(1): G144-57, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20413719

ABSTRACT

The colonic migrating motor complex (CMMC) is necessary for fecal pellet propulsion in the murine colon. We have previously shown that 5-hydroxytryptamine (5-HT) released from enterochromaffin cells activates 5-HT(3) receptors on the mucosal processes of myenteric Dogiel type II neurons to initiate the events underlying the CMMC. Our aims were to further investigate the roles of 5-HT(1A), 5-HT(3), and 5-HT(7) receptor subtypes in generating and propagating the CMMC using intracellular microelectrodes or tension recordings from the circular muscle (CM) in preparations with and without the mucosa. Spontaneous CMMCs were recorded from the CM in isolated murine colons but not in preparations without the mucosa. In mucosaless preparations, ondansetron (3 microM; 5-HT(3) antagonist) plus hexamethonium (100 microM) completely blocked spontaneous inhibitory junction potentials, depolarized the CM. Ondansetron blocked the preceding hyperpolarization associated with a CMMC. Spontaneous CMMCs and CMMCs evoked by spritzing 5-HT (10 and 100 microM) or nerve stimulation in preparations without the mucosa were blocked by SB 258719 or SB 269970 (1-5 microM; 5-HT(7) antagonists). Both NAN-190 and (S)-WAY100135 (1-5 microM; 5-HT(1A) antagonists) blocked spontaneous CMMCs and neurally evoked CMMCs in preparations without the mucosa. Both NAN-190 and (S)-WAY100135 caused an atropine-sensitive depolarization of the CM. The precursor of 5-HT, 5-hydroxytryptophan (5-HTP) (10 microM), and 5-carboxamidotryptamine (5-CT) (5 microM; 5-HT(1/5/7) agonist) increased the frequency of spontaneous CMMCs. 5-HTP and 5-CT also induced CMMCs in preparations with and without the mucosa, which were blocked by SB 258719. 5-HT(1A), 5-HT(3), and 5-HT(7) receptors, most likely on Dogiel Type II/AH neurons, are important in initiating, generating, and propagating the CMMC. Tonic inhibition of the CM appears to be driven by ongoing activity in descending serotonergic interneurons; by activating 5-HT(7) receptors on AH neurons these interneurons also contribute to the generation of the CMMC.


Subject(s)
Colon/innervation , Muscle Contraction , Muscle, Smooth/innervation , Myenteric Plexus/metabolism , Myoelectric Complex, Migrating , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Receptors, Serotonin/metabolism , 5-Hydroxytryptophan/metabolism , Animals , Electric Stimulation , Excitatory Postsynaptic Potentials , In Vitro Techniques , Inhibitory Postsynaptic Potentials , Interneurons/metabolism , Intestinal Mucosa/innervation , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects , Myenteric Plexus/drug effects , Myoelectric Complex, Migrating/drug effects , Neural Inhibition , Nicotinic Antagonists/pharmacology , Receptors, Serotonin/drug effects , Serotonin/metabolism , Serotonin 5-HT1 Receptor Antagonists , Serotonin 5-HT3 Receptor Antagonists , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Time Factors
15.
J Physiol ; 588(Pt 3): 399-421, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19948652

ABSTRACT

The spontaneous colonic migrating motor complex (CMMC) is a cyclical contractile and electrical event that is the primary motor pattern underlying fecal pellet propulsion along the murine colon. We have combined Ca(2+) imaging with immunohistochemistry to determine the role of different classes of myenteric neurons during the CMMC. Between CMMCs, myenteric neurons usually displayed ongoing but uncoordinated activity. Stroking the mucosa at the oral or anal end of the colon resulted in a CMMC (latency: 6 to 10 s; duration: 28 s) that consisted of prolonged increases in activity in many myenteric neurons that was correlated to Ca(2+) transients in and displacement of the muscle. These neurons were likely excitatory motor neurons. Activity in individual neurons during the CMMC was similar regardless of whether the CMMC occurred spontaneously or was evoked by anal or oral mucosal stimulation. This suggests that convergent interneuronal pathways exist which generate CMMCs. Interestingly, Ca(2+) transients in a subset of NOS +ve neurons were substantially reduced during the CMMC. These neurons are likely to be inhibitory motor neurons that reduce their activity during a complex (disinhibition) to allow full excitation of the muscle. Local stimulation of the mucosa evoked synchronized Ca(2+) transients in Dogiel Type II (mitotracker/calbindin-positive) neurons after a short delay (1-2 s), indicating they were the sensory neurons underlying the CMMC. These local responses were observed in hexamethonium, but were blocked by ondansetron (5-HT(3) antagonist), suggesting Dogiel Type II neurons were activated by 5-HT release from enterochromaffin cells in the mucosa. In fact, removal of the mucosa yielded no spontaneous CMMCs, although many neurons (NOS +ve and NOS ve) exhibited ongoing activity, including Dogiel Type II neurons. These results suggest that spontaneous or evoked 5-HT release from the mucosa is necessary for the activation of Dogiel Type II neurons that generate CMMCs.


Subject(s)
Calcium/metabolism , Colon/innervation , Motor Neurons/metabolism , Myenteric Plexus/metabolism , Myoelectric Complex, Migrating/physiology , Animals , Colon/metabolism , Female , Gastrointestinal Motility/physiology , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Animal , Nitric Oxide Synthase/metabolism , Serotonin/metabolism , Synaptic Transmission/physiology
16.
Am J Physiol Gastrointest Liver Physiol ; 298(2): G222-32, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19959818

ABSTRACT

Colonic migrating motor complexes (CMMCs) propel fecal contents and are altered in diseased states, including slow-transit constipation. However, the mechanisms underlying the CMMCs are controversial because it has been proposed that disinhibition (turning off of inhibitory neurotransmission) or excitatory nerve activity generate the CMMC. Therefore, our aims were to reexamine the mechanisms underlying the CMMC in the colon of wild-type and neuronal nitric oxide synthase (nNOS)(-/-) mice. CMMCs were recorded from the isolated murine large bowel using intracellular recordings of electrical activity from circular muscle (CM) combined with tension recording. Spontaneous CMMCs occurred in both wild-type (frequency: 0.3 cycles/min) and nNOS(-/-) mice (frequency: 0.4 cycles/min). CMMCs consisted of a hyperpolarization, followed by fast oscillations (slow waves) with action potentials superimposed on a slow depolarization (wild-type: 14.0 +/- 0.6 mV; nNOS(-/-): 11.2 +/- 1.5 mV). Both atropine (1 microM) and MEN 10,376 [neurokinin 2 (NK2) antagonist; 0.5 microM] added successively reduced the slow depolarization and the number of action potentials but did not abolish the fast oscillations. The further addition of RP 67580 (NK1 antagonist; 0.5 microM) blocked the fast oscillations and the CMMC. Importantly, none of the antagonists affected the resting membrane potential, suggesting that ongoing tonic inhibition of the CM was maintained. Fecal pellet propulsion, which was blocked by the NK2 or the NK1 antagonist, was slower down the longer, more constricted nNOS(-/-) mouse colon (wild-type: 47.9 +/- 2.4 mm; nNOS(-/-): 57.8 +/- 1.4 mm). These observations suggest that excitatory neurotransmission enhances pacemaker activity during the CMMC. Therefore, the CMMC is likely generated by a synergistic interaction between neural and interstitial cells of Cajal networks.


Subject(s)
Colon/innervation , Colon/physiology , Myoelectric Complex, Migrating/physiology , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Analgesics/pharmacology , Animals , Atropine/pharmacology , Enzyme Inhibitors/pharmacology , Intestinal Mucosa/innervation , Intestinal Mucosa/physiology , Isoindoles/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscarinic Antagonists/pharmacology , Muscle, Smooth/innervation , Muscle, Smooth/physiology , Myoelectric Complex, Migrating/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitroarginine/pharmacology
17.
Gastroenterology ; 136(7): 2226-36, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19268670

ABSTRACT

BACKGROUND & AIMS: In human and canine colon, both slow (slow waves, 2-8/min) and fast (myenteric potential oscillations [MPOs]; 16-20/min) electrical rhythms in the smooth muscle originate at the submucosal and myenteric borders, respectively. We used Ca(2+) imaging to investigate whether interstitial cells of Cajal (ICCs) at these borders generated distinct rhythms. METHODS: Segments of canine colon were pinned with submucosal or myenteric surface uppermost or cut in cross section. Tissues were loaded with a Ca(2+) indicator (fluo-4), and activity was monitored at 36.5 +/- 0.5 degrees C using an electron multiplying charge coupled device (EMCCD). RESULTS: Rhythmic, biphasic Ca(2+) transients (5-8/min), similar in waveform to electrical slow waves, propagated without decrement as a wave front (2-5 mm/s) through the ICC-SM network lying along the submucosal surface of the circular muscle (CM). In contrast, rhythmic intracellular Ca(2+) waves (approximately 16/min) and spontaneous reductions in Ca(2+) were observed in ICCs at the myenteric border (ICC-MY). Normally, intracellular Ca(2+) waves were unsynchronized between adjacent ICC-MY, although excitatory nerve activity synchronized activity. In addition, spontaneous reductions in Ca(2+) were observed that inhibited Ca(2+) waves. N omega-nitro-L-arginine (100 micromol/L; nitric oxide antagonist) blocked the reductions in Ca(2+) and increased the frequency (approximately 19/min) of intracellular Ca(2+) waves within ICC-MY. CONCLUSIONS: ICC-SMs form a tightly coupled network that is able to generate and propagate slow waves. In contrast, Ca(2+) transients in ICC-MYs, which are normally not synchronized, have a similar duration and frequency as MPOs. Like MPOs, their activity is inhibited by nitrergic nerves and synchronized by excitatory nerves.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Colon/innervation , Muscle Contraction/physiology , Myenteric Plexus/physiology , Action Potentials/physiology , Animals , Colon/metabolism , Dogs , Electrophysiology , Enteric Nervous System/physiology , Female , Gastrointestinal Motility/physiology , Intestinal Mucosa/innervation , Intestinal Mucosa/physiology , Male , Membrane Potentials , Microscopy, Fluorescence , Models, Animal , Myenteric Plexus/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Nitric Oxide/metabolism , Sensitivity and Specificity
18.
Gastroenterology ; 136(4): 1328-38, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19138686

ABSTRACT

BACKGROUND & AIMS: The colonic migrating motor complex (CMMC) is a motor pattern that regulates the movement of fecal matter through a rhythmic sequence of electrical activity and/or contractions along the large bowel. CMMCs have largely been studied in empty preparations; we investigated whether local reflexes generated by a fecal pellet modify the CMMC to initiate propulsive activity. METHODS: Recordings of CMMCs were made from the isolated murine large bowel, with or without a fecal pellet. Transducers were placed along the colon to record muscle tension and propulsive force on the pellet and microelectrodes were used to record electrical activity from either side of a fecal pellet, circular muscle cells oral and anal of a pellet, and in colons without the mucosa. RESULTS: Spontaneous CMMCs propagated in both an oral or anal direction. When a pellet was inserted, CMMCs increased in frequency and propagated anally, exerting propulsive force on the pellet. The amplitude of slow waves increased during the CMMC. Localized mucosal stimulation/circumferential stretch evoked a CMMC, regardless of stimulus strength. The serotonin (5-hydroxytryptamine-3) receptor antagonist ondansetron reduced the amplitude of the CMMC, the propulsive force on the pellet, and the response to mucosal stroking, but increased the apparent conduction velocity of the CMMC. Removing the mucosa abolished spontaneous CMMCs, which still could be evoked by electrical stimulation. CONCLUSIONS: The fecal pellet activates local mucosal reflexes, which release serotonin (5-hydroxytryptamine) from enterochromaffin cells, and stretch reflexes that determine the site of origin and propagation of the CMMC, facilitating propulsion.


Subject(s)
Colon/innervation , Colon/physiology , Feces , Gastrointestinal Motility/physiology , Motor Neurons/physiology , Serotonin/metabolism , Animals , Evoked Potentials/physiology , Female , Gastrointestinal Motility/drug effects , Intestinal Mucosa/innervation , Intestinal Mucosa/physiology , Male , Mice , Mice, Inbred C57BL , Microelectrodes , Muscle Contraction/physiology , Muscle, Smooth/innervation , Muscle, Smooth/physiology , Ondansetron/pharmacology , Serotonin Antagonists/pharmacology
19.
J Physiol ; 586(20): 4977-91, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18755751

ABSTRACT

We investigated intracellular Ca(2+) waves, spontaneous transient outward currents (STOCs), and membrane potentials of gastric antrum smooth muscle cells from wild-type and phospholamban-knockout mice. The NO donor sodium nitroprusside (SNP) increased intracellular Ca(2+) wave activity in wild-type antrum smooth muscle cells, but had no effect on the constitutively elevated intracellular Ca(2+) wave activity of phospholamban-knockout cells. STOC activity was also constitutively elevated in phospholamban-knockout antrum smooth muscle cells relative to wild-type cells. SNP or 8-bromo-cGMP increased the STOC activity of wild-type antrum smooth muscle cells, but had no effect on STOC activity of phospholamban-knockout cells. Iberiotoxin, but not apamin, inhibited STOC activity in wild-type and phospholamban-knockout antrum smooth muscle cells. In the presence of SNP, STOC activity in wild-type and phospholamban-knockout antrum smooth muscle cells was inhibited by ryanodine, but not 2-APB. The cGMP-dependent protein kinase inhibitor KT5823 reversed the increase in STOC activity evoked by SNP in wild-type antrum smooth muscle cells, but had no effect on STOC activity in phospholamban-knockout cells. The resting membrane potential of phospholamban-knockout antrum smooth muscle cells was hyperpolarized by approximately -6 mV compared to wild-type cells. SNP hyperpolarized the resting membrane potential of wild-type antrum smooth muscle cells to a greater extent than phospholamban-knockout antrum smooth muscles. Despite the hyperpolarized membrane potential, slow wave activity was significantly increased in phospholamban-knockout antrum smooth muscles compared to wild-type smooth muscles. These results suggest that phospholamban is an important component of the mechanisms regulating the electrical properties of gastric antrum smooth muscles.


Subject(s)
Calcium Signaling/physiology , Calcium-Binding Proteins/administration & dosage , Calcium/metabolism , Miniature Postsynaptic Potentials/physiology , Myocytes, Smooth Muscle/physiology , Pyloric Antrum/physiology , Animals , Calcium Signaling/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Miniature Postsynaptic Potentials/drug effects , Muscle, Smooth , Myocytes, Smooth Muscle/drug effects , Pyloric Antrum/cytology , Pyloric Antrum/drug effects
20.
J Physiol ; 586(17): 4225-40, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18635646

ABSTRACT

Propulsion in both small and large intestine is largely mediated by the peristaltic reflex; despite this, transit through the shorter colon is at least 10 times slower. Recently we demonstrated that elongating a segment of colon releases nitric oxide (NO) to inhibit peristalsis. The aims of this study were to determine if colonic elongation was physiologically significant, and whether elongation activated polarized intrinsic neural reflexes. Video imaging monitored fecal pellet evacuation from isolated guinea-pig colons full of pellets. Recordings were made from the circular muscle (CM) and longitudinal muscle (LM) in flat sheet preparations using either intracellular microelectrode or Ca(2+) imaging techniques. Full colons were 158.1 +/- 6.1% longer than empty colons. As each pellet was expelled, the colon shortened and pellet velocity increased exponentially (full 0.34, empty 1.01 mm s(-1)). In flat sheet preparations, maintained circumferential stretch generated ongoing peristaltic activity (oral excitatory and anal inhibitory junction potentials) and Ca(2+) waves in LM and CM. Colonic elongation (140% of its empty slack length) applied oral to the recording site abolished these activities, whereas anal elongation significantly increased the frequency and amplitude of ongoing peristaltic activity. Oral elongation inhibited the excitation produced by anal elongation; this inhibitory effect was reversed by blocking NO synthesis. Pelvic nerve stimulation elicited polarized responses that were also suppressed by NO released during colonic elongation. In conclusion, longitudinal stretch excites specific mechanosensitive ascending and descending interneurons, leading to activation of polarized reflexes. The dominance of the descending inhibitory reflex leads to slowed emptying of pellets in a naturally elongated colon.


Subject(s)
Colon/physiology , Mechanoreceptors/physiology , Physical Stimulation , Reflex/physiology , Animals , Colon/drug effects , Gastrointestinal Contents , Guinea Pigs , Hexamethonium/pharmacology , Male , Mechanotransduction, Cellular , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/physiology , Neural Pathways/physiology , Nicotinic Antagonists/pharmacology , Peristalsis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...