Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Tissue Res ; 396(3): 313-327, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38383905

RESUMEN

Understanding how the gut communicates with the brain, via sensory nerves, is of significant interest to medical science. Enteroendocrine cells (EEC) that line the mucosa of the gastrointestinal tract release neurochemicals, including the largest quantity of 5-hydroxytryptamine (5-HT). How the release of substances, like 5-HT, from enterochromaffin (EC) cells activates vagal afferent nerve endings is unresolved. We performed anterograde labelling from nodose ganglia in vivo and identified vagal afferent axons and nerve endings in the mucosa of whole-mount full-length preparations of mouse colon. We then determined the spatial relationship between mucosal-projecting vagal afferent nerve endings and EC cells in situ using 3D imaging. The mean distances between vagal afferent nerve endings in the mucosa, or nearest varicosities along vagal afferent axon branches, and the nearest EC cell were 29.6 ± 19.2 µm (n = 107, N = 6) and 25.7 ± 15.2 µm (n = 119, N = 6), respectively. No vagal afferent endings made close contacts with EC cells. The distances between EC cells and vagal afferent endings are many hundreds of times greater than known distances between pre- and post-synaptic membranes (typically 10-20 nm) that underlie synaptic transmission in vertebrates. The absence of any close physical contacts between 5-HT-containing EC cells and vagal afferent nerve endings in the mucosa leads to the inescapable conclusion that the mechanism by which 5-HT release from ECs in the colonic mucosa occurs in a paracrine fashion, to activate vagal afferents.


Asunto(s)
Colon , Células Enterocromafines , Nervio Vago , Animales , Células Enterocromafines/metabolismo , Colon/inervación , Nervio Vago/fisiología , Ratones , Ratones Endogámicos C57BL , Masculino , Terminaciones Nerviosas , Ganglio Nudoso/citología , Neuronas Aferentes
2.
J Neurosci ; 41(17): 3900-3916, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33727332

RESUMEN

Understanding the sensory mechanisms innervating the bladder is paramount to developing efficacious treatments for chronic bladder hypersensitivity conditions. The contribution of Mas-gene-related G protein-coupled receptors (Mrgpr) to bladder signaling is currently unknown. Using male and female mice, we show with single-cell RT-PCR that subpopulations of DRG neurons innervating the mouse bladder express MrgprA3 (14%) and MrgprC11 (38%), either individually or in combination, with high levels of coexpression with Trpv1 (81%-89%). Calcium imaging studies demonstrated MrgprA3 and MrgprC11 agonists (chloroquine, BAM8-22, and neuropeptide FF) activated subpopulations of bladder-innervating DRG neurons, showing functional evidence of coexpression between MrgprA3, MrgprC11, and TRPV1. In ex vivo bladder-nerve preparations, chloroquine, BAM8-22, and neuropeptide FF all evoked mechanical hypersensitivity in subpopulations (20%-41%) of bladder afferents. These effects were absent in recordings from Mrgpr-clusterΔ-/- mice. In vitro whole-cell patch-clamp recordings showed that application of an MrgprA3/C11 agonist mixture induced neuronal hyperexcitability in 44% of bladder-innervating DRG neurons. Finally, in vivo instillation of an MrgprA3/C11 agonist mixture into the bladder of WT mice induced a significant activation of dorsal horn neurons within the lumbosacral spinal cord, as quantified by pERK immunoreactivity. This MrgprA3/C11 agonist-induced activation was particularly apparent within the superficial dorsal horn and the sacral parasympathetic nuclei of WT, but not Mrgpr-clusterΔ-/- mice. This study demonstrates, for the first time, functional expression of MrgprA3 and MrgprC11 in bladder afferents. Activation of these receptors triggers hypersensitivity to distension, a critically valuable factor for therapeutic target development.SIGNIFICANCE STATEMENT Determining how bladder afferents become sensitized is the first step in finding effective treatments for common urological disorders such as overactive bladder and interstitial cystitis/bladder pain syndrome. Here we show that two of the key receptors, MrgprA3 and MrgprC11, that mediate itch from the skin are also expressed on afferents innervating the bladder. Activation of these receptors results in sensitization of bladder afferents, resulting in sensory signals being sent into the spinal cord that prematurely indicate bladder fullness. Targeting bladder afferents expressing MrgprA3 or MrgprC11 and preventing their sensitization may provide a novel approach for treating overactive bladder and interstitial cystitis/bladder pain syndrome.


Asunto(s)
Neuronas Aferentes/fisiología , Receptores Acoplados a Proteínas G/fisiología , Vejiga Urinaria/inervación , Animales , Femenino , Ganglios Espinales/fisiología , Plexo Lumbosacro/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Estimulación Física , Células del Asta Posterior/fisiología , Canales Catiónicos TRPV/fisiología
3.
Am J Physiol Gastrointest Liver Physiol ; 322(5): G523-G533, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35293258

RESUMEN

Cross talk between the gastrointestinal tract and brain is of significant relevance for human health and disease. However, our understanding of how the gut and brain communicate has been limited by a lack of techniques to identify the precise spatial relationship between extrinsic nerve endings and their proximity to specific cell types that line the inner surface of the gastrointestinal tract. We used an in vivo anterograde tracing technique, previously developed in our laboratory, to selectively label single spinal afferent axons and their nerve endings in mouse colonic mucosa. The closest three-dimensional distances between spinal afferent nerve endings and axonal varicosities to enterochromaffin (EC) cells, which contain serotonin (5-hydroxytryptamine; 5-HT), were then measured. The mean distances (± standard deviation) between any varicosity along a spinal afferent axon or its nerve ending, and the nearest EC cell, were 5.7 ± 6.0 µm (median: 3.6 µm) and 26.9 ± 18.6 µm (median: 24.1 µm), respectively. Randomization of the spatial location of EC cells revealed similar results to this actual data. These distances are ∼200-1,000 times greater than those between pre- and postsynaptic membranes (15-25 nm) that underlie synaptic transmission in the vertebrate nervous system. Our findings suggest that colonic 5-HT-containing EC cells release substances to activate centrally projecting spinal afferent nerves likely via diffusion, as such signaling is unlikely to occur with the spatial fidelity of a synapse.NEW & NOTEWORTHY We show an absence of close physical contact between spinal afferent nerves and 5-HT-containing EC cells in mouse colonic mucosa. Similar relative distances were observed between randomized EC cells and spinal afferents compared with actual data. This spatial relationship suggests that substances released from colonic 5-HT-containing EC cells are unlikely to act via synaptic transmission to neighboring spinal afferents that relay sensory information from the gut lumen to the brain.


Asunto(s)
Células Enterocromafines , Serotonina , Animales , Eje Cerebro-Intestino , Colon/metabolismo , Células Enterocromafines/metabolismo , Ratones , Serotonina/metabolismo
4.
Am J Physiol Gastrointest Liver Physiol ; 323(2): G71-G87, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35502864

RESUMEN

Colonic motor complexes (CMCs) are a major neurogenic activity in guineapig distal colon. The identity of the enteric neurons that initiate this activity is not established. Specialized intrinsic primary afferent neurons (IPANs) are a major candidate. We aimed to test this hypothesis. To do this, segments of guineapig distal colon were suspended vertically in heated organ baths and propulsive forces acting on a pellet inside the lumen were recorded by isometric force transducer while pharmacological agents were applied to affect IPAN function. In the absence of drugs, CMCs acted periodically on the pellet, generating peak propulsive forces of 12.7 ± 5 g at 0.56 ± 0.22 cpm, lasting 49 ± 17 s (215 preparations; n = 60). Most but not all CMCs were abolished by nicotinic receptor blockade to inhibit fast excitatory synaptic transmission (50/62 preparations; n = 25). Remarkably, CMCs inhibited by hexamethonium were restored by a pharmacological strategy that aimed to enhance IPAN excitability. Thus, CMCs were restored by increased smooth muscle tension (using BAY K8644, bethanechol or carbachol) and by IPAN excitation using phorbol dibutyrate; NK3 receptor agonist, senktide; and partially by αCGRP. The IPAN inhibitor, 5,6-dichloro-1-ethyl-1,3-dihydro-2H-benzimidazole-2-one (DCEBIO), decreased CMC frequency. CGRP, but not NK3-receptor antagonists, decreased CMC frequency in naive preparations. Finally, CMCs were blocked by tetrodotoxin, and this was not reversed by any drugs listed above. These results support a major role for IPANs that does not require fast synaptic transmission, in the periodic initiation of neurogenic propulsive contractions. Endogenous CGRP plays a role in determining CMC frequency, whereas further unidentified signaling pathways may determine their amplitude and duration.NEW & NOTEWORTHY The colonic motor complex (CMC) initiates propulsion in guinea pig colon. Here, CMCs evoked by an intraluminal pellet were restored during nicotinic receptor blockade by pharmacological agents that directly or indirectly enhance intrinsic primary afferent neuron (IPAN) excitability. IPANs are the only enteric neuron in colon that contain CGRP. Blocking CGRP receptors decreased CMC frequency, implicating their role in CMC initiation. The results support a role for IPANs in the initiation of CMCs.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina , Receptores Nicotínicos , Animales , Colon , Cobayas , Hexametonio/farmacología , Transmisión Sináptica
5.
Gastroenterology ; 160(7): 2451-2466.e19, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33662386

RESUMEN

BACKGROUND & AIMS: Gastrointestinal (GI) motility is regulated by serotonin (5-hydroxytryptamine [5-HT]), which is primarily produced by enterochromaffin (EC) cells in the GI tract. However, the precise roles of EC cell-derived 5-HT in regulating gastric motility remain a major point of conjecture. Using a novel transgenic mouse line, we investigated the distribution of EC cells and the pathophysiologic roles of 5-HT deficiency in gastric motility in mice and humans. METHODS: We developed an inducible, EC cell-specific Tph1CreERT2/+ mouse, which was used to generate a reporter mouse line, Tph1-tdTom, and an EC cell-depleted line, Tph1-DTA. We examined EC cell distribution, morphology, and subpopulations in reporter mice. GI motility was measured in vivo and ex vivo in EC cell-depleted mice. Additionally, we evaluated 5-HT content in biopsy and plasma specimens from patients with idiopathic gastroparesis (IG). RESULTS: Tph1-tdTom mice showed EC cells that were heterogeneously distributed throughout the GI tract with the greatest abundance in the antrum and proximal colon. Two subpopulations of EC cells were identified in the gut: self-renewal cells located at the base of the crypt and mature cells observed in the villi. Tph1-DTA mice displayed delayed gastric emptying, total GI transit, and colonic transit. These gut motility alterations were reversed by exogenous provision of 5-HT. Patients with IG had a significant reduction of antral EC cell numbers and 5-HT content, which negatively correlated with gastric emptying rate. CONCLUSIONS: The Tph1CreERT2/+ mouse provides a powerful tool to study the functional roles of EC cells in the GI tract. Our findings suggest a new pathophysiologic mechanism of 5-HT deficiency in IG.


Asunto(s)
Vaciamiento Gástrico/genética , Tránsito Gastrointestinal/genética , Serotonina/deficiencia , Animales , Línea Celular , Células Enterocromafines/fisiología , Humanos , Ratones , Ratones Transgénicos , Triptófano Hidroxilasa/metabolismo
6.
Adv Exp Med Biol ; 1383: 295-306, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36587167

RESUMEN

The enteric nervous system (ENS) is required for many cyclical patterns of motor activity along different regions of the gastrointestinal (GI) tract. What has remained mysterious is precisely how many thousands of neurons within the ENS are temporally activated to generate cyclical neurogenic contractions of GI-smooth muscle layers. This has been an especially puzzling conundrum, since the ENS consists of an extensive network of small ganglia, with each ganglion consisting of a heterogeneous population of neurons, with diverse cell soma morphologies, neurochemical and biophysical characteristics, and neural connectivity. Neuronal imaging studies of the mouse large intestine have provided major new insights into how the different classes of myenteric neurons are activated during cyclical neurogenic motor patterns, such as the colonic motor complex (CMC). It has been revealed that during CMCs (in the isolated mouse whole colon), large populations of myenteric neurons, across large spatial fields, coordinate their firing, via bursts of fast synaptic inputs at ~2 Hz. This coordinated firing of many thousands of myenteric neurons synchronously over many rows of interconnected ganglia occurs irrespective of the functional class of neuron. Aborally directed propulsion of content along the mouse colon is due, in large part, to polarity of the enteric circuits including the projections of the intrinsic excitatory and inhibitory motor neurons but still involves the fundamental ~2 Hz rhythmic activity of specific classes of enteric neurons. What remains to be determined are the mechanisms that initiate and terminate the patterned firing of large ensembles of enteric neurons during cyclic activity. This remains an exciting challenge for future studies.


Asunto(s)
Sistema Nervioso Entérico , Ratones , Animales , Sistema Nervioso Entérico/fisiología , Tracto Gastrointestinal , Colon , Neuronas Motoras/fisiología , Periodicidad
7.
Adv Exp Med Biol ; 1383: 89-103, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36587149

RESUMEN

The autonomic nervous system that regulates the gut is divided into sympathetic (SNS), parasympathetic (PNS), and enteric nervous systems (ENS). They inhibit, permit, and coordinate gastrointestinal motility, respectively. A fourth pathway, "extrinsic sensory neurons," connect gut to the central nervous system, mediating sensation. The ENS resides within the gut wall and its activities are critical for life; ENS failure to populate the gut in development is lethal without intervention."Viscerofugal neurons" are a distinctive class of enteric neurons, being the only type that escapes the gut wall. They form a unique circuit: their axons project out of the gut wall and activate sympathetic neurons, which then project back to the gut, and inhibit gut movements.For 80 years viscerofugal/sympathetic circuits were thought to have a restricted role, mediating simple sensory-motor reflexes. New data shows viscerofugal and sympathetic neurons behaving unexpectedly, compelling a re-evaluation of these circuits: both viscerofugal and sympathetic neurons transmit higher order, synchronized firing patterns that originate within the ENS. This identifies them as driving long-range motility control between different gut regions.There is need for gut motor control over distances beyond the range of ENS circuits, yet no mechanism has been identified to date. The entero-sympathetic circuits are ideally suited to meet this need. Here we provide an overview of the structure and functions of these peripheral sympathetic circuits, including new data showing the firing patterns generated by enteric networks can transmit through sympathetic neurons.


Asunto(s)
Sistema Nervioso Entérico , Sistema Nervioso Entérico/metabolismo , Sistema Nervioso Autónomo , Sistema Nervioso Simpático , Células Receptoras Sensoriales , Sistema Nervioso Central
8.
Adv Exp Med Biol ; 1383: 113-123, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36587151

RESUMEN

The characteristic motor patterns of the colon are coordinated by the enteric nervous system (ENS) and involve enterochromaffin (EC) cells, enteric glia, smooth muscle fibers, and interstitial cells. While the fundamental control mechanisms of colonic motor patterns are understood, greater complexity in the circuitry underlying motor patterns has been revealed by recent advances in the field. We review these recent advances and new findings from our laboratories that provide insights into how the ENS coordinates motor patterns in the isolated mouse colon. We contextualize these observations by describing the neuromuscular system underling the colonic motor complex (CMC) as a robust, distributed control system. Framing the colonic motor complex as a control system reveals a new perspective on the coordinated motor patterns in the colon. We test the control system by applying electrical stimulation in the isolated mouse colon to disrupt the coordination and propagation of the colonic motor complex.


Asunto(s)
Sistema Nervioso Entérico , Células Intersticiales de Cajal , Animales , Ratones , Colon , Intestino Delgado , Sistema Nervioso Entérico/fisiología , Miocitos del Músculo Liso , Motilidad Gastrointestinal/fisiología
9.
Adv Exp Med Biol ; 1383: 271-294, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36587166

RESUMEN

Over 150 years ago, methods for quantitative analysis of gastrointestinal motor patterns first appeared. Graphic representations of physiological variables were recorded with the kymograph after the mid-1800s. Changes in force or length of intestinal muscles could be quantified, however most recordings were limited to a single point along the digestive tract.In parallel, photography and cinematography with X-Rays visualised changes in intestinal shape, but were hard to quantify. More recently, the ability to record physiological events at many sites along the gut in combination with computer processing allowed construction of spatiotemporal maps. These included diameter maps (DMaps), constructed from video recordings of intestinal movements and pressure maps (PMaps), constructed using data from high-resolution manometry catheters. Combining different kinds of spatiotemporal maps revealed additional details about gut wall status, including compliance, which relates forces to changes in length. Plotting compliance values along the intestine enabled combined DPMaps to be constructed, which can distinguish active contractions and relaxations from passive changes. From combinations of spatiotemporal maps, it is possible to deduce the role of enteric circuits and pacemaker cells in the generation of complex motor patterns. Development and application of spatiotemporal methods to normal and abnormal motor patterns in animals and humans is ongoing, with further technical improvements arising from their combination with impedance manometry, magnetic resonance imaging, electrophysiology, and ultrasonography.


Asunto(s)
Motilidad Gastrointestinal , Intestino Delgado , Humanos , Animales , Motilidad Gastrointestinal/fisiología , Manometría/métodos , Grabación en Video , Músculos
10.
Adv Exp Med Biol ; 1383: 243-249, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36587163

RESUMEN

Distinguishing and characterising the different classes of neurons that make up a neural circuit has been a long-term goal for many neuroscientists. The enteric nervous system is a large but moderately simple part of the nervous system. Enteric neurons in laboratory animals have been extensively characterised morphologically, electrophysiologically, by projections and immunohistochemically. However, studies of human enteric nervous system are less advanced despite the potential availability of tissue from elective surgery (with appropriate ethics permits). Recent studies using single cell sequencing have confirmed and extended the classification of enteric neurons in mice and human, but it is not clear whether an encompassing classification has been achieved. We present preliminary data on a means to distinguish classes of myenteric neurons in specimens of human colon combining immunohistochemical, morphological, projection and size data on single cells. A method to apply multiple layers of antisera to specimens was developed, allowing up to 12 markers to be characterised in individual neurons. Applied to multi-axonal Dogiel type II neurons, this approach demonstrated that they constitute fewer than 5% of myenteric neurons, are nearly all immunoreactive for choline acetyltransferase and tachykinins. Many express the calcium-binding proteins calbindin and calretinin and they are larger than average myenteric cells. This methodology provides a complementary approach to single-cell mRNA profiling to provide a comprehensive account of the types of myenteric neurons in the human colon.


Asunto(s)
Sistema Nervioso Entérico , Plexo Mientérico , Humanos , Ratones , Animales , Plexo Mientérico/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Sistema Nervioso Entérico/metabolismo , Neuronas/fisiología , Colon/metabolismo
11.
J Physiol ; 599(20): 4561-4579, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34418078

RESUMEN

Soft faecal material is transformed into discrete, pellet-shaped faeces at the colonic flexure. Here, analysis of water content in natural faecal material revealed a decline from cecum to rectum without significant changes at the flexure. Thus, pellet formation is not explained by changes in viscosity alone. We then used video imaging of colonic wall movements with electromyography in isolated preparations containing guinea-pig proximal colon, colonic flexure and distal colon. To investigate the pellet formation process, the colonic segments were infused with artificial contents (Krebs solution and 4-6% methylcellulose) to simulate physiological faecal content flow. Remarkably, pellet formation took place in vitro, without extrinsic neural inputs. Infusion evoked slowly propagating neurogenic contractions, the proximal colon migrating motor complexes (∼0.6 cpm), which initiated pellet formation at the flexure. Lesion of the flexure, but not the proximal colon, disrupted the formation of normal individual pellets. In addition, a distinct myogenic mechanism was identified, whereby slow phasic contractions (∼1.9 cpm) initiated at the flexure and propagated short distances retrogradely into the proximal colon and antegradely into the distal colon. There were no detectable changes in the density or distribution of pacemaker-type interstitial cells of Cajal across the flexure. The findings provide new insights into how solid faecal content is generated, suggesting the major mechanisms underlying faecal pellet formation involve the unique interaction at the colonic flexure between antegrade proximal colon migrating motor complexes, organized by enteric neurons, and retrograde myogenic slow phasic contractions. Additional, as yet unidentified extrinsic and/or humoral influences appear to contribute to processing of faecal content in vivo. KEY POINTS: In herbivores, including guinea-pigs, clearly defined faecal pellets are formed at a distinct location along the large intestine (colonic flexure). The mechanism underlying the formation of these faecal pellets at this region has remained unknown. We reveal a progressive and gradual reduction in water content of faecal content along the bowel. Hence, the distinct transition from amorphous to pellet shaped faecal content could not be explained by a dramatic increase in water reabsorption from a specific site. We discovered patterns of anterograde neurogenic and retrograde myogenic motor activity that facilitate the formation of faecal pellets. The formation of 'pellet-like' boluses at the colonic flexure involves interaction of an antegrade migrating motor complex in the proximal colon and retrograde myogenic slow phasic contractions that emerge from the colonic flexure. The findings uncover intrinsic mechanisms responsible for the formation of discrete faecal scybala in the large intestine of a vertebrate.


Asunto(s)
Motilidad Gastrointestinal , Complejo Mioeléctrico Migratorio , Animales , Colon , Heces , Cobayas , Intestino Grueso
12.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G675-G687, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33624530

RESUMEN

Electrical stimulation of the enteric nervous system (ENS) is an attractive approach to modify gastrointestinal transit. Colonic motor complexes (CMCs) occur with a periodic rhythm, but the ability to elicit a premature CMC depends, at least in part, upon the intrinsic refractory properties of the ENS, which are presently unknown. The objectives of this study were to record myoelectric complexes (MCs, the electrical correlates of CMCs) in the smooth muscle and 1) determine the refractory periods of MCs, 2) inform and evaluate closed-loop stimulation to repetitively evoke MCs, and 3) identify stimulation methods to suppress MC propagation. We dissected the colon from male and female C57BL/6 mice, preserving the integrity of intrinsic circuitry while removing the extrinsic nerves, and measured properties of spontaneous and evoked MCs in vitro. Hexamethonium abolished spontaneous and evoked MCs, confirming the necessary involvement of the ENS for electrically evoked MCs. Electrical stimulation reduced the mean interval between evoked and spontaneous CMCs (24.6 ± 3.5 vs. 70.6 ± 15.7 s, P = 0.0002, n = 7). The absolute refractory period was 4.3 s (95% confidence interval (CI) = 2.8-5.7 s, R2 = 0.7315, n = 8). Electrical stimulation applied during fluid distention-evoked MCs led to an arrest of MC propagation, and following stimulation, MC propagation resumed at an increased velocity (n = 9). The timing parameters of electrical stimulation increased the rate of evoked MCs and the duration of entrainment of MCs, and the refractory period provides insight into timing considerations for designing neuromodulation strategies to treat colonic dysmotility.NEW & NOTEWORTHY Maintained physiological distension of the isolated mouse colon induces rhythmic cyclic myoelectric complexes (MCs). MCs evoked repeatedly by closed-loop electrical stimulation entrain MCs more frequently than spontaneously occurring MCs. Electrical stimulation delivered at the onset of a contraction temporarily suppresses the propagation of MC contractions. Controlled electrical stimulation can either evoke MCs or temporarily delay MCs in the isolated mouse colon, depending on timing relative to ongoing activity.


Asunto(s)
Colon/inervación , Terapia por Estimulación Eléctrica , Sistema Nervioso Entérico/fisiología , Tránsito Gastrointestinal , Músculo Liso/inervación , Complejo Mioeléctrico Migratorio , Animales , Femenino , Masculino , Mecanotransducción Celular , Ratones Endogámicos C57BL , Presión , Periodo Refractario Electrofisiológico , Factores de Tiempo
13.
Am J Physiol Regul Integr Comp Physiol ; 321(3): R317-R327, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34287078

RESUMEN

The dynamic changes in uterine contractility in response to distension are incompletely understood. Rhythmic, propagating contractions of nonpregnant uterine smooth muscle occur in the absence of nerve activity (i.e., myogenic), events that decline during pregnancy and reemerge at parturition. We therefore sought to determine how myogenic contractions of the nonpregnant uterus are affected by distension, which might provide mechanistic clues underlying distension-associated uterine conditions such as preterm birth. Uteri isolated from nulliparous adult female mice in proestrus were video imaged to generate spatiotemporal maps, and myoelectrical activity simultaneously recorded using extracellular suction electrodes. Motility patterns were examined under basal conditions and following ramped intraluminal distension with fluid to 5 and 10 cmH2O. Intraluminal distension caused pressure-dependent changes in the frequency, amplitude, propagation speed, and directionality of uterine contractions, which reversed upon pressure release. Altered burst durations of underlying smooth muscle myoelectric events were concurrently observed, although action potential spike intervals were unchanged. Voltage-gated sodium channel blockade [tetrodotoxin (TTX); 0.6 µM] attenuated both the amplitude of contractions and burst duration of action potentials, whereas all activity was abolished by L-type calcium channel blockade (nifedipine; 1 µM). These data suggest that myogenic motility patterns of the nonpregnant mouse uterus are sensitive to changes in intraluminal pressure and, at high pressures, may be modulated by voltage-gated sodium channel activity. Future studies may investigate whether similar distension-evoked changes occur in the pregnant uterus and the possible pathophysiological role of such activity in the development of preterm birth.


Asunto(s)
Motilidad Gastrointestinal/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Tetrodotoxina/farmacología , Contracción Uterina/efectos de los fármacos , Útero/efectos de los fármacos , Animales , Femenino , Ratones , Contracción Muscular/efectos de los fármacos , Músculo Liso/fisiología , Nacimiento Prematuro/fisiopatología , Contracción Uterina/fisiología , Útero/fisiología
14.
J Neurophysiol ; 123(3): 1236-1246, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31995437

RESUMEN

Techniques to identify and correlate the propagation of electrical signals (like action potentials) along neural networks are well described, using multisite recordings. In these cases, the waveform of action potentials is usually relatively stable and discriminating relevant electrical signals straightforward. However, problems can arise when attempting to identify and correlate the propagation of signals when their waveforms are unstable (e.g., fluctuations in amplitude or time course). This makes correlation of the degree of synchronization and time lag between propagating electrical events across two or more recording sites problematic. Here, we present novel techniques for the determination of the periodicity of electrical signals at individual sites. When recording from two independent sites, we present novel analytical techniques for joint determination of periodicity and time delay. The techniques presented exploit properties of the cross-correlation function, rather than utilizing the time lag at which the cross-correlation function is maximized. The approach allows determination of directionality of the spread of excitation along a neural network based on measurements of the time delay between recording sites. This new method is particularly applicable to analysis of signals in other biological systems that have unstable characteristics in waveform that show dynamic variability.NEW & NOTEWORTHY The determination of frequency(s) at which two sources are synchronized, and relative time delay between them, is a fundamental problem for a wide a range of signal-processing applications. In this methodology paper, we present novel procedures for periodicity estimation for single time series and joint periodicity and time delay estimation for two time series. The methods use properties of the cross-correlation function rather than the cross-correlation function explicitly.


Asunto(s)
Potenciales de Acción/fisiología , Sistema Nervioso Entérico/fisiología , Modelos Teóricos , Red Nerviosa/fisiología , Periodicidad , Procesamiento de Señales Asistido por Computador , Animales , Factores de Tiempo
15.
Am J Physiol Gastrointest Liver Physiol ; 318(2): G244-G253, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31790272

RESUMEN

The mechanisms underlying electrical rhythmicity in smooth muscle of the proximal colon are incompletely understood. Our aim was to identify patterns of electrical rhythmicity in smooth muscle of the proximal region of isolated whole mouse colon and characterize their mechanisms of origin. Two independent extracellular recording electrodes were used to record the patterns of electrical activity in smooth muscle of the proximal region of whole isolated mouse colon. Cross-correlation analysis was used to quantify spatial coordination of these electrical activities over increasing electrode separation distances. Four distinct neurogenic patterns of electrical rhythmicity were identified in smooth muscle of the proximal colon, three of which have not been identified and consisted of bursts of rhythmic action potentials at 1-2 Hz that were abolished by hexamethonium. These neurogenic patterns of electrical rhythmicity in smooth muscle were spatially and temporally synchronized over large separation distances (≥2 mm rosto-caudal axis). Myogenic slow waves could be recorded from the same preparations, but they showed poor spatial and temporal coordination over even short distances (≤1 mm rostro-caudal axis). It is not commonly thought that electrical rhythmicity in gastrointestinal smooth muscle is dependent upon the enteric nervous system. Here, we identified neurogenic patterns of electrical rhythmicity in smooth muscle of the proximal region of isolated mouse colon, which are dependent on synaptic transmission in the enteric nervous system. If the whole colon is studied in vitro, recordings can preserve novel neurogenic patterns of electrical rhythmicity in smooth muscle.NEW & NOTEWORTHY Previously, it has not often been thought that electrical rhythmicity in smooth muscle of the gastrointestinal tract is dependent upon the enteric nervous system. We identified patterns of electrical rhythmicity in smooth muscle of the mouse proximal colon that were abolished by hexamethonium and involved the temporal synchronization of smooth muscle membrane potential over large spatial fields. We reveal different patterns of electrical rhythmicity in colonic smooth muscle that are dependent on the ENS.


Asunto(s)
Colon/inervación , Colon/fisiología , Motilidad Gastrointestinal/fisiología , Músculo Liso/inervación , Músculo Liso/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Colon/efectos de los fármacos , Electrodos Implantados , Fenómenos Electrofisiológicos/fisiología , Sistema Nervioso Entérico/efectos de los fármacos , Sistema Nervioso Entérico/fisiología , Femenino , Bloqueadores Ganglionares/farmacología , Hexametonio/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
16.
Am J Physiol Gastrointest Liver Physiol ; 318(1): G99-G108, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31709829

RESUMEN

Cyclical propagating waves of muscle contraction have been recorded in isolated small intestine or colon, referred to here as motor complexes (MCs). Small intestinal and colonic MCs are neurogenic, occur at similar frequencies, and propagate orally or aborally. Whether they can be coordinated between the different gut regions is unclear. Motor behavior of whole length mouse intestines, from duodenum to terminal rectum, was recorded by intraluminal multisensor catheter. Small intestinal MCs were recorded in 27/30 preparations, and colonic MCs were recorded in all preparations (n = 30) with similar frequencies (0.54 ± 0.03 and 0.58 ± 0.02 counts/min, respectively). MCs propagated across the ileo-colonic junction in 10/30 preparations, forming "full intestine" MCs. The cholinesterase inhibitor physostigmine increased the probability of a full intestine MC but had no significant effect on frequency, speed, or direction. Nitric oxide synthesis blockade by Nω-nitro-l-arginine, after physostigmine, increased MC frequency in small intestine only. Hyoscine-resistant MCs were recorded in the colon but not small intestine (n = 5). All MCs were abolished by hexamethonium (n = 18) or tetrodotoxin (n = 2). The enteric neural mechanism required for motor complexes is present along the full length of both the small and large intestine. In some cases, colonic MCs can be initiated in the distal colon and propagate through the ileo-colonic junction, all the way to duodenum. In conclusion, the ileo-colonic junction provides functional neural continuity for propagating motor activity that originates in the small or large intestine.NEW & NOTEWORTHY Intraluminal manometric recordings revealed motor complexes can propagate antegradely or retrogradely across the ileo-colonic junction, spanning the entire small and large intestines. The fundamental enteric neural mechanism(s) underlying cyclic motor complexes exists throughout the length of the small and large intestine.


Asunto(s)
Colon/inervación , Sistema Nervioso Entérico/fisiología , Intestino Delgado/inervación , Complejo Mioeléctrico Migratorio , Peristaltismo , Animales , Antagonistas Colinérgicos/farmacología , Inhibidores de la Colinesterasa/farmacología , Sistema Nervioso Entérico/efectos de los fármacos , Femenino , Bloqueadores Ganglionares/farmacología , Técnicas In Vitro , Masculino , Ratones Endogámicos C57BL , Complejo Mioeléctrico Migratorio/efectos de los fármacos , Peristaltismo/efectos de los fármacos , Presión , Factores de Tiempo
17.
Cell Tissue Res ; 381(1): 25-34, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32215722

RESUMEN

There is considerable interest in understanding how contents within the gut wall (including microbiome) can activate sensory nerve endings in the gut that project to the central nervous system. However, we have only recently begun to understand the location and characteristics of extrinsic spinal afferent nerve endings that innervate the lower gastrointestinal (GI) tract. Our aim is to identify the nerve endings in the mouse distal colon that arise from single spinal afferent neurons. C57BL/6 mice were anaesthetised and single dorsal root ganglia (DRG) between lumbosacral L6-S1 were injected with dextran biotin. Mice recovered for 7 days. Animals were then euthanized and whole colons removed, fixed and stained for calcitonin-gene-related-peptide (CGRP). Single spinal afferent nerve axons were identified entering the distal colon that ramified along many rows of myenteric ganglia, often giving rise to varicose nerve endings. These same axons bifurcated in the circular muscle giving rise to 4-5 groups of branching-type intramuscular endings, where each group of endings was separated by ~ 370 µm in the rostro-caudal axis and projected 1.2 mm around the circumference. As spinal afferent axons bifurcated, their axons often showed dramatic reductions in diameter. Here, we identified in the distal colon, the characteristics of nerve endings that arise from single colorectal-projecting axons with cell bodies in DRG. These findings suggest that a population of sensory neurons in DRG can potentially detect sensory stimuli simultaneously via different morphological types of endings that lie in both colonic smooth muscle and myenteric ganglia.


Asunto(s)
Colon/inervación , Ganglios Espinales/ultraestructura , Músculo Liso/inervación , Neuronas Aferentes/ultraestructura , Células Receptoras Sensoriales/ultraestructura , Animales , Ratones , Ratones Endogámicos C57BL
18.
J Neurosci ; 38(24): 5507-5522, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29807910

RESUMEN

The enteric nervous system (ENS) contains millions of neurons essential for organization of motor behavior of the intestine. It is well established that the large intestine requires ENS activity to drive propulsive motor behaviors. However, the firing pattern of the ENS underlying propagating neurogenic contractions of the large intestine remains unknown. To identify this, we used high-resolution neuronal imaging with electrophysiology from neighboring smooth muscle. Myoelectric activity underlying propagating neurogenic contractions along murine large intestine [also referred to as colonic migrating motor complexes, (CMMCs)] consisted of prolonged bursts of rhythmic depolarizations at a frequency of ∼2 Hz. Temporal coordination of this activity in the smooth muscle over large spatial fields (∼7 mm, longitudinally) was dependent on the ENS. During quiescent periods between neurogenic contractions, recordings from large populations of enteric neurons, in mice of either sex, revealed ongoing activity. The onset of neurogenic contractions was characterized by the emergence of temporally synchronized activity across large populations of excitatory and inhibitory neurons. This neuronal firing pattern was rhythmic and temporally synchronized across large numbers of ganglia at ∼2 Hz. ENS activation preceded smooth muscle depolarization, indicating rhythmic depolarizations in smooth muscle were controlled by firing of enteric neurons. The cyclical emergence of temporally coordinated firing of large populations of enteric neurons represents a unique neural motor pattern outside the CNS. This is the first direct observation of rhythmic firing in the ENS underlying rhythmic electrical depolarizations in smooth muscle. The pattern of neuronal activity we identified underlies the generation of CMMCs.SIGNIFICANCE STATEMENT How the enteric nervous system (ENS) generates neurogenic contractions of smooth muscle in the gastrointestinal (GI) tract has been a long-standing mystery in vertebrates. It is well known that myogenic pacemaker cells exist in the GI tract [called interstitial cells of Cajal (ICCs)] that generate rhythmic myogenic contractions. However, the mechanisms underlying the generation of rhythmic neurogenic contractions of smooth muscle in the GI tract remains unknown. We developed a high-resolution neuronal imaging method with electrophysiology to address this issue. This technique revealed a novel pattern of rhythmic coordinated neuronal firing in the ENS that has never been identified. Rhythmic neuronal firing in the ENS was found to generate rhythmic neurogenic depolarizations in smooth muscle that underlie contraction of the GI tract.


Asunto(s)
Sistema Nervioso Entérico/fisiología , Músculo Liso/fisiología , Complejo Mioeléctrico Migratorio/fisiología , Animales , Femenino , Intestinos/inervación , Intestinos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroimagen/métodos
19.
J Physiol ; 597(20): 5125-5140, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31444880

RESUMEN

KEY POINTS: Enteric neural circuits enable isolated preparations of guinea-pig distal colon to propel solid and fluid contents by a self-sustaining neuromechanical loop process. In addition there are at least three neural mechanisms which are not directly involved in propulsion: cyclic motor complexes, transient neural events and distal colon migrating motor complexes. In excised guinea-pig colon we simultaneously recorded high resolution manometry, video-imaging of colonic wall movements and electrophysiological recordings from smooth muscle, which enabled us to identify mechanisms that underlie the propulsion of colonic content. The results show that the intermittent propulsion during emptying of the multiple natural faecal pellets is due to the intermittent activation of cyclic motor complexes and this is facilitated by transient neural events. Loss or dysfunction of these activities is likely to underlie disordered gastrointestinal transit. ABSTRACT: It is well known that there are different patterns of electrical activity in smooth muscle cells along different regions of the gastrointestinal tract. These different patterns can be generated by myogenic and/or neurogenic mechanisms. However, what patterns of electrical activity underlie the propulsion of natural faecal content remains unknown, particularly along the large intestine, where large quantities of water are reabsorbed and semi-solid faeces form. In this study, we developed a novel approach which enables for the first time the simultaneous recording of high resolution intraluminal manometry, electrophysiology from the smooth muscle, and spatio-temporal video imaging of colonic wall movements. Using this approach we were able to reveal the nature of enteric neuromuscular transmission and patterns of motor activity responsible for the movement of content. Three distinct neurogenic patterns of electrical activity were recorded even in the absence of propulsive movement. These were the cyclic motor complexes (CMCs), the transient neural events (TNEs) and the slowly propagating distal colonic migrating motor complexes (DCMMCs). We present evidence that the initiation of pellet propulsion is due to a cyclic motor complex (CMC) occurring oral to the pellet. Furthermore, we discovered that the intermittent propulsion of natural faecal pellets is generated by intermittent activation of CMCs; and this propulsion is facilitated by hexamethonium-sensitive TNEs. However, TNEs were not required for propulsion. The findings reveal the patterns of electrical activity that underlie propulsion of natural colonic content and demonstrate that propulsion is generated by a complex interplay between distinct enteric neural circuits.


Asunto(s)
Colon/fisiología , Motilidad Gastrointestinal/fisiología , Contracción Muscular/fisiología , Músculo Liso/fisiología , Potenciales de Acción , Animales , Electromiografía , Femenino , Cobayas , Masculino , Actividad Motora , Complejo Mioeléctrico Migratorio
20.
Am J Physiol Renal Physiol ; 316(6): F1103-F1113, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30908933

RESUMEN

Neural mechanisms of lower urinary tract symptoms in obstruction-induced bladder overactivity remain unclear. We made the first single unit recordings from different types of spinal afferents to determine the effects of bladder outlet obstruction in guinea pigs. A model of gradual bladder outlet obstruction in male guinea pigs was used to produce overactive bladder. Conscious voiding was assessed in metabolic cages, and micturition was recorded in anesthetized guinea pigs in vivo. Single unit extracellular recordings were made ex vivo from spinal afferent nerves in flat sheet preparations of the bladder. Guinea pigs with partially obstructed bladders showed a significant increase in conscious voiding frequency compared with sham-operated guinea pigs. Also, nonvoiding contractions increased significantly in both frequency and amplitude. Although spontaneous firing of low-threshold bladder afferents was increased, their stretch-induced firing was reduced. The proportion of capsaicin-sensitive low-threshold afferents increased in obstructed bladders. Interestingly, spontaneous and stretch-induced firing were both significantly increased in high-threshold afferents after obstruction. In summary, sensory signaling increased in the obstructed bladder during the filling phase. This is largely mediated by low-threshold stretch-sensitive afferents that are activated by increased local nonvoiding contractions. Increased spontaneous firing by high-threshold afferents also contributes. Our findings revealed a complex effect of bladder outlet obstruction on different types of bladder afferents that needs consideration for potential therapeutic targeting of lower urinary tract symptoms in obstruction-induced bladder overactivity.


Asunto(s)
Nervios Espinales/fisiopatología , Obstrucción del Cuello de la Vejiga Urinaria/complicaciones , Vejiga Urinaria Hiperactiva/etiología , Vejiga Urinaria/inervación , Urodinámica , Potenciales de Acción , Vías Aferentes/metabolismo , Vías Aferentes/fisiopatología , Animales , Modelos Animales de Enfermedad , Cobayas , Masculino , Mecanorreceptores/metabolismo , Umbral Sensorial , Nervios Espinales/metabolismo , Obstrucción del Cuello de la Vejiga Urinaria/metabolismo , Obstrucción del Cuello de la Vejiga Urinaria/fisiopatología , Vejiga Urinaria Hiperactiva/metabolismo , Vejiga Urinaria Hiperactiva/fisiopatología , Micción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA