Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(26): 15281-15292, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32546520

RESUMEN

Whether G protein-coupled receptors signal from endosomes to control important pathophysiological processes and are therapeutic targets is uncertain. We report that opioids from the inflamed colon activate δ-opioid receptors (DOPr) in endosomes of nociceptors. Biopsy samples of inflamed colonic mucosa from patients and mice with colitis released opioids that activated DOPr on nociceptors to cause a sustained decrease in excitability. DOPr agonists inhibited mechanically sensitive colonic nociceptors. DOPr endocytosis and endosomal signaling by protein kinase C (PKC) and extracellular signal-regulated kinase (ERK) pathways mediated the sustained inhibitory actions of endogenous opioids and DOPr agonists. DOPr agonists stimulated the recruitment of Gαi/o and ß-arrestin1/2 to endosomes. Analysis of compartmentalized signaling revealed a requirement of DOPr endocytosis for activation of PKC at the plasma membrane and in the cytosol and ERK in the nucleus. We explored a nanoparticle delivery strategy to evaluate whether endosomal DOPr might be a therapeutic target for pain. The DOPr agonist DADLE was coupled to a liposome shell for targeting DOPr-positive nociceptors and incorporated into a mesoporous silica core for release in the acidic and reducing endosomal environment. Nanoparticles activated DOPr at the plasma membrane, were preferentially endocytosed by DOPr-expressing cells, and were delivered to DOPr-positive early endosomes. Nanoparticles caused a long-lasting activation of DOPr in endosomes, which provided sustained inhibition of nociceptor excitability and relief from inflammatory pain. Conversely, nanoparticles containing a DOPr antagonist abolished the sustained inhibitory effects of DADLE. Thus, DOPr in endosomes is an endogenous mechanism and a therapeutic target for relief from chronic inflammatory pain.


Asunto(s)
Leucina Encefalina-2-Alanina/farmacología , Inflamación/complicaciones , Dolor/tratamiento farmacológico , Dolor/metabolismo , Receptores Opioides delta/agonistas , Animales , Colon/inervación , Leucina Encefalina-2-Alanina/administración & dosificación , Células HEK293 , Humanos , Ratones , Nanopartículas/administración & dosificación , Neuronas , Nociceptores/metabolismo , Receptores Opioides delta/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
2.
Am J Physiol Gastrointest Liver Physiol ; 322(2): G201-G222, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34755536

RESUMEN

Bile acids (BAs) are known to be important regulators of intestinal motility and epithelial fluid and electrolyte transport. Over the past two decades, significant advances in identifying and characterizing the receptors, transporters, and ion channels targeted by BAs have led to exciting new insights into the molecular mechanisms involved in these processes. Our appreciation of BAs, their receptors, and BA-modulated ion channels as potential targets for the development of new approaches to treat intestinal motility and transport disorders is increasing. In the current review, we aim to summarize recent advances in our knowledge of the different BA receptors and BA-modulated ion channels present in the gastrointestinal system. We discuss how they regulate motility and epithelial transport, their roles in pathogenesis, and their therapeutic potential in a range of gastrointestinal diseases.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Tracto Gastrointestinal/efectos de los fármacos , Canales Iónicos/efectos de los fármacos , Hígado/efectos de los fármacos , Humanos , Canales Iónicos/agonistas , Receptores de Calcitriol/efectos de los fármacos , Canales de Sodio/efectos de los fármacos
3.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G66-G78, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34755545

RESUMEN

Allosteric modulators (AMs) are molecules that can fine-tune signaling by G protein-coupled receptors (GPCRs). Although they are a promising therapeutic approach for treating a range of disorders, allosteric modulation of GPCRs in the context of the enteric nervous system (ENS) and digestive dysfunction remains largely unexplored. This study examined allosteric modulation of the delta opioid receptor (DOR) in the ENS and assessed the suitability of DOR AMs for the treatment of irritable bowel syndrome (IBS) symptoms using mouse models. The effects of the positive allosteric modulator (PAM) of DOR, BMS-986187, on neurogenic contractions of the mouse colon and on DOR internalization in enteric neurons were quantified. The ability of BMS-986187 to influence colonic motility was assessed both in vitro and in vivo. BMS-986187 displayed DOR-selective PAM-agonist activity and orthosteric agonist probe dependence in the mouse colon. BMS-986187 augmented the inhibitory effects of DOR agonists on neurogenic contractions and enhanced reflex-evoked DOR internalization in myenteric neurons. BMS-986187 significantly increased DOR endocytosis in myenteric neurons in response to the weakly internalizing agonist ARM390. BMS-986187 reduced the generation of complex motor patterns in the isolated intact colon. BMS-986187 reduced fecal output and diarrhea onset in the novel environment stress and castor oil models of IBS symptoms, respectively. DOR PAMs enhance DOR-mediated signaling in the ENS and have potential benefit for the treatment of dysmotility. This study provides proof of concept to support the use of GPCR AMs for the treatment of gastrointestinal motility disorders.NEW & NOTEWORTHY This study assesses the use of positive allosteric modulation as a pharmacological approach to enhance opioid receptor signaling in the enteric nervous system. We demonstrate that selective modulation of endogenous delta opioid receptor signaling can suppress colonic motility without causing constipation. We propose that allosteric modulation of opioid receptor signaling may be a therapeutic strategy to normalize gastrointestinal motility in conditions such as irritable bowel syndrome.


Asunto(s)
Sistema Nervioso Entérico/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Receptores Opioides delta/efectos de los fármacos , Xantonas/farmacología , Analgésicos Opioides/farmacología , Benzamidas/farmacología , Colon/efectos de los fármacos , Sistema Nervioso Entérico/fisiopatología , Motilidad Gastrointestinal/fisiología , Humanos , Receptores Opioides/efectos de los fármacos , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
4.
Lab Invest ; 101(7): 851-864, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33859334

RESUMEN

Endothelial and epithelial cells form physical barriers that modulate the exchange of fluid and molecules. The integrity of these barriers can be influenced by signaling through G protein-coupled receptors (GPCRs) and ion channels. Serotonin (5-HT) is an important vasoactive mediator of tissue edema and inflammation. However, the mechanisms that drive 5-HT-induced plasma extravasation are poorly defined. The Transient Receptor Potential Vanilloid 4 (TRPV4) ion channel is an established enhancer of signaling by GPCRs that promote inflammation and endothelial barrier disruption. Here, we investigated the role of TRPV4 in 5-HT-induced plasma extravasation using pharmacological and genetic approaches. Activation of either TRPV4 or 5-HT receptors promoted significant plasma extravasation in the airway and upper gastrointestinal tract of mice. 5-HT-mediated extravasation was significantly reduced by pharmacological inhibition of the 5-HT2A receptor subtype, or with antagonism or deletion of TRPV4, consistent with functional interaction between 5-HT receptors and TRPV4. Inhibition of receptors for the neuropeptides substance P (SP) or calcitonin gene-related peptide (CGRP) diminished 5-HT-induced plasma extravasation. Supporting studies assessing treatment of HUVEC with 5-HT, CGRP, or SP was associated with ERK phosphorylation. Exposure to the TRPV4 activator GSK1016790A, but not 5-HT, increased intracellular Ca2+ in these cells. However, 5-HT pre-treatment enhanced GSK1016790A-mediated Ca2+ signaling, consistent with sensitization of TRPV4. The functional interaction was further characterized in HEK293 cells expressing 5-HT2A to reveal that TRPV4 enhances the duration of 5-HT-evoked Ca2+ signaling through a PLA2 and PKC-dependent mechanism. In summary, this study demonstrates that TRPV4 contributes to 5-HT2A-induced plasma extravasation in the airways and upper GI tract, with evidence supporting a mechanism of action involving SP and CGRP release.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Pulmón/efectos de los fármacos , Serotonina , Canales Catiónicos TRPV , Tracto Gastrointestinal Superior/efectos de los fármacos , Animales , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Pulmón/citología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Serotonina/genética , Serotonina/metabolismo , Serotonina/farmacología , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Tracto Gastrointestinal Superior/citología , Tracto Gastrointestinal Superior/metabolismo
5.
Lab Invest ; 100(8): 1057-1067, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32341518

RESUMEN

Endothelial barrier disruption is a hallmark of tissue injury, edema, and inflammation. Vascular endothelial cells express the G protein-coupled receptor (GPCR) protease acctivated receptor 1 (PAR1) and the ion channel transient receptor potential vanilloid 4 (TRPV4), and these signaling proteins are known to respond to inflammatory conditions and promote edema through remodeling of cell-cell junctions and modulation of endothelial barriers. It has previously been established that signaling initiated by the related protease activated receptor 2 (PAR2) is enhanced by TRPV4 in sensory neurons and that this functional interaction plays a critical role in the development of neurogenic inflammation and nociception. Here, we investigated the PAR1-TRPV4 axis, to determine if TRPV4 plays a similar role in the control of edema mediated by thrombin-induced signaling. Using Evans Blue permeation and retention as an indication of increased vascular permeability in vivo, we showed that TRPV4 contributes to PAR1-induced vascular hyperpermeability in the airways and upper gastrointestinal tract of mice. TRPV4 contributes to sustained PAR1-induced Ca2+ signaling in recombinant cell systems and to PAR1-dependent endothelial junction remodeling in vitro. This study supports the role of GPCR-TRP channel functional interactions in inflammatory-associated changes to vascular function and indicates that TRPV4 is a signaling effector for multiple PAR family members.


Asunto(s)
Inflamación/genética , Receptor PAR-1/genética , Receptor PAR-2/genética , Transducción de Señal/genética , Canales Catiónicos TRPV/genética , Animales , Calcio/metabolismo , Permeabilidad Capilar/genética , Edema/genética , Edema/metabolismo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Inflamación/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Canales Catiónicos TRPV/metabolismo
6.
Cell Mol Life Sci ; 76(15): 3033-3050, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30904952

RESUMEN

The use of opioid analgesics is severely limited due to the development of intractable constipation, mediated through activation of mu opioid receptors (MOR) expressed by enteric neurons. The related delta opioid receptor (DOR) is an emerging therapeutic target for chronic pain, depression and anxiety. Whether DOR agonists also promote sustained inhibition of colonic transit is unknown. This study examined acute and chronic tolerance to SNC80 and ARM390, which were full and partial DOR agonists in neural pathways controlling colonic motility, respectively. Excitatory pathways developed acute and chronic tolerance to SNC80, whereas only chronic tolerance developed in inhibitory pathways. Both pathways remained functional after acute or chronic ARM390 exposure. Propagating colonic motor patterns were significantly reduced after acute or chronic SNC80 treatment, but not by ARM390 pre-treatment. These findings demonstrate that SNC80 has a prolonged inhibitory effect on propagating colonic motility. ARM390 had no effect on motor patterns and thus may have fewer gastrointestinal side-effects.


Asunto(s)
Analgésicos Opioides/farmacología , Colon/efectos de los fármacos , Tolerancia a Medicamentos , Receptores Opioides delta/metabolismo , Animales , Benzamidas/farmacología , Colon/fisiología , Estimulación Eléctrica , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Contracción Muscular/efectos de los fármacos , Neuronas/metabolismo , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo
7.
Am J Physiol Gastrointest Liver Physiol ; 316(4): G446-G452, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30702900

RESUMEN

G protein-coupled receptors (GPCRs) are essential for the neurogenic control of gastrointestinal (GI) function and are important and emerging therapeutic targets in the gut. Detailed knowledge of both the distribution and functional expression of GPCRs in the enteric nervous system (ENS) is critical toward advancing our understanding of how these receptors contribute to GI function during physiological and pathophysiological states. Equally important, but less well defined, is the complex relationship between receptor expression, ligand binding, signaling, and trafficking within enteric neurons. Neuronal GPCRs are internalized following exposure to agonists and under pathological conditions, such as intestinal inflammation. However, the relationship between the intracellular distribution of GPCRs and their signaling outputs in this setting remains a "black box". This review will briefly summarize current knowledge of agonist-evoked GPCR trafficking and location-specific signaling in the ENS and identifies key areas where future research could be focused. Greater understanding of the cellular and molecular mechanisms involved in regulating GPCR signaling in the ENS will provide new insights into GI function and may open novel avenues for therapeutic targeting of GPCRs for the treatment of digestive disorders.


Asunto(s)
Sistema Nervioso Entérico/fisiología , Enterocitos/fisiología , Transporte de Proteínas , Receptores Acoplados a Proteínas G/metabolismo , Animales , Descubrimiento de Drogas , Humanos , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Transducción de Señal
8.
Am J Physiol Gastrointest Liver Physiol ; 317(2): G79-G89, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31091149

RESUMEN

Endocytosis is a major mechanism through which cellular signaling by G protein-coupled receptors (GPCRs) is terminated. However, recent studies demonstrate that GPCRs are internalized in an active state and continue to signal from within endosomes, resulting in effects on cellular function that are distinct to those arising at the cell surface. Endocytosis inhibitors are commonly used to define the importance of GPCR internalization for physiological and pathophysiological processes. Here, we provide the first detailed examination of the effects of these inhibitors on neurogenic contractions of gastrointestinal smooth muscle, a key preliminary step to evaluate the importance of GPCR endocytosis for gut function. Inhibitors of clathrin-mediated endocytosis (Pitstop2, PS2) or G protein-coupled receptor kinase-2/3-dependent phosphorylation (Takeda compound 101, Cmpd101), significantly reduced GPCR internalization. However, they also attenuated cholinergic contractions through different mechanisms. PS2 abolished contractile responses by colonic muscle to SNC80 and morphine, which strongly and weakly internalize δ-opioid and µ-opioid receptors, respectively. PS2 did not affect the increased myogenic contractile activity following removal of an inhibitory neural influence (tetrodotoxin) but suppressed electrically evoked neurogenic contractions. Ca2+ signaling by myenteric neurons in response to exogenous ATP was unaffected by PS2, suggesting inhibitory actions on neurotransmitter release rather than neurotransmission. In contrast, Cmpd101 attenuated contractions to the cholinergic agonist carbachol, indicating direct effects on smooth muscle. We conclude that, although PS2 and Cmpd101 are effective blockers of GPCR endocytosis in enteric neurons, these inhibitors are unsuitable for the study of neurally mediated gut function due to their inhibitory effects on neuromuscular transmission and smooth muscle contractility.NEW & NOTEWORTHY Internalization of activated G protein-coupled receptors is a major determinant of the type and duration of subsequent downstream signaling events. Inhibitors of endocytosis effectively block opioid receptor internalization in enteric neurons. The clathrin-dependent endocytosis inhibitor Pitstop2 blocks effects of opioids on neurogenic contractions of the colon in an internalization-independent manner. These inhibitors also significantly impact cholinergic neuromuscular transmission. We conclude that these tools are unsuitable for examination of the contribution of neuronal G protein-coupled receptor endocytosis to gastrointestinal motility.


Asunto(s)
Benzamidas/farmacología , Clatrina/metabolismo , Colon , Endocitosis , Músculo Liso , Piridinas/farmacología , Receptores Opioides delta/metabolismo , Sulfonamidas/farmacología , Tiazolidinas/farmacología , Triazoles/farmacología , Analgésicos Opioides/farmacología , Animales , Colon/metabolismo , Colon/patología , Colon/fisiopatología , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Endosomas/metabolismo , Sistema Nervioso Entérico/metabolismo , Motilidad Gastrointestinal/fisiología , Ratones , Músculo Liso/metabolismo , Músculo Liso/patología , Músculo Liso/fisiopatología , Fosforilación/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides mu/metabolismo , Transducción de Señal , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
9.
Am J Physiol Gastrointest Liver Physiol ; 315(4): G544-G559, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29927325

RESUMEN

Endogenous opioids activate opioid receptors (ORs) in the enteric nervous system to control intestinal motility and secretion. The µ-OR mediates the deleterious side effects of opioid analgesics, including constipation, respiratory depression, and addiction. Although the δ-OR (DOR) is a promising target for analgesia, the function and regulation of DOR in the colon are poorly understood. This study provides evidence that endogenous opioids activate DOR in myenteric neurons that may regulate colonic motility. The DOR agonists DADLE, deltorphin II, and SNC80 inhibited electrically evoked contractions and induced neurogenic contractions in the mouse colon. Electrical, chemical, and mechanical stimulation of the colon evoked the release of endogenous opioids, which stimulated endocytosis of DOR in the soma and proximal neurites of myenteric neurons of transgenic mice expressing DOR fused to enhanced green fluorescent protein. In contrast, DOR was not internalized in nerve fibers within the circular muscle. Administration of dextran sulfate sodium induced acute colitis, which was accompanied by DOR endocytosis and an increased density of DOR-positive nerve fibers within the circular muscle. The potency with which SNC80 inhibited neurogenic contractions was significantly enhanced in the inflamed colon. This study demonstrates that DOR-expressing neurons in the mouse colon can be activated by exogenous and endogenous opioids. Activated DOR traffics to endosomes and inhibits neurogenic motility of the colon. DOR signaling is enhanced during intestinal inflammation. This study demonstrates functional expression of DOR by myenteric neurons and supports the therapeutic targeting of DOR in the enteric nervous system. NEW & NOTEWORTHY DOR is activated during physiologically relevant reflex stimulation. Agonist-evoked DOR endocytosis is spatially and temporally regulated. A significant proportion of DOR is internalized in myenteric neurons during inflammation. The relative proportion of all myenteric neurons that expressed DOR and the overlap with the nNOS-positive population are increased in inflammation. DOR-specific innervation of the circular muscle is increased in inflammation, and this is consistent with enhanced responsiveness to the DOR agonist SNC80.


Asunto(s)
Colitis Ulcerosa/metabolismo , Colon/metabolismo , Sistema Nervioso Entérico/metabolismo , Motilidad Gastrointestinal , Receptores Opioides delta/metabolismo , Animales , Benzamidas/farmacología , Colon/fisiología , Colon/fisiopatología , Endocitosis , Leucina Encefalina-2-Alanina/metabolismo , Sistema Nervioso Entérico/fisiología , Sistema Nervioso Entérico/fisiopatología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Muscular , Oligopéptidos/metabolismo , Piperazinas/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética
10.
Am J Physiol Gastrointest Liver Physiol ; 312(1): G85-G102, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27881401

RESUMEN

The Winnie mouse, carrying a missense mutation in Muc2, is a model for chronic intestinal inflammation demonstrating symptoms closely resembling inflammatory bowel disease (IBD). Alterations to the immune environment, morphological structure, and innervation of Winnie mouse colon have been identified; however, analyses of intestinal transit and colonic functions have not been conducted. In this study, we investigated in vivo intestinal transit in radiographic studies and in vitro motility of the isolated colon in organ bath experiments. We compared neuromuscular transmission using conventional intracellular recording between distal colon of Winnie and C57BL/6 mice and smooth muscle contractions using force displacement transducers. Chronic inflammation in Winnie mice was confirmed by detection of lipocalin-2 in fecal samples over 4 wk and gross morphological damage to the colon. Colonic transit was faster in Winnie mice. Motility was altered including decreased frequency and increased speed of colonic migrating motor complexes and increased occurrence of short and fragmented contractions. The mechanisms underlying colon dysfunctions in Winnie mice included inhibition of excitatory and fast inhibitory junction potentials, diminished smooth muscle responses to cholinergic and nitrergic stimulation, and increased number of α-smooth muscle actin-immunoreactive cells. We conclude that diminished excitatory responses occur both prejunctionally and postjunctionally and reduced inhibitory purinergic responses are potentially a prejunctional event, while diminished nitrergic inhibitory responses are probably due to a postjunction mechanism in the Winnie mouse colon. Many of these changes are similar to disturbed motor functions in IBD patients indicating that the Winnie mouse is a model highly representative of human IBD. NEW & NOTEWORTHY: This is the first study to provide analyses of intestinal transit and whole colon motility in an animal model of spontaneous chronic colitis. We found that cholinergic and purinergic neuromuscular transmission, as well as the smooth muscle cell responses to cholinergic and nitrergic stimulation, is altered in the chronically inflamed Winnie mouse colon. The changes to intestinal transit and colonic function we identified in the Winnie mouse are similar to those seen in inflammatory bowel disease patients.


Asunto(s)
Colitis/fisiopatología , Colon/fisiopatología , Motilidad Gastrointestinal/fisiología , Tránsito Gastrointestinal/fisiología , Contracción Muscular/fisiología , Transmisión Sináptica/fisiología , Animales , Colitis/genética , Modelos Animales de Enfermedad , Heces/química , Femenino , Inflamación/genética , Inflamación/fisiopatología , Lipocalina 2/análisis , Masculino , Ratones , Mucina 2/genética , Músculo Liso/fisiopatología , Mutación Missense
11.
Am J Physiol Gastrointest Liver Physiol ; 311(2): G252-66, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27365337

RESUMEN

The µ-opioid receptor (MOR) is a major regulator of gastrointestinal motility and secretion and mediates opiate-induced bowel dysfunction. Although MOR is of physiological and therapeutic importance to gut function, the cellular and subcellular distribution and regulation of MOR within the enteric nervous system are largely undefined. Herein, we defined the neurochemical coding of MOR-expressing neurons in the guinea pig gut and examined the effects of opioids on MOR trafficking and regulation. MOR expression was restricted to subsets of enteric neurons. In the stomach MOR was mainly localized to nitrergic neurons (∼88%), with some overlap with neuropeptide Y (NPY) and no expression by cholinergic neurons. These neurons are likely to have inhibitory motor and secretomotor functions. MOR was restricted to noncholinergic secretomotor neurons (VIP-positive) of the ileum and distal colon submucosal plexus. MOR was mainly detected in nitrergic neurons of the colon (nitric oxide synthase positive, 87%), with some overlap with choline acetyltransferase (ChAT). No expression of MOR by intrinsic sensory neurons was detected. [d-Ala(2), MePhe(4), Gly(ol)(5)]enkephalin (DAMGO), morphiceptin, and loperamide induced MOR endocytosis in myenteric neurons. After stimulation with DAMGO and morphiceptin, MOR recycled, whereas MOR was retained within endosomes following loperamide treatment. Herkinorin or the δ-opioid receptor agonist [d-Ala(2), d-Leu(5)]enkephalin (DADLE) did not evoke MOR endocytosis. In summary, we have identified the neurochemical coding of MOR-positive enteric neurons and have demonstrated differential trafficking of MOR in these neurons in response to established and putative MOR agonists.


Asunto(s)
Colon/inervación , Sistema Nervioso Entérico/metabolismo , Íleon/inervación , Receptores Opioides mu/metabolismo , Estómago/inervación , Analgésicos Opioides/farmacología , Animales , Neuronas Colinérgicas/metabolismo , Endocitosis , Endorfinas/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Sistema Nervioso Entérico/efectos de los fármacos , Cobayas , Loperamida/farmacología , Masculino , Neuronas Motoras/metabolismo , Neuronas Nitrérgicas/metabolismo , Transporte de Proteínas , Receptores Opioides mu/agonistas
12.
Am J Physiol Gastrointest Liver Physiol ; 307(11): G1115-29, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25301186

RESUMEN

Damage to the enteric nervous system (ENS) associated with intestinal inflammation may underlie persistent alterations to gut functions, suggesting that enteric neurons are viable targets for novel therapies. Mesenchymal stem cells (MSCs) offer therapeutic benefits for attenuation of neurodegenerative diseases by homing to areas of inflammation and exhibiting neuroprotective, anti-inflammatory, and immunomodulatory properties. In culture, MSCs release soluble bioactive factors promoting neuronal survival and suppressing inflammation suggesting that MSC-conditioned medium (CM) provides essential factors to repair damaged tissues. We investigated whether MSC and CM treatments administered by enema attenuate 2,4,6-trinitrobenzene-sulfonic acid (TNBS)-induced enteric neuropathy and motility dysfunction in the guinea pig colon. Guinea pigs were randomly assigned to experimental groups and received a single application of TNBS (30 mg/kg) followed by 1 × 10(6) human bone marrow-derived MSCs, 300 µl CM, or 300 µl unconditioned medium 3 h later. After 7 days, the effect of these treatments on enteric neurons was assessed by histological, immunohistochemical, and motility analyses. MSC and CM treatments prevented inflammation-associated weight loss and gross morphological damage in the colon; decreased the quantity of immune infiltrate in the colonic wall (P < 0.01) and at the level of the myenteric ganglia (P < 0.001); prevented loss of myenteric neurons (P < 0.05) and damage to nerve processes, changes in ChAT, and nNOS immunoreactivity (P < 0.05); and alleviated inflammation-induced colonic dysmotility (contraction speed; P < 0.001, contractions/min; P < 0.05). These results provide strong evidence that both MSC and CM treatments can effectively prevent damage to the ENS and alleviate gut dysfunction caused by TNBS-induced colitis.


Asunto(s)
Colitis/inducido químicamente , Colitis/prevención & control , Sistema Nervioso Entérico/patología , Trasplante de Células Madre Mesenquimatosas , Enfermedades del Sistema Nervioso Periférico/prevención & control , Ácido Trinitrobencenosulfónico , Animales , Movimiento Celular/fisiología , Colitis/patología , Colon/patología , Medios de Cultivo Condicionados , Femenino , Motilidad Gastrointestinal , Humanos , Masculino , Ratones , Pérdida de Peso/efectos de los fármacos
13.
J Am Heart Assoc ; 13(3): e033279, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38258657

RESUMEN

BACKGROUND: Gut dysmotility is common after ischemic stroke, but the mechanism underlying this response is unknown. Under homeostasis, gut motility is regulated by the neurons of the enteric nervous system that control contractile/relaxation activity of muscle cells in the gut wall. More recently, studies of gut inflammation revealed interactions of macrophages with enteric neurons are also involved in modulating gut motility. However, whether poststroke gut dysmotility is mediated by direct signaling to the enteric nervous system or indirectly via inflammatory macrophages is unknown. METHODS AND RESULTS: We examined these hypotheses by using a clinically relevant permanent intraluminal midcerebral artery occlusion experimental model of stroke. At 24 hours after stroke, we performed in vivo and ex vivo gut motility assays, flow cytometry, immunofluorescence, and transcriptomic analysis. Stroke-induced gut dysmotility was associated with recruitment of muscularis macrophages into the gastrointestinal tract and redistribution of muscularis macrophages away from myenteric ganglia. The permanent intraluminal midcerebral artery occlusion model caused changes in gene expression in muscularis macrophages consistent with an altered phenotype. While the size of myenteric ganglia after stroke was not altered, myenteric neurons from post-permanent intraluminal midcerebral artery occlusion mice showed a reduction in neuronal nitric oxide synthase expression, and this response was associated with enhanced intestinal smooth muscle contraction ex vivo. Finally, chemical sympathectomy with 6-hydroxydopamine prevented the loss of myenteric neuronal nitric oxide synthase expression and stroke-induced slowed gut transit. CONCLUSIONS: Our findings demonstrate that activation of the sympathetic nervous system after stroke is associated with reduced neuronal nitric oxide synthase expression in myenteric neurons, resulting in impaired smooth muscle relaxation and dysregulation of gut transit.


Asunto(s)
Sistema Nervioso Entérico , Accidente Cerebrovascular , Ratones , Animales , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Sistema Nervioso Entérico/metabolismo , Neuronas/fisiología , Relajación Muscular , Accidente Cerebrovascular/metabolismo
14.
bioRxiv ; 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38260314

RESUMEN

Background: Mechanosensation is an important trigger of physiological processes in the gastrointestinal tract. Aberrant responses to mechanical input are associated with digestive disorders, including visceral hypersensitivity. Transient Receptor Potential Vanilloid 4 (TRPV4) is a mechanosensory ion channel with proposed roles in visceral afferent signaling, intestinal inflammation, and gut motility. While TRPV4 is a potential therapeutic target for digestive disease, current mechanistic understanding of how TRPV4 may influence gut function is limited by inconsistent reports of TRPV4 expression and distribution. Methods: In this study we profiled functional expression of TRPV4 using Ca2+ imaging of wholemount preparations of the mouse, monkey, and human intestine in combination with immunofluorescent labeling for established cellular markers. The involvement of TRPV4 in colonic motility was assessed in vitro using videomapping and contraction assays. Results: The TRPV4 agonist GSK1016790A evoked Ca2+ signaling in muscularis macrophages, enteric glia, and endothelial cells. TRPV4 specificity was confirmed using TRPV4 KO mouse tissue or antagonist pre-treatment. Calcium responses were not detected in other cell types required for neuromuscular signaling including enteric neurons, interstitial cells of Cajal, PDGFRα+ cells, and intestinal smooth muscle. TRPV4 activation led to rapid Ca2+ responses by a subpopulation of glial cells, followed by sustained Ca2+ signaling throughout the enteric glial network. Propagation of these waves was suppressed by inhibition of gap junctions or Ca2+ release from intracellular stores. Coordinated glial signaling in response to GSK1016790A was also disrupted in acute TNBS colitis. The involvement of TRPV4 in the initiation and propagation of colonic motility patterns was examined in vitro. Conclusions: We reveal a previously unappreciated role for TRPV4 in the initiation of distension-evoked colonic motility. These observations provide new insights into the functional role of TRPV4 activation in the gut, with important implications for how TRPV4 may influence critical processes including inflammatory signaling and motility.

15.
Am J Physiol Gastrointest Liver Physiol ; 302(12): G1434-44, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22461022

RESUMEN

Gap junction coupling and neuromuscular transmission to smooth muscle were studied in the first 4 h after preparations were set up in vitro. Intracellular recordings were made from smooth muscle cells of guinea pig ileum. Fast inhibitory junction potentials (IJPs) were small (1.3 ± 1.0 mV) in the first 30 min but increased significantly over the first 120 min to 15.8 ± 0.9 mV (n = 12, P < 0.001). Comparable increases in slow IJPs and excitatory junction potentials were also observed. During the same period, resting membrane potential depolarized from -58.8 ± 1.4 to -47.2 ± 0.4 mV (n = 12, P < 0.001). Input resistance, estimated by intracellular current injection, decreased in parallel (P < 0.05), and dye coupling, measured by intracellular injection of carboxyfluorescein, increased (P < 0.001). Input resistance was higher and dye coupling was less in longitudinal than circular smooth muscle cells. Gap junction blockers [carbenoxolone (100 µM), 18ß-glycyrrhetinic acid (10 µM), and 2-aminoethoxydiphenyl borate (50 µM)] hyperpolarized coupled circular smooth muscle cells, reduced the amplitude of fast and slow IJPs and excitatory junction potentials, increased input resistance, and reduced dye coupling. Local application of ATP (10 mM) mimicked IJPs and showed comparable increases in amplitude over the first 120 min; carbenoxolone and 2-aminoethoxydiphenyl borate significantly reduced ATP-evoked hyperpolarizations in coupled cells. In contrast, synaptic transmission between myenteric neurons was not suppressed during the first 30 min. Gap junction coupling between circular smooth muscle cells in isolated preparations was initially disrupted but recovered over the next 120 min to a steady level. This was associated with potent effects on neuromuscular transmission and responses to exogenous ATP.


Asunto(s)
Uniones Comunicantes/fisiología , Íleon/fisiología , Miocitos del Músculo Liso/fisiología , Unión Neuromuscular/fisiología , Transmisión Sináptica/fisiología , Animales , Compuestos de Boro/farmacología , Carbenoxolona/farmacología , Uniones Comunicantes/efectos de los fármacos , Ácido Glicirretínico/farmacología , Cobayas , Íleon/efectos de los fármacos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Plexo Mientérico/efectos de los fármacos , Plexo Mientérico/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Unión Neuromuscular/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Transmisión Sináptica/efectos de los fármacos
16.
Br J Pharmacol ; 2022 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-36565295

RESUMEN

Gastrointestinal motility is tightly regulated by the enteric nervous system (ENS). Disruption of coordinated enteric nervous system activity can result in dysmotility. Pharmacological treatment options for dysmotility include targeting of G protein-coupled receptors (GPCRs) expressed by neurons of the enteric nervous system. Current GPCR-targeting drugs for motility disorders bind to the highly conserved endogenous ligand-binding site and promote indiscriminate activation or inhibition of the target receptor throughout the body. This can be associated with significant side-effect liability and a loss of physiological tone. Allosteric modulators of GPCRs bind to a distinct site from the endogenous ligand, which is typically less conserved across multiple receptor subtypes and can modulate endogenous ligand signalling. Allosteric modulation of GPCRs that are important for enteric nervous system function may provide effective relief from motility disorders while limiting side-effects. This review will focus on how allosteric modulators of GPCRs may influence gastrointestinal motility, using 5-hydroxytryptamine (5-HT), acetylcholine (ACh) and opioid receptors as examples.

17.
Neurogastroenterol Motil ; 32(2): e13787, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31999404

RESUMEN

Visceral pain is commonly associated with acute or remitting inflammatory bowel disease (IBD). In marked contrast, chronic IBD is often painless, even in the presence of active inflammation. This suggests that inflammation in itself is insufficient to sustain altered nociceptive signaling and raises the possibility that there is an endogenous analgesic system in effect in chronic disease. A new study by Basso et al. published in this issue of Neurogastroenterology & Motility provides additional support for an immune-mediated mechanism that suppresses visceral hypersensitivity. The authors examined visceral pain in the IL-10-piroxicam model of chronic colitis, which differs from other experimental IBD models in that it involves immune suppression. During active inflammation, responses by these mice to graded increases in colorectal distension were equivalent to healthy controls, consistent with normal afferent signaling. However, treatment with a peripherally restricted opioid receptor antagonist resulted in marked visceral hypersensitivity to the same stimuli. This effect was attributed to the production of endogenous opioids by colitogenic CD4+ T cells present in the mucosa. This mini-review provides a brief overview of analgesia by immune-derived opioids under inflammatory conditions and highlights how the work of Basso et al. contributes to this area of research. Potential pharmacological approaches to harness or mimic this system are provided. These strategies may prove to be an effective means through which targeted and sustained relief of IBD pain may be achieved.


Asunto(s)
Inflamación , Péptidos Opioides/metabolismo , Dolor , Animales , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Dolor/inmunología , Dolor/metabolismo
18.
Cell Mol Gastroenterol Hepatol ; 9(3): 465-483, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31759144

RESUMEN

BACKGROUND & AIMS: Functional interactions between the mu opioid receptor (MOR) and delta opioid receptor (DOR) represent a potential target for novel analgesics and may drive the effects of the clinically approved drug eluxadoline for the treatment of diarrhea-predominant irritable bowel syndrome. Although the enteric nervous system (ENS) is a likely site of action, the coexpression and potential interaction between MOR and DOR in the ENS are largely undefined. In the present study, we have characterized the distribution of MOR in the mouse ENS and examined MOR-DOR interactions by using pharmacologic and cell biology techniques. METHODS: MOR and DOR expression was defined by using MORmCherry and MORmCherry-DOR-eGFP knockin mice. MOR-DOR interactions were assessed by using DOR-eGFP internalization assays and by pharmacologic analysis of neurogenic contractions of the colon. RESULTS: Although MOR was expressed by approximately half of all myenteric neurons, MOR-positive submucosal neurons were rarely observed. There was extensive overlap between MOR and DOR in both excitatory and inhibitory pathways involved in the coordination of intestinal motility. MOR and DOR can functionally interact, as shown through heterologous desensitization of MOR-dependent responses by DOR agonists. Functional evidence suggests that MOR and DOR may not exist as heteromers in the ENS. Pharmacologic studies show no evidence of cooperativity between MOR and DOR. DOR internalizes independently of MOR in myenteric neurons, and MOR-evoked contractions are unaffected by the sequestration of DOR. CONCLUSIONS: Collectively, these findings demonstrate that although MOR and DOR are coexpressed in the ENS and functionally interact, they are unlikely to exist as heteromers under physiological conditions.


Asunto(s)
Analgésicos Opioides/farmacología , Colon/metabolismo , Sistema Nervioso Entérico/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Animales , Benzamidas/farmacología , Células CHO , Cricetulus , Sistema Nervioso Entérico/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Motilidad Gastrointestinal/fisiología , Técnicas de Sustitución del Gen , Genes Reporteros/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Proteínas Luminiscentes/genética , Ratones , Morfina/farmacología , Piperazinas/farmacología , Piperidinas/farmacología , Multimerización de Proteína/fisiología , Receptores Opioides delta/agonistas , Receptores Opioides delta/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína Fluorescente Roja
19.
Br J Pharmacol ; 173(24): 3502-3521, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27714760

RESUMEN

BACKGROUND AND PURPOSE: Oxaliplatin is a platinum-based chemotherapeutic drug used as a first-line therapy for colorectal cancer. However, its use is associated with severe gastrointestinal side-effects resulting in dose limitations and/or cessation of treatment. In this study, we tested whether oxidative stress, caused by chronic oxaliplatin treatment, induces enteric neuronal damage and colonic dysmotility. EXPERIMENTAL APPROACH: Oxaliplatin (3 mg·kg-1 per day) was administered in vivo to Balb/c mice intraperitoneally three times a week. The distal colon was collected at day 14 of treatment. Immunohistochemistry was performed in wholemount preparations of submucosal and myenteric ganglia. Neuromuscular transmission was studied by intracellular electrophysiology. Circular muscle tone was studied by force transducers. Colon propulsive activity studied in organ bath experiments and faeces were collected to measure water content. KEY RESULTS: Chronic in vivo oxaliplatin treatment resulted in increased formation of reactive oxygen species (O2 -), nitration of proteins, mitochondrial membrane depolarisation resulting in the release of cytochrome c, loss of neurons, increased inducible NOS expression and apoptosis in both the submucosal and myenteric plexuses of the colon. Oxaliplatin treatment enhanced NO-mediated inhibitory junction potentials and altered the response of circular muscles to the NO donor, sodium nitroprusside. It also reduced the frequency of colonic migrating motor complexes and decreased circular muscle tone, effects reversed by the NO synthase inhibitor, Nω-Nitro-L-arginine. CONCLUSION AND IMPLICATIONS: Our study is the first to provide evidence that oxidative stress is a key player in enteric neuropathy and colonic dysmotility leading to symptoms of chronic constipation observed in oxaliplatin-treated mice.


Asunto(s)
Antineoplásicos/farmacología , Colon/efectos de los fármacos , Seudoobstrucción Intestinal/inducido químicamente , Compuestos Organoplatinos/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Antineoplásicos/administración & dosificación , Colon/metabolismo , Colon/patología , Seudoobstrucción Intestinal/patología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino , Superóxidos/metabolismo
20.
Front Physiol ; 5: 319, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25202280

RESUMEN

Excitatory and inhibitory junction potentials of circular smooth muscle cells in guinea pig ileum and colon are suppressed 30-90 min after setting up in vitro preparations. We have previously shown this "unresponsive" period is associated with a transient loss of dye coupling between smooth muscle cells, which subsequently recovers over the ensuing 30-90 min; junction potentials recover in parallel with dye coupling (Carbone et al., 2012). Here, we investigated which components of dissection trigger the initial loss of coupling. Intracellular recordings were made from circular muscle cells of guinea pig ileum with micropipettes containing 5% carboxyfluorescein. After allowing 90-120 min for junction potentials to reach full amplitude, we re-cut all 4 edges of the preparation more than 1 mm from the recording sites. This caused a reduction in the amplitude of IJPs from 17.2 ± 0.7 mV to 9.5 ± 1.5 mV (P < 0.001, n = 12) and a significant reduction in dye coupling. Both recovered within 60 min. We repeated this experiment (n = 4), recording both 1 and 4 mm from the cut edge: both sites were equally affected by re-cutting the sides of the preparation. Equilibrated preparations were stretched to 150% of their original length, this had no significant effect on junction potentials or dye coupling. Setting up preparations in low calcium solution did not prevent the initial suppression of IJPs and dye coupling. Application of 3 µM indomethacin (n = 3), 10 µM ketotifen (n = 4) or 10 µM forskolin during dissection did not prevent the suppression of IJPs and dye coupling. If dissection damage was reduced, by leaving the mucosa and submucosa attached to the circular muscle, IJPs showed less initial suppression than in preparations where the layers were dissected off. We conclude that physical damage to the gut wall triggers loss of gap junction coupling and neuromuscular transmission, this is not due to stretch, influx of calcium ions, release of prostaglandins or mast cell degranulation. The mechanisms underlying this potent effect remain to be determined.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA