Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 81
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
J Cell Sci ; 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39219476

RÉSUMÉ

The enteric nervous system (ENS) consists of an extensive network of neurons and glial cells embedded within the wall of the gastrointestinal (GI) tract. Alterations in neuronal distribution and function are strongly associated with GI dysfunction. Current methods for assessing neuronal distribution suffer from undersampling, partly due to challenges associated with imaging and analyzing large tissue areas, and operator bias due to manual analysis. We present the Gut Analysis Toolbox (GAT), an image analysis tool designed for characterization of enteric neurons and their neurochemical coding using 2D images of GI wholemount preparations. It is developed in Fiji, has a user-friendly interface and offers rapid and accurate segmentation via custom deep learning (DL) based cell segmentation models developed using StarDist, and a ganglion segmentation model in deepImageJ. We use proximal neighbor-based spatial analysis to reveal differences in cellular distribution across gut regions using a public dataset. In summary, GAT provides an easy-to-use toolbox to streamline routine image analysis tasks in ENS research. GAT enhances throughput allowing unbiased analysis of larger tissue areas, multiple neuronal markers and numerous samples rapidly.

2.
bioRxiv ; 2024 Jan 11.
Article de Anglais | MEDLINE | ID: mdl-38260314

RÉSUMÉ

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.

3.
J Am Heart Assoc ; 13(3): e033279, 2024 Feb 06.
Article de Anglais | MEDLINE | ID: mdl-38258657

RÉSUMÉ

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.


Sujet(s)
Système nerveux entérique , Accident vasculaire cérébral , Souris , Animaux , Nitric oxide synthase type I/génétique , Nitric oxide synthase type I/métabolisme , Système nerveux entérique/métabolisme , Neurones/physiologie , Relâchement musculaire , Accident vasculaire cérébral/métabolisme
4.
Proc Natl Acad Sci U S A ; 120(22): e2220979120, 2023 05 30.
Article de Anglais | MEDLINE | ID: mdl-37216510

RÉSUMÉ

The hypothesis that sustained G protein-coupled receptor (GPCR) signaling from endosomes mediates pain is based on studies with endocytosis inhibitors and lipid-conjugated or nanoparticle-encapsulated antagonists targeted to endosomes. GPCR antagonists that reverse sustained endosomal signaling and nociception are needed. However, the criteria for rational design of such compounds are ill-defined. Moreover, the role of natural GPCR variants, which exhibit aberrant signaling and endosomal trafficking, in maintaining pain is unknown. Herein, substance P (SP) was found to evoke clathrin-mediated assembly of endosomal signaling complexes comprising neurokinin 1 receptor (NK1R), Gαq/i, and ßarrestin-2. Whereas the FDA-approved NK1R antagonist aprepitant induced a transient disruption of endosomal signals, analogs of netupitant designed to penetrate membranes and persist in acidic endosomes through altered lipophilicity and pKa caused sustained inhibition of endosomal signals. When injected intrathecally to target spinal NK1R+ve neurons in knockin mice expressing human NK1R, aprepitant transiently inhibited nociceptive responses to intraplantar injection of capsaicin. Conversely, netupitant analogs had more potent, efficacious, and sustained antinociceptive effects. Mice expressing C-terminally truncated human NK1R, corresponding to a natural variant with aberrant signaling and trafficking, displayed attenuated SP-evoked excitation of spinal neurons and blunted nociceptive responses to SP. Thus, sustained antagonism of the NK1R in endosomes correlates with long-lasting antinociception, and domains within the C-terminus of the NK1R are necessary for the full pronociceptive actions of SP. The results support the hypothesis that endosomal signaling of GPCRs mediates nociception and provides insight into strategies for antagonizing GPCRs in intracellular locations for the treatment of diverse diseases.


Sujet(s)
Endosomes , Récepteur de la neurokinine 1 , Souris , Humains , Animaux , Récepteur de la neurokinine 1/génétique , Aprépitant/pharmacologie , Substance P/pharmacologie , Récepteurs couplés aux protéines G , Douleur/traitement médicamenteux
5.
Br J Pharmacol ; 2022 Dec 24.
Article de Anglais | MEDLINE | ID: mdl-36565295

RÉSUMÉ

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.

6.
Biomaterials ; 285: 121536, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35533442

RÉSUMÉ

Soft polymer nanoparticles designed to disassemble and release an antagonist of the neurokinin 1 receptor (NK1R) in endosomes provide efficacious yet transient relief from chronic pain. These micellar nanoparticles are unstable and rapidly release cargo, which may limit the duration of analgesia. We examined the efficacy of stable star polymer nanostars containing the NK1R antagonist aprepitant-amine for the treatment of chronic pain in mice. Nanostars continually released cargo for 24 h, trafficked through the endosomal system, and disrupted NK1R endosomal signaling. After intrathecal injection, nanostars accumulated in endosomes of spinal neurons. Nanostar-aprepitant reversed mechanical, thermal and cold allodynia and normalized nociceptive behavior more efficaciously than free aprepitant in preclinical models of neuropathic and inflammatory pain. Analgesia was maintained for >10 h. The sustained endosomal delivery of antagonists from slow-release nanostars provides effective and long-lasting reversal of chronic pain.


Sujet(s)
Douleur chronique , Antagonistes du récepteur de la neurokinine-1 , Animaux , Aprépitant/pharmacologie , Aprépitant/usage thérapeutique , Douleur chronique/traitement médicamenteux , Endosomes , Souris , Antagonistes du récepteur de la neurokinine-1/pharmacologie , Antagonistes du récepteur de la neurokinine-1/usage thérapeutique , Polymères/pharmacologie
7.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article de Anglais | MEDLINE | ID: mdl-35110404

RÉSUMÉ

G protein-coupled receptors (GPCRs) regulate many pathophysiological processes and are major therapeutic targets. The impact of disease on the subcellular distribution and function of GPCRs is poorly understood. We investigated trafficking and signaling of protease-activated receptor 2 (PAR2) in colitis. To localize PAR2 and assess redistribution during disease, we generated knockin mice expressing PAR2 fused to monomeric ultrastable green fluorescent protein (muGFP). PAR2-muGFP signaled and trafficked normally. PAR2 messenger RNA was detected at similar levels in Par2-mugfp and wild-type mice. Immunostaining with a GFP antibody and RNAScope in situ hybridization using F2rl1 (PAR2) and Gfp probes revealed that PAR2-muGFP was expressed in epithelial cells of the small and large intestine and in subsets of enteric and dorsal root ganglia neurons. In healthy mice, PAR2-muGFP was prominently localized to the basolateral membrane of colonocytes. In mice with colitis, PAR2-muGFP was depleted from the plasma membrane of colonocytes and redistributed to early endosomes, consistent with generation of proinflammatory proteases that activate PAR2 PAR2 agonists stimulated endocytosis of PAR2 and recruitment of Gαq, Gαi, and ß-arrestin to early endosomes of T84 colon carcinoma cells. PAR2 agonists increased paracellular permeability of colonic epithelial cells, induced colonic inflammation and hyperalgesia in mice, and stimulated proinflammatory cytokine release from segments of human colon. Knockdown of dynamin-2 (Dnm2), the major colonocyte isoform, and Dnm inhibition attenuated PAR2 endocytosis, signaling complex assembly and colonic inflammation and hyperalgesia. Thus, PAR2 endocytosis sustains protease-evoked inflammation and nociception and PAR2 in endosomes is a potential therapeutic target for colitis.


Sujet(s)
Côlon/métabolisme , Endocytose/physiologie , Colorants fluorescents/métabolisme , Inflammation/métabolisme , Douleur/métabolisme , Récepteur de type PAR-2/métabolisme , Animaux , Arrestines/métabolisme , Membrane cellulaire/métabolisme , Endosomes/métabolisme , Femelle , Ganglions sensitifs des nerfs spinaux/métabolisme , Humains , Souris , Souris de lignée C57BL , Nociception/physiologie , Transduction du signal/physiologie
8.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G66-G78, 2022 01 01.
Article de Anglais | MEDLINE | ID: mdl-34755545

RÉSUMÉ

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.


Sujet(s)
Système nerveux entérique/effets des médicaments et des substances chimiques , Motilité gastrointestinale/effets des médicaments et des substances chimiques , Récepteur delta/effets des médicaments et des substances chimiques , Xanthones/pharmacologie , Analgésiques morphiniques/pharmacologie , Benzamides/pharmacologie , Côlon/effets des médicaments et des substances chimiques , Système nerveux entérique/physiopathologie , Motilité gastrointestinale/physiologie , Humains , Récepteurs aux opioïdes/effets des médicaments et des substances chimiques , Récepteur delta/agonistes , Récepteur mu/agonistes , Récepteur mu/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
9.
Am J Physiol Gastrointest Liver Physiol ; 322(2): G201-G222, 2022 02 01.
Article de Anglais | MEDLINE | ID: mdl-34755536

RÉSUMÉ

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.


Sujet(s)
Acides et sels biliaires/métabolisme , Tube digestif/effets des médicaments et des substances chimiques , Canaux ioniques/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Humains , Canaux ioniques/agonistes , Récepteur calcitriol/effets des médicaments et des substances chimiques , Canaux sodiques/effets des médicaments et des substances chimiques
10.
Neurosci Lett ; 770: 136377, 2022 01 23.
Article de Anglais | MEDLINE | ID: mdl-34856355

RÉSUMÉ

Transient Receptor Potential Vanilloid 4 (TRPV4) is a polymodal, non-selective cation channel that detects thermal, mechanical, and environmental cues and contributes to a range of diverse physiological processes. The effects of chronic TRPV4 stimulation and gain-of-function genetic mutations suggest that TRPV4 may also be a valuable therapeutic target for pathophysiological events including neurogenic inflammation, peripheral neuropathies, and impaired wound healing. There has been significant interest in defining how and where TRPV4 may promote inflammation and pain. Endogenous stimuli such as osmotic stress and lipid binding are established TRPV4 activators. The TRP channel family is also well-known to be controlled by 'receptor-operated' pathways. For example, G protein-coupled receptors (GPCRs) expressed by primary afferent neurons or other cells in inflammatory pathways utilize TRPV4 as an effector protein to amplify nociceptive and inflammatory signaling. Contributing to disorders including arthritis, neuropathies, and pulmonary edema, GPCRs such as the protease-activated receptor PAR2 mediate activation of kinase signaling cascades to increase TRPV4 phosphorylation, resulting in sensitization and enhanced neuronal excitability. Phospholipase activity also leads to production of polyunsaturated fatty acid lipid mediators that directly activate TRPV4. Consistent with the contribution of TRPV4 to disease, pharmacological inhibition or genetic ablation of TRPV4 can diminish receptor-mediated inflammatory events. This review outlines how receptor-mediated signaling is a major endogenous driver of TRPV4 gating and discusses key signaling pathways and emerging TRPV4 modulators such as the mechanosensitive Piezo1 ion channel. A collective understanding of how endogenous stimuli can influence TRPV4 function is critical for future therapeutic endeavors to modulate this channel.


Sujet(s)
Inflammation neurogénique/métabolisme , Nociception , Canaux cationiques TRPV/métabolisme , Animaux , Humains , Transduction du signal
11.
Lab Invest ; 101(7): 851-864, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33859334

RÉSUMÉ

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.


Sujet(s)
Perméabilité capillaire/effets des médicaments et des substances chimiques , Poumon/effets des médicaments et des substances chimiques , Sérotonine , Canaux cationiques TRPV , Tube digestif supérieur/effets des médicaments et des substances chimiques , Animaux , Cellules HEK293 , Cellules endothéliales de la veine ombilicale humaine , Humains , Poumon/cytologie , Poumon/métabolisme , Mâle , Souris , Souris de lignée C57BL , Sérotonine/génétique , Sérotonine/métabolisme , Sérotonine/pharmacologie , Canaux cationiques TRPV/génétique , Canaux cationiques TRPV/métabolisme , Tube digestif supérieur/cytologie , Tube digestif supérieur/métabolisme
12.
J Biol Chem ; 296: 100345, 2021.
Article de Anglais | MEDLINE | ID: mdl-33515548

RÉSUMÉ

G-protein-coupled receptors (GPCRs) are traditionally known for signaling at the plasma membrane, but they can also signal from endosomes after internalization to control important pathophysiological processes. In spinal neurons, sustained endosomal signaling of the neurokinin 1 receptor (NK1R) mediates nociception, as demonstrated in models of acute and neuropathic pain. An NK1R antagonist, Spantide I (Span), conjugated to cholestanol (Span-Chol), accumulates in endosomes, inhibits endosomal NK1R signaling, and causes prolonged antinociception. However, the extent to which the Chol-anchor influences long-term location and activity is poorly understood. Herein, we used fluorescent correlation spectroscopy and targeted biosensors to characterize Span-Chol over time. The Chol-anchor increased local concentration of probe at the plasma membrane. Over time we observed an increase in NK1R-binding affinity and more potent inhibition of NK1R-mediated calcium signaling. Span-Chol, but not Span, caused a persistent decrease in NK1R recruitment of ß-arrestin and receptor internalization to early endosomes. Using targeted biosensors, we mapped the relative inhibition of NK1R signaling as the receptor moved into the cell. Span selectively inhibited cell surface signaling, whereas Span-Chol partitioned into endosomal membranes and blocked endosomal signaling. In a preclinical model of pain, Span-Chol caused prolonged antinociception (>9 h), which is attributable to a three-pronged mechanism of action: increased local concentration at membranes, a prolonged decrease in NK1R endocytosis, and persistent inhibition of signaling from endosomes. Identifying the mechanisms that contribute to the increased preclinical efficacy of lipid-anchored NK1R antagonists is an important step toward understanding how we can effectively target intracellular GPCRs in disease.


Sujet(s)
Analgésiques/pharmacologie , Dihydrocholestérol/pharmacologie , Antagonistes du récepteur de la neurokinine-1/pharmacologie , Douleur/traitement médicamenteux , Substance P/analogues et dérivés , Analgésiques/composition chimique , Analgésiques/usage thérapeutique , Animaux , Membrane cellulaire/effets des médicaments et des substances chimiques , Membrane cellulaire/métabolisme , Dihydrocholestérol/analogues et dérivés , Dihydrocholestérol/usage thérapeutique , Endosomes/effets des médicaments et des substances chimiques , Endosomes/métabolisme , Cellules HEK293 , Humains , Mâle , Souris de lignée C57BL , Antagonistes du récepteur de la neurokinine-1/composition chimique , Antagonistes du récepteur de la neurokinine-1/usage thérapeutique , Douleur/métabolisme , Gestion de la douleur , Substance P/composition chimique , Substance P/pharmacologie , Substance P/usage thérapeutique
13.
Front Immunol ; 12: 828115, 2021.
Article de Anglais | MEDLINE | ID: mdl-35126384

RÉSUMÉ

Transient receptor potential vanilloid 4 (TRPV4) is a non-selective mechanosensitive ion channel expressed by various macrophage populations. Recent reports have characterized the role of TRPV4 in shaping the activity and phenotype of macrophages to influence the innate immune response to pathogen exposure and inflammation. TRPV4 has been studied extensively in the context of inflammation and inflammatory pain. Although TRPV4 activity has been generally described as pro-inflammatory, emerging evidence suggests a more complex role where this channel may also contribute to anti-inflammatory activities. However, detailed understanding of how TRPV4 may influence the initiation, maintenance, and resolution of inflammatory disease remains limited. This review highlights recent insights into the cellular processes through which TRPV4 contributes to pathological conditions and immune processes, with a focus on macrophage biology. The potential use of high-throughput and omics methods as an unbiased approach for studying the functional outcomes of TRPV4 activation is also discussed.


Sujet(s)
Régulation de l'expression des gènes , Macrophages/immunologie , Macrophages/métabolisme , Transduction du signal , Canaux cationiques TRPV/génétique , Canaux cationiques TRPV/métabolisme , Animaux , Protéines de transport , Prise en charge de la maladie , Prédisposition aux maladies , Métabolisme énergétique , Humains , Ligands , Activation des macrophages/génétique , Activation des macrophages/immunologie , Mécanotransduction cellulaire , Thérapie moléculaire ciblée , Liaison aux protéines
14.
Future Med Chem ; 13(1): 63-90, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-33319586

RÉSUMÉ

G protein-coupled receptors (GPCRs) are essential signaling proteins and tractable therapeutic targets. To develop new drug candidates, GPCR drug discovery programs require versatile, sensitive pharmacological tools for ligand binding and compound screening. With the availability of new imaging modalities and proximity-based ligand binding technologies, fluorescent ligands offer many advantages and are increasingly being used, yet labeling small molecules remains considerably more challenging relative to peptides. Focusing on recent fluorescent small molecule studies for family A GPCRs, this review addresses some of the key challenges, synthesis approaches and structure-activity relationship considerations, and discusses advantages of using high-resolution GPCR structures to inform conjugation strategies. While no single approach guarantees successful labeling without loss of affinity or selectivity, the choice of fluorophore, linker type and site of attachment have proved to be critical factors that can significantly affect their utility in drug discovery programs, and as discussed, can sometimes lead to very unexpected results.


Sujet(s)
Buprénorphine/composition chimique , Acides gras/composition chimique , Colorants fluorescents/composition chimique , Morphine/composition chimique , Ocytocine/composition chimique , Récepteurs couplés aux protéines G/composition chimique , Récepteurs couplés aux protéines G/métabolisme , Séquence d'acides aminés , Sites de fixation , Buprénorphine/métabolisme , Cristallisation , Évaluation préclinique de médicament , Acides gras/métabolisme , Transfert d'énergie par résonance de fluorescence , Humains , Ligands , Morphine/métabolisme , Imagerie optique , Ocytocine/métabolisme , Liaison aux protéines , Conformation des protéines , Relation structure-activité
15.
Front Immunol ; 11: 582358, 2020.
Article de Anglais | MEDLINE | ID: mdl-33154754

RÉSUMÉ

γδ T cells play an essential role in the immune response to many pathogens, including Plasmodium. However, long-lasting effects of infection on the γδ T cell population still remain inadequately understood. This study focused on assessing molecular and functional changes that persist in the γδ T cell population following resolution of malaria infection. We investigated transcriptional changes and memory-like functional capacity of malaria pre-exposed γδ T cells using a Plasmodiumchabaudi infection model. We show that multiple genes associated with effector function (chemokines, cytokines and cytotoxicity) and antigen-presentation were upregulated in P. chabaudi-exposed γδ T cells compared to γδ T cells from naïve mice. This transcriptional profile was positively correlated with profiles observed in conventional memory CD8+ T cells and was accompanied by enhanced reactivation upon secondary encounter with Plasmodium-infected red blood cells in vitro. Collectively our data demonstrate that Plasmodium exposure result in "memory-like imprints" in the γδ T cell population and also promotes γδ T cells that can support antigen-presentation during subsequent infections.


Sujet(s)
Paludisme/immunologie , Plasmodium chabaudi/physiologie , Lymphocytes T/immunologie , Animaux , Présentation d'antigène , Cellules cultivées , Modèles animaux de maladie humaine , Femelle , Régulation de l'expression des gènes , Humains , Mémoire immunologique , Activation des lymphocytes , Souris , Souris de lignée C57BL , Récepteur lymphocytaire T antigène, gamma-delta/métabolisme
16.
Cell Rep ; 32(10): 108100, 2020 09 08.
Article de Anglais | MEDLINE | ID: mdl-32905782

RÉSUMÉ

Mechanisms resulting in abdominal pain include altered neuro-immune interactions in the gastrointestinal tract, but the signaling processes that link immune activation with visceral hypersensitivity are unresolved. We hypothesized that enteric glia link the neural and immune systems of the gut and that communication between enteric glia and immune cells modulates the development of visceral hypersensitivity. To this end, we manipulated a major mechanism of glial intercellular communication that requires connexin-43 and assessed the effects on acute and chronic inflammation, visceral hypersensitivity, and immune responses. Deleting connexin-43 in glia protected against the development of visceral hypersensitivity following chronic colitis. Mechanistically, the protective effects of glial manipulation were mediated by disrupting the glial-mediated activation of macrophages through the macrophage colony-stimulating factor. Collectively, our data identified enteric glia as a critical link between gastrointestinal neural and immune systems that could be harnessed by therapies to ameliorate abdominal pain.


Sujet(s)
Inflammation/métabolisme , Macrophages/métabolisme , Névroglie/métabolisme , Animaux , Humains , Souris , Phénotype
17.
Proc Natl Acad Sci U S A ; 117(26): 15281-15292, 2020 06 30.
Article de Anglais | MEDLINE | ID: mdl-32546520

RÉSUMÉ

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.


Sujet(s)
2-Alanine-leucine-enképhaline/pharmacologie , Inflammation/complications , Douleur/traitement médicamenteux , Douleur/métabolisme , Récepteur delta/agonistes , Animaux , Côlon/innervation , 2-Alanine-leucine-enképhaline/administration et posologie , Cellules HEK293 , Humains , Souris , Nanoparticules/administration et posologie , Neurones , Nocicepteurs/métabolisme , Récepteur delta/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/physiologie
18.
Lab Invest ; 100(8): 1057-1067, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32341518

RÉSUMÉ

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.


Sujet(s)
Inflammation/génétique , Récepteur de type PAR-1/génétique , Récepteur de type PAR-2/génétique , Transduction du signal/génétique , Canaux cationiques TRPV/génétique , Animaux , Calcium/métabolisme , Perméabilité capillaire/génétique , Oedème/génétique , Oedème/métabolisme , Cellules HEK293 , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/physiologie , Humains , Inflammation/métabolisme , Souris de lignée C57BL , Souris knockout , Récepteur de type PAR-1/métabolisme , Récepteur de type PAR-2/métabolisme , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Canaux cationiques TRPV/métabolisme
19.
Neurogastroenterol Motil ; 32(2): e13787, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31999404

RÉSUMÉ

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.


Sujet(s)
Inflammation , Peptides opioïdes/métabolisme , Douleur , Animaux , Humains , Inflammation/immunologie , Inflammation/métabolisme , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/métabolisme , Douleur/immunologie , Douleur/métabolisme
20.
Cell Mol Gastroenterol Hepatol ; 9(3): 465-483, 2020.
Article de Anglais | MEDLINE | ID: mdl-31759144

RÉSUMÉ

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.


Sujet(s)
Analgésiques morphiniques/pharmacologie , Côlon/métabolisme , Système nerveux entérique/métabolisme , Récepteur delta/métabolisme , Récepteur mu/métabolisme , Animaux , Benzamides/pharmacologie , Cellules CHO , Cricetulus , Système nerveux entérique/effets des médicaments et des substances chimiques , Motilité gastrointestinale/effets des médicaments et des substances chimiques , Motilité gastrointestinale/physiologie , Techniques de knock-in de gènes , Gènes rapporteurs/génétique , Protéines à fluorescence verte/génétique , Humains , Protéines luminescentes/génétique , Souris , Morphine/pharmacologie , Pipérazines/pharmacologie , Pipéridines/pharmacologie , Multimérisation de protéines/physiologie , Récepteur delta/agonistes , Récepteur delta/génétique , Récepteur mu/agonistes , Récepteur mu/génétique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme ,
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE