Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 35
1.
Molecules ; 27(21)2022 Nov 02.
Article En | MEDLINE | ID: mdl-36364288

Chemotherapeutic agent-induced nausea and vomiting are the severe adverse effects that are induced by their stimulations on the peripheral and/or central emetic nerve pathways. Even though ginger has been widely used as an herbal medicine to treat emesis, mechanisms underlying its neuronal actions are still less clear. The present study aimed to determine the chemotherapeutic agent vincristine-induced effect on gastroesophageal vagal afferent nerve endings and the potential inhibitory role of ginger constituent 6-shogaol on such response. Two-photon neuron imaging studies were performed in ex vivo gastroesophageal-vagal preparations from Pirt-GCaMP6 transgenic mice. Vincristine was applied to the gastroesophageal vagal afferent nerve endings, and the evoked calcium influxes in their intact nodose ganglion neuron somas were recorded. The responsive nodose neuron population was first characterized, and the inhibitory effects of 5-HT3 antagonist palonosetron, TRPA1 antagonist HC-030031, and ginger constituent 6-shogaol were then determined. Vincristine application at gastroesophageal vagal afferent nerve endings elicited intensive calcium influxes in a sub-population of vagal ganglion neurons. These neurons were characterized by their positive responses to P2X2/3 receptor agonist α,ß-methylene ATP and TRPA1 agonist cinnamaldehyde, suggesting their nociceptive placodal nodose C-fiber neuron lineages. Pretreatment with TRPA1 selective blocker HC-030031 inhibited vincristine-induced calcium influxes in gastroesophageal nodose C-fiber neurons, indicating that TRPA1 played a functional role in mediating vincristine-induced activation response. Such inhibitory effect was comparable to that from 5-HT3 receptor antagonist palonosetron. Alternatively, pretreatment with ginger constituent 6-shogaol significantly attenuated vincristine-induced activation response. The present study provides new evidence that chemotherapeutic agent vincristine directly activates vagal nodose nociceptive C-fiber neurons at their peripheral nerve endings in the upper gastrointestinal tract. This activation response requires both TRPA1 and 5-HT3 receptors and can be attenuated by ginger constituent 6-shogaol.


Zingiber officinale , Mice , Animals , Vincristine/pharmacology , Calcium/pharmacology , Palonosetron/pharmacology , Esophagus/innervation , Action Potentials , Mice, Transgenic
2.
Molecules ; 26(13)2021 Jun 28.
Article En | MEDLINE | ID: mdl-34203134

Heartburn and non-cardiac chest pain are the predominant symptoms in many esophageal disorders, such as gastroesophageal reflux disease (GERD), non-erosive reflux disease (NERD), functional heartburn and chest pain, and eosinophilic esophagitis (EoE). At present, neuronal mechanisms underlying the process of interoceptive signals in the esophagus are still less clear. Noxious stimuli can activate a subpopulation of primary afferent neurons at their nerve terminals in the esophagus. The evoked action potentials are transmitted through both the spinal and vagal pathways to their central terminals, which synapse with the neurons in the central nervous system to induce esophageal nociception. Over the last few decades, progress has been made in our understanding on the peripheral and central neuronal mechanisms of esophageal nociception. In this review, we focus on the roles of capsaicin-sensitive vagal primary afferent nodose and jugular C-fiber neurons in processing nociceptive signals in the esophagus. We briefly compare their distinctive phenotypic features and functional responses to mechanical and chemical stimulations in the esophagus. Then, we summarize activation and/or sensitization effects of acid, inflammatory cells (eosinophils and mast cells), and mediators (ATP, 5-HT, bradykinin, adenosine, S1P) on these two nociceptive C-fiber subtypes. Lastly, we discuss the potential roles of capsaicin-sensitive esophageal afferent nerves in processing esophageal sensation and nociception. A better knowledge of the mechanism of nociceptive signal processes in primary afferent nerves in the esophagus will help to develop novel treatment approaches to relieve esophageal nociceptive symptoms, especially those that are refractory to proton pump inhibitors.


Action Potentials/drug effects , Capsaicin/therapeutic use , Esophagus/metabolism , Heartburn/diet therapy , Nociception/drug effects , Signal Transduction/drug effects , Vagus Nerve/metabolism , Animals , Esophagus/innervation , Esophagus/pathology , Heartburn/metabolism , Heartburn/pathology , Humans , Vagus Nerve/pathology
3.
Am J Physiol Gastrointest Liver Physiol ; 321(2): G149-G156, 2021 08 01.
Article En | MEDLINE | ID: mdl-34160291

Bile acid reflux in the esophagus plays a role in the pathogenesis of certain esophageal disorders, where it can induce esophageal pain and heartburn. The present study aimed to determine whether bile acid, deoxycholic acid (DCA), directly activates and sensitizes esophageal vagal nociceptive afferent C-fiber subtypes. DCA-elicited effects on vagal nodose and jugular neurons were studied by calcium imaging. Its effects on esophageal-labeled nodose and jugular neurons were then determined by patch-clamp recording. At nodose and jugular C-fiber nerve endings in the esophagus, DCA-evoked action potentials (APs) were compared by extracellular single-unit recordings in ex vivo esophageal-vagal preparations. DCA application induced calcium influxes in nodose and jugular neurons and elicited inward currents in esophageal-labeled nodose and jugular neurons. In the presence of DCA, the current densities elicited by capsaicin were enhanced in those labeled neurons. Consistently, DCA perfusion at nerve terminals in the esophagus evoked APs in about 50% of esophageal nodose and jugular C-fibers. In DCA-sensitive C-fibers, DCA perfusion also sensitized the fibers such that the subsequent response to capsaicin was amplified. Collectively, these results provide new evidence that DCA directly activates and sensitizes nociceptive nodose and jugular C-fibers in the esophagus. Such activation and sensitization effects may contribute to bile acid-induced esophageal nociceptive symptoms that are refractory to proton-pump inhibitor therapy.NEW & NOTEWORTHY Bile acid reflux in the esophagus can induce pain and heartburn in certain esophageal disorders, but the underlying neuronal mechanism is still unclear. The present study demonstrated that bile acid, deoxycholic acid (DCA), directly activates esophageal vagal afferent nodose and jugular nociceptive C-fibers and sensitizes their response to capsaicin. Such effects may contribute to bile acid-induced esophageal nociceptive symptoms that refractory to proton-pump inhibitors (PPIs) therapy.


Action Potentials , Cholagogues and Choleretics/pharmacology , Deoxycholic Acid/pharmacology , Esophagus/physiology , Nociceptors/physiology , Animals , Calcium Signaling , Cells, Cultured , Esophagus/innervation , Guinea Pigs , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/physiology , Nociceptors/drug effects , Nociceptors/metabolism , Vagus Nerve/drug effects , Vagus Nerve/physiology
4.
Pain ; 162(4): 1068-1081, 2021 04 01.
Article En | MEDLINE | ID: mdl-33021564

ABSTRACT: Mechanisms of visceral pain sensitization and referred somatic hypersensitivity remain unclear. We conducted calcium imaging in Pirt-GCaMP6s mice to gauge responses of dorsal root ganglion (DRG) neurons to visceral and somatic stimulation in vivo. Intracolonic instillation of 2,4,6-trinitrobenzene sulfonic acid (TNBS) induced colonic inflammation and increased the percentage of L6 DRG neurons that responded to colorectal distension above that of controls at day 7. Colorectal distension did not activate L4 DRG neurons. TNBS-treated mice exhibited more Evans blue extravasation than did control mice and developed mechanical hypersensitivity in low-back skin and hind paws, which are innervated by L6 and L4 DRG neurons, respectively, suggesting that colonic inflammation induced mechanical hypersensitivity in both homosegmental and heterosegmental somatic regions. Importantly, the percentage of L4 DRG neurons activated by hind paw pinch and brush stimulation and calcium responses of L6 DRG neurons to low-back brush stimulation were higher at day 7 after TNBS than those in control mice. Visceral irritation from intracolonic capsaicin instillation also increased Evans blue extravasation in hind paws and low-back skin and acutely increased the percentage of L4 DRG neurons responding to hind paw pinch and the response of L6 DRG neurons to low-back brush stimulation. These findings suggest that TNBS-induced colitis and capsaicin-induced visceral irritation may sensitize L6 DRG neurons to colorectal and somatic inputs and also increase the excitability of L4 DRG neurons that do not receive colorectal inputs. These changes may represent a potential peripheral neuronal mechanism for visceral pain sensitization and referred somatic hypersensitivity.


Ganglia, Spinal , Visceral Pain , Animals , Calcium , Disease Models, Animal , Mice , Neurons , Visceral Pain/chemically induced
6.
Neurogastroenterol Motil ; 31(6): e13585, 2019 06.
Article En | MEDLINE | ID: mdl-30947399

BACKGROUND: Ginger has been used as an herbal medicine worldwide to relieve nausea/vomiting and gastrointestinal discomfort, but the cellular and molecular mechanisms of its neuronal action remain unclear. The present study aimed to determine the effects of ginger constituent 6-shogaol on gastroesophageal vagal nodose C-fibers. METHODS: Extracellular single-unit recording and two-photon nodose neuron imaging were performed, respectively, in ex vivo gastroesophageal-vagal preparations from wild type and Pirt-GCaMP6 transgenic mice. The action potential discharge or calcium influx evoked by mechanical distension and chemical perfusions applied to the gastroesophageal vagal afferent nerve endings were recorded, respectively, at their intact neuronal cell soma in vagal nodose ganglia. The effects of 6-shogaol on nodose C-fiber neurons were then compared and determined. KEY RESULTS: Gastroesophageal application of 6-shogaol-elicited intensive calcium influxes in nodose neurons and evoked robust action potential discharges in most studied nodose C-fibers. Such activation effects were followed by a desensitized response to the second application of 6-shogaol. However, action potential discharges evoked by esophageal mechanical distension, after 6-shogaol perfusion, did not significantly change. Pretreatment with TRPA1 selective blocker HC-030031 inhibited 6-shogaol-induced action potential discharges in gastric and esophageal nodose C-fiber neurons, suggesting that TRPA1 played a role in mediating 6-shogaol-induced activation response. CONCLUSION AND INFERENCES: This study provides evidence that ginger constituent 6-shogaol directly activates vagal afferent C-fiber peripheral gastrointestinal endings. This activation leads to desensitization to subsequent application of 6-shogaol but not subsequent esophageal mechanical distension. Further investigation is required to establish a possible contribution in its anti-emetic effects.


Catechols/pharmacology , Nerve Fibers, Unmyelinated/drug effects , Neurons, Afferent/drug effects , Nodose Ganglion/drug effects , Action Potentials/drug effects , Animals , Esophagus/drug effects , Esophagus/innervation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Stomach/drug effects , Stomach/innervation
7.
Biomed Opt Express ; 10(3): 1126-1135, 2019 Mar 01.
Article En | MEDLINE | ID: mdl-30891334

We report parallel-trained deep neural networks for automated endoscopic OCT image segmentation feasible even with a limited training data set. These U-Net-based deep neural networks were trained using a modified dice loss function and manual segmentations of ultrahigh-resolution cross-sectional images collected by an 800 nm OCT endoscopic system. The method was tested on in vivo guinea pig esophagus images. Results showed its robust layer segmentation capability with a boundary error of 1.4 µm insensitive to lay topology disorders. To further illustrate its clinical potential, the method was applied to differentiating in vivo OCT esophagus images from an eosinophilic esophagitis (EOE) model and its control group, and the results clearly demonstrated quantitative changes in the top esophageal layers' thickness in the EOE model.

8.
Neurogastroenterol Motil ; 31(4): e13543, 2019 04.
Article En | MEDLINE | ID: mdl-30663188

INTRODUCTION: Acid reflux in the esophagus can induce painful sensations such as heartburn and non-cardiac chest pain. These nociceptive symptoms are initiated by activation of TRPV1-positive afferent C fibers in the esophagus. The present study aimed to explore a novel C fiber inhibition approach. We hypothesized that activation of TRPV1 by acid enabled QX-314, a membrane impermeable sodium channel blocker, to inhibit acid-induced activation of esophageal nociceptive C fiber neurons. METHOD: We determined the inhibitory effect of QX-314 in the presence of acid in guinea pig esophageal nociceptive vagal jugular C fiber neurons by both patch clamp recording in neuron soma and by extra-cellular recording at nerve terminals. KEY RESULTS: Our data demonstrated QX-314 alone did not inhibit sodium currents. However, when applied along with capsaicin to activate TRPV1, QX-314 was able to block sodium currents in esophageal-specific jugular C fiber neurons. We then showed that in the presence of acid, QX-314 significantly blocked acid-evoked activation of jugular C fiber neurons. This effect was attenuated by TRPV1 antagonist AMG9810, suggesting acid-mediated inhibitory effect of QX-314 was TRPV1-dependent. Finally, we provided evidence at nerve endings that acid-evoked action potential discharges in esophageal jugular C fibers were inhibited by QX-314 when applied in the presence of acid. CONCLUSION AND INFERENCES: Our data demonstrated that activation of TRPV1 by acid enabled membrane impermeable sodium channel blocker QX-314 to inhibit acid-induced activation in esophageal nociceptive C fibers. This supports a localized application of QX-314 in the esophagus to block esophageal nociception in acid reflux disorders.


Action Potentials/drug effects , Esophagus/innervation , Lidocaine/analogs & derivatives , Nerve Fibers, Unmyelinated/drug effects , Nociception/drug effects , Acrylamides/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Capsaicin/pharmacology , Esophagus/drug effects , Guinea Pigs , Hydrochloric Acid/pharmacology , Lidocaine/pharmacology , Patch-Clamp Techniques , TRPV Cation Channels/antagonists & inhibitors
9.
Mol Med Rep ; 18(2): 2171-2181, 2018 Aug.
Article En | MEDLINE | ID: mdl-29901151

A growing body of evidence has demonstrated that Eph/ephrin signalling may serve a central role in intestinal diseases. However, whether erythropoietin­producing hepatocellular (Eph)/ephrin signalling is associated with the development of post­infectious irritable bowel syndrome (PI­IBS) is still unknown. In the present study, the role of Eph/Ephrin signalling in lipopolysaccharide (LPS)­induced intestinal injury was evaluated in vivo and in vitro. LPS treatment significantly increased the levels of proinflammatory mediators [monocyte chemoattractant protein­1, tumour necrosis factor α, interleukin (IL)­1ß, IL­6, intercellular adhesion molecule 1 and vascular cell adhesion molecule­1], activated the EphA2­Ephrin A1, protein kinase B (Akt)­nuclear factor (NF)­κB, Src­NF­κB and Wnt/ß­catenin signalling pathways, and inhibited EphB1­Ephrin B3 signalling in colon tissues, and primary cultured enteric neuronal and glial cells. Notably, EphA2 monoclonal antibody (mAb) treatment or Ephrin B3 overexpression could partially alleviate the LPS­induced upregulation of proinflammatory mediators, and Akt­NF­κB, Src­NF­κB and Wnt/ß­catenin signalling pathways. In addition, EphA2 mAb treatment could partially inhibit LPS­induced inactivation of EphB­Ephrin B3 signalling, while Ephrin B3 overexpression could abrogate LPS­induced activation of EphA2­Ephrin A1 signalling. EphB1/Ephrin B3 signalling may antagonise the EphA2/Ephrin A1­dependent pathway following LPS treatment. The results associated with the EphA2 signaling pathway, indicated that Eph/ephrin signalling may serve a bidirectional role in LPS­induced intestinal injury. Eph/ephrin signalling may be a novel therapeutic target for LPS­induced intestinal injury and potentially PI­IBS.


Ephrin-A1/metabolism , Ephrin-B3/metabolism , Intestinal Mucosa/metabolism , Irritable Bowel Syndrome/metabolism , Lipopolysaccharides/toxicity , Receptor, EphA2/metabolism , Receptor, EphB1/metabolism , Signal Transduction/drug effects , Animals , Intestines/injuries , Intestines/pathology , Irritable Bowel Syndrome/chemically induced , Irritable Bowel Syndrome/pathology , Male , Mice , Mice, Inbred BALB C
10.
Front Pharmacol ; 9: 272, 2018.
Article En | MEDLINE | ID: mdl-29662452

Though the detailed pathological mechanism of post-infectious irritable bowel syndrome (PI-IBS) remains unclear, accumulating evidence indicates that oxidative stress and inflammation are implicated in the process of PI-IBS. Oxidative stress and inflammation are regulated by Nrf2 and NF-κB signaling pathways, respectively. EphA2, a member of Eph receptor family, promotes oxidative stress and inflammatory responses via regulation of Nrf2 and NF-κB signaling pathways in various types of human diseases. Understanding the mechanisms by which EphA2 regulate oxidative stress and inflammation in PI-IBS is important for the development of new strategies to treat PI-IBS. However, the effects of ALW-II-41-27, a novel EphA2 inhibitor on PI-IBS and the underlying molecular mechanisms have never been studied. In the present study, we showed that ALW-II-41-27 decreased gastrointestinal motility and abdominal withdrawal reflex (AWR) scores, markedly reduced the levels of oxidative stress markers [4-hydroxy-2-nonenal (4-HNE), protein carbonyl, and 8-hydroxy-2-de-axyguanine (8-OHdG)] and proinflammatory cytokines (TNF-α, IL-6, IL-17, and ICAM-1), and remarkably increased the level of anti-inflammatory cytokine (IL-10) in serum and colon of Trichinella spiralis-infected mice. Moreover, ALW-II-41-27 was effective in suppressing oxidative stress and inflammation in LPS-treated NCM460 colonic cells. Treatment of ALW-II-41-27 reversed the activation of NF-κB and inactivation of Nrf2 in LPS-treated NCM460 cells. Importantly, these protective effects of ALW-II-41-27 were partially inhibited by EphA2 KO and abolished by EphA2 overexpression. In conclusion, EphA2 may represent a promising therapeutic target for patients with PI-IBS and ALW-II-41-27 might function as a novel therapeutic agent for PI-IBS.

11.
Opt Lett ; 41(2): 250-3, 2016 Jan 15.
Article En | MEDLINE | ID: mdl-26766686

We investigated the optimal operational conditions for utilizing a broadband supercontinuum (SC) source in a portable 800 nm spectral-domain (SD) endoscopic OCT system to enable high resolution, high-sensitivity, and high-speed imaging in vivo. A SC source with a 3-dB bandwidth of ∼246 nm was employed to obtain an axial resolution of ∼2.7 µm (in air) and an optimal detection sensitivity of ∼-107 dB with an imaging speed up to 35 frames/s (at 70 k A-scans/s). The performance of the SC-based SD-OCT endoscopy system was demonstrated by imaging guinea pig esophagus in vivo, achieving image quality comparable to that acquired with a broadband home-built Ti:sapphire laser.


Endoscopy/methods , Signal-To-Noise Ratio , Tomography, Optical Coherence/methods , Animals , Guinea Pigs , Image Processing, Computer-Assisted
12.
Semin Immunopathol ; 38(3): 385-96, 2016 05.
Article En | MEDLINE | ID: mdl-26459157

Transient receptor potential (TRP) channels are predominantly distributed in both somatic and visceral sensory nervous systems and play a crucial role in sensory transduction. As the largest visceral organ system, the gastrointestinal (GI) tract frequently accommodates external inputs, which stimulate sensory nerves to initiate and coordinate sensory and motor functions in order to digest and absorb nutrients. Meanwhile, the sensory nerves in the GI tract are also able to detect potential tissue damage by responding to noxious irritants. This nocifensive function is mediated through specific ion channels and receptors expressed in a subpopulation of spinal and vagal afferent nerve called nociceptor. In the last 18 years, our understanding of TRP channel expression and function in GI sensory nervous system has been continuously improved. In this review, we focus on the expressions and functions of TRPV1, TRPA1, and TRPM8 in primary extrinsic afferent nerves innervated in the esophagus, stomach, intestine, and colon and briefly discuss their potential roles in relevant GI disorders.


Gastrointestinal Tract/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , Animals , Disease Susceptibility , Gastrointestinal Tract/innervation , Gene Expression , Humans , Multigene Family , Organ Specificity/genetics , Protein Isoforms , Sensory Receptor Cells/metabolism , Signal Transduction
13.
Am J Physiol Gastrointest Liver Physiol ; 308(6): G489-96, 2015 Mar 15.
Article En | MEDLINE | ID: mdl-25591866

Sensory transduction in esophageal afferents requires specific ion channels and receptors. TRPM8 is a new member of the transient receptor potential (TRP) channel family and participates in cold- and menthol-induced sensory transduction, but its role in visceral sensory transduction is still less clear. This study aims to determine TRPM8 function and expression in esophageal vagal afferent subtypes. TRPM8 agonist WS-12-induced responses were first determined in nodose and jugular neurons by calcium imaging and then investigated by whole cell patch-clamp recordings in Dil-labeled esophageal nodose and jugular neurons. Extracellular single-unit recordings were performed in nodose and jugular C fiber neurons using ex vivo esophageal-vagal preparations with intact nerve endings in the esophagus. TRPM8 mRNA expression was determined by single neuron RT-PCR in Dil-labeled esophageal nodose and jugular neurons. The TRPM8 agonist WS-12 elicited calcium influx in a subpopulation of jugular but not nodose neurons. WS-12 activated outwardly rectifying currents in esophageal Dil-labeled jugular but not nodose neurons in a dose-dependent manner, which could be inhibited by the TRPM8 inhibitor AMTB. WS-12 selectively evoked action potential discharges in esophageal jugular but not nodose C fibers. Consistently, TRPM8 transcripts were highly expressed in esophageal Dil-labeled TRPV1-positive jugular neurons. In summary, the present study demonstrated a preferential expression and function of TRPM8 in esophageal vagal jugular but not nodose neurons and C fiber subtypes. This provides a distinctive role of TRPM8 in esophageal sensory transduction and may lead to a better understanding of the mechanisms of esophageal sensation and nociception.


Esophagus/innervation , Nerve Fibers, Unmyelinated/metabolism , Nodose Ganglion/metabolism , Sensory Receptor Cells/metabolism , TRPM Cation Channels/metabolism , Action Potentials , Anilides/pharmacology , Animals , Benzamides/pharmacology , Calcium Signaling , Dose-Response Relationship, Drug , Guinea Pigs , Male , Menthol/analogs & derivatives , Menthol/pharmacology , Nerve Fibers, Unmyelinated/drug effects , Nodose Ganglion/drug effects , RNA, Messenger/metabolism , Sensation , Sensory Receptor Cells/drug effects , TRPM Cation Channels/drug effects , TRPM Cation Channels/genetics , TRPV Cation Channels/metabolism , Thiophenes/pharmacology , Time Factors
14.
Am J Physiol Gastrointest Liver Physiol ; 308(6): G482-8, 2015 Mar 15.
Article En | MEDLINE | ID: mdl-25591867

Transient receptor potential A1 (TRPA1) is a newly defined cationic ion channel, which selectively expresses in primary sensory afferent nerve, and is essential in mediating inflammatory nociception. Our previous study demonstrated that TRPA1 plays an important role in tissue mast cell activation-induced increase in the excitability of esophageal vagal nodose C fibers. The present study aims to determine whether prolonged antigen exposure in vivo sensitizes TRPA1 in a guinea pig model of eosinophilic esophagitis (EoE). Antigen challenge-induced responses in esophageal mucosa were first assessed by histological stains and Ussing chamber studies. TRPA1 function in vagal sensory neurons was then studied by calcium imaging and by whole cell patch-clamp recordings in 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI)-labeled esophageal vagal nodose and jugular neurons. Extracellular single-unit recordings were performed in vagal nodose and jugular C-fiber neuron subtypes using ex vivo esophageal-vagal preparations with intact nerve endings in the esophagus. Antigen challenge significantly increased infiltrations of eosinophils and mast cells in the esophagus. TRPA1 agonist allyl isothiocyanate (AITC)-induced calcium influx in nodose and jugular neurons was significantly increased, and current densities in esophageal DiI-labeled nodose and jugular neurons were also significantly increased in antigen-challenged animals. Prolonged antigen challenge decreased esophageal epithelial barrier resistance, which allowed intraesophageal-infused AITC-activating nodose and jugular C fibers at their nerve endings. Collectively, these results demonstrated that prolonged antigen challenge sensitized TRPA1 in esophageal sensory neurons and afferent C fibers. This novel finding will help us to better understand the molecular mechanism underlying esophageal sensory and motor dysfunctions in EoE.


Allergens , Eosinophilic Esophagitis/metabolism , Esophagus/innervation , Nerve Fibers, Unmyelinated/metabolism , Ovalbumin , Sensory Receptor Cells/metabolism , Transient Receptor Potential Channels/metabolism , Vagus Nerve/metabolism , Action Potentials , Animals , Calcium Signaling , Disease Models, Animal , Eosinophilic Esophagitis/immunology , Eosinophilic Esophagitis/physiopathology , Eosinophils/immunology , Eosinophils/metabolism , Guinea Pigs , Isothiocyanates/pharmacology , Male , Mast Cells/immunology , Mast Cells/metabolism , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/immunology , Nodose Ganglion/drug effects , Nodose Ganglion/immunology , Nodose Ganglion/metabolism , Sensation , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/immunology , Time Factors , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/immunology , Vagus Nerve/drug effects , Vagus Nerve/immunology , Vagus Nerve/physiopathology
15.
Am J Physiol Gastrointest Liver Physiol ; 307(4): G471-8, 2014 Aug 15.
Article En | MEDLINE | ID: mdl-24994852

Acid reflux-induced heartburn and noncardiac chest pain are processed peripherally by sensory nerve endings in the wall of the esophagus, but the underlying mechanism is still unclear. This study aims to determine the effects of acid on esophageal vagal nociceptive afferent subtypes. Extracellular single-unit recordings were performed in guinea pig vagal nodose or jugular C fiber neurons by using ex vivo esophageal-vagal preparations with intact nerve endings in the esophagus. We recorded action potentials (AP) of esophageal nodose or jugular C fibers evoked by acid perfusion and compared esophageal distension-evoked AP before and after acid perfusion. Acid perfusion for 30 min (pH range 7.4 to 5.8) did not evoke AP in nodose C fibers but significantly decreased their responses to esophageal distension, which could be recovered after washing out acid for 90 min. In jugular C fibers, acid perfusion not only evoked AP but also inhibited their responses to esophageal distension, which were not recovered after washing out acid for 120 min. Lower concentration of capsaicin perfusion mimicked acid-induced effects in nodose and jugular C fibers. Pretreatment with TRPV1 antagonist AMG9810, but not acid-sensing ion channel (ASIC) inhibitor amiloride, significantly inhibited acid-induced effects in nodose and jugular C fiber. These results demonstrate that esophageal vagal nociceptive afferent nerve subtypes display distinctive responses to acid. Acid activates jugular, but not nodose, C fibers and inhibits both of their responses to esophageal distension. These effects are mediated mainly through TRPV1. This inhibitory effect is a novel finding and may contribute to esophageal sensory/motor dysfunction in acid reflux diseases.


Acids/pharmacology , Esophagus/innervation , Nerve Fibers, Unmyelinated/drug effects , Nociception/drug effects , Nodose Ganglion/drug effects , Vagus Nerve/drug effects , Acrylamides/pharmacology , Action Potentials/drug effects , Amiloride/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Capsaicin/pharmacology , Guinea Pigs , Heartburn/physiopathology , In Vitro Techniques , Male , TRPV Cation Channels/drug effects , TRPV Cation Channels/physiology
16.
Am J Physiol Gastrointest Liver Physiol ; 307(2): G149-57, 2014 Jul 15.
Article En | MEDLINE | ID: mdl-24875100

Eosinophilic esophagitis (EoE) is characterized with eosinophils and mast cells predominated allergic inflammation in the esophagus and present with esophageal dysfunctions such as dysphagia, food impaction, and heartburn. However, the underlying mechanism of esophageal dysfunctions is unclear. This study aims to determine whether neurons in the vagal sensory ganglia are modulated in a guinea pig model of EoE. Animals were actively sensitized by ovalbumin (OVA) and then challenged with aerosol OVA inhalation for 2 wk. This results in a mild esophagitis with increases in mast cells and eosinophils in the esophageal wall. Vagal nodose and jugular neurons were disassociated, and their responses to acid, capsaicin, and transient receptor potential vanilloid type 1 (TRPV1) antagonist AMG-9810 were studied by calcium imaging and whole cell patch-clamp recording. Compared with naïve animals, antigen challenge significantly increased acid responsiveness in both nodose and jugular neurons. Their responses to capsaicin were also increased after antigen challenge. AMG-9810, at a concentration that blocked capsaicin-evoked calcium influx, abolished the increase in acid-induced activation in both nodose and jugular neurons. Vagotomy strongly attenuated those increased responses of nodose and jugular neurons to both acid and capsaicin induced by antigen challenge. These data for the first time demonstrated that prolonged antigen challenge significantly increases acid responsiveness in vagal nodose and jugular ganglia neurons. This sensitization effect is mediated largely through TRPV1 and initiated at sensory nerve endings in the peripheral tissues. Allergen-induced enhancement of responsiveness to noxious stimulation by acid in sensory nerve may contribute to the development of esophageal dysfunctions such as heartburn in EoE.


Eosinophilic Esophagitis/metabolism , Esophagus/innervation , Heartburn/metabolism , Nodose Ganglion/metabolism , Vagus Nerve/metabolism , Acrylamides/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Calcium Signaling/drug effects , Capsaicin/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Eosinophilic Esophagitis/chemically induced , Eosinophilic Esophagitis/physiopathology , Guinea Pigs , Heartburn/chemically induced , Heartburn/physiopathology , Hydrogen-Ion Concentration , Male , Membrane Potentials , Nodose Ganglion/drug effects , Nodose Ganglion/physiopathology , Nodose Ganglion/surgery , Ovalbumin , TRPV Cation Channels/drug effects , TRPV Cation Channels/metabolism , Time Factors , Vagotomy , Vagus Nerve/drug effects , Vagus Nerve/physiopathology , Vagus Nerve/surgery
17.
Opt Lett ; 39(7): 2016-9, 2014 Apr 01.
Article En | MEDLINE | ID: mdl-24686663

We present a novel design for an endoscopic imaging catheter utilizing diffractive optics for ultrahigh-resolution optical coherence tomography (OCT) imaging at 800 nm. A diffractive microlens was developed to alleviate severe chromatic aberration when a broadband light source was employed at the 800 nm wavelength range. Combined with a home-built fiber rotary joint and a broadband Ti:sapphire laser, the imaging catheter achieved a lateral resolution of 6.2 µm and an axial resolution of 3.0 µm in air. The performance of the catheter was demonstrated by three-dimensional full-circumferential endoscopic OCT imaging of guinea pig esophagus in vivo.


Tomography, Optical Coherence/methods , Animals , Endoscopy , Esophagus/cytology , Guinea Pigs , Imaging, Three-Dimensional
18.
Am J Physiol Gastrointest Liver Physiol ; 306(3): G200-7, 2014 Feb.
Article En | MEDLINE | ID: mdl-24264049

Acid reflux in the esophagus can induce esophageal painful sensations such as heartburn and noncardiac chest pain. The mechanisms underlying acid-induced esophageal nociception are not clearly understood. In our previous studies, we characterized esophageal vagal nociceptive afferents and defined their responses to noxious mechanical and chemical stimulation. In the present study, we aim to determine their responses to intraluminal acid infusion. Extracellular single-unit recordings were performed in nodose ganglion neurons with intact nerve endings in the esophagus using ex vivo esophageal-vagal preparations. Action potentials evoked by esophageal intraluminal acid perfusion were compared in naive and ovalbumin (OVA)-challenged animals, followed by measurements of transepithelial electrical resistance (TEER) and the expression of tight junction proteins (zona occludens-1 and occludin). In naive guinea pigs, intraluminal infusion with either acid (pH = 2-3) or capsaicin did not evoke an action potential discharge in esophageal nodose C fibers. In OVA-sensitized animals, following esophageal mast cell activation by in vivo OVA inhalation, intraluminal acid infusion for about 20 min started to evoke action potential discharges. This effect is further confirmed by selective mast cell activation using in vitro tissue OVA challenge in esophageal-vagal preparations. OVA inhalation leads to decreased TEER and zona occludens-1 expression, suggesting an impaired esophageal epithelial barrier function after mast cell activation. These data for the first time provide direct evidence of intraluminal acid-induced activation of esophageal nociceptive C fibers and suggest that mast cell activation may make esophageal epithelium more permeable to acid, which subsequently may increase esophageal vagal nociceptive C fiber activation.


Acids/pharmacology , Esophagus/drug effects , Evoked Potentials/physiology , Mast Cells/drug effects , Nerve Fibers, Unmyelinated/drug effects , Nodose Ganglion/cytology , Animals , Capsaicin/pharmacology , Esophagus/innervation , Esophagus/metabolism , Guinea Pigs , Male , Mast Cells/metabolism , Nerve Fibers, Unmyelinated/metabolism , Ovalbumin/pharmacology , Stimulation, Chemical , Vagus Nerve/drug effects , Vagus Nerve/metabolism
19.
Am J Physiol Gastrointest Liver Physiol ; 304(10): G908-16, 2013 May 15.
Article En | MEDLINE | ID: mdl-23471341

Sensitization of esophageal afferents plays an important role in esophageal nociception, but the mechanism is less clear. Our previous studies demonstrated that mast cell (MC) activation releases the preformed mediators histamine and tryptase, which play important roles in sensitization of esophageal vagal nociceptive C fibers. PGD2 is a lipid mediator released by activated MCs. Whether PGD2 plays a role in this sensitization process has yet to be determined. Expression of the PGD2 DP1 and DP2 receptors in nodose ganglion neurons was determined by immunofluorescence staining, Western blotting, and RT-PCR. Extracellular recordings were performed in ex vivo esophageal-vagal preparations. Action potentials evoked by esophageal distension were compared before and after perfusion of PGD2, DP1 and DP2 receptor agonists, and MC activation, with or without pretreatment with antagonists. The effect of PGD2 on 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI)-labeled esophageal nodose neurons was determined by patch-clamp recording. Our results demonstrate that DP1 and DP2 receptor mRNA and protein were expressed mainly in small- and medium-diameter neurons in nodose ganglia. PGD2 significantly increased esophageal distension-evoked action potential discharges in esophageal nodose C fibers. The DP1 receptor agonist BW 245C mimicked this effect. PGD2 directly sensitized DiI-labeled esophageal nodose neurons by decreasing the action potential threshold. Pretreatment with the DP1 receptor antagonist BW A868C significantly inhibited PGD2 perfusion- or MC activation-induced increases in esophageal distension-evoked action potential discharges in esophageal nodose C fibers. In conclusion, PGD2 plays an important role in MC activation-induced sensitization of esophageal nodose C fibers. This adds a novel mechanism of visceral afferent sensitization.


Esophagus/innervation , Mast Cells/drug effects , Neurons, Afferent/drug effects , Prostaglandin D2/pharmacology , Vagus Nerve/drug effects , Action Potentials/drug effects , Animals , Blotting, Western , Carbocyanines , Data Interpretation, Statistical , Esophagus/drug effects , Fluorescent Antibody Technique , Guinea Pigs , Male , Nerve Fibers/physiology , Nerve Fibers, Unmyelinated/drug effects , Nodose Ganglion/cytology , Nodose Ganglion/drug effects , Patch-Clamp Techniques , Physical Stimulation , Prostaglandin D2/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/agonists , Receptors, Immunologic/drug effects , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/drug effects
20.
Am J Physiol Gastrointest Liver Physiol ; 301(6): G1052-8, 2011 Dec.
Article En | MEDLINE | ID: mdl-21960520

Eosinophilic esophagitis is characterized by increased infiltration and degranulation of eosinophils in the esophagus. Whether eosinophil-derived cationic proteins regulate esophageal sensory nerve function is still unknown. Using synthetic cationic protein to investigate such effect, we performed extracellular recordings from vagal nodose or jugular neurons in ex vivo esophageal-vagal preparations with intact nerve endings in the esophagus. Nerve excitabilities were determined by comparing action potentials evoked by esophageal distensions before and after perfusion of synthetic cationic protein poly-L-lysine (PLL) with or without pretreatment with poly-L-glutamic acid (PLGA), which neutralized cationic charges of PLL. Perfusion with PLL did not evoke action potentials in esophageal nodose C fibers but increased their responses to esophageal distension. This potentiation effect lasted for 30 min after washing out of PLL. Pretreatment with PLGA significantly inhibited PLL-induced mechanohyperexcitability of esophageal nodose C fibers. In esophageal nodose Aδ fibers, perfusion with PLL did not evoke action potentials. In contrast to nodose C fibers, both the spontaneous discharges and the responses to esophageal distension in nodose Aδ fibers were decreased by perfusion with PLL, which can be restored after washing out PLL for 30-60 min. Pretreatment with PLGA attenuated PLL-induced decrease in spontaneous discharge and mechanoexcitability of esophageal nodose Aδ fibers. In esophageal jugular C fibers, PLL neither evoked action potentials nor changed their responses to esophageal distension. Collectively, these data demonstrated that synthetic cationic protein did not evoke action potential discharges of esophageal vagal afferents but had distinctive sensitization effects on their responses to esophageal distension.


Antimicrobial Cationic Peptides/pharmacology , Esophagus/innervation , Mechanoreceptors/drug effects , Neurons, Afferent/drug effects , Nodose Ganglion/drug effects , Animals , Antimicrobial Cationic Peptides/chemical synthesis , Biocompatible Materials/pharmacology , Drug Interactions , Guinea Pigs , Jugular Veins/innervation , Lactic Acid/pharmacology , Male , Mechanoreceptors/physiology , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/physiology , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/physiology , Neurons, Afferent/physiology , Nodose Ganglion/physiology , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer
...