1
|
Gupta RA, Higham JP, Pearce A, Urriola-Muñoz P, Barker KH, Paine L, Ghooraroo J, Raine T, Hockley JRF, Rahman T, St John Smith E, Brown AJH, Ladds G, Suzuki R, Bulmer DC. GPR35 agonists inhibit TRPA1-mediated colonic nociception through suppression of substance P release. Pain 2024:00006396-990000000-00727. [PMID: 39382322 DOI: 10.1097/j.pain.0000000000003399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/25/2024] [Indexed: 10/10/2024]
Abstract
ABSTRACT The development of nonopioid analgesics for the treatment of abdominal pain is a pressing clinical problem. To address this, we examined the expression of Gi/o-coupled receptors, which typically inhibit nociceptor activation, in colonic sensory neurons. This led to the identification of the orphan receptor GPR35 as a visceral analgesic drug target because of its marked coexpression with transient receptor potential ankyrin 1 (TRPA1), a mediator of noxious mechanotransduction in the bowel. Building on in silico docking simulations, we confirmed that the mast cell stabiliser, cromolyn (CS), and phosphodiesterase inhibitor, zaprinast, are agonists at mouse GPR35, promoting the activation of different Gi/o subunits. Pretreatment with either CS or zaprinast significantly attenuated TRPA1-mediated colonic nociceptor activation and prevented TRPA1-mediated mechanosensitisation. These effects were lost in tissue from GPR35-/- mice and were shown to be mediated by inhibition of TRPA1-evoked substance P (SP) release. This observation highlights the pronociceptive effect of SP and its contribution to TRPA1-mediated colonic nociceptor activation and sensitisation. Consistent with this mechanism of action, we confirmed that TRPA1-mediated colonic contractions evoked by SP release were abolished by CS pretreatment in a GPR35-dependent manner. Our data demonstrate that GPR35 agonists prevent the activation and sensitisation of colonic nociceptors through the inhibition of TRPA1-mediated SP release. These findings highlight the potential of GPR35 agonists to deliver nonopioid analgesia for the treatment of abdominal pain.
Collapse
Affiliation(s)
- Rohit A Gupta
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - James P Higham
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Abigail Pearce
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Paulina Urriola-Muñoz
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Katie H Barker
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Luke Paine
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Joshua Ghooraroo
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Tim Raine
- Department of Gastroenterology, Addenbrookes Hospital, Cambridge University Teaching Hospitals, Cambridge, United Kingdom
| | - James R F Hockley
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Alastair J H Brown
- Nxera, Steinmetz Building, Granta Park Great Abington, Cambridge, United Kingdom
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Rie Suzuki
- Nxera, Steinmetz Building, Granta Park Great Abington, Cambridge, United Kingdom
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| |
Collapse
|
2
|
Zhan D, Zhang J, Su S, Ren X, Zhao S, Zang W, Cao J. TET1 Participates in Complete Freund's Adjuvant-induced Trigeminal Inflammatory Pain by Regulating Kv7.2 in a Mouse Model. Neurosci Bull 2024; 40:707-718. [PMID: 37973721 PMCID: PMC11178721 DOI: 10.1007/s12264-023-01139-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/10/2023] [Indexed: 11/19/2023] Open
Abstract
Trigeminal inflammatory pain is one of the most severe pain-related disorders in humans; however, the underlying mechanisms remain largely unknown. In this study, we investigated the possible contribution of interaction between ten-eleven translocation methylcytosine dioxygenase 1 (TET1) and the voltage-gated K+ channel Kv7.2 (encoded by Kcnq2) to orofacial inflammatory pain in mice. We found that complete Freund's adjuvant (CFA) injection reduced the expression of Kcnq2/Kv7.2 in the trigeminal ganglion (TG) and induced orofacial inflammatory pain. The involvement of Kv7.2 in CFA-induced orofacial pain was further confirmed by Kv7.2 knockdown or overexpression. Moreover, TET1 knockdown in Tet1flox/flox mice significantly reduced the expression of Kv7.2 and M currents in the TG and led to pain-like behaviors. Conversely, TET1 overexpression by lentivirus rescued the CFA-induced decreases of Kcnq2 and M currents and alleviated mechanical allodynia. Our data suggest that TET1 is implicated in CFA-induced trigeminal inflammatory pain by positively regulating Kv7.2 in TG neurons.
Collapse
Affiliation(s)
- Dengcheng Zhan
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Neuroscience Research Institute, Zhengzhou University, Zhengzhou, 450001, China
| | - Jingjing Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Neuroscience Research Institute, Zhengzhou University, Zhengzhou, 450001, China
| | - Songxue Su
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiuhua Ren
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Sen Zhao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Neuroscience Research Institute, Zhengzhou University, Zhengzhou, 450001, China
| | - Weidong Zang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Neuroscience Research Institute, Zhengzhou University, Zhengzhou, 450001, China.
| | - Jing Cao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Neuroscience Research Institute, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
3
|
Baker CC, Sessenwein JL, Wood HM, Yu Y, Tsang Q, Alward TA, Jimenez Vargas NN, Omar AA, McDonnel A, Segal JP, Sjaarda CP, Bunnett NW, Schmidt BL, Caminero A, Boev N, Bannerman CA, Ghasemlou N, Sheth PM, Vanner SJ, Reed DE, Lomax AE. Protease-Induced Excitation of Dorsal Root Ganglion Neurons in Response to Acute Perturbation of the Gut Microbiota Is Associated With Visceral and Somatic Hypersensitivity. Cell Mol Gastroenterol Hepatol 2024; 18:101334. [PMID: 38494056 PMCID: PMC11350452 DOI: 10.1016/j.jcmgh.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND & AIMS Abdominal pain is a major symptom of diseases that are associated with microbial dysbiosis, including irritable bowel syndrome and inflammatory bowel disease. Germ-free mice are more prone to abdominal pain than conventionally housed mice, and reconstitution of the microbiota in germ-free mice reduces abdominal pain sensitivity. However, the mechanisms underlying microbial modulation of pain remain elusive. We hypothesized that disruption of the intestinal microbiota modulates the excitability of peripheral nociceptive neurons. METHODS In vivo and in vitro assays of visceral sensation were performed on mice treated with the nonabsorbable antibiotic vancomycin (50 μg/mL in drinking water) for 7 days and water-treated control mice. Bacterial dysbiosis was verified by 16s rRNA analysis of stool microbial composition. RESULTS Treatment of mice with vancomycin led to an increased sensitivity to colonic distension in vivo and in vitro and hyperexcitability of dorsal root ganglion (DRG) neurons in vitro, compared with controls. Interestingly, hyperexcitability of DRG neurons was not restricted to those that innervated the gut, suggesting a widespread effect of gut dysbiosis on peripheral pain circuits. Consistent with this, mice treated with vancomycin were more sensitive than control mice to thermal stimuli applied to hind paws. Incubation of DRG neurons from naive mice in serum from vancomycin-treated mice increased DRG neuron excitability, suggesting that microbial dysbiosis alters circulating mediators that influence nociception. The cysteine protease inhibitor E64 (30 nmol/L) and the protease-activated receptor 2 (PAR-2) antagonist GB-83 (10 μmol/L) each blocked the increase in DRG neuron excitability in response to serum from vancomycin-treated mice, as did the knockout of PAR-2 in NaV1.8-expressing neurons. Stool supernatant, but not colonic supernatant, from mice treated with vancomycin increased DRG neuron excitability via cysteine protease activation of PAR-2. CONCLUSIONS Together, these data suggest that gut microbial dysbiosis alters pain sensitivity and identify cysteine proteases as a potential mediator of this effect.
Collapse
Affiliation(s)
- Corey C Baker
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Jessica L Sessenwein
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Hannah M Wood
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Yang Yu
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Quentin Tsang
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Taylor A Alward
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | - Amal Abu Omar
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Abby McDonnel
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Julia P Segal
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Calvin P Sjaarda
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, Neuroscience Institute, New York University, New York, New York
| | - Brian L Schmidt
- Department of Molecular Pathobiology, Neuroscience Institute, New York University, New York, New York
| | - Alberto Caminero
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Nadejda Boev
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Courtney A Bannerman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Nader Ghasemlou
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; Department of Anesthesiology and Perioperative Medicine, Queen's Unversity, Kingston, Ontario, Canada
| | - Prameet M Sheth
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stephen J Vanner
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
4
|
Bhebhe CN, Higham JP, Gupta RA, Raine T, Bulmer DC. K V7 but not dual small and intermediate K Ca channel openers inhibit the activation of colonic afferents by noxious stimuli. Am J Physiol Gastrointest Liver Physiol 2023; 325:G436-G445. [PMID: 37667839 PMCID: PMC10894664 DOI: 10.1152/ajpgi.00141.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/06/2023]
Abstract
In numerous subtypes of central and peripheral neurons, small and intermediate conductance Ca2+-activated K+ (SK and IK, respectively) channels are important regulators of neuronal excitability. Transcripts encoding SK channel subunits, as well as the closely related IK subunit, are coexpressed in the soma of colonic afferent neurons with receptors for the algogenic mediators ATP and bradykinin, P2X3 and B2, highlighting the potential utility of these channels as drug targets for the treatment of abdominal pain in gastrointestinal diseases such as irritable bowel syndrome. Despite this, pretreatment with the dual SK/IK channel opener SKA-31 had no effect on the colonic afferent response to ATP, bradykinin, or noxious ramp distention of the colon. Inhibition of SK or IK channels with apamin or TRAM-34, respectively, yielded no change in spontaneous baseline afferent activity, indicating these channels are not tonically active. In contrast to its lack of effect in electrophysiological experiments, comparable concentrations of SKA-31 abolished ongoing peristaltic activity in the colon ex vivo. Treatment with the KV7 channel opener retigabine blunted the colonic afferent response to all applied stimuli. Our data therefore highlight the potential utility of KV7, but not SK/IK, channel openers as analgesic agents for the treatment of abdominal pain.NEW & NOTEWORTHY Despite marked coexpression of small (Kcnn1, Kcnn2) and intermediate (Kcnn4) conductance calcium-activated potassium channel transcripts with P2X3 (P2rx3) or bradykinin B2 (Bdkrb2) receptors in colonic sensory neurons, pharmacological activation of these channels had no effect on the colonic afferent response to ATP, bradykinin or luminal distension of the colon. This is in contrast to the robust inhibitory effect of the KV7 channel opener, retigabine.
Collapse
Affiliation(s)
- Charity N Bhebhe
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - James P Higham
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Rohit A Gupta
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Tim Raine
- Department of Gastroenterology, Addenbrookes Hospital, Cambridge University Teaching Hospitals, Cambridge, United Kingdom
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
5
|
Wang L, Yuan PQ, Taché Y. Vasculature in the mouse colon and spatial relationships with the enteric nervous system, glia, and immune cells. Front Neuroanat 2023; 17:1130169. [PMID: 37332321 PMCID: PMC10272736 DOI: 10.3389/fnana.2023.1130169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/15/2023] [Indexed: 06/20/2023] Open
Abstract
The distribution, morphology, and innervation of vasculature in different mouse colonic segments and layers, as well as spatial relationships of the vasculature with the enteric plexuses, glia, and macrophages are far from being complete. The vessels in the adult mouse colon were stained by the cardiovascular perfusion of wheat germ agglutinin (WGA)-Alexa Fluor 448 and by CD31 immunoreactivity. Nerve fibers, enteric glia, and macrophages were immunostained in the WGA-perfused colon. The blood vessels entered from the mesentery to the submucosa and branched into the capillary networks in the mucosa and muscularis externa. The capillary net formed anastomosed rings at the orifices of mucosa crypts, and the capillary rings surrounded the crypts individually in the proximal colon and more than two crypts in the distal colon. Microvessels in the muscularis externa with myenteric plexus were less dense than in the mucosa and formed loops. In the circular smooth muscle layer, microvessels were distributed in the proximal, but not the distal colon. Capillaries did not enter the enteric ganglia. There were no significant differences in microvascular volume per tissue volume between the proximal and distal colon either in the mucosa or muscularis externa containing the myenteric plexus. PGP9.5-, tyrosine hydroxylase-, and calcitonin gene-related peptide (CGRP)-immunoreactive nerve fibers were distributed along the vessels in the submucosa. In the mucosa, PGP9.5-, CGRP-, and vasoactive intestinal peptide (VIP)-immunoreactive nerves terminated close to the capillary rings, while cells and processes labeled by S100B and glial fibrillary acidic protein were distributed mainly in the lamina propria and lower portion of the mucosa. Dense Iba1 immunoreactive macrophages were closely adjacent to the mucosal capillary rings. There were a few macrophages, but no glia in apposition to microvessels in the submucosa and muscularis externa. In conclusion, in the mouse colon, (1) the differences in vasculature between the proximal and distal colon were associated with the morphology, but not the microvascular amount per tissue volume in the mucosa and muscle layers; (2) the colonic mucosa contained significantly more microvessels than the muscularis externa; and (3) there were more CGRP and VIP nerve fibers found close to microvessels in the mucosa and submucosa than in the muscle layers.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Pu-Qing Yuan
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Yvette Taché
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
6
|
Antagonism of the Muscarinic Acetylcholine Type 1 Receptor Enhances Mitochondrial Membrane Potential and Expression of Respiratory Chain Components via AMPK in Human Neuroblastoma SH-SY5Y Cells and Primary Neurons. Mol Neurobiol 2022; 59:6754-6770. [PMID: 36002781 PMCID: PMC9525428 DOI: 10.1007/s12035-022-03003-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/16/2022] [Indexed: 12/05/2022]
Abstract
Impairments in mitochondrial physiology play a role in the progression of multiple neurodegenerative conditions, including peripheral neuropathy in diabetes. Blockade of muscarinic acetylcholine type 1 receptor (M1R) with specific/selective antagonists prevented mitochondrial dysfunction and reversed nerve degeneration in in vitro and in vivo models of peripheral neuropathy. Specifically, in type 1 and type 2 models of diabetes, inhibition of M1R using pirenzepine or muscarinic toxin 7 (MT7) induced AMP-activated protein kinase (AMPK) activity in dorsal root ganglia (DRG) and prevented sensory abnormalities and distal nerve fiber loss. The human neuroblastoma SH-SY5Y cell line has been extensively used as an in vitro model system to study mechanisms of neurodegeneration in DRG neurons and other neuronal sub-types. Here, we tested the hypothesis that pirenzepine or MT7 enhance AMPK activity and via this pathway augment mitochondrial function in SH-SY5Y cells. M1R expression was confirmed by utilizing a fluorescent dye, ATTO590-labeled MT7, that exhibits great specificity for this receptor. M1R antagonist treatment in SH-SY5Y culture increased AMPK phosphorylation and mitochondrial protein expression (OXPHOS). Mitochondrial membrane potential (MMP) was augmented in pirenzepine and MT7 treated cultured SH-SY5Y cells and DRG neurons. Compound C or AMPK-specific siRNA suppressed pirenzepine or MT7-induced elevation of OXPHOS expression and MMP. Moreover, muscarinic antagonists induced hyperpolarization by activating the M-current and, thus, suppressed neuronal excitability. These results reveal that negative regulation of this M1R-dependent pathway could represent a potential therapeutic target to elevate AMPK activity, enhance mitochondrial function, suppress neuropathic pain, and enhance nerve repair in peripheral neuropathy.
Collapse
|
7
|
Lucarini E, Micheli L, Pagnotta E, Toti A, Ferrara V, Ciampi C, Margiotta F, Martelli A, Testai L, Calderone V, Matteo R, Suriano S, Troccoli A, Pecchioni N, Manera C, Mannelli LDC, Ghelardini C. The Efficacy of Camelina sativa Defatted Seed Meal against Colitis-Induced Persistent Visceral Hypersensitivity: The Relevance of PPAR α Receptor Activation in Pain Relief. Nutrients 2022; 14:nu14153137. [PMID: 35956313 PMCID: PMC9370738 DOI: 10.3390/nu14153137] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
Brassicaceae are natural sources of bioactive compounds able to promote gut health. Belonging to this plant family, Camelina sativa is an ancient oil crop rich in glucosinolates, polyunsaturated fatty acids, and antioxidants that is attracting renewed attention for its nutraceutical potential. This work aimed at investigating the therapeutic effects of a defatted seed meal (DSM) of Camelina sativa on the colon damage and the persistent visceral hypersensitivity associated with colitis in rats. Inflammation was induced by the intrarectal injection of 2,4-dinitrobenzenesulfonic acid (DNBS). The acute administration of Camelina sativa DSM (0.1–1 g kg−1) showed a dose-dependent pain-relieving effect in DNBS-treated rats. The efficacy of the meal was slightly enhanced after bioactivation with myrosinase, which increased isothiocyanate availability, and drastically decreased by pre-treating the animals with the selective peroxisome proliferator-activated receptor alpha (PPAR α) receptor antagonist GW6471. Repeated treatments with Camelina sativa DSM (1 g kg−1) meal counteracted the development, as well as the persistence, of visceral hyperalgesia in DNBS-treated animals by reducing the intestinal inflammatory damage and preventing enteric neuron damage. In conclusion, Camelina sativa meal might be employed as a nutraceutical tool to manage persistent abdominal pain in patients and to promote gut healing.
Collapse
Affiliation(s)
- Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research, and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (A.T.); (V.F.); (C.C.); (F.M.); (C.G.)
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research, and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (A.T.); (V.F.); (C.C.); (F.M.); (C.G.)
| | - Eleonora Pagnotta
- CREA—Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, 40128 Bologna, Italy; (E.P.); (R.M.)
| | - Alessandra Toti
- Department of Neuroscience, Psychology, Drug Research, and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (A.T.); (V.F.); (C.C.); (F.M.); (C.G.)
| | - Valentina Ferrara
- Department of Neuroscience, Psychology, Drug Research, and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (A.T.); (V.F.); (C.C.); (F.M.); (C.G.)
| | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research, and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (A.T.); (V.F.); (C.C.); (F.M.); (C.G.)
| | - Francesco Margiotta
- Department of Neuroscience, Psychology, Drug Research, and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (A.T.); (V.F.); (C.C.); (F.M.); (C.G.)
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (A.M.); (L.T.); (V.C.); (C.M.)
- Interdepartmental Research Centre Nutraceuticals and Food for Health—NUTRAFOOD, University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, 56126 Pisa, Italy
| | - Lara Testai
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (A.M.); (L.T.); (V.C.); (C.M.)
- Interdepartmental Research Centre Nutraceuticals and Food for Health—NUTRAFOOD, University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, 56126 Pisa, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (A.M.); (L.T.); (V.C.); (C.M.)
- Interdepartmental Research Centre Nutraceuticals and Food for Health—NUTRAFOOD, University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, 56126 Pisa, Italy
| | - Roberto Matteo
- CREA—Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, 40128 Bologna, Italy; (E.P.); (R.M.)
| | - Serafino Suriano
- CREA—Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, 71122 Foggia, Italy; (S.S.); (A.T.); (N.P.)
| | - Antonio Troccoli
- CREA—Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, 71122 Foggia, Italy; (S.S.); (A.T.); (N.P.)
| | - Nicola Pecchioni
- CREA—Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, 71122 Foggia, Italy; (S.S.); (A.T.); (N.P.)
| | - Clementina Manera
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (A.M.); (L.T.); (V.C.); (C.M.)
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research, and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (A.T.); (V.F.); (C.C.); (F.M.); (C.G.)
- Correspondence:
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research, and Child Health—NEUROFARBA—Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (A.T.); (V.F.); (C.C.); (F.M.); (C.G.)
| |
Collapse
|
8
|
Jaramillo-Polanco J, Lopez-Lopez C, Yu Y, Neary E, Hegron A, Canals M, Bunnett NW, Reed DE, Lomax AE, Vanner SJ. Opioid-Induced Pronociceptive Signaling in the Gastrointestinal Tract Is Mediated by Delta-Opioid Receptor Signaling. J Neurosci 2022; 42:3316-3328. [PMID: 35256532 PMCID: PMC9034783 DOI: 10.1523/jneurosci.2098-21.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/21/2022] Open
Abstract
Opioid tolerance (OT) leads to dose escalation and serious side effects, including opioid-induced hyperalgesia (OIH). We sought to better understand the mechanisms underlying this event in the gastrointestinal tract. Chronic in vivo administration of morphine by intraperitoneal injection in male C57BL/6 mice evoked tolerance and evidence of OIH in an assay of colonic afferent nerve mechanosensitivity; this was inhibited by the δ-opioid receptor (DOPr) antagonist naltrindole when intraperitoneally injected in previous morphine administration. Patch-clamp studies of DRG neurons following overnight incubation with high concentrations of morphine, the µ-opioid receptors (MOPr) agonist [D-Ala2, N-Me-Phe4, Gly5-ol]-Enkephalin (DAMGO) or the DOPr agonist [D-Ala2, D-Leu5]-Enkephalin evoked hyperexcitability. The pronociceptive actions of these opioids were blocked by the DOPr antagonist SDM25N but not the MOPr antagonist D-Pen-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 The hyperexcitability induced by DAMGO was reversed after a 1 h washout, but reapplication of low concentrations of DAMGO or [D-Ala2, D-Leu5]-Enkephalin restored the hyperexcitability, an effect mediated by protein kinase C. DOPr-dependent DRG neuron hyperexcitability was blocked by the endocytosis inhibitor Pitstop 2, and the weakly internalizing DOPr agonist ARM390 did not cause hyperexcitability. Bioluminescence resonance energy transfer studies in HEK cells showed no evidence of switching of G-protein signaling from Gi to a Gs pathway in response to either high concentrations or overnight incubation of opioids. Thus, chronic high-dose opioid exposure leads to opioid tolerance and features of OIH in the colon. This action is mediated by DOPr signaling and is dependent on receptor endocytosis and downstream protein kinase C signaling.SIGNIFICANCE STATEMENT Opioids are effective in the treatment of abdominal pain, but escalating doses can lead to opioid tolerance and potentially opioid-induced hyperalgesia. We found that δ-opioid receptor (DOPr) plays a central role in the development of opioid tolerance and opioid-induced hyperalgesia in colonic afferent nociceptors following prolonged exposure to high concentrations of MOPr or DOPr agonists. Furthermore, the role of DOPr was dependent on OPr internalization and activation of a protein kinase C signaling pathway. Thus, targeting DOPr or key components of the downstream signaling pathway could mitigate adverse side effects by opioids.
Collapse
Affiliation(s)
- Josue Jaramillo-Polanco
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, Ontario K7L 2V7, Canada
| | - Cintya Lopez-Lopez
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, Ontario K7L 2V7, Canada
| | - Yang Yu
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, Ontario K7L 2V7, Canada
| | - Emma Neary
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, Ontario K7L 2V7, Canada
| | - Alan Hegron
- Department of Molecular Pathobiology, Department of Neuroscience and Physiology, Neuroscience Institute, New York University, New York, New York 10010
| | - Meritxell Canals
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, United Kingdom
- Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, Nottingham, NG7 2UH, United Kingdom
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, Department of Neuroscience and Physiology, Neuroscience Institute, New York University, New York, New York 10010
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, Ontario K7L 2V7, Canada
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, Ontario K7L 2V7, Canada
| | - Stephen J Vanner
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, Ontario K7L 2V7, Canada
| |
Collapse
|
9
|
Lucarini E, Micheli L, Pagnotta E, Matteo R, Parisio C, Toti A, Ferrara V, Ciampi C, Martelli A, Testai L, Calderone V, Savino M, Russo M, Pecchioni N, Ghelardini C, Di Cesare Mannelli L. Beneficial Effects of Eruca sativa Defatted Seed Meal on Visceral Pain and Intestinal Damage Resulting from Colitis in Rats. Foods 2022; 11:foods11040580. [PMID: 35206057 PMCID: PMC8870774 DOI: 10.3390/foods11040580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/04/2022] [Accepted: 02/11/2022] [Indexed: 02/07/2023] Open
Abstract
Most therapies used in patients affected by inflammatory bowel diseases are ineffective in preventing the development of chronic visceral hypersensitivity, mainly due to inflammation-induced enteric neuroplasticity. Glucosinolates, secondary metabolites mainly of Brassicaceae with anti-inflammatory and neuroprotective properties, are effective in treating both neuropathic and arthritis pain through H2S release and Kv7 potassium channel activation. The aim of this work was to investigate the protective and anti-hyperalgesic efficacy of a defatted seed meal from Eruca sativa Mill. (Brassicaceae), rich in glucosinolates, in a rat model of colitis induced by 2,4-dinitrobenzene sulfonic acid (DNBS). The mechanisms of action were also investigated. Visceral pain was assessed by measuring the abdominal response to colorectal distension. Fifteen days after colitis induction, the acute administration of E. sativa defatted seed meal (0.1–1 g kg−1 p.o.) dose-dependently relieved pain. This effect was hampered by co-administering an H2S scavenger or a selective Kv7 blocker. Administering E. sativa (1 g kg−1) for 14 days, starting after DNBS injection, contributed to counteracting visceral pain persistence in the post-inflammatory phase of colitis by promoting colon healing from the damage and reducing enteric gliosis. E. sativa defatted seed meal might be employed as a nutraceutical tool for supporting abdominal pain relief in patients.
Collapse
Affiliation(s)
- Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (C.P.); (A.T.); (V.F.); (C.C.); (C.G.)
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (C.P.); (A.T.); (V.F.); (C.C.); (C.G.)
| | - Eleonora Pagnotta
- CREA-Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, 40128 Bologna, Italy; (E.P.); (R.M.)
| | - Roberto Matteo
- CREA-Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, 40128 Bologna, Italy; (E.P.); (R.M.)
| | - Carmen Parisio
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (C.P.); (A.T.); (V.F.); (C.C.); (C.G.)
| | - Alessandra Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (C.P.); (A.T.); (V.F.); (C.C.); (C.G.)
| | - Valentina Ferrara
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (C.P.); (A.T.); (V.F.); (C.C.); (C.G.)
| | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (C.P.); (A.T.); (V.F.); (C.C.); (C.G.)
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (A.M.); (L.T.); (V.C.)
- Interdepartmental Research Centre Nutraceuticals and Food for Health (NUTRAFOOD), University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, 56126 Pisa, Italy
| | - Lara Testai
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (A.M.); (L.T.); (V.C.)
- Interdepartmental Research Centre Nutraceuticals and Food for Health (NUTRAFOOD), University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, 56126 Pisa, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (A.M.); (L.T.); (V.C.)
- Interdepartmental Research Centre Nutraceuticals and Food for Health (NUTRAFOOD), University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, 56126 Pisa, Italy
| | - Michele Savino
- CREA-Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, 71122 Foggia, Italy; (M.S.); (M.R.); (N.P.)
| | - Mario Russo
- CREA-Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, 71122 Foggia, Italy; (M.S.); (M.R.); (N.P.)
| | - Nicola Pecchioni
- CREA-Council for Agricultural Research and Economics, Research Centre for Cereal and Industrial Crops, 71122 Foggia, Italy; (M.S.); (M.R.); (N.P.)
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (C.P.); (A.T.); (V.F.); (C.C.); (C.G.)
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy; (E.L.); (L.M.); (C.P.); (A.T.); (V.F.); (C.C.); (C.G.)
- Correspondence:
| |
Collapse
|
10
|
Lu Y, Huang J, Zhang Y, Huang Z, Yan W, Zhou T, Wang Z, Liao L, Cao H, Tan B. Therapeutic Effects of Berberine Hydrochloride on Stress-Induced Diarrhea-Predominant Irritable Bowel Syndrome Rats by Inhibiting Neurotransmission in Colonic Smooth Muscle. Front Pharmacol 2021; 12:596686. [PMID: 34594213 PMCID: PMC8476869 DOI: 10.3389/fphar.2021.596686] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/01/2021] [Indexed: 11/13/2022] Open
Abstract
The etiology of diarrhea-predominant irritable bowel syndrome (IBS-D) is complicated and closely related to neurotransmission in the gastrointestinal (GI) tract. Developing new strategies for treating this disease is a major challenge for IBS-D research. Berberine hydrochloride (BBH), the derivative of berberine, is a herbal constituent used to treat IBS. Previous studies have shown that BBH has potential anti-inflammatory, antibacterial, analgesic, and antidiarrheal effects and a wide range of biological activities, especially in regulating the release of some neurotransmitters. A modified IBS-D rat model induced by chronic restraint stress was used in all experiments to study the effects of BBH on the GI tract. This study measured the abdominal withdrawal reflex (AWR) response to graded colorectal distention (CRD; 20, 40, 60, and 80 mmHg) and observed the fecal areas of stress-induced IBS-D model. Experiments were conducted using organ bath techniques, which were performed in vitro using strips of colonic longitudinal smooth muscle. Inhibitory and excitatory neurotransmitter agents were added to each organ bath to observe contractile responses on the strips and the treatment effect exerted by BBH. The IBS-D rat model was successfully induced by chronic restraint stress, which resulted in an increased defecation frequency and visceral hypersensitivity similar to that of humans. BBH could reduce 4-h fecal areas and AWR response to CRD in IBS-D. The stress-induced IBS-D model showed upregulated colonic mRNA expression levels of 5-hydroxytryptamine-3A receptor and downregulated expression levels of neuronal nitric oxide synthase. Meanwhile, BBH could reverse this outcome. The responses of substances that regulate the contraction induced by related neurotransmission in the longitudinal smooth muscle of IBS-D colon (including the agonist of acetylcholine, carbachol; NOS inhibitor, L-NAME; and P2Y1 receptor antagonist, MRS2500) can be inhibited by BBH. In summary, BBH promotes defecation frequency and visceral hypersensitivity in IBS-D and exerts inhibitory effects on contractile responses in colonic longitudinal smooth muscle. Thus, BBH may represent a new therapeutic approach for treating IBS-D.
Collapse
Affiliation(s)
- Yulin Lu
- Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjing Huang
- Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yao Zhang
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zitong Huang
- Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiming Yan
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianran Zhou
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhesheng Wang
- Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lu Liao
- Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongying Cao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo Tan
- Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
11
|
Beck VC, Isom LL, Berg AT. Gastrointestinal Symptoms and Channelopathy-Associated Epilepsy. J Pediatr 2021; 237:41-49.e1. [PMID: 34181986 PMCID: PMC8478841 DOI: 10.1016/j.jpeds.2021.06.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/28/2021] [Accepted: 06/11/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To determine the prevalence of and identify factors associated with gastrointestinal (GI) symptoms among children with channelopathy-associated developmental and epileptic encephalopathy (DEE). STUDY DESIGN Parents of 168 children with DEEs linked to SCN1A (n = 59), KCNB1 (n = 31), or KCNQ2 (n = 78) completed online CLIRINX surveys about their children's GI symptoms. Our analysis examined the prevalence, frequency, and severity of GI symptoms, as well as DEE type, functional mobility, feeding difficulties, ketogenic diet, antiseizure medication, autism spectrum disorder (ASD), and seizures. Statistical analyses included the χ2 test, Wilcoxon rank-sum analysis, and multiple logistic regression. RESULTS GI symptoms were reported in 92 of 168 patients (55%), among whom 63 of 86 (73%) reported daily or weekly symptoms, 29 of 92 (32%) had frequent or serious discomfort, and 13 of 91 (14%) had frequent or serious appetite disturbances as a result. The prevalence of GI symptoms varied across DEE cohorts with 44% of SCN1A-DEE patients, 35% of KCNB1-DEE patients, and 71% of KCNQ2-DEE patients reporting GI symptoms in the previous month. After adjustment for DEE type, current use of ketogenic diet (6% reported), and gastrostomy tube (13% reported) were both associated with GI symptoms in a statistically, but not clinically, significant manner (P < .05). Patient age, functional mobility, feeding difficulties, ASD, and seizures were not clearly associated with GI symptoms. Overall, no individual antiseizure medication was significantly associated with GI symptoms across all DEE cohorts. CONCLUSIONS GI symptoms are common and frequently severe in patients with DEE.
Collapse
Affiliation(s)
- Veronica C Beck
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI; Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Lori L Isom
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI; Department of Pharmacology, University of Michigan, Ann Arbor, MI; Department of Neurology, University of Michigan, Ann Arbor, MI; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| | - Anne T Berg
- Division of Neurology, Epilepsy Center, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL; Department of Pediatrics, Northwestern Feinberg School of Medicine, Chicago, IL.
| |
Collapse
|
12
|
Nickerson AJ, Rajendran VM. Flupirtine enhances NHE-3-mediated Na + absorption in rat colon via an ENS-dependent mechanism. Am J Physiol Gastrointest Liver Physiol 2021; 321:G185-G199. [PMID: 34132108 PMCID: PMC8410105 DOI: 10.1152/ajpgi.00158.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 01/31/2023]
Abstract
Recent studies in our lab have shown that the KV7 channel activator, flupirtine, inhibits colonic epithelial Cl- secretion through effects on submucosal neurons of the enteric nervous system (ENS). We hypothesized that flupirtine would also stimulate Na+ absorption as a result of reduced secretory ENS input to the epithelium. To test this hypothesis, unidirectional 22Na+ fluxes were measured under voltage-clamped conditions. Pharmacological approaches using an Ussing-style recording chamber combined with immunofluorescence microscopy techniques were used to determine the effect of flupirtine on active Na+ transport in the rat colon. Flupirtine stimulated electroneutral Na+ absorption in partially seromuscular-stripped colonic tissues, while simultaneously inhibiting short-circuit current (ISC; i.e., Cl- secretion). Both of these effects were attenuated by pretreatment with the ENS inhibitor, tetrodotoxin. The Na+/H+ exchanger isoform 3 (NHE-3)-selective inhibitor, S3226, significantly inhibited flupirtine-stimulated Na+ absorption, whereas the NHE-2-selective inhibitor HOE-694 did not. NHE-3 localization near the apical membranes of surface epithelial cells was also more apparent in flupirtine-treated colon versus control. Flupirtine did not alter epithelial Na+ channel (ENaC)-mediated Na+ absorption in distal colonic tissues obtained from hyperaldosteronaemic rats and had no effect in the normal ileum but did stimulate Na+ absorption in the proximal colon. Finally, the parallel effects of flupirtine on ISC (Cl- secretion) and Na+ absorption were significantly correlated with each other. Together, these data indicate that flupirtine stimulates NHE-3-dependent Na+ absorption, likely as a result of reduced stimulatory input to the colonic epithelium by submucosal ENS neurons.NEW & NOTEWORTHY We present a novel mechanism regarding regulation of epithelial ion transport by enteric neurons. Activation of neuronal KV7 K+ channels markedly stimulates Na+ absorption and inhibits Cl- secretion across the colonic epithelium. This may be useful in developing new treatments for diarrheal disorders, such as irritable bowel syndrome with diarrhea (IBS-D).
Collapse
Affiliation(s)
- Andrew J Nickerson
- Departments of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Vazhaikkurichi M Rajendran
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Medicine, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
13
|
Nickerson AJ, Rottgen TS, Rajendran VM. Activation of KCNQ (K V7) K + channels in enteric neurons inhibits epithelial Cl - secretion in mouse distal colon. Am J Physiol Cell Physiol 2021; 320:C1074-C1087. [PMID: 33852365 PMCID: PMC8285638 DOI: 10.1152/ajpcell.00536.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022]
Abstract
Voltage-gated Kv7 (KCNQ family) K+ channels are expressed in many neuronal populations and play an important role in regulating membrane potential by generating a hyperpolarizing K+ current and decreasing cell excitability. However, the role of KV7 channels in the neural regulation of intestinal epithelial Cl- secretion is not known. Cl- secretion in mouse distal colon was measured as a function of short-circuit current (ISC), and pharmacological approaches were used to test the hypothesis that activation of KV7 channels in enteric neurons would inhibit epithelial Cl- secretion. Flupirtine, a nonselective KV7 activator, inhibited basal Cl- secretion in mouse distal colon and abolished or attenuated the effects of drugs that target various components of enteric neurotransmission, including tetrodotoxin (NaV channel blocker), veratridine (NaV channel activator), nicotine (nicotinic acetylcholine receptor agonist), and hexamethonium (nicotinic antagonist). In contrast, flupritine did not block the response to epithelium-targeted agents VIP (endogenous VPAC receptor ligand) or carbachol (nonselective cholinergic agonist). Flupirtine inhibited Cl- secretion in both full-thickness and seromuscular-stripped distal colon (containing the submucosal, but not myenteric plexus) but generated no response in epithelial T84 cell monolayers. KV7.2 and KV7.3 channel proteins were detected by immunofluorescence in whole mount preparations of the submucosa from mouse distal colon. ICA 110381 (KV7.2/7.3 specific activator) inhibited Cl- secretion comparably to flupirtine. We conclude that KV7 channel activators inhibit neurally driven Cl- secretion in the colonic epithelium and may therefore have therapeutic benefit in treating pathologies associated with hyperexcitable enteric nervous system, such as irritable bowel syndrome with diarrhea (IBS-D).
Collapse
Affiliation(s)
- Andrew J Nickerson
- Departments of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Trey S Rottgen
- Departments of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Vazhaikkurichi M Rajendran
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Medicine, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
14
|
Jalanka J, Lam C, Bennett A, Hartikainen A, Crispie F, Finnegan LA, Cotter PD, Spiller R. Colonic Gene Expression and Fecal Microbiota in Diarrhea-predominant Irritable Bowel Syndrome: Increased Toll-like Receptor 4 but Minimal Inflammation and no Response to Mesalazine. J Neurogastroenterol Motil 2021; 27:279-291. [PMID: 33795545 PMCID: PMC8026366 DOI: 10.5056/jnm20205] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/23/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022] Open
Abstract
Background/Aims Diarrhea-predominant irritable bowel syndrome (IBS-D) has been previously associated with evidence of immune activation and altered microbiota. Our aim is to assess the effect of the anti-inflammatory agent, mesalazine, on inflammatory gene expression and microbiota composition in IBS-D. Methods We studied a subset of patients (n = 43) from a previously published 12-week radomized placebo-controlled trial of mesalazine. Mucosal biopsies were assessed by immunohistochemistry and reverse transcription-polymerase chain reaction for a range of markers of inflammation, altered permeability, and sensory receptors including Toll-like receptors (TLRs) at randomization after treatment. All biopsy data were compared to 21 healthy controls. Patient’s stool microbiota composition was analysed through 16S ribosomal RNA sequencing. Results We found no evidence of increased immune activation compared to healthy controls. However, we did find increased expression of receptors in both sensory pathways and innate immune response including TLR4. Higher TLR4 expression was associated with greater urgency. TLR4 expression correlated strongly with the expression of the receptors bradykinin receptor B2, chemerin chemokine-like receptor 1, and transient receptor potential cation channel, subfamily A, member 1 as well as TLR4’s downstream adaptor myeloid differentiation factor 88. Mesalazine had minimal effect on either gene expression or microbiota composition. Conclusions Biopsies from a well-characterized IBS-D cohort showed no substantial inflammation. Mesalazine has little effect on gene expression and its previous reported effect on fecal microbiota associated with much greater inflammation found in inflammatory bowel diseases is likely secondary to reduced inflammation. Increased expression of TLR4 and correlated receptors in IBS may mediate a general increase in sensitivity to external stimuli, particularly those that signal via the TLR system.
Collapse
Affiliation(s)
- Jonna Jalanka
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Nottingham Digestive Diseases Center and NIHR Nottingham Biomedical Research Center at Nottingham University Hospitals NHS Trust, the University of Nottingham, Nottingham, Notts, UK
| | - Ching Lam
- Nottingham Digestive Diseases Center and NIHR Nottingham Biomedical Research Center at Nottingham University Hospitals NHS Trust, the University of Nottingham, Nottingham, Notts, UK
| | - Andrew Bennett
- Nottingham Digestive Diseases Center and NIHR Nottingham Biomedical Research Center at Nottingham University Hospitals NHS Trust, the University of Nottingham, Nottingham, Notts, UK.,FRAME Alternatives Laboratory, School of Life Sciences, University of Nottingham, Medical School, QMC, Nottingham, Notts, UK
| | - Anna Hartikainen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Fiona Crispie
- Teagasc Food Research Center, Moorepark, Fermoy, Co. Cork, Ireland.,APC Microbiome Ireland, Cork, Ireland
| | - Laura A Finnegan
- Teagasc Food Research Center, Moorepark, Fermoy, Co. Cork, Ireland.,APC Microbiome Ireland, Cork, Ireland
| | - Paul D Cotter
- Teagasc Food Research Center, Moorepark, Fermoy, Co. Cork, Ireland.,APC Microbiome Ireland, Cork, Ireland
| | - Robin Spiller
- Nottingham Digestive Diseases Center and NIHR Nottingham Biomedical Research Center at Nottingham University Hospitals NHS Trust, the University of Nottingham, Nottingham, Notts, UK
| |
Collapse
|
15
|
Lee MC, Nahorski MS, Hockley JRF, Lu VB, Ison G, Pattison LA, Callejo G, Stouffer K, Fletcher E, Brown C, Drissi I, Wheeler D, Ernfors P, Menon D, Reimann F, Smith ESJ, Woods CG. Human Labor Pain Is Influenced by the Voltage-Gated Potassium Channel K V6.4 Subunit. Cell Rep 2021; 32:107941. [PMID: 32697988 PMCID: PMC7383234 DOI: 10.1016/j.celrep.2020.107941] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/19/2020] [Accepted: 06/30/2020] [Indexed: 12/22/2022] Open
Abstract
By studying healthy women who do not request analgesia during their first delivery, we investigate genetic effects on labor pain. Such women have normal sensory and psychometric test results, except for significantly higher cuff pressure pain. We find an excess of heterozygotes carrying the rare allele of SNP rs140124801 in KCNG4. The rare variant KV6.4-Met419 has a dominant-negative effect and cannot modulate the voltage dependence of KV2.1 inactivation because it fails to traffic to the plasma membrane. In vivo, Kcng4 (KV6.4) expression occurs in 40% of retrograde-labeled mouse uterine sensory neurons, all of which express KV2.1, and over 90% express the nociceptor genes Trpv1 and Scn10a. In neurons overexpressing KV6.4-Met419, the voltage dependence of inactivation for KV2.1 is more depolarized compared with neurons overexpressing KV6.4. Finally, KV6.4-Met419-overexpressing neurons have a higher action potential threshold. We conclude that KV6.4 can influence human labor pain by modulating the excitability of uterine nociceptors. KCNG4 variant highly prevalent in women requiring no analgesia in childbirth KCNG4 variant encodes KV6.4Met-419; KV6.4 is a silent subunit modifying KV activity KV6.4Met-419 is retained in the cytoplasm and acts in a dominant-negative manner KV6.4Met-419 overexpression results in hypoexcitable sensory neurons
Collapse
Affiliation(s)
- Michael C Lee
- University Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.
| | - Michael S Nahorski
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - James R F Hockley
- Department of Pharmacology, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Van B Lu
- Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Gillian Ison
- University Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Luke A Pattison
- Department of Pharmacology, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Gerard Callejo
- Department of Pharmacology, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Kaitlin Stouffer
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Emily Fletcher
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Christopher Brown
- Department of Psychological Sciences, Institute of Psychology, Health and Society, University of Liverpool, Liverpool L69 7ZA, UK
| | - Ichrak Drissi
- University Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Daniel Wheeler
- University Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Patrik Ernfors
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - David Menon
- University Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Frank Reimann
- Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.
| | | | - C Geoffrey Woods
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.
| |
Collapse
|
16
|
Lawson K, Singh A, Kantsedikas I, Jenner CA, Austen DK. Flupirtine as a Potential Treatment for Fibromyalgia. JOURNAL OF EXPLORATORY RESEARCH IN PHARMACOLOGY 2021; 000:000-000. [DOI: 10.14218/jerp.2020.00043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
17
|
Hockley JR, Barker KH, Taylor TS, Callejo G, Husson ZM, Bulmer DC, Smith ESJ. Acid and inflammatory sensitisation of naked mole-rat colonic afferent nerves. Mol Pain 2020; 16:1744806920903150. [PMID: 31992138 PMCID: PMC6990608 DOI: 10.1177/1744806920903150] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Acid sensing in the gastrointestinal tract is required for gut homeostasis and the detection of tissue acidosis caused by ischaemia, inflammation and infection. In the colorectum, activation of colonic afferents by low pH contributes to visceral hypersensitivity and abdominal pain in human disease including during inflammatory bowel disease. The naked mole-rat (Heterocephalus glaber) shows no pain-related behaviour to subcutaneous acid injection and cutaneous afferents are insensitive to acid, an adaptation thought to be a consequence of the subterranean, likely hypercapnic, environment in which it lives. As such we sought to investigate naked mole-rat interoception within the gastrointestinal tract and how this differed from the mouse (Mus Musculus). Here, we show the presence of calcitonin gene-related peptide expressing extrinsic nerve fibres innervating both mesenteric blood vessels and the myenteric plexi of the smooth muscle layers of the naked mole-rat colorectum. Using ex vivo colonic-nerve electrophysiological recordings, we show differential sensitivity of naked mole-rat, compared to mouse, colonic afferents to acid and the prototypic inflammatory mediator bradykinin, but not direct mechanical stimuli. In naked mole-rat, but not mouse, we observed mechanical hypersensitivity to acid, whilst both species sensitised to bradykinin. Collectively, these findings suggest that naked mole-rat colonic afferents are capable of detecting acidic stimuli; however, their intracellular coupling to downstream molecular effectors of neuronal excitability and mechanotransduction likely differs between species.
Collapse
Affiliation(s)
- James Rf Hockley
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Katie H Barker
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Toni S Taylor
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Gerard Callejo
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Zoe M Husson
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Ewan St J Smith
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
18
|
Johnson AC, Louwies T, Ligon CO, Greenwood-Van Meerveld B. Enlightening the frontiers of neurogastroenterology through optogenetics. Am J Physiol Gastrointest Liver Physiol 2020; 319:G391-G399. [PMID: 32755304 PMCID: PMC7717115 DOI: 10.1152/ajpgi.00384.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neurogastroenterology refers to the study of the extrinsic and intrinsic nervous system circuits controlling the gastrointestinal (GI) tract. Over the past 5-10 yr there has been an explosion in novel methodologies, technologies and approaches that offer great promise to advance our understanding of the basic mechanisms underlying GI function in health and disease. This review focuses on the use of optogenetics combined with electrophysiology in the field of neurogastroenterology. We discuss how these technologies and tools are currently being used to explore the brain-gut axis and debate the future research potential and limitations of these techniques. Taken together, we consider that the use of these technologies will enable researchers to answer important questions in neurogastroenterology through fundamental research. The answers to those questions will shorten the path from basic discovery to new treatments for patient populations with disorders of the brain-gut axis affecting the GI tract such as irritable bowel syndrome (IBS), functional dyspepsia, achalasia, and delayed gastric emptying.
Collapse
Affiliation(s)
- Anthony C. Johnson
- 1Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma,2Oklahoma City Veterans Affairs Health Care System, Oklahoma City, Oklahoma,3Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Tijs Louwies
- 1Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Casey O. Ligon
- 1Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Beverley Greenwood-Van Meerveld
- 1Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma,2Oklahoma City Veterans Affairs Health Care System, Oklahoma City, Oklahoma,4Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
19
|
Yu Y, Villalobos-Hernandez EC, Pradhananga S, Baker CC, Keating C, Grundy D, Lomax AE, Reed DE. Deoxycholic acid activates colonic afferent nerves via 5-HT 3 receptor-dependent and -independent mechanisms. Am J Physiol Gastrointest Liver Physiol 2019; 317:G275-G284. [PMID: 31216174 DOI: 10.1152/ajpgi.00016.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Increased bile acids in the colon can evoke increased epithelial secretion resulting in diarrhea, but little is known about whether colonic bile acids contribute to abdominal pain. This study aimed to investigate the mechanisms underlying activation of colonic extrinsic afferent nerves and their neuronal cell bodies by a major secondary bile acid, deoxycholic acid (DCA). All experiments were performed on male C57BL/6 mice. Afferent sensitivity was evaluated using in vitro extracellular recordings from mesenteric nerves in the proximal colon (innervated by vagal and spinal afferents) and distal colon (spinal afferents only). Neuronal excitability of cultured dorsal root ganglion (DRG) and nodose ganglion (NG) neurons was examined with perforated patch clamp. Colonic 5-HT release was assessed using ELISA, and 5-HT immunoreactive enterochromaffin (EC) cells were quantified. Intraluminal DCA increased afferent nerve firing rate concentration dependently in both proximal and distal colon. This DCA-elicited increase was significantly inhibited by a 5-HT3 antagonist in the proximal colon but not in the distal colon, which may be in part due to lower 5-HT immunoreactive EC cell density and lower 5-HT levels in the distal colon following DCA stimulation. DCA increased the excitability of DRG neurons, whereas it decreased the excitability of NG neurons. DCA potentiated mechanosensitivity of high-threshold spinal afferents independent of 5-HT release. Together, this study suggests that DCA can excite colonic afferents via direct and indirect mechanisms but the predominant mechanism may differ between vagal and spinal afferents. Furthermore, DCA increased mechanosensitivity of high-threshold spinal afferents and may be a mechanism of visceral hypersensitivity.NEW & NOTEWORTHY Deoxycholic acid (DCA) directly excites spinal afferents and, to a lesser extent, indirectly via mucosal 5-HT release. DCA potentiates mechanosensitivity of high-threshold spinal afferents independent of 5-HT release. DCA increases vagal afferent firing in proximal colon via 5-HT release but directly inhibits the excitability of their cell bodies.
Collapse
Affiliation(s)
- Yang Yu
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | - Sabindra Pradhananga
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Corey C Baker
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Christopher Keating
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - David Grundy
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
20
|
Grayson M, Furr A, Ruparel S. Depiction of Oral Tumor-Induced Trigeminal Afferent Responses Using Single-Fiber Electrophysiology. Sci Rep 2019; 9:4574. [PMID: 30872649 PMCID: PMC6418205 DOI: 10.1038/s41598-019-39824-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/22/2019] [Indexed: 02/02/2023] Open
Abstract
Considerable gap in knowledge exists about the mechanisms by which oral tumors regulate peripheral sensory fibers to produce pain and altered sensations. To address this gap, we used a murine model of oral squamous cell carcinoma (OSCC) of the tongue to investigate changes in response properties of trigeminal afferent neurons. Using this model, we developed an ex vivo method for single neuron recordings of the lingual nerve from isolated tongue tissue. Our data demonstrated that the tongue tumor produced increased spontaneous firing of lingual fibers compared to control as well as produced mechanical hypersensitivity and reduced von Frey thresholds of C- and A-slow-high-threshold mechanoreceptors (HTMR) fibers but had no effect on C-LTMR, A-slow-LTMR and A-fast lingual fibers. Mechanically-insensitive fibers were also detected in lingual afferents of the control group, that were significantly decreased in tumor-bearing preparations. Collectively, using single fiber electrophysiology of lingual sensory fibers, we show that human OSCC tumors sensitize peripheral trigeminal nerve terminals, providing a unique opportunity to study mechanisms of oral cancer pain.
Collapse
Affiliation(s)
- Max Grayson
- Department of Endodontics, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Ashley Furr
- Department of Endodontics, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Shivani Ruparel
- Department of Endodontics, University of Texas Health at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
21
|
Sun H, Lin AH, Ru F, Patil MJ, Meeker S, Lee LY, Undem BJ. KCNQ/M-channels regulate mouse vagal bronchopulmonary C-fiber excitability and cough sensitivity. JCI Insight 2019; 4:124467. [PMID: 30721152 DOI: 10.1172/jci.insight.124467] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/29/2019] [Indexed: 01/06/2023] Open
Abstract
Increased airway vagal sensory C-fiber activity contributes to the symptoms of inflammatory airway diseases. The KCNQ/Kv7/M-channel is a well-known determinant of neuronal excitability, yet whether it regulates the activity of vagal bronchopulmonary C-fibers and airway reflex sensitivity remains unknown. Here we addressed this issue using single-cell RT-PCR, patch clamp technique, extracellular recording of single vagal nerve fibers innervating the mouse lungs, and telemetric recording of cough in free-moving mice. Single-cell mRNA analysis and biophysical properties of M-current (IM) suggest that KCNQ3/Kv7.3 is the major M-channel subunit in mouse nodose neurons. The M-channel opener retigabine negatively shifted the voltage-dependent activation of IM, leading to membrane hyperpolarization, increased rheobase, and suppression of both evoked and spontaneous action potential (AP) firing in nodose neurons in an M-channel inhibitor XE991-sensitive manner. Retigabine also markedly suppressed the α,β-methylene ATP-induced AP firing in nodose C-fiber terminals innervating the mouse lungs, and coughing evoked by irritant gases in awake mice. In conclusion, KCNQ/M-channels play a role in regulating the excitability of vagal airway C-fibers at both the cell soma and nerve terminals. Drugs that open M-channels in airway sensory afferents may relieve the sufferings associated with pulmonary inflammatory diseases such as chronic coughing.
Collapse
Affiliation(s)
- Hui Sun
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - An-Hsuan Lin
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Fei Ru
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mayur J Patil
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sonya Meeker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lu-Yuan Lee
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Bradley J Undem
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Abstract
Most of us live blissfully unaware of the orchestrated function that our internal organs conduct. When this peace is interrupted, it is often by routine sensations of hunger and urge. However, for >20% of the global population, chronic visceral pain is an unpleasant and often excruciating reminder of the existence of our internal organs. In many cases, there is no obvious underlying pathological cause of the pain. Accordingly, chronic visceral pain is debilitating, reduces the quality of life of sufferers, and has large concomitant socioeconomic costs. In this review, we highlight key mechanisms underlying chronic abdominal and pelvic pain associated with functional and inflammatory disorders of the gastrointestinal and urinary tracts. This includes how the colon and bladder are innervated by specialized subclasses of spinal afferents, how these afferents become sensitized in highly dynamic signaling environments, and the subsequent development of neuroplasticity within visceral pain pathways. We also highlight key contributing factors, including alterations in commensal bacteria, altered mucosal permeability, epithelial interactions with afferent nerves, alterations in immune or stress responses, and cross talk between these two adjacent organs.
Collapse
Affiliation(s)
- Luke Grundy
- Visceral Pain Research Group, College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Bedford Park, South Australia 5042, Australia; .,Centre for Nutrition and Gastrointestinal Diseases, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia 5000, Australia
| | - Andelain Erickson
- Visceral Pain Research Group, College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Bedford Park, South Australia 5042, Australia; .,Centre for Nutrition and Gastrointestinal Diseases, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia 5000, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Bedford Park, South Australia 5042, Australia; .,Centre for Nutrition and Gastrointestinal Diseases, University of Adelaide, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia 5000, Australia
| |
Collapse
|
23
|
Bautzova T, Hockley JRF, Perez-Berezo T, Pujo J, Tranter MM, Desormeaux C, Barbaro MR, Basso L, Le Faouder P, Rolland C, Malapert P, Moqrich A, Eutamene H, Denadai-Souza A, Vergnolle N, Smith ESJ, Hughes DI, Barbara G, Dietrich G, Bulmer DC, Cenac N. 5-oxoETE triggers nociception in constipation-predominant irritable bowel syndrome through MAS-related G protein-coupled receptor D. Sci Signal 2018; 11:eaal2171. [PMID: 30563864 PMCID: PMC6411128 DOI: 10.1126/scisignal.aal2171] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Irritable bowel syndrome (IBS) is a common gastrointestinal disorder that is characterized by chronic abdominal pain concurrent with altered bowel habit. Polyunsaturated fatty acid (PUFA) metabolites are increased in abundance in IBS and are implicated in the alteration of sensation to mechanical stimuli, which is defined as visceral hypersensitivity. We sought to quantify PUFA metabolites in patients with IBS and evaluate their role in pain. Quantification of PUFA metabolites by mass spectrometry in colonic biopsies showed an increased abundance of 5-oxoeicosatetraenoic acid (5-oxoETE) only in biopsies taken from patients with IBS with predominant constipation (IBS-C). Local administration of 5-oxoETE to mice induced somatic and visceral hypersensitivity to mechanical stimuli without causing tissue inflammation. We found that 5-oxoETE directly acted on both human and mouse sensory neurons as shown by lumbar splanchnic nerve recordings and Ca2+ imaging of dorsal root ganglion (DRG) neurons. We showed that 5-oxoETE selectively stimulated nonpeptidergic, isolectin B4 (IB4)-positive DRG neurons through a phospholipase C (PLC)- and pertussis toxin-dependent mechanism, suggesting that the effect was mediated by a G protein-coupled receptor (GPCR). The MAS-related GPCR D (Mrgprd) was found in mouse colonic DRG afferents and was identified as being implicated in the noxious effects of 5-oxoETE. Together, these data suggest that 5-oxoETE, a potential biomarker of IBS-C, induces somatic and visceral hyperalgesia without inflammation in an Mrgprd-dependent manner. Thus, 5-oxoETE may play a pivotal role in the abdominal pain associated with IBS-C.
Collapse
Affiliation(s)
- Tereza Bautzova
- INSERM, UMR1220, IRSD, Université de Toulouse, INRA, ENVT, UPS, Toulouse, France
| | - James R F Hockley
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB1 2PD, UK
- National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AJ, UK
| | - Teresa Perez-Berezo
- INSERM, UMR1220, IRSD, Université de Toulouse, INRA, ENVT, UPS, Toulouse, France
| | - Julien Pujo
- INSERM, UMR1220, IRSD, Université de Toulouse, INRA, ENVT, UPS, Toulouse, France
| | - Michael M Tranter
- National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AJ, UK
| | - Cleo Desormeaux
- INSERM, UMR1220, IRSD, Université de Toulouse, INRA, ENVT, UPS, Toulouse, France
| | | | - Lilian Basso
- INSERM, UMR1220, IRSD, Université de Toulouse, INRA, ENVT, UPS, Toulouse, France
| | - Pauline Le Faouder
- INSERM UMR1048, Lipidomic Core Facility, Metatoul Platform, Université de Toulouse, Toulouse, France
| | - Corinne Rolland
- INSERM, UMR1220, IRSD, Université de Toulouse, INRA, ENVT, UPS, Toulouse, France
| | - Pascale Malapert
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, Marseille, France
| | - Aziz Moqrich
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, Marseille, France
| | - Helene Eutamene
- Neuro-Gastroenterology and Nutrition Team, UMR 1331, INRA Toxalim, INP-EI-Purpan, Université de Toulouse, Toulouse, France
| | | | - Nathalie Vergnolle
- INSERM, UMR1220, IRSD, Université de Toulouse, INRA, ENVT, UPS, Toulouse, France
- Departments of Physiology & Pharmacology, and Medicine, University of Calgary Cumming School of Medicine, 3330 Hospital Drive Northwest, Calgary, Alberta T2N 4N1, Canada
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB1 2PD, UK
| | - David I Hughes
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, UK
| | - Giovanni Barbara
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Gilles Dietrich
- INSERM, UMR1220, IRSD, Université de Toulouse, INRA, ENVT, UPS, Toulouse, France
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB1 2PD, UK
- National Centre for Bowel Research and Surgical Innovation, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AJ, UK
| | - Nicolas Cenac
- INSERM, UMR1220, IRSD, Université de Toulouse, INRA, ENVT, UPS, Toulouse, France.
| |
Collapse
|
24
|
Hockley JRF, Smith ESJ, Bulmer DC. Human visceral nociception: findings from translational studies in human tissue. Am J Physiol Gastrointest Liver Physiol 2018; 315:G464-G472. [PMID: 29848022 DOI: 10.1152/ajpgi.00398.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Peripheral sensitization of nociceptors during disease has long been recognized as a leading cause of inflammatory pain. However, a growing body of data generated over the last decade has led to the increased understanding that peripheral sensitization is also an important mechanism driving abdominal pain in highly prevalent functional bowel disorders, in particular, irritable bowel syndrome (IBS). As such, the development of drugs that target pain-sensing nerves innervating the bowel has the potential to be a successful analgesic strategy for the treatment of abdominal pain in both organic and functional gastrointestinal diseases. Despite the success of recent peripherally restricted approaches for the treatment of IBS, not all drugs that have shown efficacy in animal models of visceral pain have reduced pain end points in clinical trials of IBS patients, suggesting innate differences in the mechanisms of pain processing between rodents and humans and, in particular, how we model disease states. To address this gap in our understanding of peripheral nociception from the viscera and the body in general, several groups have developed experimental systems to study nociception in isolated human tissue and neurons, the findings of which we discuss in this review. Studies of human tissue identify a repertoire of human primary afferent subtypes comparable to rodent models including a nociceptor population, the targeting of which will shape future analgesic development efforts. Detailed mechanistic studies in human sensory neurons combined with unbiased RNA-sequencing approaches have revealed fundamental differences in not only receptor/channel expression but also peripheral pain pathways.
Collapse
Affiliation(s)
- James R F Hockley
- Department of Pharmacology, University of Cambridge , Cambridge , United Kingdom
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge , Cambridge , United Kingdom
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge , Cambridge , United Kingdom
| |
Collapse
|
25
|
Yu T, Li L, Liu H, Li H, Liu Z, Li Z. KCNQ2/3/5 channels in dorsal root ganglion neurons can be therapeutic targets of neuropathic pain in diabetic rats. Mol Pain 2018; 14:1744806918793229. [PMID: 30027794 PMCID: PMC6088482 DOI: 10.1177/1744806918793229] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Diabetic neuropathic pain is poorly controlled by analgesics, and the precise molecular mechanisms underlying hyperalgesia remain unclear. The KCNQ2/3/5 channels expressed in dorsal root ganglion neurons are important in pain transmission. The expression and activity of KCNQ2/3/5 channels in dorsal root ganglion neurons in rats with diabetic neuropathic pain were investigated in this study. Methods The mRNA levels of KCNQ2/3/5 channels were analyzed by real-time polymerase chain reaction. The protein levels of KCNQ2/3/5 channels were evaluated by Western blot assay. KCNQ2/3/5 channel expression in situ in dorsal root ganglion neurons was detected by double fluorescent labeling technique. M current (IM) density and neuronal excitability were determined by whole-cell voltage and current clamp recordings. Mechanical allodynia and thermal hyperalgesia were assessed by von Frey filaments and plantar analgesia tester, respectively. Results The mRNA and protein levels of KCNQ2/3/5 channels significantly decreased, followed by the reduction of IM density and elevation of neuronal excitability of dorsal root ganglion neurons from diabetic rats. Activation of KCNQ channels with retigabine reduced the hyperexcitability and inhibition of KCNQ channels with XE991 enhanced the hyperexcitability. Administration of retigabine alleviated both mechanical allodynia and thermal hyperalgesia, while XE991 augmented both mechanical allodynia and thermal hyperalgesia in diabetic neuropathic pain in rats. Conclusion The findings elucidate the mechanisms by which downregulation of the expression and reduction of the activity of KCNQ2/3/5 channels in diabetic rat dorsal root ganglion neurons contribute to neuronal hyperexcitability, which results in hyperalgesia. These data provide intriguing evidence that activation of KCNQ2/3/5 channels might be the potential new targets for alleviating diabetic neuropathic pain symptoms.
Collapse
Affiliation(s)
- Ting Yu
- 1 Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, China.,2 Department of Physiology, Jining Medical University, Jining, China
| | - Lei Li
- 3 Department of Diagnosis, Jining Medical University, Jining, China
| | - Huaxiang Liu
- 4 Department of Rheumatology, Shandong University Qilu Hospital, Jinan, China
| | - Hao Li
- 5 Department of Orthopaedics, Shandong University Qilu Hospital, Jinan, China
| | - Zhen Liu
- 1 Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Zhenzhong Li
- 1 Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, China
| |
Collapse
|
26
|
Du X, Gao H, Jaffe D, Zhang H, Gamper N. M-type K + channels in peripheral nociceptive pathways. Br J Pharmacol 2018; 175:2158-2172. [PMID: 28800673 PMCID: PMC5980636 DOI: 10.1111/bph.13978] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/17/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022] Open
Abstract
Pathological pain is a hyperexcitability disorder. Since the excitability of a neuron is set and controlled by a complement of ion channels it expresses, in order to understand and treat pain, we need to develop a mechanistic insight into the key ion channels controlling excitability within the mammalian pain pathways and how these ion channels are regulated and modulated in various physiological and pathophysiological settings. In this review, we will discuss the emerging data on the expression in pain pathways, functional role and modulation of a family of voltage-gated K+ channels called 'M channels' (KCNQ, Kv 7). M channels are increasingly recognized as important players in controlling pain signalling, especially within the peripheral somatosensory system. We will also discuss the therapeutic potential of M channels as analgesic drug targets. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc/.
Collapse
Affiliation(s)
- Xiaona Du
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
| | - Haixia Gao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
- School of Biomedical Sciences, Faculty of Biological SciencesUniversity of LeedsLeedsUK
| | - David Jaffe
- Department of Biology, UTSA Neurosciences InstituteUniversity of Texas at San AntonioSan AntonioTXUSA
| | - Hailin Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
| | - Nikita Gamper
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and ToxicologyShijiazhuangHebei ProvinceChina
- School of Biomedical Sciences, Faculty of Biological SciencesUniversity of LeedsLeedsUK
| |
Collapse
|
27
|
Choi SI, Hwang SW. Depolarizing Effectors of Bradykinin Signaling in Nociceptor Excitation in Pain Perception. Biomol Ther (Seoul) 2018; 26:255-267. [PMID: 29378387 PMCID: PMC5933892 DOI: 10.4062/biomolther.2017.127] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 10/13/2017] [Accepted: 10/24/2017] [Indexed: 12/23/2022] Open
Abstract
Inflammation is one of the main causes of pathologic pain. Knowledge of the molecular links between inflammatory signals and pain-mediating neuronal signals is essential for understanding the mechanisms behind pain exacerbation. Some inflammatory mediators directly modulate the excitability of pain-mediating neurons by contacting the receptor molecules expressed in those neurons. For decades, many discoveries have accumulated regarding intraneuronal signals from receptor activation through electrical depolarization for bradykinin, a major inflammatory mediator that is able to both excite and sensitize pain-mediating nociceptor neurons. Here, we focus on the final effectors of depolarization, the neuronal ion channels, whose functionalities are specifically affected by bradykinin stimulation. Particular G-protein coupled signaling cascades specialized for each specific depolarizer ion channels are summarized. Some of these ion channels not only serve as downstream effectors but also play critical roles in relaying specific pain modalities such as thermal or mechanical pain. Accordingly, specific pain phenotypes altered by bradykinin stimulation are also discussed. Some members of the effector ion channels are both activated and sensitized by bradykinin-induced neuronal signaling, while others only sensitized or inhibited, which are also introduced. The present overview of the effect of bradykinin on nociceptor neuronal excitability at the molecular level may contribute to better understanding of an important aspect of inflammatory pain and help future design of further research on the components involved and pain modulating strategies.
Collapse
Affiliation(s)
- Seung-In Choi
- Department of Biomedical Sciences and Department of Physiology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Sun Wook Hwang
- Department of Biomedical Sciences and Department of Physiology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
28
|
Contribution of membrane receptor signalling to chronic visceral pain. Int J Biochem Cell Biol 2018; 98:10-23. [DOI: 10.1016/j.biocel.2018.02.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/15/2018] [Accepted: 02/19/2018] [Indexed: 12/18/2022]
|
29
|
Zhou TR, Huang JJ, Huang ZT, Cao HY, Tan B. Inhibitory effects of patchouli alcohol on stress-induced diarrhea-predominant irritable bowel syndrome. World J Gastroenterol 2018; 24:693-705. [PMID: 29456408 PMCID: PMC5807672 DOI: 10.3748/wjg.v24.i6.693] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/06/2017] [Accepted: 11/28/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To elucidate the mechanism of patchouli alcohol (PA) in treatment of rat models of diarrhea-predominant irritable bowel syndrome (IBS-D). METHODS We studied the effects of PA on colonic spontaneous motility using its cumulative log concentration (3 × 10-7 mol/L to 1 × 10-4 mol/L). We then determined the responses of the proximal and distal colon segments of rats to the following stimuli: (1) carbachol (1 × 10-9 mol/L to 1 × 10-5 mol/L); (2) neurotransmitter antagonists including Nω-nitro-L-arginine methyl ester hydrochloride (10 μmol/L) and (1R*, 2S*)-4-[2-Iodo-6-(methylamino)-9H-purin-9-yl]-2-(phosphonooxy)bicyclo[3.1.0]hexane-1-methanol dihydrogen phosphate ester tetraammonium salt (1 μmol/L); (3) agonist α,β-methyleneadenosine 5'-triphosphate trisodium salt (100 μmol/L); and (4) single KCl doses (120 mmol/L). The effects of blockers against antagonist responses were also assessed by pretreatment with PA (100 μmol/L) for 1 min. Electrical-field stimulation (40 V, 2-30 Hz, 0.5 ms pulse duration, and 10 s) was performed to observe nonadrenergic, noncholinergic neurotransmitter release in IBS-D rat colon. The ATP level of Kreb's solution was also determined. RESULTS PA exerted a concentration-dependent inhibitory effect on the spontaneous contraction of the colonic longitudinal smooth muscle, and the half maximal effective concentration (EC50) was 41.9 μmol/L. In comparison with the KCl-treated IBS-D group, the contractile response (mg contractions) in the PA + KCl-treated IBS-D group (11.87 ± 3.34) was significantly decreased in the peak tension (P < 0.01). Compared with CCh-treated IBS-D rat colon, the cholinergic contractile response of IBS-D rat colonic smooth muscle (EC50 = 0.94 μmol/L) was significantly decreased by PA (EC50 = 37.43 μmol/L) (P < 0.05). Lack of nitrergic neurotransmitter release in stress-induced IBS-D rats showed contraction effects on colonic smooth muscle. Pretreatment with PA resulted in inhibitory effect on L-NAME-induced (10 μmol/L) contraction (P < 0.05). ATP might not be the main neurotransmitter involved in inhibitory effects of PA in the colonic relaxation of stress-induced IBS-D rats. CONCLUSION PA application may serve as a new therapeutic approach for IBS-D.
Collapse
Affiliation(s)
- Tian-Ran Zhou
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Jing-Jing Huang
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Zi-Tong Huang
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Hong-Ying Cao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Bo Tan
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| |
Collapse
|