1
|
Pavlov D, Christofi FL. Use of Human In Vitro Gut Specimens for Translational Neurogastroenterology and Motility in the 21st Century. Neurogastroenterol Motil 2025:e15022. [PMID: 40296281 DOI: 10.1111/nmo.15022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/12/2025] [Accepted: 01/26/2025] [Indexed: 04/30/2025]
Abstract
There is a huge gap in our understanding of the human ENS and translating data from mice to humans that is important when developing targeted therapeutics. The ENS or "human little brain in the gut" is easily accessible for study in GI surgical or biopsy samples. This mini review is focused on the use of human gut specimens in translating laboratory data on ENS and enteric neuropathies in neurogastroenterology and motility from mice to humans. Availability of viable human gut samples, in combination with technological advances in innovative recording techniques and new in vitro models provide powerful ways to study neural activity and secretomotor function or monitor motility in health and disease with exquisite sophistication and precision. Electrophysiological recordings, optical recordings with voltage-sensitive dyes, or Ca2+ imaging (in adult or fetal gut) is used to study neural activity in human ENS in health and disease. 'First in man patch clamp recordings' is possible in isolated networks of human myenteric ganglia, opening the door for patch-seq. The human ENS at single cell resolution (snRNA-seq) revealed cell-diversity, similarities and differences between human and mouse in vitro. Visceral afferent recordings are used for mechanosensation and pain signaling in humans. Stem cell therapies may hold future promise for patients with enteric neuropathies. A greater focus on the human ENS and enteric neuropathies (i.e. IBS, FD, postoperative ileus, CIPO, chronic constipation, Hirschsprung Disease, infection, gastroparesis, Parkinson's disease, IBD, visceral pain) is one important step for consideration in developing potential therapeutics before proceeding to more expensive and complex clinical trials in patients to treat GI Disorders and Diseases.
Collapse
Affiliation(s)
- Dmitrii Pavlov
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Fievos L Christofi
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
2
|
Meng MY, Paine LW, Sagnat D, Bello I, Oldroyd S, Javid F, Harper MT, Hockley JRF, St John Smith E, Owens RM, Alric L, Buscail E, Welsh F, Vergnolle N, Bulmer DC. TRPV4 stimulates colonic afferents through mucosal release of ATP and glutamate. Br J Pharmacol 2025; 182:1324-1340. [PMID: 39626870 DOI: 10.1111/bph.17408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Abdominal pain is a leading cause of morbidity for people living with gastrointestinal disease. Whereas the transient receptor potential vanilloid 4 (TRPV4) ion channel has been implicated in the pathogenesis of abdominal pain, the relative paucity of TRPV4 expression in colon-projecting sensory neurons suggests that non-neuronal cells may contribute to TRPV4-mediated nociceptor stimulation. EXPERIMENTAL APPROACH Changes in murine colonic afferent activity were examined using ex vivo electrophysiology in tissues with the gut mucosa present or removed. ATP and glutamate release were measured by bioluminescence assays from human colon organoid cultures and mouse colon. Dorsal root ganglion sensory neuron activity was evaluated by Ca2+ imaging when cultured alone or co-cultured with colonic mucosa. KEY RESULTS Bath application of TRPV4 agonist GSK1016790A elicited a robust increase in murine colonic afferent activity, which was abolished by removing the gut mucosa. GSK1016790A promoted ATP and glutamate release from human colon organoid cultures and mouse colon. Inhibition of ATP degradation in mouse colon enhanced the afferent response to GSK1016790A. Pretreatment with purinoceptor or glutamate receptor antagonists attenuated and abolished the response to GSK1016790A when given alone or in combination, respectively. Sensory neurons co-cultured with colonic mucosal cells produced a marked increase in intracellular Ca2+ to GSK1016790A compared with neurons cultured alone. CONCLUSION AND IMPLICATIONS Our data indicate that mucosal release of ATP and glutamate is responsible for the stimulation of colonic afferents following TRPV4 activation. These findings highlight an opportunity to target the gut mucosa for the development of new visceral analgesics.
Collapse
Affiliation(s)
- Michelle Y Meng
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Luke W Paine
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - David Sagnat
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Ivana Bello
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Sophie Oldroyd
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Farideh Javid
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | | | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Laurent Alric
- Internal Medicine Department of Digestive Disease, CHU Toulouse-Rangueil and Université de Toulouse, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Etienne Buscail
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
- Department of Surgery, CHU Toulouse-Rangueil and Université de Toulouse, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Fraser Welsh
- BioPharmaceuticals R&D, AstraZeneca, Neuroscience, Cambridge, UK
| | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
- Department of Physiology and Pharmacology, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
3
|
Ten Barge JA, van den Bosch GE, Slater R, van den Hoogen NJ, Reiss IKM, Simons SHP. Visceral Pain in Preterm Infants with Necrotizing Enterocolitis: Underlying Mechanisms and Implications for Treatment. Paediatr Drugs 2025; 27:201-220. [PMID: 39752054 PMCID: PMC11829917 DOI: 10.1007/s40272-024-00676-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 01/04/2025]
Abstract
Necrotizing enterocolitis (NEC) is a relatively rare but very severe gastrointestinal disease primarily affecting very preterm infants. NEC is characterized by excessive inflammation and ischemia in the intestines, and is associated with prolonged, severe visceral pain. Despite its recognition as a highly painful disease, current pain management for NEC is often inadequate, and research on optimal analgesic therapy for these patients is lacking. Insight into the mechanisms underlying intestinal pain in infants with NEC-visceral pain-could help identify the most effective analgesics for these vulnerable patients. Therefore, this comprehensive review aims to provide an overview of visceral nociception, including transduction, transmission, modulation, and experience, and discuss the implications for analgesic therapy in preterm infants with NEC. The transmission of visceral pain differs from that of somatic pain, contributing to the diffuse nature of visceral pain. Studies evaluating the effectiveness of analgesics for treating visceral pain in infants are scarce. However, research in visceral pain models highlights agents that may be particularly effective for treating visceral pain based on their mechanisms of action. Further research is necessary to determine whether agents that have shown promise for treating visceral pain in preclinical studies and adults are effective in infants with NEC as well.
Collapse
Affiliation(s)
- Judith A Ten Barge
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands.
| | - Gerbrich E van den Bosch
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | | | - Irwin K M Reiss
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Sinno H P Simons
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
4
|
Baidoo N, Sanger GJ. The human colon: Evidence for degenerative changes during aging and the physiological consequences. Neurogastroenterol Motil 2024:e14848. [PMID: 38887160 DOI: 10.1111/nmo.14848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/16/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND The incidence of constipation increases among the elderly (>65 years), while abdominal pain decreases. Causes include changes in lifestyle (e.g., diet and reduced exercise), disease and medications affecting gastrointestinal functions. Degenerative changes may also occur within the colo-rectum. However, most evidence is from rodents, animals with relatively high rates of metabolism and accelerated aging, with considerable variation in time course. In humans, cellular and non-cellular changes in the aging intestine are poorly investigated. PURPOSE To examine all available studies which reported the effects of aging on cellular and tissue functions of human isolated colon, noting the region studied, sex and age of tissue donors and study size. The focus on human colon reflects the ability to access full-thickness tissue over a wide age range, compared with other gastrointestinal regions. Details are important because of natural human variability. We found age-related changes within the muscle, in the enteric and nociceptor innervation, and in the submucosa. Some involve all regions of colon, but the ascending colon appears more vulnerable. Changes can be cell- and sublayer-dependent. Mechanisms are unclear but may include development of "senescent-like" and associated inflammaging, perhaps associated with increased mucosal permeability to harmful luminal contents. In summary, reduced nociceptor innervation can explain diminished abdominal pain among the elderly. Degenerative changes within the colon wall may have little impact on symptoms and colonic functions, because of high "functional reserve," but are likely to facilitate the development of constipation during age-related challenges (e.g., lifestyle, disease, and medications), now operating against a reduced functional reserve.
Collapse
Affiliation(s)
- Nicholas Baidoo
- School of Life Sciences, University of Westminster, London, UK
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gareth J Sanger
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
5
|
Oldroyd P, Oldroyd S, Meng M, Makwana R, Sanger G, Bulmer D, Malliaras GG, Owens RM. Stretchable Device for Simultaneous Measurements of Contractility and Electrophysiology of Neuromuscular Tissue in the Gastrointestinal Tract. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312735. [PMID: 38290128 DOI: 10.1002/adma.202312735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/15/2024] [Indexed: 02/01/2024]
Abstract
Devices interfacing with biological tissues can provide valuable insights into function, disease, and metabolism through electrical and mechanical signals. However, certain neuromuscular tissues, like those in the gastrointestinal tract, undergo significant strains of up to 40%. Conventional inextensible devices cannot capture the dynamic responses in these tissues. This study introduces electrodes made from poly(3,4-ethylenedioxythiophene):poly(styrenesulfonate) (PEDOT:PSS) and polydimethylsiloxane (PDMS) that enable simultaneous monitoring of electrical and mechanical responses of gut tissue. The soft PDMS layers conform to tissue surfaces during gastrointestinal movement. Dopants, including Capstone FS-30 and polyethylene glycol, are explored to enhance the conductivity, electrical sensitivity to strain, and stability of the PEDOT:PSS. The devices are fabricated using shadow masks and solution-processing techniques, providing a faster and simpler process than traditional clean-room-based lithography. Tested on ex vivo mouse colon and human stomach, the device recorded voltage changes of up to 300 µV during contraction and distension consistent with muscle activity, while simultaneously recording resistance changes of up to 150% due to mechanical strain. These devices detect and respond to chemical stimulants and blockers, and can induce contractions through electrical stimulation. They hold great potential for studying and treating complex disorders like irritable bowel syndrome and gastroparesis.
Collapse
Affiliation(s)
- Poppy Oldroyd
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, CB3 0FA, UK
| | - Sophie Oldroyd
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, CB3 0AS, UK
| | - Michelle Meng
- Department of Pharmacology, Tennis Ct Rd, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Rajesh Makwana
- Blizard Institute, Queen Mary University of London, Cambridge, E1 2AT, UK
| | - Gareth Sanger
- Blizard Institute, Queen Mary University of London, Cambridge, E1 2AT, UK
| | - David Bulmer
- Department of Pharmacology, Tennis Ct Rd, University of Cambridge, Cambridge, CB2 1PD, UK
| | - George G Malliaras
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, CB3 0FA, UK
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, CB3 0AS, UK
| |
Collapse
|
6
|
King JW, Bennett ASW, Wood HM, Baker CC, Alsaadi H, Topley M, Vanner SA, Reed DE, Lomax AE. Expression and function of transient receptor potential melastatin 3 in the spinal afferent innervation of the mouse colon. Am J Physiol Gastrointest Liver Physiol 2024; 326:G176-G186. [PMID: 38084411 DOI: 10.1152/ajpgi.00230.2023] [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: 10/11/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 01/27/2024]
Abstract
Abdominal pain is a cardinal symptom of inflammatory bowel disease (IBD). Transient receptor potential (TRP) channels contribute to abdominal pain in preclinical models of IBD, and TRP melastatin 3 (TRPM3) has recently been implicated in inflammatory bladder and joint pain in rodents. We hypothesized that TRPM3 is involved in colonic sensation and is sensitized during colitis. We used immunohistochemistry, ratiometric Ca2+ imaging, and colonic afferent nerve recordings in mice to evaluate TRPM3 protein expression in colon-projecting dorsal root ganglion (DRG) neurons, as well as functional activity in DRG neurons and colonic afferent nerves. Colitis was induced using dextran sulfate sodium (DSS) in drinking water. TRPM3 protein expression was observed in 76% of colon-projecting DRG neurons and was often colocalized with calcitonin gene-related peptide. The magnitudes of intracellular Ca2+ transients in DRG neurons in response to the TRPM3 agonists CIM-0216 and pregnenolone sulfate sodium were significantly greater in neurons from mice with colitis compared with controls. In addition, the percentage of DRG neurons from mice with colitis that responded to CIM-0216 was significantly increased. CIM-0216 also increased the firing rate of colonic afferent nerves from control and mice with colitis. The TRPM3 inhibitor isosakuranetin inhibited the mechanosensitive response to distension of wide dynamic range afferent nerve units from mice with colitis but had no effect in control mice. Thus, TRPM3 contributes to colonic sensory transduction and may be a potential target for treating pain in IBD.NEW & NOTEWORTHY This is the first study to characterize TRPM3 protein expression and function in colon-projecting DRG neurons. A TRPM3 agonist excited DRG neurons and colonic afferent nerves from healthy mice. TRPM3 agonist responses in DRG neurons were elevated during colitis. Inhibiting TRPM3 reduced the firing of wide dynamic range afferent nerves from mice with colitis but had no effect in control mice.
Collapse
Affiliation(s)
- James W King
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Aidan S W Bennett
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Hannah M Wood
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Corey C Baker
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Hanin Alsaadi
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Max Topley
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Stephen A Vanner
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
7
|
Rajasekhar P, Carbone SE, Johnston ST, Nowell CJ, Wiklendt L, Crampin EJ, She Y, DiCello JJ, Saito A, Sorensen L, Nguyen T, Lee KM, Hamilton JA, King SK, Eriksson EM, Spencer NJ, Gulbransen BD, Veldhuis NA, Poole DP. TRPV4 is expressed by enteric glia and muscularis macrophages of the colon but does not play a prominent role in colonic motility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574831. [PMID: 38260314 PMCID: PMC10802399 DOI: 10.1101/2024.01.09.574831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Mechanosensation is an important trigger of physiological processes in the gastrointestinal tract. Aberrant responses to mechanical input are associated with digestive disorders, including visceral hypersensitivity. Transient Receptor Potential Vanilloid 4 (TRPV4) is a mechanosensory ion channel with proposed roles in visceral afferent signaling, intestinal inflammation, and gut motility. While TRPV4 is a potential therapeutic target for digestive disease, current mechanistic understanding of how TRPV4 may influence gut function is limited by inconsistent reports of TRPV4 expression and distribution. Methods In this study we profiled functional expression of TRPV4 using Ca2+ imaging of wholemount preparations of the mouse, monkey, and human intestine in combination with immunofluorescent labeling for established cellular markers. The involvement of TRPV4 in colonic motility was assessed in vitro using videomapping and contraction assays. Results The TRPV4 agonist GSK1016790A evoked Ca2+ signaling in muscularis macrophages, enteric glia, and endothelial cells. TRPV4 specificity was confirmed using TRPV4 KO mouse tissue or antagonist pre-treatment. Calcium responses were not detected in other cell types required for neuromuscular signaling including enteric neurons, interstitial cells of Cajal, PDGFRα+ cells, and intestinal smooth muscle. TRPV4 activation led to rapid Ca2+ responses by a subpopulation of glial cells, followed by sustained Ca2+ signaling throughout the enteric glial network. Propagation of these waves was suppressed by inhibition of gap junctions or Ca2+ release from intracellular stores. Coordinated glial signaling in response to GSK1016790A was also disrupted in acute TNBS colitis. The involvement of TRPV4 in the initiation and propagation of colonic motility patterns was examined in vitro. Conclusions We reveal a previously unappreciated role for TRPV4 in the initiation of distension-evoked colonic motility. These observations provide new insights into the functional role of TRPV4 activation in the gut, with important implications for how TRPV4 may influence critical processes including inflammatory signaling and motility.
Collapse
Affiliation(s)
- Pradeep Rajasekhar
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
- Centre for Dynamic Imaging, WEHI, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Simona E Carbone
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Stuart T Johnston
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Lukasz Wiklendt
- College of Medicine & Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | - Edmund J Crampin
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Yinghan She
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Jesse J DiCello
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Ayame Saito
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Luke Sorensen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Thanh Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Kevin Mc Lee
- Department of Medicine, The University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3010, Australia
| | - John A Hamilton
- Department of Medicine, The University of Melbourne, Royal Melbourne Hospital, Parkville, VIC 3010, Australia
| | - Sebastian K King
- Department of Paediatric Surgery, The Royal Children's Hospital, Parkville, VIC 3052, Australia
- Surgical Research, Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Emily M Eriksson
- Population Health and Immunity, WEHI, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Nick J Spencer
- College of Medicine & Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | | | - Nicholas A Veldhuis
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Daniel P Poole
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| |
Collapse
|
8
|
Expression of TRP Channels in Colonic Mucosa of IBS-D Patients and Its Correlation with the Severity of the Disease. Gastroenterol Res Pract 2022; 2022:7294775. [PMID: 35677724 PMCID: PMC9168202 DOI: 10.1155/2022/7294775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 11/18/2022] Open
Abstract
Aim Lots of researches have endeavored to elucidate the pathogenetic mechanism of visceral hypersensitivity in order to guide the therapy of diarrhea predominant-irritable bowel syndrome (IBS-D). Transient receptor potential (TRP) channels and their role in visceral nociception have been vastly investigated. We investigated the expression of TRP channels in IBS-D colonic biopsies and its correlation with the severity of the disease. Methods Sigmoid biopsies were obtained from 34 IBS-D patients and 28 healthy controls (HCs). IBS-D was diagnosed according to Rome IV criteria. Their clinical parameters were assessed through questionnaires. Expression of TRPV1, TRPV4, TRPA1, TRPM2, and TRPM8 was evaluated with immunohistology staining. Results Expression levels of TRPV1, TRPV4, and TRPA1 in the colonic mucosa of IBS-D patients were significantly higher than those in HCs (p < 0.05), while there was no obvious difference of TRPM2 and TRPM8 expression between IBS-D patients and HCs. In addition, the expression levels of TRPV1 and TRPA1, but TRPV4, in the colonic mucosa correlated positively with the severity of diseases (r = 0.6303 and 0.4506, respectively, p < 0.05). Conclusions Expression of TRPV1, TRPA1, and TRPV4 in the colonic mucosa was enhanced in IBS-D patients compared with HCs with the former two correlated with the severity of the disease. TRP channels might be promising biomarkers in the diagnosis and estimate of the severity in IBS-D.
Collapse
|
9
|
Hartrick CT. Exploiting Injury-Induced Peripheral Opioid Receptor Changes in Novel Analgesic Development for Chronic Pain. FRONTIERS IN PAIN RESEARCH 2022; 3:883164. [PMID: 35557854 PMCID: PMC9090307 DOI: 10.3389/fpain.2022.883164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
|
10
|
Aguilera-Lizarraga J, Hussein H, Boeckxstaens GE. Immune activation in irritable bowel syndrome: what is the evidence? Nat Rev Immunol 2022; 22:674-686. [PMID: 35296814 DOI: 10.1038/s41577-022-00700-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2022] [Indexed: 12/15/2022]
Abstract
Irritable bowel syndrome (IBS) is a chronic functional gastrointestinal disorder that is characterized by abdominal pain and an altered defecation pattern. It affects between 5 and 20% of the general population and can seriously impact quality of life. The pathophysiology of IBS is rather complex and multifactorial including, for example, altered signalling by the gut-brain axis, dysbiosis, abnormal visceral pain signalling and intestinal immune activation. The latter has gained particular interest in recent years, with growing insight into the bidirectional communication between the nervous system and the immune system. In this Review, we detail the current evidence suggesting that immune activation contributes to the pathology seen in patients with IBS and discuss the potential mechanisms involved. Moreover, we describe how immune mediators, particularly those released by mast cells, can directly activate or sensitize pain-transmitting nerves, leading to increased pain signalling and abdominal pain. Finally, we discuss the potential of interventions targeting immune activation as a new therapeutic strategy for patients suffering from IBS.
Collapse
Affiliation(s)
- Javier Aguilera-Lizarraga
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Hind Hussein
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Guy E Boeckxstaens
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.
| |
Collapse
|
11
|
Brierley SM, Grundy L, Castro J, Harrington AM, Hannig G, Camilleri M. Guanylate cyclase-C agonists as peripherally acting treatments of chronic visceral pain. Trends Pharmacol Sci 2022; 43:110-122. [PMID: 34865885 PMCID: PMC8760167 DOI: 10.1016/j.tips.2021.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 02/03/2023]
Abstract
Irritable bowel syndrome (IBS) is a chronic gastrointestinal disorder characterized by abdominal pain and altered bowel habit that affects ~11% of the global population. Over the past decade, preclinical and clinical studies have revealed a variety of novel mechanisms relating to the visceral analgesic effects of guanylate cyclase-C (GC-C) agonists. Here we discuss the mechanisms by which GC-C agonists target the GC-C/cyclic guanosine-3',5'-monophosphate (cGMP) pathway, resulting in visceral analgesia as well as clinically relevant relief of abdominal pain and other sensations in IBS patients. Due to the preponderance of evidence we focus on linaclotide, a 14-amino acid GC-C agonist with very low oral bioavailability that acts within the gut. Collectively, the weight of experimental and clinical evidence supports the concept that GC-C agonists act as peripherally acting visceral analgesics.
Collapse
Affiliation(s)
- Stuart M. Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, 5042, AUSTRALIA.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, AUSTRALIA.,Discipline of Medicine, University of Adelaide, North Terrace, Adelaide, South Australia 5000, AUSTRALIA.,Corresponding Author: Prof. Stuart M. Brierley, Ph.D. Visceral Pain Research Group, Level 7, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA 5000, AUSTRALIA.
| | - Luke Grundy
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, 5042, AUSTRALIA.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, AUSTRALIA
| | - Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, 5042, AUSTRALIA.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, AUSTRALIA
| | - Andrea M. Harrington
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, 5042, AUSTRALIA.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, AUSTRALIA
| | | | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiologic Research Program, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
12
|
Beckers AB, Wilms E, Mujagic Z, Kajtár B, Csekő K, Weerts ZZRM, Vork L, Troost FJ, Kruimel JW, Conchillo JM, Helyes Z, Masclee AAM, Keszthelyi D, Jonkers DMAE. Age-Related Decrease in Abdominal Pain and Associated Structural- and Functional Mechanisms: An Exploratory Study in Healthy Individuals and Irritable Bowel Syndrome Patients. Front Pharmacol 2022; 12:806002. [PMID: 34975501 PMCID: PMC8716827 DOI: 10.3389/fphar.2021.806002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction: The world population is ageing, resulting in increased prevalence of age-related comorbidities and healthcare costs. Limited data are available on intestinal health in elderly populations. Structural and functional changes, including altered visceroperception, may lead to altered bowel habits and abdominal symptoms in healthy individuals and irritable bowel syndrome (IBS) patients. Our aim was to explore age-related changes in gastrointestinal symptoms and underlying mechanisms. Methods: In total, 780 subjects (IBS patients n = 463, healthy subjects n = 317) from two separate studies were included. Subjects were divided into different age groups ranging from young adult to elderly. Demographics and gastrointestinal symptom scores were collected from all participants using validated questionnaires. A subset of 233 IBS patients and 103 controls underwent a rectal barostat procedure to assess visceral hypersensitivity. Sigmoid biopsies were obtained from 10 healthy young adults and 10 healthy elderly. Expression of the visceral pain-associated receptors transient receptor potential (TRP) Ankyrin 1 (TRPA1) and Vanilloid 1 (TRPV1) genes were investigated by quantitative RT-PCR and immunofluorescence. Results: Both elderly IBS and healthy individuals showed significantly lower scores for abdominal pain (p < 0.001) and indigestion (p < 0.05) as compared to respective young adults. Visceral hypersensitivity was less common in elderly than young IBS patients (p < 0.001). Relative TRPA1 gene transcription, as well as TRPA1 and TRPV1 immunoreactivity were significantly lower in healthy elderly versus healthy young adults (p < 0.05). Conclusions: Our findings show an age-related decrease in abdominal pain perception. This may in part be related to decreased TRPA1 and/or TRPV1 receptor expression. Further studies are needed to reveal precise underlying mechanisms and the associations with intestinal health.
Collapse
Affiliation(s)
- Abraham B Beckers
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ellen Wilms
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Zlatan Mujagic
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Béla Kajtár
- Department of Pathology, Clinical Centre, Medical School, University of Pecs, Pécs, Hungary
| | - Kata Csekő
- Department of Pharmacology and Pharmacotherapy, Medical School and Molecular Pharmacology Research Group, Szentágothai Research Centre, University of Pecs, Pécs, Hungary
| | - Zsa Zsa R M Weerts
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Lisa Vork
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Freddy J Troost
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Joanna W Kruimel
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - José M Conchillo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School and Molecular Pharmacology Research Group, Szentágothai Research Centre, University of Pecs, Pécs, Hungary.,PharmInVivo Ltd, Pécs, Hungary
| | - Ad A M Masclee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Daniel Keszthelyi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Daisy M A E Jonkers
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
13
|
Neural signalling of gut mechanosensation in ingestive and digestive processes. Nat Rev Neurosci 2022; 23:135-156. [PMID: 34983992 DOI: 10.1038/s41583-021-00544-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2021] [Indexed: 12/29/2022]
Abstract
Eating and drinking generate sequential mechanosensory signals along the digestive tract. These signals are communicated to the brain for the timely initiation and regulation of diverse ingestive and digestive processes - ranging from appetite control and tactile perception to gut motility, digestive fluid secretion and defecation - that are vital for the proper intake, breakdown and absorption of nutrients and water. Gut mechanosensation has been investigated for over a century as a common pillar of energy, fluid and gastrointestinal homeostasis, and recent discoveries of specific mechanoreceptors, contributing ion channels and the well-defined circuits underlying gut mechanosensation signalling and function have further expanded our understanding of ingestive and digestive processes at the molecular and cellular levels. In this Review, we discuss our current understanding of the generation of mechanosensory signals from the digestive periphery, the neural afferent pathways that relay these signals to the brain and the neural circuit mechanisms that control ingestive and digestive processes, focusing on the four major digestive tract parts: the oral and pharyngeal cavities, oesophagus, stomach and intestines. We also discuss the clinical implications of gut mechanosensation in ingestive and digestive disorders.
Collapse
|
14
|
Peiris M, Weerts ZZRM, Aktar R, Masclee AAM, Blackshaw A, Keszthelyi D. A putative anti-inflammatory role for TRPM8 in irritable bowel syndrome-An exploratory study. Neurogastroenterol Motil 2021; 33:e14170. [PMID: 34145938 DOI: 10.1111/nmo.14170] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 04/08/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Chronic and recurring pain is a characteristic symptom in irritable bowel syndrome (IBS). Altered signaling between immune cells and sensory neurons within the gut may promote generation of pain symptoms. As transient receptor potential melastatin 8 (TRPM8) agonists, such as L-menthol in peppermint oil, have shown to attenuate IBS pain symptoms, we began investigating potential molecular mechanisms. METHODS Colonic biopsy tissues were collected from patients with IBS and controls, in two separate cohorts. Immunohistochemistry was performed to identify TRPM8 localization. Quantitative PCR was performed to measure mucosal mRNA levels of TRPM8. In addition, functional experiments with the TRPM8 agonist icilin were performed ex vivo to examine cytokine release from biopsies. Daily diaries were collected to ascertain pain symptoms. RESULTS In biopsy tissue from IBS patients, we showed that TRPM8 immunoreactivity is colocalized with immune cells predominantly of the dendritic cell lineage, in close approximation to nerve endings, and TRPM8 protein and mRNA expression was increased in IBS patients compared to controls (p < 0.001). TRPM8 mRNA expression showed a significant positive association with abdominal pain scores (p = 0.015). Treatment of IBS patient biopsies with icilin reduced release of inflammatory cytokines IL-1β, IL-6, and TNF-α (p < 0.05). CONCLUSIONS AND INFERENCES These data indicate TRPM8 may have important anti-inflammatory properties and by this virtue can impact neuro-immune disease mechanisms in IBS.
Collapse
Affiliation(s)
- Madusha Peiris
- Wingate Institute for Neurogastroenterology, Centre for Neuroscience, Trauma & Surgery, Blizard Institute, Queen Mary University of London, London, UK
| | - Zsa Zsa R M Weerts
- Division of Gastroenterology & Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Rubina Aktar
- Wingate Institute for Neurogastroenterology, Centre for Neuroscience, Trauma & Surgery, Blizard Institute, Queen Mary University of London, London, UK
| | - Ad A M Masclee
- Division of Gastroenterology & Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ashley Blackshaw
- Wingate Institute for Neurogastroenterology, Centre for Neuroscience, Trauma & Surgery, Blizard Institute, Queen Mary University of London, London, UK
| | - Daniel Keszthelyi
- Division of Gastroenterology & Hepatology, Department of Internal Medicine, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Despite the wide prevalence of gastro-esophageal reflux disease (GERD), the neurophysiological mechanisms underlying heartburn perception in the esophagus of patients with GERD remains incompletely understood. Recent studies have highlighted the potential influence sensory afferent nerves innervating the oesophageal epithelium may have on heartburn pathogenesis. The purpose of this review is to consider the current understanding of esophageal afferent neuronal innervation, including the nociceptive role of acid-sensing receptors expressed on these sensory nerves, in relation to pain perception in the esophagus of GERD patients. RECENT FINDINGS Central and peripheral pathways of sensitization following noxious stimulation of nociceptive receptors expressed on afferent nerves can regulate the strength of sensory nerve activation in the esophagus, which can result in the amplification or suppression of afferent signal transmission. The localization and characterization of mucosal sensory afferent nerves vary between GERD phenotypes and may explain the heterogeneity of symptom perception in patients with apparently similar levels of reflux. SUMMARY In this review, we discuss the relevance of afferent esophageal innervation in heartburn perception, with a particular focus on the pathways of reflux-induced activation of nociceptive nerves.
Collapse
|
16
|
Middleton SJ, Barry AM, Comini M, Li Y, Ray PR, Shiers S, Themistocleous AC, Uhelski ML, Yang X, Dougherty PM, Price TJ, Bennett DL. Studying human nociceptors: from fundamentals to clinic. Brain 2021; 144:1312-1335. [PMID: 34128530 PMCID: PMC8219361 DOI: 10.1093/brain/awab048] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/26/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic pain affects one in five of the general population and is the third most important cause of disability-adjusted life-years globally. Unfortunately, treatment remains inadequate due to poor efficacy and tolerability. There has been a failure in translating promising preclinical drug targets into clinic use. This reflects challenges across the whole drug development pathway, from preclinical models to trial design. Nociceptors remain an attractive therapeutic target: their sensitization makes an important contribution to many chronic pain states, they are located outside the blood-brain barrier, and they are relatively specific. The past decade has seen significant advances in the techniques available to study human nociceptors, including: the use of corneal confocal microscopy and biopsy samples to observe nociceptor morphology, the culture of human nociceptors (either from surgical or post-mortem tissue or using human induced pluripotent stem cell derived nociceptors), the application of high throughput technologies such as transcriptomics, the in vitro and in vivo electrophysiological characterization through microneurography, and the correlation with pain percepts provided by quantitative sensory testing. Genome editing in human induced pluripotent stem cell-derived nociceptors enables the interrogation of the causal role of genes in the regulation of nociceptor function. Both human and rodent nociceptors are more heterogeneous at a molecular level than previously appreciated, and while we find that there are broad similarities between human and rodent nociceptors there are also important differences involving ion channel function, expression, and cellular excitability. These technological advances have emphasized the maladaptive plastic changes occurring in human nociceptors following injury that contribute to chronic pain. Studying human nociceptors has revealed new therapeutic targets for the suppression of chronic pain and enhanced repair. Cellular models of human nociceptors have enabled the screening of small molecule and gene therapy approaches on nociceptor function, and in some cases have enabled correlation with clinical outcomes. Undoubtedly, challenges remain. Many of these techniques are difficult to implement at scale, current induced pluripotent stem cell differentiation protocols do not generate the full diversity of nociceptor populations, and we still have a relatively poor understanding of inter-individual variation in nociceptors due to factors such as age, sex, or ethnicity. We hope our ability to directly investigate human nociceptors will not only aid our understanding of the fundamental neurobiology underlying acute and chronic pain but also help bridge the translational gap.
Collapse
Affiliation(s)
- Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Allison M Barry
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Maddalena Comini
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Yan Li
- Department of Anesthesia and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pradipta R Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Andreas C Themistocleous
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK.,Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Megan L Uhelski
- Department of Anesthesia and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xun Yang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Patrick M Dougherty
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
17
|
Functional and anatomical deficits in visceral nociception with age: a mechanism of silent appendicitis in the elderly? Pain 2021; 161:773-786. [PMID: 31790010 DOI: 10.1097/j.pain.0000000000001764] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The ability to sense visceral pain during appendicitis is diminished with age leading to delay in seeking health care and poorer clinical outcomes. To understand the mechanistic basis of this phenomenon, we examined visceral nociception in aged mouse and human tissue. Inflamed and noninflamed appendixes were collected from consenting patients undergoing surgery for the treatment of appendicitis or bowel cancer. Supernatants were generated by incubating samples in buffer and used to stimulate multiunit activity in intestinal preparations, or single-unit activity from teased fibres in colonic preparations, of young and old mice. Changes in afferent innervation with age were determined by measuring the density of calcitonin gene-related peptide-positive afferent fibres and by counting dorsal root ganglia back-labelled by injection of tracer dye into the wall of the colon. Finally, the effect of age on nociceptor function was studied in mouse and human colon. Afferent responses to appendicitis supernatants were greatly impaired in old mice. Further investigation revealed this was due to a marked reduction in the afferent innervation of the bowel and a substantial impairment in the ability of the remaining afferent fibres to transduce noxious stimuli. Translational studies in human tissue demonstrated a significant reduction in the multiunit but not the single-unit colonic mesenteric nerve response to capsaicin with age, indicative of a loss of nociceptor innervation. Our data demonstrate that anatomical and functional deficits in nociception occur with age, underpinning the atypical or silent presentation of appendicitis in the elderly.
Collapse
|
18
|
Guo T, Patel S, Shah D, Chi L, Emadi S, Pierce DM, Han M, Brumovsky PR, Feng B. Optical clearing reveals TNBS-induced morphological changes of VGLUT2-positive nerve fibers in mouse colorectum. Am J Physiol Gastrointest Liver Physiol 2021; 320:G644-G657. [PMID: 33533318 PMCID: PMC8238166 DOI: 10.1152/ajpgi.00363.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/12/2021] [Accepted: 01/27/2021] [Indexed: 01/31/2023]
Abstract
Colorectal hypersensitivity and sensitization of both mechanosensitive and mechanically insensitive afferents develop after intracolonic instillation of 2,4,6-trinitrobenzenesulfonic acid (TNBS) in the mouse, a model of postinfectious irritable bowel syndrome. In mice in which ∼80% of extrinsic colorectal afferents were labeled genetically using the promotor for vesicular glutamate transporter type 2 (VGLUT2), we systematically quantified the morphology of VGLUT2-positive axons in mouse colorectum 7-28 days following intracolonic TNBS treatment. After removal, the colorectum was distended (20 mmHg), fixed with paraformaldehyde, and optically cleared to image VGLUT2-positive axons throughout the colorectal wall thickness. We conducted vector path tracing of individual axons to allow systematic quantification of nerve fiber density and shape. Abundant VGLUT2-positive nerve fibers were present in most layers of the colorectum, except the serosal and longitudinal muscular layers. A small percentage of VGLUT2-positive myenteric plexus neurons was also detected. Intracolonic TNBS treatment significantly reduced the number of VGLUT2-positive nerve fibers in submucosal, myenteric plexus, and mucosal layers at day 7 post-TNBS, which mostly recovered by day 28. We also found that almost all fibers in the submucosa were meandering and curvy, with ∼10% showing pronounced curviness (quantified by the linearity index). TNBS treatment resulted in a significant reduction of the proportions of pronounced curvy fibers in the rectal region at 28 days post-TNBS. Altogether, the present morphological study reveals profound changes in the distribution of VGLUT2-positive fibers in mouse colorectum undergoing TNBS-induced colitis and draws attention to curvy fibers in the submucosa with potential roles in visceral nociception.NEW & NOTEWORTHY We conducted genetic labeling and optical clearing to visualize extrinsic sensory nerve fibers in whole-mount colorectum, which revealed widespread presence of axons in the submucosal layer. Remarkably, axons in the submucosa were meandering and curvy, in contrast to axons in other layers generally aligned with the basal tissues. Intracolonic TNBS treatment led to pronounced changes of nerve fiber density and curviness, suggesting nerve fiber morphologies as potentially contributing factors to sensory sensitization.
Collapse
Affiliation(s)
- Tiantian Guo
- Department of Biomedical Engineering, University of Connecticut, Mansfield, Connecticut
| | - Shivam Patel
- Department of Physiology and Neurobiology, University of Connecticut, Mansfield, Connecticut
| | - Dhruv Shah
- Department of Molecular and Cell Biology, University of Connecticut, Mansfield, Connecticut
| | - Ling Chi
- Department of Physiology and Neurobiology, University of Connecticut, Mansfield, Connecticut
| | - Sharareh Emadi
- Department of Biomedical Engineering, University of Connecticut, Mansfield, Connecticut
| | - David M Pierce
- Department of Biomedical Engineering, University of Connecticut, Mansfield, Connecticut
- Department of Mechanical Engineering, University of Connecticut, Mansfield, Connecticut
| | - Martin Han
- Department of Biomedical Engineering, University of Connecticut, Mansfield, Connecticut
| | - Pablo R Brumovsky
- Instituto de Investigaciones en Medicina Traslacional, National Scientific and Technical Research Council, Austral University, Buenos Aires, Argentina
| | - Bin Feng
- Department of Biomedical Engineering, University of Connecticut, Mansfield, Connecticut
- Department of Physiology and Neurobiology, University of Connecticut, Mansfield, Connecticut
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Feng B, Guo T. Visceral pain from colon and rectum: the mechanotransduction and biomechanics. J Neural Transm (Vienna) 2020; 127:415-429. [PMID: 31598778 PMCID: PMC7141966 DOI: 10.1007/s00702-019-02088-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 09/28/2019] [Indexed: 12/14/2022]
Abstract
Visceral pain is the cardinal symptom of functional gastrointestinal (GI) disorders such as the irritable bowel syndrome (IBS) and the leading cause of patients' visit to gastroenterologists. IBS-related visceral pain usually arises from the distal colon and rectum (colorectum), an intraluminal environment that differs greatly from environment outside the body in chemical, biological, thermal, and mechanical conditions. Accordingly, visceral pain is different from cutaneous pain in several key psychophysical characteristics, which likely underlies the unsatisfactory management of visceral pain by drugs developed for other types of pain. Colorectal visceral pain is usually elicited from mechanical distension/stretch, rather than from heating, cutting, pinching, or piercing that usually evoke pain from the skin. Thus, mechanotransduction, i.e., the encoding of colorectal mechanical stimuli by sensory afferents, is crucial to the underlying mechanisms of GI-related visceral pain. This review will focus on colorectal mechanotransduction, the process of converting colorectal mechanical stimuli into trains of action potentials by the sensory afferents to inform the central nervous system (CNS). We will summarize neurophysiological studies on afferent encoding of colorectal mechanical stimuli, highlight recent advances in our understanding of colorectal biomechanics that plays critical roles in mechanotransduction, and review studies on mechano-sensitive ion channels in colorectal afferents. This review calls for focused attention on targeting colorectal mechanotransduction as a new strategy for managing visceral pain, which can also have an added benefit of limited CNS side effects, because mechanotransduction arises from peripheral organs.
Collapse
Affiliation(s)
- Bin Feng
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269-3247, USA.
| | - Tiantian Guo
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, Storrs, CT, 06269-3247, USA
| |
Collapse
|
22
|
Rivas-Ramírez P, Reboreda A, Rueda-Ruzafa L, Herrera-Pérez S, Lamas JA. PIP 2 Mediated Inhibition of TREK Potassium Currents by Bradykinin in Mouse Sympathetic Neurons. Int J Mol Sci 2020; 21:ijms21020389. [PMID: 31936257 PMCID: PMC7014146 DOI: 10.3390/ijms21020389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/30/2019] [Accepted: 01/02/2020] [Indexed: 12/17/2022] Open
Abstract
Bradykinin (BK), a hormone inducing pain and inflammation, is known to inhibit potassium M-currents (IM) and to increase the excitability of the superior cervical ganglion (SCG) neurons by activating the Ca2+-calmodulin pathway. M-current is also reduced by muscarinic agonists through the depletion of membrane phosphatidylinositol 4,5-biphosphate (PIP2). Similarly, the activation of muscarinic receptors inhibits the current through two-pore domain potassium channels (K2P) of the “Tandem of pore-domains in a Weakly Inward rectifying K+ channel (TWIK)-related channels” (TREK) subfamily by reducing PIP2 in mouse SCG neurons (mSCG). The aim of this work was to test and characterize the modulation of TREK channels by bradykinin. We used the perforated-patch technique to investigate riluzole (RIL) activated currents in voltage- and current-clamp experiments. RIL is a drug used in the palliative treatment of amyotrophic lateral sclerosis and, in addition to blocking voltage-dependent sodium channels, it also selectively activates the K2P channels of the TREK subfamily. A cell-attached patch-clamp was also used to investigate TREK-2 single channel currents. We report here that BK reduces spike frequency adaptation (SFA), inhibits the riluzole-activated current (IRIL), which flows mainly through TREK-2 channels, by about 45%, and reduces the open probability of identified single TREK-2 channels in cultured mSCG cells. The effect of BK on IRIL was precluded by the bradykinin receptor (B2R) antagonist HOE-140 (d-Arg-[Hyp3, Thi5, d-Tic7, Oic8]BK) but also by diC8PIP2 which prevents PIP2 depletion when phospholipase C (PLC) is activated. On the contrary, antagonizing inositol triphosphate receptors (IP3R) using 2-aminoethoxydiphenylborane (2-APB) or inhibiting protein kinase C (PKC) with bisindolylmaleimide did not affect the inhibition of IRIL by BK. In conclusion, bradykinin inhibits TREK-2 channels through the activation of B2Rs resulting in PIP2 depletion, much like we have demonstrated for muscarinic agonists. This mechanism implies that TREK channels must be relevant for the capture of information about pain and visceral inflammation.
Collapse
|
23
|
Mihara H, Boudaka A, Tominaga M, Sugiyama T. Transient Receptor Potential Vanilloid 4 Regulation of Adenosine Triphosphate Release by the Adenosine Triphosphate Transporter Vesicular Nucleotide Transporter, a Novel Therapeutic Target for Gastrointestinal Baroreception and Chronic Inflammation. Digestion 2020; 101:6-11. [PMID: 31770754 PMCID: PMC6979422 DOI: 10.1159/000504021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 02/04/2023]
Abstract
BACKGROUND Transient receptor potential vanilloid 4 (TRPV4) is activated by stretch (mechanical), warm temperature, some epoxyeicosatrienoic acids, and lipopolysaccharide. TRPV4 is expressed throughout the gastrointestinal epithelia and its activation induces adenosine triphosphate (ATP) exocytosis that is involved in visceral hypersensitivity. As an ATP transporter, vesicular nucleotide transporter (VNUT) mediates ATP storage in secretory vesicles and ATP release via exocytosis upon stimulation. SUMMARY TRPV4 is sensitized under inflammatory conditions by a variety of factors, including proteases and serotonin, whereas methylation-dependent silencing of TRPV4 expression is associated with various pathophysiological conditions. Gastrointestinal epithelia also release ATP in response to hypo-osmolality or acid through molecular mechanisms that remain unclear. These synergistically released ATP could be involved in visceral hypersensitivity. Low concentrations of the first generation bisphosphate, clodronate, were recently reported to inhibit VNUT activity and thus clodronate may be a safe and potent therapeutic option to treat visceral pain. Key Messages: This review focuses on: (1) ATP and TRPV4 activities in gastrointestinal epithelia; (2) factors that could modulate TRPV4 activity in gastrointestinal epithelia; and (3) the inhibition of VNUT as a potential novel therapeutic strategy for functional gastrointestinal disorders.
Collapse
Affiliation(s)
- Hiroshi Mihara
- aCenter for Medical Education and Career Development, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan,bDepartment of Gastroenterology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan,*Hiroshi Mihara, Center for Medical Education and Career Development, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194 (Japan), E-Mail
| | - Ammar Boudaka
- cDepartment of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Makoto Tominaga
- dDivision of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Toshiro Sugiyama
- eResearch Division of Molecular Targeting Therapy and Prevention of GI Cancer, Hokkaido University, Sapporo, Japan
| |
Collapse
|
24
|
Alaimo A, Rubert J. The Pivotal Role of TRP Channels in Homeostasis and Diseases throughout the Gastrointestinal Tract. Int J Mol Sci 2019; 20:ijms20215277. [PMID: 31652951 PMCID: PMC6862298 DOI: 10.3390/ijms20215277] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022] Open
Abstract
The transient receptor potential (TRP) channels superfamily are a large group of proteins that play crucial roles in cellular processes. For example, these cation channels act as sensors in the detection and transduction of stimuli of temperature, small molecules, voltage, pH, and mechanical constrains. Over the past decades, different members of the TRP channels have been identified in the human gastrointestinal (GI) tract playing multiple modulatory roles. Noteworthy, TRPs support critical functions related to the taste perception, mechanosensation, and pain. They also participate in the modulation of motility and secretions of the human gut. Last but not least, altered expression or activity and mutations in the TRP genes are often related to a wide range of disorders of the gut epithelium, including inflammatory bowel disease, fibrosis, visceral hyperalgesia, irritable bowel syndrome, and colorectal cancer. TRP channels could therefore be promising drug targets for the treatment of GI malignancies. This review aims at providing a comprehensive picture of the most recent advances highlighting the expression and function of TRP channels in the GI tract, and secondly, the description of the potential roles of TRPs in relevant disorders is discussed reporting our standpoint on GI tract–TRP channels interactions.
Collapse
Affiliation(s)
- Alessandro Alaimo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Povo (Tn), Italy.
| | - Josep Rubert
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Povo (Tn), Italy.
| |
Collapse
|
25
|
Hockley JRF, Taylor TS, Callejo G, Wilbrey AL, Gutteridge A, Bach K, Winchester WJ, Bulmer DC, McMurray G, Smith ESJ. Single-cell RNAseq reveals seven classes of colonic sensory neuron. Gut 2019; 68:633-644. [PMID: 29483303 PMCID: PMC6580772 DOI: 10.1136/gutjnl-2017-315631] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 02/02/2018] [Accepted: 02/10/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Integration of nutritional, microbial and inflammatory events along the gut-brain axis can alter bowel physiology and organism behaviour. Colonic sensory neurons activate reflex pathways and give rise to conscious sensation, but the diversity and division of function within these neurons is poorly understood. The identification of signalling pathways contributing to visceral sensation is constrained by a paucity of molecular markers. Here we address this by comprehensive transcriptomic profiling and unsupervised clustering of individual mouse colonic sensory neurons. DESIGN Unbiased single-cell RNA-sequencing was performed on retrogradely traced mouse colonic sensory neurons isolated from both thoracolumbar (TL) and lumbosacral (LS) dorsal root ganglia associated with lumbar splanchnic and pelvic spinal pathways, respectively. Identified neuronal subtypes were validated by single-cell qRT-PCR, immunohistochemistry (IHC) and Ca2+-imaging. RESULTS Transcriptomic profiling and unsupervised clustering of 314 colonic sensory neurons revealed seven neuronal subtypes. Of these, five neuronal subtypes accounted for 99% of TL neurons, with LS neurons almost exclusively populating the remaining two subtypes. We identify and classify neurons based on novel subtype-specific marker genes using single-cell qRT-PCR and IHC to validate subtypes derived from RNA-sequencing. Lastly, functional Ca2+-imaging was conducted on colonic sensory neurons to demonstrate subtype-selective differential agonist activation. CONCLUSIONS We identify seven subtypes of colonic sensory neurons using unbiased single-cell RNA-sequencing and confirm translation of patterning to protein expression, describing sensory diversity encompassing all modalities of colonic neuronal sensitivity. These results provide a pathway to molecular interrogation of colonic sensory innervation in health and disease, together with identifying novel targets for drug development.
Collapse
Affiliation(s)
- James R F Hockley
- Department of Pharmacology, University of Cambridge, Cambridge, UK,Neuroscience and Pain Research Unit, Pfizer, Cambridge, UK
| | - Toni S Taylor
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Gerard Callejo
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Anna L Wilbrey
- Neuroscience and Pain Research Unit, Pfizer, Cambridge, UK
| | | | - Karsten Bach
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
26
|
Balemans D, Aguilera-Lizarraga J, Florens MV, Jain P, Denadai-Souza A, Viola MF, Alpizar YA, Van Der Merwe S, Vanden Berghe P, Talavera K, Vanner S, Wouters MM, Boeckxstaens GE. Histamine-mediated potentiation of transient receptor potential (TRP) ankyrin 1 and TRP vanilloid 4 signaling in submucosal neurons in patients with irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol 2019; 316:G338-G349. [PMID: 30629470 DOI: 10.1152/ajpgi.00116.2018] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Previously, we showed histamine-mediated sensitization of transient receptor potential (TRP) vanilloid 1 (TRPV1) in patients with irritable bowel syndrome (IBS). Sensitization of TRP ankyrin 1 (TRPA1) and TRP vanilloid 4 (TRPV4) are also involved in aberrant pain perception in preclinical models of somatic pain. Here, we hypothesize that in parallel with TRPV1, histamine sensitizes TRPA1 and TRPV4, contributing to increased visceral pain in patients with IBS. Rectal biopsies were collected from patients with IBS and healthy subjects (HS) to study neuronal sensitivity to TRPA1 and TRPV4 agonists (cinnamaldehyde and GSK1016790A) using intracellular Ca2+ imaging. In addition, the effect of supernatants of rectal biopsies on patients with IBS and HS was assessed on TRPA1 and TRPV4 responses in murine dorsal root ganglion (DRG) sensory neurons. Finally, we evaluated the role of histamine and histamine 1 receptor (H1R) in TRPA1 and TRPV4 sensitization. Application of TRPA1 and TRPV4 agonists evoked significantly higher peak amplitudes and percentage of responding submucosal neurons in biopsies of patients with IBS compared with HS. In HS, pretreatment with histamine significantly increased the Ca2+ responses to cinnamaldehyde and GSK1016790A, an effect prevented by H1R antagonism. IBS supernatants, but not of HS, sensitized TRPA1 and TRPV4 on DRG neurons. This effect was reproduced by histamine and prevented by H1R antagonism. We demonstrate that the mucosal microenvironment in IBS contains mediators, such as histamine, which sensitize TRPV4 and TRPA1 via H1R activation, most likely contributing to increased visceral pain perception in IBS. These data further underscore H1R antagonism as potential treatment for IBS. NEW & NOTEWORTHY We provide evidence for histamine-mediated transient receptor potential (TRP) ankyrin 1 and TRP vanilloid 4 sensitization in irritable bowel syndrome (IBS) via histamine 1 receptor (H1R) activation, most likely contributing to increased visceral pain perception. Our results reveal a general role of sensory TRP channels as histamine effectors in the pathophysiology of IBS and provide novel mechanistic insights into the therapeutic potential of H1R antagonism in IBS.
Collapse
Affiliation(s)
- D Balemans
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, Katholieke Universiteit Leuven , Leuven , Belgium
| | - J Aguilera-Lizarraga
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, Katholieke Universiteit Leuven , Leuven , Belgium
| | - M V Florens
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, Katholieke Universiteit Leuven , Leuven , Belgium
| | - P Jain
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, Katholieke Universiteit Leuven , Leuven , Belgium
| | - A Denadai-Souza
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, Katholieke Universiteit Leuven , Leuven , Belgium
| | - M F Viola
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, Katholieke Universiteit Leuven , Leuven , Belgium
| | - Y A Alpizar
- Laboratory of Ion Channel Research and Transient Receptor Potential Channel Research Platform, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven , Leuven , Belgium.,Vlaams Instituut voor Biotechnologie Center for Brain & Disease Research, Katholieke Universiteit Leuven , Belgium
| | - S Van Der Merwe
- Department of Hepatology, University Hospital Leuven, and Department of Chronic Diseases, Metabolism and Ageing, Katholieke Universiteit Leuven , Leuven , Belgium
| | - P Vanden Berghe
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, Katholieke Universiteit Leuven , Leuven , Belgium
| | - K Talavera
- Laboratory of Ion Channel Research and Transient Receptor Potential Channel Research Platform, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven , Leuven , Belgium.,Vlaams Instituut voor Biotechnologie Center for Brain & Disease Research, Katholieke Universiteit Leuven , Belgium
| | - S Vanner
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University , Kingston , Canada
| | - M M Wouters
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, Katholieke Universiteit Leuven , Leuven , Belgium
| | - G E Boeckxstaens
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, Katholieke Universiteit Leuven , Leuven , Belgium
| |
Collapse
|
27
|
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
|
28
|
Brierley SM, Hibberd TJ, Spencer NJ. Spinal Afferent Innervation of the Colon and Rectum. Front Cell Neurosci 2018; 12:467. [PMID: 30564102 PMCID: PMC6288476 DOI: 10.3389/fncel.2018.00467] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/16/2018] [Indexed: 12/12/2022] Open
Abstract
Despite their seemingly elementary roles, the colon and rectum undertake a variety of key processes to ensure our overall wellbeing. Such processes are coordinated by the transmission of sensory signals from the periphery to the central nervous system, allowing communication from the gut to the brain via the "gut-brain axis". These signals are transmitted from the peripheral terminals of extrinsic sensory nerve fibers, located within the wall of the colon or rectum, and via their axons within the spinal splanchnic and pelvic nerves to the spinal cord. Recent studies utilizing electrophysiological, anatomical and gene expression techniques indicate a surprisingly diverse set of distinct afferent subclasses, which innervate all layers of the colon and rectum. Combined these afferent sub-types allow the detection of luminal contents, low- and high-intensity stretch or contraction, in addition to the detection of inflammatory, immune, and microbial mediators. To add further complexity, the proportions of these afferents vary within splanchnic and pelvic pathways, whilst the density of the splanchnic and pelvic innervation also varies along the colon and rectum. In this review we traverse this complicated landscape to elucidate afferent function, structure, and nomenclature to provide insights into how the extrinsic sensory afferent innervation of the colon and rectum gives rise to physiological defecatory reflexes and sensations of discomfort, bloating, urgency, and pain.
Collapse
Affiliation(s)
- Stuart M Brierley
- Visceral Pain Research Group, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide, Adelaide, SA, Australia
| | - Timothy J Hibberd
- Visceral Neurophysiology Laboratory, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Nick J Spencer
- Visceral Neurophysiology Laboratory, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| |
Collapse
|
29
|
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
|
30
|
De Gregori M, Belfer I, De Giorgio R, Marchesini M, Muscoli C, Rondanelli M, Martini D, Mena P, Arranz LI, Lorente-Cebrián S, Perna S, Villarini A, Salamone M, Allegri M, Schatman ME. Second edition of SIMPAR's "Feed Your Destiny" workshop: the role of lifestyle in improving pain management. J Pain Res 2018; 11:1627-1636. [PMID: 30214272 PMCID: PMC6118253 DOI: 10.2147/jpr.s160660] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
This review is aimed to summarize the latest data regarding pain and nutrition, which have emerged during the second edition of Feed Your Destiny (FYD). Theme presentations and interactive discussions were held at a workshop on March 30, 2017, in Florence, Italy, during the 9th Annual Meeting of Study in Multidisciplinary Pain Research, where an international faculty, including recognized experts in nutrition and pain, reported the scientific evidence on this topic from various perspectives. Presentations were divided into two sections. In the initial sessions, we analyzed the outcome variables and methods of measurement for health claims pertaining to pain proposed under Regulation EC No 1924/2006 of the European Parliament and of the Council of 20 December 2006 on nutrition and health claims made on foods. Moreover, we evaluated how the Mediterranean diet can have a potential impact on pain, gastrointestinal disorders, obesity, cancer, and aging. Second, we discussed the evidence regarding vitamin D as a nutraceutical that may contribute to pain control, evaluating the interindividual variability of pain nature and nurture, and the role of micro-RNAs (miRNAs), polyunsaturated omega 3 fatty acids, and phenolic compounds, with a final revision of the clinical role of nutrition in tailoring pain therapy. The key take-home message provided by the FYD workshop was that a balanced, personalized nutritional regimen might play a role as a synergic strategy that can improve management of chronic pain through a precision medicine approach.
Collapse
Affiliation(s)
- Manuela De Gregori
- Pain Therapy Service, Fondazione IRCCS Polclinico San Matteo, Pavia, Italy, .,Study in Multidisciplinary Pain Research Group, Parma, Italy, .,Young Against Pain Group, Parma, Italy,
| | - Inna Belfer
- Study in Multidisciplinary Pain Research Group, Parma, Italy, .,Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Roberto De Giorgio
- Department of Clinical Sciences, Nuovo Arcispedale S. Anna, University of Ferrara, Ferrara, Italy
| | - Maurizio Marchesini
- Study in Multidisciplinary Pain Research Group, Parma, Italy, .,Young Against Pain Group, Parma, Italy, .,Anesthesia, Intensive Care and Pain Therapy Service, Azienda Ospedaliero, Universitaria of Parma, Parma, Italy
| | - Carolina Muscoli
- Department of Health Sciences, Institute of Research for Food Safety and Health, University "Magna Graecia" of Catanzaro, Parma, Italy
| | - Mariangela Rondanelli
- Study in Multidisciplinary Pain Research Group, Parma, Italy, .,Department of Public Health, Section of Human Nutrition and Dietetics, Azienda di Servizi alla Persona di Pavia, University of Pavia, Pavia, Italy
| | - Daniela Martini
- Human Nutrition Unit, Department of Food & Drugs, University of Parma, Parma, Italy
| | - Pedro Mena
- Human Nutrition Unit, Department of Food & Drugs, University of Parma, Parma, Italy
| | - Laura Isabel Arranz
- Study in Multidisciplinary Pain Research Group, Parma, Italy, .,Department of Nutrition, Food Sciences and Gastronomy, University of Barcelona, Barcelona, Spain
| | - Silvia Lorente-Cebrián
- Study in Multidisciplinary Pain Research Group, Parma, Italy, .,Department of Nutrition, Food Science and Physiology, Faculty of Pharmacy, Center for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Simone Perna
- Department of Public Health, Section of Human Nutrition and Dietetics, Azienda di Servizi alla Persona di Pavia, University of Pavia, Pavia, Italy
| | - Anna Villarini
- Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maurizio Salamone
- Study in Multidisciplinary Pain Research Group, Parma, Italy, .,Science department, Metagenics Italia srl, Milano, Italy.,Società internazionale di Neuropsicocardiologia, Trapani, Italy
| | - Massimo Allegri
- Study in Multidisciplinary Pain Research Group, Parma, Italy, .,Anesthesia and Intensive Care Service - IRCCS MultiMedica Hospital, Sesto San Giovanni, Milano, Italy
| | - Michael E Schatman
- Study in Multidisciplinary Pain Research Group, Parma, Italy, .,Research and Network Development, Boston Pain Care, Waltham, MA, USA.,Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
31
|
Rizopoulos T, Papadaki-Petrou H, Assimakopoulou M. Expression Profiling of the Transient Receptor Potential Vanilloid (TRPV) Channels 1, 2, 3 and 4 in Mucosal Epithelium of Human Ulcerative Colitis. Cells 2018; 7:E61. [PMID: 29914124 PMCID: PMC6025154 DOI: 10.3390/cells7060061] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/11/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022] Open
Abstract
The Transient Receptor Potential (TRP) family of selective and non-selective ion channels is well represented throughout the mammalian gastrointestinal track. Several members of the Transient Receptor Potential Vanilloid (TRPV) subfamily have been identified in contributing to modulation of mobility, secretion and sensitivity of the human intestine. Previous studies have focused on the detection of TRPV mRNA levels in colon tissue of patients with inflammatory bowel disease (IBD) whereas little information exists regarding TRPV channel expression in the colonic epithelium. The aim of this study was to evaluate the expression levels of TRPV1, TRPV2, TRPV3 and TRPV4 in mucosa epithelial cells of colonic biopsies from patients with ulcerative colitis (UC) in comparison to colonic resections from non-IBD patients (control group). Immunohistochemistry, using specific antibodies and quantitative analyses of TRPV-immunostained epithelial cells, was performed in semi-serial sections of the samples. TRPV1 expression was significantly decreased whereas TRPV4 expression was significantly increased in the colonic epithelium of UC patients compared to patients in the control group (p < 0.05). No significant difference for TRPV2 and TRPV3 expression levels between UC and control specimens was detected (p > 0.05). There was no correlation between TRPV channel expression and the clinical features of the disease (p > 0.05). Further investigation is needed to clarify the role of TRPV channels in human bowel inflammatory response.
Collapse
Affiliation(s)
- Theodoros Rizopoulos
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, Rion 26504, Greece.
| | - Helen Papadaki-Petrou
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, Rion 26504, Greece.
| | - Martha Assimakopoulou
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, Rion 26504, Greece.
| |
Collapse
|
32
|
Nedi T, White PJ, Coupar IM, Irving HR. Effect of the 5-HT 4 receptor agonist tegaserod on the expression of GRK2 and GRK6 in the rat gastrointestinal tract. BMC Res Notes 2018; 11:362. [PMID: 29884229 PMCID: PMC5994065 DOI: 10.1186/s13104-018-3495-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 06/06/2018] [Indexed: 12/31/2022] Open
Abstract
Objective Tegaserod is a 5-hydroxytryptamine type 4 (5-HT4) receptor agonist, formerly used in treating constipation predominant irritable bowel syndrome, which desensitizes 5-HT4 receptors in rat oesophagus and colon in vitro. Desensitization of 5-HT4 receptors is regulated by G-protein coupled receptor kinases. This study was designed to assess the effect of 5-HT4 receptor activation on the expression of GRK2 and GRK6 in the rat oesophagus and distal colon by acute administration of tegaserod. Results Rats were treated with a single dose of tegaserod (5 mg/kg) and tissue samples of the oesophagus and distal colon were prepared and level of GRK2 and GRK6 protein expression was determined using western blotting. The immunodensity of GRK2 and GRK6 was normalized against the loading control β-actin and compared with control animals. Acute administration of tegaserod for 1, 2, 3, 4, 6, and 8 h did not change significantly the immunodensity of GRK2 or GRK6 in the oesophagus or GRK2 in the distal colon when compared with control animals. This may indicate that the basal level of GRK2 and GRK6 expression is sufficient to regulate the desensitization of 5-HT4 receptors in acute drug treatment.
Collapse
Affiliation(s)
- Teshome Nedi
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC, 3052, Australia.,School of Pharmacy, College of Health Sciences, Addis Ababa University, PO Box 1176, Addis Ababa, Ethiopia
| | - Paul J White
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Ian M Coupar
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Helen R Irving
- Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC, 3052, Australia. .,La Trobe Institute for Molecular Science, La Trobe University, PO Box 199, Bendigo, VIC, 3552, Australia.
| |
Collapse
|
33
|
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]
|
34
|
Peiris M, Hockley JR, Reed DE, Smith ESJ, Bulmer DC, Blackshaw LA. Peripheral K V7 channels regulate visceral sensory function in mouse and human colon. Mol Pain 2018; 13:1744806917709371. [PMID: 28566000 PMCID: PMC5456027 DOI: 10.1177/1744806917709371] [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: 12/18/2022] Open
Abstract
Background Chronic visceral pain is a defining symptom of many gastrointestinal disorders. The KV7 family (KV7.1–KV7.5) of voltage-gated potassium channels mediates the M current that regulates excitability in peripheral sensory nociceptors and central pain pathways. Here, we use a combination of immunohistochemistry, gut-nerve electrophysiological recordings in both mouse and human tissues, and single-cell qualitative real-time polymerase chain reaction of gut-projecting sensory neurons, to investigate the contribution of peripheral KV7 channels to visceral nociception. Results Immunohistochemical staining of mouse colon revealed labelling of KV7 subtypes (KV7.3 and KV7.5) with CGRP around intrinsic enteric neurons of the myenteric plexuses and within extrinsic sensory fibres along mesenteric blood vessels. Treatment with the KV7 opener retigabine almost completely abolished visceral afferent firing evoked by the algogen bradykinin, in agreement with significant co-expression of mRNA transcripts by single-cell qualitative real-time polymerase chain reaction for KCNQ subtypes and the B2 bradykinin receptor in retrogradely labelled extrinsic sensory neurons from the colon. Retigabine also attenuated responses to mechanical stimulation of the bowel following noxious distension (0–80 mmHg) in a concentration-dependent manner, whereas the KV7 blocker XE991 potentiated such responses. In human bowel tissues, KV7.3 and KV7.5 were expressed in neuronal varicosities co-labelled with synaptophysin and CGRP, and retigabine inhibited bradykinin-induced afferent activation in afferent recordings from human colon. Conclusions We show that KV7 channels contribute to the sensitivity of visceral sensory neurons to noxious chemical and mechanical stimuli in both mouse and human gut tissues. As such, peripherally restricted KV7 openers may represent a viable therapeutic modality for the treatment of gastrointestinal pathologies.
Collapse
Affiliation(s)
- Madusha Peiris
- 1 Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - James Rf Hockley
- 2 Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - David E Reed
- 3 GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | | | - David C Bulmer
- 1 Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - L Ashley Blackshaw
- 1 Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
35
|
Reed DE, Vanner SJ. Emerging studies of human visceral nociceptors. Am J Physiol Gastrointest Liver Physiol 2017; 312:G201-G207. [PMID: 28007748 DOI: 10.1152/ajpgi.00391.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 01/31/2023]
Abstract
Animal studies have led to significant advances in our understanding of pain mechanisms in the intestine that could lead to altered signaling in disorders such as irritable bowel syndrome. However, how these translate to the human afferent nervous system is unclear. Recent studies have demonstrated that it is possible to use a variety of techniques, including electrophysiological recordings, to begin to examine these concepts in humans. This mini-review examines these studies to explore how well animal studies translate to humans suffering from irritable bowel syndrome, highlights some of the advantages and technical limitations of these approaches, and identifies some priorities for future studies using human tissues.
Collapse
Affiliation(s)
- David E Reed
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, Ontario, Canada
| | - Stephen J Vanner
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
36
|
Ng KS, Brookes SJ, Montes-Adrian NA, Mahns DA, Gladman MA. Electrophysiological characterization of human rectal afferents. Am J Physiol Gastrointest Liver Physiol 2016; 311:G1047-G1055. [PMID: 27789454 PMCID: PMC5298880 DOI: 10.1152/ajpgi.00153.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 10/17/2016] [Indexed: 01/31/2023]
Abstract
It is presumed that extrinsic afferent nerves link the rectum to the central nervous system. However, the anatomical/functional existence of such nerves has never previously been demonstrated in humans. Therefore, we aimed to identify and make electrophysiological recordings in vitro from extrinsic afferents, comparing human rectum to colon. Sections of normal rectum and colon were procured from anterior resection and right hemicolectomy specimens, respectively. Sections were pinned and extrinsic nerves dissected. Extracellular visceral afferent nerve activity was recorded. Neuronal responses to chemical [capsaicin and "inflammatory soup" (IS)] and mechanical (Von Frey probing) stimuli were recorded and quantified as peak firing rate (range) in 1-s intervals. Twenty-eight separate nerve trunks from eight rectums were studied. Of these, spontaneous multiunit afferent activity was recorded in 24 nerves. Peak firing rates increased significantly following capsaicin [median 6 (range 3-25) spikes/s vs. 2 (1-4), P < 0.001] and IS [median 5 (range 2-18) spikes/s vs. 2 (1-4), P < 0.001]. Mechanosensitive "hot spots" were identified in 16 nerves [median threshold 2.0 g (range 1.4-6.0 g)]. In eight of these, the threshold decreased after IS [1.0 g (0.4-1.4 g)]. By comparison, spontaneous activity was recorded in only 3/30 nerves studied from 10 colons, and only one hot spot (threshold 60 g) was identified. This study confirms the anatomical/functional existence of extrinsic rectal afferent nerves and characterizes their chemo- and mechanosensitivity for the first time in humans. They have different electrophysiological properties to colonic afferents and warrant further investigation in disease states.
Collapse
Affiliation(s)
- Kheng-Seong Ng
- 1Academic Colorectal Unit, Sydney Medical School, Concord, University of Sydney, Sydney, Australia; ,2Enteric Neuroscience and Gastrointestinal Research Group, ANZAC Research Institute, University of Sydney, Sydney, Australia;
| | - Simon J. Brookes
- 3Discipline of Human Physiology, FMST, School of Medicine, Flinders University, Adelaide, Australia; and
| | - Noemi A. Montes-Adrian
- 2Enteric Neuroscience and Gastrointestinal Research Group, ANZAC Research Institute, University of Sydney, Sydney, Australia;
| | - David A. Mahns
- 4Department of Integrative Physiology, School of Medicine, Western Sydney University, Sydney, Australia
| | - Marc A. Gladman
- 1Academic Colorectal Unit, Sydney Medical School, Concord, University of Sydney, Sydney, Australia; ,2Enteric Neuroscience and Gastrointestinal Research Group, ANZAC Research Institute, University of Sydney, Sydney, Australia;
| |
Collapse
|