101
|
Darbaky Y, Evrard B, Patrier S, Falenta J, Garcin S, Tridon A, Dapoigny M, Silberberg C, Nivoliez A, Diop L. Oral probiotic treatment of Lactobacillus rhamnosus Lcr35 ® prevents visceral hypersensitivity to a colonic inflammation and an acute psychological stress. J Appl Microbiol 2016; 122:188-200. [PMID: 27718511 DOI: 10.1111/jam.13320] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 09/05/2016] [Accepted: 10/05/2016] [Indexed: 12/16/2022]
Abstract
AIMS This study evaluated the efficacy of a repeated oral treatment with two active pharmaceutical ingredients (Lcr Lenio® and Lcr Restituo® ) derivated from the probiotic bacterial strain Lactobacillus rhamnosus Lcr35® in two animal models mimicking different features of irritable bowel syndrome (IBS). IBS is characterized by visceral pain associated with alteration of bowel transit. IBS patients present visceral hypersensitivity with peripheral and central origins. METHODS AND RESULTS The injection of 2,4,6-trinitrobenzenesulfonic acid (TNBS) into the proximal colon as well as an acute partial restraint stress (PRS) produces colonic hypersensitivity measured in conscious rats by a decrease in pain threshold in response to distal colonic distension. Visceral hypersensitivity was produced by injection of TNBS 7 days before colonic distension or by acute PRS on testing day. Treatments were performed once a day during eight consecutive days. CONCLUSIONS This study indicates that an 8-day probiotic treatment (Lcr Lenio and Lcr Restituo) produces an antihypersensitivity activity in both TNBS and PRS visceral pain models. As this probiotic strain attenuates peripherally and centrally induced visceral hypersensitivity in rats, it may be active in treatment of IBS symptoms. An immunomodulatory effect of the probiotics was highlighted in the TNBS model on the IL-23 secretion, suggesting a mechanism of action involving a regulation of the local IL-23/Th17 immune activation. SIGNIFICANCE AND IMPACT OF THE STUDY Two formulas of Lcr35® probiotic strain show very encouraging results for the treatment of IBS patients. Further studies are needed to better understand the role and mechanisms of probiotics on the pathogenesis of IBS.
Collapse
Affiliation(s)
| | - B Evrard
- Laboratoire d'Immunologie, Université d'Auvergne-Clermont 1, Clermont-Ferrand, France
| | - S Patrier
- Département Recherche et Développement-Biose®, Arpajon-sur-Cère, France
| | - J Falenta
- Laboratoire d'Immunologie, Université d'Auvergne-Clermont 1, Clermont-Ferrand, France
| | - S Garcin
- Laboratoire d'Immunologie, Université d'Auvergne-Clermont 1, Clermont-Ferrand, France
| | - A Tridon
- Laboratoire d'Immunologie, Université d'Auvergne-Clermont 1, Clermont-Ferrand, France
| | - M Dapoigny
- Médecine Digestive, Centre Hospitalier Universitaire (CHU) Estaing, CHU Clermont Université, Clermont-Ferrand, France
| | | | - A Nivoliez
- Département Recherche et Développement-Biose®, Arpajon-sur-Cère, France
| | - L Diop
- ANS Biotech, Riom, France
| |
Collapse
|
102
|
Genetics, Mucosal Inflammation and the Environment in Post-Infectious Chronic Gut Syndromes. ACTA ACUST UNITED AC 2016. [DOI: 10.1038/ajgsup.2016.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
103
|
Nullens S, Deiteren A, Jiang W, Keating C, Ceuleers H, Francque S, Grundy D, De Man JG, De Winter BY. In Vitro Recording of Mesenteric Afferent Nerve Activity in Mouse Jejunal and Colonic Segments. J Vis Exp 2016:54576. [PMID: 27805592 PMCID: PMC5092238 DOI: 10.3791/54576] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Afferent nerves not only convey information concerning normal physiology, but also signal disturbed homeostasis and pathophysiological processes of the different organ systems from the periphery towards the central nervous system. As such, the increased activity or 'sensitization' of mesenteric afferent nerves has been allocated an important role in the pathophysiology of visceral hypersensitivity and abdominal pain syndromes. Mesenteric afferent nerve activity can be measured in vitro in an isolated intestinal segment that is mounted in a purpose-built organ bath and from which the splanchnic nerve is isolated, allowing researchers to directly assess nerve activity adjacent to the gastrointestinal segment. Activity can be recorded at baseline in standardized conditions, during distension of the segment or following the addition of pharmacological compounds delivered intraluminally or serosally. This technique allows the researcher to easily study the effect of drugs targeting the peripheral nervous system in control specimens; besides, it provides crucial information on how neuronal activity is altered during disease. It should be noted however that measuring afferent neuronal firing activity only constitutes one relay station in the complex neuronal signaling cascade, and researchers should bear in mind not to overlook neuronal activity at other levels (e.g., dorsal root ganglia, spinal cord or central nervous system) in order to fully elucidate the complex neuronal physiology in health and disease. Commonly used applications include the study of neuronal activity in response to the administration of lipopolysaccharide, and the study of afferent nerve activity in animal models of irritable bowel syndrome. In a more translational approach, the isolated mouse intestinal segment can be exposed to colonic supernatants from IBS patients. Furthermore, a modification of this technique has been recently shown to be applicable in human colonic specimens.
Collapse
Affiliation(s)
- Sara Nullens
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp
| | - Annemie Deiteren
- Visceral Pain Group, Discipline of Medicine, University of Adelaide
| | - Wen Jiang
- Department of Biomedical Sciences, University of Sheffield
| | - Christopher Keating
- Department of Pharmacy, Pharmacology and Postgraduate Medicine, University of Hertfordshire
| | - Hannah Ceuleers
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital
| | - David Grundy
- Department of Biomedical Sciences, University of Sheffield
| | - Joris G De Man
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp
| | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp;
| |
Collapse
|
104
|
Reiss D, Ceredig RA, Secher T, Boué J, Barreau F, Dietrich G, Gavériaux-Ruff C. Mu and delta opioid receptor knockout mice show increased colonic sensitivity. Eur J Pain 2016; 21:623-634. [PMID: 27748566 DOI: 10.1002/ejp.965] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Opiates act through opioid receptors to diminish pain. Here, we investigated whether mu (MOR) and delta (DOR) receptor endogenous activity assessed in the whole mouse body or in particular at peripheral receptors on primary nociceptive neurons, control colonic pain. METHODS We compared global MOR and DOR receptor knockout (KO) mice, mice with a conditional deletion of MOR and DOR in Nav1.8-positive nociceptive primary afferent neurons (cKO), and control floxed mice of both genders for visceral sensitivity. Visceromotor responses to colorectal distension (CRD) and macroscopic colon scores were recorded on naïve mice and mice with acute colitis induced by 3% dextran sodium sulphate (DSS) for 5 days. Transcript expression for opioid genes and cytokines was measured by quantitative RT-PCR. RESULTS Naïve MOR and DOR global KO mice show increased visceral sensitivity that was not observed in cKO mice. MOR and preproenkephalin (Penk) were the most expressed opioid genes in colon. MOR KO mice had augmented kappa opioid receptor and Tumour-Necrosis-Factor-α and diminished Penk transcript levels while DOR, preprodynorphin and Interleukin-1β were unchanged. Global MOR KO females had a thicker colon than floxed females. No alteration was detected in DOR mutant animals. A 5-day DSS treatment led to comparable hypersensitivity in the different mouse lines. CONCLUSION Our results suggest that mu and delta opioid receptor global endogenous activity but not activity at the peripheral Nav1.8 neurons contribute to visceral sensitivity in naïve mice, and that endogenous MOR and DOR tones were insufficient to elicit analgesia after 5-day DSS-induced colitis. SIGNIFICANCE Knockout mice for mu and delta opioid receptor have augmented colon sensitivity in the CRD assay. It shows endogenous mu and delta opioid analgesia that may be explored as potential targets for alleviating chronic intestinal pain.
Collapse
Affiliation(s)
- D Reiss
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
| | - R A Ceredig
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Institut des Neurosciences Cellulaires et Intégratives INCI, UPR3212, Strasbourg, France
| | - T Secher
- Institut de Recherche en Santé Digestive IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - J Boué
- Institut de Recherche en Santé Digestive IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - F Barreau
- Institut de Recherche en Santé Digestive IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - G Dietrich
- Institut de Recherche en Santé Digestive IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - C Gavériaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Ecole Supérieure de Biotechnologie de Strasbourg, Université de Strasbourg, France
| |
Collapse
|
105
|
Holtmann GJ, Ford AC, Talley NJ. Pathophysiology of irritable bowel syndrome. Lancet Gastroenterol Hepatol 2016; 1:133-146. [DOI: 10.1016/s2468-1253(16)30023-1] [Citation(s) in RCA: 256] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 06/20/2016] [Accepted: 06/24/2016] [Indexed: 11/25/2022]
|
106
|
Changes in the Interstitial Cells of Cajal and Immunity in Chronic Psychological Stress Rats and Therapeutic Effects of Acupuncture at the Zusanli Point (ST36). EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:1935372. [PMID: 27594888 PMCID: PMC4987473 DOI: 10.1155/2016/1935372] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 03/03/2016] [Accepted: 05/18/2016] [Indexed: 02/06/2023]
Abstract
Now, chronic psychological stress (CPS) related diseases are increasing. Many CPS patients have gastrointestinal complaints, immune suppression, and immune imbalance. Increasing evidence is indicating that acupuncture (AP) at the Zusanli point (ST36) can alleviate functional gastrointestinal disorders (FGID), immune suppression, and immune imbalance. However, few studies have investigated the potential mechanisms. In this study, CPS rat models were established, and electroacupuncture (EA) at ST36 was done for CPS rats. Daily food intake, weight, intestinal sensitivity, the morphology of interstitial cell of Cajal (ICC) in the small intestine, and serum indexes were measured. The study found that, in CPS rats, EA at ST36 could improve food intake, weight, visceral hypersensitivity, and immunity; in CPS rats, in small intestine, the morphology of ICCs was abnormal and the number was decreased, which may be part causes of gastrointestinal motility dysfunction. EA at ST36 showed useful therapeutic effects. The mechanisms may be partially related to its repairing effects on ICCs damages; in CPS rats, there were immune suppression and immune imbalance, which may be part causes of visceral hypersensitivity. EA at ST36 showed useful therapeutic effects. The mechanisms may be partially related to its regulation on immunity.
Collapse
|
107
|
Hughes PA, Costello SP, Bryant RV, Andrews JM. Opioidergic effects on enteric and sensory nerves in the lower GI tract: basic mechanisms and clinical implications. Am J Physiol Gastrointest Liver Physiol 2016; 311:G501-13. [PMID: 27469369 DOI: 10.1152/ajpgi.00442.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 07/21/2016] [Indexed: 01/31/2023]
Abstract
Opioids are one of the most prescribed drug classes for treating acute pain. However, chronic use is often associated with tolerance as well as debilitating side effects, including nausea and dependence, which are mediated by the central nervous system, as well as constipation emerging from effects on the enteric nervous system. These gastrointestinal (GI) side effects limit the usefulness of opioids in treating pain in many patients. Understanding the mechanism(s) of action of opioids on the nervous system that shows clinical benefit as well as those that have unwanted effects is critical for the improvement of opioid drugs. The opioidergic system comprises three classical receptors (μ, δ, κ) and a nonclassical receptor (nociceptin), and each of these receptors is expressed to varying extents by the enteric and intestinal extrinsic sensory afferent nerves. The purpose of this review is to discuss the role that the opioidergic system has on enteric and extrinsic afferent nerves in the lower GI tract in health and diseases of the lower GI tract, particularly inflammatory bowel disease and irritable bowel syndrome, and the implications of opioid treatment on clinical outcomes. Consideration is also given to emerging developments in our understanding of the immune system as a novel source of endogenous opioids and the mechanisms underlying opioid tolerance, including the potential influence of opioid receptor splice variants and heteromeric complexes.
Collapse
Affiliation(s)
- Patrick A Hughes
- Centre for Nutrition and Gastrointestinal Disease, Department of Medicine, University of Adelaide and South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia; School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia;
| | - Samuel P Costello
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, South Australia, Australia; and Department of Gastroenterology, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| | - Robert V Bryant
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, South Australia, Australia; and
| | - Jane M Andrews
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, South Australia, Australia; and
| |
Collapse
|
108
|
Correlation of Serum β-Endorphin and the Quality of Life in Allergic Rhinitis. DISEASE MARKERS 2016; 2016:2025418. [PMID: 27647946 PMCID: PMC5014966 DOI: 10.1155/2016/2025418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 08/02/2016] [Indexed: 11/18/2022]
Abstract
Background. Allergic rhinitis (AR) significantly impairs the quality of life of the patients; however, a questionnaire alone is an insufficient and subjective measure of this condition. Obtaining an objective clinical assessment of the level of impairment will be valuable for its treatment. β-Endorphin is one of the most important mediators of both mental state and specific immunity. Thus, we investigated the possibility of using β-endorphin as a biomarker for evaluating the impairment level in AR. Methods. This study included 48 patients with AR and 32 healthy volunteers. The serum β-endorphin level was determined by enzyme immunoassay, and the serum-specific IgE and total IgE levels were determined by immunoblot assay. The Rhinoconjunctivitis Quality of Life Questionnaire (RQLQ) was used to assess the impairment level in the symptom duration. Results. The β-endorphin concentration was significantly decreased in AR patients compared to the healthy controls (p = 0.000, p < 0.05). There was significant negative correlation between the impairment level and serum β-endorphin level (correlation coefficient: −0.468; p = 0.001; p < 0.05), but there was no association between the serum β-endorphin and total IgE levels (p = 0.947, p > 0.05). Conclusion. β-Endorphin is a systemic biomarker that has the potential to assess the impairment level in AR and may therefore be a novel therapeutic target for the treatment of AR.
Collapse
|
109
|
Ratanasirintrawoot S, Israsena N. Stem Cells in the Intestine: Possible Roles in Pathogenesis of Irritable Bowel Syndrome. J Neurogastroenterol Motil 2016; 22:367-82. [PMID: 27184041 PMCID: PMC4930294 DOI: 10.5056/jnm16023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/08/2016] [Indexed: 12/13/2022] Open
Abstract
Irritable bowel syndrome is one of the most common functional gastrointestinal (GI) disorders that significantly impair quality of life in patients. Current available treatments are still not effective and the pathophysiology of this condition remains unclearly defined. Recently, research on intestinal stem cells has greatly advanced our understanding of various GI disorders. Alterations in conserved stem cell regulatory pathways such as Notch, Wnt, and bone morphogenic protein/TGF-β have been well documented in diseases such as inflammatory bowel diseases and cancer. Interaction between intestinal stem cells and various signals from their environment is important for the control of stem cell self-renewal, regulation of number and function of specific intestinal cell types, and maintenance of the mucosal barrier. Besides their roles in stem cell regulation, these signals are also known to have potent effects on immune cells, enteric nervous system and secretory cells in the gut, and may be responsible for various aspects of pathogenesis of functional GI disorders, including visceral hypersensitivity, altered gut motility and low grade gut inflammation. In this article, we briefly summarize the components of these signaling pathways, how they can be modified by extrinsic factors and novel treatments, and provide evidenced support of their roles in the inflammation processes. Furthermore, we propose how changes in these signals may contribute to the symptom development and pathogenesis of irritable bowel syndrome.
Collapse
Affiliation(s)
- Sutheera Ratanasirintrawoot
- Stem Cell and Cell Therapy Research Unit, Chulalongkorn University, Bangkok, Thailand.,Department of Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nipan Israsena
- Stem Cell and Cell Therapy Research Unit, Chulalongkorn University, Bangkok, Thailand.,Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
110
|
Basso L, Boué J, Mahiddine K, Blanpied C, Robiou-du-Pont S, Vergnolle N, Deraison C, Dietrich G. Endogenous analgesia mediated by CD4(+) T lymphocytes is dependent on enkephalins in mice. J Neuroinflammation 2016; 13:132. [PMID: 27245576 PMCID: PMC4888630 DOI: 10.1186/s12974-016-0591-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/20/2016] [Indexed: 12/31/2022] Open
Abstract
Background T cell-derived opioids play a key role in the control of inflammatory pain. However, the nature of opioids produced by T cells is still matter of debate in mice. Whereas β-endorphin has been found in T lymphocytes by using antibody-based methods, messenger RNA (mRNA) quantification shows mainly mRNA encoding for enkephalins. The objective of the study is to elucidate the nature of T cell-derived opioids responsible for analgesia and clarify discrepancy of the results at the protein and genetic levels. Methods CD4+ T lymphocytes were isolated from wild-type and enkephalin-deficient mice. mRNA encoding for β-endorphin and enkephalin was quantified by RT-qPCR. The binding of commercially available polyclonal anti-endorphin antibodies to lymphocytes from wild-type or enkephalin knockout mice was assessed by cytofluorometry. Opioid-mediated analgesic properties of T lymphocytes from wild-type and enkephalin-deficient mice were compared in a model of inflammation-induced somatic pain by measuring sensitivity to mechanical stimuli using calibrated von Frey filaments. Results CD4+ T lymphocytes expressed high level of mRNA encoding for enkephalins but not for β-endorphin in mice. Anti-β-endorphin polyclonal IgG antibodies are specific for β-endorphin but cross-react with enkephalins. Anti-β-endorphin polyclonal antibodies bound to wild-type but not enkephalin-deficient CD4+ T lymphocytes. Endogenous regulation of inflammatory pain by wild-type T lymphocytes was completely abolished when T lymphocytes were deficient in enkephalins. Pain behavior of immune-deficient (i.e., without B and T lymphocytes) mice was superimposable to that of mice transferred with enkephalin-deficient lymphocytes. Conclusions Rabbit polyclonal anti-β-endorphin serum IgG bind to CD4+ T lymphocytes because of their cross-reactivity towards enkephalins. Thus, staining of T lymphocytes by anti-β-endorphin polyclonal IgG reported in most of studies in mice is because of their binding to enkephalins. In mice, CD4+ T lymphocytes completely lose their analgesic opioid-mediated activity when lacking enkephalins.
Collapse
Affiliation(s)
- Lilian Basso
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Jérôme Boué
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Karim Mahiddine
- CPTP, Université de Toulouse, CNRS, INSERM, UPS, Toulouse, France
| | | | | | | | - Céline Deraison
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Gilles Dietrich
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.
| |
Collapse
|
111
|
Hughes PA. Sleeping in on pancreatic cancer pain: Schwann cell secreted IL-6 pushes snooze on the pain alarm. Gut 2016; 65:897-8. [PMID: 26864073 DOI: 10.1136/gutjnl-2015-311272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/18/2016] [Indexed: 12/08/2022]
|
112
|
Sobolewska-Włodarczyk A, Włodarczyk M, Storr M, Fichna J. Clinical potential of eluxadoline in the treatment of diarrhea-predominant irritable bowel syndrome. Ther Clin Risk Manag 2016; 12:771-5. [PMID: 27257381 PMCID: PMC4874728 DOI: 10.2147/tcrm.s83722] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Diarrhea-predominant irritable bowel syndrome (IBS-D) belongs to the group of functional gastrointestinal disorders and is characterized by abdominal pain in conjunction with diarrhea. The incidence of IBS-D is currently increasing, leading to a heavy economic burden for patients and health care systems worldwide. Recent studies suggest eluxadoline as an attractive new tool for the treatment of patients with IBS-D. Eluxadoline is an orally active μ- and κ-opioid receptor agonist and δ-opioid receptor antagonist, with powerful antidiarrheal and analgesic activity. Eluxadoline is believed to act locally in the enteric nervous system, and has no adverse effects in the central nervous system. In this review, we discuss the most recent findings on the mechanism of action of eluxadoline and the results of the clinical trials in patients with IBS-D. We also discuss possible side effects and analyze the potential of eluxadoline to be used in the treatment of IBS-D.
Collapse
Affiliation(s)
| | - Marcin Włodarczyk
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | | | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
113
|
Enck P, Aziz Q, Barbara G, Farmer AD, Fukudo S, Mayer EA, Niesler B, Quigley EMM, Rajilić-Stojanović M, Schemann M, Schwille-Kiuntke J, Simren M, Zipfel S, Spiller RC. Irritable bowel syndrome. Nat Rev Dis Primers 2016; 2:16014. [PMID: 27159638 PMCID: PMC5001845 DOI: 10.1038/nrdp.2016.14] [Citation(s) in RCA: 633] [Impact Index Per Article: 70.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disease with a high population prevalence. The disorder can be debilitating in some patients, whereas others may have mild or moderate symptoms. The most important single risk factors are female sex, younger age and preceding gastrointestinal infections. Clinical symptoms of IBS include abdominal pain or discomfort, stool irregularities and bloating, as well as other somatic, visceral and psychiatric comorbidities. Currently, the diagnosis of IBS is based on symptoms and the exclusion of other organic diseases, and therapy includes drug treatment of the predominant symptoms, nutrition and psychotherapy. Although the underlying pathogenesis is far from understood, aetiological factors include increased epithelial hyperpermeability, dysbiosis, inflammation, visceral hypersensitivity, epigenetics and genetics, and altered brain-gut interactions. IBS considerably affects quality of life and imposes a profound burden on patients, physicians and the health-care system. The past decade has seen remarkable progress in our understanding of functional bowel disorders such as IBS that will be summarized in this Primer.
Collapse
Affiliation(s)
- Paul Enck
- Department of Internal Medicine VI (Psychosomatic Medicine and Psychotherapy), University Hospital Tübingen, Tübingen, Germany
| | - Qasim Aziz
- Wingate Institute of Neurogastroenterology, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Giovanni Barbara
- Department of Medical and Surgical Sciences, St. Orsola-Malpighi Hospital, Bologna, Italy
| | - Adam D Farmer
- Wingate Institute of Neurogastroenterology, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Shin Fukudo
- Department of Behavioural Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Emeran A Mayer
- Oppenheimer Center for Neurobiology of Stress, Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Beate Niesler
- Department of Human Molecular Genetics, University of Heidelberg, Heidelberg, Germany
| | - Eamonn M M Quigley
- Lynda K and David M Underwood Center for Digestive Disorders, Division of Gastroenterology and Hepatology, Houston Methodist Hospital, Weill Cornell Medical College, Houston, Texas, USA
| | - Mirjana Rajilić-Stojanović
- Department of Biochemical Engineering and Biotechnology, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Michael Schemann
- Department of Human Biology, Technical University Munich, Freising-Weihenstephan, Germany
| | - Juliane Schwille-Kiuntke
- Department of Internal Medicine VI (Psychosomatic Medicine and Psychotherapy), University Hospital Tübingen, Tübingen, Germany
| | - Magnus Simren
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stephan Zipfel
- Department of Internal Medicine VI (Psychosomatic Medicine and Psychotherapy), University Hospital Tübingen, Tübingen, Germany
| | - Robin C Spiller
- NIHR Nottingham Digestive Diseases Biomedical Research Unit, University of Nottingham, Nottingham, UK
| |
Collapse
|
114
|
Mahurkar S, Polytarchou C, Iliopoulos D, Pothoulakis C, Mayer EA, Chang L. Genome-wide DNA methylation profiling of peripheral blood mononuclear cells in irritable bowel syndrome. Neurogastroenterol Motil 2016; 28:410-22. [PMID: 26670691 PMCID: PMC4760882 DOI: 10.1111/nmo.12741] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/30/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is a stress-sensitive disorder. Environmental factors including stress can trigger epigenetic changes, which have not been well-studied in IBS. We performed a pilot study investigating genome-wide DNA methylation of IBS patients and healthy controls (HCs) to identify potential epigenetic markers and associated pathways. Additionally, we investigated relationships of epigenetic changes in selected genes with clinical traits. METHODS Twenty-seven IBS patients (59% women; 10 IBS-diarrhea, 8 IBS-constipation, 9 IBS-mixed) and 23 age- and sex-matched HCs were examined. DNA methylation from peripheral blood mononuclear cells (PBMCs) was measured using HM450 BeadChip, and representative methylation differences were confirmed by bisulphite sequencing. Gene expression was measured using quantitative PCR. Gastrointestinal (GI) and non-GI symptoms were measured using validated questionnaires. Associations were tested using non-parametric methods. KEY RESULTS Genome-wide DNA methylation profiling of IBS patients compared with HCs identified 133 differentially methylated positions (DMPs) (mean difference ≥10%; p < 0.05). These genes were associated with gene ontology terms including glutathione metabolism related to oxidative stress and neuropeptide hormone activity. Validation by sequencing confirmed differential methylation of subcommissural organ (SCO)-Spondin (SSPO), glutathione-S-transferases mu 5 (GSTM5), and tubulin polymerization promoting protein genes. Methylation of two promoter CpGs in GSTM5 was associated with epigenetic silencing. Epigenetic changes in SSPO gene were positively correlated with hospital anxiety and depression scores in IBS patients (r > 0.4 and false discovery rate <0.05). CONCLUSIONS & INFERENCES This study is the first to comprehensively explore the methylome of IBS patients. We identified DMPs in novel candidate genes which could provide new insights into disease mechanisms; however, these preliminary findings warrant confirmation in larger, independent studies.
Collapse
Affiliation(s)
- Swapna Mahurkar
- Oppenheimer Center for Neurobiology of Stress at UCLA; Division of Digestive Diseases, David Geffen School of Medicine at UCLA
| | - Christos Polytarchou
- Center for Systems Biomedicine, Division of Digestive Diseases, David Geffen School of Medicine at UCLA
| | - Dimitrios Iliopoulos
- Center for Systems Biomedicine, Division of Digestive Diseases, David Geffen School of Medicine at UCLA
| | - Charalabos Pothoulakis
- Department of Medicine, Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA
| | - Emeran A. Mayer
- Oppenheimer Center for Neurobiology of Stress at UCLA; Division of Digestive Diseases, David Geffen School of Medicine at UCLA
| | - Lin Chang
- Oppenheimer Center for Neurobiology of Stress at UCLA; Division of Digestive Diseases, David Geffen School of Medicine at UCLA
| |
Collapse
|
115
|
Azizian M, Basati G, Abangah G, Mahmoudi MR, Mirzaei A. Contribution of Blastocystishominis subtypes and associated inflammatory factors in development of irritable bowel syndrome. Parasitol Res 2016; 115:2003-9. [DOI: 10.1007/s00436-016-4942-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 01/26/2016] [Indexed: 01/01/2023]
|
116
|
Altered Ion Channel/Receptor Expression and Function in Extrinsic Sensory Neurons: The Cause of and Solution to Chronic Visceral Pain? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 891:75-90. [PMID: 27379637 DOI: 10.1007/978-3-319-27592-5_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The gastrointestinal tract is unique in that it is innervated by several distinct populations of neurons, whose cell bodies are either intrinsic (enteric, viscerofugal) or extrinsic (sympathetic, sensory afferents) to the wall of the gut. We are usually completely unaware of the continuous, complicated orchestra of functions that these neurons conduct. However, for patients with Inflammatory Bowel Disease (IBD) or functional gastrointestinal disorders, such as Functional Dyspepsia (FD) and Irritable Bowel Syndrome (IBS) altered gastrointestinal motility, discomfort and pain are common, debilitating symptoms. Whilst bouts of inflammation underlie the symptoms associated with IBD, over the past few years there is increased pre-clinical and clinical evidence that infection and inflammation are key risk factors for the development of several functional gastrointestinal disorders, in particular IBS. There is a strong correlation between prior exposure to gut infection and symptom occurrence; with the duration and severity of the initial illness the strongest associated risk factors. This review discusses the current body of evidence for neuroplasticity during inflammation and how in many cases fails to reset back to normal, long after healing of the damaged tissues. Recent evidence suggests that the altered expression and function of key ion channels and receptors within extrinsic sensory neurons play fundamental roles in the aberrant pain sensation associated with these gastrointestinal diseases and disorders.
Collapse
|
117
|
Campaniello MA, Harrington AM, Martin CM, Ashley Blackshaw L, Brierley SM, Hughes PA. Activation of colo-rectal high-threshold afferent nerves by Interleukin-2 is tetrodotoxin-sensitive and upregulated in a mouse model of chronic visceral hypersensitivity. Neurogastroenterol Motil 2016; 28:54-63. [PMID: 26468044 DOI: 10.1111/nmo.12696] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 09/02/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Chronic visceral pain is a defining feature of irritable bowel syndrome (IBS). IBS patients often show alterations in innate and adaptive immune function which may contribute to symptoms. Immune mediators are known to modulate the activity of viscero-sensory afferent nerves, but the focus has been on the innate immune system. Interleukin-2 (IL-2) is primarily associated with adaptive immune responses but its effects on colo-rectal afferent function in health or disease are unknown. METHODS Myeloperoxidase (MPO) activity determined the extent of inflammation in health, acute trinitrobenzene-sulfonic acid (TNBS) colitis, and in our post-TNBS colitis model of chronic visceral hypersensitivity (CVH). The functional effects of IL-2 on high-threshold colo-rectal afferents and the expression of IL-2R and NaV 1.7 mRNA in colo-rectal dorsal root ganglia (DRG) neurons were compared between healthy and CVH mice. KEY RESULTS MPO activity was increased during acute colitis, but subsided to levels comparable to health in CVH mice. IL-2 caused direct excitation of colo-rectal afferents that was blocked by tetrodotoxin. IL-2 did not affect afferent mechanosensitivity in health or CVH. However, an increased proportion of afferents responded directly to IL-2 in CVH mice compared with controls (73% vs 33%; p < 0.05), and the abundance of IL-2R and NaV 1.7 mRNA was increased 3.5- and 2-fold (p < 0.001 for both) in colo-rectal DRG neurons. CONCLUSIONS & INFERENCES IL-2, an immune mediator from the adaptive arm of the immune response, affects colo-rectal afferent function, indicating these effects are not restricted to innate immune mediators. Colo-rectal afferent sensitivity to IL-2 is increased long after healing from inflammation.
Collapse
Affiliation(s)
- M A Campaniello
- Centre for Nutritional and Gastrointestinal Diseases, Department of Medicine, University of Adelaide and South Australian Health Medical Health Research Institute, Adelaide, SA, Australia
| | - A M Harrington
- Centre for Nutritional and Gastrointestinal Diseases, Department of Medicine, University of Adelaide and South Australian Health Medical Health Research Institute, Adelaide, SA, Australia
| | - C M Martin
- Centre for Nutritional and Gastrointestinal Diseases, Department of Medicine, University of Adelaide and South Australian Health Medical Health Research Institute, Adelaide, SA, Australia
| | - L Ashley Blackshaw
- Neurogastroenterology Group, Blizard Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - S M Brierley
- Centre for Nutritional and Gastrointestinal Diseases, Department of Medicine, University of Adelaide and South Australian Health Medical Health Research Institute, Adelaide, SA, Australia
| | - P A Hughes
- Centre for Nutritional and Gastrointestinal Diseases, Department of Medicine, University of Adelaide and South Australian Health Medical Health Research Institute, Adelaide, SA, Australia
| |
Collapse
|
118
|
Jarrett ME, Cain KC, Barney PG, Burr RL, Naliboff BD, Shulman R, Zia J, Heitkemper MM. Balance of Autonomic Nervous System Predicts Who Benefits from a Self-management Intervention Program for Irritable Bowel Syndrome. J Neurogastroenterol Motil 2015; 22:102-11. [PMID: 26459461 PMCID: PMC4699727 DOI: 10.5056/jnm15067] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 07/09/2015] [Accepted: 08/04/2015] [Indexed: 12/22/2022] Open
Abstract
Background/Aims To determine if potential biomarkers can be used to identify subgroups of people with irritable bowel syndrome (IBS) who will benefit the most or the least from a comprehensive self-management (CSM) intervention. Methods In a two-armed randomized controlled trial a CSM (n = 46) was compared to a usual care (n = 46) group with follow-up at 3 and 6 months post randomization. Biomarkers obtained at baseline included heart rate variability, salivary cortisol, interleukin-10 produced by unstimulated peripheral blood mononuclear cells, and lactulose/mannitol ratio. Linear mixed models were used to test whether these biomarkers predicted improvements in the primary outcomes including daily abdominal pain, Gastrointestinal Symptom score and IBS-specific quality of life. Results The nurse-delivered 8-session CSM intervention is more effective than usual care in reducing abdominal pain, reducing Gastrointestinal Symptom score, and enhancing quality of life. Participants with lower nighttime high frequency heart rate variability (vagal modulation) and increased low frequency/high frequency ratio (sympathovagal balance) had less benefit from CSM on abdominal pain. Salivary cortisol, IL-10, and lactulose/mannitol ratio were not statistically significant in predicting CSM benefit. Baseline symptom severity interacts with treatment, namely the benefit of CSM is greater in those with higher baseline symptoms. Conclusions Cognitively-focused therapies may be less effective in reducing abdominal pain in IBS patients with higher sympathetic tone. Whether this a centrally-mediated patient characteristic or related to heightened arousal remains to be determined.
Collapse
Affiliation(s)
- Monica E Jarrett
- Department of Biobehavioral Nursing and Health Systems, University of Washington, Seattle, WA, USA
| | - Kevin C Cain
- Deptartment of Biostatistics and Office for Nursing Research, University of Washington, Seattle, WA, USA
| | - Pamela G Barney
- Department of Biobehavioral Nursing and Health Systems, University of Washington, Seattle, WA, USA
| | - Robert L Burr
- Department of Biobehavioral Nursing and Health Systems, University of Washington, Seattle, WA, USA
| | - Bruce D Naliboff
- Center for Neurovisceral Sciences & Women's Health, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Jasmine Zia
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Margaret M Heitkemper
- Department of Biobehavioral Nursing and Health Systems, University of Washington, Seattle, WA, USA
| |
Collapse
|
119
|
Ke J, Qi R, Liu C, Xu Q, Wang F, Zhang L, Lu G. Abnormal regional homogeneity in patients with irritable bowel syndrome: A resting-state functional MRI study. Neurogastroenterol Motil 2015; 27:1796-803. [PMID: 26403620 DOI: 10.1111/nmo.12692] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/26/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Task-related brain imaging research has implicated abnormal central processing of visceral sensation in irritable bowel syndrome (IBS). However, how brain function of IBS patients is altered during resting-state remains to be determined. We investigated spontaneous brain activity of patients with IBS using regional homogeneity (ReHo) analysis in resting-state functional magnetic resonance imaging (rs-fMRI). METHODS Thirty-one patients with diarrhea-predominant IBS and 32 age- and sex- matched healthy controls underwent clinical assessments and rs-fMRI scanning. ReHo maps were acquired by calculating the Kendall's coefficient of concordance and compared between the IBS group and the control group. The effects of psychological disturbance on group differences were assessed by including anxiety and depression levels as covariates in the statistical analyses. Multiple regression analyses were conducted to examine the relationship between ReHo values and disease duration, symptom severity, and pain intensity. KEY RESULTS Compared with controls, IBS patients showed increased ReHo in the postcentral gyrus and thalamus and decreased ReHo in the anterior cingulate cortex and prefrontal cortex. The inclusion of anxiety and depression as covariates did not alter ReHo differences between the two groups. Furthermore, significant correlations were found between clinical indices and ReHo values in some brain regions in the IBS group. CONCLUSIONS & INFERENCES IBS patients have abnormal local synchronization of spontaneous brain activity in regions involved in visceral afferent processing, emotional arousal, and cognitive modulation. Combining rs-fMRI and ReHo analysis seems to be a valuable approach to investigate the neural basis of IBS.
Collapse
Affiliation(s)
- J Ke
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - R Qi
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - C Liu
- Department of Gastroenterology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Q Xu
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - F Wang
- Department of Gastroenterology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - L Zhang
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - G Lu
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
120
|
Keely S, Walker MM, Marks E, Talley NJ. Immune dysregulation in the functional gastrointestinal disorders. Eur J Clin Invest 2015; 45:1350-9. [PMID: 26444549 DOI: 10.1111/eci.12548] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/03/2015] [Indexed: 12/14/2022]
Abstract
Gastrointestinal conditions may be broadly classified into two: organic and functional disease, with functional disorders accounting for the majority of patients with chronic gastrointestinal symptoms. Functional gastrointestinal disorders (FGIDs) present with no obvious pathology or well-accepted biochemical mechanism and, as such, treatment strategies are limited and focus on symptoms rather than cure. Irritable bowel syndrome and functional dyspepsia are the most widely recognised FGIDs, and there is a growing body of evidence to suggest an underlying inflammatory phenotype in subsets with these conditions. Here, we discuss the current knowledge of immune involvement in FGIDs and the commonalities between the different manifestations of FGIDs and propose a new hypothesis, potentially defining an underlying immunopathological basis of these conditions.
Collapse
Affiliation(s)
- Simon Keely
- School of Biomedical Sciences & Pharmacy, University of Newcastle & Vaccine and Asthma (VIVA) Program, Hunter Medical Research Institute, Callaghan, NSW, Australia
| | - Marjorie M Walker
- School of Medicine & Public Health, University of Newcastle, Callaghan, NSW, Australia
| | - Ellen Marks
- School of Biomedical Sciences & Pharmacy, University of Newcastle & Vaccine and Asthma (VIVA) Program, Hunter Medical Research Institute, Callaghan, NSW, Australia
| | - Nicholas J Talley
- School of Medicine & Public Health, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
121
|
Holzer P, Hassan AM, Jain P, Reichmann F, Farzi A. Neuroimmune pharmacological approaches. Curr Opin Pharmacol 2015; 25:13-22. [PMID: 26426677 PMCID: PMC4830400 DOI: 10.1016/j.coph.2015.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 02/07/2023]
Abstract
Intestinal inflammation is a major health problem which impairs the quality of life, impacts mental health and is exacerbated by stress and psychiatric disturbances which, in turn, can affect disease prognosis and response to treatment. Accumulating evidence indicates that the immune system is an important interface between intestinal inflammation and the enteric, sensory, central and autonomic nervous systems. In addition, the neuroimmune interactions originating from the gastrointestinal tract are orchestrated by the gut microbiota. This article reviews some major insights into this complex homeostatic network that have been achieved during the past two years and attempts to put these advances into perspective with novel opportunities of pharmacological intervention.
Collapse
Affiliation(s)
- Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, A-8010 Graz, Austria.
| | - Ahmed M Hassan
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, A-8010 Graz, Austria
| | - Piyush Jain
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, A-8010 Graz, Austria
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, A-8010 Graz, Austria
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, A-8010 Graz, Austria
| |
Collapse
|
122
|
Abstract
Visceral pain is diffusely localized, referred into other tissues, frequently not correlated with visceral traumata, preferentially accompanied by autonomic and somatomotor reflexes, and associated with strong negative affective feelings. It belongs together with the somatic pain sensations and non-painful body sensations to the interoception of the body. (1) Visceral pain is correlated with the excitation of spinal (thoracolumbar, sacral) visceral afferents and (with a few exceptions) not with the excitation of vagal afferents. Spinal visceral afferents are polymodal and activated by adequate mechanical and chemical stimuli. All groups of spinal visceral afferents can be sensitized (e.g., by inflammation). Silent mechanoinsensitive spinal visceral afferents are recruited by inflammation. (2) Spinal visceral afferent neurons project into the laminae I, II (outer part IIo) and V of the spinal dorsal horn over several segments, medio-lateral over the whole width of the dorsal horn and contralateral. Their activity is synaptically transmitted in laminae I, IIo and deeper laminae to viscero-somatic convergent neurons that receive additionally afferent synaptic (mostly nociceptive) input from the skin and from deep somatic tissues of the corresponding dermatomes, myotomes and sclerotomes. (3) The second-order neurons consist of excitatory and inhibitory interneurons (about 90 % of all dorsal horn neurons) and tract neurons activated monosynaptically in lamina I by visceral afferent neurons and di- or polysynaptically in deeper laminae. (4) The sensitization of viscero-somatic convergent neurons (central sensitization) is dependent on the sensitization of spinal visceral afferent neurons, local spinal excitatory and inhibitory interneurons and supraspinal endogenous control systems. The mechanisms of this central sensitization have been little explored. (5) Viscero-somatic tract neurons project through the contralateral ventrolateral tract and presumably other tracts to the lower and upper brain stem, the hypothalamus and via the thalamus to various cortical areas. (6) Visceral pain is presumably (together with other visceral sensations and nociceptive as well as non-nociceptive somatic body sensations) primarily represented in the posterior dorsal insular cortex (primary interoceptive cortex). This cortex receives in primates its spinal synaptic inputs mainly from lamina I tract neurons via the ventromedial posterior nucleus of the thalamus. (7) The transmission of activity from visceral afferents to second-order neurons in spinal cord is modulated in an excitatory and inhibitory way by endogenous anti- and pronociceptive control systems in the lower and upper brain stem. These control systems are under cortical control. (8) Visceral pain is referred to deep somatic tissues, to the skin and to other visceral organs. This referred pain consists of spontaneous pain and mechanical hyperalgesia. The mechanisms underlying referred pain and the accompanying tissue changes have been little explored.
Collapse
Affiliation(s)
- W Jänig
- Physiologisches Institut, Christian-Albrechts-Universität, Olshausenstr. 40, 24098, Kiel, Deutschland,
| |
Collapse
|
123
|
Yu X, Yu M, Liu Y, Yu S. TRP channel functions in the gastrointestinal tract. Semin Immunopathol 2015; 38:385-96. [PMID: 26459157 DOI: 10.1007/s00281-015-0528-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 09/07/2015] [Indexed: 12/14/2022]
Abstract
Transient receptor potential (TRP) channels are predominantly distributed in both somatic and visceral sensory nervous systems and play a crucial role in sensory transduction. As the largest visceral organ system, the gastrointestinal (GI) tract frequently accommodates external inputs, which stimulate sensory nerves to initiate and coordinate sensory and motor functions in order to digest and absorb nutrients. Meanwhile, the sensory nerves in the GI tract are also able to detect potential tissue damage by responding to noxious irritants. This nocifensive function is mediated through specific ion channels and receptors expressed in a subpopulation of spinal and vagal afferent nerve called nociceptor. In the last 18 years, our understanding of TRP channel expression and function in GI sensory nervous system has been continuously improved. In this review, we focus on the expressions and functions of TRPV1, TRPA1, and TRPM8 in primary extrinsic afferent nerves innervated in the esophagus, stomach, intestine, and colon and briefly discuss their potential roles in relevant GI disorders.
Collapse
Affiliation(s)
- Xiaoyun Yu
- Division of Gastroenterology & Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 945, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Mingran Yu
- Division of Gastroenterology & Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 945, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Yingzhe Liu
- Division of Gastroenterology & Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 945, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Shaoyong Yu
- Division of Gastroenterology & Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 945, 720 Rutland Ave, Baltimore, MD, 21205, USA.
| |
Collapse
|
124
|
Benson S, Rebernik L, Wegner A, Kleine-Borgmann J, Engler H, Schlamann M, Forsting M, Schedlowski M, Elsenbruch S. Neural circuitry mediating inflammation-induced central pain amplification in human experimental endotoxemia. Brain Behav Immun 2015; 48:222-31. [PMID: 25882910 DOI: 10.1016/j.bbi.2015.03.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND & AIMS To elucidate the brain mechanisms underlying inflammation-induced visceral hyperalgesia in humans, in this functional magnetic resonance imaging (fMRI) study we tested if intravenous administration of lipopolysaccharide (LPS) involves altered central processing of visceral pain stimuli. METHODS In this randomized, double-blind, placebo-controlled fMRI study, 26 healthy male subjects received either an intravenous injection of low-dose LPS (N=14, 0.4 ng/kg body weight) or placebo (N=12, control group). Plasma cytokines (TNF-α, IL-6), body temperature, plasma cortisol and mood were assessed at baseline and up to 6 h post-injection. At baseline and 2 h post-injection (test), rectal pain thresholds and painful rectal distension-induced blood oxygen level-dependent (BOLD) responses in brain regions-of-interest were assessed. To address specificity for visceral pain, BOLD responses to non-painful rectal distensions and painful somatic stimuli (i.e., punctuate mechanical stimulation) were also analyzed as control stimuli. RESULTS Compared to the control group, LPS-treated subjects demonstrated significant and transient increases in TNF-α, IL-6, body temperature and cortisol, along with impaired mood. In response to LPS, rectal pain thresholds decreased in trend, along with enhanced up-regulation of rectal pain-induced BOLD responses within the posterior insula, dorsolateral prefrontal (DLPFC), anterior midcingulate (aMCC) and somatosensory cortices (all FWE-corrected p<0.05). Within the LPS group, more pronounced cytokine responses correlated significantly with enhanced rectal pain-induced neural activation in DLPFC and aMCC. No significant LPS effects were observed on neural responses to non-painful rectal distensions or mechanical stimulation. CONCLUSIONS These findings support that peripheral inflammatory processes affect visceral pain thresholds and the central processing of sensory-discriminative aspects of visceral pain.
Collapse
Affiliation(s)
- Sven Benson
- Institute of Medical Psychology & Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Laura Rebernik
- Institute of Medical Psychology & Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany; Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Alexander Wegner
- Clinic for Trauma Surgery, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Julian Kleine-Borgmann
- Institute of Medical Psychology & Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Harald Engler
- Institute of Medical Psychology & Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Marc Schlamann
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Michael Forsting
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology & Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Sigrid Elsenbruch
- Institute of Medical Psychology & Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany.
| |
Collapse
|
125
|
Humenick A, Chen BN, Wiklendt L, Spencer NJ, Zagorodnyuk VP, Dinning PG, Costa M, Brookes SJH. Activation of intestinal spinal afferent endings by changes in intra-mesenteric arterial pressure. J Physiol 2015; 593:3693-709. [PMID: 26010893 DOI: 10.1113/jp270378] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/18/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS A major class of mechano-nociceptors to the intestine have mechanotransduction sites on extramural and intramural arteries and arterioles ('vascular afferents'). These sensory neurons can be activated by compression or axial stretch of vessels. Using isolated preparations we showed that increasing intra-arterial pressure, within the physiological range, activated mechano-nociceptors on vessels in intact mesenteric arcades, but not in isolated arteries. This suggests that distortion of the branching vascular tree is the mechanical adequate stimulus for these sensory neurons, rather than simple distension. The same rises in pressure also activated intestinal peristalsis in a partially capsaicin-sensitive manner indicating that pressure-sensitive vascular afferents influence enteric circuits. The results identify the mechanical adequate stimulus for a major class of mechano-nociceptors with endings on blood vessels supplying the gut wall; these afferents have similar endings to ones supplying other viscera, striated muscle and dural vessels. ABSTRACT Spinal sensory neurons innervate many large blood vessels throughout the body. Their activation causes the hallmarks of neurogenic inflammation: vasodilatation through the release of the neuropeptide calcitonin gene-related peptide and plasma extravasation via tachykinins. The same vasodilator afferent neurons show mechanical sensitivity, responding to crushing, compression or axial stretch of blood vessels - responses which activate pain pathways and which can be modified by cell damage and inflammation. In the present study, we tested whether spinal afferent axons ending on branching mesenteric arteries ('vascular afferents') are sensitive to increased intravascular pressure. From a holding pressure of 5 mmHg, distension to 20, 40, 60 or 80 mmHg caused graded, slowly adapting increases in firing of vascular afferents. Many of the same afferent units showed responses to axial stretch, which summed with responses evoked by raised pressure. Many vascular afferents were also sensitive to raised temperature, capsaicin and/or local compression with von Frey hairs. However, responses to raised pressure in single, isolated vessels were negligible, suggesting that the adequate stimulus is distortion of the arterial arcade rather than distension per se. Increasing arterial pressure often triggered peristaltic contractions in the neighbouring segment of intestine, an effect that was mimicked by acute exposure to capsaicin (1 μm) and which was reduced after desensitisation to capsaicin. These results indicate that sensory fibres with perivascular endings are sensitive to pressure-induced distortion of branched arteries, in addition to compression and axial stretch, and that they contribute functional inputs to enteric motor circuits.
Collapse
Affiliation(s)
- A Humenick
- Discipline of Human Physiology and Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, South Australia
| | - B N Chen
- Discipline of Human Physiology and Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, South Australia
| | - L Wiklendt
- Discipline of Human Physiology and Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, South Australia
| | - N J Spencer
- Discipline of Human Physiology and Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, South Australia
| | - V P Zagorodnyuk
- Discipline of Human Physiology and Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, South Australia
| | - P G Dinning
- Discipline of Human Physiology and Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, South Australia
| | - M Costa
- Discipline of Human Physiology and Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, South Australia
| | - S J H Brookes
- Discipline of Human Physiology and Centre for Neuroscience, School of Medicine, Flinders University, Adelaide, South Australia
| |
Collapse
|
126
|
Hungin APS, Becher A, Cayley B, Heidelbaugh JJ, Muris JWM, Rubin G, Seifert B, Russell A, De Wit NJ. Irritable bowel syndrome: an integrated explanatory model for clinical practice. Neurogastroenterol Motil 2015; 27:750-63. [PMID: 25703486 DOI: 10.1111/nmo.12524] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 01/13/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Although irritable bowel syndrome (IBS) is a symptom-based diagnosis, clinicians' management of and communication about the disorder is often hampered by an unclear conceptual understanding of the nature of the problem. We aimed to elucidate an integrated explanatory model (EM) for IBS from the existing literature for pragmatic use in the clinical setting. METHODS Systematic and exploratory literature searches were performed in PubMed to identify publications on IBS and EMs. KEY RESULTS The searches did not identify a single, integrated EM for IBS. However, three main hypotheses were elucidated that could provide components with which to develop an IBS EM: (i) altered peripheral regulation of gut function (including sensory and secretory mechanisms); (ii) altered brain-gut signaling (including visceral hypersensitivity); and (iii) psychological distress. Genetic polymorphisms and epigenetic changes may, to some degree, underlie the etiology and pathophysiology of IBS and could increase the susceptibility to developing the disorder. The three model components also fit into one integrated explanation for abdominal symptoms and changes in stool habit. Additionally, IBS may share a common pathophysiological mechanism with other associated functional syndromes. CONCLUSIONS & INFERENCES It was possible to elucidate an integrated, three-component EM as a basis for clinicians to conceptualize the nature of IBS, with the potential to contribute to better diagnosis and management, and dialog with sufferers.
Collapse
Affiliation(s)
- A P S Hungin
- School of Medicine, Pharmacy and Health, Durham University, Stockton-on-Tees, UK
| | - A Becher
- School of Medicine, Pharmacy and Health, Durham University, Stockton-on-Tees, UK.,Research and Evaluation Unit, Oxford PharmaGenesis Ltd, Oxford, UK
| | - B Cayley
- Department of Family Medicine, University of Wisconsin, Madison, WI, USA
| | - J J Heidelbaugh
- Departments of Family Medicine and Urology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | - J W M Muris
- Department of Family Medicine, Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - G Rubin
- School of Medicine, Pharmacy and Health, Durham University, Stockton-on-Tees, UK
| | - B Seifert
- Institute of General Practice, Charles University, Praha, Czech Republic
| | - A Russell
- Department of Anthropology, Durham University, Durham, UK
| | - N J De Wit
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
127
|
Ostertag D, Buhner S, Michel K, Pehl C, Kurjak M, Götzberger M, Schulte-Frohlinde E, Frieling T, Enck P, Phillip J, Schemann M. Reduced Responses of Submucous Neurons from Irritable Bowel Syndrome Patients to a Cocktail Containing Histamine, Serotonin, TNFα, and Tryptase (IBS-Cocktail). Front Neurosci 2015; 9:465. [PMID: 26733780 PMCID: PMC4679876 DOI: 10.3389/fnins.2015.00465] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/23/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND AIMS Malfunctions of enteric neurons are believed to play an important role in the pathophysiology of irritable bowel syndrome (IBS). Our aim was to investigate whether neuronal activity in biopsies from IBS patients is altered in comparison to healthy controls (HC). METHODS Activity of human submucous neurons in response to electrical nerve stimulation and local application of nicotine or a mixture of histamine, serotonin, tryptase, and TNF-α (IBS-cocktail) was recorded in biopsies from 17 HC and 35 IBS patients with the calcium-sensitive-dye Fluo-4 AM. The concentrations of the mediators resembeled those found in biopsy supernatants or blood. Neuronal activity in guinea-pig submucous neurons was studied with the voltage-sensitive-dye di-8-ANEPPS. RESULTS Activity in submucous ganglia in response to nicotine or electrical nerve stimulation was not different between HC and IBS patients (P = 0.097 or P = 0.448). However, the neuronal response after application of the IBS-cocktail was significantly decreased (P = 0.039) independent of whether diarrhea (n = 12), constipation (n = 5) or bloating (n = 5) was the predominant symptom. In agreement with this we found that responses of submucous ganglia conditioned by overnight incubation with IBS mucosal biopsy supernatant to spritz application of this supernatant was significantly reduced (P = 0.019) when compared to incubation with HC supernatant. CONCLUSION We demonstrated for the first time reduced neuronal responses in mucosal IBS biopsies to an IBS mediator cocktail. While excitability to classical stimuli of enteric neurons was comparable to HC, the activation by the IBS-cocktail was decreased. This was very likely due to desensitization to mediators constantly released by mucosal and immune cells in the gut wall of IBS patients.
Collapse
Affiliation(s)
| | - Sabine Buhner
- Human Biology, Technische Universität MünchenFreising, Germany
| | - Klaus Michel
- Human Biology, Technische Universität MünchenFreising, Germany
| | | | | | | | | | | | - Paul Enck
- Academic Hospital TübingenTübingen, Germany
| | | | - Michael Schemann
- Human Biology, Technische Universität MünchenFreising, Germany
- *Correspondence: Michael Schemann
| |
Collapse
|
128
|
Veldhuis NA, Poole DP, Grace M, McIntyre P, Bunnett NW. The G protein-coupled receptor-transient receptor potential channel axis: molecular insights for targeting disorders of sensation and inflammation. Pharmacol Rev 2015; 67:36-73. [PMID: 25361914 DOI: 10.1124/pr.114.009555] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Sensory nerves are equipped with receptors and ion channels that allow them to detect and respond to diverse chemical, mechanical, and thermal stimuli. These sensory proteins include G protein-coupled receptors (GPCRs) and transient receptor potential (TRP) ion channels. A subclass of peptidergic sensory nerves express GPCRs and TRP channels that detect noxious, irritant, and inflammatory stimuli. Activation of these nerves triggers protective mechanisms that lead to withdrawal from danger (pain), removal of irritants (itch, cough), and resolution of infection (neurogenic inflammation). The GPCR-TRP axis is central to these mechanisms. Signals that emanate from the GPCR superfamily converge on the small TRP family, leading to channel sensitization and activation, which amplify pain, itch, cough, and neurogenic inflammation. Herein we discuss how GPCRs and TRP channels function independently and synergistically to excite sensory nerves that mediate noxious and irritant responses and inflammation in the skin and the gastrointestinal and respiratory systems. We discuss the signaling mechanisms that underlie the GPCR-TRP axis and evaluate how new information about the structure of GPCRs and TRP channels provides insights into their functional interactions. We propose that a deeper understanding of the GPCR-TRP axis may facilitate the development of more selective and effective therapies to treat dysregulated processes that underlie chronic pain, itch, cough, and inflammation.
Collapse
Affiliation(s)
- Nicholas A Veldhuis
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Daniel P Poole
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Megan Grace
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Peter McIntyre
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| | - Nigel W Bunnett
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia (N.A.V., D.P.P., N.W.B); Departments of Genetics (N.A.V.), Anatomy and Neuroscience (D.P.P.), and Pharmacology (N.W.B.), The University of Melbourne, Melbourne, Victoria, Australia; School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia (M.G., P.M.); and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia (N.W.B.)
| |
Collapse
|
129
|
Abstract
The aetiology and pathology of IBS, a functional bowel disorder thought to lack an organic cause, is largely unknown. However, studies suggest that various features, such as altered composition of the gut microbiota, together with increased intestinal permeability, a changed balance in the enteroendocrine system and a dysregulated immune system in the gut, most likely have an important role in IBS. Exactly how these entities act together and give rise to symptoms is still unknown, but an altered gut microbiota composition could lead to dysregulation of the intestinal barrier as well as the enteroendocrine and the immune systems, which (through interactions with the nervous system) might generate symptoms. This Review highlights the crosstalk between the gut microbiota, the enteroendocrine system, the immune system and the role of intestinal permeability in patients with IBS.
Collapse
|
130
|
Acupuncture for visceral pain: neural substrates and potential mechanisms. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:609594. [PMID: 25614752 PMCID: PMC4295157 DOI: 10.1155/2014/609594] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 12/13/2014] [Accepted: 12/13/2014] [Indexed: 12/17/2022]
Abstract
Visceral pain is the most common form of pain caused by varied diseases and a major reason for patients to seek medical consultation. Despite much advances, the pathophysiological mechanism is still poorly understood comparing with its somatic counterpart and, as a result, the therapeutic efficacy is usually unsatisfactory. Acupuncture has long been used for the management of numerous disorders in particular pain and visceral pain, characterized by the high therapeutic benefits and low adverse effects. Previous findings suggest that acupuncture depresses pain via activation of a number of neurotransmitters or modulators including opioid peptides, serotonin, norepinephrine, and adenosine centrally and peripherally. It endows us, by advancing the understanding of the role of ion channels and gut microbiota in pain process, with novel perspectives to probe the mechanisms underlying acupuncture analgesia. In this review, after describing the visceral innervation and the relevant afferent pathways, in particular the ion channels in visceral nociception, we propose three principal mechanisms responsible for acupuncture induced benefits on visceral pain. Finally, potential topics are highlighted regarding the future studies in this field.
Collapse
|
131
|
Blackshaw LA. Transient receptor potential cation channels in visceral sensory pathways. Br J Pharmacol 2014; 171:2528-36. [PMID: 24641218 DOI: 10.1111/bph.12641] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 01/09/2014] [Accepted: 01/20/2014] [Indexed: 01/03/2023] Open
Abstract
The extensive literature on this subject is in direct contrast to the limited range of clinical uses for ligands of the transient receptor potential cation channels (TRPs) in diseases of the viscera. TRPV1 is the most spectacular example of this imbalance, as it is in other systems, but it is nonetheless the only TRP target that is currently targeted clinically in bladder sensory dysfunction. It is not clear why this discrepancy exists, but a likely answer is in the promiscuity of TRPs as sensors and transducers for environmental mechanical and chemical stimuli. This review first describes the different sensory pathways from the viscera, and on which nociceptive and non-nociceptive neurones within these pathways TRPs are expressed. They not only fulfil roles as both mechano- and chemo-sensors on visceral afferents, but also form an effector mechanism for cell activation after activation of GPCR and cytokine receptors. Their role may be markedly changed in diseased states, including chronic pain and inflammation. Pain presents the most obvious potential for further development of therapeutic interventions targeted at TRPs, but forms of inflammation are emerging as likely to benefit also. However, despite much basic research, we are still at the beginning of exploring such potential in visceral sensory pathways.
Collapse
Affiliation(s)
- L Ashley Blackshaw
- Wingate Institute for Neurogastroenterology, Centre for Digestive Diseases, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
132
|
La JH, Gebhart GF. Condition-specific role of colonic inflammatory molecules in persistent functional colorectal hypersensitivity in the mouse. Neurogastroenterol Motil 2014; 26:1730-42. [PMID: 25307695 PMCID: PMC4245395 DOI: 10.1111/nmo.12455] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/16/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND A low-level inflammation has been hypothesized to mediate visceral hypersensitivity in functional bowel disorders that persist after or even in the absence of gut inflammation. We aimed to test the efficacy of a steroidal anti-inflammatory treatment, and identify local inflammatory molecules mediating post- and non-inflammatory colorectal hypersensitivity using two mouse models. METHODS Visceromotor responses to colorectal distension were quantified as a measure of colorectal sensitivity. On day 1, mice received intracolonic saline (control), trinitrobenzenesulfonic acid (postinflammatory on day 15), or acidified hypertonic saline (non-inflammatory). Colorectal sensitivity before (day 10) and after (day 15) 4-day dexamethasone (Dex) treatment was compared, and colonic gene expression of inflammatory molecules was quantified. KEY RESULTS Dexamethasone effectively inhibited gene expression of inflammatory molecules such as interleukin (IL)-1β and mast cell protease-1 in the colon, but did not attenuate colorectal hypersensitivity in either model. Gene expression of inflammatory molecules in the colon did not differ between control and the non-inflammatory model, but the postinflammatory model showed increased IL-10 and tight junction protein 2, and decreased IL-6, transforming growth factor (TGF)-β, a precursor of β-endorphin, occludin, and mucin 2. While no common molecule explained colorectal hypersensitivity in these models, hypersensitivity was positively correlated with TGF-β2 mRNA in control, and with IL-1β, inhibin βA, and prostaglandin E2 synthase in the Dex-treated postinflammatory model. In the non-inflammatory model, cyclooxygenase-2 mRNA was negatively correlated with colorectal sensitivity. CONCLUSIONS & INFERENCES These results suggest that persistent functional colorectal hypersensitivity is mediated by condition-specific mediators whose gene expression in the colon is not inevitably sensitive to steroidal anti-inflammatory treatment.
Collapse
Affiliation(s)
- J.-H. La
- Center for Pain Research; Department of Anesthesiology; University of Pittsburgh School of Medicine; Pittsburgh PA USA
| | - G. F. Gebhart
- Center for Pain Research; Department of Anesthesiology; University of Pittsburgh School of Medicine; Pittsburgh PA USA
| |
Collapse
|
133
|
Thompson GL, Canals M, Poole DP. Biological redundancy of endogenous GPCR ligands in the gut and the potential for endogenous functional selectivity. Front Pharmacol 2014; 5:262. [PMID: 25506328 PMCID: PMC4246669 DOI: 10.3389/fphar.2014.00262] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 11/12/2014] [Indexed: 01/27/2023] Open
Abstract
This review focuses on the existence and function of multiple endogenous agonists of the somatostatin and opioid receptors with an emphasis on their expression in the gastrointestinal tract. These agonists generally arise from the proteolytic cleavage of prepropeptides during peptide maturation or from degradation of peptides by extracellular or intracellular endopeptidases. In other examples, endogenous peptide agonists for the same G protein-coupled receptors can be products of distinct genes but contain high sequence homology. This apparent biological redundancy has recently been challenged by the realization that different ligands may engender distinct receptor conformations linked to different intracellular signaling profiles and, as such the existence of distinct ligands may underlie mechanisms to finely tune physiological responses. We propose that further characterization of signaling pathways activated by these endogenous ligands will provide invaluable insight into the mechanisms governing biased agonism. Moreover, these ligands may prove useful in the design of novel therapeutic tools to target distinct signaling pathways, thereby favoring desirable effects and limiting detrimental on-target effects. Finally we will discuss the limitations of this area of research and we will highlight the difficulties that need to be addressed when examining endogenous bias in tissues and in animals.
Collapse
Affiliation(s)
- Georgina L Thompson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences Parkville, VIC, Australia
| | - Meritxell Canals
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences Parkville, VIC, Australia
| | - Daniel P Poole
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences Parkville, VIC, Australia ; Department of Anatomy and Neuroscience, The University of Melbourne Parkville, VIC, Australia
| |
Collapse
|
134
|
Hughes PA, Moretta M, Lim A, Grasby DJ, Bird D, Brierley SM, Liebregts T, Adam B, Blackshaw LA, Holtmann G, Bampton P, Hoffmann P, Andrews JM, Zola H, Krumbiegel D. Immune derived opioidergic inhibition of viscerosensory afferents is decreased in Irritable Bowel Syndrome patients. Brain Behav Immun 2014; 42:191-203. [PMID: 25063707 DOI: 10.1016/j.bbi.2014.07.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 06/30/2014] [Accepted: 07/01/2014] [Indexed: 12/29/2022] Open
Abstract
Alterations in the neuro-immune axis contribute toward viscerosensory nerve sensitivity and symptoms in Irritable Bowel Syndrome (IBS). Inhibitory factors secreted from immune cells inhibit colo-rectal afferents in health, and loss of this inhibition may lead to hypersensitivity and symptoms. We aimed to determine the immune cell type(s) responsible for opioid secretion in humans and whether this is altered in patients with IBS. The β-endorphin content of specific immune cell lineages in peripheral blood and colonic mucosal biopsies were compared between healthy subjects (HS) and IBS patients. Peripheral blood mononuclear cell (PBMC) supernatants from HS and IBS patients were applied to colo-rectal sensory afferent endings in mice with post-inflammatory chronic visceral hypersensitivity (CVH). β-Endorphin was identified predominantly in monocyte/macrophages relative to T or B cells in human PBMC and colonic lamina propria. Monocyte derived β-endorphin levels and colonic macrophage numbers were lower in IBS patients than healthy subjects. PBMC supernatants from healthy subjects had greater inhibitory effects on colo-rectal afferent mechanosensitivity than those from IBS patients. The inhibitory effects of PBMC supernatants were more prominent in CVH mice compared to healthy mice due to an increase in μ-opioid receptor expression in dorsal root ganglia neurons in CVH mice. Monocyte/macrophages are the predominant immune cell type responsible for β-endorphin secretion in humans. IBS patients have lower monocyte derived β-endorphin levels than healthy subjects, causing less inhibition of colonic afferent endings. Consequently, altered immune function contributes toward visceral hypersensitivity in IBS.
Collapse
Affiliation(s)
- Patrick A Hughes
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; Leukocyte Biology Laboratory, Women's and Children's Health Research Institute, North Adelaide, SA 5006, Australia.
| | - Melissa Moretta
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Amanda Lim
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Dallas J Grasby
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Daniel Bird
- Leukocyte Biology Laboratory, Women's and Children's Health Research Institute, North Adelaide, SA 5006, Australia
| | - Stuart M Brierley
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Tobias Liebregts
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Birgit Adam
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - L Ashley Blackshaw
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Gerald Holtmann
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Peter Bampton
- Department of Gastroenterology, Flinders Medical Centre, Flinders University, Bedford Park, SA 5042, Australia
| | - Peter Hoffmann
- Adelaide Proteomics Centre, School of Molecular Biomedical Science, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Jane M Andrews
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Heddy Zola
- Leukocyte Biology Laboratory, Women's and Children's Health Research Institute, North Adelaide, SA 5006, Australia; Discipline of Paediatrics, Faculty of Health Sciences, University of Adelaide, SA 5005, Australia
| | - Doreen Krumbiegel
- Leukocyte Biology Laboratory, Women's and Children's Health Research Institute, North Adelaide, SA 5006, Australia; Discipline of Paediatrics, Faculty of Health Sciences, University of Adelaide, SA 5005, Australia; SA Pathology, Adelaide, SA 5000, Australia
| |
Collapse
|
135
|
Associations among gut permeability, inflammatory markers, and symptoms in patients with irritable bowel syndrome. J Gastroenterol 2014; 49:1467-76. [PMID: 24435814 PMCID: PMC4102674 DOI: 10.1007/s00535-013-0919-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 11/21/2013] [Indexed: 02/04/2023]
Abstract
BACKGROUND Alterations in gastrointestinal (GI) permeability and immune measures are present in some patients with irritable bowel syndrome (IBS) but the relationship to symptoms is poorly defined. In adults with IBS, we compared permeability, unstimulated peripheral blood monocyte (PBMC) interleukin-10 (IL-10) levels, IBS life interference, and GI and psychological distress symptoms. METHODS In 88 women and 18 men with IBS, GI permeability was quantitated as percent recovery of urinary sucrose and the lactulose/mannitol (L/M) ratio. IL-10 was measured in supernatants from 72-h incubated, unstimulated PBMCs. Participants completed a 4-week daily diary recording IBS life interference on daily activities and work, IBS symptoms, and psychological distress symptoms. They also completed the Brief Symptom Inventory. RESULTS The L/M ratio but not percent sucrose recovery was significantly correlated with IBS interference with activities and work and retrospectively measured anxiety and depression. Unstimulated PBMC production of IL-10 correlated significantly with IBS interference with daily work, IBS symptom score, and abdominal pain. We identified a subgroup of IBS subjects with higher IL-10 and/or higher L/M ratio who had substantially higher IBS interference and IBS symptom scores. CONCLUSIONS Our findings suggest a distinct subgroup of IBS patients with alterations in gut barrier function. This subgroup is characterized by increased GI permeability and/or increased PBMC production of IL-10. These physiologic alterations reflect more severe IBS as measured by interference of IBS with daily activities and daily IBS symptoms.
Collapse
|
136
|
Abstract
Painful sensation is a hallmark of microbe-induced inflammation. This inflammatory pain is downregulated a few days after infection by opioids locally released by effector T lymphocytes generated in response to microbe-derived antigens. This review focuses on the endogenous regulation of inflammatory pain associated with adaptive T-cell response and puts in perspective the clinical consequences of the opioid-mediated analgesic activity of colitogenic T lymphocytes in inflammatory bowel disease.
Collapse
|
137
|
Abstract
The gastrointestinal tract is innervated by several distinct populations of neurons, whose cell bodies either reside within (intrinsic) or outside (extrinsic) the gastrointestinal wall. Normally, most individuals are unaware of the continuous, complicated functions of these neurons. However, for patients with gastrointestinal disorders, such as IBD and IBS, altered gastrointestinal motility, discomfort and pain are common, debilitating symptoms. Although bouts of intestinal inflammation underlie the symptoms associated with IBD, increasing preclinical and clinical evidence indicates that infection and inflammation are also key risk factors for the development of other gastrointestinal disorders. Notably, a strong correlation exists between prior exposure to gut infection and symptom occurrence in IBS. This Review discusses the evidence for neuroplasticity (structural, synaptic or intrinsic changes that alter neuronal function) affecting gastrointestinal function. Such changes are evident during inflammation and, in many cases, long after healing of the damaged tissues, when the nervous system fails to reset back to normal. Neuroplasticity within distinct populations of neurons has a fundamental role in the aberrant motility, secretion and sensation associated with common clinical gastrointestinal disorders. To find appropriate therapeutic treatments for these disorders, the extent and time course of neuroplasticity must be fully appreciated.
Collapse
|
138
|
Lin YM, Li F, Shi XZ. Mechanical stress is a pro-inflammatory stimulus in the gut: in vitro, in vivo and ex vivo evidence. PLoS One 2014; 9:e106242. [PMID: 25180799 PMCID: PMC4152012 DOI: 10.1371/journal.pone.0106242] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 07/28/2014] [Indexed: 12/14/2022] Open
Abstract
Aims Inflammatory infiltrates and pro-inflammatory mediators are found increased in obstructive and functional bowel disorders, in which lumen distention is present. However, what caused the low level inflammation is not well known. We tested the hypothesis that lumen distention- associated mechanical stress may induce expression of specific inflammatory mediators in gut smooth muscle. Methods Static mechanical stretch (18% elongation) was applied in vitro in primary culture of rat colonic circular smooth muscle cells (RCCSMCs) with a Flexercell FX-4000 Tension Plus System. Mechanical distention in vivo was induced in rats with an obstruction band placed in the distal colon. Results In the primary culture of RCCSMCs, we found that static stretch significantly induced mRNA expression of iNOS, IL-6, and MCP-1 in 3 hours by 6.0(±1.4), 2.5(±0.5), and 2.2(±0.5) fold (n = 6∼8, p<0.05), respectively. However, gene expression of TNF-α, IL-1β, and IL-8 was not significantly affected by mechanical stretch. In the in vivo model of colon obstruction, we found that gene expression of iNOS, IL-6, and MCP-1 is also significantly increased in a time-dependent manner in the mechanically distended proximal segment, but not in the sham controls or distal segments. The conditioned medium from the muscle strips of the stretched proximal segment, but not the distal segment or control, significantly induced translocation and phosphorylation of NF-κB p65. This treatment further increased mRNA expression of inflammatory mediators in the naïve cells. However, treatment of the conditioned medium from the proximal segment with neutralizing antibody against rat IL-6 significantly attenuated the activation of NF-κB and gene expression of inflammatory mediators. Conclusions Our studies demonstrate that mechanical stress induces gene expression of inflammatory mediators i.e. iNOS, IL-6, and MCP-1 in colonic SMC. Further ex vivo study showed that mechanical stress functions as a pro-inflammatory stimulus in the gut.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/pharmacology
- Cells, Cultured
- Chemokines/genetics
- Chemokines/metabolism
- Colon/pathology
- Culture Media, Conditioned/pharmacology
- Gastrointestinal Tract/drug effects
- Gastrointestinal Tract/pathology
- Inflammation/pathology
- Inflammation Mediators/metabolism
- Intestinal Obstruction/pathology
- Male
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- NF-kappa B/metabolism
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type II/genetics
- Nitric Oxide Synthase Type II/metabolism
- Phosphorylation/drug effects
- Protein Transport/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Stress, Mechanical
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- You-Min Lin
- Division of Gastroenterology, Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Feng Li
- Division of Gastroenterology, Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Xuan-Zheng Shi
- Division of Gastroenterology, Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
139
|
Ford AC, Moayyedi P, Lacy BE, Lembo AJ, Saito YA, Schiller LR, Soffer EE, Spiegel BMR, Quigley EMM. American College of Gastroenterology monograph on the management of irritable bowel syndrome and chronic idiopathic constipation. Am J Gastroenterol 2014; 109 Suppl 1:S2-26; quiz S27. [PMID: 25091148 DOI: 10.1038/ajg.2014.187] [Citation(s) in RCA: 391] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Alexander C Ford
- 1] Leeds Gastroenterology Institute, St James's University Hospital, Leeds, UK [2] First author on the monograph, but is not a member of the Task Force
| | - Paul Moayyedi
- 1] Farncombe Family Digestive Health Research Institute, Division of Gastroenterology, McMaster University, Hamilton, Ontario, Canada [2] Conducted systematic reviews with the support of A.C. Ford, and carried out the technical analyses of the data independent of the Task Force
| | - Brian E Lacy
- Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Anthony J Lembo
- Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | - Lawrence R Schiller
- Baylor University Medical Center, Digestive Health Associates of Texas, Dallas, Texas, USA
| | - Edy E Soffer
- Division of Gastroenterology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Brennan M R Spiegel
- UCLA School of Medicine, UCLA/VA Center for Outcomes Research and Education (CORE), Los Angeles, California, USA
| | - Eamonn M M Quigley
- Division of Gastroenterology and Hepatology, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Texas, USA
| | | |
Collapse
|
140
|
Buhner S, Braak B, Li Q, Kugler EM, Klooker T, Wouters M, Donovan J, Vignali S, Mazzuoli-Weber G, Grundy D, Boeckxstaens G, Schemann M. Neuronal activation by mucosal biopsy supernatants from irritable bowel syndrome patients is linked to visceral sensitivity. Exp Physiol 2014; 99:1299-311. [DOI: 10.1113/expphysiol.2014.080036] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Sabine Buhner
- Human Biology; Technische Universität München; Freising Germany
| | - Breg Braak
- Department of Gastroenterology and Hepatology; Academic Medical Center; Amsterdam The Netherlands
| | - Qin Li
- Human Biology; Technische Universität München; Freising Germany
- Department of Physiology; Shangdong University; Shangdong China
| | | | - Tamira Klooker
- Department of Gastroenterology and Hepatology; Academic Medical Center; Amsterdam The Netherlands
| | - Mira Wouters
- Translational Research Center for Gastrointestinal Disorders; University Hospital Gasthuisberg; Catholic University of Leuven; Leuven Belgium
| | - Jemma Donovan
- Department of Biomedical Science; University of Sheffield; Sheffield UK
| | - Sheila Vignali
- Human Biology; Technische Universität München; Freising Germany
| | | | - David Grundy
- Department of Biomedical Science; University of Sheffield; Sheffield UK
| | - Guy Boeckxstaens
- Department of Gastroenterology and Hepatology; Academic Medical Center; Amsterdam The Netherlands
- Translational Research Center for Gastrointestinal Disorders; University Hospital Gasthuisberg; Catholic University of Leuven; Leuven Belgium
| | | |
Collapse
|
141
|
Bashashati M, Rezaei N, Shafieyoun A, McKernan DP, Chang L, Öhman L, Quigley EM, Schmulson M, Sharkey KA, Simrén M. Cytokine imbalance in irritable bowel syndrome: a systematic review and meta-analysis. Neurogastroenterol Motil 2014; 26:1036-48. [PMID: 24796536 DOI: 10.1111/nmo.12358] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 04/06/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder of unknown etiology; although infection and inflammation have recently been considered as important etiologic agents. A recent meta-analysis showed correlations between cytokine [interleukin-10 (IL-10) and tumor necrosis factor (TNF)] gene polymorphisms and IBS; however, it is still unknown whether patients with IBS have different cytokine profiles compared to healthy population. METHODS To determine the relationships between serum/plasma levels or mucosal expression of IL-10/TNF-α and IBS, we conducted a systematic review and meta-analysis based on case-control studies retrieved from PubMed and EMBASE search through August 2013. Standardized mean difference (SMD) was generated by using the inverse variance method. Heterogeneity was assessed based on I(2) values. KEY RESULTS Serum/plasma levels of TNF-α tended to be higher in IBS vs controls (p = 0.09); this reached significance in IBS subtypes vs controls and in female patients with IBS. However, serum/plasma levels of IL-10 were not significantly different in IBS patients vs controls. Further analysis of serum/plasma IL-10 levels in IBS subtypes did not show any difference; however, analysis based on gender showed a significantly lower serum/plasma IL-10 levels in male patients with IBS vs male controls (p = 0.02). Colonic IL-10 mRNA had a significantly lower expression in IBS vs control (p = 0.001). CONCLUSIONS & INFERENCES There is an imbalance of proinflammatory TNF-α, and anti-inflammatory IL-10, cytokines in IBS. Stratifying IBS patients based on cytokine profile may represent an opportunity for personalized treatment of this condition.
Collapse
Affiliation(s)
- M Bashashati
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Hughes PA, Castro J, Harrington AM, Isaacs N, Moretta M, Hicks GA, Urso DM, Brierley SM. Increased κ-opioid receptor expression and function during chronic visceral hypersensitivity. Gut 2014; 63:1199-200. [PMID: 24285775 DOI: 10.1136/gutjnl-2013-306240] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Patrick A Hughes
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, South Australia, Australia Department of Gastroenterology and Hepatology, Hanson Institute, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Joel Castro
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, South Australia, Australia Department of Gastroenterology and Hepatology, Hanson Institute, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Andrea M Harrington
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, South Australia, Australia Department of Gastroenterology and Hepatology, Hanson Institute, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Nicole Isaacs
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Melissa Moretta
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | | | - David M Urso
- Tioga Pharmaceuticals Inc., San Diego, California, USA
| | - Stuart M Brierley
- Nerve-Gut Research Laboratory, Discipline of Medicine, Faculty of Health Sciences, The University of Adelaide, Adelaide, South Australia, Australia Department of Gastroenterology and Hepatology, Hanson Institute, Royal Adelaide Hospital, Adelaide, South Australia, Australia Discipline of Physiology, Faculty of Health Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
143
|
Nilius B, Szallasi A. Transient receptor potential channels as drug targets: from the science of basic research to the art of medicine. Pharmacol Rev 2014; 66:676-814. [PMID: 24951385 DOI: 10.1124/pr.113.008268] [Citation(s) in RCA: 377] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The large Trp gene family encodes transient receptor potential (TRP) proteins that form novel cation-selective ion channels. In mammals, 28 Trp channel genes have been identified. TRP proteins exhibit diverse permeation and gating properties and are involved in a plethora of physiologic functions with a strong impact on cellular sensing and signaling pathways. Indeed, mutations in human genes encoding TRP channels, the so-called "TRP channelopathies," are responsible for a number of hereditary diseases that affect the musculoskeletal, cardiovascular, genitourinary, and nervous systems. This review gives an overview of the functional properties of mammalian TRP channels, describes their roles in acquired and hereditary diseases, and discusses their potential as drug targets for therapeutic intervention.
Collapse
Affiliation(s)
- Bernd Nilius
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| | - Arpad Szallasi
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| |
Collapse
|
144
|
Reed DE, Blackshaw LA. Inhibition of visceral nociceptors. Front Pharmacol 2014; 5:72. [PMID: 24782775 PMCID: PMC3995067 DOI: 10.3389/fphar.2014.00072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 03/27/2014] [Indexed: 12/12/2022] Open
Affiliation(s)
- David E Reed
- Neurogastroenterolohy Group, Centre for Digestive Diseases, Blizard Institute, Queen Mary University of London London, UK
| | - L Ashley Blackshaw
- Neurogastroenterolohy Group, Centre for Digestive Diseases, Blizard Institute, Queen Mary University of London London, UK
| |
Collapse
|
145
|
Nasser Y, Boeckxstaens GE, Wouters MM, Schemann M, Vanner S. Using human intestinal biopsies to study the pathogenesis of irritable bowel syndrome. Neurogastroenterol Motil 2014; 26:455-69. [PMID: 24602069 DOI: 10.1111/nmo.12316] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 01/15/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Although animal models of the irritable bowel syndrome (IBS) have provided important insights, there are no models that fully express the features of this complex condition. One alternative approach is the use of human intestinal biopsies obtained during endoscopic procedures to examine peripheral mechanisms in this disorder. These studies have served to confirm the existence of peripheral pathways in humans with IBS and have provided many new mechanistic insights. Two general approaches have been employed; one approach has been to examine the biological activity of mediators within the mucosal tissue of IBS patients and the other has been to examine changes in the structural properties of key signaling pathways contained within the biopsies. Using these approaches, important changes have been discovered involving the enteric nervous system and the extrinsic sensory pathway (dorsal root ganglia neurons), the immune system, and epithelial signaling in IBS patients compared to healthy subjects. PURPOSE This review will systematically explore these mechanistic pathways, highlight the implications of these novel findings and discuss some of the important limitations of this approach.
Collapse
Affiliation(s)
- Y Nasser
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | | | | | | | | |
Collapse
|
146
|
Sharkey KA, Savidge TC. Reprint of: Role of enteric neurotransmission in host defense and protection of the gastrointestinal tract. Auton Neurosci 2014; 182:70-82. [PMID: 24674836 DOI: 10.1016/j.autneu.2014.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 12/11/2013] [Indexed: 12/11/2022]
Abstract
Host defense is a vital role played by the gastrointestinal tract. As host to an enormous and diverse microbiome, the gut has evolved an elaborate array of chemical and physicals barriers that allow the digestion and absorption of nutrients without compromising the mammalian host. The control of such barrier functions requires the integration of neural, humoral, paracrine and immune signaling, involving redundant and overlapping mechanisms to ensure, under most circumstances, the integrity of the gastrointestinal epithelial barrier. Here we focus on selected recent developments in the autonomic neural control of host defense functions used in the protection of the gut from luminal agents, and discuss how the microbiota may potentially play a role in enteric neurotransmission. Key recent findings include: the important role played by subepithelial enteric glia in modulating intestinal barrier function, identification of stress-induced mechanisms evoking barrier breakdown, neural regulation of epithelial cell proliferation, the role of afferent and efferent vagal pathways in regulating barrier function, direct evidence for bacterial communication to the enteric nervous system, and microbial sources of enteric neurotransmitters. We discuss these new and interesting developments in our understanding of the role of the autonomic nervous system in gastrointestinal host defense.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.
| | - Tor C Savidge
- Texas Children's Microbiome Center, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
147
|
Selenoether oxytocin analogues have analgesic properties in a mouse model of chronic abdominal pain. Nat Commun 2014; 5:3165. [DOI: 10.1038/ncomms4165] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 12/20/2013] [Indexed: 12/13/2022] Open
|
148
|
Neuropeptides and the microbiota-gut-brain axis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 817:195-219. [PMID: 24997035 DOI: 10.1007/978-1-4939-0897-4_9] [Citation(s) in RCA: 289] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuropeptides are important mediators both within the nervous system and between neurons and other cell types. Neuropeptides such as substance P, calcitonin gene-related peptide and neuropeptide Y (NPY), vasoactive intestinal polypeptide, somatostatin and corticotropin-releasing factor are also likely to play a role in the bidirectional gut-brain communication. In this capacity they may influence the activity of the gastrointestinal microbiota and its interaction with the gut-brain axis. Current efforts in elucidating the implication of neuropeptides in the microbiota-gut-brain axis address four information carriers from the gut to the brain (vagal and spinal afferent neurons; immune mediators such as cytokines; gut hormones; gut microbiota-derived signalling molecules) and four information carriers from the central nervous system to the gut (sympathetic efferent neurons; parasympathetic efferent neurons; neuroendocrine factors involving the adrenal medulla; neuroendocrine factors involving the adrenal cortex). Apart from operating as neurotransmitters, many biologically active peptides also function as gut hormones. Given that neuropeptides and gut hormones target the same cell membrane receptors (typically G protein-coupled receptors), the two messenger roles often converge in the same or similar biological implications. This is exemplified by NPY and peptide YY (PYY), two members of the PP-fold peptide family. While PYY is almost exclusively expressed by enteroendocrine cells, NPY is found at all levels of the gut-brain and brain-gut axis. The function of PYY-releasing enteroendocrine cells is directly influenced by short chain fatty acids generated by the intestinal microbiota from indigestible fibre, while NPY may control the impact of the gut microbiota on inflammatory processes, pain, brain function and behaviour. Although the impact of neuropeptides on the interaction between the gut microbiota and brain awaits to be analysed, biologically active peptides are likely to emerge as neural and endocrine messengers in orchestrating the microbiota-gut-brain axis in health and disease.
Collapse
|
149
|
Abstract
The transient receptor potential ankyrin subtype 1 protein (TRPA1) is a nonselective cation channel permeable to Ca(2+), Na(+), and K(+). TRPA1 is a promiscuous chemical nocisensor that is also involved in noxious cold and mechanical sensation. It is present in a subpopulation of Aδ- and C-fiber nociceptive sensory neurons as well as in other sensory cells including epithelial cells. In primary sensory neurons, Ca(2+) and Na(+) flowing through TRPA1 into the cell cause membrane depolarization, action potential discharge, and neurotransmitter release both at peripheral and central neural projections. In addition to being activated by cysteine and lysine reactive electrophiles and oxidants, TRPA1 is indirectly activated by pro-inflammatory agents via the phospholipase C signaling pathway, in which cytosolic Ca(2+) is an important regulator of channel gating. The finding that non-electrophilic compounds, including menthol and cannabinoids, activate TRPA1 may provide templates for the design of non-tissue damaging activators to fine-tune the activity of TRPA1 and raises the possibility that endogenous ligands sharing binding sites with such non-electrophiles exist and regulate TRPA1 channel activity. TRPA1 is promising as a drug target for novel treatments of pain, itch, and sensory hyperreactivity in visceral organs including the airways, bladder, and gastrointestinal tract.
Collapse
Affiliation(s)
- Peter M Zygmunt
- Clinical and Experimental Pharmacology, Clinical Chemistry, Department of Laboratory Medicine, Lund University, Skåne University Hospital, SE-221 85, Lund, Sweden,
| | | |
Collapse
|
150
|
Role of enteric neurotransmission in host defense and protection of the gastrointestinal tract. Auton Neurosci 2013; 181:94-106. [PMID: 24412639 DOI: 10.1016/j.autneu.2013.12.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 12/11/2013] [Indexed: 12/24/2022]
Abstract
Host defense is a vital role played by the gastrointestinal tract. As host to an enormous and diverse microbiome, the gut has evolved an elaborate array of chemical and physicals barriers that allow the digestion and absorption of nutrients without compromising the mammalian host. The control of such barrier functions requires the integration of neural, humoral, paracrine and immune signaling, involving redundant and overlapping mechanisms to ensure, under most circumstances, the integrity of the gastrointestinal epithelial barrier. Here we focus on selected recent developments in the autonomic neural control of host defense functions used in the protection of the gut from luminal agents, and discuss how the microbiota may potentially play a role in enteric neurotransmission. Key recent findings include: the important role played by subepithelial enteric glia in modulating intestinal barrier function, identification of stress-induced mechanisms evoking barrier breakdown, neural regulation of epithelial cell proliferation, the role of afferent and efferent vagal pathways in regulating barrier function, direct evidence for bacterial communication to the enteric nervous system, and microbial sources of enteric neurotransmitters. We discuss these new and interesting developments in our understanding of the role of the autonomic nervous system in gastrointestinal host defense.
Collapse
|