Advertisement
Review| Volume 7, ISSUE 2, P433-445, 2019

Enteric Glia: A New Player in Abdominal Pain

  • Wilmarie Morales-Soto
    Affiliations
    Neuroscience Program, Department of Physiology, Michigan State University, East Lansing, Michigan
    Search for articles by this author
  • Brian D. Gulbransen
    Correspondence
    Correspondence Address correspondence to: Brian D. Gulbransen, PhD, Neuroscience Program and Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, Michigan 48824. fax: (517) 355-5125.
    Affiliations
    Neuroscience Program, Department of Physiology, Michigan State University, East Lansing, Michigan
    Search for articles by this author
Open AccessPublished:November 24, 2018DOI:https://doi.org/10.1016/j.jcmgh.2018.11.005
      Chronic abdominal pain is the most common gastrointestinal issue and contributes to the pathophysiology of functional bowel disorders and inflammatory bowel disease. Current theories suggest that neuronal plasticity and broad alterations along the brain-gut axis contribute to the development of chronic abdominal pain, but the specific mechanisms involved in chronic abdominal pain remain incompletely understood. Accumulating evidence implicates glial cells in the development and maintenance of chronic pain. Astrocytes and microglia in the central nervous system and satellite glia in dorsal root ganglia contribute to chronic pain states through reactive gliosis, the modification of glial networks, and the synthesis and release of neuromodulators. In addition, new data suggest that enteric glia, a unique type of peripheral glia found within the enteric nervous system, have the potential to modify visceral perception through interactions with neurons and immune cells. Understanding these emerging roles of enteric glia is important to fully understand the mechanisms that drive chronic pain and to identify novel therapeutic targets. In this review, we discuss enteric glial cell signaling mechanisms that have the potential to influence chronic abdominal pain.

      Graphical abstract

      Keywords

      Abbreviations used in this paper:

      ATP (adenosine triphosphate), BDNF (brain-derived neurotrophic factor), Cx-43 (connexin-43), DRG (dorsal root ganglia), GFAP (glial fibrillary acidic protein), GI (gastrointestinal), IBD (irritable bowel disease), IBS (irritable bowel syndrome), IL (interleukin), TNF (tumor necrosis factor), TRPV1 (transient receptor potential vanilloid receptor type-1)
      Chronic abdominal pain is a common gastrointestinal issue, but the mechanisms that produce abdominal pain are unresolved. Here, we discuss evidence suggesting that glia, and in particular, enteric glia, function as central players in mechanisms that produce abdominal pain.
      Abdominal pain is the most common gastrointestinal (GI) issue and results in more than 15 million office visits per year.
      • Peery A.F.
      • Crockett S.D.
      • Barritt A.S.
      • Dellon E.S.
      • Eluri S.
      • Gangarosa L.M.
      • Jensen E.T.
      • Lund J.L.
      • Pasricha S.
      • Runge T.
      • Schmidt M.
      • Shaheen N.J.
      • Sandler R.S.
      Burden of gastrointestinal, liver, and pancreatic diseases in the United States.
      Chronic abdominal pain affects at least 10%–15% of the general population and is a characteristic feature of functional bowel disorders, such as irritable bowel syndrome (IBS), the inflammatory bowel diseases (IBDs), and is a growing problem associated with chronic opioid use.
      • Mansfield K.E.
      • Sim J.
      • Jordan J.L.
      • Jordan K.P.
      A systematic review and meta-analysis of the prevalence of chronic widespread pain in the general population.
      • Longstreth G.F.
      • Thompson G.W.
      • Chey W.D.
      • Houghton L.A.
      • Mearin F.
      • Spiller R.C.
      Functional bowel disorders.
      • Regueiro M.
      • Greer J.B.
      • Szigethy E.
      Etiology and treatment of pain and psychosocial issues in patients with inflammatory bowel diseases.
      Acute abdominal pain is associated with inflammatory flare-ups in IBD, but a large subset of patients that suffer from IBD eventually develop chronic abdominal pain even after achieving clinical remission. This condition, called comorbid IBS (loosely termed “IBS-IBD”), is present in up to 46%–59% of patients with Crohn’s disease and 36%–38% of patients with ulcerative colitis in remission.
      • Keohane J.
      • O’Mahony C.
      • O’Mahony L.
      • O’Mahony S.
      • Quigley E.M.
      • Shanahan F.
      Irritable bowel syndrome–type symptoms in patients with inflammatory bowel disease: a real association or reflection of occult inflammation?.
      • Minderhoud I.M.
      • Oldenburg B.
      • Wismeijer J.A.
      • Henegouwen G.P.
      • Smout A.
      IBS-like symptoms in patients with inflammatory bowel disease in remission; relationships with quality of life and coping behavior.
      • Halpin S.J.
      • Ford A.C.
      Prevalence of symptoms meeting criteria for irritable bowel syndrome in inflammatory bowel disease: systematic review and meta-analysis.
      Abdominal pain linked to these disorders is common in adults and pediatric patients and is a major contributing factor to their low quality of life and high morbidity. As a result, the US Food and Drug Administration now requires the use of abdominal pain as a patient-reported outcome to assess the efficacy of new therapies for Crohn’s disease.
      • Regueiro M.
      • Greer J.B.
      • Szigethy E.
      Etiology and treatment of pain and psychosocial issues in patients with inflammatory bowel diseases.
      The mechanisms that drive the development of chronic abdominal pain remain largely unresolved and this presents a major barrier to progress in the development of new therapies. Current therapies for IBD, for example, primarily focus on controlling inflammation with amino salicylates, corticosteroids, immunomodulators, and biologic agents. Although these agents clearly benefit the treatment of active inflammation, they do not address abdominal pain.
      • Colombel J.-F.
      • Mahadevan U.
      Inflammatory bowel disease 2017: innovations and changing paradigms.
      Opioids are the current frontline therapy for chronic abdominal pain, but chronic opioid use has serious complications and produces a condition called narcotic bowel syndrome that is characterized by abdominal pain.
      • Camilleri M.
      • Lembo A.
      • Katzka D.A.
      Opioids in gastroenterology: treating adverse effects and creating therapeutic benefits.
      This is a serious and growing issue that requires a more sophisticated understanding of the causal mechanisms to permit the development of more effective therapies.
      Current theories suggest that neuronal plasticity involving the sensitization of visceral afferent sensory nerve fibers and broad alterations to the brain-gut axis contribute to the development of chronic abdominal pain.
      • Ji R.-R.
      • Chamessian A.
      • Zhang Y.-Q.
      Pain regulation by non-neuronal cells and inflammation.
      • Mayer E.A.
      • Tillisch K.
      The brain-gut axis in abdominal pain syndromes.
      Much of this theory is based on evidence demonstrating changes in neuronal sensitivity, firing patterns, and network activity in the periphery, brain, and spinal cord.
      • Gold M.S.
      • Gebhart G.F.
      Nociceptor sensitization in pain pathogenesis.
      • Cervero F.
      • Laird J.
      Understanding the signaling and transmission of visceral nociceptive events.
      • Basbaum A.I.
      • Bautista D.M.
      • Scherrer G.
      • Julius D.
      Cellular and molecular mechanisms of pain.
      Although little is still known regarding the mechanisms that drive these alterations to neurons and their networks, it is increasingly clear that these properties are regulated by glia.
      Here we discuss evidence supporting the concept that changes to glia contribute to mechanisms involved in the development of abdominal pain. We primarily focus on enteric glia in this review because of their potential to interact with nociceptors in the intestine and the potential consequences of these interactions on the generation of abdominal pain. Furthermore, the topic of central glia in pain transmission has been well covered by recent reviews.
      • Ji R.-R.
      • Chamessian A.
      • Zhang Y.-Q.
      Pain regulation by non-neuronal cells and inflammation.
      • Ji R.-R.R.
      • Berta T.
      • Nedergaard M.
      Glia and pain: is chronic pain a gliopathy?.
      However, readers should note that alterations to central glia play a major role in pain transmission and, in fact, much of the understanding of glia in neuropathic pain conditions comes from research on glia in central circuits. A more sophisticated understanding of how central and peripheral glia regulate neural activity and how changes to these properties contributes to altered transmission or perception of sensory stimuli in the intestine holds great potential to advance the field of abdominal pain research and to identify novel therapeutic targets. In this review we aim to amass the existing evidence implicating enteric glia as major contributors to peripheral sensitization of nerve afferents and highlight their potential roles in the cause of chronic abdominal pain.

      Known Mechanisms That Contribute to Abdominal Pain

      Visceral hypersensitivity is a major mechanism that contributes to abdominal pain in IBS and IBD.
      • Simrén M.
      • Törnblom H.
      • Palsson O.S.
      • van Tilburg M.A.
      • Oudenhove L.
      • Tack J.
      • Whitehead W.E.
      Visceral hypersensitivity is associated with GI symptom severity in functional GI disorders: consistent findings from five different patient cohorts.
      This involves alterations along the brain-gut axis, a network of afferent and efferent neural pathways that link cognitive, emotional, and autonomic centers in the brain to neuroendocrine centers, the enteric nervous system, the gut microbiome, and the immune system. The sensitization of visceral afferent nerve fibers is driven by neuroplasticity and leads to a skewed perception of sensory stimuli. Nociceptive information is transduced by the peripheral axon terminals of primary afferent neurons whose cell bodies reside in dorsal root ganglia (DRG). These “nociceptors” are the axon terminals of polymodal C fibers that transduce a variety of potentially noxious stimuli (mechanical, chemical, and thermal) to the brain. The sensitivity of nociceptors is one of the most important factors that gates the transmission of noxious information and alterations to nociceptor sensitivity in GI disorders can lead to an increased perception of pain (hyperalgesia) or the painful perception to innocuous stimuli (allodynia).
      Immune activation and neuroplasticity are 2 key mechanisms involved in the generation of chronic abdominal pain. Nociceptor sensitivity is profoundly altered by inflammatory mediators.
      • Stein C.
      • Machelska H.
      Modulation of peripheral sensory neurons by the immune system: implications for pain therapy.
      Active inflammation, such as an IBD flare-up or acute gastroenteritis, involves the release of proinflammatory mediators that alter nociceptor sensitivity. Inflammatory mediators including adenosine triphosphate (ATP),
      • Cook S.
      • McCleskey E.
      Cell damage excites nociceptors through release of cytosolic ATP.
      histamine,
      • Wouters M.M.
      • Balemans D.
      • Wanrooy S.
      • Dooley J.
      • Cibert-Goton V.
      • Alpizar Y.A.
      • Valdez-Morales E.E.
      • Nasser Y.
      • Veldhoven P.P.
      • Vanbrabant W.
      • der Merwe S.
      • Mols R.
      • Ghesquière B.
      • Cirillo C.
      • Kortekaas I.
      • Carmeliet P.
      • Peetermans W.E.
      • Vermeire S.
      • Rutgeerts P.
      • Augustijns P.
      • Hellings P.W.
      • Belmans A.
      • Vanner S.
      • Bulmer D.C.
      • Talavera K.
      • Berghe P.
      • Liston A.
      • Boeckxstaens G.E.
      Histamine receptor H1–mediated sensitization of TRPV1 mediates visceral hypersensitivity and symptoms in patients with irritable bowel syndrome.
      interleukin (IL)-1β,
      • Binshtok A.M.
      • Wang H.
      • Zimmermann K.
      • Amaya F.
      • Vardeh D.
      • Shi L.
      • Brenner G.J.
      • Ji R.-R.
      • Bean B.P.
      • Woolf C.J.
      • Samad T.A.
      Nociceptors are interleukin-1β sensors.
      proteases,
      • Amadesi S.
      • Cottrell G.S.
      • Divino L.
      • Chapman K.
      • Grady E.F.
      • Bautista F.
      • Karanjia R.
      • Barajas-Lopez C.
      • Vanner S.
      • Vergnolle N.
      • Bunnett N.W.
      Protease-activated receptor 2 sensitizes TRPV1 by protein kinase Cɛ- and A-dependent mechanisms in rats and mice.
      and bradykinins
      • Bandell M.
      • Story G.M.
      • Hwang S.
      • Viswanath V.
      • Eid S.R.
      • Petrus M.J.
      • Earley T.J.
      • Patapoutian A.
      Noxious cold ion channel TRPA1 is activated by pungent compounds and bradykinin.
      cause pain by interacting with receptors on nociceptors. Other inflammatory mediators, such as IL-6
      • Obreja O.
      • Biasio W.
      • Andratsch M.
      • Lips K.
      • Rathee P.
      • Ludwig A.
      • Rose-John S.
      • Kress M.
      Fast modulation of heat-activated ionic current by proinflammatory interleukin 6 in rat sensory neurons.
      and tumor-necrosis factor (TNF)-α,
      • Czeschik J.
      • Hagenacker T.
      • Schäfers M.
      • Büsselberg D.
      TNF-α differentially modulates ion channels of nociceptive neurons.
      lead to the sensitization of nociceptors through indirect mechanisms. Nociceptors, themselves, also contribute to local inflammation (termed “neuroinflammation”) through axon reflexes that produce inflammatory mediators including substance P, neurokinin A, ATP, and calcitonin-gene related peptide.
      Neuroimmune activation following a psychological stressor or peripheral inflammation results in neuroplastic changes along nerve afferents that contribute to visceral hyperalgesia.
      • Azpiroz F.
      • Bouin M.
      • Camilleri M.
      • Mayer E.
      • Poitras P.
      • Serra J.
      • Spiller R.
      Mechanisms of hypersensitivity in IBS and functional disorders.
      • Sengupta J.N.
      Sensory nerves.
      Neuroimmune modulators drive neuroplasticity through mechanisms that include the downregulation of glutamate transporters and the subsequent upregulation of synaptic glutamate and the ionotropic glutamate receptor, N-methyl-d-aspartate, signaling.
      • Li J.
      • McRoberts J.A.
      • Ennes H.S.
      • Trevisani M.
      • Nicoletti P.
      • Mittal Y.
      • Mayer E.A.
      Experimental colitis modulates the functional properties of NMDA receptors in dorsal root ganglia neurons.
      • Sung B.
      • Lim G.
      • Mao J.
      Altered expression and uptake activity of spinal glutamate transporters after nerve injury contribute to the pathogenesis of neuropathic pain in rats.
      Neuroplastic changes in the expression and function of transient receptor potential vanilloid receptor type-1 (TRPV1) channels present on nociceptors innervating the intestinal mucosa are also implicated in visceral hypersensitivity in IBS.
      • Jones C.R.
      • Xu L.
      • Gebhart G.
      The mechanosensitivity of mouse colon afferent fibers and their sensitization by inflammatory mediators require transient receptor potential vanilloid 1 and acid-sensing ion channel 3.
      Increased levels of TRPV1 are present in patients with idiopathic rectal hypersensitivity
      • Chan C.
      • Facer P.
      • Davis J.
      • Smith G.
      • Egerton J.
      • Bountra C.
      • Williams N.
      • Anand P.
      Sensory fibers expressing capsaicin receptor TRPV1 in patients with rectal hypersensitivity and fecal urgency.
      and an increased density of TRPV1 immunoreactive nerve fibers is present in the colonic mucosa of patients with IBS, correlating with the degree of abdominal pain experienced.
      • Akbar A.
      • Yiangou Y.
      • Facer P.
      • Walters J.
      • Anand P.
      • Ghosh S.
      Increased capsaicin receptor TRPV1-expressing sensory fibers in irritable bowel syndrome and their correlation with abdominal pain.
      Alterations to emotional circuits of the brain, primarily those associated with stress and anxiety, contribute to visceral hypersensitivity in patients with IBS.
      • Larauche M.
      • Mulak A.
      • Taché Y.
      Stress-related alterations of visceral sensation: animal models for irritable bowel syndrome study.
      Various stressors, chronic and acute, regulate visceral pain responses in animal models
      • Ren T.-H.
      • Wu J.
      • Yew D.
      • Ziea E.
      • Lao L.
      • Leung W.-K.
      • Berman B.
      • Hu P.-J.
      • Sung J.J.
      Effects of neonatal maternal separation on neurochemical and sensory response to colonic distension in a rat model of irritable bowel syndrome.
      • Tramullas M.
      • Finger B.C.
      • Moloney R.D.
      • Golubeva A.V.
      • Moloney G.
      • Dinan T.G.
      • Cryan J.F.
      Toll-like receptor 4 regulates chronic stress-induced visceral pain in mice.
      and are associated with an increase in symptom severity in functional
      • Elsenbruch S.
      • Rosenberger C.
      • Bingel U.
      • Forsting M.
      • Schedlowski M.
      • Gizewski E.R.
      Patients with irritable bowel syndrome have altered emotional modulation of neural responses to visceral stimuli.
      and inflammatory GI disorders.
      • Drewes A.
      • Frkjær J.
      • Larsen E.
      • Reddy H.
      • Arendt-Nielsen L.
      • Gregersen H.
      Pain and mechanical properties of the rectum in patients with active ulcerative colitis.
      • Bielefeldt K.
      • Davis B.
      • Binion D.G.
      Pain and inflammatory bowel disease.
      Brain regions associated with visceral pain, emotional arousal, and attention are consistently activated in neuroimaging studies of patients with IBS.
      • Tillisch K.
      • Mayer E.A.
      • Labus J.S.
      Quantitative meta-analysis identifies brain regions activated during rectal distension in irritable bowel syndrome.
      Furthermore, alterations to gray matter density in areas associated with corticolimbic inhibition, stress, and arousal circuits indicate altered activity in the brains of patients with IBS.
      • Seminowicz D.A.
      • Labus J.S.
      • Bueller J.A.
      • Tillisch K.
      • Naliboff B.D.
      • Bushnell C.M.
      • Mayer E.A.
      Regional gray matter density changes in brains of patients with irritable bowel syndrome.
      The activation of these regions contributes to acute and chronic stress-induced hyperalgesia in the colon, in part, by stimulating the release of factors, such as corticotropin-releasing factor.
      • Larauche M.
      • Bradesi S.
      • Million M.
      • McLean P.
      • Taché Y.
      • Mayer E.A.
      • McRoberts J.A.
      Corticotropin-releasing factor type 1 receptors mediate the visceral hyperalgesia induced by repeated psychological stress in rats.
      • La J.-H.
      Peripheral corticotropin releasing hormone mediates post-inflammatory visceral hypersensitivity in rats.

      Glial Cells in Abdominal Pain Pathways

      A plethora of emerging studies link nerve fiber sensitization with alterations to glial cells. Increased glial activity is characteristic of all forms of pain and glia contribute to the transition from acute to chronic pain through interactions with neurons that alter neurotransmission.
      • Ji R.-R.R.
      • Berta T.
      • Nedergaard M.
      Glia and pain: is chronic pain a gliopathy?.
      • Cairns B.E.
      • Arendt-Nielsen L.
      • Sacerdote P.
      Perspectives in pain research 2014: neuroinflammation and glial cell activation: the cause of transition from acute to chronic pain?.
      For example, increases in astrocyte and microglial cell markers are present in animal models of acute pain, inflammatory pain, and neuropathic pain.
      • Stuesse S.L.
      • Crisp T.
      • McBurney D.L.
      • Schechter J.B.
      • Lovell J.A.
      • Cruce W.L.R.
      Neuropathic pain in aged rats: behavioral responses and astrocytic activation.
      • Coyle D.E.
      Partial peripheral nerve injury leads to activation of astroglia and microglia which parallels the development of allodynic behavior.
      • Sweitzer S.M.
      • Colburn R.W.
      • Rutkowski M.
      • DeLeo J.A.
      Acute peripheral inflammation induces moderate glial activation and spinal IL-1β expression that correlates with pain behavior in the rat.
      • Tanga F.
      • Raghavendra V.
      • DeLeo J.
      Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain.
      Structural changes and altered signaling in peripheral glial cells, such as satellite glia in DRG, are implicated in abdominal pain pathways.
      • Hanani M.
      Role of satellite glial cells in gastrointestinal pain.
      Furthermore, enteric glial cells, a unique class of peripheral glia found within the enteric nervous system, have bidirectional interactions with neurons in the gut that impact both gut reflexes in health and neuroinflammation.
      • Brown I.A.
      • Gulbransen B.D.
      The antioxidant glutathione protects against enteric neuron death in situ, but its depletion is protective during colitis.
      • Grubišić V.
      • Gulbransen B.D.
      Enteric glia: the most alimentary of all glia.
      • McClain J.L.
      • Fried D.E.
      • Gulbransen B.D.
      Agonist-evoked Ca2+ signaling in enteric glia drives neural programs that regulate intestinal motility in mice.
      • Brown I.A.
      • McClain J.L.
      • Watson R.E.
      • Patel B.A.
      • Gulbransen B.D.
      Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide.
      • McClain J.L.
      • Grubišić V.
      • Fried D.
      • Gomez-Suarez R.A.
      • Leinninger G.M.
      • Sévigny J.
      • Parpura V.
      • Gulbransen B.D.
      Ca2+ responses in enteric glia are mediated by connexin-43 hemichannels and modulate colonic transit in mice.
      • Grubišić V.
      • Gulbransen B.D.
      Enteric glial activity regulates secretomotor function in the mouse colon but does not acutely affect gut permeability.
      Therefore, an emerging theme is that glia present at all sites along ascending and descending pathways that transduce and modify nociceptive information deriving from the intestine have the potential to broadly influence visceral perception (Figure 1).
      Figure thumbnail gr1
      Figure 1Major populations of glia that contribute to chronic pain. Astrocytes and microglia are present in the central nervous system and primarily affect chronic pain pathways in the brain and in the spinal cord. Satellite glia are peripheral glial cells located in the dorsal root ganglia where they contribute to the sensitization of dorsal root ganglion cells. Enteric glial cells are located within the intestine and form part of the enteric nervous system. Enteric glia are located adjacent to the nerve endings of primary afferent fibers and intercellular interactions between enteric glia and neurons have the potential to influence chronic pain.
      Most of the understanding of glia in pain transmission stems from studies of glia at the first synapse in ascending pain pathways in the dorsal horn of the spinal cord. Microglia and astrocytes are the major glial cell types implicated in altering pain transmission at this level. Microglial activation in the spinal cord occurs in response to injury or inflammation and is reflected by increased cell number and altered gene expression profiles.
      • Ji R.-R.R.
      • Berta T.
      • Nedergaard M.
      Glia and pain: is chronic pain a gliopathy?.
      • Tanga F.
      • Raghavendra V.
      • DeLeo J.
      Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain.
      • Gosselin R.-D.
      • Suter M.R.
      • Ji R.-R.
      • Decosterd I.
      Glial cells and chronic pain.
      • Guo W.
      • Wang H.
      • Watanabe M.
      • Shimizu K.
      • Zou S.
      • LaGraize S.C.
      • Wei F.
      • Dubner R.
      • Ren K.
      Glial–cytokine–neuronal interactions underlying the mechanisms of persistent pain.
      Likewise, spinal astrocytes become activated and undergo reactive gliosis in animal models of neuropathic pain and increased expression of glial fibrillary acidic protein (GFAP), a marker of reactive astrocytes, mirrors the intensity of pain hypersensitivity.
      • Tanga F.
      • Raghavendra V.
      • DeLeo J.
      Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain.
      • Guo W.
      • Wang H.
      • Watanabe M.
      • Shimizu K.
      • Zou S.
      • LaGraize S.C.
      • Wei F.
      • Dubner R.
      • Ren K.
      Glial–cytokine–neuronal interactions underlying the mechanisms of persistent pain.
      • Romero-Sandoval A.
      • Chai N.
      • Nutile-McMenemy N.
      • DeLeo J.A.
      A comparison of spinal Iba1 and GFAP expression in rodent models of acute and chronic pain.
      The inhibition of astrocyte activity results in a significant attenuation of inflammatory hyperalgesia, suggesting that astrocytic activation is at least partly necessary for the development of pain.
      • Guo W.
      • Wang H.
      • Watanabe M.
      • Shimizu K.
      • Zou S.
      • LaGraize S.C.
      • Wei F.
      • Dubner R.
      • Ren K.
      Glial–cytokine–neuronal interactions underlying the mechanisms of persistent pain.
      Evidence from studies using in vivo optogenetics suggests that the selective activation of spinal astrocytes is sufficient to increase pain hypersensitivity.
      • Nam Y.
      • Kim J.-H.
      • Kim J.-H.
      • Jha M.
      • Jung J.
      • Lee M.-G.
      • Choi I.-S.
      • Jang I.-S.
      • Lim D.
      • Hwang S.-H.
      • Cho H.-J.
      • Suk K.
      Reversible induction of pain hypersensitivity following optogenetic stimulation of spinal astrocytes.
      There is still much controversy surrounding glial optogenetic data, because several studies suggest that the activation of astrocytes driven by channelrhodopsin is an effect of cell swelling and is not driven by normal calcium signaling pathways.
      • Cho W.-H.
      • Barcelon E.
      • Lee S.
      Optogenetic glia manipulation: possibilities and future prospects.
      Alternative methods of glial-specific activation that more closely reflect physiological mechanisms, such as designer receptors exclusively activated by designer drugs, should be implemented to further validate these findings.
      Glial activity in the spinal cord contributes to neuroplasticity, in part by stimulating the release of glial-derived substances termed “gliotransmitters.” Active glial cells produce several proinflammatory mediators that contribute to the activation and sensitization of nerve fibers to central sensitization in the dorsal horn.
      • Ji R.-R.
      • Chamessian A.
      • Zhang Y.-Q.
      Pain regulation by non-neuronal cells and inflammation.
      For example, TNF-α produced by spinal microglia and astrocytes contributes to hyperalgesia and allodynia in preclinical pain models.
      • Zhong Y.
      • Zhou L.-J.
      • Ren W.-J.
      • Xin W.-J.
      • Li Y.-Y.
      • Zhang T.
      • Liu X.-G.
      The direction of synaptic plasticity mediated by C-fibers in spinal dorsal horn is decided by Src-family kinases in microglia: the role of tumor necrosis factor-α.
      • Gao Y.
      • Zhang L.
      • Glia J.R.
      Spinal injection of TNF-α-activated astrocytes produces persistent pain symptom mechanical allodynia by releasing monocyte chemoattractant protein-1.
      TNF-α released by astrocytes and microglia also contributes to central sensitization and neuroplasticity through mechanisms that involve the downregulation of glutamate transporters and subsequently, glutamate excitotoxicity.
      • Sung B.
      • Lim G.
      • Mao J.
      Altered expression and uptake activity of spinal glutamate transporters after nerve injury contribute to the pathogenesis of neuropathic pain in rats.
      Other key gliotransmitters, such as purines, activate nerve fibers directly
      • Cook S.
      • McCleskey E.
      Cell damage excites nociceptors through release of cytosolic ATP.
      and in animal models of chronic pain, astrocytes increase the release of ATP and the production of proalgesic mediators, such as glutamate, cytokines, and chemokines.
      • Ji R.-R.
      • Chamessian A.
      • Zhang Y.-Q.
      Pain regulation by non-neuronal cells and inflammation.
      Glial mechanisms similar to those occurring in the spinal cord may also affect processing in the brain. Although less is known regarding the contributions of glia in the brain to pain conditions, increased astrocyte activity is observed in the nucleus of the solitary tract following colonic inflammation.
      • Sun Y.-N.
      • Luo J.-Y.
      • Rao Z.-R.
      • Lan L.
      • Duan L.
      GFAP and Fos immunoreactivity in lumbo-sacral spinal cord and medulla oblongata after chronic colonic inflammation in rats.
      Likewise, microglial activation within the thalamus occurs following nociceptive spinal cord injury.
      • Zhao P.
      • Waxman S.G.
      • Hains B.C.
      Modulation of thalamic nociceptive processing after spinal cord injury through remote activation of thalamic microglia by cysteine–cysteine chemokine ligand 21.
      Alterations to glia in the brain are likely associated with the emotional experience of pain and can be correlated with mood disorders, such as depression and anxiety.
      • Rajkowska G.
      • Miguel-Hidalgo J.
      Gliogenesis and glial pathology in depression.
      Multiple populations of peripheral glial cells actively contribute to modifying pain transmission through effects on nociceptors. For example, satellite glia play crucial roles in visceral sensitization. Satellite glia are located in peripheral sensory ganglia where they envelop neuronal cell bodies.
      • Hanani M.
      Satellite glial cells in sympathetic and parasympathetic ganglia: In search of function.
      A recent review by Hanani
      • Hanani M.
      Role of satellite glial cells in gastrointestinal pain.
      thoroughly covered the topic of satellite glia in the development and maintenance of chronic pain. To briefly summarize, satellite glia undergo cell division and upregulate their production of GFAP following inflammation or injury to peripheral nerves and express and release proinflammatory cytokines including TNF-α and IL-1β. In addition, satellite glia increase gap-junction mediated coupling following nerve injury and inflammation. These changes to satellite glia are linked to GI pain and are also implicated in chronic abdominal pain in IBS.

      Potential Roles of Enteric Glia in Abdominal Pain

      Enteric glial cells are a unique class of peripheral glia that are associated with neurons in the enteric nervous system. Enteric glia play instrumental roles in gut reflexes that regulate motility and secretion, influence the epithelial barrier, and modulate neuroinflammation.
      • Grubišić V.
      • Gulbransen B.D.
      Enteric glia: the most alimentary of all glia.
      • Gulbransen B.D.
      Do nerves make bowels irritable?.
      Yet how enteric glia might influence visceral hypersensitivity and nociceptor sensitization is still relatively unknown.
      Enteric glia, particularly those associated with neurons in the myenteric plexus, share some morphologic and functional properties with astrocytes that might suggest similar roles in pain transmission. For example, enteric glia express molecular markers, such as GFAP
      • Jessen K.
      • Mirsky R.
      Astrocyte-like glia in the peripheral nervous system: an immunohistochemical study of enteric glia.
      and S100β
      • Cocchia D.
      • Michetti F.
      • Ferri G.-L.
      • Polak J.M.
      • Probert L.
      • Marangos P.J.
      Evidence for the presence of S-100 protein in the glial component of the human enteric nervous system.
      ; provide trophic and protective support to enteric neurons
      • Gulbransen B.D.
      • Sharkey K.A.
      Novel functional roles for enteric glia in the gastrointestinal tract.
      • von Boyen G.B.
      • Steinkamp M.
      • Reinshagen M.
      • Schäfer K.H.
      • Adler G.
      • Kirsch J.
      Proinflammatory cytokines increase glial fibrillary acidic protein expression in enteric glia.
      ; exhibit activity encoded by intracellular calcium signaling in response to neurotransmitters; and release gliotransmitters through membrane channels composed of connexin-43 (Cx-43).
      • McClain J.L.
      • Grubišić V.
      • Fried D.
      • Gomez-Suarez R.A.
      • Leinninger G.M.
      • Sévigny J.
      • Parpura V.
      • Gulbransen B.D.
      Ca2+ responses in enteric glia are mediated by connexin-43 hemichannels and modulate colonic transit in mice.
      Connexins are a major structural component of gap junctions and hemichannels in glia and connexin-mediated glial mechanisms play a crucial role in development of chronic neuropathic pain.
      • Chen M.J.
      • Kress B.
      • Han X.
      • Moll K.
      • Peng W.
      • Ji R.
      • Nedergaard M.
      Astrocytic CX43 hemichannels and gap junctions play a crucial role in development of chronic neuropathic pain following spinal cord injury.
      Bidirectional communication between enteric neurons and glia mediated by the Cx-43-dependent release of enteric gliotransmitters regulates gut reflexes that control motility and secretions
      • McClain J.L.
      • Fried D.E.
      • Gulbransen B.D.
      Agonist-evoked Ca2+ signaling in enteric glia drives neural programs that regulate intestinal motility in mice.
      • McClain J.L.
      • Grubišić V.
      • Fried D.
      • Gomez-Suarez R.A.
      • Leinninger G.M.
      • Sévigny J.
      • Parpura V.
      • Gulbransen B.D.
      Ca2+ responses in enteric glia are mediated by connexin-43 hemichannels and modulate colonic transit in mice.
      • Grubišić V.
      • Gulbransen B.D.
      Enteric glial activity regulates secretomotor function in the mouse colon but does not acutely affect gut permeability.
      • Fried D.E.
      • Watson R.E.
      • Robson S.C.
      • Gulbransen B.D.
      Ammonia modifies enteric neuromuscular transmission through glial γ-aminobutyric acid signaling.
      and the Cx-43-dependent release of gliotransmitters, such as ATP, during neuroinflammation drives neurodegeneration through the activation of neuronal P2X7 receptors.
      • Brown I.A.
      • McClain J.L.
      • Watson R.E.
      • Patel B.A.
      • Gulbransen B.D.
      Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide.
      • Gulbransen B.D.
      • Bashashati M.
      • Hirota S.A.
      • Gui X.
      • Roberts J.A.
      • MacDonald J.A.
      • Muruve D.A.
      • McKay D.M.
      • Beck P.L.
      • Mawe G.M.
      Activation of neuronal P2X7 receptor–pannexin-1 mediates death of enteric neurons during colitis.
      Enteric glia are intimately associated with nociceptors in the intestine and the activation of TRPV1+ sensory neurons within the myenteric plexus elicits glial activity and gliotransmitter release through similar mechanisms.
      • Delvalle N.
      • Dharshika C.
      • Morales-Soto W.
      • Fried D.E.
      • Gaudette L.
      • Gulbransen B.D.
      Communication between enteric neurons, glia, and nociceptors underlies the effects of tachykinins on neuroinflammation.
      Whether gliotransmitters subsequently have direct or indirect effects on nociceptor activity or sensitivity is still unknown, but this remains an active area of research.
      Enteric glia are targets of stress, which is a primary component in the development of visceral hypersensitivity. Several forms of stress, such as chronic stress and early life stress, alter glial signaling and contribute to pain sensitivity in IBS.
      • Larauche M.
      • Mulak A.
      • Taché Y.
      Stress-related alterations of visceral sensation: animal models for irritable bowel syndrome study.
      The contribution of enteric glia to stress-induced hyperalgesia is still undefined. However, early life stress causes structural changes in enteric glial cells that are correlated with inefficient gastric motility.
      • Fujikawa Y.
      • Tominaga K.
      • Tanaka F.
      • Tanigawa T.
      • Watanabe T.
      • Fujiwara Y.
      • Arakawa T.
      Enteric glial cells are associated with stress-induced colonic hyper-contraction in maternally separated rats.
      Furthermore, stress activates sympathetic pathways in the intestine that activate enteric glia via purinergic transmission.
      • Gulbransen B.D.
      • Bains J.S.
      • Sharkey K.A.
      Enteric glia are targets of the sympathetic innervation of the myenteric plexus in the guinea pig distal colon.
      Enteric glia contribute to intestinal inflammatory and immune responses
      • Cabarrocas J.
      • Savidge T.C.
      • Liblau R.S.
      Role of enteric glial cells in inflammatory bowel disease.
      and interact with, and express, cytokines and immunoregulatory signals that contribute to altered nociceptive signaling.
      • Cabarrocas J.
      • Savidge T.C.
      • Liblau R.S.
      Role of enteric glial cells in inflammatory bowel disease.
      • Tjwa E.
      • Bradley J.M.
      • Keenan C.M.
      • Kroese A.
      • Sharkey K.A.
      Interleukin-1β activates specific populations of enteric neurons and enteric glia in the guinea pig ileum and colon.
      • Rühl A.
      • Franzke S.
      • Collins Stremmel W.
      Interleukin-6 expression and regulation in rat enteric glial cells.
      Altered immune responses are key components in the development of chronic pain and mediators that sensitize nociceptors, such as neurokinin A,
      • Delvalle N.
      • Dharshika C.
      • Morales-Soto W.
      • Fried D.E.
      • Gaudette L.
      • Gulbransen B.D.
      Communication between enteric neurons, glia, and nociceptors underlies the effects of tachykinins on neuroinflammation.
      substance P,
      • Delvalle N.
      • Dharshika C.
      • Morales-Soto W.
      • Fried D.E.
      • Gaudette L.
      • Gulbransen B.D.
      Communication between enteric neurons, glia, and nociceptors underlies the effects of tachykinins on neuroinflammation.
      proteases,
      • Garrido R.
      • Segura B.
      • Zhang W.
      • Mulholland M.
      Presence of functionally active protease-activated receptors 1 and 2 in myenteric glia.
      and TRPV4
      • Cenac N.
      • Bautzova T.
      • Faouder P.
      • Veldhuis N.A.
      • Poole D.P.
      • Rolland C.
      • Bertrand J.
      • Liedtke W.
      • Dubourdeau M.
      • Bertrand-Michel J.
      • Zecchi L.
      • Stanghellini V.
      • Bunnett N.W.
      • Barbara G.
      • Vergnolle N.
      Quantification and potential functions of endogenous agonists of transient receptor potential channels in patients with irritable bowel syndrome.
      agonists, all elicit responses in enteric glia. This raises the possibility that enteric glia contribute to nociceptor sensitization caused by these mediators. In support, perturbing enteric glial metabolism with fluorocitrate reduces visceromotor responses in mice.
      • Wang P.
      • Du C.
      • Chen F.-X.
      • Li C.-Q.
      • Yu Y.-B.
      • Han T.
      • Akhtar S.
      • Zuo X.-L.
      • Tan X.-D.
      • Li Y.-Q.
      BDNF contributes to IBS-like colonic hypersensitivity via activating the enteroglia-nerve unit.
      Together, these multiple lines of evidence suggest that crosstalk between enteric glia and nociceptors contributes to mechanisms that drive visceral hypersensitivity (Figure 2). The precise mechanisms by which enteric glia influence nociceptors are still being discovered. Yet new data discussed in the following sections are offering promising leads that could identify novel molecular targets for new abdominal pain therapies.
      Figure thumbnail gr2
      Figure 2Overview of potential enteric glial cell mechanisms that contribute to the sensitization of nociceptors. Highlighted are pathways that directly and indirectly sensitize nociceptors. Enteric glial activity can be influenced by several factors, such as inflammation, stress, neurotransmission, and/or the gut microbiota. Enteric glia release neuromodulators that act on primary afferent neurons by binding to receptors on nociceptor nerve endings or by sensitizing nociceptors. Enteric glia possess ectoenzymes that remove neuromodulators from the extracellular space. Enteric glia also have the potential to indirectly modulate nociceptive neurons by activating immune cells either through the release of immune cell regulators or by acting as antigen-presenting cells. GABA, γ-aminobutyric acid.

      Potential Mechanisms for Direct Modulation of Nociceptors by Enteric Glia

      Enteric glia play active roles in neurotransmission in the intestine by generating and regulating the availability of neurotransmitters. Many of the transmitter systems regulated by enteric glia are involved in the activation and sensitization of nociceptors. For example, enteric glia are a primary source of ATP, which contributes to cell-to-cell communication in health and disease and acts on P2X receptors expressed by nociceptors.
      • Cook S.
      • McCleskey E.
      Cell damage excites nociceptors through release of cytosolic ATP.
      • Brown I.A.
      • McClain J.L.
      • Watson R.E.
      • Patel B.A.
      • Gulbransen B.D.
      Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide.
      • Berta T.
      • Park C.-K.
      • Xu Z.-Z.
      • Xie R.-G.
      • Liu T.
      • Lü N.
      • Liu Y.-C.
      • Ji R.-R.
      Extracellular caspase-6 drives murine inflammatory pain via microglial TNF-α secretion.
      • Song D.
      • Li Y.
      • Tang D.
      • Huang L.
      • Yuan Y.
      Neuron-glial communication mediated by TNF-α and glial activation in dorsal root ganglia in visceral inflammatory hypersensitivity.
      Purines and purinergic signaling from glial cells are altered during inflammation and are strongly implicated in the process of visceral hypersensitivity and abdominal pain.
      • Xu G.-Y.
      • Shenoy M.
      • Winston J.H.
      • Mittal S.
      • Pasricha P.J.
      P2X receptor-mediated visceral hyperalgesia in a rat model of chronic visceral hypersensitivity.
      • Deiteren A.
      • van der Linden L.
      • de Wit A.
      • Ceuleers H.
      • Buckinx R.
      • Timmermans J.-P.
      • Moreels T.G.
      • Pelckmans P.A.
      • Man J.G.
      • Winter B.Y.
      P2x3 receptors mediate visceral hypersensitivity during acute chemically-induced colitis and in the post-inflammatory phase via different mechanisms of sensitization.
      Glial cells, including enteric glia, also possess ectoenzymes, such as NTPDase2, that are responsible for the degradation of ATP and other transmitters.
      • Brown I.A.
      • McClain J.L.
      • Watson R.E.
      • Patel B.A.
      • Gulbransen B.D.
      Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide.
      • Lavoie E.G.
      • Gulbransen B.D.
      • Martín-Satué M.
      • Aliagas E.
      • Sharkey K.A.
      • Sévigny J.
      Ectonucleotidases in the digestive system: focus on NTPDase3 localization.
      • Brisevac D.
      • Bajic A.
      • Bjelobaba I.
      • Milosevic M.
      • Stojiljkovic M.
      • Beyer C.
      • Clarner T.
      • Kipp M.
      • Nedeljkovic N.
      Expression of ecto-nucleoside triphosphate diphosphohydrolase1-3 (NTPDase1–3) by cortical astrocytes after exposure to pro-inflammatory factors in vitro.
      Astrocytic NTPDase2 expression is altered during inflammation and could contribute to the sensitization of sensory neurons by increasing purinergic signaling.
      • Brisevac D.
      • Bajic A.
      • Bjelobaba I.
      • Milosevic M.
      • Stojiljkovic M.
      • Beyer C.
      • Clarner T.
      • Kipp M.
      • Nedeljkovic N.
      Expression of ecto-nucleoside triphosphate diphosphohydrolase1-3 (NTPDase1–3) by cortical astrocytes after exposure to pro-inflammatory factors in vitro.
      The similarities between enteric glia and astrocytes suggest that a similar behavior could be observed in enteric glia. Glial ATP release is triggered by the excitation of enteric glial cells, intracellular calcium signaling, and the subsequent opening of Cx-43 hemichannels,
      • Brown I.A.
      • McClain J.L.
      • Watson R.E.
      • Patel B.A.
      • Gulbransen B.D.
      Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide.
      which is a molecular mechanism highly implicated in the development of nociceptive pain and inflammatory pain.
      • Brown I.A.
      • McClain J.L.
      • Watson R.E.
      • Patel B.A.
      • Gulbransen B.D.
      Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide.
      • Chen M.J.
      • Kress B.
      • Han X.
      • Moll K.
      • Peng W.
      • Ji R.
      • Nedergaard M.
      Astrocytic CX43 hemichannels and gap junctions play a crucial role in development of chronic neuropathic pain following spinal cord injury.
      • Tonkin R.S.
      • Bowles C.
      • Perera C.J.
      • Keating B.A.
      • Makker P.
      • Duffy S.S.
      • Lees J.G.
      • Tran C.
      • Don A.S.
      • Fath T.
      • Liu L.
      • O’Carroll S.J.
      • Nicholson L.
      • Green C.R.
      • Gorrie C.
      • Moalem-Taylor G.
      Attenuation of mechanical pain hypersensitivity by treatment with Peptide5, a connexin-43 mimetic peptide, involves inhibition of NLRP3 inflammasome in nerve-injured mice.
      In spinal astrocytes, Cx-43 expression is highly elevated following spinal cord injury and the inhibition of Cx-43 reduces allodynia and hyperalgesia.
      • Chen M.J.
      • Kress B.
      • Han X.
      • Moll K.
      • Peng W.
      • Ji R.
      • Nedergaard M.
      Astrocytic CX43 hemichannels and gap junctions play a crucial role in development of chronic neuropathic pain following spinal cord injury.
      • Tonkin R.S.
      • Bowles C.
      • Perera C.J.
      • Keating B.A.
      • Makker P.
      • Duffy S.S.
      • Lees J.G.
      • Tran C.
      • Don A.S.
      • Fath T.
      • Liu L.
      • O’Carroll S.J.
      • Nicholson L.
      • Green C.R.
      • Gorrie C.
      • Moalem-Taylor G.
      Attenuation of mechanical pain hypersensitivity by treatment with Peptide5, a connexin-43 mimetic peptide, involves inhibition of NLRP3 inflammasome in nerve-injured mice.
      • Choi S.-R.
      • Roh D.-H.
      • Yoon S.-Y.
      • Kwon S.-G.
      • Choi H.-S.
      • Han H.-J.
      • Beitz A.J.
      • Lee J.-H.
      Astrocyte sigma-1 receptors modulate connexin 43 expression leading to the induction of below-level mechanical allodynia in spinal cord injured mice.
      A direct link between Cx-43 on enteric glial cells and nociceptors sensitization has yet to emerge. However, because of the implications of astroglial Cx-43 in chronic pain and the analogous nature of enteric glia, it is likely that enteric Cx-43 is readily involved in the mechanisms of abdominal pain. In support, we recently showed that bidirectional communication between nociceptors and enteric glia involves glial Cx-43.
      • Delvalle N.
      • Dharshika C.
      • Morales-Soto W.
      • Fried D.E.
      • Gaudette L.
      • Gulbransen B.D.
      Communication between enteric neurons, glia, and nociceptors underlies the effects of tachykinins on neuroinflammation.
      Proinflammatory cytokines including IL-6, IL-1β, and TNF-α play major roles in the development of intestinal pathology and are known contributors to the development and maintenance of inflammatory and neuropathic pain.
      • Ji R.-R.
      • Chamessian A.
      • Zhang Y.-Q.
      Pain regulation by non-neuronal cells and inflammation.
      • Ji R.-R.R.
      • Berta T.
      • Nedergaard M.
      Glia and pain: is chronic pain a gliopathy?.
      These cytokines are increased during IBS and IBD.
      • Scully P.
      • McKernan D.P.
      • Keohane J.
      • Groeger D.
      • Shanahan F.
      • Dinan T.G.
      • Quigley E.M.
      Plasma cytokine profiles in females with irritable bowel syndrome and extra-intestinal co-morbidity.
      • Sawa Y.
      • Oshitani N.
      • Adachi K.
      • Higuchi K.
      • Matsumoto T.
      • Arakawa T.
      Comprehensive analysis of intestinal cytokine messenger RNA profile by real-time quantitative polymerase chain reaction in patients with inflammatory bowel disease.
      Astrocytes, microglia, and satellite glia produce proinflammatory cytokines, such as IL-1β, during inflammation and nerve injury.
      • White F.A.
      • Jung H.
      • Miller R.J.
      Chemokines and the pathophysiology of neuropathic pain.
      • Clark A.K.
      • Staniland A.A.
      • Marchand F.
      • Kaan T.K.
      • McMahon S.B.
      • Malcangio M.
      P2x7-dependent release of interleukin-1β and nociception in the spinal cord following lipopolysaccharide.
      • Zhang J.-M.
      • An J.
      Cytokines, inflammation, and pain.
      • Wen Y.-R.
      • Suter M.R.
      • Ji R.-R.
      • Yeh G.-C.
      • Wu Y.-S.
      • Wang K.-C.
      • Kohno T.
      • Sun W.-Z.
      • Wang C.-C.
      Activation of p38 mitogen-activated protein kinase in spinal microglia contributes to incision-induced mechanical allodynia.
      • Takeda M.
      • Takahashi M.
      • Matsumoto S.
      Contribution of the activation of satellite glia in sensory ganglia to pathological pain.
      • Bashashati M.
      • Moradi M.
      • Sarosiek I.
      Interleukin-6 in irritable bowel syndrome: a systematic review and meta-analysis of IL-6 (-G174C) and circulating IL-6 levels.
      Similarly, enteric glia produce, and respond to cytokines including IL-6 and IL-1β.
      • Tjwa E.
      • Bradley J.M.
      • Keenan C.M.
      • Kroese A.
      • Sharkey K.A.
      Interleukin-1β activates specific populations of enteric neurons and enteric glia in the guinea pig ileum and colon.
      • Rühl A.
      • Franzke S.
      • Collins Stremmel W.
      Interleukin-6 expression and regulation in rat enteric glial cells.
      • Takac B.
      • Mihaljević S.
      • Stefanić M.
      • Glavas-Obrovac L.
      • Kibel A.
      • Samardzija M.
      Importance of interleukin 6 in pathogenesis of inflammatory bowel disease.
      Experimental studies support a proinflammatory, pronociceptive role for IL-6 and the upregulation of IL-6 correlates with the development of hyperalgesia and allodynia in animal models.
      • DeLeo J.
      • Colburn R.
      • Nichols M.
      • Malhotra A.
      Interleukin-6-mediated hyperalgesia/allodynia and increased spinal IL-6 expression in a rat mononeuropathy model.
      • Okamoto K.
      • Martin D.P.
      • Schmelzer J.D.
      • Mitsui Y.
      • Low P.A.
      Pro- and anti-inflammatory cytokine gene expression in rat sciatic nerve chronic constriction injury model of neuropathic pain.
      • Murphy P.
      • Ramer M.
      • Borthwick L.
      • Gauldie J.
      • Richardson P.
      • Bisby M.
      Endogenous interleukin-6 contributes to hypersensitivity to cutaneous stimuli and changes in neuropeptides associated with chronic nerve constriction in mice.
      Blocking IL-6 production and activity reduces or alleviates nociceptor sensitization.
      • Ferreira S.H.
      • Lorenzetti B.B.
      • Poole S.
      Bradykinin initiates cytokine-mediated inflammatory hyperalgesia.
      • Xu X.-J.
      • Hao J.-X.
      • Andell-Jonsson S.
      • Poli V.
      • Bartfai T.
      • Wiesenfeld-Hallin Z.
      Nociceptive responses in interleukin-6-deficient mice to peripheral inflammation and peripheral nerve section.
      Similarly, IL-1β has pronociceptive effects and treatment with IL-1β in vivo causes increased visceromotor responses to colorectal distention in animal models of inflammatory pain.
      • Nozu T.
      • Miyagishi S.
      • Nozu R.
      • Takakusaki K.
      • Okumura T.
      Repeated water avoidance stress induces visceral hypersensitivity: role of interleukin-1, interleukin-6, and peripheral corticotropin-releasing factor.
      Enteric glia also interact with TNF-α signaling pathways and TNF-α is heavily implicated in the development of visceral hypersensitivity. Microglia and satellite glia increase their expression of TNF-α during nerve injury and inflammation
      • Berta T.
      • Park C.-K.
      • Xu Z.-Z.
      • Xie R.-G.
      • Liu T.
      • Lü N.
      • Liu Y.-C.
      • Ji R.-R.
      Extracellular caspase-6 drives murine inflammatory pain via microglial TNF-α secretion.
      • Song D.
      • Li Y.
      • Tang D.
      • Huang L.
      • Yuan Y.
      Neuron-glial communication mediated by TNF-α and glial activation in dorsal root ganglia in visceral inflammatory hypersensitivity.
      • Liu Y.
      • Zhou L.-J.
      • Wang J.
      • Li D.
      • Ren W.-J.
      • Peng J.
      • Wei X.
      • Xu T.
      • Xin W.-J.
      • Pang R.-P.
      • Li Y.-Y.
      • Qin Z.-H.
      • Murugan M.
      • Mattson M.P.
      • Wu L.-J.
      • Liu X.-G.
      TNF-α differentially regulates synaptic plasticity in the hippocampus and spinal cord by microglia-dependent mechanisms after peripheral nerve injury.
      and the excitation of DRG neurons in mouse colitis models stimulates TNF-α production.
      • Song D.
      • Li Y.
      • Tang D.
      • Huang L.
      • Yuan Y.
      Neuron-glial communication mediated by TNF-α and glial activation in dorsal root ganglia in visceral inflammatory hypersensitivity.
      The exogenous application of TNF-α increases DRG neuron activity and visceral pain responses in trinitrobenzene sulfonic acid colitis, which is reversed by the administration of anti-TNF-α.
      • Song D.
      • Li Y.
      • Tang D.
      • Huang L.
      • Yuan Y.
      Neuron-glial communication mediated by TNF-α and glial activation in dorsal root ganglia in visceral inflammatory hypersensitivity.
      Evidence regarding the production of TNF-α by enteric glia is still controversial.
      • Cabarrocas J.
      • Savidge T.C.
      • Liblau R.S.
      Role of enteric glial cells in inflammatory bowel disease.
      However, TNF-α does alter enteric glial cell behavior and exposure to TNF-α induces increased GFAP expression and calcium signaling in enteric glia.
      • von Boyen G.B.
      • Steinkamp M.
      • Reinshagen M.
      • Schäfer K.H.
      • Adler G.
      • Kirsch J.
      Proinflammatory cytokines increase glial fibrillary acidic protein expression in enteric glia.
      • Liñán-Rico A.
      • Turco F.
      • Ochoa-Cortes F.
      • Harzman A.
      • Needleman B.J.
      • Arsenescu R.
      • Abdel-Rasoul M.
      • Fadda P.
      • Grants I.
      • Whitaker E.
      • Cuomo R.
      • Christofi F.L.
      Molecular signaling and dysfunction of the human reactive enteric glial cell phenotype: implications for GI infection, IBD, POI, neurological, motility, and GI disorders.
      It is possible that these effects are mediated through modifications to Cx-43 hemichannels based on the known effects of TNF-α in spinal cord injury.
      • Kárpáti A.
      • Yoshikawa T.
      • Nakamura T.
      • Iida T.
      • Matsuzawa T.
      • Kitano H.
      • Harada R.
      • Yanai K.
      Histamine elicits glutamate release from cultured astrocytes.
      • Zhu J.
      • Qu C.
      • Lu X.
      • Zhang S.
      Activation of microglia by histamine and substance P.
      Neurotrophic factors, such as nerve growth factor, brain-derived neurotrophic factor (BDNF), and neurotrophin-3, are also associated with neuropathic pain and inflammatory processes.
      • Siniscalco D.
      • Giordano C.
      • Rossi F.
      • Maione S.
      • de Novellis V.
      Role of neurotrophins in neuropathic pain.
      Enteric glia are a significant source, and target of neurotrophins in the intestine and alterations in the expression of neurotrophins and their receptors are associated with changes in enteric glial activity.
      • Xia C.-M.
      • Gulick M.A.
      • Yu S.J.
      • Grider J.R.
      • Murthy K.S.
      • Kuemmerle J.F.
      • Akbarali H.I.
      • Qiao L.-Y.
      Up-regulation of brain-derived neurotrophic factor in primary afferent pathway regulates colon-to-bladder cross-sensitization in rat.
      • Liu W.
      • Yue W.
      • Wu R.
      Effects of diabetes on expression of glial fibrillary acidic protein and neurotrophins in rat colon.
      The upregulation of BDNF expression by DRG neurons contributes to visceral pain
      • Xia C.-M.
      • Gulick M.A.
      • Yu S.J.
      • Grider J.R.
      • Murthy K.S.
      • Kuemmerle J.F.
      • Akbarali H.I.
      • Qiao L.-Y.
      Up-regulation of brain-derived neurotrophic factor in primary afferent pathway regulates colon-to-bladder cross-sensitization in rat.
      and enteric glia in the colon of patients with IBS highly express the BDNF receptor TrkB.
      • Wang P.
      • Du C.
      • Chen F.-X.
      • Li C.-Q.
      • Yu Y.-B.
      • Han T.
      • Akhtar S.
      • Zuo X.-L.
      • Tan X.-D.
      • Li Y.-Q.
      BDNF contributes to IBS-like colonic hypersensitivity via activating the enteroglia-nerve unit.
      • Levanti M.B.
      • Esteban I.
      • Ciriaco E.
      • Pérez-Piñera P.
      • Cabo R.
      • García-Suarez O.
      • Pardo B.
      • Silos-Santiago I.
      • Cobo J.
      • Vega J.A.
      Enteric glial cells express full-length TrkB and depend on TrkB expression for normal development.
      In addition, recent data suggest that BDNF-induced colonic hypersensitivity involves reactive enteric glia because visceral pain responses and GFAP expression were reduced in BDNF knockout mice.
      • Wang P.
      • Du C.
      • Chen F.-X.
      • Li C.-Q.
      • Yu Y.-B.
      • Han T.
      • Akhtar S.
      • Zuo X.-L.
      • Tan X.-D.
      • Li Y.-Q.
      BDNF contributes to IBS-like colonic hypersensitivity via activating the enteroglia-nerve unit.
      Similarly, nerve growth factor secretion by cultured enteric glia and mRNA levels of nerve growth factor and its receptor TrkA are increased in the presence of lipopolysaccharide, TNF-α, and IL-1β.
      • Boyen V.G.
      • Steinkamp M.
      • Reinshagen M.
      • Schäfer K.-H.
      • Adler G.
      • Kirsch J.
      Nerve growth factor secretion in cultured enteric glia cells is modulated by proinflammatory cytokines.
      Given the multiple biologic roles of neurotrophins in the maintenance of neuronal survival and function,
      • Siniscalco D.
      • Giordano C.
      • Rossi F.
      • Maione S.
      • de Novellis V.
      Role of neurotrophins in neuropathic pain.
      changes in glial neurotrophin expression and/or signaling could contribute to the neuroplasticity involved in visceral hypersensitivity.
      The gut microbiome has emerged as an essential component of the brain-gut axis and alterations in the composition of the microbiome are observed in several GI and pain conditions.
      • Chichlowski M.
      • Rudolph C.
      Visceral pain and gastrointestinal microbiome.
      Altered fecal microbiota in patients with IBS may even predict severity.
      • Quigley E.
      The gut-brain axis and the microbiome: clues to pathophysiology and opportunities for novel management strategies in irritable bowel syndrome (IBS).
      The microbiota produce a range of neuroactive compounds, such as the neurotransmitters γ-aminobutyric acid, serotonin, norepinephrine, and dopamine,
      • Moloney R.D.
      • Johnson A.C.
      • O’Mahony S.M.
      • Dinan T.G.
      • Meerveld B.
      • Cryan J.F.
      Stress and the microbiota–gut–brain axis in visceral pain: relevance to irritable bowel syndrome.
      and other bacterial products, such as lipopolysaccharide
      • Ochoa-Cortes F.
      • Ramos-Lomas T.
      • Miranda-Morales M.
      • Spreadbury I.
      • Ibeakanma C.
      • Barajas-Lopez C.
      • Vanner S.
      Bacterial cell products signal to mouse colonic nociceptive dorsal root ganglia neurons.
      and formyl peptides.
      • Husebye E.
      • Hellström P.
      • Midtvedt T.
      Intestinal microflora stimulates myoelectric activity of rat small intestine by promoting cyclic initiation and aboral propagation of migrating myoelectric complex.
      This suggests that the gut microbiome could influence pain pathways, but mechanistic links between the gut microbiome, sensory nerves, and alterations in the brain-gut axis are still relatively undefined. New data suggest that some effects of the microbiome could be exerted through effects on enteric glial function and development.
      • Kabouridis P.S.
      • Lasrado R.
      • McCallum S.
      • Chng S.H.
      • Snippert H.J.
      • Clevers H.
      • Pettersson S.
      • Pachnis V.
      The gut microbiota keeps enteric glial cells on the move; prospective roles of the gut epithelium and immune system.
      The generation of the mucosal enteric glial cell network parallels the maturation of the gut microbiome
      • Nicholson J.K.
      • Holmes E.
      • Kinross J.
      • Burcelin R.
      • Gibson G.
      • Jia W.
      • Pettersson S.
      Host-gut microbiota metabolic interactions.
      • Kabouridis P.S.
      • Lasrado R.
      • McCallum S.
      • Chng S.
      • Snippert H.J.
      • Clevers H.
      • Pettersson S.
      • Pachnis V.
      Microbiota controls the homeostasis of glial cells in the gut lamina propria.
      and the impaired mucosal glial population in germ-free mice can be restored by introducing a normal gut microbiome.
      • Kabouridis P.S.
      • Lasrado R.
      • McCallum S.
      • Chng S.
      • Snippert H.J.
      • Clevers H.
      • Pettersson S.
      • Pachnis V.
      Microbiota controls the homeostasis of glial cells in the gut lamina propria.
      The lipopolysaccharide receptor, TLR4, is located on enteric glial cells and is increased in mice following exposure to stress.
      • Tramullas M.
      • Finger B.C.
      • Moloney R.D.
      • Golubeva A.V.
      • Moloney G.
      • Dinan T.G.
      • Cryan J.F.
      Toll-like receptor 4 regulates chronic stress-induced visceral pain in mice.
      • Xu S.
      • Qin B.
      • Shi A.
      • Zhao J.
      • Guo X.
      • Dong L.
      Oxytocin inhibited stress induced visceral hypersensitivity, enteric glial cells activation, and release of proinflammatory cytokines in maternal separated rats.
      Visceromotor responses in maternally separated mice can be suppressed with oxytocin, which also causes a downregulation of enteric glial cell activity and TLR4 expression. This is indicative of a TLR4-dependent mechanism of enteric glial cells in modulating visceral pain responses.
      • Xu S.
      • Qin B.
      • Shi A.
      • Zhao J.
      • Guo X.
      • Dong L.
      Oxytocin inhibited stress induced visceral hypersensitivity, enteric glial cells activation, and release of proinflammatory cytokines in maternal separated rats.
      • Chow A.K.
      • Gulbransen B.D.
      Potential roles of enteric glia in bridging neuroimmune communication in the gut.
      • Bhave S.
      • Gade A.
      • Kang M.
      • Hauser K.F.
      • Dewey W.L.
      • Akbarali H.I.
      Connexin-purinergic signaling in enteric glia mediates the prolonged effect of morphine on constipation.
      The gut microbiome also produces several gliomodulators, such as proteases that directly act on enteric glial cells and are implicated in the pathophysiology of visceral hypersensitivity.
      • Garrido R.
      • Segura B.
      • Zhang W.
      • Mulholland M.
      Presence of functionally active protease-activated receptors 1 and 2 in myenteric glia.
      • Moloney R.D.
      • Johnson A.C.
      • O’Mahony S.M.
      • Dinan T.G.
      • Meerveld B.
      • Cryan J.F.
      Stress and the microbiota–gut–brain axis in visceral pain: relevance to irritable bowel syndrome.
      • Ceuleers H.
      • Spaendonk H.
      • Hanning N.
      • Heirbaut J.
      • Lambeir A.-M.
      • Joossens J.
      • Augustyns K.
      • Man J.G.
      • Meester I.
      • Winter B.Y.
      Visceral hypersensitivity in inflammatory bowel diseases and irritable bowel syndrome: The role of proteases.
      However, the significance of interactions between the microbiome and glia in the generation of visceral pain is still unknown and requires additional mechanistic studies to unravel.

      Potential Mechanisms of Indirect Modulation of Nociceptors by Enteric Glia

      Inflammation is a key component in the development of chronic pain
      • Ji R.-R.
      • Chamessian A.
      • Zhang Y.-Q.
      Pain regulation by non-neuronal cells and inflammation.
      • Zhang J.-M.
      • An J.
      Cytokines, inflammation, and pain.
      and abnormalities in immune responses are considered one of the primary causes leading to the development of IBS.
      • Barbara G.
      • Cremon C.
      • Giorgio R.
      • Dothel G.
      • Zecchi L.
      • Bellacosa L.
      • Carini G.
      • Stanghellini V.
      • Corinaldesi R.
      Mechanisms underlying visceral hypersensitivity in irritable bowel syndrome.
      Approximately 10% of all cases of IBS are associated with a previous inflammatory insult, such as a bacterial infection.
      • El-Salhy M.
      Irritable bowel syndrome: diagnosis and pathogenesis.
      Immune cells play an important role in the sensitization of sensory nerve fibers during inflammation and contribute to the development of acute and chronic pain.
      • Ji R.-R.
      • Chamessian A.
      • Zhang Y.-Q.
      Pain regulation by non-neuronal cells and inflammation.
      Their active role in pain is primarily caused by the release of proinflammatory mediators that modulate ion channel expression in primary afferent fibers, such as TRPV1 receptors.
      • Franchi S.
      • Sacerdote P.
      • Panerai A.
      The prokineticin system: an interface between neural inflammation and pain.
      • Lis K.
      • Grygorowicz T.
      • Cudna A.
      • Szymkowski D.E.
      • Bałkowiec-Iskra E.
      Inhibition of TNF reduces mechanical orofacial hyperalgesia induced by Complete Freund’s Adjuvant by a TRPV1-dependent mechanism in mice.
      • Utsumi D.
      • Matsumoto K.
      • Tsukahara T.
      • Amagase K.
      • Tominaga M.
      • Kato S.
      Transient receptor potential vanilloid 1 and transient receptor potential ankyrin 1 contribute to the progression of colonic inflammation in dextran sulfate sodium-induced colitis in mice: Links to calcitonin gene-related peptide and substance P.
      • Spencer N.J.
      • Magnúsdóttir E.I.
      • Jakobsson J.
      • Kestell G.
      • Chen B.N.
      • Morris D.
      • Brookes S.J.
      • Lagerström M.C.
      CGRPα within the Trpv1-Cre population contributes to visceral nociception.
      Histamine, for example, activates nerve fibers and contributes to neurogenic inflammation and pain.
      • Rosa A.
      • Fantozzi R.
      The role of histamine in neurogenic inflammation.
      Blocking H1 histamine receptors expressed by primary sensory afferent neurons in mice reduces pain responses after cutaneous injection of formalin to the paw.
      • Mobarakeh J.
      • Torkaman-Boutorabi A.
      • Rahimi A.
      • Ghasri S.
      • Nezhad R.
      • Hamzely A.
      • Sima B.
      • Takahashi K.
      • Nunoki K.
      • Yanai K.
      Interaction of histamine and calcitonin gene-related peptide in the formalin induced pain perception in rats.
      Recently, the concept that both central and peripheral glial cells function as immunoregulatory cells has emerged.
      • Chow A.K.
      • Gulbransen B.D.
      Potential roles of enteric glia in bridging neuroimmune communication in the gut.
      Glial cell–mast cell communication is critical for the development of inflammation.
      • Skaper S.D.
      • Facci L.
      • Zusso M.
      • Giusti P.
      Neuroinflammation, mast cells, and glia: dangerous liaisons.
      • Skaper S.D.
      • Facci L.
      • Giusti P.
      Mast cells, glia and neuroinflammation: partners in crime?.
      Histamine clearance is an essential process for avoiding excessive histaminergic neuronal activation. Astrocytes play a prominent role in histamine clearance via histamine N-methyltransferase, a histamine metabolizing enzyme.
      • Yoshikawa T.
      • Naganuma F.
      • Iida T.
      • Nakamura T.
      • Harada R.
      • Mohsen A.S.
      • Kasajima A.
      • Sasano H.
      • Yanai K.
      Molecular mechanism of histamine clearance by primary human astrocytes.
      Astrocyte activity is modulated via histamine receptors on their cell membrane
      • Kárpáti A.
      • Yoshikawa T.
      • Nakamura T.
      • Iida T.
      • Matsuzawa T.
      • Kitano H.
      • Harada R.
      • Yanai K.
      Histamine elicits glutamate release from cultured astrocytes.
      and microglial activation through histamine results in the subsequent release of inflammatory mediators.
      • Zhu J.
      • Qu C.
      • Lu X.
      • Zhang S.
      Activation of microglia by histamine and substance P.
      Enteric glia exhibit the potential to function through similar pathways because their activity is also increased in the presence of histamine.
      • Kimball B.C.
      • Mulholland M.W.
      Enteric glia exhibit P2U receptors that increase cytosolic calcium by a phospholipase C-dependent mechanism.
      Current evidence supports the notion that enteric glia modify immune responses by the secretion of cytokines and other chemical transmitters and/or by acting as antigen-presenting cells. Glial proinflammatory mediators including S100B, ATP, and nitric oxide increase oxidative stress in the local environment and contribute to neuronal cell death and the release of apoptotic signaling that triggers immune responses.
      • Brown I.A.
      • McClain J.L.
      • Watson R.E.
      • Patel B.A.
      • Gulbransen B.D.
      Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide.
      • Liñán-Rico A.
      • Turco F.
      • Ochoa-Cortes F.
      • Harzman A.
      • Needleman B.J.
      • Arsenescu R.
      • Abdel-Rasoul M.
      • Fadda P.
      • Grants I.
      • Whitaker E.
      • Cuomo R.
      • Christofi F.L.
      Molecular signaling and dysfunction of the human reactive enteric glial cell phenotype: implications for GI infection, IBD, POI, neurological, motility, and GI disorders.
      • Turco F.
      • Sarnelli G.
      • Cirillo C.
      • Palumbo I.
      • Giorgi F.
      • D’Alessandro A.
      • Cammarota M.
      • Giuliano M.
      • Cuomo R.
      Enteroglial-derived S100B protein integrates bacteria-induced Toll-like receptor signalling in human enteric glial cells.
      • MacEachern S.J.
      • Patel B.A.
      • Keenan C.M.
      • Dicay M.
      • Chapman K.
      • McCafferty D.-M.
      • Savidge T.C.
      • Beck P.L.
      • MacNaughton W.K.
      • Sharkey K.A.
      Inhibiting inducible nitric oxide synthase in enteric glia restores electrogenic ion transport in mice with colitis.
      Enteric glia are also capable of expressing major histocompatibility complex II molecules and ligands necessary for T-cell signaling.
      • Turco F.
      • Sarnelli G.
      • Cirillo C.
      • Palumbo I.
      • Giorgi F.
      • D’Alessandro A.
      • Cammarota M.
      • Giuliano M.
      • Cuomo R.
      Enteroglial-derived S100B protein integrates bacteria-induced Toll-like receptor signalling in human enteric glial cells.
      • Geboes K.
      Histological classification of chronic gastritis: An iconoclastic view.
      • da Silveira A.
      • de Oliveira E.C.
      • Neto S.G.
      • Luquetti A.O.
      • Fujiwara R.
      • Oliveira R.
      • Brehmer A.
      Enteroglial cells act as antigen-presenting cells in chagasic megacolon.
      Several types of immune cells are involved in inflammatory pain, particularly in the persistence of hyperalgesia.
      • Ibiza S.
      • García-Cassani B.
      • Ribeiro H.
      • Carvalho T.
      • Almeida L.
      • Marques R.
      • Misic A.M.
      • Bartow-McKenney C.
      • Larson D.M.
      • Pavan W.J.
      • Eberl G.
      • Grice E.A.
      • Veiga-Fernandes H.
      Glial-cell-derived neuroregulators control type 3 innate lymphoid cells and gut defence.
      Immune cell activity can also contribute to neuropathic pain because of damage to peripheral nerves or to the central nervous system.
      • Ji R.-R.
      • Chamessian A.
      • Zhang Y.-Q.
      Pain regulation by non-neuronal cells and inflammation.
      Together these data show that enteric glia can modulate immune cells, which in turn can influence nociception.

      Potential Roles of Enteric Glia in Opioid-Induced Hyperalgesia

      Current treatments targeting chronic pain are limited and often involve the use of opioids.
      • Dunn K.
      • Saunders K.W.
      • Rutter C.M.
      • Banta-Green C.J.
      • Merrill J.O.
      • Sullivan M.D.
      • Weisner C.M.
      • Silverberg M.J.
      • Campbell C.I.
      • Psaty B.M.
      • Korff M.
      Opioid prescriptions for chronic pain and overdose: a cohort study.
      However, opioid use has serious side effects that include persistent constipation and antinociceptive tolerance that limit clinical efficacy. Opioid-induced hyperalgesia can result from chronic opioid use and produce an IBS-like syndrome termed “narcotic bowel syndrome.”
      • Chu L.F.
      • Angst M.S.
      • Clark D.
      Opioid-induced hyperalgesia in humans: molecular mechanisms and clinical considerations.
      • Grunkemeier D.M.
      • Cassara J.E.
      • Dalton C.B.
      • Drossman D.A.
      The narcotic bowel syndrome: clinical features, pathophysiology, and management.
      Narcotic bowel syndrome is a subset of opioid bowel dysfunction characterized by recurring abdominal pain that worsens with the continued or escalating use of narcotics.
      • Watkins L.R.
      • Hutchinson M.R.
      • Johnston I.N.
      • Maier S.F.
      Glia: novel counter-regulators of opioid analgesia.
      Glial cells play a role in modulating opioid behavior.
      • Watkins L.R.
      • Hutchinson M.R.
      • Johnston I.N.
      • Maier S.F.
      Glia: novel counter-regulators of opioid analgesia.
      • Rattan A.K.
      • Tejwani G.A.
      Effect of chronic treatment with morphine, midazolam and both together on dynorphin(1–13) levels in the rat.
      • Ji R.R.
      Targeting microglial purinergic signaling to improve morphine analgesia.
      Chronic opioid treatment causes glial release of nitric oxide, prostaglandins, excitatory amino acids, and growth factors. It also causes changes in purinergic signaling and increases the release of glial mediators that enhance the release of pain transmission.
      • Watkins L.R.
      • Hutchinson M.R.
      • Johnston I.N.
      • Maier S.F.
      Glia: novel counter-regulators of opioid analgesia.
      Opiates modulate central glial activity by directly binding to the glial (μ)-opioid receptor causing the release of proinflammatory cytokines or indirectly through the release of dynorphins.
      • Rattan A.K.
      • Tejwani G.A.
      Effect of chronic treatment with morphine, midazolam and both together on dynorphin(1–13) levels in the rat.
      Chronic exposure to opioids leads to robust astrocyte and microglia reactions and induces hyperalgesia.
      • Ji R.R.
      Targeting microglial purinergic signaling to improve morphine analgesia.
      Acute opioid treatment also produces marked satellite glia reaction in DRG.
      • Berta T.
      • Liu T.
      • Liu Y.C.
      • Xu Z.Z.
      • Ji R.R.
      Acute morphine activates satellite glial cells and up-regulates IL-1β in dorsal root ganglia in mice via matrix metalloprotease-9.
      The bulk of these effects result from altered glial purinergic signaling. Chronic opioid treatment leads to the upregulation of P2X4 and P2X7 receptors in spinal microglia and contributes to opioid tolerance.
      • Berta T.
      • Liu T.
      • Liu Y.C.
      • Xu Z.Z.
      • Ji R.R.
      Acute morphine activates satellite glial cells and up-regulates IL-1β in dorsal root ganglia in mice via matrix metalloprotease-9.
      • Horvath R.
      • DeLeo J.A.
      Morphine enhances microglial migration through modulation of P2X4 receptor signaling.
      • Zhou Z.
      • Peng X.
      • Hao S.
      • Fink D.J.
      • Mata M.
      HSV-mediated transfer of interleukin-10 reduces inflammatory pain through modulation of membrane tumor necrosis factor α in spinal cord microglia.
      Similar effects are observed in enteric glial cells. Long-term morphine treatment enhances purinergic activity in enteric glia that subsequently leads to barrier dysfunction and constipation through mechanisms that require glial Cx-43 activity.
      • Bhave S.
      • Gade A.
      • Kang M.
      • Hauser K.F.
      • Dewey W.L.
      • Akbarali H.I.
      Connexin-purinergic signaling in enteric glia mediates the prolonged effect of morphine on constipation.
      Opioid exposure upregulates proinflammatory cytokine release and contributes to morphine tolerance.
      • Hutchinson M.R.
      • Coats B.D.
      • Lewis S.S.
      • Zhang Y.
      • Sprunger D.B.
      • Rezvani N.
      • Baker E.M.
      • Jekich B.M.
      • Wieseler J.L.
      • Somogyi A.A.
      • Martin D.
      • Poole S.
      • Judd C.M.
      • Maier S.F.
      • Watkins L.R.
      Proinflammatory cytokines oppose opioid-induced acute and chronic analgesia.
      Chronic morphine exposure causes the release of TNF-α, IL-1β, and IL-6 from glial cells, all of which are known contributors to chronic pain.
      • Wei F.
      • Guo W.
      • Zou S.
      • Ren K.
      • Dubner R.
      Supraspinal glial–neuronal interactions contribute to descending pain facilitation.
      Chronic opioid exposure induces glial responses through TLR4 and reinforces the rewarding effects of opioids. Blocking TLR4 signaling attenuates the development of analgesic tolerance and hyperalgesia.
      • Hutchinson M.R.
      • Zhang Y.
      • Shridhar M.
      • Evans J.H.
      • Buchanan M.M.
      • Zhao T.X.
      • Slivka P.F.
      • Coats B.D.
      • Rezvani N.
      • Wieseler J.
      • Hughes T.S.
      • Landgraf K.E.
      • Chan S.
      • Fong S.
      • Phipps S.
      • Falke J.J.
      • Leinwand L.A.
      • Maier S.F.
      • Yin H.
      • Rice K.C.
      • Watkins L.R.
      Evidence that opioids may have toll-like receptor 4 and MD-2 effects.
      • Nicotra L.
      • Loram L.C.
      • Watkins L.R.
      • Hutchinson M.R.
      Toll-like receptors in chronic pain.
      Enteric glia express TLR receptors and TLR4-deficient mice fail to develop opioid-induced bowel disfunctions.
      • Beckett E.A.
      • Staikopoulos V.
      • Hutchinson M.R.
      Differential effect of morphine on gastrointestinal transit, colonic contractions and nerve-evoked relaxations in Toll-Like Receptor deficient mice.
      Although these findings implicate enteric glial cells as playing a major role in morphine-induced bowel disfunction and opioid tolerance, more in-depth studies are needed to clearly define the role of glia in opioid-induced hyperalgesia.

      Conclusions

      New therapies are needed to combat the common clinical issue of chronic abdominal pain. However, their development is limited primarily by the incomplete understanding of mechanisms that drive the development and maintenance visceral pain. Recent discoveries suggest that significant new insight into these mechanisms could be gained by focusing on glial cells. Glia are intertwined with mechanisms that produce nociceptive hyperalgesia and allodynia in pathologic conditions. Specifically, glia are responsive to neurotransmitters in pain circuits and produce, secrete, and regulate modulators that activate and sensitize nociceptors. Moreover, glial cells actively contribute to processes involved in neuroplasticity. The precise mechanisms whereby peripheral glia contribute to neuroplasticity in pain pathologies is still a developing area, but current discoveries suggest that these mechanisms are involved in the transition from acute to chronic pain.
      The contributions of glial cells to pain hypersensitivity are not centralized, but in fact require the involvement of several glial cell types along the brain-gut axis. Existing knowledge regarding enteric glia suggests that they are involved in visceral hypersensitivity and chronic pain. Enteric glia are primed for immune and stress responses. Increasing evidence supports their role in nociception, because they can influence nerve fiber activity either directly by releasing pronociceptive modulators or indirectly by interacting with the immune system. Their reactivity is also linked with altered ion channel signaling on nociceptor nerve endings. In addition to stress and inflammatory visceral hypersensitivity, enteric glia also show promise in playing a key role in the mechanisms of opioid-induced hyperalgesia and narcotic bowel syndrome.
      Enteric glia signaling involving Cx-43 is currently the most well-described glial mechanism that contributes to neuroinflammation and intercellular communication.
      • Brown I.A.
      • McClain J.L.
      • Watson R.E.
      • Patel B.A.
      • Gulbransen B.D.
      Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide.
      • McClain J.L.
      • Grubišić V.
      • Fried D.
      • Gomez-Suarez R.A.
      • Leinninger G.M.
      • Sévigny J.
      • Parpura V.
      • Gulbransen B.D.
      Ca2+ responses in enteric glia are mediated by connexin-43 hemichannels and modulate colonic transit in mice.
      • Grubišić V.
      • Gulbransen B.D.
      Enteric glial activity regulates secretomotor function in the mouse colon but does not acutely affect gut permeability.
      However, the therapeutic potential of this target is untested. Existing drugs that modulate Cx-43 are poorly selective and interact with connexin hemichannels and gap junctions in multiple tissues and cell types, which could potentially compromise normal gut function. This suggests that targeting mechanisms upstream or downstream of glial Cx-43 may prove more effective. In support, a clinical trial targeting downstream neuronal mechanisms with the P2X7 receptor antagonist AZD9056 showed significant promise in the treatment of visceral pain in patients with Crohn’s disease.
      • Eser A.
      • Colombel J.F.
      • Rutgeerts P.
      • Vogelsang H.
      • Braddock M.
      • Persson T.
      • Reinisch W.
      Safety and efficacy of an oral inhibitor of the purinergic receptor P2X7 in adult patients with moderately to severely active Crohn’s disease: a randomized placebo-controlled, double-blind, phase IIa study.
      Other potential targets on enteric glia to modulate pain are less defined but targeting enteric glial IL-1β and IL-6 signaling could be promising because both contribute to pain pathologies.
      • Okamoto K.
      • Martin D.P.
      • Schmelzer J.D.
      • Mitsui Y.
      • Low P.A.
      Pro- and anti-inflammatory cytokine gene expression in rat sciatic nerve chronic constriction injury model of neuropathic pain.
      • Xu X.-J.
      • Hao J.-X.
      • Andell-Jonsson S.
      • Poli V.
      • Bartfai T.
      • Wiesenfeld-Hallin Z.
      Nociceptive responses in interleukin-6-deficient mice to peripheral inflammation and peripheral nerve section.
      • Nozu T.
      • Miyagishi S.
      • Nozu R.
      • Takakusaki K.
      • Okumura T.
      Repeated water avoidance stress induces visceral hypersensitivity: role of interleukin-1, interleukin-6, and peripheral corticotropin-releasing factor.
      Similarly, modulating glial PAR and neurotrophin signaling may also serve to regulate glial activity and potentially reduce visceral hypersensitivity.
      • Esposito G.
      • Capoccia E.
      • Turco F.
      • Palumbo I.
      • Lu J.
      • Steardo A.
      • Cuomo R.
      • Sarnelli G.
      • Steardo L.
      Palmitoylethanolamide improves colon inflammation through an enteric glia/toll like receptor 4-dependent PPAR-α activation.
      • Qiao L.-Y.
      Neurotrophin signaling and visceral hypersensitivity.
      Further studies should focus on elucidating the role of enteric glial cells in chronic abdominal pain, because these cell types are essential in maintaining intestinal homeostasis and are highly active elements in GI disorders. In general, glia demonstrate great promise in filling the existing gaps in knowledge with regards to chronic abdominal pain and may become a considerable target for therapeutic approaches.

      Acknowledgments

      The manuscript was written by Wilmarie Morales-Soto and edited by Brian D. Gulbransen.

      References

        • Peery A.F.
        • Crockett S.D.
        • Barritt A.S.
        • Dellon E.S.
        • Eluri S.
        • Gangarosa L.M.
        • Jensen E.T.
        • Lund J.L.
        • Pasricha S.
        • Runge T.
        • Schmidt M.
        • Shaheen N.J.
        • Sandler R.S.
        Burden of gastrointestinal, liver, and pancreatic diseases in the United States.
        Gastroenterology. 2015; 149: 1731-1741
        • Mansfield K.E.
        • Sim J.
        • Jordan J.L.
        • Jordan K.P.
        A systematic review and meta-analysis of the prevalence of chronic widespread pain in the general population.
        Pain. 2016; 157: 55-64
        • Longstreth G.F.
        • Thompson G.W.
        • Chey W.D.
        • Houghton L.A.
        • Mearin F.
        • Spiller R.C.
        Functional bowel disorders.
        Gastroenterology. 2006; 130: 1480-1491
        • Regueiro M.
        • Greer J.B.
        • Szigethy E.
        Etiology and treatment of pain and psychosocial issues in patients with inflammatory bowel diseases.
        Gastroenterology. 2017; 152: 430-439
        • Keohane J.
        • O’Mahony C.
        • O’Mahony L.
        • O’Mahony S.
        • Quigley E.M.
        • Shanahan F.
        Irritable bowel syndrome–type symptoms in patients with inflammatory bowel disease: a real association or reflection of occult inflammation?.
        Am J Gastroenterol. 2010; 105: 1789-1794
        • Minderhoud I.M.
        • Oldenburg B.
        • Wismeijer J.A.
        • Henegouwen G.P.
        • Smout A.
        IBS-like symptoms in patients with inflammatory bowel disease in remission; relationships with quality of life and coping behavior.
        Dig Dis Sci. 2004; 49: 469-474
        • Halpin S.J.
        • Ford A.C.
        Prevalence of symptoms meeting criteria for irritable bowel syndrome in inflammatory bowel disease: systematic review and meta-analysis.
        Am J Gastroenterol. 2012; 107: 1474-1482
        • Colombel J.-F.
        • Mahadevan U.
        Inflammatory bowel disease 2017: innovations and changing paradigms.
        Gastroenterology. 2017; 152: 309-312
        • Camilleri M.
        • Lembo A.
        • Katzka D.A.
        Opioids in gastroenterology: treating adverse effects and creating therapeutic benefits.
        Clin Gastroenterol Hepatol. 2017; 15: 1338-1349
        • Ji R.-R.
        • Chamessian A.
        • Zhang Y.-Q.
        Pain regulation by non-neuronal cells and inflammation.
        Science. 2016; 354: 572-577
        • Mayer E.A.
        • Tillisch K.
        The brain-gut axis in abdominal pain syndromes.
        Annu Rev Med. 2011; 62: 381-396
        • Gold M.S.
        • Gebhart G.F.
        Nociceptor sensitization in pain pathogenesis.
        Nat Med. 2010; 16: 1248-1257
        • Cervero F.
        • Laird J.
        Understanding the signaling and transmission of visceral nociceptive events.
        J Neurobiol. 2004; 61: 45-54
        • Basbaum A.I.
        • Bautista D.M.
        • Scherrer G.
        • Julius D.
        Cellular and molecular mechanisms of pain.
        Cell. 2009; 139: 267-284
        • Ji R.-R.R.
        • Berta T.
        • Nedergaard M.
        Glia and pain: is chronic pain a gliopathy?.
        Pain. 2013; 154: S10-S28
        • Simrén M.
        • Törnblom H.
        • Palsson O.S.
        • van Tilburg M.A.
        • Oudenhove L.
        • Tack J.
        • Whitehead W.E.
        Visceral hypersensitivity is associated with GI symptom severity in functional GI disorders: consistent findings from five different patient cohorts.
        Gut. 2018; 67: 255-262
        • Stein C.
        • Machelska H.
        Modulation of peripheral sensory neurons by the immune system: implications for pain therapy.
        Pharmacol Rev. 2011; 63: 860-881
        • Cook S.
        • McCleskey E.
        Cell damage excites nociceptors through release of cytosolic ATP.
        Pain. 2002; 95: 41-47
        • Wouters M.M.
        • Balemans D.
        • Wanrooy S.
        • Dooley J.
        • Cibert-Goton V.
        • Alpizar Y.A.
        • Valdez-Morales E.E.
        • Nasser Y.
        • Veldhoven P.P.
        • Vanbrabant W.
        • der Merwe S.
        • Mols R.
        • Ghesquière B.
        • Cirillo C.
        • Kortekaas I.
        • Carmeliet P.
        • Peetermans W.E.
        • Vermeire S.
        • Rutgeerts P.
        • Augustijns P.
        • Hellings P.W.
        • Belmans A.
        • Vanner S.
        • Bulmer D.C.
        • Talavera K.
        • Berghe P.
        • Liston A.
        • Boeckxstaens G.E.
        Histamine receptor H1–mediated sensitization of TRPV1 mediates visceral hypersensitivity and symptoms in patients with irritable bowel syndrome.
        Gastroenterology. 2016; 150: 875-887
        • Binshtok A.M.
        • Wang H.
        • Zimmermann K.
        • Amaya F.
        • Vardeh D.
        • Shi L.
        • Brenner G.J.
        • Ji R.-R.
        • Bean B.P.
        • Woolf C.J.
        • Samad T.A.
        Nociceptors are interleukin-1β sensors.
        J Neurosci. 2008; 28: 14062-14073
        • Amadesi S.
        • Cottrell G.S.
        • Divino L.
        • Chapman K.
        • Grady E.F.
        • Bautista F.
        • Karanjia R.
        • Barajas-Lopez C.
        • Vanner S.
        • Vergnolle N.
        • Bunnett N.W.
        Protease-activated receptor 2 sensitizes TRPV1 by protein kinase Cɛ- and A-dependent mechanisms in rats and mice.
        J Physiol. 2006; 575: 555-571
        • Bandell M.
        • Story G.M.
        • Hwang S.
        • Viswanath V.
        • Eid S.R.
        • Petrus M.J.
        • Earley T.J.
        • Patapoutian A.
        Noxious cold ion channel TRPA1 is activated by pungent compounds and bradykinin.
        Neuron. 2004; 41: 849-857
        • Obreja O.
        • Biasio W.
        • Andratsch M.
        • Lips K.
        • Rathee P.
        • Ludwig A.
        • Rose-John S.
        • Kress M.
        Fast modulation of heat-activated ionic current by proinflammatory interleukin 6 in rat sensory neurons.
        Brain. 2005; 128: 1634-1641
        • Czeschik J.
        • Hagenacker T.
        • Schäfers M.
        • Büsselberg D.
        TNF-α differentially modulates ion channels of nociceptive neurons.
        Neurosci Lett. 2008; 434: 293-298
        • Azpiroz F.
        • Bouin M.
        • Camilleri M.
        • Mayer E.
        • Poitras P.
        • Serra J.
        • Spiller R.
        Mechanisms of hypersensitivity in IBS and functional disorders.
        Neurogastroenterol Motil. 2007; 19: 62-88
        • Sengupta J.N.
        Sensory nerves.
        Handb Exp Pharmacol. 2009; 194: 31-74
        • Li J.
        • McRoberts J.A.
        • Ennes H.S.
        • Trevisani M.
        • Nicoletti P.
        • Mittal Y.
        • Mayer E.A.
        Experimental colitis modulates the functional properties of NMDA receptors in dorsal root ganglia neurons.
        Am J Physiol Gastrointest Liver Physiol. 2006; 291: G219-G228
        • Sung B.
        • Lim G.
        • Mao J.
        Altered expression and uptake activity of spinal glutamate transporters after nerve injury contribute to the pathogenesis of neuropathic pain in rats.
        J Neurosci. 2003; 23: 2899-2910
        • Jones C.R.
        • Xu L.
        • Gebhart G.
        The mechanosensitivity of mouse colon afferent fibers and their sensitization by inflammatory mediators require transient receptor potential vanilloid 1 and acid-sensing ion channel 3.
        J Neurosci. 2005; 25: 10981-10989
        • Chan C.
        • Facer P.
        • Davis J.
        • Smith G.
        • Egerton J.
        • Bountra C.
        • Williams N.
        • Anand P.
        Sensory fibers expressing capsaicin receptor TRPV1 in patients with rectal hypersensitivity and fecal urgency.
        Lancet. 2003; 361: 385-391
        • Akbar A.
        • Yiangou Y.
        • Facer P.
        • Walters J.
        • Anand P.
        • Ghosh S.
        Increased capsaicin receptor TRPV1-expressing sensory fibers in irritable bowel syndrome and their correlation with abdominal pain.
        Gut. 2008; 57: 923-929
        • Larauche M.
        • Mulak A.
        • Taché Y.
        Stress-related alterations of visceral sensation: animal models for irritable bowel syndrome study.
        J Neurogastroenterol Motil. 2011; 17: 213-234
        • Ren T.-H.
        • Wu J.
        • Yew D.
        • Ziea E.
        • Lao L.
        • Leung W.-K.
        • Berman B.
        • Hu P.-J.
        • Sung J.J.
        Effects of neonatal maternal separation on neurochemical and sensory response to colonic distension in a rat model of irritable bowel syndrome.
        Am J Physiol Gastrointest Liver Physiol. 2006; 292: G849-G856
        • Tramullas M.
        • Finger B.C.
        • Moloney R.D.
        • Golubeva A.V.
        • Moloney G.
        • Dinan T.G.
        • Cryan J.F.
        Toll-like receptor 4 regulates chronic stress-induced visceral pain in mice.
        Biol Psychiatry. 2014; 76: 340-348
        • Elsenbruch S.
        • Rosenberger C.
        • Bingel U.
        • Forsting M.
        • Schedlowski M.
        • Gizewski E.R.
        Patients with irritable bowel syndrome have altered emotional modulation of neural responses to visceral stimuli.
        Gastroenterology. 2010; 139: 1310-1319
        • Drewes A.
        • Frkjær J.
        • Larsen E.
        • Reddy H.
        • Arendt-Nielsen L.
        • Gregersen H.
        Pain and mechanical properties of the rectum in patients with active ulcerative colitis.
        Inflamm Bowel Dis. 2006; 12: 294-303
        • Bielefeldt K.
        • Davis B.
        • Binion D.G.
        Pain and inflammatory bowel disease.
        Inflamm Bowel Dis. 2009; 15: 778-788
        • Tillisch K.
        • Mayer E.A.
        • Labus J.S.
        Quantitative meta-analysis identifies brain regions activated during rectal distension in irritable bowel syndrome.
        Gastroenterology. 2011; 140: 91-100
        • Seminowicz D.A.
        • Labus J.S.
        • Bueller J.A.
        • Tillisch K.
        • Naliboff B.D.
        • Bushnell C.M.
        • Mayer E.A.
        Regional gray matter density changes in brains of patients with irritable bowel syndrome.
        Gastroenterology. 2010; 139: 48-57
        • Larauche M.
        • Bradesi S.
        • Million M.
        • McLean P.
        • Taché Y.
        • Mayer E.A.
        • McRoberts J.A.
        Corticotropin-releasing factor type 1 receptors mediate the visceral hyperalgesia induced by repeated psychological stress in rats.
        Am J Physiol Gastrointest Liver Physiol. 2008; 294: G1033-G1040
        • La J.-H.
        Peripheral corticotropin releasing hormone mediates post-inflammatory visceral hypersensitivity in rats.
        World J Gastroenterol. 2008; 14: 731-736
        • Cairns B.E.
        • Arendt-Nielsen L.
        • Sacerdote P.
        Perspectives in pain research 2014: neuroinflammation and glial cell activation: the cause of transition from acute to chronic pain?.
        Scand J Pain. 2015; 6: 3-6
        • Stuesse S.L.
        • Crisp T.
        • McBurney D.L.
        • Schechter J.B.
        • Lovell J.A.
        • Cruce W.L.R.
        Neuropathic pain in aged rats: behavioral responses and astrocytic activation.
        Exp Brain Res. 2001; 137: 219-227
        • Coyle D.E.
        Partial peripheral nerve injury leads to activation of astroglia and microglia which parallels the development of allodynic behavior.
        Glia. 1998; 23: 75-83
        • Sweitzer S.M.
        • Colburn R.W.
        • Rutkowski M.
        • DeLeo J.A.
        Acute peripheral inflammation induces moderate glial activation and spinal IL-1β expression that correlates with pain behavior in the rat.
        Brain Res. 1999; 829: 209-221
        • Tanga F.
        • Raghavendra V.
        • DeLeo J.
        Quantitative real-time RT-PCR assessment of spinal microglial and astrocytic activation markers in a rat model of neuropathic pain.
        Neurochem Int. 2004; 45: 397-407
        • Hanani M.
        Role of satellite glial cells in gastrointestinal pain.
        Front Cell Neurosci. 2015; 9: 412
        • Brown I.A.
        • Gulbransen B.D.
        The antioxidant glutathione protects against enteric neuron death in situ, but its depletion is protective during colitis.
        Am J Physiol Gastrointest Liver Physiol. 2018; 314: G39-G52
        • Grubišić V.
        • Gulbransen B.D.
        Enteric glia: the most alimentary of all glia.
        J Physiol. 2017; 595: 557-570
        • McClain J.L.
        • Fried D.E.
        • Gulbransen B.D.
        Agonist-evoked Ca2+ signaling in enteric glia drives neural programs that regulate intestinal motility in mice.
        Cell Mol Gastroenterol Hepatol. 2015; 1: 631-645
        • Brown I.A.
        • McClain J.L.
        • Watson R.E.
        • Patel B.A.
        • Gulbransen B.D.
        Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide.
        Cell Mol Gastroenterol Hepatol. 2016; 2: 77-91
        • McClain J.L.
        • Grubišić V.
        • Fried D.
        • Gomez-Suarez R.A.
        • Leinninger G.M.
        • Sévigny J.
        • Parpura V.
        • Gulbransen B.D.
        Ca2+ responses in enteric glia are mediated by connexin-43 hemichannels and modulate colonic transit in mice.
        Gastroenterology. 2014; 146: 497-507
        • Grubišić V.
        • Gulbransen B.D.
        Enteric glial activity regulates secretomotor function in the mouse colon but does not acutely affect gut permeability.
        J Physiol. 2017; 595: 3409-3424
        • Gosselin R.-D.
        • Suter M.R.
        • Ji R.-R.
        • Decosterd I.
        Glial cells and chronic pain.
        Neuroscientist. 2010; 16: 519-531
        • Guo W.
        • Wang H.
        • Watanabe M.
        • Shimizu K.
        • Zou S.
        • LaGraize S.C.
        • Wei F.
        • Dubner R.
        • Ren K.
        Glial–cytokine–neuronal interactions underlying the mechanisms of persistent pain.
        J Neurosci. 2007; 27: 6006-6018
        • Romero-Sandoval A.
        • Chai N.
        • Nutile-McMenemy N.
        • DeLeo J.A.
        A comparison of spinal Iba1 and GFAP expression in rodent models of acute and chronic pain.
        Brain Res. 2008; 1219: 116-126
        • Nam Y.
        • Kim J.-H.
        • Kim J.-H.
        • Jha M.
        • Jung J.
        • Lee M.-G.
        • Choi I.-S.
        • Jang I.-S.
        • Lim D.
        • Hwang S.-H.
        • Cho H.-J.
        • Suk K.
        Reversible induction of pain hypersensitivity following optogenetic stimulation of spinal astrocytes.
        Cell Rep. 2016; 17: 3049-3061
        • Cho W.-H.
        • Barcelon E.
        • Lee S.
        Optogenetic glia manipulation: possibilities and future prospects.
        Exp Neurobiol. 2016; 25: 197-204
        • Zhong Y.
        • Zhou L.-J.
        • Ren W.-J.
        • Xin W.-J.
        • Li Y.-Y.
        • Zhang T.
        • Liu X.-G.
        The direction of synaptic plasticity mediated by C-fibers in spinal dorsal horn is decided by Src-family kinases in microglia: the role of tumor necrosis factor-α.
        Brain Behav Immun. 2010; 24: 874-880
        • Gao Y.
        • Zhang L.
        • Glia J.R.
        Spinal injection of TNF-α-activated astrocytes produces persistent pain symptom mechanical allodynia by releasing monocyte chemoattractant protein-1.
        Glia. 2010; 58: 1871-1880
        • Sun Y.-N.
        • Luo J.-Y.
        • Rao Z.-R.
        • Lan L.
        • Duan L.
        GFAP and Fos immunoreactivity in lumbo-sacral spinal cord and medulla oblongata after chronic colonic inflammation in rats.
        World J Gastroenterol. 2005; 11: 4827-4832
        • Zhao P.
        • Waxman S.G.
        • Hains B.C.
        Modulation of thalamic nociceptive processing after spinal cord injury through remote activation of thalamic microglia by cysteine–cysteine chemokine ligand 21.
        J Neurosci. 2007; 27: 8893-8902
        • Rajkowska G.
        • Miguel-Hidalgo J.
        Gliogenesis and glial pathology in depression.
        CNS Neurol Disord Drug Targets. 2007; 6: 219-233
        • Hanani M.
        Satellite glial cells in sympathetic and parasympathetic ganglia: In search of function.
        Brain Res Rev. 2010; 64: 304-327
        • Gulbransen B.D.
        Do nerves make bowels irritable?.
        Am J Physiol Gastrointest Liver Physiol. 2018; 315: G126-G127
        • Jessen K.
        • Mirsky R.
        Astrocyte-like glia in the peripheral nervous system: an immunohistochemical study of enteric glia.
        J Neurosci. 1983; 3: 2206-2218
        • Cocchia D.
        • Michetti F.
        • Ferri G.-L.
        • Polak J.M.
        • Probert L.
        • Marangos P.J.
        Evidence for the presence of S-100 protein in the glial component of the human enteric nervous system.
        Nature. 1982; 297: 409-410
        • Gulbransen B.D.
        • Sharkey K.A.
        Novel functional roles for enteric glia in the gastrointestinal tract.
        Nat Rev Gastroenterol Hepatol. 2012; 9: 625-632
        • von Boyen G.B.
        • Steinkamp M.
        • Reinshagen M.
        • Schäfer K.H.
        • Adler G.
        • Kirsch J.
        Proinflammatory cytokines increase glial fibrillary acidic protein expression in enteric glia.
        Gut. 2004; 53: 222-228
        • Chen M.J.
        • Kress B.
        • Han X.
        • Moll K.
        • Peng W.
        • Ji R.
        • Nedergaard M.
        Astrocytic CX43 hemichannels and gap junctions play a crucial role in development of chronic neuropathic pain following spinal cord injury.
        Glia. 2012; 60: 1660-1670
        • Fried D.E.
        • Watson R.E.
        • Robson S.C.
        • Gulbransen B.D.
        Ammonia modifies enteric neuromuscular transmission through glial γ-aminobutyric acid signaling.
        Am J Physiol Gastrointest Liver Physiol. 2017; 313: G570-G580
        • Gulbransen B.D.
        • Bashashati M.
        • Hirota S.A.
        • Gui X.
        • Roberts J.A.
        • MacDonald J.A.
        • Muruve D.A.
        • McKay D.M.
        • Beck P.L.
        • Mawe G.M.
        Activation of neuronal P2X7 receptor–pannexin-1 mediates death of enteric neurons during colitis.
        Nat Med. 2012; 18: 600-604
        • Delvalle N.
        • Dharshika C.
        • Morales-Soto W.
        • Fried D.E.
        • Gaudette L.
        • Gulbransen B.D.
        Communication between enteric neurons, glia, and nociceptors underlies the effects of tachykinins on neuroinflammation.
        Cell Mol Gastroenterol Hepatol. 2018; 6: 321-344
        • Fujikawa Y.
        • Tominaga K.
        • Tanaka F.
        • Tanigawa T.
        • Watanabe T.
        • Fujiwara Y.
        • Arakawa T.
        Enteric glial cells are associated with stress-induced colonic hyper-contraction in maternally separated rats.
        Neurogastroenterol Motil. 2015; 27: 1010-1023
        • Gulbransen B.D.
        • Bains J.S.
        • Sharkey K.A.
        Enteric glia are targets of the sympathetic innervation of the myenteric plexus in the guinea pig distal colon.
        J Neurosci. 2010; 30: 6801-6809
        • Cabarrocas J.
        • Savidge T.C.
        • Liblau R.S.
        Role of enteric glial cells in inflammatory bowel disease.
        Glia. 2003; 41: 81-93
        • Tjwa E.
        • Bradley J.M.
        • Keenan C.M.
        • Kroese A.
        • Sharkey K.A.
        Interleukin-1β activates specific populations of enteric neurons and enteric glia in the guinea pig ileum and colon.
        Am J Physiol Gastrointest Liver Physiol. 2003; 285: G1268-G1276
        • Rühl A.
        • Franzke S.
        • Collins Stremmel W.
        Interleukin-6 expression and regulation in rat enteric glial cells.
        Am J Physiol Gastrointest Liver Physiol. 2001; 280: G1163-G1171
        • Garrido R.
        • Segura B.
        • Zhang W.
        • Mulholland M.
        Presence of functionally active protease-activated receptors 1 and 2 in myenteric glia.
        J Neurochem. 2002; 83: 556-564
        • Cenac N.
        • Bautzova T.
        • Faouder P.
        • Veldhuis N.A.
        • Poole D.P.
        • Rolland C.
        • Bertrand J.
        • Liedtke W.
        • Dubourdeau M.
        • Bertrand-Michel J.
        • Zecchi L.
        • Stanghellini V.
        • Bunnett N.W.
        • Barbara G.
        • Vergnolle N.
        Quantification and potential functions of endogenous agonists of transient receptor potential channels in patients with irritable bowel syndrome.
        Gastroenterology. 2015; 149: 433-444
        • Wang P.
        • Du C.
        • Chen F.-X.
        • Li C.-Q.
        • Yu Y.-B.
        • Han T.
        • Akhtar S.
        • Zuo X.-L.
        • Tan X.-D.
        • Li Y.-Q.
        BDNF contributes to IBS-like colonic hypersensitivity via activating the enteroglia-nerve unit.
        Sci Rep. 2016; 6: 20320
        • Berta T.
        • Park C.-K.
        • Xu Z.-Z.
        • Xie R.-G.
        • Liu T.
        • Lü N.
        • Liu Y.-C.
        • Ji R.-R.
        Extracellular caspase-6 drives murine inflammatory pain via microglial TNF-α secretion.
        J Clin Invest. 2014; 124: 1173-1186
        • Song D.
        • Li Y.
        • Tang D.
        • Huang L.
        • Yuan Y.
        Neuron-glial communication mediated by TNF-α and glial activation in dorsal root ganglia in visceral inflammatory hypersensitivity.
        Am J Physiol Gastrointest Liver Physiol. 2014; 306: G788-G795
        • Xu G.-Y.
        • Shenoy M.
        • Winston J.H.
        • Mittal S.
        • Pasricha P.J.
        P2X receptor-mediated visceral hyperalgesia in a rat model of chronic visceral hypersensitivity.
        Gut. 2008; 57: 1230-1237
        • Deiteren A.
        • van der Linden L.
        • de Wit A.
        • Ceuleers H.
        • Buckinx R.
        • Timmermans J.-P.
        • Moreels T.G.
        • Pelckmans P.A.
        • Man J.G.
        • Winter B.Y.
        P2x3 receptors mediate visceral hypersensitivity during acute chemically-induced colitis and in the post-inflammatory phase via different mechanisms of sensitization.
        PloS One. 2015; 10: e0123810
        • Lavoie E.G.
        • Gulbransen B.D.
        • Martín-Satué M.
        • Aliagas E.
        • Sharkey K.A.
        • Sévigny J.
        Ectonucleotidases in the digestive system: focus on NTPDase3 localization.
        Am J Physiol Gastrointest Liver Physiol. 2011; 300: G608-G620
        • Brisevac D.
        • Bajic A.
        • Bjelobaba I.
        • Milosevic M.
        • Stojiljkovic M.
        • Beyer C.
        • Clarner T.
        • Kipp M.
        • Nedeljkovic N.
        Expression of ecto-nucleoside triphosphate diphosphohydrolase1-3 (NTPDase1–3) by cortical astrocytes after exposure to pro-inflammatory factors in vitro.
        J Mol Neurosci. 2013; 51: 871-879
        • Tonkin R.S.
        • Bowles C.
        • Perera C.J.
        • Keating B.A.
        • Makker P.
        • Duffy S.S.
        • Lees J.G.
        • Tran C.
        • Don A.S.
        • Fath T.
        • Liu L.
        • O’Carroll S.J.
        • Nicholson L.
        • Green C.R.
        • Gorrie C.
        • Moalem-Taylor G.
        Attenuation of mechanical pain hypersensitivity by treatment with Peptide5, a connexin-43 mimetic peptide, involves inhibition of NLRP3 inflammasome in nerve-injured mice.
        Exp Neurol. 2018; 300: 1-12
        • Choi S.-R.
        • Roh D.-H.
        • Yoon S.-Y.
        • Kwon S.-G.
        • Choi H.-S.
        • Han H.-J.
        • Beitz A.J.
        • Lee J.-H.
        Astrocyte sigma-1 receptors modulate connexin 43 expression leading to the induction of below-level mechanical allodynia in spinal cord injured mice.
        Neuropharmacology. 2016; 111: 34-46
        • Scully P.
        • McKernan D.P.
        • Keohane J.
        • Groeger D.
        • Shanahan F.
        • Dinan T.G.
        • Quigley E.M.
        Plasma cytokine profiles in females with irritable bowel syndrome and extra-intestinal co-morbidity.
        Am J Gastroenterol. 2010; 105: 2235-2243
        • Sawa Y.
        • Oshitani N.
        • Adachi K.
        • Higuchi K.
        • Matsumoto T.
        • Arakawa T.
        Comprehensive analysis of intestinal cytokine messenger RNA profile by real-time quantitative polymerase chain reaction in patients with inflammatory bowel disease.
        Int J Mol Med. 2003; 11: 175-179
        • White F.A.
        • Jung H.
        • Miller R.J.
        Chemokines and the pathophysiology of neuropathic pain.
        Proc Natl Acad Sci. 2007; 104: 20151-20158
        • Clark A.K.
        • Staniland A.A.
        • Marchand F.
        • Kaan T.K.
        • McMahon S.B.
        • Malcangio M.
        P2x7-dependent release of interleukin-1β and nociception in the spinal cord following lipopolysaccharide.
        J Neurosci. 2010; 30: 573-582
        • Zhang J.-M.
        • An J.
        Cytokines, inflammation, and pain.
        Int Anesthesiol Clin. 2007; 45: 27-37
        • Wen Y.-R.
        • Suter M.R.
        • Ji R.-R.
        • Yeh G.-C.
        • Wu Y.-S.
        • Wang K.-C.
        • Kohno T.
        • Sun W.-Z.
        • Wang C.-C.
        Activation of p38 mitogen-activated protein kinase in spinal microglia contributes to incision-induced mechanical allodynia.
        Anesthesiology. 2009; 110: 155-165
        • Takeda M.
        • Takahashi M.
        • Matsumoto S.
        Contribution of the activation of satellite glia in sensory ganglia to pathological pain.
        Neurosci Biobehav Rev. 2009; 33: 784-792
        • Bashashati M.
        • Moradi M.
        • Sarosiek I.
        Interleukin-6 in irritable bowel syndrome: a systematic review and meta-analysis of IL-6 (-G174C) and circulating IL-6 levels.
        Cytokine. 2017; 99: 132-138
        • Takac B.
        • Mihaljević S.
        • Stefanić M.
        • Glavas-Obrovac L.
        • Kibel A.
        • Samardzija M.
        Importance of interleukin 6 in pathogenesis of inflammatory bowel disease.
        Coll Antropol. 2014; 38: 659-664
        • DeLeo J.
        • Colburn R.
        • Nichols M.
        • Malhotra A.
        Interleukin-6-mediated hyperalgesia/allodynia and increased spinal IL-6 expression in a rat mononeuropathy model.
        J Interferon Cytokine Res. 1996; 16: 695-700
        • Okamoto K.
        • Martin D.P.
        • Schmelzer J.D.
        • Mitsui Y.
        • Low P.A.
        Pro- and anti-inflammatory cytokine gene expression in rat sciatic nerve chronic constriction injury model of neuropathic pain.
        Exp Neurol. 2001; 169: 386-391
        • Murphy P.
        • Ramer M.
        • Borthwick L.
        • Gauldie J.
        • Richardson P.
        • Bisby M.
        Endogenous interleukin-6 contributes to hypersensitivity to cutaneous stimuli and changes in neuropeptides associated with chronic nerve constriction in mice.
        Eur J Neurosci. 1999; 11: 2243-2253
        • Ferreira S.H.
        • Lorenzetti B.B.
        • Poole S.
        Bradykinin initiates cytokine-mediated inflammatory hyperalgesia.
        Br J Ph