Skip to main content
Gut logoLink to Gut
. 2006 Apr;55(4):445–447. doi: 10.1136/gut.2005.079046

Histamine, mast cells, and the enteric nervous system in the irritable bowel syndrome, enteritis, and food allergies

J D Wood
PMCID: PMC1856149  PMID: 16531524

Short abstract

There is altered expression of histamine H1 and H2 receptor subtypes in mucosal biopsies from the terminal ileum and large intestine of patients with symptoms of food allergy and/or irritable bowel syndrome

Keywords: histamine, intestine, irritable bowel syndrome, food allergy, autonomic nervous system, neurogastroenterology


The research article by Sander and colleagues1 in this issue of Gut, reports their results for expression of histamine receptor subtypes in the human intestinal tract from normal individuals and patients with symptoms of the irritable bowel syndrome (IBS) and/or food allergies (see page 498). Work of this nature was overdue because most of the available histological and functional data for histamine receptors in the small and large intestine were obtained from animal models. The authors' principal findings for the human bowel are in general agreement with the animal literature that reports on expression of the histamine H1, H2, and H4 receptor subtypes in the enteric nervous system (ENS), intestinal musculature, mucosal epithelium, and immune/inflammatory cells. In contrast, the finding by Sander and colleagues1 that histamine H3 receptors are not expressed in the human bowel was unexpected in view of the clearcut evidence for functional involvement of the H3 receptor subtype in the nervous control of motility, secretion, and blood flow in guinea pig intestine, which serves as the primary animal model.2,3,4,5

The authors' evidence for altered expression of histamine H1 and H2 receptor subtypes in mucosal biopsies from the terminal ileum and large intestine of patients with symptoms of food allergy and/or IBS is consistent with current concepts for the involvement of histamine release from enteric mast cells and its paracrine signalling function in the ENS as an underlying factor in these two disorders.5,6,7,8 Histamine is not expressed by enteric neurones and is not a neurotransmitter in the ENS.9 Its signalling function is paracrine in nature through release from enteric mast cells and inflammatory granulocytes. Mastocytosis and presumably elevated availability of histamine are present in microscopic colitis, parasitic infections, IBS, and no doubt additional functional gastrointestinal disorders associated with symptoms of cramping abdominal pain, watery diarrhoea, and defecation urgency.6,8,10,11,12,13,14,15,16,17

The appearance of histamine H2 receptors in human myenteric ganglia is reminiscent of expression of the H2 receptor subtype in the guinea pig ENS. Binding of histamine to H2 receptors on enteric neuronal cell bodies in the guinea pig, either during exogenous application of histamine or by degranulation of neighbouring mast cells, elevates neuronal excitability characterised by firing of longlasting trains of nerve impulses.18,19,20,21 In the case of submucosal secretomotor neurones, elevated firing rates increase the volume of mucosal secretions of electrolytes and H2O and thereby increase the liquidity of the intestinal contents, which in turn can underlie neurogenic secretory diarrhoea.22 For musculomotor neurones in the myenteric plexus, histamine H2 evoked firing alters contractile behaviour of the muscularis externa that is coordinated with organised secretory patterns.23,24 Similar outcomes for release of histamine and its actions at the H2 neuronal receptors, now reported by Sander and colleagues1 for human ENS, can be reasonably assumed. Nevertheless, progress in understanding specific pathophysiological malfunctions and therapeutic improvisation requires that future human research be pursued along the lines of what has been done in basic science models.

Excitation of ENS neuronal perikarya is one of the significant actions of histamine at the H2 receptor subtype. A second important action, which has been well documented in the guinea pig enteric ENS but not in humans, is suppression of synaptic transmission.2,19,25 Exposure of the ENS to histamine, either by exogenous application in vitro or by release from sensitised mast cells in response to allergins (for example, food proteins or infectious organisms), suppresses neurotransmitter release at four important information transmission nodes in the neural microcircuitry. Which are: (1) fast excitatory nicotinic synapses; (2) slow excitatory synapses where serotonin, substance P, calcitonin gene related peptide, and ATP are among the putative neurotransmitters; (3) slow inhibitory synapses, especially on submucosal secretomotor neurones, where norepinephrine release from the sympathetic innervation and somatostatin released from intrinsic neurons are inhibitory neurotransmitters; and (4) sympathetic neurovascular junctions.

Inhibition of neurotransmission in each of these cases is presynaptic. Stimulation of presynaptic inhibitory receptors by histamine suppresses the release of neurotransmitter from the presynaptic axonal terminal and thereby inhibits transmission of neural signals. Inhibition of transmission at the multitude of nicotinic synapses in the enteric neural networks would be expected to prevent “call‐up” of selective behavioural programmes or to selectively activate a specific programme in the ENS library of programmes (for example, intestinal defence).5 Suppression of slow excitatory transmission, either at selective slow synapses or in combination with suppression of fast nicotinic transmission, is probably also involved in generation of the pattern of defensive intestinal behaviour, which can be demonstrated during exposure to sensitising antigens in previously sensitised animals. Slow inhibitory postsynaptic potentials (IPSPs) in submucosal secretomotor neurones impose a braking action on neurogenic secretion that is removed when histamine is applied experimentally or released from enteric mast cells in sensitised animals. Removal of the sympathetic brake from secretomotor neurones is a factor underlying the diarrhoeal states associated with allergic responses and mucosal inflammation.2 Suppression of norepinephrine release at submucosal neurovascular junctions removes the sympathetic braking action on blood flow, which in effect supports stimulation of neurogenic mucosal secretion.4

Several types of presynaptic inhibitory receptors are expressed in the ENS, one of which is a histaminergic receptor. The presynaptic histaminergic inhibitory receptor in the guinea pig ENS belongs to the histamine H3 receptor subtype. The slow IPSPs in guinea pig secretomotor neurones, which are mediated by release of norepinephrine and somatostatin, are suppressed by histamine.2 Selective histamine H3 agonists, but not histamine H1 or H2 agonists, act presynaptically to suppress IPSPs, and selective H3 antagonists, but not H1 or H2 antagonists, block both the effects of exogenously applied histamine and the effects of histamine released from mast cells in sensitised animal preparations.2,19,20,21,25 Likewise, suppression of excitatory neurotransmission at other neural synapses and neurovascular junctions reflects histamine H3 mediated inhibition of neurotransmitter release.4

Absence of the histamine H3 receptor subtype from human bowel, as reported by Sander and colleagues,1 was unexpected and is paradoxical in view of the evidence in the literature for its expression and importance in the animal model. Data to explain the paradox are not readily available. On the one hand, failure to find the human receptor with any of three valid methods (that is, immunohistochemistry, western blot, or reverse transcription‐polymerase chain reaction) strongly supports the conclusion that the H3 receptor is not expressed in human bowel. On the other hand, evidence from physiological studies convincingly supports expression and important functional significance of the receptor in the guinea pig model. This is a dilemma raised by Sander and colleagues.1

The importance of histamine release from enteric mast cells in terms of intestinal symptoms, which are associated with human allergy, IBS and brain‐gut interactions in stress is widely supported and convincing.12,13,14,15,26 Symptoms of watery diarrhoea, urgency, cramping abdominal pain, and intestinal hypersensitivity to distension in humans appear in general to have a counterpart in animal models, whether it is a guinea pig, rat, or canine model.5,6 These symptoms are perceived as side effects of the “running” of a specific ENS neural programme that has evolved as a defensive mechanism for rapid expulsion from the intestine of a threat to the integrity of the whole animal. If this is indeed the case, then the mechanisms of histaminergic call‐up of programmed intestinal defence are not expected to differ much across mammalian species. Most of the results reported by Sander and colleagues1 are consistent with this concept, except for the absence of the histamine H3 receptor subtype. Histaminergic presynaptic inhibition that removes the sympathetic brake on secretion and mucosal blood flow would seem to be a necessary requirement in the “running” of the secretory component of the neural defence programme that “flushes” threatening agents and organisms from the mucosa and maintains them in suspension in a fluid filled intestine awaiting clearance by powerful propulsive motility.

In view of the importance of immune/inflammatory cells and histamine signalling in the ENS, thorough understanding for the human gut is imperative. A credible start in this direction has been made by Sander and colleagues.1 Now, neurogastroenterological research must determine whether presynaptic inhibition in the ENS has the same significance for the common symptoms of food allergy, mucosal inflammation, and brain‐gut interactions in stress in humans, as is known to exist in animal models. If this proves to be the case, then additional investigation will be needed to determine if it might be mediated by a histamine receptor other than the H3 subtype.

Footnotes

Conflict of interest: None declared.

References

  • 1.Sander L E, Lorentz A, Sellge G.et al Selective expression of histamine receptors H1R, H2R, and H4R, but not H3R, in the human intestinal tract. Gut 200655498–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Liu S, Xia Y, Hu H‐Z.et al Histamine H3 receptor‐mediated suppression of inhibitory synaptic transmission in the guinea‐pig submucous plexus. Eur J Pharmacol 200039749–54. [DOI] [PubMed] [Google Scholar]
  • 3.Blandizzi C, Tognetti M, Colucci R.et al Histamine H(3) receptors mediate inhibition of noradrenaline release from intestinal sympathetic nerves. Br J Pharmacol 20001291387–1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ishikawa S, Sperelakis N. A novel class (H3) of histamine receptors on perivascular nerve terminals. Nature 1987327158–160. [DOI] [PubMed] [Google Scholar]
  • 5.Wood J D. Enteric neuroimmuno physiology and pathophysiology. Gastroenterology 2004127635–657. [DOI] [PubMed] [Google Scholar]
  • 6.Wood J D. Neuro‐pathophysiology of IBS. J Clin Gastroenterol 200235(suppl)11–22. [Google Scholar]
  • 7.Barbara G, De Giorgio R, Stanghellini V.et al New pathophysiological mechanisms in irritable bowel syndrome. Aliment Pharmacol Ther 200420(suppl 2)1–9. [DOI] [PubMed] [Google Scholar]
  • 8.Barbara G, Stanghellini V, De Giorgio R.et al Activated mast cells in proximity to colonic nerves correlate with abdominal pain in irritable bowel syndrome. Gastroenterology 2004126693–702. [DOI] [PubMed] [Google Scholar]
  • 9.Panula P, Kaartinen M, Macklin M.et al Histamine‐containing peripheral neuronal and endocrine systems. J Histochem Cytochem 198533933–941. [DOI] [PubMed] [Google Scholar]
  • 10.O'Sullivan M, Clayton N, Breslin N P.et al Increased mast cells in the irritable bowel syndrome. Neurogastroenterol Motil 200012449–457. [DOI] [PubMed] [Google Scholar]
  • 11.Baum C A, Bhatia P, Miner P B., Jr Increased colonic mucosal mast cells associated with severe watery diarrhea and microscopic colitis. Dig Dis Sci 1989341462–1465. [DOI] [PubMed] [Google Scholar]
  • 12.Siddiqui A A, Miner P B., Jr The role of mast cells in common gastrointestinal diseases. Curr Allergy Asthma Rep 2004447–54. [DOI] [PubMed] [Google Scholar]
  • 13.Weston A P, Biddle W L, Bhatia P S.et al Terminal ileal mucosal mast cells in irritable bowel syndrome. Dig Dis Sci 1993381590–1595. [DOI] [PubMed] [Google Scholar]
  • 14.Libel R, Biddle W L, Miner P B., Jr Evaluation of anorectal physiology in patients with increased mast cells. Dig Dis Sci 199338877–881. [DOI] [PubMed] [Google Scholar]
  • 15.Miner P B., Jr The role of the mast cell in clinical gastrointestinal disease with special reference to systemic mastocytosis. J Invest Dermatol 19919640–3S. [PubMed] [Google Scholar]
  • 16.Russell D A, Castro G A. Immunological regulation of colonic ion transport. Am J Physiol 1989256G396–G403. [DOI] [PubMed] [Google Scholar]
  • 17.Alizadeh H, Castro G A, Weems W A. Intrinsic jejunal propulsion in the guinea pig during parasitism with Trichinella spiralis. Gastroenterology 198793784–790. [DOI] [PubMed] [Google Scholar]
  • 18.Nemeth P R, Ort C A, Wood J D. Intracellular study of effects of histamine on electrical behaviour of myenteric neurones in guinea‐pig small intestine. J Physiol 1984355411–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Frieling T, Cooke H J, Wood J D. Histamine receptors on submucous neurons in guinea pig colon. Am J Physiol 1993264G74–G80. [DOI] [PubMed] [Google Scholar]
  • 20.Frieling T, Cooke H J, Wood J D. Neuroimmune communication in the submucous plexus of guinea pig colon after sensitization to milk antigen. Am J Physiol 1994267G1087–G1093. [DOI] [PubMed] [Google Scholar]
  • 21.Frieling T, Palmer J M, Cooke H J.et al Neuroimmune communication in the submucous plexus of guinea pig colon after infection with Trichinella spiralis. Gastroenterology 19941071602–1609. [DOI] [PubMed] [Google Scholar]
  • 22.Cooke H J. Neurotransmitters in neuronal reflexes regulating intestinal secretion. Ann N Y Acad Sci 200091577–80. [DOI] [PubMed] [Google Scholar]
  • 23.Cooke H J, Wang Y Z, Rogers R. Coordination of Cl secretion and contraction by a histamine H2‐receptor agonist in guinea pig distal colon. Am J Physiol 1993265G973–G978. [DOI] [PubMed] [Google Scholar]
  • 24.Bozarov A V, Wang Y ‐ Z, Javed N.et al New method to study coordination of secretory and motility reflexes using sonomicrometry and voltage clamp apparatus in flat intestinal sheets. Gastroenterology 2005128(suppl 2)A608 [Google Scholar]
  • 25.Tamura K, Palmer J M, Wood J D. Presynaptic inhibition produced by histamine at nicotinic synapses in enteric ganglia. Neuroscience 198825171–179. [DOI] [PubMed] [Google Scholar]
  • 26.Santos J, Saperas E, Nogueiras C, Malagelada J R.et al Release of mast cell mediators into the jejunum by cold pain stress in humans. Gastroenterology 1998114640–648. [DOI] [PubMed] [Google Scholar]

Articles from Gut are provided here courtesy of BMJ Publishing Group

RESOURCES