intestinal serotonin signalling in irritable bowel syndrome

10 downloads 4893 Views 304KB Size Report
Jan 27, 2006 - Review article: intestinal serotonin signalling in irritable bowel syndrome ... University of Vermont College of. Medicine .... 5-HT release. SERT. 5-HIAA. Serum 5-HT. Reference. « fl fl. « fl. 5. «1. › 13. « ..... AGA technical review.
Alimentary Pharmacology & Therapeutics

Review article: intestinal serotonin signalling in irritable bowel syndrome G. M. MAWE* , M. D. COATES* & P. L. MOSES 

Departments of *Anatomy and Neurobiology and  Medicine, University of Vermont College of Medicine, Burlington, VT, USA Correspondence to: Dr G. M. Mawe, Given Building D403A, Department of Anatomy and Neurobiology, 89 Beaumont Ave, University of Vermont, Burlington, VT 05405, USA. E-mail: [email protected]

Publication data Submitted 8 December 2005 First decision 3 January 2006 Resubmitted 27 January 2006 Accepted 27 January 2006

SUMMARY Alterations in motility, secretion and visceral sensation are hallmarks of irritable bowel syndrome. As all of these aspects of gastrointestinal function involve serotonin signalling between enterochromaffin cells and sensory nerve fibres in the mucosal layer of the gut, potential alterations in mucosal serotonin signalling have been explored as a possible mechanism of altered function and sensation in irritable bowel syndrome. Literature related to intestinal serotonin signalling in normal and pathophysiological conditions has been searched and summarized. Elements of serotonin signalling that are altered in irritable bowel syndrome include: enterochromaffin cell numbers, serotonin content, tryptophan hydroxylase message levels, 5-hydroxyindoleacedic acid levels, serum serotonin levels and expression of the serotonin-selective reuptake transporter. Both genetic and epigenetic factors could contribute to decreased serotonin-selective reuptake transporter in irritable bowel syndrome. A serotonin-selective reuptake transporter gene promoter polymorphism may cause a genetic predisposition, and inflammatory mediators can induce serotonin-selective reuptake transporter downregulation. While a psychiatric co-morbidity exists with IBS, changes in mucosal serotonin handling support the concept that there is a gastrointestinal component to the aetiology of irritable bowel syndrome. Additional studies will be required to gain a more complete understanding of changes in serotonin signalling that are occurring, their cause and effect relationship, and which of these changes have pathophysiological consequences. Aliment Pharmacol Ther 23, 1067–1076

ª 2006 The Author Journal compilation ª 2006 Blackwell Publishing Ltd doi:10.1111/j.1365-2036.2006.02858.x

1067

1068 G . M . M A W E et al.

INTRODUCTION Irritable bowel syndrome (IBS) is a common gastrointestinal (GI) disorder associated with alterations in motility, secretion and visceral sensation. A range of clinical symptoms characterize this disorder, including altered stool frequency and form, abdominal pain and bloating. Impairment in quality of life is common for the significant proportion of the population who suffer from IBS.1–3 Specific clinical criteria differentiate IBS from other functional GI disorders as well as from inflammatory bowel disease (IBD) and other non-functional GI disorders. There are no specific biological, radiographic, endoscopic or physiological markers that have been identified in this disorder, and therefore, the diagnosis of IBS is based solely on these clinical criteria. Despite extensive interest and investigation, the pathogenesis of IBS remains unclear. Improved understanding of the enteric nervous system (ENS), and the neuroenteric pathways involved in GI function, may provide a conceptual framework to integrate previously described motility, sensory, and cognitive features of the disease and to identify new therapeutic targets. In this article, we will specifically review the evolving understanding of the important roles of serotonin (5-HT) as a signalling molecule in the gut and the pathophysiology of functional GI disorders like IBS. We have searched and summarized relevant literature related to 5-HT signalling in the mucosa of the intestines under normal conditions and changes in key elements of 5-HT signalling that have been reported in IBS and in response to inflammation.

THE ROLES OF SEROTONIN AS A SIGNALLING MOLECULE IN THE GUT The ENS is by far the most vast and complex component of our peripheral nervous system.4 Distinguishing features of the ENS include the quantity of neurones, the variety of neurotransmitters and associated receptors, and the existence of intrinsic reflex circuitry. The number of neurones in the human small intestines alone (100 000 000) approximates the number of neurones in the entire spinal cord. Almost every known neurotransmitter can be found in the ENS, and most receptors associated with these neurotransmitters are also found in the ENS. The GI tract is the only part of the body with neural reflexes that are housed entirely within the organ. Unlike reflexes associated with neural control of other organ systems or smooth

muscle, complete neural circuits that include sensory neurones, interneurones, and motor neurones are contained within the wall of the intestines, and these circuits are responsible for motility, secretion and vascular tone in the gut. One of the signalling molecules with an unambiguous physiological role in the ENS is 5-HT.5 5-HT is often thought of as a neurotransmitter exclusive to the central nervous system (CNS), because of its well-defined roles in depression, arousal, pain pathways and other CNS functions. However, the major source of bioavailable 5-HT in the human body is located in the bowel, primarily in a subset of epithelial cells called enterochromaffin (EC) cells.6 EC cells express the enzymatic machinery, including the rate limiting enzyme, tryptophan hydroxylase (TpH), to synthesize 5-HT which is then stored in secretory granules. 5-HT is released from EC cells in response to luminal stimuli, including mechanical forces. Once released, 5-HT acts on receptors located on the processes of sensory neurones that pass into the lamina propria. These include branches of intrinsic sensory neurones whose cell bodies are located in submucosal and myenteric ganglia, as well as sensory neurones located in spinal (dorsal root) ganglia and vagal (nodose) ganglia (see Figure 1). As a result, release of 5-HT from EC cells initiates activation of motor, secretory and vasodilatory reflexes, as well as stimulation of afferent signals to the brain and spinal cord. An important facet of efficient intercellular signalling is the termination of the signal. In the case of 5-HT signalling in the brain, nerve terminals that release 5-HT express a serotonin-selective reuptake transporter (SERT), so they are uniquely capable of terminating the signals that they initiate.7, 8 5-HT neurotransmission in the brain can be augmented by compounds that function as selective serotonin reuptake inhibitors (SSRIs). These drugs, fluoxetine (Prozac), citalopram (Celexa) and paroxetine (Paxil) and related compounds, increase 5-HT availability in the synaptic cleft by inhibiting SERT. The intestinal mucosa has an enormous capacity to remove 5-HT from the interstitial space because all of the epithelial cells that line the luminal surface of the gut express SERT.5, 9 Therefore, once 5-HT is released from EC cells and acts on local receptors, it is transported into epithelial cells by SERT. Postprandially, 5-HT also enters the bloodstream where it is rapidly taken up by platelets, which also express SERT.10 In fact, 5-HT found in platelets arises primarily from 5HT released from EC cells.11, 12 ª 2006 The Authors, Aliment Pharmacol Ther 23, 1067–1076 Journal compilation ª 2006 Blackwell Publishing Ltd

REVIEW: INTESTINAL SEROTONIN SIGNALLING IN IBS 1069

Figure 1. Schematic diagram depicting the branch patterns of intrinsic and extrinsic primary afferent neurones that innervate the intestinal mucosa. Intrinsic primary afferent nerve fibres originate from neurones with cell bodies located in the myenteric and submucosal plexuses. These neurones are involved in local activities such as motility, secretion and vasodilation within the intestines. Extrinsic primary afferent nerves arise from neurones with cell bodies located in the nodose ganglia (vagal afferents) and dorsal root ganglia (spinal afferents). Extrinsic afferents transmit signals related to digestive reflexes, satiety, pain and discomfort from the gut to the central nervous system. As all these types of primary afferent neurones extend processes into the lamina propria of the intestines, they can be activated by serotonin (5-HT) released from enterochromaffin (EC) cells that are located in the crypt epithelium.

EVIDENCE FOR ALTERED SEROTONIN SIGNALLING IN IBS It is becoming increasingly clear that changes in 5-HT signalling occur in IBS. Due to the importance of 5-HT as an intercellular signalling molecule in intrinsic and extrinsic gut reflexes, various elements of 5-HT signalling have been evaluated to determine whether it is altered in IBS.5, 13–17 These include EC cell numbers, TpH message levels, 5-HT content, 5-HT release, SERT immunoreactivity, SERT message levels and plateletfree serum 5-HT levels. The findings of these studies are summarized in Tables 1–3.

A common feature of the studies conducted to date is that they have all reported changes in one or more aspects of 5-HT signalling in IBS. However, the results of these investigations are not entirely in harmony. For example, various combinations of changes in EC cell populations and 5-HT content have been reported in the different forms of IBS. One of these studies reported that no changes in 5-HT release were detected in IBS-D or IBS-C under basal or stimulated conditions.5 If this finding reflects the physiological nature of 5-HT release in these individuals, it would indicate that the same amount of 5-HT is being released regardless of possible changes in EC cell numbers or

Table 1. Diarrhoea-predominant IBS EC cells

5-HT

TpH mRNA

5-HT release

SERT

«





«



5-HIAA

« «

› indicates increase; fl indicates significant decrease; « indicates no significant change; 1 Measured in urine samples.

ª 2006 The Authors, Aliment Pharmacol Ther 23, 1067–1076 Journal compilation ª 2006 Blackwell Publishing Ltd

Reference



13

5 1

«

Serum 5-HT

14

1070 G . M . M A W E et al.

Table 2. Constipation-predominant IBS EC cells

5-HT

TpH mRNA

5-HT release

SERT

«

fl › «



«



«

5-HIAA

Serum 5-HT

Reference 5

« fl

14



15

Serum 5-HT

Reference

› indicates increase; fl indicates significant decrease; « indicates no significant change.

Table 3. Postinfectious IBS (with diarrhoea symptoms) EC cells › › «

5-HT

TpH mRNA

5-HT release

SERT

5-HIAA

17 16

«





15

› indicates increase; fl indicates significant decrease; « indicates no significant change.

5-HT content, and therefore changes in 5-HT signalling upstream of release may be irrelevant. When interpreting data related to elements of 5-HT signalling downstream of 5-HT release, both consistencies and contradictions exist. In the case of IBS-D, Coates et al.5 reported a decrease in SERT expression, indicating that the capacity of the epithelial cells to sequester locally released 5-HT would be compromised. Consistent with this, Dunlop et al.15 reported an increase in serum 5-HT levels, which was also reported in IBS-D. On the other hand, a decrease in SERT expression was also reported in IBS-C whereas a decrease in serum 5-HT was observed in this form of IBS. Further studies will have to be conducted to validate the findings that have been reported to date and to resolve the discrepancies that exist.

IS DECREASED SERT EXPRESSION IN IBS DUE TO GENETIC AND/OR EPIGENETIC FACTORS? The 5-HT transporter, SERT, is a highly regulated protein. For example, the genetic make-up of an individual appears to influence their level of SERT expression.18 Furthermore, activation of G-proteincoupled receptors can lead to changes in SERT by modulating its transcription by moving the transporter to or away from the plasma membrane, and by

altering the phosphorylation state of the protein. It is therefore possible that genetic and/or epigenetic factors could affect SERT expression and function in the intestinal epithelium.

Genetic factors It is possible that individuals with IBS have a genetic predisposition for decreased SERT expression. The human SERT gene is located on human chromosome 17; it spans 37.8 kb and is composed of 14 exons that encode a 630 amino acid protein.19, 20 One possible explanation for the variability in SERT expression amongst individuals is the SERT gene-linked polymorphic region (SERT-LPR).21 The SERT-LPR, comprising long (l) and short (s) variants, is located 1.2 kb upstream of the transcription start site in exon 1, and may influence the level of transcription. The l variant contains 16 copies of a 20–23 base pair sequence whereas the s variant contains 14 copies of this sequence. Lymphoblast transfection studies with expression vectors containing the long or short variants linked to a reporter gene indicate that the long variant directs higher levels of transcription compared with the short variant [see Murphy et al. (18) for review]. Consistent with this, higher SERT mRNA levels and higher rates of 5-HT uptake were measured in lymphoblasts of l/l homozygotes compared with those ª 2006 The Authors, Aliment Pharmacol Ther 23, 1067–1076 Journal compilation ª 2006 Blackwell Publishing Ltd

REVIEW: INTESTINAL SEROTONIN SIGNALLING IN IBS 1071

containing at least one copy of the s allele.22 The l variant was also associated with higher rates of 5-HT uptake, and higher levels of 5-HT binding in platelets.23–25 It is important to note, however, that not all studies have reported a correlation between SERT-LPR alleles and SERT expression. Analysis of SERT mRNA levels in a large number of permanent lymphoblast cell lines failed to detect a correlation with SERT-LPR genotype.26 In addition, no correlation between SERT-LPR alleles and promoter activity was observed with expression vectors assayed in COS-727, 28 or PC12 cells.22, 27 Furthermore, no statistically significant correlations have been detected between SERT-LPR alleles and binding of SERT ligands in the brains of healthy volunteers.29, 30 A number of studies have examined possible relationships between SERT-LPR alleles and psychiatric disorders, anxiety-related personality traits, suicide, alcoholism and response to antidepressants.18 Although many studies have reported correlations between SERT-LPR alleles and a given clinical phenotype, others have not. The relationship between SERT-LPR and IBS is unclear at this point, as data from the four studies conducted to date are somewhat contradictory. The first such study31 examined possible associations between SERT-LPR and different clinical patterns of IBS, including IBS-C, IBS-D and IBS with alternating symptoms (IBS-A) in a Turkish population. The distributions of SERT polymorphisims were comparable between the healthy subjects and the IBS population as a whole. However, genotype variations were detected amongst the IBS populations, with the s/s genotype being more predominant in individuals with IBS-C and the l/s genotype being more predominant in individuals with IBS-D. The second study32 investigated genotypes related to SERT-LPR and a2 adrenoreceptors in individuals in the upper Midwestern USA with lower GI symptoms (n ¼ 274) that were divided into IBS-D, IBS-C, IBS-A and chronic pain. In this study, no difference was detected between the SERTLPR genotypes of control individuals vs. the lower GI symptom population as a whole, or controls vs. IBS-D (n ¼ 128), IBS-C (n ¼ 90) or IBS-A (n ¼ 38). Results of this study did identify combinations of polymorphisms that were associated with high symptom scores indicating that the SERT-LPR genotype may predispose a given individual to lower GI symptoms. The authors concluded that future studies of interactions between ª 2006 The Authors, Aliment Pharmacol Ther 23, 1067–1076 Journal compilation ª 2006 Blackwell Publishing Ltd

candidate genes associated with motor, sensory and/or behavioural functions would be of interest. A third study,33 conducted in Korea, did not detect a relationship between SERT-LPR genotype in controls (n ¼ 56) vs. the IBS population as a whole (n ¼ 33) or the IBSC (n ¼ 12), IBS-D (n ¼ 15) and IBS-A (n ¼ 6) subgroups. They reported that s/s was the predominant genotype in healthy volunteers (n ¼ 56) as well as in individuals with IBS. The fourth study34 involved comparison of SERT-LPR genotype in a large cohort of North American Caucasian females with IBS-D (n ¼ 194) vs. North American Caucasian female healthy volunteers (n ¼ 448). In this investigation, a significant association was observed between IBS-D and the s/s genotype of SERT-LPR. Correlative support for a genetic predisposition for decreased SERT expression comes from a study that involved an analysis of SERT-binding kinetics in platelets. Similar to serotonergic neurones and intestinal epithelial cells, platelets express SERT on their outer membranes and they act as sponges for 5-HT that enters the bloodstream. In a study by Bellini et al., SERT-binding kinetics of platelets from individuals with IBS-D were compared with those of healthy volunteers.35 Both the maximal binding capacity and dissociation constant were lower in individuals with IBS-D, suggesting a lower density of SERT of platelets in individuals with IBS-D. The study demonstrating the most dramatic correlation between IBS and SERT-LPR was the study performed by Hicks and colleagues34 that involved the largest sample size and this study involved a relatively homogeneous population (North American Caucasian females). The finding that a link between SERT-LPR and a specific subtype of IBS, but not in IBS as a whole, or subtypes of IBS in heterogeneous populations supports the concept that a universal aetiology for IBS does not exist. It is likely that a variety of conditions, triggers and contributing factors result in the wide array of symptoms that are grouped into the descriptor, IBS. While it is not yet possible to draw a firm conclusion regarding a genetic predisposition for IBS, this is clearly a promising avenue to explore, and any links that are discovered could possibly lead to more effective treatment plans.

Epigenetic factors In addition to being influenced by means of genetic polymorphisms, SERT expression and function can also

1072 G . M . M A W E et al.

be modulated via second messengers that are activated by homologous (5-HT) and heterologous receptors.8, 36 For example, 5-HT, cholera toxin and forskolin increase SERT expression, while cyclic adenosine monophosphate (cAMP) activators increase SERT activity. SSRIs decrease SERT expression, and PKC activation decreases SERT activity. Furthermore, adenosine receptor stimulation activates two different protein kinase G (PKG)dependent pathways that increase 5-HT uptake by moving SERT to the cytosolic membrane and by phosphorylating SERT via a mitogen activated protein (MAP) kinase-dependent mechanism. Various elements of 5-HT signalling are altered in IBD5, 37–39 (see Table 4). In addition, 5-HT content, EC cell numbers, release and SERT expression are altered in a number animal models of intestinal inflammation, including trinitro benzenesulphonic acid (TNBS) colitis40 and ileitis41 in guinea-pig, TNBS colitis in mouse,42 Trichinella spiralis ileitis in mouse,43 Citrobacter rodentium colitis in mouse44 and dextran sodium sulphate colitis in rat45 (see Table 5).

The changes that have been detected in the EC cell population and 5-HT levels in postinfectious IBS (PI-IBS) may be brought on by the inflammatory response associated with the infection. The data from experimental animals, which represent genetically homogeneous populations, indicate that the inflammatory process can lead to changes in EC cells as well as SERT expression by enterocytes. This concept is supported by data we presented at the 2005 meeting of the American Motility Society demonstrating that tumour necrosis factor alpha and c-interferon decrease SERT function in CACO-2 cells, a human colonic epithelial cell line.46 Furthermore, the decrease in SERT immunoreactivity that is detected in T. spiralis-infected mouse intestine persists following recovery from inflammation.43 It is worth noting that EC cells and mucosal 5-HT levels are consistently found to be elevated in animal models of intestinal inflammation and in PI-IBS (Tables 4 and 5), whereas this trend is either not detected or not as consistently observed in IBD, IBS-D or IBS-C (Tables 1–3). As the animal models

Table 4. Mucosal serotonin signalling in inflammatory bowel disease Form of IBD UC CD & UC UC CD & UC

EC cells

5-HT

TpH mRNA

5-HT release

SERT



fl fl



«



5-HIAA

Reference



37

5

38

fl ›1

39

CD, Crohn’s disease; UC, ulcerative colitis. › indicates increase; fl indicates significant decrease; « indicates no significant change. 1 Measured area rather than the number of EC cells.

Table 5. Mucosal serotonin signalling in animal models of intestinal inflammation Model Guinea-pig TNBS colitis Guinea-pig TNBS ileitis Mouse TNBS colitis Mouse T. spiralis ileitis Mouse Citrobacter rodentium ileitis Rat DSS colitis

EC cells

5-HT

5-HT release

SERT

Reference

› › ›1

› › « ›

›2 › «

fl fl fl fl fl

40

fl ›

› ›

41 42 43 44 45

TNBS, trinitro benzenesulphonic acid; DSS, dextran sodium sulphate. 1 May be due to mast cell hyperplasia as mast cells contain 5-HT in mouse. 2 May be due to decreased 5-HT reuptake rather than increased release.

ª 2006 The Authors, Aliment Pharmacol Ther 23, 1067–1076 Journal compilation ª 2006 Blackwell Publishing Ltd

REVIEW: INTESTINAL SEROTONIN SIGNALLING IN IBS 1073

and PI-IBS involve relatively short time points, it is possible that EC cell hyperplasia occurs and 5-HT levels are elevated during the inflammatory response and then rebound to normal or below normal levels over time.

THE CAUSE AND EFFECT RELATIONSHIP OF ALTERED SEROTONIN SIGNALLING IN IBS Serotonin is clearly an important signalling molecule in the activation of motor and secretory reflexes and in the activation of sensory signals from the gut to the CNS. The findings that are described above demonstrate that 5-HT signalling is altered in IBS-D, IBS-C and PI-IBS, but the cause and effect relationship of epigenetic changes in the elements of 5-HT signalling is unclear. In other words, we do not know whether changes in 5-HT signalling contribute to the alterations in GI function and sensation that are the hallmarks of IBS, and/or if elements of 5-HT are altered in response to disrupted function and sensation. We do not yet know whether 5-HT signalling changes in response to altered gut function, but several lines of evidence support the concept that altered 5-HT signalling can lead to changes in gut function. For example, transgenic mice lacking the gene for SERT typically exhibit symptoms similar to those of IBS-D, but some mice are more similar to IBS-C, as they have decreased colonic motility.47 In vitro studies involving evaluation of propulsive motility in the guinea-pig distal colon also demonstrate that changes in 5-HT signalling can affect motility. For example, administration of low concentrations of the SSRI, fluoxetine (Prozac), increases the rate of propulsive motility at low concentrations and slows motility at higher concentrations.40, 48 Furthermore, administration of desensitizing concentrations of 5-HT decreases propulsive motility in vivo. These findings support the concept that peristaltic reflex is complicated, and while it can be augmented by slight stimulation, over-stimulation can lead to decreased efficiency, possibly through receptor desensitization in the case of increased 5-HT availability.

THE RELATIONSHIP BETWEEN SEROTONIN RECEPTOR DRUGS AND THE PATHOPHYSIOLOGY OF IBS Receptors that have been identified for 5-HT include the following: 5-HT1A-E, P, 5-HT2A,B,C, 5-HT3, 5-HT4, ª 2006 The Authors, Aliment Pharmacol Ther 23, 1067–1076 Journal compilation ª 2006 Blackwell Publishing Ltd

5-HT5, 5-HT6 and 5-HT7. Of these, 5-HT1A, 5-HT1P, 5-HT2, 5-HT3, 5-HT4 and 5-HT7 receptors have been identified in the gut, with 5-HT2 receptors being located primarily on muscle and epithelium, and the others being located primarily on nerves.49 Treatment strategies for IBS involving 5-HT-related compounds are currently directed at the predominant symptoms, and 5-HT3 and 5-HT4 receptors have been the targets of choice. The 5-HT3 receptor is a ligandgated ion channel,50 whereas the 5-HT4 receptor is a seven-transmembrane domain, G-protein-coupled receptor.51 The 5-HT4 receptor is linked to GS, and activation of this receptor leads to an increase in cyclic AMP. While both types of receptors can undergo desensitization, 5-HT3 receptors exhibit rapid desensitization upon sustained exposure to an agonist.52 This property of 5-HT3 receptors bring into question clinical potential of 5-HT3 agonists, particularly those with full agonist properties, and would make 5-HT3 receptors particularly susceptible to increased availability of 5-HT when SERT is decreased. In IBS-D, the 5-HT3 receptor antagonists such as alosetron are thought to work primarily by inhibiting the activation of 5-HT3 receptors located on the mucosal processes of intrinsic and extrinsic primary afferents. Inhibiting 5-HT3 receptors located on intrinsic sensory neurones can diminish motor and secretory reflex activity, and decreasing the activation of extrinsic sensory neurones, which transmit signals to the CNS, inhibits sensory signals leading to pain and discomfort.53 5-HT4 receptor agonists such as tegaserod are used as a treatment for IBS-C. It is not yet clear whether 5-HT4 receptors are located on the mucosal processes of intrinsic and extrinsic sensory neurones, but if these receptors are present at this location, 5-HT4 agonists would augment the activation of motor and secretory reflex activity. Electrophysiological and anatomical studies have demonstrated that 5-HT4 receptors are located on nerve terminals throughout the ENS that mediate excitatory synaptic transmission.54–56 Activation of these presynaptic 5-HT4 receptors leads to an increase in the amount of transmitters, such as acetylcholine, that are released from these terminals, and therefore enhances reflex responses. It is not understood whether 5-HT3 antagonists and/ or 5-HT4 agonists have an effect on the 5-HT signalling anomalies that have been detected in IBS, or whether their mechanisms of action simply involve increasing or decreasing reflex activity at sites further

1074 G . M . M A W E et al.

downstream in the circuitry. It is possible that increased 5-HT availability contributes to the symptoms of IBS-D, and if this is the case, an antagonist working at the site of 5-HT signalling between EC cells and sensory nerves would be operating at the site of disrupted 5-HT signalling. Conversely, if 5-HT4 receptors are also located on mucosal projections of afferent nerves, as some data suggest,57 5-HT4 agonists may promote the activation of enteric reflexes at the site of disrupted 5-HT signalling, and thereby relieve constipation. The presynaptic action of 5-HT4 agonists on nerve terminals in enteric neural circuitry is downstream from the site of disrupted mucosal 5-HT signalling. In an intriguing report by Tonini et al., 5-HT7 receptors were found to mediate intestinal smooth muscle relaxation and accommodation. Endogenous 5-HT was involved in smooth muscle accommodation in the preparatory phase of peristalsis by direct activation of 5-HT7 receptors. The authors suggest that abnormal stimulation of the 5-HT7 receptor may contribute to certain clinical syndromes like IBS and that the 5-HT7 receptor may be a reasonable candidate for therapeutic intervention in some patients.58

CONCLUDING REMARKS The studies summarized here provide compelling evidence for a role for altered mucosal 5-HT signalling in IBS, as well as in IBD and other GI disorders. While the data reported in these studies represent snapshots of 5-HT signalling in what are generally chronic conditions, it is clear that the sequence of events responsible for 5-HT signalling can be remodelled in response to various physiological and pathophysiological conditions. Although the cause and effect relationship of these changes has not been established, it is quite likely that altered 5-HT signalling contributes to abnormal gut function and heightened sensitivity in IBS. Further studies will be required to gain a more complete picture of the changes that are occurring, and which of these changes have pathophysiological consequences. One part of the picture that is missing is the status of 5-HT receptors in IBS, and also in IBD. Similar to SERT, 5-HT receptor expression is dynamic and could be affected by the amount of 5-HT that is available and by other factors such as inflammatory mediators. For example, adaptive changes in 5-HT3 receptor expression by enteric neurones have been detected in SERT knockout mice.59 Studies of 5-HT receptor expression in the intestinal mucosa have been hampered by the fact

that mRNA for these receptors is in the neuronal cell bodies, and is therefore not acquired in a mucosal biopsy. As effective receptor-selective antisera become available, progress may be made in this area. A comprehensive understanding of mucosal 5-HT signalling in IBS and IBD will require a thorough appreciation of the status of receptors that mediate the actions of 5-HT. It is highly unlikely that one given defect or alteration is responsible for the various changes in gut function and sensitivity that are encountered in disorders of GI function. In other words, a universal aetiology for IBS is not likely to exist. For example, in addition to the alterations in 5-HT signalling that are summarized here, it is clear that a co-morbidity exists between psychiatric disorders and IBS, and that many cases of IBS-D involve a previous infectious inflammatory event (postinfectious IBS). Furthermore, mounting evidence suggests a role of the stress hormone, corticotropin-releasing factor, in IBS.60 Therefore, the variability reported in many IBS studies may be due to the heterogeneity of the patient population. In line with this, it is unlikely that a given therapy will be highly effective in all individuals with IBS, or even a subtype of IBS. Further elucidation, at the molecular level, of the various changes that contribute to the symptoms of the various forms of IBS will hopefully enhance our ability to treat these individuals effectively. In order to make progress in this regard, it will be crucial for investigators to take care in defining the phenotypes of the individuals that are included in a given study, including their predominant GI symptoms, their psychiatric and past medical histories. Furthermore, DNA samples should be acquired for genotyping of these individuals as we identify candidate genes. This type of approach will improve our understanding of the role of 5-HT and other contributing factors in IBS. While the Rome criteria are adequate for making the diagnosis of IBS, assays for detecting molecular abnormalities that are found to contribute to the disorder may assist in identifying those individuals who are most likely to respond to a given treatment plan. This is a significant goal that would ease the frustration and burden of health care providers and patients alike.

ACKNOWLEDGEMENTS This work was supported by NIH grants DK62267, P20 COBRE grant RR16435 and NS050919, and a research grant from Novartis Pharmaceuticals. ª 2006 The Authors, Aliment Pharmacol Ther 23, 1067–1076 Journal compilation ª 2006 Blackwell Publishing Ltd

REVIEW: INTESTINAL SEROTONIN SIGNALLING IN IBS 1075

REFERENCES 1 Hahn BA, Yan S, Strassels S. Impact of irritable bowel syndrome on quality of life and resource use in the United States and United Kingdom. Digestion 1999; 60: 77–81. 2 Drossman DA, Camilleri M, Mayer EA, Whitehead WE. AGA technical review on irritable bowel syndrome. Gastroenterology 2002; 123: 2108–31. 3 Brandt LJ, Bjorkman D, Fennerty MB, et al. Systematic review on the management of irritable bowel syndrome in North America. Am J Gastroenterol 2002; 97 (Suppl. 11): S7–26. 4 Gershon MD. Nerves, reflexes, and the enteric nervous system: pathogenesis of the irritable bowel syndrome. J Clin Gastroenterol 2005; 39 (Suppl. 3): S184–93. 5 Coates MD, Mahoney CR, Linden DR, et al. Molecular defects in mucosal serotonin content and decreased serotonin reuptake transporter in ulcerative colitis and irritable bowel syndrome. Gastroenterology 2004; 126: 1657–64. 6 Erspamer V. Occurrence and distribution of 5-hydroxytryptamine (enteramine) in the living organism. Z Vitam Horm Fermentforsch 1957; 9: 74–96. 7 Blakely RD, Berson HE, Fremeau RT Jr, et al. Cloning and expression of a functional serotonin transporter from rat brain. Nature 1991; 354: 66–70. 8 Blakely RD, Defelice LJ, Galli A. Biogenic amine neurotransmitter transporters: just when you thought you knew them. Physiology (Bethesda) 2005; 20: 225–31. 9 Chen JX, Pan H, Rothman TP, Wade PR, Gershon MD. Guinea pig 5-HT transporter: cloning, expression, distribution, and function in intestinal sensory reception. Am J Physiol 1998; 275 (Pt 1): G433–48. 10 Da Prada M, Tranzer JP, Pletscher A. Storage of 5-hydroxytryptamine in human blood platelets. Experientia 1972; 28: 1328–9. 11 Erspamer V, Testini A. Observations on the release and turnover rate of 5-hydroxytryptamine in the gastrointestinal tract. J Pharm Pharmacol 1959; 11: 618–23. 12 Bertaccini G. Tissue 5-hydroxytryptomine and urinary 5-hyroxyindoleacetic acid after partial or total removal of the

13

14

15

16

17

18

19

20

21

22

23

gastro-intestinal tract in the rat. J Physiol (Lond) 1960; 153: 239–49. Bearcroft CP, Perrett D, Farthing MJ. Postprandial plasma 5-hydroxytryptamine in diarrhoea predominant irritable bowel syndrome: a pilot study. Gut 1998; 42: 42–6. Miwa J, Echizen H, Matsueda K, Umeda N. Patients with constipation-predominant irritable bowel syndrome (IBS) may have elevated serotonin concentrations in colonic mucosa as compared with diarrhea-predominant patients and subjects with normal bowel habits. Digestion 2001; 63: 188–94. Dunlop SP, Coleman NS, Blackshaw E, et al. Abnormalities of 5-hydroxytryptamine metabolism in irritable bowel syndrome. Clin Gastroenterol Hepatol 2005; 3: 349–57. Dunlop SP, Jenkins D, Neal KR, Spiller RC. Relative importance of enterochromaffin cell hyperplasia, anxiety, and depression in postinfectious IBS. Gastroenterology 2003; 125: 1651–9. Spiller RC, Jenkins D, Thornley JP, et al. Increased rectal mucosal enteroendocrine cells, T lymphocytes, and increased gut permeability following acute Campylobacter enteritis and in post-dysenteric irritable bowel syndrome. Gut 2000; 47: 804–11. Murphy DL, Lerner A, Rudnick G, Lesch KP. Serotonin transporter: gene, genetic disorders, and pharmacogenetics. Mol Interv 2004; 4: 109–23. Lesch KP, Balling U, Gross J, et al. Organization of the human serotonin transporter gene. J Neural Transm Gen Sect 1994; 95: 157–62. Ramamoorthy S, Bauman AL, Moore KR, et al. Antidepressant-and cocainesensitive human serotonin transporter: molecular cloning, expression, and chromosomal localization. Proc Natl Acad Sci U S A 1993; 90: 2542–6. Lesch KP, Mossner R. Genetically driven variation in serotonin uptake: is there a link to affective spectrum, neurodevelopmental, and neurodegenerative disorders? Biol Psychiatry 1998; 44: 179–92. Lesch KP, Bengel D, Heils A, et al. Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region. Science 1996; 274: 1527–31. Greenberg BD, Tolliver TJ, Huang SJ, Li Q, Bengel D, Murphy DL. Genetic variation in the serotonin transporter promo-

ª 2006 The Authors, Aliment Pharmacol Ther 23, 1067–1076 Journal compilation ª 2006 Blackwell Publishing Ltd

24

25

26

27

28

29

30

31

32

33

ter region affects serotonin uptake in human blood platelets. Am J Med Genet 1999; 88: 83–7. Nobile M, Begni B, Giorda R, et al. Effects of serotonin transporter promoter genotype on platelet serotonin transporter functionality in depressed children and adolescents. J Am Acad Child Adolesc Psychiatry 1999; 38: 1396–402. Stoltenberg SF, Twitchell GR, Hanna GL, et al. Serotonin transporter promoter polymorphism, peripheral indexes of serotonin function, and personality measures in families with alcoholism. Am J Med Genet 2002; 114: 230–4. Hranilovic D, Stefulj J, Schwab S, et al. Serotonin transporter promoter and intron 2 polymorphisms: relationship between allelic variants and gene expression. Biol Psychiatry 2004; 55: 1090–4. Sakai K, Nakamura M, Ueno S, et al. The silencer activity of the novel human serotonin transporter linked polymorphic regions. Neurosci Lett 2002; 327: 13–6. Mortensen OV, Thomassen M, Larsen MB, Whittemore SR, Wiborg O. Functional analysis of a novel human serotonin transporter gene promoter in immortalized raphe cells. Brain Res Mol Brain Res 1999; 68: 141–8. Willeit M, Stastny J, Pirker W, et al. No evidence for in vivo regulation of midbrain serotonin transporter availability by serotonin transporter promoter gene polymorphism. Biol Psychiatry 2001; 50: 8–12. Shioe K, Ichimiya T, Suhara T, et al. No association between genotype of the promoter region of serotonin transporter gene and serotonin transporter binding in human brain measured by PET. Synapse 2003; 48: 184–8. Pata C, Erdal ME, Derici E, Yazar A, Kanik A, Ulu O. Serotonin transporter gene polymorphism in irritable bowel syndrome. Am J Gastroenterol 2002; 97: 1780–4. Kim HJ, Camilleri M, Carlson PJ, et al. Association of distinct alpha(2) adrenoceptor and serotonin transporter polymorphisms with constipation and somatic symptoms in functional gastrointestinal disorders. Gut 2004; 53: 829–37. Lee DY, Park H, Kim WH, Lee SI, Seo YJ, Choi YC. Serotonin transporter gene

1076 G . M . M A W E et al.

34

35

36

37

38

39

40

41

42

polymorphism in healthy adults and patients with irritable bowel syndrome. Korean J Gastroenterol 2004; 43: 18– 22. Yeo A, Boyd P, Lumsden S, et al. Association between a functional polymorphism in the serotonin transporter gene and diarrhoea predominant irritable bowel syndrome in women. Gut 2004; 53: 1452–8. Bellini M, Rappelli L, Blandizzi C, et al. Platelet serotonin transporter in patients with diarrhea-predominant irritable bowel syndrome both before and after treatment with alosetron. Am J Gastroenterol 2003; 98: 2705–11. Blakely RD, Bauman AL. Biogenic amine transporters: regulation in flux. Curr Opin Neurobiol 2000; 10: 328–36. Magro F, Vieira-Coelho MA, Fraga S, et al. Impaired synthesis or cellular storage of norepinephrine, dopamine, and 5-hydroxytryptamine in human inflammatory bowel disease. Dig Dis Sci 2002; 47: 216–24. Ahonen A, Kyosola K, Penttila O. Enterochromaffin cells in macrophages in ulcerative colitis and irritable colon. Ann Clin Res 1976; 8: 1–7. El-Salhy M, Danielsson A, Stenling R, Grimelius L. Colonic endocrine cells in inflammatory bowel disease. J Intern Med 1997; 242: 413–9. Linden DR, Chen JX, Gershon MD, Sharkey KA, Mawe GM. Serotonin availability is increased in mucosa of guinea pigs with TNBS-induced colitis. Am J Physiol Gastrointest Liver Physiol 2003; 285: G207–16. O’Hara JR, Ho W, Linden DR, Mawe GM, Sharkey KA. Enteroendocrine cells and serotonin availability are altered in the mucosa of guinea pigs with TNBS ileitis. Am J Physiol Gastrointest Liver Physiol 2004; 287: G998–1007. Linden DR, Foley KF, McQuoid C, Simpson J, Sharkey KA, Mawe GM. Serotonin transporter function and expression are reduced in mice with

43

44

45

46

47

48

49

50

51

TNBS-induced colitis. Neurogastroenterol Motil 2005; 17: 565–74. Wheatcroft J, Wakelin D, Smith A, Mahoney CR, Mawe GM, Spiller R. Enterochromaffin cell hyperplasia and decreased serotonin transporter in a mouse model of postinfectious bowel dysfunction. Neurogastroenterol Motil 2005; 17: 863–70. O’Hara JR, Skinn AC, MacNaughton WK, Sherman PM, Sharkey KA. Consequences of Citrobacter rodentium infection on enteroendocrine cells and the enteric nervous system in the mouse colon. Cell Microbiol 2006; in press. Oshima S, Fujimura M, Fukimiya M. Changes in number of serotonin-containing cells and serotonin levels in the intestinal mucosa of rats with colitis induced by dextran sodium sulfate. Histochem Cell Biol 1999; 112: 257–63. Mawe GM, Ciolino A, Foley KF. Inflammatory cytokines decrease serotonin transporter function in colonic epithelial cells. Neurogastroenterol Motil 2005; 17: 613. Chen JJ, Li Z, Pan H, et al. Maintenance of serotonin in the intestinal mucosa and ganglia of mice that lack the highaffinity serotonin transporter: Abnormal intestinal motility and the expression of cation transporters. J Neurosci 2001; 21: 6348–61. Wade PR, Chen J, Jaffe B, Kassem IS, Blakely RD, Gershon MD. Localization and function of a 5-HT transporter in crypt epithelia of the gastrointestinal tract. J Neurosci 1996; 16: 2352–64. Gershon MD. Review article: serotonin receptors and transporters–roles in normal and abnormal gastrointestinal motility. Aliment Pharmacol Ther 2004; 20 (Suppl. 7): 3–14. Costall B, Naylor RJ. 5-HT3 receptors. Curr Drug Targets CNS Neurol Disord 2004; 3: 27–37. Bockaert J, Claeysen S, Compan V, Dumuis A. 5-HT4 receptors. Curr Drug Targets CNS Neurol Disord 2004; 3: 39–51.

52 Galligan JJ. Ligand-gated ion channels in the enteric nervous system. Neurogastroenterol Motil 2002; 14: 611–23. 53 Kozlowski CM, Green A, Grundy D, Boissonade FM, Bountra C. The 5-HT(3) receptor antagonist alosetron inhibits the colorectal distention induced depressor response and spinal c-fos expression in the anaesthetised rat. Gut 2000; 46: 474–80. 54 Liu M, Geddis MS, Wen Y, Setlik W, Gershon MD. Expression and function of 5-HT4 receptors in the mouse enteric nervous system. Am J Physiol Gastrointest Liver Physiol 2005; 289: G1148–63. 55 Pan H, Galligan JJ. 5-HT1A and 5-HT4 receptors mediate inhibition and facilitation of fast synaptic transmission in enteric neurons. Am J Physiol 1994; 266 (Pt 1): G230–8. 56 Galligan JJ, Pan H, Messori E. Signalling mechanism coupled to 5-hydroxytryptamine4 receptor-mediated facilitation of fast synaptic transmission in the guinea-pig ileum myenteric plexus. Neurogastroenterol Motil 2003; 15: 523–9. 57 Grider JR. Desensitization of the peristaltic reflex induced by mucosal stimulation with the selective 5-HT4 agonist, tegaserod. Am J Physiol Gastrointest Liver Physiol 2006; 290: G319–27. 58 Tonini M, Vicini R, Cervio E, et al. 5-HT(7) Receptors modulate peristalsis and accommodation in the guinea pig ileum. Gastroenterology 2005; 129: 1557–66. 59 Liu MT, Rayport S, Jiang Y, Murphy DL, Gershon MD. Expression and function of 5-HT3 receptors in the enteric neurons of mice lacking the serotonin transporter. Am J Physiol Gastrointest Liver Physiol 2002; 283: G1398–411. 60 Tache Y, Martinez V, Million M, Wang L. Stress and the gastrointestinal tract III. Stress-related alterations of gut motor function: role of brain corticotropin-releasing factor receptors. Am J Physiol Gastrointest Liver Physiol 2001; 280: G173–7.

ª 2006 The Authors, Aliment Pharmacol Ther 23, 1067–1076 Journal compilation ª 2006 Blackwell Publishing Ltd