Topic Highlight Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 28, 2015; 21(40): 11353-11361
Published online Oct 28, 2015. doi: 10.3748/wjg.v21.i40.11353
Genetic epidemiology of irritable bowel syndrome
Jasbir Makker, Sridhar Chilimuri, Jonathan N Bella, Department of Medicine, Bronx-Lebanon Hospital Center and Albert Einstein College of Medicine, Bronx, NY 10457, United States
Author contributions: Makker J wrote the manuscript; Chilimuri S and Bella JN is the senior author who critically reviewed and revised the manuscript.
Conflict-of-interest statement: Authors have no conflict of interest to disclose.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Jonathan N Bella, MD, Department of Medicine, Bronx-Lebanon Hospital Center and Albert Einstein College of Medicine, 12th Floor, 1650 Grand Concourse, Bronx, NY 10457, United States. jonnbella@earthlink.net
Telephone: +1-718-5185222 Fax: +1-718-5185585
Received: April 20, 2015
Peer-review started: April 21, 2015
First decision: May 18, 2015
Revised: June 16, 2015
Accepted: August 25, 2015
Article in press: August 25, 2015
Published online: October 28, 2015

Abstract

Irritable bowel syndrome (IBS) is the most common functional gastrointestinal disorder characterized by presence of abdominal pain or discomfort associated with altered bowel habits. It has three main subtypes - constipation predominant IBS (C-IBS), diarrhea predominant IBS (D-IBS) and IBS with mixed features of both diarrhea as well as constipation (M-IBS). Its pathophysiology and underlying mechanisms remain elusive. It is traditionally believed that IBS is a result of multiple factors including hypersensitivity of the bowel, altered bowel motility, inflammation and stress. Initial studies have shown familial aggregation of IBS suggesting shared genetic or environmental factors. Twin studies of IBS from different parts of world have shown higher concordance rates among monozygotic twins than dizygotic twins, and thus suggesting a genetic component to this disorder. Multiple studies have tried to link single-nucleotide polymorphisms (SNPs) to IBS but there is little evidence that these SNPs are functional. Various molecules have been studied and investigated by the researchers. Serotonin, a known neurotransmitter and a local hormone in the enteric nervous system, has been most extensively explored. At this time, the underlying gene pathways, genes and functional variants linked with IBS remain unknown and the promise of genetically-determined risk prediction and personalize medicine remain unfulfilled. However, molecular biological technologies continue to evolve rapidly and genetic investigations offer much promise in the intervention, treatment and prevention of IBS.

Key Words: Irritable bowel syndrome, Single-nucleotide polymorphism, Serotonin, Familial aggregation, Genetics

Core tip: Irritable bowel syndrome (IBS) is believed to result from interplay of several factors including hypersensitivity of the bowel, altered bowel motility, inflammation and stress. Familial aggregation of cases and twin studies underscore the genetic basis of IBS. Different researchers have studied several candidate genes but the evidence so far linking IBS to specific genes is inconsistent and weak. Genome wide association studies that can examine several common genetic variants are needed to design newer drugs and diagnostic methods.



INTRODUCTION

Irritable bowel syndrome (IBS) is the most common functional gastrointestinal disorder with a worldwide prevalence rates ranging between 7%-21%[1]. It is a clinical diagnosis that does not require diagnostic testing unless needed to exclude other diagnostic possibilities. It is characterized by presence of abdominal pain or discomfort associated with altered bowel habits. Rome III criteria that assess the relationship between abdominal pain or discomfort, stool form and change in bowel frequency is the most accepted criteria used in clinical practice for making a clinical diagnosis[2]. Three clinical types have been recognized based on altered bowel motility and the resulting predominant feature - constipation predominant IBS (C-IBS), diarrhea predominant IBS (D-IBS) and IBS with mixed features of both diarrhea as well as constipation (M-IBS).

The genetic basis of IBS has been suggested by familial aggregation and twin studies. Furthermore, over the last decade, several candidate genes have been identified that are potentially linked to IBS. This review will further elaborate on our current understanding of genetic epidemiology of IBS.

GENETIC EPIDEMIOLOGY OF IBS
Familial aggregation of IBS

Mankind is familiar with IBS for more than a century but its pathophysiology and underlying mechanisms remain elusive. It is traditionally believed that IBS is a result of multiple factors including hypersensitivity of the bowel, altered bowel motility, inflammation and stress. Initial studies have shown familial aggregation of IBS suggesting that genes or shared environmental exposures contribute to the development of IBS (Table 1). In one of the earlier studies by Whorwell et al[3] while investigating the non colonic symptoms of IBS, investigators found that 33% of IBS patients had a family member with IBS as compared to only 2% in the control group (P < 0.0001). In an interesting study by Levy et al[4] it was revealed that children of parents who had IBS made 20% more ambulatory care visits than the children of parents without IBS (P = 0.0001). In the same study, researchers also showed that these children who had a parent with IBS made 50% more visits for gastrointestinal symptoms as compared to control group (P = 0.0001). In the study by Locke et al[5], a survey of people residing in Olmsted county of Minnesota with functional gastrointestinal disorders showed higher odds (OR = 2.3; 95%CI: 1.3-3.9) of reporting a relative with gastrointestinal symptoms among these individuals. Studies by Kalantar et al[6] and Saito et al[7] are the other examples where relatives of IBS patients and controls were interviewed and it was determined that statistically higher percentage of IBS patients’ relatives had IBS as compared to control patients’ relatives (17% vs 7%, OR = 2.7; 95%CI: 1.2-6.3 and 37% vs 16%, P = 0.002 respectively). In another study by Saito et al[8] besides showing aggregation of IBS cases among family members, lack of any association in spouses was also shown. In a recent large nationwide case cohort study from Sweden, a higher odds ratio of IBS was found in first, second and third degree relatives of IBS patients (OR for first degree relatives: 1.75-1.90, for second degree relatives: 1.10-1.78 and third degree relatives: 1.11)[9]. These studies further support the concept of shared genes or shared environmental exposures.

Table 1 Familial aggregation studies.
Authors (Year)Number of IBS patientsConclusion
Whorwell et al[3] (1986)100Significant number of IBS patient had another family member with IBS
Levy et al[4] (2000)373Children of parents with IBS made more visits for gastrointestinal symptoms
Locke et al[5] (2000)643People with functional gastrointestinal disorders had higher odds of reporting a relative with similar symptoms
Kalantar et al[6] (2003)181IBS prevalence was higher among IBS patients' relatives
Saito et al[7] (2008)50Statistically higher percentage of IBS patients’ relatives had IBS
Saito et al[8] (2010)47750% of IBS patients had at least another relative with IBS
Waehrens et al[9] (2015)51952Increased risk of IBS among first, second and third degree relatives of IBS patients

Similarly, twin studies have also demonstrated the role of genes in IBS. The first twin study from Australia by Morris-Yates et al[10] and another study involving Swedish twin pairs by Svedberg et al[11] emphasized the genetic basis of IBS. Studies by Levy et al[12] and Bengtson et al[13] involving 281 twin pairs in United States and 3334 twin pairs in Norway respectively showed higher concordance rate for IBS among monozygotic twins than in dizygotic twins (17.2% vs 8.4%, P = 0.03; 22.4% vs 9.1%, P = 0.011 respectively). In the study by Lembo et al[14], 986 twin pairs from Minnesota twin registry were surveyed and their results concur with results of the studies from the other countries and showed the higher rate of IBS among monozygotic twins. Though all these twin studies mentioned above underscore the genetic basis of IBS, the study by Mohammed et al[15] did not come to the same conclusion and found no difference in concordance rates between monozygotic and dizygotic twins.

CANDIDATE GENE STUDIES AND IBS
Serotonin and IBS

Many investigators have focused on specific candidate genes and whether variation in these genes is associated with IBS. Serotonin (5-hydroxytryptamine, 5-HT) is the most widely studied molecule due to its role in brain-gut axis and abundance in the gastrointestinal tract. Majority of body serotonin is present in gastrointestinal tract where it is synthesized mainly by enterochromaffin cells (EC cells) and some of it by myenteric plexus neurons. Serotonin is a known neurotransmitter and a local hormone in the enteric nervous system. It is released from EC cells in response to variety of stimuli and exerts its local paracrine effects through serotonin receptors. There are seven serotonin receptors identified so far named as 5-HT1 to 5-HT7. Out of these seven receptors, 5HT1 to 5-HT4 and 5HT7 play a key role in mediating intestinal responses. The effect of serotonin molecule initiated after its binding to serotonin receptor is eventually terminated by its uptake through a serotonin reuptake transporter (SERT)[16].

There is evidence that patients with IBS have defects in serotonergic signaling. Almost four decades ago, it was discovered that patients with IBS have more number of EC cells as compared to controls[17,18]. Since then numerous studies have explored the relationship between EC cells, serotonin and IBS. Study by Bearcroft et al[19] demonstrated that patients with D-IBS have higher blood levels of serotonin. Similarly studies by Dunlop et al[20] and Atkinson et al[21] have shown higher blood levels of serotonin in D-IBS patients and lower blood levels of serotonin in C- IBS patients. These authors also measured the levels of 5-HIAA (5-hydroxyindoleacetic acid), a 5-HT metabolite, in the rectal mucosal biopsy specimens and blood to obtain 5-HT/5-HIAA ratios as a surrogate marker for serotonin turnover. Based on analysis of 5-HT/5-HIAA ratios in blood and mucosal biopsies, serotonin release defects in C-IBS and serotonin uptake defects in D-IBS were suggested. In the search for an explanation to these differences SERT functions, polymorphism of SERT gene has been extensively investigated.

The SERT gene, also known as solute carrier family 6 member 4 (SLC6A4), was mapped to chromosome 17q11.2 by Ramamoorthy et al[22] in 1993. Polymorphic loci that affect the expression and function of SERT gene have been identified. There is a GC base pair rich repetitive sequence located at 5’ regulatory end of the SERT gene and is labeled as 5-HT transporter linked promoter region (5-HTT LPR). Polymorphism due to deletion or insertion of 44 base pairs in this region resulting in a long (L) and short (S) allele was first discovered by Heils et al[23]. Another common polymorphism of SERT gene results from a variable number of 17 base pair repeats (VNTR: variable number tandem repeats) in the intron 2 of gene[24]. VNTR has 4 alleles described with 9, 10, 11 and 12 repeats.

The evidence on the relationship between different genotypes and their phenotypic expression comes from a study conducted by Lesch et al[25]. Lesch et al[25] found that S/S genotype as compared to other genotypes L/L and S/L had less 5-HT uptake resulting in higher blood levels of 5-HT. Based on studies from Dunlop et al[20] and Atkinson et al[21] where defects in serotonin uptake were suggested in D-IBS, S/S genotype was expected to be associated with D-IBS. Several researchers have explored the association between IBS and SERT gene since then but have come up with conflicting results. In 2002 for the first time, Pata et al[26] demonstrated higher percentage of C-IBS patients to have S/S genotype and D-IBS to have L/S genotype. In the same year Camilleri et al[27] investigated the association between SERT polymorphism and response to Alosetron (5-HT3 receptor antagonist) and found higher response rates to the drug in IBS individuals with L/L genotype. According to the study by Lee et al[28] in the Korean population there was no association between IBS and SERT gene polymorphism. However, another Korean study by Park et al[29] found significantly higher frequency of S/S genotype among patients with D-IBS, contrary to results of study by Pata et al[26] mentioned above. Similarly, in the North American study by Yeo et al[30] S/S genotype was found to be associated with D-IBS female patients but Saito et al[31] found a significantly higher number of S/S genotype with M-IBS patients. In the study involving Indian IBS patients a significant association was found between S/S genotype and C-IBS[32]. In a Chinese study by Li et al[33] allele frequency of the L/L genotype was significantly higher in the C-IBS group. In the same study, researchers also demonstrated poor response to Tegaserod (5HT4 partial selective agonist) associated with L/L genotype.

Another polymorphism pertaining to a single nucleotide polymorphism locus, rs25531 that is located immediately upstream of 5-HTTLPR was described by Kohen et al[34]. It has two - A and G alleles. Hu et al[35] showed that A variant of L allele (designated as LA) yields higher SERT expression as compared to G variant of L allele (designated as LG). This implies that LG (G variant of rs25531 with L allele) actually behaves as the low expressing S allele. With respect to VNTR polymorphism, one study[36] found a significant association between the VNTR polymorphism and IBS but other studies by Yeo et al[30], Pata et al[26] and Li et al[33] did not find any significant association.

To investigate the possible association between IBS subtypes and SERT polymorphism, Van Kerkhoven et al[37] conducted a meta-analysis of eight studies in 2007 that found no significant association between SERT polymorphism and IBS subtypes. Another meta-analysis conducted in 2013 that included more recent studies conducted since the first meta-analysis by Van Kerkhoven et al in 2007, concluded a positive association between SERT polymorphism and C-IBS[38]. In another meta-analysis from 2013 performed by Areeshi et al[39] that included twelve studies comprising 2068 IBS patients, no association was found between SERT polymorphism and IBS overall. However, when the studies were stratified according the country of origin, significant association was found in American and Asian studies. The most recent meta-analysis with the largest sample size, involving 25 studies comprising of 3443 IBS patients found a positive association between SERT polymorphism and IBS but this association was found only in the East Asian population and not in the Caucasian population[40]. One probable explanation for this ethnic difference, as evidenced by this meta-analysis, could be the significantly lower frequency of L allele among the East Asian population as compared to Caucasian controls.

Besides the most widely studied polymorphism involving SERT LPR discussed above, several other polymorphism loci have been explored. These include polymorphisms of gene involving other serotonin receptors - 5-HT2A gene[41-43] and 5-HT3E gene[44] and yet again the studies have yielded either conflicting results or need further validation in other ethnic groups.

Tryptophan hydroxylase (TPH), the rate-limiting enzyme in 5-HT biosynthetic pathway has two isoforms - TPH1 and TPH2 encoded by genes on chromosome 11 and 12 respectively. Jun et al[45] found no association between TPH1 gene single nucleotide polymorphism and risk of developing IBS, however found significant association with severity as well as number of days with diarrhea in IBS patients. TPH2 gene polymorphism was also tested and shown to be associated with reduced risk of having IBS but statistically this difference was barely significant. Similar to this study by Jun et al, no association was found between genotype frequencies and IBS in the study conducted by Grasberger et al[46]. But the CC genotype was found to be more prevalent in IBS-D patients in the study. Researchers that investigated the colonic mucosa levels of TPH1 mRNA in IBS patients found significantly reduced TPH1 mRNA levels in both IBS subtypes[47].

CYTOKINES AND IBS

Cytokines are important mediators of inflammatory and immune responses. Cytokines have an established role in the inflammation of gastrointestinal tract, however the role in IBS is not yet clear. Genes involving various cytokines and inflammatory pathways have been studied. These involve genes for tumor necrosis factor-alpha (TNF-α), Transforming growth factor beta 1 (TGF-β1), interleukin-8 (IL-8) and interleukin-10 (IL-10). Production of these cytokines is under the genetic control and based on polymorphism involving genes coding these cytokines, high and low cytokine producing alleles been identified.

IL-10 gene polymorphism located at position 1082 constituting G to A substitution yields two types of alleles - allele G associated with high production of IL-10 whereas allele A associated with low production of IL-10[48]. Similarly for TNF-α, allele A and for TGF-β1, alleles T and G are identified as high producers[49]. Gonsalkorale et al[50], found significantly less number of high producer alleles of IL-10 in patients with IBS. Subsequently there were studies that highlighted the relationship between IBS and IL-10[51,52] but on the other hand others showed no association among the two[53]. Similarly the studies that explored the role of other cytokines like TNF-α also had conflicting results. Chang et al[52] did not find any positive association between IBS and TNF-α but van der Veek et al[53] on the other hand found that high producer genotypes of TNF-α were more prevalent in patients with IBS. Inconsistent findings of these and other studies led to a meta-analysis by Bashashati et al[54] that comprised of five studies with 529 IBS patients and looked at three cytokines - IL-10, TNF-α and TGF-β1. The meta-analysis found a decreased risk of IBS among high producer genotypes of IL-10 (1082, G/G), positive correlation between TNF (G/A genotype) and Asian IBS patients and failed to find any significant association between IBS and TGF-β1 gene polymorphism. Another more recent and larger meta-analysis by Bashashati et al[55] comprising of nine studies found no difference in blood IL-10 levels and IBS patients but based on gender stratification a significant association was found among men with IBS and lower IL-10 levels. With regards to TNF-α, the meta-analysis found that all IBS subtypes and women had significantly higher blood levels of TNF-α. In another meta-analysis comprising 8 studies with 928 IBS patients, all three known polymorphisms of IL-10 gene - rs1800870 (1082 G/A), rs1800871 (819C/T), and rs1800872 (592A/C) were explored. The meta-analysis concluded that one of the polymorphism was significantly associated with IBS in Caucasians (rs1800870), second (rs1800872) associated with IBS in Asians and third (rs1800871) had no association to IBS[56].

CANNABINOIDS

Δ9-Tetrahydrocannabinol, active ingredient of cannabis, acts through two cannabinoid receptors - CB1 and CB2. While CB1 mediates neurotransmitter release in the peripheral and central neuronal pathways, CB2 is associated with immune functions. CB1 receptors are present throughout the gastrointestinal tract and endocannabinoids mediate their gastrointestinal effects through these receptors. Action of the endocannabinoids is terminated by their uptake and subsequent metabolism by the enzyme fatty acid amide hydrolase (FAAH)[57]. Gene coding for CB1 receptor has been mapped to chromosome 6 and polymorphism involving AAT triplet microsatellite flanking 3’ end has been described in literature[58]. In a Korean study by Park et al[59] a significant association was found between IBS and the AAT triplet genotype with more than 10 repeats. Similar results were replicated by another Chinese study conducted by Jiang et al[60]. These authors also studied another single nucleotide polymorphism (rs324420) involving FAAH gene and found that A/A genotype was present less frequently in patients with IBS as compared to healthy controls but the difference was not statistically significant. In another study by Camilleri et al[61] comprising mainly of Caucasians investigated the association between phenotypic expression (colonic transit and rectal sensation) and genetic variations - AAT polymorphism as well as another polymorphism locus (rs806378) of CB1 receptor gene. The group found no association between AAT repeat polymorphism and phenotypic expression - colonic transit and rectal sensation. However, a significant association between D-IBS and colonic transit as well sensation rating of gas was found.

OTHER MEDIATORS

TNFSF15, also known as TL1a, is a member of tumor necrosis factor super family (TNFSF) of ligands. It binds to death receptor 3 (DR3) and mediates T cell proliferation, secretion of inflammatory cytokines and T cell immune responses. Polymorphism of TNFSF15 gene has been well established to be associated with inflammatory bowel disease. Its association with C-IBS was shown in a study by Zucchelli et al[62], whereas Swan et al[63] found it to be associated with D-IBS.

Cholecystokinin (CCK), a gastrointestinal tract peptide hormone acts through two receptors - CCK1 and CCK2. Genetic polymorphism of CCK1 was found more commonly among C-IBS and M-IBS patients in a study by Park et al[64]. In another study evaluating the effects of a CCK1 antagonist in C-IBS patients, gastric emptying was accelerated but no effect on overall colonic motility was found[65].

Enzyme catechol-o-methyltransferase (COMT) that degrades the catecholamines including adrenaline, noradrenaline and dopamine is another possible molecule related to IBS. Both, COMT gene polymorphism as well as IBS, have been shown to be associated with pain[66] and anxiety[67]. On these grounds the relationship between COMT gene polymorphism and IBS was also explored. Karling et al found that val/val allele of COMT gene encoding high COMT activity enzyme was more common among D-IBS patients. On the contrary, Chinese researchers Wang et al[68] found val/val allele to be protective against IBS, whereas the other allele met/met encoding reduced COMT activity enzyme significantly associated with elderly D-IBS patients. Interestingly in another study by Hall et al[69] investigating the placebo effect, the cases with allele (met/met) were found to be most responsive to placebos.

Single nucleotide polymorphism C825T on gene encoding β-3 subunit of the guanine nucleotide binding protein (GNβ3) was also explored. Studies from United States by Andresen et al[70] and Saito et al[71] found no relationship between this polymorphism and IBS patients. On the contrary, significant relationship among the GNB3 polymorphism and Korean as well Greek IBS patients was shown by Lee et al[72] and Markoutsaki et al[73] respectively. But other Korean study[74] as well as study from China did not show any association of the same polymorphism to IBS[68].

Hypothalamic hormone, corticotropin releasing hormone (CRH), which mediates its effects through two receptors - CRH1 and CRH2 has also been studied. Sato et al[75] explored three single nucleotide polymorphism of CRH1 gene and found significant association with IBS symptoms.

Mediators of autonomic system also have been implicated in pathogenesis of IBS. Association between autonomic dysregulation and IBS has been already shown in other studies[76,77]. α2-adrenergic receptors that are present on both presynaptic as well as postsynaptic locations and have three subtypes - 2A, 2B and 2C, are encoded by gene on chromosome 10. A 12 base pair deletion polymorphism of adrenergic receptor 2C (denoted α2C Del 322-325) and another single nucleotide polymorphism involving adrenergic receptor 2A gene (C1291G) were studied by Kim et al[78] Both the polymorphisms were found to have significantly higher odds (odds ratio 2.48 and 1.66 respectively) to be associated with C-IBS. In contrast, Sikander et al[79] in Indian IBS patients and Choi et al[80] in Korean IBS patients found an association between adrenergic receptor 2A gene polymorphism and D-IBS. Whereas in the study by Camilleri et al[81] no association was found between adrenergic receptor 2A gene polymorphism and IBS. Cholinergic muscarinic receptor type 3 gene polymorphism (rs3738435) was investigated by Onodera et al[82] Though no association was found between the polymorphism and different IBS subtypes, researchers found the polymorphism to be associated with the duration of disease.

So far the expedition to find a genetic basis for IBS has included wide array of studies ranging from epidemiological familial aggregation studies to gene polymorphism testing studies. Though these studies in several ways have shown the possible hereditary component of IBS but most of the studies have provided only weak evidence between the two. In the last decade gene search studies have focused more on detecting gene polymorphisms. Several possible gene polymorphisms have been shown to be associated with IBS as a result of growing interest in gene polymorphisms. Even though the P values of these small studies show strength of association between the polymorphism tested and IBS but it cannot be taken as equivalent to genetic basis of IBS.

Genome-wide association studies

In addition to candidate gene-based association studies, association studies have also been conducted in a systematic, genome-wide manner. Genome wide association studies (GWAS) utilize high throughput genotyping techniques to assay hundreds of thousands of the most common form of genetic variation, the single-nucleotide polymorphism (SNP) and relate genotypes at these variants to the phenotypes of diseases and other traits[83]. This approach permits the interrogation of much of the common variation in the entire genome in thousands (or even hundreds of thousands) of unrelated individuals, achieving a higher positional resolution than is reasonably possible. In the first and so far the only GWAS by Weronica et al linkage was shown between IBS and two genes mapped to locus 7p22.1. The two genes mapped to this locus were KDEL receptor 2 gene (KDELR2) and glutamate receptor ionotropic delta 2 interacting protein (GRID2IP)[84,85].

FUTURE DIRECTIONS

The field of genetics exploring the underlying potential IBS genes is rapidly evolving. However, huge gaps in our understanding of this complex disorder remain and need further study. Genetic research efforts that are focused on studying the candidate gene polymorphism have so far provided the weak evidence at the best. Gene linkage studies and genome wide association studies are needed to understand the deficiencies in our current knowledge. In the future studies, it will be also of importance to explore how the environment influences these newly discovered genes.

IBS is a complex functional gastrointestinal disorder that poses a great challenge to both, the affected patients and the physicians treating the ones affected. Historically, underlying pathophysiology is believed to be interplay of hypersensitive gut with key contributions from psychological, social and environmental factors. Genetic basis has also been suggested by familial aggregation of cases as well as high concordance rate among monozygotic twins than dizygotic twins. Gene hunt efforts, so far, have explored several potential candidate genes (Table 2) and shown association between polymorphic gene loci and different IBS phenotypes. While studies so far have been less than emphatic in their results due to small sample sizes, varying criteria used to diagnose IBS, heterogeneity of methods used and ethnic differences of the participants tested, there is convincing evidence that a proportion of IBS are due to additive genetic effects. As an alternative to current gene specific candidate driven approach, larger and non-candidate gene driven studies in the form of GWAS are needed to understand this common and complex disorder. Such genome wide studies may provide much needed insight into the genetic susceptibility of IBS and could aid in development of novel therapeutic strategies to diagnose, treat and prevent this disorder.

Table 2 Candidate genes associated with irritable bowel syndrome.
Gene studiedPolymorphism
Serotonin or 5-HT reuptake transporter (SERT) gene –SLC6A45-HTT LPR – S and L alleles[23]
rs25531 – A and G alleles[34]
VNTR - alleles with 9, 10, 11 and 12 repeats[24]
5HT2A1438 (G/A) and 102 (C/T)[41]
5HT3Ac.-42C>T[85]
5HT3Ers62625044 - 76 (G/A)[44]
Tryptophan hydroxylase 1 (TPH1)rs4537731 – 6526 (A/G)[45]
rs684302
rs21105
rs1800532 – 218 (A/C)
rs1799913 – 779 (A/C)
Tryptophan hydroxylase 2 (TPH2)rs4570625 – 709 (G/T)[45]
Tumor necrosis factor–alpha (TNF-α)308 (G/A)[53]
Transforming growth factor beta 1 (TGF-β1)869 (T/C)[54]
915 (G/C)[54]
Interleukin-10 (IL-10)rs1800870 - 1082 (G/A)[50]
rs1800871 - 819 (C/T)[53]
rs1800872 - 592 (A/C)[56]
Cannabinoid receptor 1 (CB1)AAT triplets[59]
rs324420[60]
rs806378[61]
Tumor necrosis factor super family 15 (TNFSF15)rs4263839[62]
Cholecystokinin 1(CCK1)779 (T/C)[64]
Catechol-o-methyltransferase (COMT)rs 4680 - 158 (Val/Met)[68]
Guanine nucleotide binding protein (GNβ3)rs 5443 - 825 (C/T)[68]
Corticotropin releasing hormone receptor 1(CRH1)rs110402[75]
rs242924
rs7209436
α2-adrenergic receptor 2A1291 (C/G)[80]
α2-adrenergic receptor 2CDel 322-325[78]
Cholinergic muscarinic receptor type 3rs3738435[82]
Footnotes

P- Reviewer: Amarapurkar DN, Chiarioni G, Ocker M S- Editor: Ma YJ L- Editor: A E- Editor: Ma S

References
1.  Lovell RM, Ford AC. Global prevalence of and risk factors for irritable bowel syndrome: a meta-analysis. Clin Gastroenterol Hepatol. 2012;10:712-721.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1251]  [Cited by in F6Publishing: 1292]  [Article Influence: 107.7]  [Reference Citation Analysis (1)]
2.  Longstreth GF, Thompson WG, Chey WD, Houghton LA, Mearin F, Spiller RC. Functional bowel disorders. Gastroenterology. 2006;130:1480-1491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3413]  [Cited by in F6Publishing: 3271]  [Article Influence: 181.7]  [Reference Citation Analysis (1)]
3.  Whorwell PJ, McCallum M, Creed FH, Roberts CT. Non-colonic features of irritable bowel syndrome. Gut. 1986;27:37-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 381]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
4.  Levy RL, Whitehead WE, Von Korff MR, Feld AD. Intergenerational transmission of gastrointestinal illness behavior. Am J Gastroenterol. 2000;95:451-456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 147]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
5.  Locke GR, Zinsmeister AR, Talley NJ, Fett SL, Melton LJ. Familial association in adults with functional gastrointestinal disorders. Mayo Clin Proc. 2000;75:907-912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 194]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
6.  Kalantar JS, Locke GR, Zinsmeister AR, Beighley CM, Talley NJ. Familial aggregation of irritable bowel syndrome: a prospective study. Gut. 2003;52:1703-1707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 126]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
7.  Saito YA, Zimmerman JM, Harmsen WS, De Andrade M, Locke GR, Petersen GM, Talley NJ. Irritable bowel syndrome aggregates strongly in families: a family-based case-control study. Neurogastroenterol Motil. 2008;20:790-797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 27]  [Reference Citation Analysis (0)]
8.  Saito YA, Petersen GM, Larson JJ, Atkinson EJ, Fridley BL, de Andrade M, Locke GR, Zimmerman JM, Almazar-Elder AE, Talley NJ. Familial aggregation of irritable bowel syndrome: a family case-control study. Am J Gastroenterol. 2010;105:833-841.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 75]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
9.  Waehrens R, Ohlsson H, Sundquist J, Sundquist K, Zöller B. Risk of irritable bowel syndrome in first-degree, second-degree and third-degree relatives of affected individuals: a nationwide family study in Sweden. Gut. 2015;64:215-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 32]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
10.  Morris-Yates A, Talley NJ, Boyce PM, Nandurkar S, Andrews G. Evidence of a genetic contribution to functional bowel disorder. Am J Gastroenterol. 1998;93:1311-1317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 189]  [Article Influence: 7.3]  [Reference Citation Analysis (1)]
11.  Svedberg P, Johansson S, Wallander MA, Hamelin B, Pedersen NL. Extra-intestinal manifestations associated with irritable bowel syndrome: a twin study. Aliment Pharmacol Ther. 2002;16:975-983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
12.  Levy RL, Jones KR, Whitehead WE, Feld SI, Talley NJ, Corey LA. Irritable bowel syndrome in twins: heredity and social learning both contribute to etiology. Gastroenterology. 2001;121:799-804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 299]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
13.  Bengtson MB, Rønning T, Vatn MH, Harris JR. Irritable bowel syndrome in twins: genes and environment. Gut. 2006;55:1754-1759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 128]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
14.  Lembo A, Zaman M, Jones M, Talley NJ. Influence of genetics on irritable bowel syndrome, gastro-oesophageal reflux and dyspepsia: a twin study. Aliment Pharmacol Ther. 2007;25:1343-1350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 80]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
15.  Mohammed I, Cherkas LF, Riley SA, Spector TD, Trudgill NJ. Genetic influences in irritable bowel syndrome: a twin study. Am J Gastroenterol. 2005;100:1340-1344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 76]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
16.  Sikander A, Rana SV, Prasad KK. Role of serotonin in gastrointestinal motility and irritable bowel syndrome. Clin Chim Acta. 2009;403:47-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 170]  [Article Influence: 11.3]  [Reference Citation Analysis (1)]
17.  Ahonen A, Kyösola K, Penttilä O. Enterochromaffin cells in macrophages in ulcerative colitis and irritable colon. Ann Clin Res. 1976;8:1-7.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Kyösola K, Penttilä O, Salaspuro M. Rectal mucosal adrenergic innervation and enterochromaffin cells in ulcerative colitis and irritable colon. Scand J Gastroenterol. 1977;12:363-367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 76]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
19.  Bearcroft CP, Perrett D, Farthing MJ. Postprandial plasma 5-hydroxytryptamine in diarrhoea predominant irritable bowel syndrome: a pilot study. Gut. 1998;42:42-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 243]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
20.  Dunlop SP, Coleman NS, Blackshaw E, Perkins AC, Singh G, Marsden CA, Spiller RC. Abnormalities of 5-hydroxytryptamine metabolism in irritable bowel syndrome. Clin Gastroenterol Hepatol. 2005;3:349-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 254]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
21.  Atkinson W, Lockhart S, Whorwell PJ, Keevil B, Houghton LA. Altered 5-hydroxytryptamine signaling in patients with constipation- and diarrhea-predominant irritable bowel syndrome. Gastroenterology. 2006;130:34-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 251]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
22.  Ramamoorthy S, Bauman AL, Moore KR, Han H, Yang-Feng T, Chang AS, Ganapathy V, Blakely RD. Antidepressant- and cocaine-sensitive human serotonin transporter: molecular cloning, expression, and chromosomal localization. Proc Natl Acad Sci USA. 1993;90:2542-2546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 587]  [Cited by in F6Publishing: 603]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
23.  Heils A, Teufel A, Petri S, Stöber G, Riederer P, Bengel D, Lesch KP. Allelic variation of human serotonin transporter gene expression. J Neurochem. 1996;66:2621-2624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1523]  [Cited by in F6Publishing: 1501]  [Article Influence: 53.6]  [Reference Citation Analysis (0)]
24.  Lesch KP, Balling U, Gross J, Strauss K, Wolozin BL, Murphy DL, Riederer P. Organization of the human serotonin transporter gene. J Neural Transm Gen Sect. 1994;95:157-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 426]  [Cited by in F6Publishing: 412]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
25.  Lesch KP, Bengel D, Heils A, Sabol SZ, Greenberg BD, Petri S, Benjamin J, Müller CR, Hamer DH, Murphy DL. Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region. Science. 1996;274:1527-1531.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3630]  [Cited by in F6Publishing: 3400]  [Article Influence: 121.4]  [Reference Citation Analysis (0)]
26.  Pata C, Erdal ME, Derici E, Yazar A, Kanik A, Ulu O. Serotonin transporter gene polymorphism in irritable bowel syndrome. Am J Gastroenterol. 2002;97:1780-1784.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 113]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
27.  Camilleri M, Atanasova E, Carlson PJ, Ahmad U, Kim HJ, Viramontes BE, McKinzie S, Urrutia R. Serotonin-transporter polymorphism pharmacogenetics in diarrhea-predominant irritable bowel syndrome. Gastroenterology. 2002;123:425-432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 213]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
28.  Lee DY, Park H, Kim WH, Lee SI, Seo YJ, Choi YC. [Serotonin transporter gene polymorphism in healthy adults and patients with irritable bowel syndrome]. Korean J Gastroenterol. 2004;43:18-22.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Park JM, Choi MG, Park JA, Oh JH, Cho YK, Lee IS, Kim SW, Choi KY, Chung IS. Serotonin transporter gene polymorphism and irritable bowel syndrome. Neurogastroenterol Motil. 2006;18:995-1000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 68]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
30.  Yeo A, Boyd P, Lumsden S, Saunders T, Handley A, Stubbins M, Knaggs A, Asquith S, Taylor I, Bahari B. Association between a functional polymorphism in the serotonin transporter gene and diarrhoea predominant irritable bowel syndrome in women. Gut. 2004;53:1452-1458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 202]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
31.  Saito YA, Locke GR, Zimmerman JM, Holtmann G, Slusser JP, de Andrade M, Petersen GM, Talley NJ. A genetic association study of 5-HTT LPR and GNbeta3 C825T polymorphisms with irritable bowel syndrome. Neurogastroenterol Motil. 2007;19:465-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 55]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
32.  Sikander A, Rana SV, Sinha SK, Prasad KK, Arora SK, Sharma SK, Singh K. Serotonin transporter promoter variant: Analysis in Indian IBS patients and control population. J Clin Gastroenterol. 2009;43:957-961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 38]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
33.  Li Y, Nie Y, Xie J, Tang W, Liang P, Sha W, Yang H, Zhou Y. The association of serotonin transporter genetic polymorphisms and irritable bowel syndrome and its influence on tegaserod treatment in Chinese patients. Dig Dis Sci. 2007;52:2942-2949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 58]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
34.  Kohen R, Jarrett ME, Cain KC, Jun SE, Navaja GP, Symonds S, Heitkemper MM. The serotonin transporter polymorphism rs25531 is associated with irritable bowel syndrome. Dig Dis Sci. 2009;54:2663-2670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 37]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
35.  Hu XZ, Lipsky RH, Zhu G, Akhtar LA, Taubman J, Greenberg BD, Xu K, Arnold PD, Richter MA, Kennedy JL. Serotonin transporter promoter gain-of-function genotypes are linked to obsessive-compulsive disorder. Am J Hum Genet. 2006;78:815-826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 890]  [Cited by in F6Publishing: 917]  [Article Influence: 50.9]  [Reference Citation Analysis (0)]
36.  Wang BM, Wang YM, Zhang WM, Zhang QY, Liu WT, Jiang K, Zhang J. [Serotonin transporter gene polymorphism in irritable bowel syndrome]. Zhonghua Nei Ke Zazhi. 2004;43:439-441.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Van Kerkhoven LA, Laheij RJ, Jansen JB. Meta-analysis: a functional polymorphism in the gene encoding for activity of the serotonin transporter protein is not associated with the irritable bowel syndrome. Aliment Pharmacol Ther. 2007;26:979-986.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 67]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
38.  Dai C, Zheng CQ, Jiang M. Letter: serotonin transporter gene polymorphisms and the irritable bowel syndrome. Aliment Pharmacol Ther. 2013;37:657-658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
39.  Areeshi MY, Haque S, Panda AK, Mandal RK. A serotonin transporter gene (SLC6A4) polymorphism is associated with reduced risk of irritable bowel syndrome in American and Asian population: a meta-analysis. PLoS One. 2013;8:e75567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
40.  Zhang ZF, Duan ZJ, Wang LX, Yang D, Zhao G, Zhang L. The serotonin transporter gene polymorphism (5-HTTLPR) and irritable bowel syndrome: a meta-analysis of 25 studies. BMC Gastroenterol. 2014;14:23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 50]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
41.  Pata C, Erdal E, Yazc K, Camdeviren H, Ozkaya M, Ulu O. Association of the -1438 G/A and 102 T/C polymorphism of the 5-Ht2A receptor gene with irritable bowel syndrome 5-Ht2A gene polymorphism in irritable bowel syndrome. J Clin Gastroenterol. 2004;38:561-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 44]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
42.  Villani AC, Lemire M, Thabane M, Belisle A, Geneau G, Garg AX, Clark WF, Moayyedi P, Collins SM, Franchimont D. Genetic risk factors for post-infectious irritable bowel syndrome following a waterborne outbreak of gastroenteritis. Gastroenterology. 2010;138:1502-1513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 186]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
43.  Markoutsaki T, Karantanos T, Gazouli M, Anagnou NP, Karamanolis DG. 5-HT2A receptor gene polymorphisms and irritable bowel syndrome. J Clin Gastroenterol. 2011;45:514-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 14]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
44.  Kapeller J, Houghton LA, Mönnikes H, Walstab J, Möller D, Bönisch H, Burwinkel B, Autschbach F, Funke B, Lasitschka F. First evidence for an association of a functional variant in the microRNA-510 target site of the serotonin receptor-type 3E gene with diarrhea predominant irritable bowel syndrome. Hum Mol Genet. 2008;17:2967-2977.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 143]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
45.  Jun S, Kohen R, Cain KC, Jarrett ME, Heitkemper MM. Associations of tryptophan hydroxylase gene polymorphisms with irritable bowel syndrome. Neurogastroenterol Motil. 2011;23:233-29, e116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 27]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
46.  Grasberger H, Chang L, Shih W, Presson AP, Sayuk GS, Newberry RD, Karagiannides I, Pothoulakis C, Mayer E, Merchant JL. Identification of a functional TPH1 polymorphism associated with irritable bowel syndrome bowel habit subtypes. Am J Gastroenterol. 2013;108:1766-1774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
47.  Coates MD, Mahoney CR, Linden DR, Sampson JE, Chen J, Blaszyk H, Crowell MD, Sharkey KA, Gershon MD, Mawe GM. Molecular defects in mucosal serotonin content and decreased serotonin reuptake transporter in ulcerative colitis and irritable bowel syndrome. Gastroenterology. 2004;126:1657-1664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 554]  [Cited by in F6Publishing: 551]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
48.  Turner DM, Williams DM, Sankaran D, Lazarus M, Sinnott PJ, Hutchinson IV. An investigation of polymorphism in the interleukin-10 gene promoter. Eur J Immunogenet. 1997;24:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1254]  [Cited by in F6Publishing: 1327]  [Article Influence: 49.1]  [Reference Citation Analysis (0)]
49.  Perrey C, Pravica V, Sinnott PJ, Hutchinson IV. Genotyping for polymorphisms in interferon-gamma, interleukin-10, transforming growth factor-beta 1 and tumour necrosis factor-alpha genes: a technical report. Transpl Immunol. 1998;6:193-197.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Gonsalkorale WM, Perrey C, Pravica V, Whorwell PJ, Hutchinson IV. Interleukin 10 genotypes in irritable bowel syndrome: evidence for an inflammatory component? Gut. 2003;52:91-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 217]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
51.  O'Mahony L, McCarthy J, Kelly P, Hurley G, Luo F, Chen K, O’Sullivan GC, Kiely B, Collins JK, Shanahan F. Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology. 2005;128:541-551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1003]  [Cited by in F6Publishing: 912]  [Article Influence: 48.0]  [Reference Citation Analysis (0)]
52.  Chang L, Adeyemo M, Karagiannides I, Videlock EJ, Bowe C, Shih W, Presson AP, Yuan PQ, Cortina G, Gong H. Serum and colonic mucosal immune markers in irritable bowel syndrome. Am J Gastroenterol. 2012;107:262-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 108]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
53.  van der Veek PP, van den Berg M, de Kroon YE, Verspaget HW, Masclee AA. Role of tumor necrosis factor-alpha and interleukin-10 gene polymorphisms in irritable bowel syndrome. Am J Gastroenterol. 2005;100:2510-2516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 145]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
54.  Bashashati M, Rezaei N, Bashashati H, Shafieyoun A, Daryani NE, Sharkey KA, Storr M. Cytokine gene polymorphisms are associated with irritable bowel syndrome: a systematic review and meta-analysis. Neurogastroenterol Motil. 2012;24:1102-e566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
55.  Bashashati M, Rezaei N, Shafieyoun A, McKernan DP, Chang L, Öhman L, Quigley EM, Schmulson M, Sharkey KA, Simrén M. Cytokine imbalance in irritable bowel syndrome: a systematic review and meta-analysis. Neurogastroenterol Motil. 2014;26:1036-1048.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 114]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
56.  Qin SY, Jiang HX, Lu DH, Zhou Y. Association of interleukin-10 polymorphisms with risk of irritable bowel syndrome: a meta-analysis. World J Gastroenterol. 2013;19:9472-9480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
57.  Coutts AA, Izzo AA. The gastrointestinal pharmacology of cannabinoids: an update. Curr Opin Pharmacol. 2004;4:572-579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 71]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
58.  Dawson E. Identification of a highly polymorphic triplet repeat marker for the brain cannabinoid receptor gene: Use in linkage and association studies of schizophrenia. Schizophr Res. 1995;15:37.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Park JM, Choi MG, Cho YK, Lee IS, Kim SW, Choi KY, Chung IS. Cannabinoid receptor 1 gene polymorphism and irritable bowel syndrome in the Korean population: a hypothesis-generating study. J Clin Gastroenterol. 2011;45:45-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 33]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
60.  Jiang Y, Nie Y, Li Y, Zhang L. Association of cannabinoid type 1 receptor and fatty acid amide hydrolase genetic polymorphisms in Chinese patients with irritable bowel syndrome. J Gastroenterol Hepatol. 2014;29:1186-1191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
61.  Camilleri M, Kolar GJ, Vazquez-Roque MI, Carlson P, Burton DD, Zinsmeister AR. Cannabinoid receptor 1 gene and irritable bowel syndrome: phenotype and quantitative traits. Am J Physiol Gastrointest Liver Physiol. 2013;304:G553-G560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
62.  Zucchelli M, Camilleri M, Andreasson AN, Bresso F, Dlugosz A, Halfvarson J, Törkvist L, Schmidt PT, Karling P, Ohlsson B. Association of TNFSF15 polymorphism with irritable bowel syndrome. Gut. 2011;60:1671-1677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 101]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
63.  Swan C, Duroudier NP, Campbell E, Zaitoun A, Hastings M, Dukes GE, Cox J, Kelly FM, Wilde J, Lennon MG. Identifying and testing candidate genetic polymorphisms in the irritable bowel syndrome (IBS): association with TNFSF15 and TNFα. Gut. 2013;62:985-994.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 125]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
64.  Park SY, Rew JS, Lee SM, Ki HS, Lee KR, Cheo JH, Kim HI, Noh DY, Joo YE, Kim HS. Association of CCK(1) Receptor Gene Polymorphisms and Irritable Bowel Syndrome in Korean. J Neurogastroenterol Motil. 2010;16:71-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
65.  Cremonini F, Camilleri M, McKinzie S, Carlson P, Camilleri CE, Burton D, Thomforde G, Urrutia R, Zinsmeister AR. Effect of CCK-1 antagonist, dexloxiglumide, in female patients with irritable bowel syndrome: a pharmacodynamic and pharmacogenomic study. Am J Gastroenterol. 2005;100:652-663.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 82]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
66.  Loggia ML, Jensen K, Gollub RL, Wasan AD, Edwards RR, Kong J. The catechol-O-methyltransferase (COMT) val158met polymorphism affects brain responses to repeated painful stimuli. PLoS One. 2011;6:e27764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 44]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
67.  Domschke K, Deckert J, O’donovan MC, Glatt SJ. Meta-analysis of COMT val158met in panic disorder: ethnic heterogeneity and gender specificity. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:667-673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 119]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
68.  Wang Y, Wu Z, Qiao H, Zhang Y. A genetic association study of single nucleotide polymorphisms in GNβ3 and COMT in elderly patients with irritable bowel syndrome. Med Sci Monit. 2014;20:1246-1254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
69.  Hall KT, Lembo AJ, Kirsch I, Ziogas DC, Douaiher J, Jensen KB, Conboy LA, Kelley JM, Kokkotou E, Kaptchuk TJ. Catechol-O-methyltransferase val158met polymorphism predicts placebo effect in irritable bowel syndrome. PLoS One. 2012;7:e48135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 151]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
70.  Andresen V, Camilleri M, Kim HJ, Stephens DA, Carlson PJ, Talley NJ, Saito YA, Urrutia R, Zinsmeister AR. Is there an association between GNbeta3-C825T genotype and lower functional gastrointestinal disorders? Gastroenterology. 2006;130:1985-1994.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 52]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
71.  Saito YA, Larson JJ, Atkinson EJ, Ryu E, Almazar AE, Petersen GM, Talley NJ. The role of 5-HTT LPR and GNβ3 825C>T polymorphisms and gene-environment interactions in irritable bowel syndrome (IBS). Dig Dis Sci. 2012;57:2650-2657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 21]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
72.  Lee HJ, Lee SY, Choi JE, Kim JH, Sung IK, Park HS, Jin CJ. G protein beta3 subunit, interleukin-10, and tumor necrosis factor-alpha gene polymorphisms in Koreans with irritable bowel syndrome. Neurogastroenterol Motil. 2010;22:758-763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 35]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
73.  Markoutsaki T, Karantanos T, Gazouli M, Anagnou NP, Ladas SD, Karamanolis DG. Serotonin transporter and G protein beta 3 subunit gene polymorphisms in Greeks with irritable bowel syndrome. Dig Dis Sci. 2011;56:3276-3280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
74.  Kim HG, Lee KJ, Lim SG, Jung JY, Cho SW. G-Protein Beta3 Subunit C825T Polymorphism in Patients With Overlap Syndrome of Functional Dyspepsia and Irritable Bowel Syndrome. J Neurogastroenterol Motil. 2012;18:205-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
75.  Sato N, Suzuki N, Sasaki A, Aizawa E, Obayashi T, Kanazawa M, Mizuno T, Kano M, Aoki M, Fukudo S. Corticotropin-releasing hormone receptor 1 gene variants in irritable bowel syndrome. PLoS One. 2012;7:e42450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 23]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
76.  Gupta V, Sheffield D, Verne GN. Evidence for autonomic dysregulation in the irritable bowel syndrome. Dig Dis Sci. 2002;47:1716-1722.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.0]  [Reference Citation Analysis (0)]
77.  Salvioli B, Pellegatta G, Malacarne M, Pace F, Malesci A, Pagani M, Lucini D. Autonomic nervous system dysregulation in irritable bowel syndrome. Neurogastroenterol Motil. 2015;27:423-430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 48]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
78.  Kim HJ, Camilleri M, Carlson PJ, Cremonini F, Ferber I, Stephens D, McKinzie S, Zinsmeister AR, Urrutia R. Association of distinct alpha(2) adrenoceptor and serotonin transporter polymorphisms with constipation and somatic symptoms in functional gastrointestinal disorders. Gut. 2004;53:829-837.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Sikander A, Rana SV, Sharma SK, Sinha SK, Arora SK, Prasad KK, Singh K. Association of alpha 2A adrenergic receptor gene (ADRAlpha2A) polymorphism with irritable bowel syndrome, microscopic and ulcerative colitis. Clin Chim Acta. 2010;411:59-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
80.  Choi YJ, Hwang SW, Kim N, Park JH, Oh JC, Lee DH. Association Between SLC6A4 Serotonin Transporter Gene Lainked Polymorphic Region and ADRA2A -1291C>G and Irritable Bowel Syndrome in Korea. J Neurogastroenterol Motil. 2014;20:388-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
81.  Camilleri M, Busciglio I, Carlson P, McKinzie S, Burton D, Baxter K, Ryks M, Zinsmeister AR. Candidate genes and sensory functions in health and irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol. 2008;295:G219-G225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 53]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
82.  Onodera S, Chiba T, Sugai T, Habano W. A genetic association between ß3-aderenoceptor and cholinergic receptor muscarinic 3 polymorphisms in irritable bowel syndrome. Hepatogastroenterology. 2011;58:1474-1478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
83.  D'Amato M. Genes and functional GI disorders: from casual to causal relationship. Neurogastroenterol Motil. 2013;25:638-649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
84.  Ek WE, Reznichenko A, Ripke S, Niesler B, Zucchelli M, Rivera NV, Schmidt PT, Pedersen NL, Magnusson P, Talley NJ. Exploring the genetics of irritable bowel syndrome: a GWA study in the general population and replication in multinational case-control cohorts. Gut. 2014;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 87]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
85.  Kilpatrick LA, Labus JS, Coveleskie K, Hammer C, Rappold G, Tillisch K, Bueller JA, Suyenobu B, Jarcho JM, McRoberts JA. The HTR3A polymorphism c. -42C>T is associated with amygdala responsiveness in patients with irritable bowel syndrome. Gastroenterology. 2011;140:1943-1951.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 69]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]