Review Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Sep 7, 2013; 19(33): 5402-5420
Published online Sep 7, 2013. doi: 10.3748/wjg.v19.i33.5402
What does irritable bowel syndrome share with non-alcoholic fatty liver disease?
Antonella Scalera, Matteo Nicola Dario Di Minno, Giovanni Tarantino, Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, 80131 Naples, Italy
Giovanni Tarantino, INT “Fondazione Pascale” - Cancer Research Center of Mercogliano, 83013 Mercogliano (Av), Italy
Author contributions: Tarantino G designed research and analyzed literature data; Di Minno MND critically reviewed the manuscript; Scalera A wrote the paper.
Correspondence to: Giovanni Tarantino, MD, Professor, Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Via Sergio Pansini, 5, 80131 Naples, Italy. tarantin@unina.it
Telephone: +39-81-7462024 Fax: +39-81-5466152
Received: February 25, 2013
Revised: June 5, 2013
Accepted: July 23, 2013
Published online: September 7, 2013

Abstract

Non-alcoholic fatty liver disease (NAFLD) and irritable bowel syndrome (IBS) are two very common diseases in the general population. To date, there are no studies that highlight a direct link between NAFLD and IBS, but some recent reports have found an interesting correlation between obesity and IBS. A systematic PubMed database search was conducted highlighting that common mechanisms are involved in many of the local and systemic manifestations of NAFLD, leading to an increased cardiovascular risk, and IBS, leading to microbial dysbiosis, impaired intestinal barrier and altered intestinal motility. It is not known when considering local and systemic inflammation/immune system activation, which one has greater importance in NAFLD and IBS pathogenesis. Also, the nervous system is implicated. In fact, inflammation participates in the development of mood disorders, such as anxiety and depression, characteristics of obesity and consequently of NAFLD and, on the other hand, in intestinal hypersensitivity and dysmotility.

Key Words: Non-alcoholic fatty liver disease, Irritable bowel syndrome, Low grade chronic inflammation, Cytokines

Core tip: The link between non-alcoholic fatty liver disease (NAFLD) and irritable bowel syndrome (IBS) should be carefully evaluated in future research, representing an intriguing field of investigation. A better understanding of the role of systemic inflammation and activation of the immune system may be necessary to clarify obscure points of NAFLD and IBS pathogenesis, and therefore it can be helpful in the development of new therapies.



INTRODUCTION

Fat accumulation in the liver in the absence of specific causes of hepatic steatosis, such as alcohol consumption, with or without liver inflammation and its consequences, is described as non-alcoholic fatty liver disease (NAFLD)[1]. To date, there are no studies that highlight the link between NAFLD and irritable bowel syndrome (IBS), but some recent reports have found an interesting correlation between obesity and IBS. A relationship between body mass index (BMI) and IBS-like symptoms seems to exist[2,3]. Moreover, in IBS subjects a high BMI is associated with significantly faster colonic and recto-sigmoid transit and high stool frequency[4]. Cremonini et al[5] have compared obese binge eaters and non binge eaters to healthy controls and have evidenced that obese subjects more frequently have constipation, diarrhea, straining and flatus regardless of the eating disorder, and that obese binge eaters are characterized by more recurrent upper and lower gastrointestinal symptoms. Clements et al[6] have highlighted that obese patients experience more severe gastrointestinal symptoms than healthy controls, and that after laparoscopic Roux-en-Y gastric bypass they have increased abdominal pain, gastroesophageal reflux disease, sleep disturbance and IBS symptoms. The development of small intestinal bacterial overgrowth (SIBO) may explicate, perhaps partially, the incidence of IBS symptoms in obese subjects with previous abdominal surgery, and in this case the bacterial overgrowth may be the consequence of changes in the function and in the morphological structure of the gut[7].

NAFLD pathogenesis is strictly allied to metabolic syndrome, insulin resistance and obesity[8,9] but inflammation plays an equally important role. Day et al[10] have developed in 1998 the classical “two-hit” theory: liver fat accumulation is the “first-hit”, linked to obesity, insulin resistance and metabolic syndrome, while the “second-hit” is activated by hepatic inflammation, together with oxidative stress and endotoxemia, which have a key role in the progression to non-alcoholic steatohepatitis (NASH) and, over time, to fibrosis, until the development of cirrhosis[11].

Actually, this classic view has been revised, because a frank distinction between “first-hit” and “second-hit” is not easy to make, leading to the “multiple-hit theory”[12]. Recent studies have shown that, independently from fat accumulation in the liver, obesity systemically leads to activation of the immune system and low-chronic inflammation from the first stage of the disease[13].

Obesity and hepatic fat accumulation are hypothetically implicated in IBS genesis or development. Moreover, an initial correlation between IBS and NAFLD can be suggested by some interesting data. In the pathogenesis of NAFLD and NASH, there is a strong involvement of the gastrointestinal system, as evidenced by many studies on the so-called “gut-liver axis”, aiming to comprehend the role of gut microbiota, SIBO and intestinal permeability dysfunction[14]. On the other hand, hepatic fat accumulation and hepatic inflammation in NAFLD subjects[15] and gastrointestinal symptoms in IBS subjects[16] both improve after therapy with probiotics.

IBS, one of the most common gastrointestinal disorders with an estimated prevalence of 7%-10% worldwide[17], is characterized by abdominal pain/discomfort, changes in bowel habits and no association with organic cause. Despite the fact that in IBS subjects macroscopically evident pathological lesions at colonoscopy have not been found, molecular biology and in-depth histological investigations have revealed the activation of the immune system. A key piece of evidence is that the exposure of rodent[18-20] or human[21] tissues or cell cultures[22] to mucosal or luminal mediators from IBS subjects leads to impaired nervous stimulation or intestinal barrier damage. A proportion of patients develop IBS symptoms after infectious gastroenteritis, or in a remission state from microscopic colitis, Crohn’s disease and ulcerative colitis, or on a gluten-free diet for celiac disease[23].

Inflammation and immune system activation may be the mechanisms linking two apparently very different diseases, and the purpose of our review is to collect key evidence supporting their relationship and therefore to explain the pathophysiological link between the intestine and the liver, which is exquisitely firstly anatomical and consequently also functional.

IMMUNITY IN NAFLD

A low-grade chronic inflammation underlies all NAFLD entities/stages and can develop and promote the liver damage[13].

Innate and adaptive immune pathways are activated in obesity and many findings show that adipose tissue inflammation exacerbates hepatic steatosis and promotes non-alcoholic steatohepatitis (NASH). Obese individuals more frequently develop infectious diseases[24-26] as complications after surgery[27,28], and an increased BMI is associated with enhanced risk of infections in institutionalized geriatric patients[29].

The adipose tissue has an important role in regulating energy utilization, vascular functions and immune system homeostasis[30]. C-reactive protein (CRP)[8], interleukin (IL)-6[31], fibrinogen and plasminogen activator inhibitor-1[32] levels are higher in obese patients compared to healthy subjects. Stanton et al[33] have recently found that obese mice, after high fat and high cholesterol diets, express abnormal levels of macrophages and inflammation-associated genes in adipose tissue and in liver.

Obesity can influence liver metabolism directly, via circulating free fatty acids (FFA), and indirectly, via proinflammatory cytokine production. FFA and other lipids in hepatocytes are involved in production of reactive oxygen species, mitochondrial dysfunction and endoplasmatic reticulum stress. They have proapoptotic capacity and can stimulate proinflammatory signaling pathways[30]. FFA from adipose tissue, food and intestinal bacteria can bind toll like receptors (TLR) expressed on immune cells systemically and also in the liver, and enhance the hepatic expression of TLR-4 and TLR-2[34], these being receptors fundamental to the activity of immune system.

The presence of a dysregulation of the immune system in NAFLD has been firstly evidenced by the modification in immune cell populations in the liver. Natural killer (NK) cell circulating levels are reduced in obese rats[35]; meanwhile in the liver of NASH subjects their concentration is increased[36]. These cells have anti-fibrotic effects and produce apoptosis directly[37] and via interferon gamma (IFNγ) production[38] from hepatic stellate cells (HSC), which have a major role in liver fibrosis[39]. In the light of the strict resemblance between NASH and alcoholic hepatitis, Jeong et al[40] have detected that alcohol contributes to the anti-fibrotic effect of IFNγ and NK cells in animals.

Another immune cell population, natural killer T (NKT) cells, which express NK cell markers and α/β T cell receptors, are reduced in steatotic, obese mice[41,42] and in humans[43]. NKT cells are able to produce both T helper (Th) 1 and Th2 cytokines but their depletion in NAFLD has been associated with Th1 polarization of hepatic T cells in mice[44,45].

Two T helper cell subsets were recently discovered and are strictly related to the innate immune response. Th17 cells on one side and Treg cells on the other balance tolerance and elicitation of immune responses[46]. Th17 cells produce IL-17, IL-21 and IL-22, and require transforming growth factor-β (TGF-β) and IL-6 for their differentiation[47], the same cytokines that inhibit Treg cells. A Chinese study group has recently discovered that oxidative stress induces Treg cell apoptosis in mice with fatty livers[48] and subsequently has found also that Th17 cells are increased in the liver of animal and human NASH models[49].

Kupffer cells (KC) are liver macrophages involved in the response to such stressors as infections, ischemia and toxins[50] and they are also implicated in liver inflammation and NASH progression[51].

Tumor necrosis factor (TNF) -α, a cytokine produced by KCs, hepatocytes, and abdominal fat, is associated with the development in rodents[52,53] and in humans[54-57] of insulin resistance, NAFLD and NASH. The role of TNF-α in NAFLD may be due to its capacity to induce hepatocyte apoptosis, insulin resistance and to regulate KC activation locally[58,59]. Moreover, TNF-α regulates hepatic lipid metabolism[60].

In a NASH animal model involving choline-deficient diet fed rats it was found that there was an increase in serum and portal alanine aminotransferase levels and hepatic TNF-α, IFNγ and TLR4. Higher TNF-α levels were detected in KCs and, most importantly, increased TNF-α, TLR4 expression, and macrophage/dendritic cell populations were found in ileal tissue specimens, demonstrating also the involvement of the gut in steatotic liver damage[61].

To date, it is debatable whether circulating levels of TNF-α may discriminate the presence of NAFLD in obese subjects or in subjects with metabolic syndrome[62,63], but they seem to be useful in the non-invasive diagnosis of hepatic fibrosis in NASH[64].

IL-6 is a polyvalent cytokine with proinflammatory and prooncogenic activity, and it supports hematopoiesis[65] and is a predictive marker of insulin resistance and cardiovascular diseases[66]. In animal[67] and human[68,69] models respectively, hepatic and serum IL-6 levels are higher in NAFLD. Initially this cytokine was considered hepatoprotective because it reduces oxidative stress and prevents mitochondrial dysfunction in animal models[70,71]. Moreover, there are contrasting data on IL-6 production in the liver of NAFLD subjects[57,72]. IL-6, with TNF-α, suppresses adiponectin levels; meanwhile, TNF-α stimulates the production of leptin[73,74]. Adiponectin is an adipocytokine with anti-inflammatory properties and it decreases in subjects with increased liver fat concentration[75]. Leptin has opposite effects; it activates neutrophils and innate immune system[76], is associated with obesity and may contribute to NAFLD progression[77]. IL-6 production is also enhanced by TNF-α and IL-1 and can act with paracrine and endocrine mechanisms to activate IL-6 signaling systemically and peripherally in other organs such as liver and muscle[13]. FFA and IL-17 synergistically induce IL-6 production; on the other hand IL-6, with TGF-β1, enhances Th17 response in in vitro HepG2 cell models[49]. Tarantino et al[78] have also observed that, surprisingly, NAFLD subjects have increased TGF-β1 blood levels compared with those with chronic hepatitis C.

An anti-inflammatory cytokine, IL-10, is protective for hepatic steatosis, as seen in IL-10 deficient mice[79] as well as in NAFLD humans[80], and the inhibition of IL-10 promotes hepatic steatosis, enhances the expression of proinflammatory cytokines and impairs insulin signal transduction[81]. Main data on the pathophysiological role of inflammatory cytokines in NAFLD are summarized in Table 1.

Table 1 Principal findings on inflammatory cytokines in non-alcoholic fatty liver disease in humans, and in in vitro and animal models.
Principal findings
TNF-α
In vitro: FFA induce TNF-α gene expression[60]. KC and hepatocytes from NAFLD produce ↑ TNF-α and ↑ lipid peroxidation and accumulation[59,61]. TNF-α induces hepatocyte apoptosis[59]
Animal: TNF-α regulates KC apoptosis[58]. Hepatic, portal blood and intestinal TNF-α is ↑[52,53,61]
Human: Circulating levels are ↑ in NAFLD and NASH[57,68]. Contrasting data on simple FL[55,62]. They correlate with activity and progression of NAFLD[64] but do not differentiate mild to severe NASH[60]. NASH subjects have also ↑PBMCs TNF-α, IL-6 and IL-8 production[68]. TNF-α mRNA expression is ↑ in liver and fat of NASH compared with NAFLD[57], but there are contrasting data[55,238] TNF-α polymorphism is most frequent in NAFLD and correlates also with IR[56]
IL-6
In vitro: FFA induces IL-6 expression in hepatic cell cultures[72] and enhances Th17 response[49]
Animal: IL-6, TNF-α, IL-8 production is ↑ in liver and muscle of NAFLD mice[64]. Possible hepatoprotective role[70,71]
Human: ↑ IL-6 blood levels and other inflammatory and cytonecrosis indexes in NAFLD and NASH subjects compared to controls and obese[57,68,69]. IL-6 is an index of NASH activity and progression[72]. Normal levels of IL-6 and normal spleen longitudinal diameter may be useful in excluding NASH from NAFLD[34]. IL-6 tissue expression is controversial in liver of NAFLD[57,72]
IL-8
In vitro: IL-8 with TNF-α are ↑ in NAFLD and in NASH compared to FL[64]. FFA induces IL-8 expression[60]
Human: Blood levels of IL-8, IL-6 and TNF-α are ↑ in NASH[68,69]
IL-1β
Animal: NAFLD rats express similar IL-1β, TNF-α and IL-6 levels in liver and in muscle[64]
Human: TNF-α, IL-6 and IL-1β blood levels are ↑ in NAFLD and NASH[68,69]
TGF-β1
In vitro: IL-17 and FFA induce IL-6 in hepatocytes and IL-6, with TGF-β1, enhance Th17 response[49]
Human: TGF-β1 blood levels in NAFLD are ↑ than CHC[78]
IL-10
Animal: After IL-10 inhibition, TNF-α, IL-6 and IL-1β levels increase in liver of HFD mice[81]. IL-10 knock-out mice have ↑ FFA plasma levels and hepatic TG[79]
Human: In NAFLD and obese children, lower IL-10 blood levels correlate with markers of visceral and subcutaneous fat, insulin, HOMA-IR, ALT, AST and GGT[77]
IL-17
In vitro: IL-17 and FFA induce IL-6 production[49]
Animal: LPS-induced liver injury ameliorated after IL-17 blockade in HFD rats[49]
Th2 cytokines (IL-4, IL-5, IL-13)
Animal: Rats genetically oriented to a Th1 response develop steatosis and lobular inflammation more than others oriented toTh2 response[44,45]

Brun et al[82] have observed that HSCs isolated from genetically obese and diabetic mice show more pronounced fibrogenic responses induced by lipopolysaccharide (LPS) than HSCs from lean animals. Thus, HSCs are more sensitive to bacterial endotoxins, because genetically obese mice have an impaired intestinal permeability leading to increased portal endotoxemia. To expand on the evidence that systemic inflammation is also related to intestinal inflammation, a recent study undertaken by Kant et al[83] has found that weight loss in obese subjects reduces fecal calprotectin levels. Precedent studies have pointed out that circulating calprotectin levels are related to increased BMI[84,85]. As detailed later, the intestine, and especially intestinal inflammation, is closely related to NAFLD pathogenesis.

IMMUNITY IN IBS

In IBS subjects a low chronic inflammation is present and many other immune phenomena are also points of contact with hepatic steatosis.

The intestinal mucosa physiologically contains immune cells much more than other organs and tissues, and this is mainly due to its anatomical configuration and function as the first barrier of the organism[86]. In the “irritated” gut there is an increased population of immune cells in the small and large intestine, as reported in many studies[87,88]. Moreover, the inflammatory infiltrate is lower than in ulcerative colitis (UC) but is similar to that revealed in microscopic colitis[89]. These findings, with others discussed later, lead to the theory that IBS could be considered as an inflammatory disease.

The adaptive immune system is involved in the low grade inflammation of the gut, specifically, CD3+, CD4+ and CD8+ T cell count is increased[89-91] in the gut and in the peripheral blood of IBS subjects.

The innate immune response is also implicated in IBS pathogenesis. An increased number of mast cells are found in the small[92] and large[93] intestine. These cells are in close contact with enteric nerve endings[94] and this is an important factor in the neuronal stimulation that underlies the establishment of typical IBS symptoms[95]. Braak et al[96] are discordant on this point because they have observed a decreased number of mast cells, macrophages and T cells in IBS subjects. Moreover, they do not find visceral hypersensitivity or abnormal stress response.

Few reports have examined other immune cells involved in the innate immune system in IBS. NK cells[97] and neutrophils[98] may be hyper-activated but, to determine their role in intestinal inflammation, more studies are needed.

Contrasting data are reported on the monocyte/macrophage population. These cells were reduced[99] or normal[90] in number in the gut of IBS patients compared to controls but they may be hyper-activated, as seen by increased calprotectin expression[90]. Calprotectin is a calcium-binding protein produced by phagocytes with proinflammatory activity, such as leukocyte recruitment[100]. Fecal calprotectin may be useful in the differential diagnosis between inflammatory bowel diseases (IBD) and IBS[101]. Moreover, other authors have observed that patients with IBD and IBS-like symptoms have significantly higher fecal calprotectin levels than those with IBD but without IBS symptoms[102]. Shulman et al[103] have shown that fecal calprotectin concentration is greater in children with IBS and functional abdominal pain compared to controls, and also in the same population there is an impaired permeability in the proximal and distal gut.

There are contrasting data on the role of Treg cells, a T cell subpopulation with regulatory functions in IBS: these cells seem to be normally or under-expressed in intestinal tissues and blood of IBS subjects[104,105], even though previously Chadwick et al[88] have observed increased CD25+ T cell population in the lamina propria of IBS subjects. The role of Th17 cells in the pathophysiology of IBS is still unexplored but, recently, Andoh et al[106] have summarized the main evidence on the role of this subpopulation in intestinal inflammation. It would be interesting to see if IBS might be involved in the dysregulation between Th17 and Treg cells as shown in NAFLD.

Studies on proinflammatory cytokine production in IBS have evidenced the activation of both the innate and adaptive immune systems. Indeed, different study methods were used to explore the systemic cytokine production and results were not always concordant[107].

IL-6 and TNFα are the most studied inflammatory cytokines in IBS. In many reports blood levels of TNFα and IL-6 are increased[108-112]. Similar results are reported in cultured peripheral blood mononuclear cells[108,111]. Studies on Peripheral blood mononuclear cells (PBMCs) have also noticed decreased levels of the anti-inflammatory IL-10[112,113], in agreement with the systemic inflammatory state in IBS. Moreover, the IL-10 high producer genotype seems to be protective against IBS, whereas IL-10 low producer, and maybe even TGF-1β intermediate producer genotypes, are a risk factor for IBS development[114]. In IBS mice, IL-6 may enhance colonic cells neuronal activation and their absorption/secretory responses[115]. The intestinal cytokine production is poorly understood[116-119], and, as described in a recent review by Ortiz-Lucas et al[120], only IL-1β expression is clearly increased in post-infectious IBS (PI-IBS)[121]. On the contrary, Hughes et al[122] have observed increased cytokine expression in supernatants of mice with IBS and that visceral neurons express receptors for IL-6, TNF-β, IL-1β and IL-10, confirming the role of these pro-inflammatory cytokines in gut homeostasis.

Th 2 cytokines were also considered in recent reports: in animals Th 2 cytokines enhance intestinal motility[123] and in IBS subjects stimulated PBMCs produce more IL-5 and IL-13 than controls[124].

Cytokines have several roles in the development of IBS symptoms. For example, TNF-α can act on the peripheral nervous system as well as on the central nervous system (CNS) to develop a symptom burden of hypersensitivity, nausea, emesis, gastric hypomotility, anorexia and fever[125,126]. IL-6 is able to stimulate submucosal neurons in IBS animal models[127], most probably via a TLR-mediated mechanism[128]. TNF-α and IL-6 are also implicated in intestinal barrier integrity[129] (Table 2).

Table 2 Principal findings on inflammatory cytokines in irritable bowel syndrome in humans, and in in vitro and animal models.
Principal findings
TNF-α
Animal: D-IBS supernatants have ↑ levels of proinflammatory cytokines and they cause hypersensitivity in mouse colonic afferent endings[122]
Human: IBS has ↑ circulating TNF-α levels[109,112], especially D-IBS[112] or in patients with comorbidities such as fibromyalgia, premenstrual dysmorphic disorder and chronic fatigue syndrome[109]. Baseline and LPS-stimulated levels in PBMCs of proinflammatory cytokines as TNF-α, in IBD and D-IBS, are ↑ and are related to symptom intensity[108]. TLR-2, TLR-4 and TLR-5 antagonists induce TNF-α production[128]. No difference in TNF-α and other proinflammatory cytokine production (IL-6 and IL-1β) in the gut of IBS subjects compared to controls[116]
IL-6
In vitro: No differences in colonic production between IBS and controls 116. IL-6 have excitatory action on colonic cells from IBS rats producing neuronal activation and absorption/secretory responses[115]
Animal: IL-6 colonic secretion is ↑ in IBS rats and activate submucosal neurons[127]
Human: IL-6 blood levels are ↑ in all IBS subtypes[109-111]. IL-6 levels are related to ACTH response and ∆ACTH/∆Cortisol ratio[110]. Baseline and LPS or TLR agonist-stimulated PBMC levels are ↑ in IBS[108]
IL-8
In vitro: Reduced expression of mRNA of IL-8 in ex vivo biopsy cultures[116]
Human: Circulating levels of IL-8 are ↑ in IBS[109-111,119]. TLR-3 and TLR-7 agonists induce IL-8 production in PBMCs[128]
IL-1β
Animal: In stressed rats with previous acute colitis IL-1β mRNA expression is ↓[117]
Human: ↑ IL-1β levels in IBS[108,128], in C-IBS and in D-IBS[108]. With TNF-α, IL-1β↑ levels are found in IBS subjects with fibromyalgia, premenstrual dysmorphic disorder and chronic fatigue syndrome[109]. IL-1β↑ production in PBMCs stimulated by antiCD3/CD28 antibody[91] and by TLR-4 and TLR-5 agonists[128]. Increased IL-1β expression in rectum of PI-IBS[121]
TGF-1β
Animal: No different expression of TGF-β1 protein in colon of IBS rats[11]
Human: TGF-1β intermediate producers may be at risk of developing IBS[114]
IL-10
Human: IBS subjects have ↓ circulating levels of IL-10[112]. Altered IL-10/IL-12 ratio in PBMCs with Th1 proinflammatory state[113]. IL-10 levels are ↓ and IFNγ levels are ↑ in colon of PI-IBS compared to non PI-IBS and controls[119]. IL-10 high producer genotype is protective against IBS[114]
Th2 cytokines (IL-4, IL-5, IL-13)
Animal: Th2 cytokines may have a role in intestinal hypercontractility[123]
Human: Stimulated PBMCs IL-5 and IL-13 levels are ↑ in IBS[124]
NAFLD AND IBS MAY BE RELATED

The above-mentioned evidence suggests that innate immunity is a main pathogenetic component of both NAFLD and IBS. But, how does the immune system work in patients with both NAFLD and IBS In other words, is the similar action of pro-inflammatory cytokines, such as IL-6 and TNF-α, the only one that can be found on the immune system side

The metabolic syndrome, which often anticipates or is detected in conjunction with NAFLD, leads to a state of chronic inflammation, systemic or local (hepatic)[12], but to date it is still unclear which one of the two types has a greater impact on these patients, even if a lot of evidence favors the former[13]. A very similar scenario, but with partly different participants, is possible in IBS. Although the disease has not been overtly related to an inflammatory systemic disease, as happens for the metabolic syndrome, nevertheless, IBS is characterized by hyper-activation of the immune system and general inflammation. Indeed, many researchers have struggled to find a similar component at local level, studying the intestinal cytokine production, but they have not always had a favorable outcome[107,120]. In some subsets of IBS patients, such as diarrhoea-predominant IBS (D-IBS) and IBS developing following infective gastroenteritis (PI-IBS), there is often a frank intestinal inflammation[108,119]. On the contrary, in C-IBS a systemic inflammation is not always associated with a local counterpart or is less apparent than in D-IBS[122].

NAFLD and IBS are classically defined as different diseases. NAFLD is related to the metabolic syndrome, obesity, diabetes and insulin resistance and IBS is a functional intestinal disease closest to psychological disorders such as depression and anxiety, certainly not to liver diseases. But, surprisingly, there are many points of contact, such as the dysfunction of the intestinal microbiota, the impaired intestinal barrier, intestinal dysmotility and brain-gut axis dysfunction, which are fundamental to their pathogenesis, being related to the immune activation and inflammation.

Thus, principal questions are: Can metabolic liver disease affect the functions of the gastrointestinal tract leading to syndromic manifestations typical of IBS and may the bowel dysfunction lead or otherwise support the development of a chronic hepatic inflammatory state

GUT MICROBIOTA

The gut microbiota is a composite member of our body. Intestinal bacteria interact with the intestinal epithelial barrier and subsequently with extraintestinal organs performing physiological and pathological actions.

This close contact makes the microbiota important for the metabolism of nutrients and energy delivery[130], the intestinal barrier function[131], the natural tropism of the intestinal wall[132] and ensures the maturation of intestinal immune tolerance and the immune response[133].

The dysregulation of the intestinal bacterial milieu is a component of NAFLD and IBS. Recent reports have also shown both in NAFLD and in IBS an important role for TLR. These are receptors that characterize the innate immunity and link specific molecules such as pathogen-associated molecular patterns, LPS, and danger-associated molecular patterns[134]. These receptors are able to elicit the innate immune response once activated (they induce the expression of proinflammatory chemokines, cytokines and adhesion molecules on immune cells)[135]. In NAFLD and in IBS this role is consistently related to the alteration of gut microbiota, impaired intestinal permeability and impaired intestinal motility[136,137].

Changes in microbiota composition and simultaneous or subsequent dysregulation of intestinal permeability let PAMPs and TLRs be in strict contact in the deeper layers of the intestinal wall and thus lead to stimulation of the innate immune response[138].

Despite the fact that the roles of TLRs in the liver of NAFLD and NASH are well established[137], only recently have the activity of TLRs in IBS been studied. Ohman et al[139] have observed increased expression of TLR2 on circulating monocytes in IBS. A study from McKernan et al[128] demonstrated that the TLR-induced cytokine release (IL-1β, IL-6, IL-8 and TNF-α) was enhanced in blood from IBS subjects. The TLR mRNA production in the gut mucosa of mice with colonic visceral hypersensitivity was studied and significant increases were seen[140]. Similar results were found in humans[141].

TLRs are fundamental in T-cell differentiation and activation, particularly for Th17 and Treg cells[142]. In the gut, bacterial products[143], acute phase proteins[144] and proinflammatory cytokines such as IL-6 and TGF-β[145] promote Th17 response, meanwhile IL-25 and IL-23[146] produced by epithelial cells inhibit it.

Obesity and NAFLD

In the literature there are few reports on the intestinal microbiota composition in NAFLD. The role of intestinal dysbiosis in these patients may be assumed by reports on microbiota present in obese subjects or by indirect data on the action of bacterial products from the gut delivered to the liver in NAFLD.

Obese patients are characterized by low intestinal bacterial diversity. They have a reduced Bacteroides and increased Firmicutes population compared to controls, and this proportion improves with weight loss[147]. Studying the microbiome, the same group has found that obese patients exhibit impaired bacterial gene expression[148].

Animal models have shown that the intestinal microbiota may have an important role in energy harvesting and fat storage. Germ-free mice seem to be protected from diet-induced weight gain[149] most probably because intestinal bacteria are involved in the fermentation of polysaccharides to monosaccharide and in the metabolism of short chain fatty acids[150]. The microbiota can also enhance the lipoprotein lipase activity because it reduces the expression of the fasting-induced adipocyte factor in the intestinal epithelium resulting in enhanced FFA storage in adipocytes[149].

LPS produced by intestinal bacteria constitutes the outer membrane of Gram-negative bacteria and can elicit an immune response acting as an endotoxin. LPS may also have a role in the development of obesity, low-grade inflammation and insulin resistance[151]. An elegant study by Cani et al[152] noticed that high-fat diet induces LPS production in mice and probably its abnormal absorption through the intestinal epithelium may be fat-dependent. The same study has evidenced that endotoxemia induces weight gain, intrahepatic triglyceride accumulation and hepatic insulin resistance, leading to increased expression of TLR4 and proinflammatory cytokines (TNF-α, IL-6, IL-1 and PAI1) in muscle, adipose tissue and liver.

The correlation between intestinal dysbiosis and lipid accumulation in the liver is evidenced by recent research by de Wit et al[153]: in mice, a diet with high concentration of palm oil induces higher weight gain and liver triglyceride concentration, reduces microbial diversity and increases Firmicutes/Bacteroidetes ratio compared to one high in polyunsaturated fatty acids. The fecal microbiota of women following a choline-deficient diet, which induces steatosis, varies during choline depletion and correlates with changes in liver fat concentration, showing modifications in Gammaproteobacteria and Erysipelotrichi populations[154].

IBS

Intestinal dysbiosis is also involved in the development of IBS symptoms. The intestinal microbiota modulates intestinal motility and sensitivity[155]. An animal study has observed that oral antibiotic therapy perturbs the intestinal microbiota, reduces lactobacilli and decreases bacteroides and enterococci populations, and affects pain perception and viscero-motor responses in the gut. The myoelectrical activity in the gut is also altered in germ-free animals and it reversed after colonization[156]. The supernatant made from Escherichia coli Nissle 1917 stimulates smooth muscle cells and enhances colonic contractility[157], and also Lactobacillus rhamnosus GG has a dose- and time-dependent effect on the acetylcholine-stimulated contraction of human colonic muscle cells[158]. Lactobacillus rhamnosus also has a protective role in pain prevention in animal models[159].

The intestinal bacterial population inhabits a complex environment and its composition varies throughout the gut. It is necessary to distinguish at least three different types of microbiota evaluated in different studies: the luminal microbiota, within the intestinal lumen; the mucosal microbiota that adheres to the intestinal wall; and the fecal microbiota, excreted in feces. In IBS subjects, studies on fecal microbiota have found increased facultative and anaerobic bacteria (as Escherichia coli and Clostridium) and decreased lactobacilli and bifidobacteria[160,161]. Later studies used molecular techniques because most bacterial species in the gut are not cultivable; a recent report of the Rome foundation reviewed principal results[162]. The majority of reports have studied fecal microbiota while only a few are focused on the mucosal flora. Furthermore, different molecular techniques are carried out and other limitations may explain that data shown are often contradictory or inconsistent. Moreover, the evidence that SIBO is frequently found in IBS subjects[163], especially in diarrhoea-predominant IBS (D-IBS)[164], and that IBS can develop following infective gastroenteritis (PI-IBS)[165] confirms the role of gut dysbiosis in the IBS pathogenesis.

INTESTINAL PERMEABILITY

A single layer of cells composes the intestinal epithelium, a selective filter and barrier for exogenous substances and water[129]. The ways to pass the epithelial layer are mainly two: transcellular and paracellular[166].

The regulation of the paracellular pathway is mainly due to complex structures localized at the apical-lateral and along the lateral membrane between the cells of the intestinal epithelium: desmosomes, adherent junctions and tight junctions (TJs)[167].

TJs regulate selective paracellular ionic solute transport, prevent the passage of luminal antigens, microorganisms and toxins, but also regulate the tropism of enterocytes[168]. TJs are so called “kissing points”, fusion points where there is no space between two enterocytes[166], and are formed by different transmembrane proteins: tricerullin, occludin, claudins and junctional adhesion molecules, which seal together adjacent cells and cytoskeleton[169].

Several stimuli can modulate the intestinal permeability, but bacterial toxins inter alia are able to modify the localization of TJ proteins directly[170] or via the release of proinflammatory cytokines such as TNF-α, IFN-γ[171] and IL-6[172] that per se can reduce the expression of zonula occludens-1 (ZO-1), occludin and claudin.

NAFLD and NASH

In a recent review, Ilan[151] have focused on the role of bacterial translocation in NASH. The bacterial translocation is intimately connected with liver damage from the first step of lipid accumulation in the liver to the development of steatohepatitis, passing through the activation of the innate immune system and mitochondrial dysfunction.

Many animal and human studies have focused on the microbial dysbiosis in NAFLD and to date the endotoxemia, subsequent to bacterial translocation from the gut to the liver through the venous portal system, is an important factor in the development of NASH[173]. The mechanisms that lead up to endotoxemia are bacterial overgrowth and impaired intestinal barrier. Sabaté et al[174], and previously Wigg et al[175], have pointed out that obese subjects have an increased prevalence of SIBO and this condition correlates with severe hepatic steatosis.

Obese mice have a modified distribution of occludin and ZO-1 in the intestinal mucosa in combination with a lower intestinal resistance and higher circulating levels of inflammatory cytokines and portal endotoxemia[82]. Similar results are found in mice with fructose-induced steatosis: treatment with metformin leads to a decrease in hepatic triglyceride accumulation and plasma alanine-aminotransferase levels and protection against the loss of the TJ proteins occludin and ZO-1 in the duodenum[176].

In humans, an immunohistochemical analysis of duodenal expression of ZO-1 performed by Miele et al[177] has highlighted that subjects with biopsy-proven NAFLD have increased gut permeability and high prevalence of SIBO, and that both correlate with the severity of steatosis. Also, NASH subjects have a higher prevalence of SIBO, related to enhanced expression of TLR-4 and release of IL-8[178]. The presence of endotoxins in portal blood is found also in cirrhotic patients and is related to an impaired intestinal barrier function[179]. Non-cirrhotic NAFLD subjects have increased LPS[180] and LPS-binding protein serum levels[181]. Probiotic treatment of obese mice leads to a lower intestinal permeability and improved TJ function, a lower plasma LPS and cytokine concentration and a decreased hepatic expression of inflammatory and oxidative stress markers[182]. Recently, the association between metabolic syndrome, gut microbiota dysregulation and impaired intestinal barrier has been further confirmed in an animal model where dietary obese rats show reduced expression of ZO-1 in the gut and higher TNF-α levels in combination with reduced Lactobacillus and increased Oscillibacter fecal population. Moreover, TNF-α and IL-6 mRNA levels were higher in mesenteric fat[183].

IBS

The impaired intestinal permeability is not only a key factor in the development of NAFLD and NASH. Other inflammatory gastrointestinal diseases such as Crohn’s disease, UC, bacterial infections caused by Escherichia coli, Clostridium difficile and Vibrio cholera, anti-inflammatory agents associated enteritis and IBS are involved. In vivo studies have observed that IBS patients have an impaired intestinal barrier function[87,90]. Nevertheless, it is likely that these findings are specific only to D-IBS and PI-IBS subjects and in other IBS subtypes similar results are not found[87,184].

In IBS, intestinal dysbiosis is an important factor participating in damaging the intestinal barrier through the activation of the immune system[185] even though another possible cause of impaired intestinal barrier is the exposure to chronic stress. In healthy animals and humans, acute or chronic stress enhances the intestinal permeability to water and also to macromolecules, and IBS subjects are more sensitive to physical and mental stressors compared to healthy subjects[110].

It has been explicated that in IBS subjects there is a low grade inflammation in the gut. Mast cells and T lymphocytes represent the majority of intestinal inflammatory infiltrate and mast cells are also involved in the regulation of motor and visceral responses in the intestine[19,21,88].

The intestinal permeability is controlled by mast cells, via histamine, serotonin 5-hydroxytryptamine (5-HT) and protease production[21]. Proteases are markedly increased in the mucosa of IBS subjects[18,186] and supernatants rich in proteases from D-IBS subjects are able to evoke epithelial dysfunction and allodynia in healthy mice[20]. In addition, colonic soluble mediators in supernatants from IBS subjects are able to reproduce permeability alterations in Caco-2 cells and decrease ZO-1 expression[22]. A recent study by Martínez et al[187] confirms this hypothesis because it has been demonstrated that activated mast cells induce the downregulation of ZO-1 in intestinal epithelium.

Another class of TJ proteins, claudins, is involved too; in fact, claudin-1 and claudin-4 levels are decreased in the small and large intestine of D-IBS patients, whereas claudin-1 and claudin-3 were elevated in constipation-predominant IBS (C-IBS) patients[188].

INTESTINAL MOTILITY

Intestinal motor and sensory functions are influenced by the immune system to activate a mechanism of defense from noxious agents in the intestinal lumen[189].

Mice infected with Trichinella spiralis develop muscle hyper-contractility in the gut[190] but these effects disappear in animal models of athymic and CD4+ cell-deficient mice[191], encouraging the hypothesis of a role for the immune system and inflammation in intestinal motor functions. Th2 cytokine production was associated with enhanced motor functions and appropriate helminthic elimination. On the other hand, the response with a reduced intestinal motility of Th1, but interestingly also of Th17 cells, seems to be involved in small intestine motor functions. In this setting, specifically IL-17 induces smooth muscle cell contraction[192].

Among Th2 cytokines, IL-13 is secreted by CD4+ cells and by many other immune cellular types of innate immunity, as the so called “innate helper cells”, which can be found normally in the gut and in blood. IL-13 has, in low concentrations, regulatory effects, increasing IL-10 and decreasing IL-17 levels, but, when up-regulated, it leads to inflammatory modifications and hyper-contractility of smooth muscle in the gut[193].

In agreement with these findings, the production of 5-HT, one of the most important neurotransmitters of intestinal motility[194], is also influenced by immune response and cytokine production and its secretion seems to be enhanced by Th2 and reduced by Th1 response[195]. 5-HT is synthesized and secreted by enterochromaffin cells (EC) and acts on receptors located on the processes of intrinsic and extrinsic primary afferent neurons in the lamina propria of the gut to initiate peristaltic and secretory reflexes[196]. The 5-HT transporter (SERT) is the physiological inhibitor, it is expressed by enterocytes and removes 5-HT from the intestinal space by internalizing it[197].

Obesity and NAFLD

In high-fat diet fed mice a slower gastric emptying was found, as well as modified intestinal hormone production: higher plasma leptin and cholecystokinin (CCK) concentrations and lower plasma ghrelin levels were found[198]. Covasa et al[199] have shown that in high-fat diet fed mice there is a reduction in CCK-induced and oleate-induced inhibition of gastric motility.

In obese rats, after Roux-en-Y gastric bypass, an increase in peptide YY and a decrease in ghrelin concentrations occurred. These hormonal modifications may contribute to weight loss by decreasing the food intake and slowing the gastric emptying and transit time[200].

A recent study by Hyland et al[201] confirms the presence of an impaired intestinal motility, a modified submucosal nerve function and a decreased electrogenic glucose transport in obese rats. The author hypothesizes that the loss of motor control may lead to an altered host defense and intestinal dysbiosis, and the adapted glucose transport may be a control mechanism in the restriction of nutrient absorption.

Obese subjects have an accelerated esophageal and gastric motility and impaired gastrointestinal hormone secretion[202,203]. Vazquez Roque et al[204] have detected a lower postprandial gastric volume in obese subjects. A recent report disputes their data: in newborns, fasting and postprandial gallbladder volumes and gastric emptying were similar between obese and lean subjects, but in obese pre-adolescents, and even more in adults, a larger fasting gallbladder volume with slower postprandial gastric emptying was found[205].

Small and large intestinal motility are also involved, as reported by Xing et al[206]. As we see above, SIBO is most frequently viewed in obese subjects and it has been associated with an altered pattern of migrating motor complexes (MMC) in the small intestine[207].

The role of intestinal dysmotility in liver cirrhosis is confirmed by numerous data[208]. Vice versa, in NAFLD, only a few studies have focused on impaired intestinal motility, although obesity, which is one of the most important etiological factors of NAFLD, is strictly related to impaired intestinal motility. Initial studies have found that NAFLD[209] and non-alcoholic cirrhosis[210] subjects have a prolonged orocecal transit time.

Interestingly, an up-to-date study correlates 5-HT3 antagonists to reduced endotoxin levels in the portal system, attenuated liver fat content, inflammation, and cell necrosis, improved TNF-α levels and increased TJ expression in the duodenum of obese, leptin-deficient mice[211]. The same group has confirmed these data and has found that SERT deficiency causes hepatic steatosis and impaired intestinal permeability[212]. These findings suggest that obesity, and consequently NAFLD, are affected by impaired gut motility and most probably the impaired intestinal barrier, the gut inflammation and also neuronal signaling are key points in their maintenance.

IBS

IBS subjects frequently report upper gastrointestinal symptoms such as functional dyspepsia[213]. Impaired lower esophageal motility and delayed gastric emptying are frequently viewed[214] and should be related to small-bowel dysmotility[215].

Many studies have focused attention on the small intestine and large intestine gut dysmotility in IBS subjects. As reviewed elsewhere, studies on MMCs and clustered activity as well as intestinal transit for the small intestine and on myoelectrical activity, intraluminal pressure recordings and transit for the large intestine confirm this hypothesis[216].

In the small intestine of IBS subjects, alterations in the periodicity of MMCs are found[217]. Kellow et al[218,219] have demonstrated that MMCs have a shorter periodicity in D-IBS, whereas in C-IBS this is longer.

EC cell numbers in the intestinal wall are increased[220,221] and postprandial 5-HT levels are increased in platelet-poor plasma[222] of IBS subjects, especially in PI-IBS. 5-HT signaling is involved in the pathogenesis of intestinal dysmotility and hypersensitivity; indeed 5-HT modulators are used in IBS therapy[223]. 5-HT4 agonists accelerate colonic transit and are useful in constipation unresponsive to laxative treatment, while 5-HT3 antagonists inhibit colonic secretion and motility, and visceral sensation, and for this reason are used in D-IBS.

Moses et al[224] have found that SERT was less expressed in C-IBS and UC colonic biopsy specimens. Camilleri et al[225] have shown that SERT polymorphisms may influence colonic motility in patients with D-IBS and may influence the response to a 5-HT3 antagonist.

In the colon of IBS subjects activated mast cells in proximity to mucosal innervations may contribute to pain perception[93] and are correlated with 5-HT release by intestinal EC cells[226]. Interestingly, Mizutani et al[123] have observed that in an animal model of IBS, muscle hyper-contractility is related to an increased Th2 cytokine profile (IL-4 and IL-13). Even if these data confirm the role of immune activation in gut motility alteration, it is mandatory to observe that in IBS, and especially in D-IBS and PI-IBS, there is an enhanced gut motor activity even though these IBS subtypes are often related to a Th1 cytokine profile, at least in peripheral blood or in PBMCs.

However, there are no reports on the possible role of IL-17 and Th17 in IBS; meanwhile, IL-13 production by PBMCs is higher compared to controls[124].

Recent studies have shown that bacterial products may regulate gastrointestinal motor functions[227,228], but intestinal motility may also influence the gut microbiota composition[229]. Pimentel et al[230] for the first time demonstrated that the impaired intestinal motility may be related to SIBO in IBS subjects, but subsequent contrasting data have questioned this theory[163]. Moreover, as has been described before, IBS and NAFLD are characterized by an intestinal dysbiosis and only a proportion of subjects meet diagnostic criteria for SIBO.

CNS INVOLVEMENT

A recent review by Capuron et al[231] has focused on how the immune system can affect the CNS and contribute to the development of neuropsychiatric disorders such as depression, with particular relevance to cytokine signaling. Cytokines are involved in production, function and reuptake of several neurotransmitters, such as 5-HT. They affect the hypothalamic-pituitary-adrenal (HPA) axis and can modify the neuronal architecture, neuronal plasticity and aging, and neuronal circuits in CNS.

As previously described, 5-HT is an important neurotransmitter of the enteric nervous system (ENS) but it is also fundamental to CNS functioning. 5-HT, produced from tryptophan, plays a major role in the modulation of brain-gut axis[232]. The brain-gut axis is constituted peripherally of ENS communicating with the gut wall and centrally with the CNS and HPA axis[233]. The gastrointestinal system and the brain communicate in bi-directional mode, both of them influencing each other (the so called top-down and bottom-up model developed in functional GI disorder studies)[234]. The HPA axis is composed of corticotropin-releasing hormone (CRH), produced in the hypothalamic para-ventricular nucleus, which stimulates adrenocorticotropin (ACTH) production in the anterior pituitary gland that in turn induces the adrenal cortex to produce cortisol in response to various stressors[235].

In animal and human models the turnover of 5-HT in the brain is altered by acute and chronic exposure to proinflammatory cytokines[236,237].

Cytokines stimulate CRH, ACTH and cortisol production and in chronic states influence the diurnal cortisol curve because they stimulate inflammatory signaling that reduces glucocorticoid receptor functions and expression leading to decreased responsiveness to glucocorticoids.

Obesity and NAFLD

Recently, animal studies have shown that in the hippocampus and cortex of high-fat fed mice there is increased production of inflammatory products[238,239] and systemic inflammation is also related to cognitive dysfunctions[240,241]. Depression and depressed serotoninergic state are strictly related to metabolic syndrome and obesity[242,243]. Tarantino et al[244] have studied urinary 5-hydroxy-3-indoleacetic acid, a 5-HT metabolite, in depressed and obese/overweight subjects and have found that it negatively correlates with dysthymia and depression status.

Alteration in the HPA axis is well established in obese patients and chronic stress with hyper-alimentation is an important factor in its development[245]. Although there are contrasting data on urinary free cortisol (UFC) in obese subjects, a recent study has evidenced in NAFLD subjects increased UFC and cortisol serum concentrations after dexamethasone suppression, both correlated with hepatic inflammation and fibrosis stage[246]. Moreover, in a human model, cortisol clearance is increased in NAFLD subjects and is correlated with insulin sensitivity[247]. Peripherally, cytokines such as TNF-α and leptin stimulate 11β-HSD1, an enzyme required for the activation of cortisone to cortisol[248]. Also leptin and ghrelin increased levels are related to HPA axis dysregulation in obese subjects[245].

Finally, early life stress predisposes to overweight and insulin resistance, at least in animal models[249].

IBS

Hypersensitivity and brain alterations, investigated with different study methods, have been found in the last 15 years in IBS subjects; and, despite often contradictory data, there is strong evidence of dysregulation in pain and other stimuli perception[250]. Moreover, mood disorders (depression, anxiety) and other psychiatric disorders (eating disorders, posttraumatic stress syndrome, panic attack, etc.) are frequent, evidencing the role of gut-brain dysfunction in these patients[107].

As has been mentioned above, the majority of reports on 5-HT in IBS have studied its intestinal production; meanwhile, few are focused on its systemic production. Clarke et al[251] have found that IBS subjects degrade tryptophan more via the kynurenine pathway, an alternative metabolic way producing neurotransmitters other than 5-HT. Subsequently, the same group has found that kinurein from blood of IBS subjects can influence TLR expression[252] in an in vitro model.

The main evidence on HPA dysregulation in IBS[250] is the following: CRH and ACTH stimulate colonic secretion, intestinal motility, visceral sensitivity and anxiety. Principal brain regions influenced by HPA axis are the amygdala and hippocampus. In IBS there are increased HPA axis responses to stressors such as meals, hormonal stimuli, and mental stress compared to controls. Fatigue and depression are associated with increased mast cell counts in the colonic mucosa of IBS subjects, confirming the role of gut-brain dysfunction in IBS[253]. Indeed, a key question still unresolved is whether the SNC dysfunction is the primum movens of the gut inflammation and consequently the visceral hypersensitivity and dysmotility in IBS or whether the gut inflammation represents the main cause of subsequent SNC and systemic disorder.

UNANSWERED QUESTIONS

Could weight loss ameliorate IBS symptoms by influencing intestinal microbiota Is there a relationship between NAFLD severity and IBS symptoms Could patients suffering from IBS be at major risk to develop NASH Are circulating levels of inflammatory cytokines overlapping in IBS subjects and NAFLD Could intestinal dysbiosis affect CVD risk via NAFLD[254]

Footnotes

P- Reviewer Hahm KB S- Editor Gou SX L- Editor Logan S E- Editor Li JY

References
1.  Adams LA, Angulo P. Recent concepts in non-alcoholic fatty liver disease. Diabet Med. 2005;22:1129-1133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 200]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
2.  Delgado-Aros S, Locke GR, Camilleri M, Talley NJ, Fett S, Zinsmeister AR, Melton LJ. Obesity is associated with increased risk of gastrointestinal symptoms: a population-based study. Am J Gastroenterol. 2004;99:1801-1806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 193]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
3.  Talley NJ, Quan C, Jones MP, Horowitz M. Association of upper and lower gastrointestinal tract symptoms with body mass index in an Australian cohort. Neurogastroenterol Motil. 2004;16:413-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 81]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
4.  Sadik R, Björnsson E, Simrén M. The relationship between symptoms, body mass index, gastrointestinal transit and stool frequency in patients with irritable bowel syndrome. Eur J Gastroenterol Hepatol. 2010;22:102-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 51]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
5.  Cremonini F, Camilleri M, Clark MM, Beebe TJ, Locke GR, Zinsmeister AR, Herrick LM, Talley NJ. Associations among binge eating behavior patterns and gastrointestinal symptoms: a population-based study. Int J Obes (Lond). 2009;33:342-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 55]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
6.  Clements RH, Gonzalez QH, Foster A, Richards WO, McDowell J, Bondora A, Laws HL. Gastrointestinal symptoms are more intense in morbidly obese patients and are improved with laparoscopic Roux-en-Y gastric bypass. Obes Surg. 2003;13:610-614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 78]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
7.  Bures J, Cyrany J, Kohoutova D, Förstl M, Rejchrt S, Kvetina J, Vorisek V, Kopacova M. Small intestinal bacterial overgrowth syndrome. World J Gastroenterol. 2010;16:2978-2990.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 383]  [Cited by in F6Publishing: 332]  [Article Influence: 23.7]  [Reference Citation Analysis (3)]
8.  Tarantino G, Colicchio P, Conca P, Finelli C, Di Minno MN, Tarantino M, Capone D, Pasanisi F. Young adult obese subjects with and without insulin resistance: what is the role of chronic inflammation and how to weigh it non-invasively. J Inflamm (Lond). 2009;6:6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 52]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
9.  Larter CZ, Chitturi S, Heydet D, Farrell GC. A fresh look at NASH pathogenesis. Part 1: the metabolic movers. J Gastroenterol Hepatol. 2010;25:672-690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 123]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
10.  Day CP, James OF. Steatohepatitis: a tale of two “hits”? Gastroenterology. 1998;114:842-845.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Portincasa P, Grattagliano I, Palmieri VO, Palasciano G. Nonalcoholic steatohepatitis: recent advances from experimental models to clinical management. Clin Biochem. 2005;38:203-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 67]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
12.  Tilg H, Moschen AR. Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology. 2010;52:1836-1846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1543]  [Cited by in F6Publishing: 1629]  [Article Influence: 116.4]  [Reference Citation Analysis (0)]
13.  Tarantino G, Savastano S, Colao A. Hepatic steatosis, low-grade chronic inflammation and hormone/growth factor/adipokine imbalance. World J Gastroenterol. 2010;16:4773-4783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 131]  [Cited by in F6Publishing: 148]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
14.  Compare D, Coccoli P, Rocco A, Nardone OM, De Maria S, Cartenì M, Nardone G. Gut--liver axis: the impact of gut microbiota on non alcoholic fatty liver disease. Nutr Metab Cardiovasc Dis. 2012;22:471-476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 298]  [Cited by in F6Publishing: 296]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
15.  Machado MV, Cortez-Pinto H. Gut microbiota and nonalcoholic fatty liver disease. Ann Hepatol. 2012;11:440-449.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Ringel Y, Ringel-Kulka T. The rationale and clinical effectiveness of probiotics in irritable bowel syndrome. J Clin Gastroenterol. 2011;45 Suppl:S145-S148.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Brandt LJ, Chey WD, Foxx-Orenstein AE, Schiller LR, Schoenfeld PS, Spiegel BM, Talley NJ, Quigley EM. An evidence-based position statement on the management of irritable bowel syndrome. Am J Gastroenterol. 2009;104 Suppl 1:S1-S35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 258]  [Article Influence: 17.2]  [Reference Citation Analysis (1)]
18.  Cenac N, Andrews CN, Holzhausen M, Chapman K, Cottrell G, Andrade-Gordon P, Steinhoff M, Barbara G, Beck P, Bunnett NW. Role for protease activity in visceral pain in irritable bowel syndrome. J Clin Invest. 2007;117:636-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 432]  [Cited by in F6Publishing: 431]  [Article Influence: 25.4]  [Reference Citation Analysis (0)]
19.  Barbara G, Wang B, Stanghellini V, de Giorgio R, Cremon C, Di Nardo G, Trevisani M, Campi B, Geppetti P, Tonini M. Mast cell-dependent excitation of visceral-nociceptive sensory neurons in irritable bowel syndrome. Gastroenterology. 2007;132:26-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 531]  [Cited by in F6Publishing: 540]  [Article Influence: 31.8]  [Reference Citation Analysis (0)]
20.  Gecse K, Róka R, Ferrier L, Leveque M, Eutamene H, Cartier C, Ait-Belgnaoui A, Rosztóczy A, Izbéki F, Fioramonti J. Increased faecal serine protease activity in diarrhoeic IBS patients: a colonic lumenal factor impairing colonic permeability and sensitivity. Gut. 2008;57:591-599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 221]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
21.  Buhner S, Li Q, Vignali S, Barbara G, De Giorgio R, Stanghellini V, Cremon C, Zeller F, Langer R, Daniel H. Activation of human enteric neurons by supernatants of colonic biopsy specimens from patients with irritable bowel syndrome. Gastroenterology. 2009;137:1425-1434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 262]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
22.  Piche T, Barbara G, Aubert P, Bruley des Varannes S, Dainese R, Nano JL, Cremon C, Stanghellini V, De Giorgio R, Galmiche JP. Impaired intestinal barrier integrity in the colon of patients with irritable bowel syndrome: involvement of soluble mediators. Gut. 2009;58:196-201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in F6Publishing: 385]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
23.  Barbara G, De Giorgio R, Stanghellini V, Cremon C, Salvioli B, Corinaldesi R. New pathophysiological mechanisms in irritable bowel syndrome. Aliment Pharmacol Ther. 2004;20 Suppl 2:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 134]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
24.  Nessim SJ, Komenda P, Rigatto C, Verrelli M, Sood MM. Frequency and microbiology of peritonitis and exit-site infection among obese peritoneal dialysis patients. Perit Dial Int. 2012;33:167-174.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Paiva LV, Nomura RM, Dias MC, Zugaib M. Maternal obesity in high-risk pregnancies and postpartum infectious complications. Rev Assoc Med Bras. 2012;58:453-458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
26.  Easterbrook JD, Dunfee RL, Schwartzman LM, Jagger BW, Sandouk A, Kash JC, Memoli MJ, Taubenberger JK. Obese mice have increased morbidity and mortality compared to non-obese mice during infection with the 2009 pandemic H1N1 influenza virus. Influenza Other Respi Viruses. 2011;5:418-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 53]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
27.  Kerkhoffs GM, Servien E, Dunn W, Dahm D, Bramer JA, Haverkamp D. The influence of obesity on the complication rate and outcome of total knee arthroplasty: a meta-analysis and systematic literature review. J Bone Joint Surg Am. 2012;94:1839-1844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in F6Publishing: 350]  [Article Influence: 29.2]  [Reference Citation Analysis (0)]
28.  Dowsey MM, Choong PF. Obesity is a major risk factor for prosthetic infection after primary hip arthroplasty. Clin Orthop Relat Res. 2008;466:153-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 207]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
29.  Dorner TE, Schwarz F, Kranz A, Freidl W, Rieder A, Gisinger C. Body mass index and the risk of infections in institutionalised geriatric patients. Br J Nutr. 2010;103:1830-1835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 25]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
30.  Federico A, D’Aiuto E, Borriello F, Barra G, Gravina AG, Romano M, De Palma R. Fat: a matter of disturbance for the immune system. World J Gastroenterol. 2010;16:4762-4772.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 49]  [Cited by in F6Publishing: 52]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
31.  Tarantino G, Conca P, Pasanisi F, Ariello M, Mastrolia M, Arena A, Tarantino M, Scopacasa F, Vecchione R. Could inflammatory markers help diagnose nonalcoholic steatohepatitis. Eur J Gastroenterol Hepatol. 2009;21:504-511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 126]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
32.  Barbato A, Iacone R, Tarantino G, Russo O, Sorrentino P, Avallone S, Galletti F, Farinaro E, Della Valle E, Strazzullo P. Relationships of PAI-1 levels to central obesity and liver steatosis in a sample of adult male population in southern Italy. Intern Emerg Med. 2009;4:315-323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
33.  Stanton MC, Chen SC, Jackson JV, Rojas-Triana A, Kinsley D, Cui L, Fine JS, Greenfeder S, Bober LA, Jenh CH. Inflammatory Signals shift from adipose to liver during high fat feeding and influence the development of steatohepatitis in mice. J Inflamm (Lond). 2011;8:8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 97]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
34.  Wolowczuk I, Verwaerde C, Viltart O, Delanoye A, Delacre M, Pot B, Grangette C. Feeding our immune system: impact on metabolism. Clin Dev Immunol. 2008;2008:639803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 157]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
35.  Lamas O, Martínez JA, Marti A. Energy restriction restores the impaired immune response in overweight (cafeteria) rats. J Nutr Biochem. 2004;15:418-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 50]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
36.  Kahraman A, Schlattjan M, Kocabayoglu P, Yildiz-Meziletoglu S, Schlensak M, Fingas CD, Wedemeyer I, Marquitan G, Gieseler RK, Baba HA. Major histocompatibility complex class I-related chains A and B (MIC A/B): a novel role in nonalcoholic steatohepatitis. Hepatology. 2010;51:92-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 82]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
37.  Radaeva S, Sun R, Jaruga B, Nguyen VT, Tian Z, Gao B. Natural killer cells ameliorate liver fibrosis by killing activated stellate cells in NKG2D-dependent and tumor necrosis factor-related apoptosis-inducing ligand-dependent manners. Gastroenterology. 2006;130:435-452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 435]  [Cited by in F6Publishing: 441]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
38.  Jeong WI, Park O, Radaeva S, Gao B. STAT1 inhibits liver fibrosis in mice by inhibiting stellate cell proliferation and stimulating NK cell cytotoxicity. Hepatology. 2006;44:1441-1451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in F6Publishing: 213]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
39.  Safadi R, Friedman SL. Hepatic fibrosis-role of hepatic stellate cell activation. MedGenMed. 2002;4:27.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Jeong WI, Park O, Gao B. Abrogation of the antifibrotic effects of natural killer cells/interferon-gamma contributes to alcohol acceleration of liver fibrosis. Gastroenterology. 2008;134:248-258.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 180]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
41.  Guebre-Xabier M, Yang S, Lin HZ, Schwenk R, Krzych U, Diehl AM. Altered hepatic lymphocyte subpopulations in obesity-related murine fatty livers: potential mechanism for sensitization to liver damage. Hepatology. 2000;31:633-640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 154]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
42.  Li Z, Soloski MJ, Diehl AM. Dietary factors alter hepatic innate immune system in mice with nonalcoholic fatty liver disease. Hepatology. 2005;42:880-885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 232]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
43.  Kremer M, Thomas E, Milton RJ, Perry AW, van Rooijen N, Wheeler MD, Zacks S, Fried M, Rippe RA, Hines IN. Kupffer cell and interleukin-12-dependent loss of natural killer T cells in hepatosteatosis. Hepatology. 2010;51:130-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 119]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
44.  Maina V, Sutti S, Locatelli I, Vidali M, Mombello C, Bozzola C, Albano E. Bias in macrophage activation pattern influences non-alcoholic steatohepatitis (NASH) in mice. Clin Sci (Lond). 2012;122:545-553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 56]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
45.  Li Z, Oben JA, Yang S, Lin H, Stafford EA, Soloski MJ, Thomas SA, Diehl AM. Norepinephrine regulates hepatic innate immune system in leptin-deficient mice with nonalcoholic steatohepatitis. Hepatology. 2004;40:434-441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 87]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
46.  Weaver CT, Hatton RD. Interplay between the TH17 and TReg cell lineages: a (co-)evolutionary perspective. Nat Rev Immunol. 2009;9:883-889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 296]  [Cited by in F6Publishing: 308]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
47.  Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 Cells. Annu Rev Immunol. 2009;27:485-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3754]  [Cited by in F6Publishing: 3614]  [Article Influence: 240.9]  [Reference Citation Analysis (0)]
48.  Ma X, Hua J, Mohamood AR, Hamad AR, Ravi R, Li Z. A high-fat diet and regulatory T cells influence susceptibility to endotoxin-induced liver injury. Hepatology. 2007;46:1519-1529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 129]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
49.  Tang Y, Bian Z, Zhao L, Liu Y, Liang S, Wang Q, Han X, Peng Y, Chen X, Shen L. Interleukin-17 exacerbates hepatic steatosis and inflammation in non-alcoholic fatty liver disease. Clin Exp Immunol. 2011;166:281-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 220]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
50.  Diehl AM. Nonalcoholic steatosis and steatohepatitis IV. Nonalcoholic fatty liver disease abnormalities in macrophage function and cytokines. Am J Physiol Gastrointest Liver Physiol. 2002;282:G1-G5.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Park JW, Jeong G, Kim SJ, Kim MK, Park SM. Predictors reflecting the pathological severity of non-alcoholic fatty liver disease: comprehensive study of clinical and immunohistochemical findings in younger Asian patients. J Gastroenterol Hepatol. 2007;22:491-497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 64]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
52.  Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature. 1997;389:610-614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1684]  [Cited by in F6Publishing: 1604]  [Article Influence: 59.4]  [Reference Citation Analysis (0)]
53.  Koca SS, Bahcecioglu IH, Poyrazoglu OK, Ozercan IH, Sahin K, Ustundag B. The treatment with antibody of TNF-alpha reduces the inflammation, necrosis and fibrosis in the non-alcoholic steatohepatitis induced by methionine- and choline-deficient diet. Inflammation. 2008;31:91-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 105]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
54.  Dandona P, Weinstock R, Thusu K, Abdel-Rahman E, Aljada A, Wadden T. Tumor necrosis factor-alpha in sera of obese patients: fall with weight loss. J Clin Endocrinol Metab. 1998;83:2907-2910.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Hui JM, Hodge A, Farrell GC, Kench JG, Kriketos A, George J. Beyond insulin resistance in NASH: TNF-alpha or adiponectin. Hepatology. 2004;40:46-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 671]  [Cited by in F6Publishing: 668]  [Article Influence: 33.4]  [Reference Citation Analysis (0)]
56.  Valenti L, Fracanzani AL, Dongiovanni P, Santorelli G, Branchi A, Taioli E, Fiorelli G, Fargion S. Tumor necrosis factor alpha promoter polymorphisms and insulin resistance in nonalcoholic fatty liver disease. Gastroenterology. 2002;122:274-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 231]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
57.  Coulon S, Francque S, Colle I, Verrijken A, Blomme B, Heindryckx F, De Munter S, Prawitt J, Caron S, Staels B. Evaluation of inflammatory and angiogenic factors in patients with non-alcoholic fatty liver disease. Cytokine. 2012;59:442-449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 80]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
58.  Kitamura K, Nakamoto Y, Akiyama M, Fujii C, Kondo T, Kobayashi K, Kaneko S, Mukaida N. Pathogenic roles of tumor necrosis factor receptor p55-mediated signals in dimethylnitrosamine-induced murine liver fibrosis. Lab Invest. 2002;82:571-583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 52]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
59.  Zhang W, Kudo H, Kawai K, Fujisaka S, Usui I, Sugiyama T, Tsukada K, Chen N, Takahara T. Tumor necrosis factor-alpha accelerates apoptosis of steatotic hepatocytes from a murine model of non-alcoholic fatty liver disease. Biochem Biophys Res Commun. 2010;391:1731-1736.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 43]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
60.  Chavez-Tapia NC, Rosso N, Tiribelli C. Effect of intracellular lipid accumulation in a new model of non-alcoholic fatty liver disease. BMC Gastroenterol. 2012;12:20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 98]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
61.  Tsujimoto T, Kawaratani H, Kitazawa T, Uemura M, Fukui H. Innate immune reactivity of the ileum-liver axis in nonalcoholic steatohepatitis. Dig Dis Sci. 2012;57:1144-1151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
62.  Lucero D, Zago V, López GI, Graffigna M, Fainboim H, Miksztowicz V, Meroño T, Belli S, Levalle O, Wikinski R. Pro-inflammatory and atherogenic circulating factors in non-alcoholic fatty liver disease associated to metabolic syndrome. Clin Chim Acta. 2011;412:143-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
63.  Lebensztejn DM, Kowalczuk D, Tarasów E, Skiba E, Kaczmarski M. Tumor necrosis factor alpha and its soluble receptors in obese children with NAFLD. Adv Med Sci. 2010;55:74-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
64.  Jarrar MH, Baranova A, Collantes R, Ranard B, Stepanova M, Bennett C, Fang Y, Elariny H, Goodman Z, Chandhoke V. Adipokines and cytokines in non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2008;27:412-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 303]  [Cited by in F6Publishing: 306]  [Article Influence: 19.1]  [Reference Citation Analysis (0)]
65.  Kishimoto T. IL-6: from its discovery to clinical applications. Int Immunol. 2010;22:347-352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 525]  [Cited by in F6Publishing: 548]  [Article Influence: 39.1]  [Reference Citation Analysis (0)]
66.  Kopp HP, Kopp CW, Festa A, Krzyzanowska K, Kriwanek S, Minar E, Roka R, Schernthaner G. Impact of weight loss on inflammatory proteins and their association with the insulin resistance syndrome in morbidly obese patients. Arterioscler Thromb Vasc Biol. 2003;23:1042-1047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 272]  [Cited by in F6Publishing: 264]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
67.  Cai D, Yuan M, Frantz DF, Melendez PA, Hansen L, Lee J, Shoelson SE. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Nat Med. 2005;11:183-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1612]  [Cited by in F6Publishing: 1671]  [Article Influence: 87.9]  [Reference Citation Analysis (0)]
68.  Kugelmas M, Hill DB, Vivian B, Marsano L, McClain CJ. Cytokines and NASH: a pilot study of the effects of lifestyle modification and vitamin E. Hepatology. 2003;38:413-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 375]  [Cited by in F6Publishing: 353]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
69.  Tarantino G, Marra M, Contaldo F, Pasanisi F. Basal metabolic rate in morbidly obese patients with non-alcoholic fatty liver disease. Clin Invest Med. 2008;31:E24-E29.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Cressman DE, Greenbaum LE, DeAngelis RA, Ciliberto G, Furth EE, Poli V, Taub R. Liver failure and defective hepatocyte regeneration in interleukin-6-deficient mice. Science. 1996;274:1379-1383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1181]  [Cited by in F6Publishing: 1172]  [Article Influence: 41.9]  [Reference Citation Analysis (0)]
71.  El-Assal O, Hong F, Kim WH, Radaeva S, Gao B. IL-6-deficient mice are susceptible to ethanol-induced hepatic steatosis: IL-6 protects against ethanol-induced oxidative stress and mitochondrial permeability transition in the liver. Cell Mol Immunol. 2004;1:205-211.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Wieckowska A, Papouchado BG, Li Z, Lopez R, Zein NN, Feldstein AE. Increased hepatic and circulating interleukin-6 levels in human nonalcoholic steatohepatitis. Am J Gastroenterol. 2008;103:1372-1379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 395]  [Cited by in F6Publishing: 422]  [Article Influence: 26.4]  [Reference Citation Analysis (0)]
73.  Tilg H, Moschen AR. Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol. 2006;6:772-783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2065]  [Cited by in F6Publishing: 2136]  [Article Influence: 118.7]  [Reference Citation Analysis (0)]
74.  La Cava A, Matarese G. The weight of leptin in immunity. Nat Rev Immunol. 2004;4:371-379.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Kolak M, Westerbacka J, Velagapudi VR, Wågsäter D, Yetukuri L, Makkonen J, Rissanen A, Häkkinen AM, Lindell M, Bergholm R. Adipose tissue inflammation and increased ceramide content characterize subjects with high liver fat content independent of obesity. Diabetes. 2007;56:1960-1968.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 241]  [Cited by in F6Publishing: 244]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
76.  Procaccini C, Jirillo E, Matarese G. Leptin as an immunomodulator. Mol Aspects Med. 2012;33:35-45.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Angulo P, Alba LM, Petrovic LM, Adams LA, Lindor KD, Jensen MD. Leptin, insulin resistance, and liver fibrosis in human nonalcoholic fatty liver disease. J Hepatol. 2004;41:943-949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 141]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
78.  Tarantino G, Conca P, Riccio A, Tarantino M, Di Minno MN, Chianese D, Pasanisi F, Contaldo F, Scopacasa F, Capone D. Enhanced serum concentrations of transforming growth factor-beta1 in simple fatty liver: is it really benign. J Transl Med. 2008;6:72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 68]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
79.  den Boer MA, Voshol PJ, Schröder-van der Elst JP, Korsheninnikova E, Ouwens DM, Kuipers F, Havekes LM, Romijn JA. Endogenous interleukin-10 protects against hepatic steatosis but does not improve insulin sensitivity during high-fat feeding in mice. Endocrinology. 2006;147:4553-4558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 63]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
80.  Mager DR, Yap J, Rodriguez-Dimitrescu C, Mazurak V, Ball G, Gilmour S. Anthropometric measures of visceral and subcutaneous fat are important in the determination of metabolic dysregulation in boys and girls at risk for nonalcoholic fatty liver disease. Nutr Clin Pract. 2013;28:101-111.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Cintra DE, Pauli JR, Araújo EP, Moraes JC, de Souza CT, Milanski M, Morari J, Gambero A, Saad MJ, Velloso LA. Interleukin-10 is a protective factor against diet-induced insulin resistance in liver. J Hepatol. 2008;48:628-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 122]  [Article Influence: 7.6]  [Reference Citation Analysis (1)]
82.  Brun P, Castagliuolo I, Di Leo V, Buda A, Pinzani M, Palù G, Martines D. Increased intestinal permeability in obese mice: new evidence in the pathogenesis of nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver Physiol. 2007;292:G518-G525.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 592]  [Cited by in F6Publishing: 600]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
83.  Kant P, Fazakerley R, Hull MA. Faecal calprotectin levels before and after weight loss in obese and overweight subjects. Int J Obes (Lond). 2013;37:317-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 23]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
84.  Catalán V, Gómez-Ambrosi J, Rodríguez A, Ramírez B, Rotellar F, Valentí V, Silva C, Gil MJ, Fernández-Real JM, Salvador J. Increased levels of calprotectin in obesity are related to macrophage content: impact on inflammation and effect of weight loss. Mol Med. 2011;17:1157-1167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 91]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
85.  Mortensen OH, Nielsen AR, Erikstrup C, Plomgaard P, Fischer CP, Krogh-Madsen R, Lindegaard B, Petersen AM, Taudorf S, Pedersen BK. Calprotectin--a novel marker of obesity. PLoS One. 2009;4:e7419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 87]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
86.  Furness JB, Kunze WA, Clerc N. Nutrient tasting and signaling mechanisms in the gut. II. The intestine as a sensory organ: neural, endocrine, and immune responses. Am J Physiol. 1999;277:G922-G928.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Dunlop SP, Hebden J, Campbell E, Naesdal J, Olbe L, Perkins AC, Spiller RC. Abnormal intestinal permeability in subgroups of diarrhea-predominant irritable bowel syndromes. Am J Gastroenterol. 2006;101:1288-1294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 336]  [Cited by in F6Publishing: 342]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
88.  Chadwick VS, Chen W, Shu D, Paulus B, Bethwaite P, Tie A, Wilson I. Activation of the mucosal immune system in irritable bowel syndrome. Gastroenterology. 2002;122:1778-1783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 564]  [Cited by in F6Publishing: 563]  [Article Influence: 25.6]  [Reference Citation Analysis (0)]
89.  Cremon C, Gargano L, Morselli-Labate AM, Santini D, Cogliandro RF, De Giorgio R, Stanghellini V, Corinaldesi R, Barbara G. Mucosal immune activation in irritable bowel syndrome: gender-dependence and association with digestive symptoms. Am J Gastroenterol. 2009;104:392-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 244]  [Cited by in F6Publishing: 259]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
90.  Spiller RC, Jenkins D, Thornley JP, Hebden JM, Wright T, Skinner M, Neal KR. Increased rectal mucosal enteroendocrine cells, T lymphocytes, and increased gut permeability following acute Campylobacter enteritis and in post-dysenteric irritable bowel syndrome. Gut. 2000;47:804-811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 805]  [Cited by in F6Publishing: 792]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
91.  Ohman L, Isaksson S, Lundgren A, Simrén M, Sjövall H. A controlled study of colonic immune activity and beta7+ blood T lymphocytes in patients with irritable bowel syndrome. Clin Gastroenterol Hepatol. 2005;3:980-986.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
92.  Walker MM, Talley NJ, Prabhakar M, Pennaneac’h CJ, Aro P, Ronkainen J, Storskrubb T, Harmsen WS, Zinsmeister AR, Agreus L. Duodenal mastocytosis, eosinophilia and intraepithelial lymphocytosis as possible disease markers in the irritable bowel syndrome and functional dyspepsia. Aliment Pharmacol Ther. 2009;29:765-773.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Akbar A, Yiangou Y, Facer P, Walters JR, Anand P, Ghosh S. Increased capsaicin receptor TRPV1-expressing sensory fibres in irritable bowel syndrome and their correlation with abdominal pain. Gut. 2008;57:923-929.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Barbara G, Stanghellini V, De Giorgio R, Cremon C, Cottrell GS, Santini D, Pasquinelli G, Morselli-Labate AM, Grady EF, Bunnett NW. Activated mast cells in proximity to colonic nerves correlate with abdominal pain in irritable bowel syndrome. Gastroenterology. 2004;126:693-702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1004]  [Cited by in F6Publishing: 973]  [Article Influence: 48.7]  [Reference Citation Analysis (0)]
95.  Barbara G, Cremon C, Carini G, Bellacosa L, Zecchi L, De Giorgio R, Corinaldesi R, Stanghellini V. The immune system in irritable bowel syndrome. J Neurogastroenterol Motil. 2011;17:349-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 135]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
96.  Braak B, Klooker TK, Wouters MM, Welting O, van der Loos CM, Stanisor OI, van Diest S, van den Wijngaard RM, Boeckxstaens GE. Mucosal immune cell numbers and visceral sensitivity in patients with irritable bowel syndrome: is there any relationship. Am J Gastroenterol. 2012;107:715-726.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 62]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
97.  Motzer SA, Jarrett M, Heitkemper MM, Tsuji J. Natural killer cell function and psychological distress in women with and without irritable bowel syndrome. Biol Res Nurs. 2002;4:31-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 21]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
98.  Kristjánsson G, Venge P, Wanders A, Lööf L, Hällgren R. Clinical and subclinical intestinal inflammation assessed by the mucosal patch technique: studies of mucosal neutrophil and eosinophil activation in inflammatory bowel diseases and irritable bowel syndrome. Gut. 2004;53:1806-1812.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  O’Sullivan M, Clayton N, Breslin NP, Harman I, Bountra C, McLaren A, O’Morain CA. Increased mast cells in the irritable bowel syndrome. Neurogastroenterol Motil. 2000;12:449-457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 340]  [Cited by in F6Publishing: 365]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
100.  Roth J, Vogl T, Sorg C, Sunderkötter C. Phagocyte-specific S100 proteins: a novel group of proinflammatory molecules. Trends Immunol. 2003;24:155-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
101.  Grad C, David L, Portincasa P, Dumitraşcu DL. Diagnostic value of calprotectin in irritable bowel syndrome and in inflammatory bowel disease. Rom J Intern Med. 2012;50:3-6.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Keohane J, O’Mahony C, O’Mahony L, O’Mahony S, Quigley EM, Shanahan F. Irritable bowel syndrome-type symptoms in patients with inflammatory bowel disease: a real association or reflection of occult inflammation. Am J Gastroenterol. 2010;105:1788, 1789-1794; quiz 1795.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 167]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
103.  Shulman RJ, Eakin MN, Czyzewski DI, Jarrett M, Ou CN. Increased gastrointestinal permeability and gut inflammation in children with functional abdominal pain and irritable bowel syndrome. J Pediatr. 2008;153:646-650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 140]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
104.  Ohman L, Isaksson S, Lindmark AC, Posserud I, Stotzer PO, Strid H, Sjövall H, Simrén M. T-cell activation in patients with irritable bowel syndrome. Am J Gastroenterol. 2009;104:1205-1212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 116]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
105.  Holmén N, Isaksson S, Simrén M, Sjövall H, Ohman L. CD4+CD25+ regulatory T cells in irritable bowel syndrome patients. Neurogastroenterol Motil. 2007;19:119-125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
106.  Andoh A, Ogawa A, Bamba S, Fujiyama Y. Interaction between interleukin-17-producing CD4+ T cells and colonic subepithelial myofibroblasts: what are they doing in mucosal inflammation. J Gastroenterol. 2007;42 Suppl 17:29-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
107.  Scalera A, Loguercio C. Focus on irritable bowel syndrome. Eur Rev Med Pharmacol Sci. 2012;16:1155-1171.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Liebregts T, Adam B, Bredack C, Röth A, Heinzel S, Lester S, Downie-Doyle S, Smith E, Drew P, Talley NJ. Immune activation in patients with irritable bowel syndrome. Gastroenterology. 2007;132:913-920.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 452]  [Cited by in F6Publishing: 493]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
109.  Scully P, McKernan DP, Keohane J, Groeger D, Shanahan F, Dinan TG, Quigley EM. Plasma cytokine profiles in females with irritable bowel syndrome and extra-intestinal co-morbidity. Am J Gastroenterol. 2010;105:2235-2243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 129]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
110.  Dinan TG, Quigley EM, Ahmed SM, Scully P, O’Brien S, O’Mahony L, O’Mahony S, Shanahan F, Keeling PW. Hypothalamic-pituitary-gut axis dysregulation in irritable bowel syndrome: plasma cytokines as a potential biomarker. Gastroenterology. 2006;130:304-311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 440]  [Cited by in F6Publishing: 440]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
111.  Dinan TG, Clarke G, Quigley EM, Scott LV, Shanahan F, Cryan J, Cooney J, Keeling PW. Enhanced cholinergic-mediated increase in the pro-inflammatory cytokine IL-6 in irritable bowel syndrome: role of muscarinic receptors. Am J Gastroenterol. 2008;103:2570-2576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 108]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
112.  Schmulson M, Pulido-London D, Rodriguez O, Morales-Rochlin N, Martinez-García R, Gutierrez-Ruiz MC, López-Alvarenga JC, Robles-Díaz G, Gutiérrez-Reyes G. Lower serum IL-10 is an independent predictor of IBS among volunteers in Mexico. Am J Gastroenterol. 2012;107:747-753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 40]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
113.  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: 38]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
114.  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)]
115.  O’Malley D, Liston M, Hyland NP, Dinan TG, Cryan JF. Colonic soluble mediators from the maternal separation model of irritable bowel syndrome activate submucosal neurons via an interleukin-6-dependent mechanism. Am J Physiol Gastrointest Liver Physiol. 2011;300:G241-G252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 57]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
116.  Macsharry J, O’Mahony L, Fanning A, Bairead E, Sherlock G, Tiesman J, Fulmer A, Kiely B, Dinan TG, Shanahan F. Mucosal cytokine imbalance in irritable bowel syndrome. Scand J Gastroenterol. 2008;43:1467-1476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 125]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
117.  Collins SM, McHugh K, Jacobson K, Khan I, Riddell R, Murase K, Weingarten HP. Previous inflammation alters the response of the rat colon to stress. Gastroenterology. 1996;111:1509-1515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 102]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
118.  Sun X, Lan C, An Y, Sun Y. Colonic expression of Runx3 protein and TGF-β(1) and their correlation in patients with irritable bowel syndrome. Asian Pac J Trop Med. 2011;4:547-549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
119.  Chen J, Zhang Y, Deng Z. Imbalanced shift of cytokine expression between T helper 1 and T helper 2 (Th1/Th2) in intestinal mucosa of patients with post-infectious irritable bowel syndrome. BMC Gastroenterol. 2012;12:91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 52]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
120.  Ortiz-Lucas M, Saz-Peiró P, Sebastián-Domingo JJ. Irritable bowel syndrome immune hypothesis. Part two: the role of cytokines. Rev Esp Enferm Dig. 2010;102:711-717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 16]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
121.  Gwee KA, Collins SM, Read NW, Rajnakova A, Deng Y, Graham JC, McKendrick MW, Moochhala SM. Increased rectal mucosal expression of interleukin 1beta in recently acquired post-infectious irritable bowel syndrome. Gut. 2003;52:523-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 323]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
122.  Hughes PA, Harrington AM, Castro J, Liebregts T, Adam B, Grasby DJ, Isaacs NJ, Maldeniya L, Martin CM, Persson J. Sensory neuro-immune interactions differ between Irritable Bowel Syndrome subtypes. Gut. 2012;Jul 27; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 149]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
123.  Mizutani T, Akiho H, Khan WI, Murao H, Ogino H, Kanayama K, Nakamura K, Takayanagi R. Persistent gut motor dysfunction in a murine model of T-cell-induced enteropathy. Neurogastroenterol Motil. 2010;22:196-203, e65.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Kindt S, Van Oudenhove L, Broekaert D, Kasran A, Ceuppens JL, Bossuyt X, Fischler B, Tack J. Immune dysfunction in patients with functional gastrointestinal disorders. Neurogastroenterol Motil. 2009;21:389-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 111]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
125.  Hermann GE, Holmes GM, Rogers RC. TNF(alpha) modulation of visceral and spinal sensory processing. Curr Pharm Des. 2005;11:1391-1409.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
126.  Hermann GE, Rogers RC. TNFalpha: a trigger of autonomic dysfunction. Neuroscientist. 2008;14:53-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 54]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
127.  O’Malley D, Dinan TG, Cryan JF. Altered expression and secretion of colonic interleukin-6 in a stress-sensitive animal model of brain-gut axis dysfunction. J Neuroimmunol. 2011;235:48-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 27]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
128.  McKernan DP, Gaszner G, Quigley EM, Cryan JF, Dinan TG. Altered peripheral toll-like receptor responses in the irritable bowel syndrome. Aliment Pharmacol Ther. 2011;33:1045-1052.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 98]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
129.  Al-Sadi R, Boivin M, Ma T. Mechanism of cytokine modulation of epithelial tight junction barrier. Front Biosci (Landmark Ed). 2009;14:2765-2778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 344]  [Cited by in F6Publishing: 414]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
130.  Jumpertz R, Le DS, Turnbaugh PJ, Trinidad C, Bogardus C, Gordon JI, Krakoff J. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am J Clin Nutr. 2011;94:58-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 801]  [Cited by in F6Publishing: 782]  [Article Influence: 60.2]  [Reference Citation Analysis (0)]
131.  Ashida H, Ogawa M, Kim M, Mimuro H, Sasakawa C. Bacteria and host interactions in the gut epithelial barrier. Nat Chem Biol. 2012;8:36-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 218]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
132.  Tappenden KA, Deutsch AS. The physiological relevance of the intestinal microbiota--contributions to human health. J Am Coll Nutr. 2007;26:679S-683S.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Gigante G, Tortora A, Ianiro G, Ojetti V, Purchiaroni F, Campanale M, Cesario V, Scarpellini E, Gasbarrini A. Role of gut microbiota in food tolerance and allergies. Dig Dis. 2011;29:540-549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
134.  Fukata M, Vamadevan AS, Abreu MT. Toll-like receptors (TLRs) and Nod-like receptors (NLRs) in inflammatory disorders. Semin Immunol. 2009;21:242-253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 217]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
135.  Maher JJ, Leon P, Ryan JC. Beyond insulin resistance: Innate immunity in nonalcoholic steatohepatitis. Hepatology. 2008;48:670-678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 160]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
136.  Rodríguez-Fandiño O, Hernández-Ruiz J, Schmulson M. From cytokines to toll-like receptors and beyond-current knowledge and future research needs in irritable bowel syndrome. J Neurogastroenterol Motil. 2010;16:363-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
137.  Miura K, Seki E, Ohnishi H, Brenner DA. Role of toll-like receptors and their downstream molecules in the development of nonalcoholic Fatty liver disease. Gastroenterol Res Pract. 2010;2010:362847.  [PubMed]  [DOI]  [Cited in This Article: ]
138.  Abreu MT. Toll-like receptor signalling in the intestinal epithelium: how bacterial recognition shapes intestinal function. Nat Rev Immunol. 2010;10:131-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 856]  [Cited by in F6Publishing: 866]  [Article Influence: 61.9]  [Reference Citation Analysis (0)]
139.  Ohman L, Lindmark AC, Isaksson S, Posserud I, Strid H, Sjövall H, Simrén M. Increased TLR2 expression on blood monocytes in irritable bowel syndrome patients. Eur J Gastroenterol Hepatol. 2012;24:398-405.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  McKernan DP, Nolan A, Brint EK, O’Mahony SM, Hyland NP, Cryan JF, Dinan TG. Toll-like receptor mRNA expression is selectively increased in the colonic mucosa of two animal models relevant to irritable bowel syndrome. PLoS One. 2009;4:e8226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 55]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
141.  Brint EK, MacSharry J, Fanning A, Shanahan F, Quigley EM. Differential expression of toll-like receptors in patients with irritable bowel syndrome. Am J Gastroenterol. 2011;106:329-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 153]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
142.  Jin B, Sun T, Yu XH, Yang YX, Yeo AE. The effects of TLR activation on T-cell development and differentiation. Clin Dev Immunol. 2012;2012:836485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 128]  [Article Influence: 10.7]  [Reference Citation Analysis (1)]
143.  Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, Littman DR, Benoist C, Mathis D. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity. 2010;32:815-827.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1157]  [Cited by in F6Publishing: 1159]  [Article Influence: 82.8]  [Reference Citation Analysis (0)]
144.  Ather JL, Ckless K, Martin R, Foley KL, Suratt BT, Boyson JE, Fitzgerald KA, Flavell RA, Eisenbarth SC, Poynter ME. Serum amyloid A activates the NLRP3 inflammasome and promotes Th17 allergic asthma in mice. J Immunol. 2011;187:64-73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 185]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
145.  Atarashi K, Nishimura J, Shima T, Umesaki Y, Yamamoto M, Onoue M, Yagita H, Ishii N, Evans R, Honda K. ATP drives lamina propria T(H)17 cell differentiation. Nature. 2008;455:808-812.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 815]  [Cited by in F6Publishing: 815]  [Article Influence: 50.9]  [Reference Citation Analysis (0)]
146.  Zaph C, Du Y, Saenz SA, Nair MG, Perrigoue JG, Taylor BC, Troy AE, Kobuley DE, Kastelein RA, Cua DJ. Commensal-dependent expression of IL-25 regulates the IL-23-IL-17 axis in the intestine. J Exp Med. 2008;205:2191-2198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 225]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
147.  Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022-1023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5789]  [Cited by in F6Publishing: 5927]  [Article Influence: 348.6]  [Reference Citation Analysis (0)]
148.  Turnbaugh PJ, Gordon JI. The core gut microbiome, energy balance and obesity. J Physiol. 2009;587:4153-4158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 689]  [Cited by in F6Publishing: 670]  [Article Influence: 44.7]  [Reference Citation Analysis (0)]
149.  Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA. 2004;101:15718-15723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3967]  [Cited by in F6Publishing: 4117]  [Article Influence: 205.9]  [Reference Citation Analysis (4)]
150.  Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027-1031.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7820]  [Cited by in F6Publishing: 8109]  [Article Influence: 477.0]  [Reference Citation Analysis (1)]
151.  Ilan Y. Leaky gut and the liver: a role for bacterial translocation in nonalcoholic steatohepatitis. World J Gastroenterol. 2012;18:2609-2618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 135]  [Cited by in F6Publishing: 129]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
152.  Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava F, Tuohy KM, Chabo C. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761-1772.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4095]  [Cited by in F6Publishing: 4129]  [Article Influence: 242.9]  [Reference Citation Analysis (0)]
153.  de Wit N, Derrien M, Bosch-Vermeulen H, Oosterink E, Keshtkar S, Duval C, de Vogel-van den Bosch J, Kleerebezem M, Müller M, van der Meer R. Saturated fat stimulates obesity and hepatic steatosis and affects gut microbiota composition by an enhanced overflow of dietary fat to the distal intestine. Am J Physiol Gastrointest Liver Physiol. 2012;303:G589-G599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 283]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
154.  Spencer MD, Hamp TJ, Reid RW, Fischer LM, Zeisel SH, Fodor AA. Association between composition of the human gastrointestinal microbiome and development of fatty liver with choline deficiency. Gastroenterology. 2011;140:976-986.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 430]  [Cited by in F6Publishing: 514]  [Article Influence: 39.5]  [Reference Citation Analysis (0)]
155.  Collins SM, Denou E, Verdu EF, Bercik P. The putative role of the intestinal microbiota in the irritable bowel syndrome. Dig Liver Dis. 2009;41:850-853.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 84]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
156.  Husebye E, Hellström PM, Midtvedt T. Intestinal microflora stimulates myoelectric activity of rat small intestine by promoting cyclic initiation and aboral propagation of migrating myoelectric complex. Dig Dis Sci. 1994;39:946-956.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 126]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
157.  Bär F, Von Koschitzky H, Roblick U, Bruch HP, Schulze L, Sonnenborn U, Böttner M, Wedel T. Cell-free supernatants of Escherichia coli Nissle 1917 modulate human colonic motility: evidence from an in vitro organ bath study. Neurogastroenterol Motil. 2009;21:559-566, e16-17.  [PubMed]  [DOI]  [Cited in This Article: ]
158.  Guarino MP, Altomare A, Stasi E, Marignani M, Severi C, Alloni R, Dicuonzo G, Morelli L, Coppola R, Cicala M. Effect of acute mucosal exposure to Lactobacillus rhamnosus GG on human colonic smooth muscle cells. J Clin Gastroenterol. 2008;42 Suppl 3 Pt 2:S185-S190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 34]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
159.  Kamiya T, Wang L, Forsythe P, Goettsche G, Mao Y, Wang Y, Tougas G, Bienenstock J. Inhibitory effects of Lactobacillus reuteri on visceral pain induced by colorectal distension in Sprague-Dawley rats. Gut. 2006;55:191-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 161]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
160.  Balsari A, Ceccarelli A, Dubini F, Fesce E, Poli G. The fecal microbial population in the irritable bowel syndrome. Microbiologica. 1982;5:185-194.  [PubMed]  [DOI]  [Cited in This Article: ]
161.  Wyatt GM, Bayliss CE, Lakey AF, Bradley HK, Hunter JO, Jones VA. The faecal flora of two patients with food-related irritable bowel syndrome during challenge with symptom-provoking foods. J Med Microbiol. 1988;26:295-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
162.  Simrén M, Barbara G, Flint HJ, Spiegel BM, Spiller RC, Vanner S, Verdu EF, Whorwell PJ, Zoetendal EG. Intestinal microbiota in functional bowel disorders: a Rome foundation report. Gut. 2013;62:159-176.  [PubMed]  [DOI]  [Cited in This Article: ]
163.  Ford AC, Spiegel BM, Talley NJ, Moayyedi P. Small intestinal bacterial overgrowth in irritable bowel syndrome: systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2009;7:1279-1286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 276]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
164.  Ghoshal UC, Kumar S, Mehrotra M, Lakshmi C, Misra A. Frequency of small intestinal bacterial overgrowth in patients with irritable bowel syndrome and chronic non-specific diarrhea. J Neurogastroenterol Motil. 2010;16:40-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 60]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
165.  Thabane M, Kottachchi DT, Marshall JK. Systematic review and meta-analysis: The incidence and prognosis of post-infectious irritable bowel syndrome. Aliment Pharmacol Ther. 2007;26:535-544.  [PubMed]  [DOI]  [Cited in This Article: ]
166.  Tsukita S, Furuse M, Itoh M. Multifunctional strands in tight junctions. Nat Rev Mol Cell Biol. 2001;2:285-293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1851]  [Cited by in F6Publishing: 1834]  [Article Influence: 79.7]  [Reference Citation Analysis (0)]
167.  Farquhar MG, Palade GE. Junctional complexes in various epithelia. J Cell Biol. 1963;17:375-412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2475]  [Cited by in F6Publishing: 2656]  [Article Influence: 102.2]  [Reference Citation Analysis (0)]
168.  Förster C. Tight junctions and the modulation of barrier function in disease. Histochem Cell Biol. 2008;130:55-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 392]  [Cited by in F6Publishing: 424]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
169.  Groschwitz KR, Hogan SP. Intestinal barrier function: molecular regulation and disease pathogenesis. J Allergy Clin Immunol. 2009;124:3-20; quiz 21-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1145]  [Cited by in F6Publishing: 1056]  [Article Influence: 70.4]  [Reference Citation Analysis (1)]
170.  Chen ML, Pothoulakis C, LaMont JT. Protein kinase C signaling regulates ZO-1 translocation and increased paracellular flux of T84 colonocytes exposed to Clostridium difficile toxin A. J Biol Chem. 2002;277:4247-4254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 130]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
171.  Bruewer M, Luegering A, Kucharzik T, Parkos CA, Madara JL, Hopkins AM, Nusrat A. Proinflammatory cytokines disrupt epithelial barrier function by apoptosis-independent mechanisms. J Immunol. 2003;171:6164-6172.  [PubMed]  [DOI]  [Cited in This Article: ]
172.  Suzuki T, Yoshinaga N, Tanabe S. Interleukin-6 (IL-6) regulates claudin-2 expression and tight junction permeability in intestinal epithelium. J Biol Chem. 2011;286:31263-31271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 336]  [Cited by in F6Publishing: 377]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
173.  Cesaro C, Tiso A, Del Prete A, Cariello R, Tuccillo C, Cotticelli G, Del Vecchio Blanco C, Loguercio C. Gut microbiota and probiotics in chronic liver diseases. Dig Liver Dis. 2011;43:431-438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 120]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
174.  Sabaté JM, Jouët P, Harnois F, Mechler C, Msika S, Grossin M, Coffin B. High prevalence of small intestinal bacterial overgrowth in patients with morbid obesity: a contributor to severe hepatic steatosis. Obes Surg. 2008;18:371-377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 173]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
175.  Wigg AJ, Roberts-Thomson IC, Dymock RB, McCarthy PJ, Grose RH, Cummins AG. The role of small intestinal bacterial overgrowth, intestinal permeability, endotoxaemia, and tumour necrosis factor alpha in the pathogenesis of non-alcoholic steatohepatitis. Gut. 2001;48:206-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 610]  [Cited by in F6Publishing: 598]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
176.  Spruss A, Kanuri G, Stahl C, Bischoff SC, Bergheim I. Metformin protects against the development of fructose-induced steatosis in mice: role of the intestinal barrier function. Lab Invest. 2012;92:1020-1032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 123]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
177.  Miele L, Valenza V, La Torre G, Montalto M, Cammarota G, Ricci R, Mascianà R, Forgione A, Gabrieli ML, Perotti G. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology. 2009;49:1877-1887.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 938]  [Cited by in F6Publishing: 982]  [Article Influence: 65.5]  [Reference Citation Analysis (0)]
178.  Shanab AA, Scully P, Crosbie O, Buckley M, O’Mahony L, Shanahan F, Gazareen S, Murphy E, Quigley EM. Small intestinal bacterial overgrowth in nonalcoholic steatohepatitis: association with toll-like receptor 4 expression and plasma levels of interleukin 8. Dig Dis Sci. 2011;56:1524-1534.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 141]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
179.  Bellot P, Francés R, Such J. Pathological bacterial translocation in cirrhosis: pathophysiology, diagnosis and clinical implications. Liver Int. 2013;33:31-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 171]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
180.  Sakaguchi S, Takahashi S, Sasaki T, Kumagai T, Nagata K. Progression of alcoholic and non-alcoholic steatohepatitis: common metabolic aspects of innate immune system and oxidative stress. Drug Metab Pharmacokinet. 2011;26:30-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 92]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
181.  Ruiz AG, Casafont F, Crespo J, Cayón A, Mayorga M, Estebanez A, Fernadez-Escalante JC, Pons-Romero F. Lipopolysaccharide-binding protein plasma levels and liver TNF-alpha gene expression in obese patients: evidence for the potential role of endotoxin in the pathogenesis of non-alcoholic steatohepatitis. Obes Surg. 2007;17:1374-1380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 195]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
182.  Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, Geurts L, Naslain D, Neyrinck A, Lambert DM. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut. 2009;58:1091-1103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1735]  [Cited by in F6Publishing: 1740]  [Article Influence: 116.0]  [Reference Citation Analysis (0)]
183.  Lam YY, Ha CW, Campbell CR, Mitchell AJ, Dinudom A, Oscarsson J, Cook DI, Hunt NH, Caterson ID, Holmes AJ. Increased gut permeability and microbiota change associate with mesenteric fat inflammation and metabolic dysfunction in diet-induced obese mice. PLoS One. 2012;7:e34233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 377]  [Cited by in F6Publishing: 425]  [Article Influence: 35.4]  [Reference Citation Analysis (0)]
184.  Thabane M, Marshall JK. Post-infectious irritable bowel syndrome. World J Gastroenterol. 2009;15:3591-3596.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 101]  [Cited by in F6Publishing: 108]  [Article Influence: 7.2]  [Reference Citation Analysis (3)]
185.  Ohman L, Simrén M. Pathogenesis of IBS: role of inflammation, immunity and neuroimmune interactions. Nat Rev Gastroenterol Hepatol. 2010;7:163-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 399]  [Cited by in F6Publishing: 370]  [Article Influence: 26.4]  [Reference Citation Analysis (0)]
186.  Róka R, Rosztóczy A, Leveque M, Izbéki F, Nagy F, Molnár T, Lonovics J, Garcia-Villar R, Fioramonti J, Wittmann T. A pilot study of fecal serine-protease activity: a pathophysiologic factor in diarrhea-predominant irritable bowel syndrome. Clin Gastroenterol Hepatol. 2007;5:550-555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 95]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
187.  Martínez C, Vicario M, Ramos L, Lobo B, Mosquera JL, Alonso C, Sánchez A, Guilarte M, Antolín M, de Torres I. The jejunum of diarrhea-predominant irritable bowel syndrome shows molecular alterations in the tight junction signaling pathway that are associated with mucosal pathobiology and clinical manifestations. Am J Gastroenterol. 2012;107:736-746.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 157]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
188.  Kong WM, Gong J, Dong L, Xu JR. Changes of tight junction claudin-1, -3, -4 protein expression in the intestinal mucosa in patients with irritable bowel syndrome. Nanfang Yike Daxue Xuebao. 2007;27:1345-1347.  [PubMed]  [DOI]  [Cited in This Article: ]
189.  Khan WI, Collins SM. Gut motor function: immunological control in enteric infection and inflammation. Clin Exp Immunol. 2006;143:389-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 75]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
190.  Vallance BA, Blennerhassett PA, Collins SM. Increased intestinal muscle contractility and worm expulsion in nematode-infected mice. Am J Physiol. 1997;272:G321-G327.  [PubMed]  [DOI]  [Cited in This Article: ]
191.  Vallance BA, Galeazzi F, Collins SM, Snider DP. CD4 T cells and major histocompatibility complex class II expression influence worm expulsion and increased intestinal muscle contraction during Trichinella spiralis infection. Infect Immun. 1999;67:6090-6097.  [PubMed]  [DOI]  [Cited in This Article: ]
192.  Akiho H, Ihara E, Motomura Y, Nakamura K. Cytokine-induced alterations of gastrointestinal motility in gastrointestinal disorders. World J Gastrointest Pathophysiol. 2011;2:72-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 66]  [Cited by in F6Publishing: 68]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
193.  Mannon P, Reinisch W. Interleukin 13 and its role in gut defence and inflammation. Gut. 2012;61:1765-1773.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 80]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
194.  Sharkey KA, Mawe GM. Neuroimmune and epithelial interactions in intestinal inflammation. Curr Opin Pharmacol. 2002;2:669-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
195.  Motomura Y, Ghia JE, Wang H, Akiho H, El-Sharkawy RT, Collins M, Wan Y, McLaughlin JT, Khan WI. Enterochromaffin cell and 5-hydroxytryptamine responses to the same infectious agent differ in Th1 and Th2 dominant environments. Gut. 2008;57:475-481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 63]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
196.  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:G433-G448.  [PubMed]  [DOI]  [Cited in This Article: ]
197.  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;16:2352-2364.  [PubMed]  [DOI]  [Cited in This Article: ]
198.  Li J, Ma W, Wang S. Slower gastric emptying in high-fat diet induced obese rats is associated with attenuated plasma ghrelin and elevated plasma leptin and cholecystokinin concentrations. Regul Pept. 2011;171:53-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
199.  Covasa M, Ritter RC. Adaptation to high-fat diet reduces inhibition of gastric emptying by CCK and intestinal oleate. Am J Physiol Regul Integr Comp Physiol. 2000;278:R166-R170.  [PubMed]  [DOI]  [Cited in This Article: ]
200.  Suzuki S, Ramos EJ, Goncalves CG, Chen C, Meguid MM. Changes in GI hormones and their effect on gastric emptying and transit times after Roux-en-Y gastric bypass in rat model. Surgery. 2005;138:283-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 98]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
201.  Hyland NP, Rybicka JM, Ho W, Pittman QJ, Macnaughton WK, Sharkey KA. Adaptation of intestinal secretomotor function and nutrient absorption in response to diet-induced obesity. Neurogastroenterol Motil. 2010;22:602-e171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
202.  Fornari F, Callegari-Jacques SM, Dantas RO, Scarsi AL, Ruas LO, de Barros SG. Obese patients have stronger peristalsis and increased acid exposure in the esophagus. Dig Dis Sci. 2011;56:1420-1426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
203.  Little TJ, Horowitz M, Feinle-Bisset C. Modulation by high-fat diets of gastrointestinal function and hormones associated with the regulation of energy intake: implications for the pathophysiology of obesity. Am J Clin Nutr. 2007;86:531-541.  [PubMed]  [DOI]  [Cited in This Article: ]
204.  Vazquez Roque MI, Camilleri M, Stephens DA, Jensen MD, Burton DD, Baxter KL, Zinsmeister AR. Gastric sensorimotor functions and hormone profile in normal weight, overweight, and obese people. Gastroenterology. 2006;131:1717-1724.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 89]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
205.  Di Ciaula A, Wang DQ, Portincasa P. Gallbladder and gastric motility in obese newborns, pre-adolescents and adults. J Gastroenterol Hepatol. 2012;27:1298-1305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
206.  Xing J, Chen JD. Alterations of gastrointestinal motility in obesity. Obes Res. 2004;12:1723-1732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 106]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
207.  Madrid AM, Poniachik J, Quera R, Defilippi C. Small intestinal clustered contractions and bacterial overgrowth: a frequent finding in obese patients. Dig Dis Sci. 2011;56:155-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 45]  [Article Influence: 3.5]  [Reference Citation Analysis (1)]
208.  Lata J, Jurankova J, Kopacova M, Vitek P. Probiotics in hepatology. World J Gastroenterol. 2011;17:2890-2896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 49]  [Cited by in F6Publishing: 45]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
209.  Soza A, Riquelme A, González R, Alvarez M, Pérez-Ayuso RM, Glasinovic JC, Arrese M. Increased orocecal transit time in patients with nonalcoholic fatty liver disease. Dig Dis Sci. 2005;50:1136-1140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 31]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
210.  Nagasako CK, de Oliveira Figueiredo MJ, de Souza Almeida JR, Lorena SL, Akasaka HM, Pavan CR, Yamanaka A, Pereira TS, Soares EC, Mesquita MA. Investigation of autonomic function and orocecal transit time in patients with nonalcoholic cirrhosis and the potential influence of these factors on disease outcome. J Clin Gastroenterol. 2009;43:884-889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
211.  Haub S, Ritze Y, Ladel I, Saum K, Hubert A, Spruss A, Trautwein C, Bischoff SC. Serotonin receptor type 3 antagonists improve obesity-associated fatty liver disease in mice. J Pharmacol Exp Ther. 2011;339:790-798.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
212.  Haub S, Kanuri G, Volynets V, Brune T, Bischoff SC, Bergheim I. Serotonin reuptake transporter (SERT) plays a critical role in the onset of fructose-induced hepatic steatosis in mice. Am J Physiol Gastrointest Liver Physiol. 2010;298:G335-G344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 68]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
213.  Corsetti M, Caenepeel P, Fischler B, Janssens J, Tack J. Impact of coexisting irritable bowel syndrome on symptoms and pathophysiological mechanisms in functional dyspepsia. Am J Gastroenterol. 2004;99:1152-1159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 115]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
214.  Sarnelli G, Caenepeel P, Geypens B, Janssens J, Tack J. Symptoms associated with impaired gastric emptying of solids and liquids in functional dyspepsia. Am J Gastroenterol. 2003;98:783-788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 356]  [Cited by in F6Publishing: 345]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
215.  Evans PR, Bak YT, Shuter B, Hoschl R, Kellow JE. Gastroparesis and small bowel dysmotility in irritable bowel syndrome. Dig Dis Sci. 1997;42:2087-2093.  [PubMed]  [DOI]  [Cited in This Article: ]
216.  Gunnarsson J, Simrén M. Peripheral factors in the pathophysiology of irritable bowel syndrome. Dig Liver Dis. 2009;41:788-793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
217.  Larsson MH, Simrén M, Thomas EA, Bornstein JC, Lindström E, Sjövall H. Elevated motility-related transmucosal potential difference in the upper small intestine in the irritable bowel syndrome. Neurogastroenterol Motil. 2007;19:812-820.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 35]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
218.  Kellow JE, Phillips SF. Altered small bowel motility in irritable bowel syndrome is correlated with symptoms. Gastroenterology. 1987;92:1885-1893.  [PubMed]  [DOI]  [Cited in This Article: ]
219.  Kellow JE, Eckersley GM, Jones M. Enteric and central contributions to intestinal dysmotility in irritable bowel syndrome. Dig Dis Sci. 1992;37:168-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
220.  Dunlop SP, Jenkins D, Neal KR, Spiller RC. Relative importance of enterochromaffin cell hyperplasia, anxiety, and depression in postinfectious IBS. Gastroenterology. 2003;125:1651-1659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 418]  [Cited by in F6Publishing: 445]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
221.  Lee KJ, Kim YB, Kim JH, Kwon HC, Kim DK, Cho SW. The alteration of enterochromaffin cell, mast cell, and lamina propria T lymphocyte numbers in irritable bowel syndrome and its relationship with psychological factors. J Gastroenterol Hepatol. 2008;23:1689-1694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 119]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
222.  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 F6Publishing: 5]  [Reference Citation Analysis (0)]
223.  Spiller R, Garsed K. Infection, inflammation, and the irritable bowel syndrome. Dig Liver Dis. 2009;41:844-849.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 97]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
224.  Moses PL, Bannon C, Linden DR, Crowell MD, Sharkey KA, Mawe GM. Evidence for altered serotonin signalling in IBD and constipation predominant IBS. Amer J Gastroenterol. 2002;97:S274.  [PubMed]  [DOI]  [Cited in This Article: ]
225.  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)]
226.  Cremon C, Carini G, Wang B, Vasina V, Cogliandro RF, De Giorgio R, Stanghellini V, Grundy D, Tonini M, De Ponti F. Intestinal serotonin release, sensory neuron activation, and abdominal pain in irritable bowel syndrome. Am J Gastroenterol. 2011;106:1290-1298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 156]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
227.  Anitha M, Vijay-Kumar M, Sitaraman SV, Gewirtz AT, Srinivasan S. Gut microbial products regulate murine gastrointestinal motility via Toll-like receptor 4 signaling. Gastroenterology. 2012;143:1006-16.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 264]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
228.  Tattoli I, Petitta C, Scirocco A, Ammoscato F, Cicenia A, Severi C. Microbiota, innate immune system, and gastrointestinal muscle: ongoing studies. J Clin Gastroenterol. 2012;46 Suppl:S6-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
229.  Quigley EM. Microflora modulation of motility. J Neurogastroenterol Motil. 2011;17:140-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 85]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
230.  Pimentel M, Soffer EE, Chow EJ, Kong Y, Lin HC. Lower frequency of MMC is found in IBS subjects with abnormal lactulose breath test, suggesting bacterial overgrowth. Dig Dis Sci. 2002;47:2639-2643.  [PubMed]  [DOI]  [Cited in This Article: ]
231.  Capuron L, Miller AH. Immune system to brain signaling: neuropsychopharmacological implications. Pharmacol Ther. 2011;130:226-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 709]  [Cited by in F6Publishing: 750]  [Article Influence: 57.7]  [Reference Citation Analysis (0)]
232.  Kim DY, Camilleri M. Serotonin: a mediator of the brain-gut connection. Am J Gastroenterol. 2000;95:2698-2709.  [PubMed]  [DOI]  [Cited in This Article: ]
233.  Collins SM, Bercik P. The relationship between intestinal microbiota and the central nervous system in normal gastrointestinal function and disease. Gastroenterology. 2009;136:2003-2014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 390]  [Cited by in F6Publishing: 377]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
234.  Stasi C, Rosselli M, Bellini M, Laffi G, Milani S. Altered neuro-endocrine-immune pathways in the irritable bowel syndrome: the top-down and the bottom-up model. J Gastroenterol. 2012;47:1177-1185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 93]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
235.  Kyrou I, Chrousos GP, Tsigos C. Stress, visceral obesity, and metabolic complications. Ann N Y Acad Sci. 2006;1083:77-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 203]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
236.  Anisman H, Merali Z, Poulter MO, Hayley S. Cytokines as a precipitant of depressive illness: animal and human studies. Curr Pharm Des. 2005;11:963-972.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 140]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
237.  Raison CL, Woolwine BJ, Demetrashvili MF, Borisov AS, Weinreib R, Staab JP, Zajecka JM, Bruno CJ, Henderson MA, Reinus JF. Paroxetine for prevention of depressive symptoms induced by interferon-alpha and ribavirin for hepatitis C. Aliment Pharmacol Ther. 2007;25:1163-1174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 96]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
238.  De Souza CT, Araujo EP, Bordin S, Ashimine R, Zollner RL, Boschero AC, Saad MJ, Velloso LA. Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology. 2005;146:4192-4199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 799]  [Cited by in F6Publishing: 793]  [Article Influence: 41.7]  [Reference Citation Analysis (0)]
239.  Zhang X, Dong F, Ren J, Driscoll MJ, Culver B. High dietary fat induces NADPH oxidase-associated oxidative stress and inflammation in rat cerebral cortex. Exp Neurol. 2005;191:318-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 207]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
240.  Bilbo SD, Tsang V. Enduring consequences of maternal obesity for brain inflammation and behavior of offspring. FASEB J. 2010;24:2104-2115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 341]  [Cited by in F6Publishing: 360]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
241.  Ghareeb DA, Hafez HS, Hussien HM, Kabapy NF. Non-alcoholic fatty liver induces insulin resistance and metabolic disorders with development of brain damage and dysfunction. Metab Brain Dis. 2011;26:253-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
242.  Faith MS, Butryn M, Wadden TA, Fabricatore A, Nguyen AM, Heymsfield SB. Evidence for prospective associations among depression and obesity in population-based studies. Obes Rev. 2011;12:e438-e453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 209]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
243.  Herrera-Marquez R, Hernandez-Rodriguez J, Medina-Serrano J, Boyzo-Montes de Oca A, Manjarrez-Gutierrez G. Association of metabolic syndrome with reduced central serotonergic activity. Metab Brain Dis. 2011;26:29-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
244.  Tarantino G, Savastano S, Colao A, Polichetti G, Capone D. Urinary excretion of 5-hydroxy-3-indoleacetic acid in dystimic/depressed, adult obese women: what correlations to hepatic steatosis. Int J Immunopathol Pharmacol. 2011;24:769-779.  [PubMed]  [DOI]  [Cited in This Article: ]
245.  Bose M, Oliván B, Laferrère B. Stress and obesity: the role of the hypothalamic-pituitary-adrenal axis in metabolic disease. Curr Opin Endocrinol Diabetes Obes. 2009;16:340-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 210]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
246.  Targher G, Bertolini L, Rodella S, Zoppini G, Zenari L, Falezza G. Associations between liver histology and cortisol secretion in subjects with nonalcoholic fatty liver disease. Clin Endocrinol (Oxf). 2006;64:337-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 70]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
247.  Holt HB, Wild SH, Postle AD, Zhang J, Koster G, Umpleby M, Shojaee-Moradie F, Dewbury K, Wood PJ, Phillips DI. Cortisol clearance and associations with insulin sensitivity, body fat and fatty liver in middle-aged men. Diabetologia. 2007;50:1024-1032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 30]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
248.  Tomlinson JW, Moore J, Cooper MS, Bujalska I, Shahmanesh M, Burt C, Strain A, Hewison M, Stewart PM. Regulation of expression of 11beta-hydroxysteroid dehydrogenase type 1 in adipose tissue: tissue-specific induction by cytokines. Endocrinology. 2001;142:1982-1989.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 45]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
249.  Kaufman D, Banerji MA, Shorman I, Smith EL, Coplan JD, Rosenblum LA, Kral JG. Early-life stress and the development of obesity and insulin resistance in juvenile bonnet macaques. Diabetes. 2007;56:1382-1386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 108]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
250.  Fichna J, Storr MA. Brain-Gut Interactions in IBS. Front Pharmacol. 2012;3:127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 86]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
251.  Clarke G, Fitzgerald P, Cryan JF, Cassidy EM, Quigley EM, Dinan TG. Tryptophan degradation in irritable bowel syndrome: evidence of indoleamine 2,3-dioxygenase activation in a male cohort. BMC Gastroenterol. 2009;9:6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 94]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
252.  Clarke G, McKernan DP, Gaszner G, Quigley EM, Cryan JF, Dinan TG. A Distinct Profile of Tryptophan Metabolism along the Kynurenine Pathway Downstream of Toll-Like Receptor Activation in Irritable Bowel Syndrome. Front Pharmacol. 2012;3:90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 72]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
253.  Piche T, Saint-Paul MC, Dainese R, Marine-Barjoan E, Iannelli A, Montoya ML, Peyron JF, Czerucka D, Cherikh F, Filippi J. Mast cells and cellularity of the colonic mucosa correlated with fatigue and depression in irritable bowel syndrome. Gut. 2008;57:468-473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 141]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
254.  Tarantino G, Finelli C, Colao A, Capone D, Tarantino M, Grimaldi E, Chianese D, Gioia S, Pasanisi F, Contaldo F. Are hepatic steatosis and carotid intima media thickness associated in obese patients with normal or slightly elevated gamma-glutamyl-transferase. J Transl Med. 2012;10:50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]