Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Nov 14, 2014; 20(42): 15624-15631
Published online Nov 14, 2014. doi: 10.3748/wjg.v20.i42.15624
Gut microbiota-related complications in cirrhosis
Isabel Gómez-Hurtado, José Such, Rubén Francés, CIBERehd-Liver Unit, Hospital General Universitario Alicante, Alicante 03010, Spain
Yolanda Sanz, Instituto de Agroquímica y Tecnología de los Alimentos, Consejo Superior de Investigaciones Científicas (IATA-CSIC), Carrer Catedràtic Agustín Escardino Benlloch, 7, Paterna, València 46980, Spain
Author contributions: Gómez-Hurtado I performed manuscript writing; Such J performed clinical considerations and manuscript writing; Sanz Y performed microbiological considerations and manuscript wrtitng; Francés R performed design and manuscript writing.
Supported by Grants PI13/1443 from Instituto de Salud Carlos III, Madrid, Spain; and No. AGL2011-25169 from MINECO, Madrid, Spain
Correspondence to: Rubén Francés, PhD, CIBERehd-Liver Unit, Hospital General Universitario Alicante, C/Pintor Baeza 12, Alicante 03010, Spain. frances_rub@gva.es
Telephone: +34-965-913928 Fax: +34-965-913922
Received: February 28, 2014
Revised: April 16, 2014
Accepted: May 19, 2014
Published online: November 14, 2014

Abstract

Gut microbiota plays an important role in cirrhosis. The liver is constantly challenged with commensal bacteria and their products arriving through the portal vein in the so-called gut-liver axis. Bacterial translocation from the intestinal lumen through the intestinal wall and to mesenteric lymph nodes is facilitated by intestinal bacterial overgrowth, impairment in the permeability of the intestinal mucosal barrier, and deficiencies in local host immune defences. Deranged clearance of endogenous bacteria from portal and systemic circulation turns the gut into the major source of bacterial-related complications. Liver function may therefore be affected by alterations in the composition of the intestinal microbiota and a role for commensal flora has been evidenced in the pathogenesis of several complications arising in end-stage liver disease such as hepatic encephalopathy, splanchnic arterial vasodilatation and spontaneous bacterial peritonitis. The use of antibiotics is the main therapeutic pipeline in the management of these bacteria-related complications. However, other strategies aimed at preserving intestinal homeostasis through the use of pre-, pro- or symbiotic formulations are being studied in the last years. In this review, the role of intestinal microbiota in the development of the most frequent complications arising in cirrhosis and the different clinical and experimental studies conducted to prevent or improve these complications by modifying the gut microbiota composition are summarized.

Key Words: Cirrhosis, Encephalopathy, Portal hypertension, Spontaneous bacterial peritonitis, Microbiota, Bacterial translocation

Core tip: The close relationship between the most frequent complications arising in patients with cirrhosis and the gut microbiota has been intensively studied in the last years and has enhanced the relevance of the constant communication between the gut and the liver in the management of patients with cirrhosis. This review intends to integrate the existing knowledge of the role of intestinal microbiota plays in the development of these complications and the evidence on the possible efficacy of gut-microbiota modulating strategies to help in their management.



INTRODUCTION

Cirrhosis is the common end-stage liver histologic distortion for several hepatic diseases, characterized by the presence of regenerative nodules that causes portal hypertension. This fact in turn alters intestinal motility inducing intestinal bacterial overgrowth (IBO)[1]. The close relationship between the most frequent complications arising in patients with cirrhosis and the gut microbiota has been intensively studied in the last years and has enhanced the relevance of the constant communication between the gut and the liver in the management of patients with cirrhosis[2]. Complications such as hepatic encephalopathy (HE), spontaneous bacterial peritonitis (SBP) and variceal bleeding are directly caused or aggravated by the translocation of enteric bacteria or their products into the blood of cirrhotic patients.

The current pathogenic mechanism to explain the passage of bacteria or their products from the intestinal lumen through the intestinal barrier and to mesenteric lymph nodes is defined as bacterial translocation (BT)[3,4]. IBO, impairment in intestinal permeability, and deficiencies in local host immune defences are the major mechanisms postulated to favour BT in cirrhosis[5,6]. Any of these abnormalities deranges clearance of endogenous bacteria from portal and systemic circulation, turning the gut into the major source of bacterial-related complications.

The large bowel is the most densely populated natural environment known, containing roughly 1014 bacterial cells[7], which represents around ten times more microbial cells than eukaryotic cells. Gut microbiota is a large and diverse population of living microorganisms (approximately 500-1500 different bacterial species[8]), susceptible to environmental and pathophysiological alterations[9]. Original studies on mice showed that gastrointestinal microbiota is mainly composed by Enterobacteriaceae, Lactobacillus, Bacteroides and Clostridium[10,11]. More recent studies using next generation sequencing techniques (NGS) that allow a deeper analysis of the microbiota have reveled alterations in phylotypes previously overlooked using conventional techniques in patients with liver cirrhosis[12,13].

These experimental studies have brought the attention of clinicians to new therapeutic strategies targeting gut microbiota composition and function and aimed at preventing BT episodes and inflammation in cirrhosis. In fact, a recent publication by Kakiyama et al[14] provides interesting data in which among other reasons, dysbiosis is occurring in patients with cirrhosis in part due to low bile acid input into the gut. In this regard, prebiotics, probotics or symbiotics have been evaluated in different clinical studies in the last years[15].

This review intends to integrate the existing knowledge of the role of intestinal microbiota plays in the development of the most frequent complications arising in cirrhosis and the evidence on the possible efficacy of gut-microbiota modulating strategies to help in the management of these complications.

HE

HE is a serious and progressive neuropsychiatric condition that occurs in patients with advanced cirrhosis. Overt HE presents as a wide spectrum of clinical signs and symptoms, ranging in severity from mild confusion to life-threatening coma. Minimal HE is a more subtle form of the condition; it is characterized by deficits in cognitive function that can be detected with specialized tools. HE is considered to complicate up to 25% of presentations of acute liver failure[16], and is associated with poor survival[17,18].

Ammonia is a key factor in the pathogenesis of HE in cirrhotic patients[19]. The association between ammonia neurotoxicity and HE was first suggested by studies in dogs with porto-cava anastomosis[20]. Ammonia is generated in the intestines from dietary nitrogen, primarily from the breakdown of urea by urease present in colonic microbiota[21]. Under normal conditions, ammonia is metabolized to urea in the liver, but in the setting of liver failure, ammonia can bypass the liver into systemic circulation, inducing oxidative stress by generation of free radicals, and the nitrotyrosination of proteins in the brain[22]. This process is critical for mithochondrial function, and may cause failure of normal neurotransmission. Ammonia is taken up by astrocytes in the brain, and converted to glutamine where it exerts an osmotic effect and result in astrocyte swelling and brain edema[23-25].

Several studies show that gut microbiota is altered in cirrhotic patients with HE. More specifically, a quantitative change in the Bacteroides/Firmicutes ratio, with the prevalence of potentially pathogenic bacteria (e.g., Enterobacteriaceae)[13,26] and the reduction of commensals (Lachnospiraceae)[26] have been described. Liu et al[27] demonstrated a significant fecal overgrowth of potentially pathogenic Escherichia coli (E. coli) and Staphylococcus spp. in the gut microbiota of cirrhotic patients with minimal HE. Other studies have shown that patients with cirrhosis and HE had higher concentration of Enterobacteriaceae and Alcaligenaceae compared with control subjects and cirrhotic patients without HE[26]. This study shows, as well, that specific bacterial families (Alcaligenaceae, Porphyromonadaceae, Enterobacteriaceae) are strongly associated with cognition and inflammation in HE[26]. Bajaj et al[26,28,29] also studied the relationship between gut microbiota, inflammation and cognition in cirrhotic patients. Increased Alcaligenaceae abundance was significantly associated with poor cognitive performance, whereas Enterobacteriaceae were associated with worsening inflammation in the cirrhosis group. Alcaligenaceae are Proteobacteria that degrade urea to produce ammonia, which may explain his association with poor cognitive function[30]. Finally, in patients with HE, markers of Th17 and immune response were highly correlated with gut microbiome components, possibly indicating a synergy between inflammation and cognition with microbiome changes[31,32]. This evidence supports previous data revealing the components of the gut microbiota are required for the differentiation of Th17 cells in mouse models of autoimmune disease[33].

Treatment of HE is based in part on manipulation of the gut microbiota[34], with the aim to decrease the production and intestinal absorption of ammonia[35]. Lactulose (4-O-β-d-galactopyranosyl-d-fructose) is the most widely used treatment in HD; it lowers the colonic pH as a result of the production of organic acids by bacterial fermentation, inducing an environment that is both hostile to the survival of urease-producing gut bacteria, such as Klebsiella spp. and Proteus spp, facilitating the growth of acid resistant, non-urease-producing species, such as lactobacilli and bifidobacteria. This approach firstly reduces the ammonia production in the colonic lumen and secondly the acidification of colonic environment reduces the absorption of ammonia by nonionic diffusion[35].

Rifaximin is a non-absorbable antibiotic that is known that improve cognitive function in HE, especially its subclinical form, minimal HE, but the precise mechanism of its action is unclear[36]. A recent clinical trial by Bajaj et al[36] demonstrates that rifaximin is associated with reduction in endotoxemia in patients with cirrhosis and minimal HE, with a modest change in the stool microbiota composition, reducing Veillonellaceae and increasing Eubacteriaceae. After rifaximin therapy, there was also a significant change in the serum metabolome characterized by a specific increase in serum fatty acids. Although rifaximin is more effective than lactulose in the maintenance of remission and decreased re-admission in patients with minimal HE, it was not as cost-effective as lactulose[37].

The administration of probiotics that may modulate the gut microbiota composition or function in HE has had as primary objective to reduce the total amount of ammonia that reaches the portal system; this effect can be mediated through various mechanisms[38]. Several studies have shown the efficacy of some probiotics in the treatment of minimal HE and the prevention of overt HE episodes[39]. The first studies with probiotics in patients with cirrhosis and HE[40,41], compared the effects of administration of Enterococcus faecium SF68 vs lactulose on ammoniemia and clinical scores of HE. In these randomized studies, the probiotic SF68 showed similar results to lactulose during the treatment period. The therapeutic effect was maintained even during the washout period but only in the group treated with probiotics, suggesting that SF68 transiently colonize the colon and, consequently, this could favor the persistence of the effects.

In a prospective, randomized controlled trial, probiotics (including strains of the species Bifidobacterium breve, Bifidobacterium longum, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus paracasei, Lactobacillus bulgaricus, Streptococcus thermophilus, 110 billion CFU, available as VSL#3) were found to be effective in preventing HE in patients with cirrhosis[42]. However, in a double-blind placebo-controlled study that included patients with cirrhosis and at least one major complication of cirrhosis in the past, probiotic supplementation did not show beneficial effects on portal hypertension or decreased hepatic synthetic function, in patients with compensated cirrhosis[43]. Nevertheless, results pointed toward a possible positive effect of this probiotic in patients with above normal baseline ammonia levels.

An interesting study in rats with experimental cirrhosis demonstrated that a strain of the probiotic Lactobacillus plantarum is able to decrease amoniemia[44]. Considering the importance of ammonia in the pathogenesis of HE[45], this study suggests that treatment with this probiotic may be useful in patients with cirrhosis to treat or prevent this complication. Also, several studies[27,46-48] have yielded interesting results with synbiotics both in presence of overt HE, but also in minimal HE, as detected by improvement of all psychometric evaluations. However, the correlation between ammonia concentration and HE severity in patients with cirrhosis of liver is still poor[49]. Actually, it is common to find patients with symptoms of overt HE with just moderately elevated arterial ammonia levels[50]. This is probably related to the fact that HE is believed to be the consequence of different disturbances developed in advanced cirrhosis, and not only of hyperammoniemia[51,52].

SPLANCHNIC ARTERIAL VASODILATION

Splanchnic arterial vasodilation is the principal factor in the pathogenesis of the hyperdynamic circulatory syndrome that occurs in patients with cirrhosis and portal hypertension[53-55]. Increased synthesis and vascular release of the potent vasodilator nitric oxide (NO) is considered to be its main cause. Activation of the endothelial and inducible forms of nitric oxide synthase (eNOS and iNOS respectively) are considered to be the main sources of the elevated levels of NO in the splanchnic circulation. While activation of eNOS is due mostly to shear stress, exposure to bacteria and their products, directly or involving host cytokines, has been associated with activation of iNOS and increased release of NO[56] which in turn exacerbates further the circulatory disarrangement of cirrhosis[57,58]. TNF-α is also known to be involved in the pathogenesis of the hyperdynamic circulatory syndrome in portal hypertension[59,60].

Esophageal varices develop in response to the portal hypertension. Compared with other collaterals, gastroesophageal varices are important because of their risk of rupture and bleeding. Esophageal variceal bleeding is a serious event with 30% mortality per episode, and the likelihood of rebleeding reaches 30% to 40% in the first 6 wk without adequate therapy.

The management of variceal bleeding requires a complex approach[61] including vaso-pressors[62], use of prophylactic antibiotics[63,64], emergency endoscopic therapy[65,66], access to salvage therapy with trans-jugular intrahepatic portosystemic shunting (TIPS) in the most severe cases[67], as well as improvements in general supportive and critical care.

The use of prophylactic antibiotics (norfloxacin or ceftriaxone) in the setting of acute variceal bleeding is standard practice because it is known that decreases the rate of bacterial infection, risk of early rebleeding, and mortality[68,69]. However, antibiotic prophylaxis is associated with risk of infection with multi-resistant strains. The effect of the quinolone on the enteric bacterial load not only decreases the risk of bacterial infections but also increases systemic vascular resistance, improving haemodynamics[70].

Studies have suggested that chronic manipulation of the intestinal microbiota could have beneficial effects on the treatment of portal hypertension, but a few studies[37,43,71-73] have investigated the administration of probiotics to patients with variceal bleeding. In an experimental model of pre-hepatic portal hypertension[73], bacteriotherapy with a strain of Lactobacillus acidophilus or Lactobacillus GG. did not cause significant changes in BT between the animals treated with probiotic (82%) and those treated with placebo (75%), probably because of their ineffectivenes to modulate the microbiota composition.

In general no portal hypotensive effect has been observed when administering probiotics. In two different studies[74,75], the VSL#3 probiotic (a mix of lactic acid bacteria) used in patients with compensated or early decompensated cirrhosis and hepatic venous pressure gradient > 10 mmHg, resulted in no reduction in portal pressure. In one of them though[75], probiotic administration helped to decrease plasma endotoxin that suggests possible beneficial effects of probiotics for this patient population. In a recent study performed in cirrhotic patients having large esophageal varices without history of variceal bleeding[71], adjunctive probiotic VSL#3 improved the response rate to propranolol therapy and was safe and well tolerated in patients with cirrhosis. A double-blind placebo-controlled study that included patients with liver cirrhosis and at least one major complication of cirrhosis in the past (like variceal bleeding), and where the participants were randomly assigned to receive different probiotic capsules or placebo[43], did not show any significant clinical or laboratory effect of probiotic administration in patients with cirrhosis.

SBP

Bacterial infections are a frequent complication in patients with decompensated cirrhosis, with an incidence at the time of admission or during the hospitalization of about 32% in the most recent prospective studies[76]. Among them, SBP is the most common type of infection, induced in up to 70%-80% of cases by aerobic Gram-negative enteric organism, mainly E. coli and Klebsiella pneumoniae[77-79]. SBP is the infection of ascitic fluid in the absence of any primary intra-abdominal source, and it is considered to be present if the ascitic neutrophil count is in excess of 250 per microliter[69], with either positive or negative microbiological culture[79,80].

Broad-spectrum intravenous antibiotic therapy should be commenced immediately after diagnosed paracentesis in patients in whom SBP is clinically suspected or after the demonstration of a raised ascitic neutrophil count, without waiting for the result of the culture. SBP is usually treated with antibiotics using third generation cephalosporins. Cefotaxime or similar third-generation cephalosporin for a total course of 5 d is the treatment of choice[81-84]. Oral ofloxacin has been shown in one randomized controlled trial to be as effective as intravenous cefotaxime for a subgroup of patients with SBP and absence of risk factors[85]. Ciprofloxacin or levofloxacin can be used in patients who are intolerant to beta lactams. Patients surviving a SBP episode have a recurrence rate at 1 year of approximately 70%. In this setting indefinite selective intestinal decontamination with oral Norfloxacin reduces the risk of SBP recurrence from 50% to 20% at 2 years[86].

An increased frequency of infections due to quinolone-resistant (either Gram positive or negative) and multiresistant bacteria has recently been recognized though in patients being treated with norfloxacin, and this is associated with failure to control the infection with empiric antibiotic therapy and increased mortality[87]. New alternative strategies are needed for the empirical prophylaxis of nosocomial infections in cirrhosis. There have been several studies looking for alternative therapies to the use of Norfloxacin in high-risk patients with cirrhosis. In an experimental model in rats with CCl4-induced cirrhosis, long-term prophylactic trimethoprim-sulfamethoxazole administration delayed the development of ascites and reduced the incidence of Gram-negative bacteria translocation, without increasing Gram-positive episodes, suggesting that trimethoprim-sulfamethoxazole might be a good alternative to norfloxacin[88]. Another study showed that ciprofloxacin administered in a weekly dose was ineffective in selective intestinal decontamination, and was not useful in preventing SBP[89]. Rifaximin has been shown to decrease portal-hypertension related complications in a prospective randomized trial[90]. Taken together, available information suggests that blockade of episodes of BT by using oral antibiotics decreases the risk of development of different types of complications, and not only infections. Their use though, predisposes patients to develop infections by QR and MR bacteria. New therapeutic options are needed.

Proinflammatory cytokines are involved in the process of BT. Administration of anti TNF has been shown to significantly decrease BT in a rat model of cirrhosis[91]. This approach however has not been pursued further for the risk of inducing infections. Pentoxyfilline is also an anti TNF drug that has been shown to have a similar efficacy to norfloxacin in the prevention of BT and SBP in rats with CCl4-induced cirrhosis and ascites[92]. This is in line with an experimental study in CCl4-induced cirrhosis in rats showing that a prolonged oxidative stress in the intestine, accompanied by changes in gut microbiota composition, might facilitate translocation across the mucosa, resulting in complications such as SBP[93]. In fact, pentoxifylline administration, but not norfloxacin, decreases oxidative stress in cecal mucosa.

Probiotics appear therefore as attractive alternatives to antibiotics. They are often components of the commensal intestinal microbiota and have been shown to exert a number of beneficial effects mainly in animal models and to a lesser extent in humans. Results reported in the literature are still conflicting regarding efficacy. In a rat model of experimental CCl4-induced cirrhosis and ascites[94], Lactobacillus GG was unable to prevent both BT and infection of the ascitic fluid. In another study using Lactobacillus johnsonii (L. johnsonii) LA1 with antioxidants[95] in rats with CCl4-induced cirrhosis, there was a decrease in intestinal enterobacteria and in BT compared to untreated control rats, and there was a reduction in malonyldialdehyde (MDA) levels, an index of intestinal oxidative damage; however, in this study, the probiotic was not administered in the absence of antioxidants to be able to attribute a role to the probiotic per se. A recent work using the same animal model without antioxidants[96], indicate that L. johnsonii La1 alone has no significant effect on intestinal microbiota and BT in this experimental model of cirrhosis. The present data suggest that the beneficial effects observed in the previous study were mainly due to the antioxidant effect of vitamin C and glutamate.

A double-blind, randomized-controlled trial was conducted in cirrhotic patients who had either recovered from SBP (secondary prophylaxis) or who were at a high risk for the development of SBP[97] to assess the efficacy of administration of probiotics as secondary prophylaxis of infections. Norfloxacin (400 mg/d) with probiotic capsules (strains of Enterococcus faecalis, Clostridium butyricum, Bacillus mesentericus JPC, Bacillus coagulans) or norfloxacin with a placebo were given and the occurrence of SBP within a period of 6 mo, and side effects of therapy and mortality were recorded. Results were similar between groups, showing that the addition of probiotics to norfloxacin does not improve its efficacy in primary or secondary prophylaxis of SBP or in reducing the mortality in cirrhotic patients with ascites.

Stadlbauer et al[98] observed that treatment with Lactobacillus casei Shirota in cirrhotic patients of alcoholic etiology improved the phagocytic activity of neutrophils, decreased the levels of soluble TNF receptor 1 and 2 after stimulation with endotoxin and decreased overexpression of TLR4, indicating that the beneficial effect of the probiotic would result, at least in part, from the modification of the antigenic stimulus and IL10 secretion.

Recently, our group has evaluated the effects of Bifidobacterium pseudocatenulatum CECT7765 on BT and the liver status in a experimental model of cirrhosis in Balb/c mice by oral administration of CCl4, resulting in an improved gut barrier integrity and showing a beneficial effect against induced bacterial antigen translocation with E. coli[99].

CONCLUSION

As reviewed here, many studies have demonstrated the relevance of the interaction between the gut and the liver in the development of bacteria-related complications in cirrhosis. Changes in the microbiota composition have been shown to disturb intestinal immune homeostasis, which plays a central role in the proper management of the host-microbe crosstalk, not only favouring the translocation of pro-inflammatory bacterial products but also impairing the immunological host defence mechanisms against them and facilitating the production of soluble mediators that further complicate the liver disease.

While the use of antibiotics continues being an effective strategy for the management of these complications, other alternatives have emerged in the attempt to avoid the adverse impact of antibiotic therapy on the microbiota composition and function, given its relevance in keeping the intestinal homeostasis. These strategies include the use of pre-, pro- or synbiotic formulations, and experimental faecal transplantation aimed at restoring a “healthier” microbiota environment in patients with decompensated cirrhosis. Nevertheless, in contrast to abundant clinical evidence of efficacy and safety of the use of antibiotics in these patients, data available regarding the efficacy of these other strategies mainly comes from individual experimental studies[100]. The results of ongoing research in this area and the design of further studies on alternative microbiota-based interventions to prevent or to modulate bacteria-related complications in cirrhosis will shed light into the intimate interaction between the gut and the liver that lay down the development of these complications and into the design of more efficient strategies to re-shape the microbiota.

Footnotes

P- Reviewer: Ortiz LT S- Editor: Ma N L- Editor: A E- Editor: Ma S

References
1.  Madrid AM, Cumsille F, Defilippi C. Altered small bowel motility in patients with liver cirrhosis depends on severity of liver disease. Dig Dis Sci. 1997;42:738-742.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Garcia-Tsao G, Wiest R. Gut microflora in the pathogenesis of the complications of cirrhosis. Best Pract Res Clin Gastroenterol. 2004;18:353-372.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Berg RD, Garlington AW. Translocation of certain indigenous bacteria from the gastrointestinal tract to the mesenteric lymph nodes and other organs in a gnotobiotic mouse model. Infect Immun. 1979;23:403-411.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Wiest R, Garcia-Tsao G. Bacterial translocation (BT) in cirrhosis. Hepatology. 2005;41:422-433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 492]  [Cited by in F6Publishing: 484]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
5.  Guarner C, Runyon BA, Young S, Heck M, Sheikh MY. Intestinal bacterial overgrowth and bacterial translocation in cirrhotic rats with ascites. J Hepatol. 1997;26:1372-1378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 160]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
6.  Guarner C, Soriano G. Bacterial translocation and its consequences in patients with cirrhosis. Eur J Gastroenterol Hepatol. 2005;17:27-31.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Savage DC. Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol. 1977;31:107-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1659]  [Cited by in F6Publishing: 1428]  [Article Influence: 30.4]  [Reference Citation Analysis (0)]
8.  Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489:220-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3051]  [Cited by in F6Publishing: 3198]  [Article Influence: 266.5]  [Reference Citation Analysis (0)]
9.  Guarner F, Malagelada JR. Gut flora in health and disease. Lancet. 2003;361:512-519.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Dubos R, Schaedler RW, Costello R, Hoet P. Indigenous, normal, and autochthonous flora of the gastrointestinal tract. J Exp Med. 1965;122:67-76.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Schaedler RW, Dubos R, Costello R. The development of the bacterial flora in the gastrointestinal tract of mice. J Exp Med. 1965;122:59-66.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Fouts DE, Torralba M, Nelson KE, Brenner DA, Schnabl B. Bacterial translocation and changes in the intestinal microbiome in mouse models of liver disease. J Hepatol. 2012;56:1283-1292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 232]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
13.  Chen Y, Yang F, Lu H, Wang B, Chen Y, Lei D, Wang Y, Zhu B, Li L. Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology. 2011;54:562-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 674]  [Cited by in F6Publishing: 701]  [Article Influence: 53.9]  [Reference Citation Analysis (1)]
14.  Kakiyama G, Pandak WM, Gillevet PM, Hylemon PB, Heuman DM, Daita K, Takei H, Muto A, Nittono H, Ridlon JM. Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. J Hepatol. 2013;58:949-955.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 503]  [Cited by in F6Publishing: 549]  [Article Influence: 49.9]  [Reference Citation Analysis (0)]
15.  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)]
16.  Bernal W, Hall C, Karvellas CJ, Auzinger G, Sizer E, Wendon J. Arterial ammonia and clinical risk factors for encephalopathy and intracranial hypertension in acute liver failure. Hepatology. 2007;46:1844-1852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 271]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
17.  Bajaj JS. Review article: the modern management of hepatic encephalopathy. Aliment Pharmacol Ther. 2010;31:537-547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 161]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
18.  Córdoba J. New assessment of hepatic encephalopathy. J Hepatol. 2011;54:1030-1040.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 160]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
19.  Jalan R, Shawcross D, Davies N. The molecular pathogenesis of hepatic encephalopathy. Int J Biochem Cell Biol. 2003;35:1175-1181.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Shawcross DL, Olde Damink SW, Butterworth RF, Jalan R. Ammonia and hepatic encephalopathy: the more things change, the more they remain the same. Metab Brain Dis. 2005;20:169-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 41]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
21.  Huizenga JR, Gips CH, Tangerman A. The contribution of various organs to ammonia formation: a review of factors determining the arterial ammonia concentration. Ann Clin Biochem. 1996;33:23-30.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Norenberg MD. Oxidative and nitrosative stress in ammonia neurotoxicity. Hepatology. 2003;37:245-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 101]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
23.  Shawcross DL, Wright G, Olde Damink SW, Jalan R. Role of ammonia and inflammation in minimal hepatic encephalopathy. Metab Brain Dis. 2007;22:125-138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 190]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
24.  Wong F, Bernardi M, Balk R, Christman B, Moreau R, Garcia-Tsao G, Patch D, Soriano G, Hoefs J, Navasa M. Sepsis in cirrhosis: report on the 7th meeting of the International Ascites Club. Gut. 2005;54:718-725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 264]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
25.  Butterworth RF. Pathophysiology of hepatic encephalopathy: a new look at ammonia. Metab Brain Dis. 2002;17:221-227.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Bajaj JS, Ridlon JM, Hylemon PB, Thacker LR, Heuman DM, Smith S, Sikaroodi M, Gillevet PM. Linkage of gut microbiome with cognition in hepatic encephalopathy. Am J Physiol Gastrointest Liver Physiol. 2012;302:G168-G175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 347]  [Cited by in F6Publishing: 373]  [Article Influence: 31.1]  [Reference Citation Analysis (0)]
27.  Liu Q, Duan ZP, Ha DK, Bengmark S, Kurtovic J, Riordan SM. Synbiotic modulation of gut flora: effect on minimal hepatic encephalopathy in patients with cirrhosis. Hepatology. 2004;39:1441-1449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 444]  [Cited by in F6Publishing: 426]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
28.  Bajaj JS, Gillevet PM, Patel NR, Ahluwalia V, Ridlon JM, Kettenmann B, Schubert CM, Sikaroodi M, Heuman DM, Crossey MM. A longitudinal systems biology analysis of lactulose withdrawal in hepatic encephalopathy. Metab Brain Dis. 2012;27:205-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 61]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
29.  Bajaj JS, Hylemon PB, Ridlon JM, Heuman DM, Daita K, White MB, Monteith P, Noble NA, Sikaroodi M, Gillevet PM. Colonic mucosal microbiome differs from stool microbiome in cirrhosis and hepatic encephalopathy and is linked to cognition and inflammation. Am J Physiol Gastrointest Liver Physiol. 2012;303:G675-G685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 354]  [Cited by in F6Publishing: 375]  [Article Influence: 31.3]  [Reference Citation Analysis (0)]
30.  Obata T, Goto Y, Kunisawa J, Sato S, Sakamoto M, Setoyama H, Matsuki T, Nonaka K, Shibata N, Gohda M. Indigenous opportunistic bacteria inhabit mammalian gut-associated lymphoid tissues and share a mucosal antibody-mediated symbiosis. Proc Natl Acad Sci USA. 2010;107:7419-7424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 178]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
31.  Keshavarzian A, Holmes EW, Patel M, Iber F, Fields JZ, Pethkar S. Leaky gut in alcoholic cirrhosis: a possible mechanism for alcohol-induced liver damage. Am J Gastroenterol. 1999;94:200-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 287]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
32.  Mutlu E, Keshavarzian A, Engen P, Forsyth CB, Sikaroodi M, Gillevet P. Intestinal dysbiosis: a possible mechanism of alcohol-induced endotoxemia and alcoholic steatohepatitis in rats. Alcohol Clin Exp Res. 2009;33:1836-1846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 260]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
33.  Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA. 2011;108 Suppl 1:4615-4622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 881]  [Cited by in F6Publishing: 946]  [Article Influence: 67.6]  [Reference Citation Analysis (0)]
34.  Prasad S, Dhiman RK, Duseja A, Chawla YK, Sharma A, Agarwal R. Lactulose improves cognitive functions and health-related quality of life in patients with cirrhosis who have minimal hepatic encephalopathy. Hepatology. 2007;45:549-559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 412]  [Cited by in F6Publishing: 369]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
35.  Riordan SM, Williams R. Treatment of hepatic encephalopathy. N Engl J Med. 1997;337:473-479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 305]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
36.  Bajaj JS, Heuman DM, Sanyal AJ, Hylemon PB, Sterling RK, Stravitz RT, Fuchs M, Ridlon JM, Daita K, Monteith P. Modulation of the metabiome by rifaximin in patients with cirrhosis and minimal hepatic encephalopathy. PLoS One. 2013;8:e60042.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 302]  [Article Influence: 27.5]  [Reference Citation Analysis (0)]
37.  Rahimi RS, Rockey DC. End-stage liver disease complications. Curr Opin Gastroenterol. 2013;29:257-263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
38.  Solga SF. Probiotics can treat hepatic encephalopathy. Med Hypotheses. 2003;61:307-313.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Dhiman RK. Gut microbiota and hepatic encephalopathy. Metab Brain Dis. 2013;28:321-326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
40.  Loguercio C, Del Vecchio Blanco C, Coltorti M. Enterococcus lactic acid bacteria strain SF68 and lactulose in hepatic encephalopathy: a controlled study. J Int Med Res. 1987;15:335-343.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Loguercio C, Abbiati R, Rinaldi M, Romano A, Del Vecchio Blanco C, Coltorti M. Long-term effects of Enterococcus faecium SF68 versus lactulose in the treatment of patients with cirrhosis and grade 1-2 hepatic encephalopathy. J Hepatol. 1995;23:39-46.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Lunia MK, Sharma BC, Sharma P, Sachdeva S, Srivastava S. Probiotics prevent hepatic encephalopathy in patients with cirrhosis: a randomized controlled trial. Clin Gastroenterol Hepatol. 2014;12:1003-8.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 125]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
43.  Pereg D, Kotliroff A, Gadoth N, Hadary R, Lishner M, Kitay-Cohen Y. Probiotics for patients with compensated liver cirrhosis: a double-blind placebo-controlled study. Nutrition. 2011;27:177-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
44.  Nicaise C, Prozzi D, Viaene E, Moreno C, Gustot T, Quertinmont E, Demetter P, Suain V, Goffin P, Devière J. Control of acute, chronic, and constitutive hyperammonemia by wild-type and genetically engineered Lactobacillus plantarum in rodents. Hepatology. 2008;48:1184-1192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
45.  Córdoba J, Mínguez B. Hepatic encephalopathy. Semin Liver Dis. 2008;28:70-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 97]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
46.  Malaguarnera M, Greco F, Barone G, Gargante MP, Malaguarnera M, Toscano MA. Bifidobacterium longum with fructo-oligosaccharide (FOS) treatment in minimal hepatic encephalopathy: a randomized, double-blind, placebo-controlled study. Dig Dis Sci. 2007;52:3259-3265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 127]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
47.  Bajaj JS, Saeian K, Christensen KM, Hafeezullah M, Varma RR, Franco J, Pleuss JA, Krakower G, Hoffmann RG, Binion DG. Probiotic yogurt for the treatment of minimal hepatic encephalopathy. Am J Gastroenterol. 2008;103:1707-1715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 184]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
48.  Dhiman RK, Chawla YK. Minimal hepatic encephalopathy: time to recognise and treat. Trop Gastroenterol. 2008;29:6-12.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Shawcross DL, Sharifi Y, Canavan JB, Yeoman AD, Abeles RD, Taylor NJ, Auzinger G, Bernal W, Wendon JA. Infection and systemic inflammation, not ammonia, are associated with Grade 3/4 hepatic encephalopathy, but not mortality in cirrhosis. J Hepatol. 2011;54:640-649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 179]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
50.  Arora S, Martin CL, Herbert M. Myth: interpretation of a single ammonia level in patients with chronic liver disease can confirm or rule out hepatic encephalopathy. CJEM. 2006;8:433-435.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Butterworth RF. Hepatic encephalopathy: a central neuroinflammatory disorder? Hepatology. 2011;53:1372-1376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 164]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
52.  Tranah TH, Vijay GK, Ryan JM, Shawcross DL. Systemic inflammation and ammonia in hepatic encephalopathy. Metab Brain Dis. 2013;28:1-5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 83]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
53.  Colombato LA, Albillos A, Groszmann RJ. Temporal relationship of peripheral vasodilatation, plasma volume expansion and the hyperdynamic circulatory state in portal-hypertensive rats. Hepatology. 1992;15:323-328.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Schrier RW, Arroyo V, Bernardi M, Epstein M, Henriksen JH, Rodés J. Peripheral arterial vasodilation hypothesis: a proposal for the initiation of renal sodium and water retention in cirrhosis. Hepatology. 1988;8:1151-1157.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Groszmann RJ. Hyperdynamic circulation of liver disease 40 years later: pathophysiology and clinical consequences. Hepatology. 1994;20:1359-1363.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Francés R, Muñoz C, Zapater P, Uceda F, Gascón I, Pascual S, Pérez-Mateo M, Such J. Bacterial DNA activates cell mediated immune response and nitric oxide overproduction in peritoneal macrophages from patients with cirrhosis and ascites. Gut. 2004;53:860-864.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Bellot P, García-Pagán JC, Francés R, Abraldes JG, Navasa M, Pérez-Mateo M, Such J, Bosch J. Bacterial DNA translocation is associated with systemic circulatory abnormalities and intrahepatic endothelial dysfunction in patients with cirrhosis. Hepatology. 2010;52:2044-2052.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 153]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
58.  Meyer J, Traber LD, Nelson S, Lentz CW, Nakazawa H, Herndon DN, Noda H, Traber DL. Reversal of hyperdynamic response to continuous endotoxin administration by inhibition of NO synthesis. J Appl Physiol (1985). 1992;73:324-328.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Lopez-Talavera JC, Cadelina G, Olchowski J, Merrill W, Groszmann RJ. Thalidomide inhibits tumor necrosis factor alpha, decreases nitric oxide synthesis, and ameliorates the hyperdynamic circulatory syndrome in portal-hypertensive rats. Hepatology. 1996;23:1616-1621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 29]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
60.  Lopez-Talavera JC, Merrill WW, Groszmann RJ. Tumor necrosis factor alpha: a major contributor to the hyperdynamic circulation in prehepatic portal-hypertensive rats. Gastroenterology. 1995;108:761-767.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Jairath V, Rehal S, Logan R, Kahan B, Hearnshaw S, Stanworth S, Travis S, Murphy M, Palmer K, Burroughs A. Acute variceal haemorrhage in the United Kingdom: patient characteristics, management and outcomes in a nationwide audit. Dig Liver Dis. 2014;46:419-426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 59]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
62.  Ioannou G, Doust J, Rockey DC. Terlipressin for acute esophageal variceal hemorrhage. Cochrane Database Syst Rev. 2001;CD002147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 75]  [Article Influence: 3.3]  [Reference Citation Analysis (1)]
63.  Bernard B, Grangé JD, Khac EN, Amiot X, Opolon P, Poynard T. Antibiotic prophylaxis for the prevention of bacterial infections in cirrhotic patients with gastrointestinal bleeding: a meta-analysis. Hepatology. 1999;29:1655-1661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 594]  [Cited by in F6Publishing: 606]  [Article Influence: 24.2]  [Reference Citation Analysis (0)]
64.  Chavez-Tapia NC, Barrientos-Gutierrez T, Tellez-Avila FI, Soares-Weiser K, Uribe M. Antibiotic prophylaxis for cirrhotic patients with upper gastrointestinal bleeding. Cochrane Database Syst Rev. 2010;CD002907.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 63]  [Article Influence: 4.5]  [Reference Citation Analysis (1)]
65.  Sclerotherapy after first variceal hemorrhage in cirrhosis. A randomized multicenter trial. The Copenhagen Esophageal Varices Sclerotherapy Project. N Engl J Med. 1984;311:1594-1600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 259]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
66.  Triantos CK, Goulis J, Patch D, Papatheodoridis GV, Leandro G, Samonakis D, Cholongitas E, Burroughs AK. An evaluation of emergency sclerotherapy of varices in randomized trials: looking the needle in the eye. Endoscopy. 2006;38:797-807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 39]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
67.  Boyer TD, Haskal ZJ. The role of transjugular intrahepatic portosystemic shunt in the management of portal hypertension. Hepatology. 2005;41:386-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 315]  [Cited by in F6Publishing: 286]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
68.  Fernández J, Ruiz del Arbol L, Gómez C, Durandez R, Serradilla R, Guarner C, Planas R, Arroyo V, Navasa M. Norfloxacin vs ceftriaxone in the prophylaxis of infections in patients with advanced cirrhosis and hemorrhage. Gastroenterology. 2006;131:1049-156; quiz 1285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 308]  [Cited by in F6Publishing: 227]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
69.  Rimola A, García-Tsao G, Navasa M, Piddock LJ, Planas R, Bernard B, Inadomi JM. Diagnosis, treatment and prophylaxis of spontaneous bacterial peritonitis: a consensus document. International Ascites Club. J Hepatol. 2000;32:142-153.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Albillos A, de la Hera A, González M, Moya JL, Calleja JL, Monserrat J, Ruiz-del-Arbol L, Alvarez-Mon M. Increased lipopolysaccharide binding protein in cirrhotic patients with marked immune and hemodynamic derangement. Hepatology. 2003;37:208-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 320]  [Cited by in F6Publishing: 320]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
71.  Gupta N, Kumar A, Sharma P, Garg V, Sharma BC, Sarin SK. Effects of the adjunctive probiotic VSL#3 on portal haemodynamics in patients with cirrhosis and large varices: a randomized trial. Liver Int. 2013;33:1148-1157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 64]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
72.  Rahimi RS, Rockey DC. Complications of cirrhosis. Curr Opin Gastroenterol. 2012;28:223-229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 33]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
73.  Wiest R, Chen F, Cadelina G, Groszmann RJ, Garcia-Tsao G. Effect of Lactobacillus-fermented diets on bacterial translocation and intestinal flora in experimental prehepatic portal hypertension. Dig Dis Sci. 2003;48:1136-1141.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Jayakumar S, Carbonneau M, Hotte N, Befus AD, St Laurent C, Owen R, McCarthy M, Madsen K, Bailey RJ, Ma M. VSL#3 ® probiotic therapy does not reduce portal pressures in patients with decompensated cirrhosis. Liver Int. 2013;33:1470-1477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
75.  Tandon P, Moncrief K, Madsen K, Arrieta MC, Owen RJ, Bain VG, Wong WW, Ma MM. Effects of probiotic therapy on portal pressure in patients with cirrhosis: a pilot study. Liver Int. 2009;29:1110-1115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 50]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
76.  Fernández J, Navasa M, Gómez J, Colmenero J, Vila J, Arroyo V, Rodés J. Bacterial infections in cirrhosis: epidemiological changes with invasive procedures and norfloxacin prophylaxis. Hepatology. 2002;35:140-148.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Caly WR, Strauss E. A prospective study of bacterial infections in patients with cirrhosis. J Hepatol. 1993;18:353-358.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Yoshida H, Hamada T, Inuzuka S, Ueno T, Sata M, Tanikawa K. Bacterial infection in cirrhosis, with and without hepatocellular carcinoma. Am J Gastroenterol. 1993;88:2067-2071.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Such J, Runyon BA. Spontaneous bacterial peritonitis. Clin Infect Dis. 1998;27:669-74; quiz 675-6.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Guarner C, Soriano G. Spontaneous bacterial peritonitis. Semin Liver Dis. 1997;17:203-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 118]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
81.  Runyon BA, Akriviadis EA, Sattler FR, Cohen J. Ascitic fluid and serum cefotaxime and desacetyl cefotaxime levels in patients treated for bacterial peritonitis. Dig Dis Sci. 1991;36:1782-1786.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Runyon BA, McHutchison JG, Antillon MR, Akriviadis EA, Montano AA. Short-course versus long-course antibiotic treatment of spontaneous bacterial peritonitis. A randomized controlled study of 100 patients. Gastroenterology. 1991;100:1737-1742.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Rimola A, Salmerón JM, Clemente G, Rodrigo L, Obrador A, Miranda ML, Guarner C, Planas R, Solá R, Vargas V. Two different dosages of cefotaxime in the treatment of spontaneous bacterial peritonitis in cirrhosis: results of a prospective, randomized, multicenter study. Hepatology. 1995;21:674-679.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Ricart E, Soriano G, Novella MT, Ortiz J, Sàbat M, Kolle L, Sola-Vera J, Miñana J, Dedéu JM, Gómez C. Amoxicillin-clavulanic acid versus cefotaxime in the therapy of bacterial infections in cirrhotic patients. J Hepatol. 2000;32:596-602.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Navasa M, Follo A, Llovet JM, Clemente G, Vargas V, Rimola A, Marco F, Guarner C, Forné M, Planas R. Randomized, comparative study of oral ofloxacin versus intravenous cefotaxime in spontaneous bacterial peritonitis. Gastroenterology. 1996;111:1011-1017.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Ginés P, Rimola A, Planas R, Vargas V, Marco F, Almela M, Forné M, Miranda ML, Llach J, Salmerón JM. Norfloxacin prevents spontaneous bacterial peritonitis recurrence in cirrhosis: results of a double-blind, placebo-controlled trial. Hepatology. 1990;12:716-724.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Fernández J, Acevedo J, Castro M, Garcia O, de Lope CR, Roca D, Pavesi M, Sola E, Moreira L, Silva A. Prevalence and risk factors of infections by multiresistant bacteria in cirrhosis: a prospective study. Hepatology. 2012;55:1551-1561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 403]  [Cited by in F6Publishing: 392]  [Article Influence: 32.7]  [Reference Citation Analysis (0)]
88.  Guarner C, Runyon BA, Heck M, Young S, Sheikh MY. Effect of long-term trimethoprim-sulfamethoxazole prophylaxis on ascites formation, bacterial translocation, spontaneous bacterial peritonitis, and survival in cirrhotic rats. Dig Dis Sci. 1999;44:1957-1962.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Terg R, Llano K, Cobas SM, Brotto C, Barrios A, Levi D, Wasen W, Bartellini MA. Effects of oral ciprofloxacin on aerobic gram-negative fecal flora in patients with cirrhosis: results of short- and long-term administration, with daily and weekly dosages. J Hepatol. 1998;29:437-442.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Vlachogiannakos J, Viazis N, Vasianopoulou P, Vafiadis I, Karamanolis DG, Ladas SD. Long-term administration of rifaximin improves the prognosis of patients with decompensated alcoholic cirrhosis. J Gastroenterol Hepatol. 2013;28:450-455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 159]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
91.  Francés R, Chiva M, Sánchez E, González-Navajas JM, Llovet T, Zapater P, Soriano G, Muñoz C, Balanzó J, Pérez-Mateo M. Bacterial translocation is downregulated by anti-TNF-alpha monoclonal antibody administration in rats with cirrhosis and ascites. J Hepatol. 2007;46:797-803.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Corradi F, Brusasco C, Fernández J, Vila J, Ramirez MJ, Seva-Pereira T, Fernández-Varo G, Mosbah IB, Acevedo J, Silva A. Effects of pentoxifylline on intestinal bacterial overgrowth, bacterial translocation and spontaneous bacterial peritonitis in cirrhotic rats with ascites. Dig Liver Dis. 2012;44:239-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
93.  Natarajan SK, Ramamoorthy P, Thomas S, Basivireddy J, Kang G, Ramachandran A, Pulimood AB, Balasubramanian KA. Intestinal mucosal alterations in rats with carbon tetrachloride-induced cirrhosis: changes in glycosylation and luminal bacteria. Hepatology. 2006;43:837-846.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Bauer TM, Fernández J, Navasa M, Vila J, Rodés J. Failure of Lactobacillus spp. to prevent bacterial translocation in a rat model of experimental cirrhosis. J Hepatol. 2002;36:501-506.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Chiva M, Soriano G, Rochat I, Peralta C, Rochat F, Llovet T, Mirelis B, Schiffrin EJ, Guarner C, Balanzó J. Effect of Lactobacillus johnsonii La1 and antioxidants on intestinal flora and bacterial translocation in rats with experimental cirrhosis. J Hepatol. 2002;37:456-462.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Soriano G, Sánchez E, Guarner C, Schiffrin EJ. Lactobacillus johnsonii La1 without antioxidants does not decrease bacterial translocation in rats with carbon tetrachloride-induced cirrhosis. J Hepatol. 2012;57:1395-1396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
97.  Pande C, Kumar A, Sarin SK. Addition of probiotics to norfloxacin does not improve efficacy in the prevention of spontaneous bacterial peritonitis: a double-blind placebo-controlled randomized-controlled trial. Eur J Gastroenterol Hepatol. 2012;24:831-839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
98.  Stadlbauer V, Mookerjee RP, Hodges S, Wright GA, Davies NA, Jalan R. Effect of probiotic treatment on deranged neutrophil function and cytokine responses in patients with compensated alcoholic cirrhosis. J Hepatol. 2008;48:945-951.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 213]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
99.  Moratalla A, Gómez-Hurtado I, Santacruz A, Moya Á, Peiró G, Zapater P, González-Navajas JM, Giménez P, Such J, Sanz Y. Protective effect of Bifidobacterium pseudocatenulatum CECT7765 against induced bacterial antigen translocation in experimental cirrhosis. Liver Int. 2014;34:850-858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
100.  Quigley EM, Monsour HP. The gut microbiota and the liver: implications for clinical practice. Expert Rev Gastroenterol Hepatol. 2013;7:723-732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]