Topic Highlight Open Access
Copyright ©2006 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Jun 14, 2006; 12(22): 3487-3495
Published online Jun 14, 2006. doi: 10.3748/wjg.v12.i22.3487
Ursodeoxycholic acid treatment of vanishing bile duct syndromes
Thomas Pusl, Ulrich Beuers, Department of Medicine II, Klinikum Grosshadern, University of Munich, Germany
Author contributions: All authors contributed equally to the work.
Correspondence to: Ulrich Beuers, MD, Department of Medicine II, Klinikum Grosshadern, Marchioninistrasse 15, 81377 Munich, Germany. beuers@med.uni-muenchen.de
Telephone: +49-89-70955272 Fax: +49-89-70955271
Received: January 31, 2006
Revised: March 5, 2006
Accepted: March 14, 2006
Published online: June 14, 2006

Abstract

Vanishing bile duct syndromes (VBDS) are characterized by progressive loss of small intrahepatic ducts caused by a variety of different diseases leading to chronic cholestasis, cirrhosis, and premature death from liver failure. The majority of adult patients with VBDS suffer from primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC). Ursodeoxycholic acid (UDCA), a hydrophilic dihydroxy bile acid, is the only drug currently approved for the treatment of patients with PBC, and anticholestatic effects have been reported for several other cholestatic syndromes. Several potential mechanisms of action of UDCA have been proposed including stimulation of hepatobiliary secretion, inhibition of apoptosis and protection of cholangiocytes against toxic effects of hydrophobic bile acids.

Key Words: Cholestasis, Primary biliary cirrhosis, Primary sclerosing cholangitis, Secretion, Signaling, Transport, Ursodeoxycholic acid, Vanishing bile duct syndrome



INTRODUCTION

Cholestasis is defined as an impairment of bile flow and failure to adequately secrete cholephilic compounds into bile often associated with clinical manifestations such as jaundice and pruritus and biochemical alterations such as an elevation in serum alkaline phosphatase and γ-glutamyl transpeptidase. The liver damage observed in chronic cholestasis has long been attributed to accumulation and retention of potentially toxic bile acids within hepatocytes. Chronic cholestasis is the main feature of vanishing bile duct syndromes (VBDS) characterized by progressive loss of small intrahepatic ducts caused by a variety of different diseases. In children, biliary atresia and Alagille syndrome are rare ductopenic diseases classified as VBDS. The majority of adult patients with VBDS suffer from primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC). Other diseases associated with ductopenia include autoimmune cholangitis, chronic hepatic allograft rejection, graft-versus-host disease (GVHD), chronic cholestatic sarcoidosis and ischemic (vascular) cholangiopathies. Among the various causes of VBDS, drugs have an increasing importance. Drug reactions are related especially to antibiotics, phenothiazine derivates and carbamazepine. It is increasingly clear that immunopathogenetic mechanisms involving innate and adaptive immune responses contribute to ductopenia in most of these diseases. Prognosis varies from hepatic failure and death or liver transplantation to resolution of cholestasis and normal liver function.

Ursodeoxycholic acid (UDCA) is a hydrophilic dihydroxy bile acid (chemical structure: 3α, 7β-dihydroxy-5β-cholanoic acid) which was first identified in the bile of the Chinese black bear[1]. UDCA is a physiologic bile acid also present in man albeit in a low concentration of about 3% of the bile acid pool, where it is formed by 7β-epimerization of the primary bile acid chenodeoxycholic acid in the gut by intestinal bacteria[2,3]. It has been used for centuries in traditional Chinese medicine for the treatment of liver diseases. Reports from Japan and Europe first revealed that UDCA was able to dissolve gallstones[4-6]. In 1985, Leuschner and coworkers observed improved serum liver tests in patients with chronic active hepatitis treated with UDCA for gallstone dissolution[7], and similar observations were made previously in Japan. Since then, various trials have shown the beneficial effect of UDCA for different cholestatic syndromes.

PHARMACOKINETICS AND METABOLISM OF UDCA

After oral administration, UDCA is absorbed by passive nonionic diffusion mainly in the small intestine (-80%) and less in the colon (-20%) following solubilization in mixed micelles of endogenous bile acids in the proximal jejunum[2,8]. Its absorption rate is enhanced when it is given with a meal and may be decreased in patients with cholestasis and decreased biliary secretion of endogenous bile acids[9]. After intestinal absorption, UDCA is taken up from the portal blood by the hepatocytes at their sinusoidal domain via specific bile acid transporters namely, NTCP and OATP, conjugated mainly with glycine and to a lesser extent with taurine and is subsequently transported across the canalicular domain into the bile ducts via another bile acid carrier molecule, designated BSEP[2,10]. UDCA conjugates reach the small intestine and are reabsorbed mainly from the distal ileum via an active Na+-dependent transport mechanism undergoing an effective enterohepatic circulation. Non-absorbed UDCA and UDCA conjugates pass into the colon and, after bacterial deamidation of conjugates, are mostly converted to lithocholic acid by intestinal bacteria and eliminated via the feces[2]. Only minute amounts of the insoluble lithocholic acid are reabsorbed via the colonic mucosa, sulphated in the liver, secreted into bile and excreted in the feces. Under continuous oral treatment at pharmacological doses (13-15 mg/kg per day) UDCA becomes the predominant bile acid in the liver and the systemic circulation, comprising 40% to 60% of total bile acid[2,8].

MECHANISMS OF ACTION OF URSO-DEOXYCHOLIC ACID

The mechanisms underlying the beneficial effects of UDCA in cholestasis are being increasingly unraveled[11-14]. Initial research interest was focused on changes in bile acid pool composition, hepatocyte membrane protective effects, immunomodulatory effects, and bicarbonate-rich hypercholeresis induced by UDCA. Over the past decade, it has, however, become evident that UDCA is a potent intracellular signaling agent that induces stimulation of impaired hepatocellular secretion, anti-apoptotic effects and may mediate cholangiocyte protection. Depending on the pathophysiology and the stage of the underlying liver disease, the predominant mechanisms of action of UDCA may vary.

Stimulation of hepatobiliary secretion

Cholestatic liver diseases are characterized by an impairment of hepatobiliary secretion. As a consequence, bile acids and other potentially toxic cholephiles accumulate in the hepatocyte and may lead to liver cell injury, apoptosis and necrosis. UDCA stimulates biliary secretion of bile acids and other organic compounds (e.g. bilirubin glucuronides, glutathione conjugates, bromosulfophthalein) in various experimental models such as the isolated hepatocytes, isolated perfused rat liver and bile fistula rat model and counteracts cholestasis induced by hydrophobic bile acids in rat liver[15-22]. In line with these observations, biliary secretion of bile acids and phospholipids is stimulated and elevated serum levels of the hydrophobic bile acid, chenodeoxycholic acid, and of bilirubin are decreased in patients with PBC and PSC during UDCA treatment[23-28]. Thus, the beneficial effects of UDCA in cholestatic liver disease may be partly due to the enhanced elimination of toxic compounds from the hepatocytes. The secretory capacity of the hepatocytes is determined by the number and activity of transporter proteins in the canalicular membrane. UDCA stimulates the expression of transporter proteins for biliary secretion in the liver and the targeting and insertion of transporter molecules into the canalicular membrane at a transcriptional and posttranscriptional level[15,29-31]. As such, UDCA stimulates the overall gene expression of both canalicular (Mrp2, Bsep) and alternative basolateral carriers (Mrp3, Mrp4) in mouse liver, which facilitates alternative efflux of bile salts and other organic anions into the systemic circulation[32,33]. UDCA also stimulates murine renal (Mrp2, Mrp4) and intestinal (Mrp2, Mrp3) efflux transport proteins, resulting in an increased overall elimination capacity for potentially toxic biliary compounds[32]. In addition to these transcriptional effects, UDCA also stimulates vesicular exocytosis and insertion of transporter proteins into the canalicular membrane by modulating complex intracellular signalling cascades including calcium, protein kinase C and different mitogen-activated protein kinases[15,31,34-36]. Moreover, UDCA may also directly activate canalicular transporters through modification of their phosphorylation status[37]. While effects of UDCA on mRNA and protein levels of transporters may be important for long-term regulation, the effects on the insertion into the canalicular membrane and the activity of transporters may determine short-term regulation of secretion. In conclusion, UDCA modulates hepatobiliary secretion by transcriptional and posttranscriptional mechanisms in experimental models in vivo and in vitro. Thus, upregulation of synthesis, apical targeting and insertion, and activation of key canalicular transporters may represent key mechanisms to explain the anticholestatic action of UDCA in patients with cholestatic liver disease.

Inhibition of apoptosis

Apoptosis, or programmed cell death, is an important mechanism of cell death in cholestatic liver diseases[14,38,39]. For example, apoptotic features causing bile duct loss have been observed more frequently in liver tissue from patients with PBC than in normal controls[40]. Toxic bile acids can induce apoptosis in hepatocytes at concentrations comparable to those found in chronic cholestasis and the mechanisms of bile acid-induced apoptosis have increasingly been understood (reviewed by Higuchi et al[41]). Endogenous hydrophobic bile acids such as glycochenodeoxycholic acid (GCDCA) or glycodeoxycholic acid (GDCA) induce apoptosis by ligand-independent activation of the Fas death-receptor, followed by activation of caspase 8 and Bid, a pro-apoptotic member of the Bcl-2 protein family, which chaperones another pro-apototic Bcl-2 molecule, Bax, to the mitochondrial membrane inducing mitochondrial membrane permeability transition (MMPT). MMPT causes a sudden increase in permeability of the inner mitochondrial membrane to ions followed by mitochondrial swelling and release of cytochrome c from the intermembrane space to the cytosol. In the cytosol, cytochrome c interacts with the apoptotic protease-activating factor 1 (APAF-1), which leads to activation of caspase 9 and subsequently causes apoptotic cell death[40,42].

A number of studies showed that UDCA can block apoptosis in vitro as well as in vivo in the rat and in human hepatocytes by interrupting classic pathways of apoptosis[43-45]. Antiapoptotic effects of UDCA were associated with a reduction of the MMPT and mitochondrial cytochrome c release[14,45]. UDCA protects rat hepatocytes against bile acid-induced apoptosis by preventing bile acid-induced, c-Jun N-terminal kinase-dependent Fas trafficking to the plasma membrane[46]. Apoptosis can not only be inhibited by blocking pro-apoptotic pathways, but also by activation of intracellular survival signals. Indeed, UDCA was shown to induce a survival signal in hepatocytes via activation of the epidermal growth factor receptor (EGFR) and mitogen-activated protein kinase (MAPK) that may contribute to the antiapoptotic effect[47]. Thus, UDCA appears to be a bile acid that partially activates pro-apoptotic signals, but also simultaneously a strong MAPK-dependent survival signal, which makes UDCA an overall non-toxic bile acid. Although intriguing, the impact of antiapoptotic mechanisms for the beneficial effects of UDCA in cholestatic liver disease remains unclear at present.

Protection of cholangiocytes against toxic effects of hydrophobic bile acids

Hydrophobic bile acids damage cell membranes of hepatocytes and cholangiocytes and exert extracellular cytotoxicity at millimolar concentrations present in bile[48-50]. Indeed, levels of hydrophobic bile acids are about 1000-fold higher in bile than in serum even under physiological conditions. The proposed mechanisms of bile acid-induced cell damage extend from simply binding to plasma membranes to the induction of apoptosis or even necrosis. UDCA has been shown to counteract hydrophobic bile acid-induced membrane disruption in vitro presumably by alteration of the structure and composition of simple and mixed micelles rather than by direct membrane interactions[50,51]. UDCA treatment at therapeutic doses of 13-15 mg/kg per day renders the bile acid composition of bile less hydrophobic leading to a relative enrichment of UDCA to about 40%-50% of total biliary bile acids[8]. To achieve this effect in isolated cells or whole organ a high (millimolar) concentration of UDCA is required. Thus, the membrane protective effects of UDCA play a role mainly at the bile duct level, since high concentrations of bile acids are only present within the biliary tree[52]. UDCA decreases the degree of cholangiocellular injury, portal inflammation and ductular proliferation in the Mdr2-knockout mouse, an animal model of cholestasis which shares morphological features with PSC resulting from the toxic effect of bile acids on the biliary epithelium in the absence of protective mixed micelle formation with phospholipids[53,54]. In line with these observations, the (peri-) portal inflammatory reaction is less severe in patients with PBC and PSC under UDCA treatment as compared to those treated with placebo[27,28,55,56]. The effects of UDCA conjugates on cholangiocytes are apparently mediated by Ca2+- and protein kinase Cα-dependent mechanisms which have been implicated in stimulation of biliary secretion in cholestatic hepatocytes as outlined above. Although the protection against the consequences of bile duct destruction is likely to be one mechanism of action of UDCA, the underlying pathophysiologic events leading to the ongoing bile duct destruction are probably not influenced by UDCA.

UDCA IN VANISHING BILE DUCT SYN-DROMES
Primary biliary cirrhosis

Primary biliary cirrhosis (PBC) is a chronic cholestatic liver disease characterized by chronic inflammation and destruction of intrahepatic bile ductules, which leads to progressive ductopenia. ultimately resulting in fibrosis and biliary cirrhosis. The prevalence differs considerably in different geographic areas, ranging from 40 to 400 per million. It primarily affects women, with a female to male ratio as high as 10 to 1[57,58]. Current theories on the pathogenesis of PBC favor the hypothesis that the disease develops as a result of an inappropriate immune response following stimulation by an environmental or infectious agent. The pathogenetic mechanism is believed to be caused by a defect in immunologic tolerance, resulting in the activation and expansion of self-antigen specific T and B lymphocyte clones and the production of circulating autoantibodies in addition to a myriad of cytokines and other inflammatory mediators. The serologic hallmark of PBC is the presence of autoantibodies to mitochondria, especially to the E2 component of the pyruvate dehydrogenase complex (PDC)[59]. 50% to 60% of patients are asymptomatic at diagnosis, and the disease is initially detected on the basis of screening liver-biochemistry profiles. Fatigue and pruritus are the most common presenting symptoms[60], occurring in up to 78% and 70% of patients, respectively. Jaundice develops usually later, often associated with progression of the disease[61]. Eventually, complications of cirrhosis and portal hypertension, such as ascites, variceal bleeding, and hepatic encephalopathy, develop.

UDCA is now the mainstay of therapy for PBC. In 1987 Poupon and co-workers reported benefit from UDCA treatment[62]. Since then, several randomized double-blind placebo-controlled studies have shown that UDCA improves biochemical parameters including serum bilirubin levels which are used as a prognostic marker for PBC[25,27,55,63-66]. Indeed, it has been shown that the mean survival in patients with serum bilirubin levels above 34 μmol/L is 4 years; in those with values of more than 103 μmol/L only 2.1 years[67]. This prognostic value of serum bilirubin is similar in UDCA-treated patients as in non-treated patients. The benefit from UDCA therapy on liver fibrosis progression in PBC has been shown by Corpechot et al[68] using Markov modeling: A fivefold decrease in progression rate from early-stage disease toward extensive fibrosis or cirrhosis was found in UDCA-treated patients compared with placebo-treated patients. At 4 years, the probability of UDCA-treated patients to remain in early stage disease was 76% (95% confidence interval: 58%-88%), as compared with 29% (15%-52%) in placebo-treated patients. These findings are consistent with recent data showing that a 2-year treatment with UDCA reduces periportal necroinflammatory lesions, improves ductular proliferation, and delays the progression of histologic stage when given at the earlier stages (stages I-II) of the disease[69]. When started later in the disease course, UDCA can still ameliorate inflammation and ductular proliferation but is not capable of reversing fibrosis.

What is still more debatable is the effect of UDCA treatment on long-term survival. A combined analysis of three larger randomized trials, in which patients with primary biliary cirrhosis were randomly assigned to receive UDCA (n = 273) or placebo (n = 275) suggested that survival free of liver transplantation was significantly improved in patients who received UDCA treatment for 4 years[70]. This benefit was seen in patients with moderate to severe disease but not in those with mild disease (serum bilirubin concentration < 23.9 μmol/L or histologic stage I or II) probably because progression to end-stage disease did not occur in the short time interval of the study in patients with mild disease[70]. Lindor et al showed that survival of patients treated up to 6 years with UDCA was increased as compared with a placebo group or the predicted survival from the Mayo model[71]. In a large cohort of UDCA-treated PBC patients (225 patients) followed for up to 10 years survival free of liver transplantation was significantly higher than survival predicted by the Mayo model and 10-year survival among UDCA-treated patients is slightly lower than that of an age- and sex-matched population, the difference being mainly explained by mortality among cirrhotic patients[72]. However, the efficacy of UDCA in PBC has also been questioned. The extended follow-up of another clinical trial[73], as well as a randomized controlled trial with the longest follow-up[55], did not show a favorable effect of UDCA on the incidence of death or liver transplantation. In addition, a systematic review and meta-analysis of 11 randomized controlled trials, including 1272 patients, and six reports of the switch-over phases did not find a beneficial effect of UDCA on the incidence of liver-related death, liver transplantation, or the development of complications of liver disease[74]. Similarly, another meta-analysis of 16 randomized clinical trials evaluating UDCA against placebo (n = 15) or no intervention (n = 1) in 1422 patients did not detect a significant effect of UDCA on mortality[75]. However, these meta-analyses included trials of different length of follow-up, mostly only up to 24 mo, which were performed with various doses of UDCA, in part less than 13-15 mg/kg per day, a dose currently regarded as optimal for PBC. Thus, these meta-analyses have to be interpreted with caution, because survival analyses in a disease with a very long natural history over decades are ideally based on longer follow-up periods. In a further analysis including five studies with a follow-up of at least 4 years, a 32% reduction in the risk of death or need of liver transplantation was reported in patients treated with UDCA[76]. Nonetheless, to clarify the true efficacy of UDCA in the long-term treatment of PBC, additional data are needed. At present, the general recommendation is to treat PBC patients with UDCA at a dose of 13-15 mg/kg per day. Treatment should be started as early as possible, since patients with mild or moderate disease are likely to have a greater benefit from therapy than those with advanced disease.

Primary sclerosing cholangitis (PSC)

Primary sclerosing cholangitis (PSC) is a rare chronic cholestatic disease of the intra- and extrahepatic bile ducts that is generally progressive and leads to end-stage liver disease with a median survival from diagnosis to death or liver transplantation, currently estimated at 12 years[77-80]. PSC is characterized by diffuse inflammation, concentric obliterative fibrosis and progressive stricturing and dilatation of the biliary tree. The etiology and pathogenesis is unknown, but as in PBC, immune dysregulation plays an important role in the development of the disease. In Western populations, the estimated prevalence of PSC is 8.5 cases per 100 000 persons[81] and 70%-80% of PSC patients have or will develop inflammatory bowel disease. Of importance, patients with PSC have an increased risk of developing cholangiocarcinoma with an estimated yearly incidence of 1.5% after diagnosis of PSC[82-85] and patients with both PSC and ulcerative colitis may be at higher risk for developing colorectal aneuploidy, dysplasia, or cancer than UC patients without PSC[86,87]. Based on the beneficial effect of UDCA in patients with PBC, UDCA was evaluated in a number of small trials for patients with PSC in the early 1990s[28,88,89]. Although small, these studies observed significant biochemical and histologic improvements for patients receiving UDCA. Similar to the treatment of PBC, no major side effects were reported. In contrast to these promising results, the first large randomized placebo-controlled study including 105 patients with PSC could not find a clinical benefit of UDCA treatment with respect to time to treatment failure or histologic findings in a dose of 13-15 mg/kg per day during a mean follow-up period of 2.2 years[26]. A meta-analysis of six randomized clinical trials showed no difference between UDCA and placebo in the effects of incidence on death, treatment failure (including liver transplantation, varices, ascites, and encephalopathy), liver histological deterioration or liver cholangiographic deterioration[90]. However, most trials were small with an average sample size of 37 patients and the follow-up period might have been too short to show a clinical benefit, since PSC has usually a long natural history of over a decade. A large percentage of the patients had advanced disease and as in PBC, these patients might respond less to medical treatment than patients in earlier stages of disease. Finally, UDCA therapy alone will not suffice to treat the bile duct strictures typical for PSC, but may need additional mechanical intervention. Indeed, major bile duct strictures develop during UDCA treatment[91]. Based on these observation, UDCA treatment was combined with endoscopic dilatation of bile duct strictures in an 8-year prospective study. The survival of patients receiving this combination therapy was significantly better than the calculated survival without treatment[92]. As biliary enrichment of UDCA is expected to be lower in cholestasis and because of discouraging results with standard doses of UDCA (13 -15 mg/kg per day), use of high doses of UDCA in PSC has a rationale and has been evaluated by three groups[93-95]. The first study compared high-dose UDCA (20 mg/kg per day) (n = 13) to placebo (n = 13) for 2 years in 26 patients with PSC and demonstrated a significant improvement in serum levels of AP and γ-GT (no effect on bilirubin and albumin levels), and a significant reduction in progression of cholangiographic features and liver fibrosis as assessed by disease staging without significant side effects. UDCA did not improve symptoms[94]. Harnois et al[93] reported similar results in a 1-year open-label study of 30 PSC patients treated with UDCA at a dose of 25-30 mg/kg per day. Changes in the Mayo risk score at 1 year of treatment and projected survival at 4 year were compared with that observed in patients randomized to placebo or UDCA at a dose of 13-15 mg/kg per day in a previous study. A significant improvement in serum alkaline phosphatase activity, AST, albumin and total bilirubin occurred at 1 year of therapy with high-dose UDCA. The expected survival at 4 years was significantly improved for patients in the high-UDCA group when compared with a historical placebo control, but not between the dose of 13-15 mg/kg per day and placebo. As in the first study, high-dose UDCA was well tolerated. Finally, in the biggest placebo-controlled prospective study in PSC ever performed, a total of 219 patients were randomized to 17 to 23 mg/kg per day of UDCA (n = 110) or placebo (n = 109) for 5 years[95]. No statistically significant benefit from 20 mg/kg UDCA on survival without liver transplantation or prevention in cholangiocarcinoma in PSC was shown in this study. However, there was a tendency to improved survival in the UDCA-treated patients and the study was underpowered to show a possible positive effect.

UDCA has also been shown to have colon cancer chemopreventive effects in preclinical studies[96,97]. In addition, two clinical studies suggested that UDCA also decreases the risk for developing colorectal neoplasia in UC patients with PSC[98,99]. In a cross-sectional study by Tung et al involving 59 patients with ulcerative colitis and primary sclerosing cholangitis who were undergoing colonoscopic surveillance for colonic dysplasia, UDCA use was strongly associated with decreased prevalence of colonic dysplasia (odds ratio, 0.18, 95% confidence interval, 0.05 to 0.61; P = 0.005)[98]. In a blinded, prospective study by Pardi et al[99], 52 patients with ulcerative colitis and PSC were randomized to receive UDCA or placebo and were followed-up for a total of 355 person-years. Those originally assigned to receive UDCA had a relative risk of 0.26 for the development of dysplasia or cancer (95% confidence interval, 0.06-0.92; P = 0.034) suggesting a significant chemoprotective effect for UDCA in these patients.

In conclusion, UDCA therapy (13-20 mg/kg per day) of PSC is warranted and should be initiated as soon as possible and be continued life-long. In addition, major strictures should be dilated regularly in centers with experienced endoscopists. In patients with PSC and ulcerative colitis, who carry a considerable lifetime risk for colorectal neoplasia, UDCA may act as a chemopreventive drug. Due to the unresolved issues of optimal dosing and the true long-term benefit of UDCA in PSC, patients should be included in trials if possible.

Intrahepatic cholestasis of pregnancy

Intrahepatic cholestasis of pregnancy (ICP) is the most common pregnancy-related liver disorder characterized by pruritus associated with a mild or moderate increase in serum aminotransferases and serum bile acids starting in the second or third trimester of pregnancy and disappearing after delivery. ICP is associated with increased risk of intrauterine fetal death and premature delivery[100,101]. There is increasing evidence that gen-etically determined dysfunction in the canalicular ABC transporters bile salt export pump and multidrug resistance protein 3 might be risk factors for ICP development[102]. In small controlled trials and several observational trials, UDCA improved maternal pruritus and serum liver chemistries such as serum bilirubin and transaminases, and diminished the number of premature deliveries. The medication seems to be well tolerated by pregnant women and no adverse effects in mothers or newborns have been detected[103-105]. A recent prospective, randomized trial

(n = 84) comparing cholestyramine (8 g daily) and UDCA (8-10 mg/kg daily) for the treatment of ICP further supported these findings. Pruritus was more effectively reduced by UDCA than cholestyramine. Newborns were delivered significantly closer to term by patients treated with UDCA, than those treated with cholestyramine. Serum alanine and aspartate aminotransferase activities were markedly reduced by 78.5% and 73.8%, respectively, after ursodeoxycholic acid, but by only 21.4%, each, after cholestyramine therapy. Ursodeoxycholic acid, but not cholestyramine was free of adverse effects[106]. In ICP, the relief of cholestasis by UDCA has been suggested to be due to stimulation of vesicular exocytosis resulting in mobilization of an increased number of transport proteins to the canalicular membrane and, thereby, stimulation of transport systems involved in the biliary secretion of steroid mono- and disulfates. In conclusion, data that are available support the use of UDCA as first-line therapy for ICP in the third trimester.

Hepatic complications of allogeneic bone marrow transplantation

Hepatic graft-versus-host disease (GVHD) is a frequent complication after bone-marrow transplantation usually associated with a cholestatic picture characterized by elevated serum alkaline phosphatase and hyperbili-rubinemia attributed to damage of the small bile ducts. UDCA has been reported to be effective in the treatment of manifest GVHD[107,108]. In an open-label study 12 patients with refractory chronic GVHD of the liver after allogeneic bone marrow transplantation were given UDCA (10 to 15 mg/kg per day) for 6 wk. Serum tests of cholestatic liver injury showed improvement compared with baseline. After discontinuation of UDCA therapy, 11 patients were followed for 6 additional weeks. All showed significant worsening in liver function test results. Symptom scores were unchanged throughout the study. No adverse effects were observed[107]. In addition, the efficacy of UDCA has also been reported as a prophylaxis for veno-occlusive disease (VOD) of the liver after allogeneic bone marrow transplantation. In a randomized, double-blind, placebo-controlled study including 67 consecutive patients undergoing transplantation with allogeneic bone marrow with a preoperative regimen of busulfan plus cyclophosphamide were randomly assigned to receive UDCA 300 mg twice daily or placebo. The incidence of VOD was 40% in placebo recipients and 15% in the UDCA-treated recipients (P = 0.03)[109]. Thus, further studies are certainly needed, but UDCA may be considered for prophylaxis of VOD and treatment of GVHD of the liver.

Cystic fibrosis

Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the CFTR (cystic fibrosis transmembrane conductance regulator) gene resulting in the secretion of viscous bile. This may lead to the formation of bile duct plugs within small bile ducts, biliary obstruction and ultimately result in (focal to multilobular) biliary cirrhosis. Hepatobiliary complications increase with patient age and up to 7% of children and young adults with CF may present with liver cirrhosis[110]. Early uncontrolled studies showed that UDCA significantly improved laboratory tests and nutritional status of patients with CF[111-113]. A small randomized, placebo-controlled study showed clinical, biochemical and nutritional improvement in CF patients treated with UDCA (15 mg/kg per day) for one year[114]. Improvement of liver histology of CF patients treated with UDCA (10-15 mg/kg per day) for 2 years has been reported as well[115]. A higher dose of 20 mg/kg per day may be more efficious than lower doses (5-15 mg/kg per day)[111,116]. Thus, UDCA may be considered as potentially effective in CF patients. However, whether UDCA affects the natural history of liver disease in CF-particularly amelioration of the complications of portal hypertension, need for liver transplantation, or a measurable survival is unknown and further well-designed, adequately powered, randomized controlled trials should address these issues. Stimulation of biliary secretion may be the major mechanism for the therapeutic effect. Interestingly, it has been hypothesized that the therapeutic effect of UDCA is mediated in part by stimulation of ATP release into bile and subsequent ATP-induced bicarbonate secretion by bile-duct epithelia via calcium-dependent chloride channels distinct from the CFTR[117].

Footnotes

S- Editor Pan BR E- Editor Bai SH

References
1.  Hagey LR, Crombie DL, Espinosa E, Carey MC, Igimi H, Hofmann AF. Ursodeoxycholic acid in the Ursidae: biliary bile acids of bears, pandas, and related carnivores. J Lipid Res. 1993;34:1911-1917.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Hofmann AF. Pharmacology of ursodeoxycholic acid, an enterohepatic drug. Scand J Gastroenterol Suppl. 1994;204:1-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 130]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
3.  Poupon R, Poupon RE. Ursodeoxycholic acid therapy of chronic cholestatic conditions in adults and children. Pharmacol Ther. 1995;66:1-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 56]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
4.  Sugata F, Shimizu M. [Retrospective studies on gallstone disappearance (author's transl)]. Nihon Shokakibyo Gakkai Zasshi. 1974;71:75-80.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Bachrach WH, Hofmann AF. Ursodeoxycholic acid in the treatment of cholesterol cholelithiasis. part I. Dig Dis Sci. 1982;27:737-761.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 201]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
6.  Makino I, Shinozaki K, Yoshino K, Nakagawa S. [Dissolution of cholesterol gallstones by long-term administration of ursodeoxycholic acid]. Nihon Shokakibyo Gakkai Zasshi. 1975;72:690-702.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Leuschner U, Leuschner M, Sieratzki J, Kurtz W, Hübner K. Gallstone dissolution with ursodeoxycholic acid in patients with chronic active hepatitis and two years follow-up. A pilot study. Dig Dis Sci. 1985;30:642-649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 133]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
8.  Rubin RA, Kowalski TE, Khandelwal M, Malet PF. Ursodiol for hepatobiliary disorders. Ann Intern Med. 1994;121:207-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 56]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
9.  Sauer P, Benz C, Rudolph G, Klöters-Plachky P, Stremmel W, Stiehl A. Influence of cholestasis on absorption of ursodeoxycholic acid. Dig Dis Sci. 1999;44:817-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
10.  Kullak-Ublick GA, Stieger B, Hagenbuch B, Meier PJ. Hepatic transport of bile salts. Semin Liver Dis. 2000;20:273-292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 229]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
11.  Beuers U, Boyer JL, Paumgartner G. Ursodeoxycholic acid in cholestasis: potential mechanisms of action and therapeutic applications. Hepatology. 1998;28:1449-1453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 151]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
12.  Paumgartner G, Beuers U. Ursodeoxycholic acid in cholestatic liver disease: mechanisms of action and therapeutic use revisited. Hepatology. 2002;36:525-531.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 484]  [Cited by in F6Publishing: 447]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
13.  Trauner M, Graziadei IW. Review article: mechanisms of action and therapeutic applications of ursodeoxycholic acid in chronic liver diseases. Aliment Pharmacol Ther. 1999;13:979-996.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 119]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
14.  Lazaridis KN, Gores GJ, Lindor KD. Ursodeoxycholic acid 'mechanisms of action and clinical use in hepatobiliary disorders'. J Hepatol. 2001;35:134-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 282]  [Cited by in F6Publishing: 258]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
15.  Beuers U, Bilzer M, Chittattu A, Kullak-Ublick GA, Keppler D, Paumgartner G, Dombrowski F. Tauroursodeoxycholic acid inserts the apical conjugate export pump, Mrp2, into canalicular membranes and stimulates organic anion secretion by protein kinase C-dependent mechanisms in cholestatic rat liver. Hepatology. 2001;33:1206-1216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 178]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
16.  Kitani K, Ohta M, Kanai S. Tauroursodeoxycholate prevents biliary protein excretion induced by other bile salts in the rat. Am J Physiol. 1985;248:G407-G417.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Schölmerich J, Baumgartner U, Miyai K, Gerok W. Tauroursodeoxycholate prevents taurolithocholate-induced cholestasis and toxicity in rat liver. J Hepatol. 1990;10:280-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 49]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
18.  Heuman DM, Mills AS, McCall J, Hylemon PB, Pandak WM, Vlahcevic ZR. Conjugates of ursodeoxycholate protect against cholestasis and hepatocellular necrosis caused by more hydrophobic bile salts. In vivo studies in the rat. Gastroenterology. 1991;100:203-211.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Heuman DM, Pandak WM, Hylemon PB, Vlahcevic ZR. Conjugates of ursodeoxycholate protect against cytotoxicity of more hydrophobic bile salts: in vitro studies in rat hepatocytes and human erythrocytes. Hepatology. 1991;14:920-926.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 160]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
20.  Ohiwa T, Katagiri K, Hoshino M, Hayakawa T, Nakai T. Tauroursodeoxycholate and tauro-beta-muricholate exert cytoprotection by reducing intrahepatocyte taurochenodeoxycholate content. Hepatology. 1993;17:470-476.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Deroubaix X, Coche T, Depiereux E, Feytmans E. Saturation of hepatic transport of taurocholate in rats in vivo. Am J Physiol. 1991;260:G189-G196.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Galan AI, Jimenez R, Muñoz ME, Gonzalez J. Effects of ursodeoxycholate on maximal biliary secretion of bilirubin in the rat. Biochem Pharmacol. 1990;39:1175-1180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
23.  Jazrawi RP, de Caestecker JS, Goggin PM, Britten AJ, Joseph AE, Maxwell JD, Northfield TC. Kinetics of hepatic bile acid handling in cholestatic liver disease: effect of ursodeoxycholic acid. Gastroenterology. 1994;106:134-142.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Poupon RE, Chrétien Y, Poupon R, Paumgartner G. Serum bile acids in primary biliary cirrhosis: effect of ursodeoxycholic acid therapy. Hepatology. 1993;17:599-604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 97]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
25.  Heathcote EJ, Cauch-Dudek K, Walker V, Bailey RJ, Blendis LM, Ghent CN, Michieletti P, Minuk GY, Pappas SC, Scully LJ. The Canadian Multicenter Double-blind Randomized Controlled Trial of ursodeoxycholic acid in primary biliary cirrhosis. Hepatology. 1994;19:1149-1156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 241]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
26.  Lindor KD. Ursodiol for primary sclerosing cholangitis. Mayo Primary Sclerosing Cholangitis-Ursodeoxycholic Acid Study Group. N Engl J Med. 1997;336:691-695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 421]  [Cited by in F6Publishing: 366]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
27.  Poupon RE, Balkau B, Eschwège E, Poupon R. A multicenter, controlled trial of ursodiol for the treatment of primary biliary cirrhosis. UDCA-PBC Study Group. N Engl J Med. 1991;324:1548-1554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 570]  [Cited by in F6Publishing: 530]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
28.  Beuers U, Spengler U, Kruis W, Aydemir U, Wiebecke B, Heldwein W, Weinzierl M, Pape GR, Sauerbruch T, Paumgartner G. Ursodeoxycholic acid for treatment of primary sclerosing cholangitis: a placebo-controlled trial. Hepatology. 1992;16:707-714.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 324]  [Cited by in F6Publishing: 286]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
29.  Medina JF, Martínez-Ansó JJ, Prieto J. Decreased anion exchanger 2 immunoreactivity in the liver of patients with primary biliary cirrhosis. Hepatology. 1997;25:12-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 152]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
30.  Fickert P, Zollner G, Fuchsbichler A, Stumptner C, Pojer C, Zenz R, Lammert F, Stieger B, Meier PJ, Zatloukal K. Effects of ursodeoxycholic and cholic acid feeding on hepatocellular transporter expression in mouse liver. Gastroenterology. 2001;121:170-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 210]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
31.  Kurz AK, Graf D, Schmitt M, Vom Dahl S, Häussinger D. Tauroursodesoxycholate-induced choleresis involves p38(MAPK) activation and translocation of the bile salt export pump in rats. Gastroenterology. 2001;121:407-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 121]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
32.  Zollner G, Fickert P, Fuchsbichler A, Silbert D, Wagner M, Arbeiter S, Gonzalez FJ, Marschall HU, Zatloukal K, Denk H. Role of nuclear bile acid receptor, FXR, in adaptive ABC transporter regulation by cholic and ursodeoxycholic acid in mouse liver, kidney and intestine. J Hepatol. 2003;39:480-488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 133]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
33.  Trauner M, Meier PJ, Boyer JL. Molecular pathogenesis of cholestasis. N Engl J Med. 1998;339:1217-1227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 579]  [Cited by in F6Publishing: 497]  [Article Influence: 19.1]  [Reference Citation Analysis (1)]
34.  Beuers U, Nathanson MH, Isales CM, Boyer JL. Tauroursodeoxycholic acid stimulates hepatocellular exocytosis and mobilizes extracellular Ca++ mechanisms defective in cholestasis. J Clin Invest. 1993;92:2984-2993.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 119]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
35.  Beuers U, Throckmorton DC, Anderson MS, Isales CM, Thasler W, Kullak-Ublick GA, Sauter G, Koebe HG, Paumgartner G, Boyer JL. Tauroursodeoxycholic acid activates protein kinase C in isolated rat hepatocytes. Gastroenterology. 1996;110:1553-1563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 108]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
36.  Schliess F, Kurz AK, vom Dahl S, Häussinger D. Mitogen-activated protein kinases mediate the stimulation of bile acid secretion by tauroursodeoxycholate in rat liver. Gastroenterology. 1997;113:1306-1314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 85]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
37.  Noe J, Hagenbuch B, Meier PJ, St-Pierre MV. Characterization of the mouse bile salt export pump overexpressed in the baculovirus system. Hepatology. 2001;33:1223-1231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 87]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
38.  Yoon JH, Gores GJ. Death receptor-mediated apoptosis and the liver. J Hepatol. 2002;37:400-410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 145]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
39.  Guicciardi ME, Gores GJ. Ursodeoxycholic acid cytoprotection: dancing with death receptors and survival pathways. Hepatology. 2002;35:971-973.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 21]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
40.  Harada K, Ozaki S, Gershwin ME, Nakanuma Y. Enhanced apoptosis relates to bile duct loss in primary biliary cirrhosis. Hepatology. 1997;26:1399-1405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 119]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
41.  Higuchi H, Gores GJ. Mechanisms of liver injury: an overview. Curr Mol Med. 2003;3:483-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 96]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
42.  Faubion WA, Guicciardi ME, Miyoshi H, Bronk SF, Roberts PJ, Svingen PA, Kaufmann SH, Gores GJ. Toxic bile salts induce rodent hepatocyte apoptosis via direct activation of Fas. J Clin Invest. 1999;103:137-145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 420]  [Cited by in F6Publishing: 430]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
43.  Benz C, Angermüller S, Töx U, Klöters-Plachky P, Riedel HD, Sauer P, Stremmel W, Stiehl A. Effect of tauroursodeoxycholic acid on bile-acid-induced apoptosis and cytolysis in rat hepatocytes. J Hepatol. 1998;28:99-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 107]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
44.  Benz C, Angermüller S, Otto G, Sauer P, Stremmel W, Stiehl A. Effect of tauroursodeoxycholic acid on bile acid-induced apoptosis in primary human hepatocytes. Eur J Clin Invest. 2000;30:203-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 70]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
45.  Rodrigues CM, Fan G, Wong PY, Kren BT, Steer CJ. Ursodeoxycholic acid may inhibit deoxycholic acid-induced apoptosis by modulating mitochondrial transmembrane potential and reactive oxygen species production. Mol Med. 1998;4:165-178.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Graf D, Kurz AK, Fischer R, Reinehr R, Häussinger D. Taurolithocholic acid-3 sulfate induces CD95 trafficking and apoptosis in a c-Jun N-terminal kinase-dependent manner. Gastroenterology. 2002;122:1411-1427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 85]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
47.  Qiao L, Yacoub A, Studer E, Gupta S, Pei XY, Grant S, Hylemon PB, Dent P. Inhibition of the MAPK and PI3K pathways enhances UDCA-induced apoptosis in primary rodent hepatocytes. Hepatology. 2002;35:779-789.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 115]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
48.  Hofmann AF. Bile Acids: The Good, the Bad, and the Ugly. News Physiol Sci. 1999;14:24-29.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Güldütuna S, Zimmer G, Imhof M, Bhatti S, You T, Leuschner U. Molecular aspects of membrane stabilization by ursodeoxycholate [see comment]. Gastroenterology. 1993;104:1736-1744.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Heuman DM, Bajaj RS, Lin Q. Adsorption of mixtures of bile salt taurine conjugates to lecithin-cholesterol membranes: implications for bile salt toxicity and cytoprotection. J Lipid Res. 1996;37:562-573.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Heuman DM, Bajaj R. Ursodeoxycholate conjugates protect against disruption of cholesterol-rich membranes by bile salts. Gastroenterology. 1994;106:1333-1341.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Heuman DM. Hepatoprotective properties of ursodeoxycholic acid. Gastroenterology. 1993;104:1865-1870.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Van Nieuwkerk CM, Elferink RP, Groen AK, Ottenhoff R, Tytgat GN, Dingemans KP, Van Den Bergh Weerman MA, Offerhaus GJ. Effects of Ursodeoxycholate and cholate feeding on liver disease in FVB mice with a disrupted mdr2 P-glycoprotein gene. Gastroenterology. 1996;111:165-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 134]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
54.  Fickert P, Zollner G, Fuchsbichler A, Stumptner C, Weiglein AH, Lammert F, Marschall HU, Tsybrovskyy O, Zatloukal K, Denk H. Ursodeoxycholic acid aggravates bile infarcts in bile duct-ligated and Mdr2 knockout mice via disruption of cholangioles. Gastroenterology. 2002;123:1238-1251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 231]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
55.  Parés A, Caballería L, Rodés J, Bruguera M, Rodrigo L, García-Plaza A, Berenguer J, Rodríguez-Martínez D, Mercader J, Velicia R. Long-term effects of ursodeoxycholic acid in primary biliary cirrhosis: results of a double-blind controlled multicentric trial. UDCA-Cooperative Group from the Spanish Association for the Study of the Liver. J Hepatol. 2000;32:561-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 205]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
56.  Stiehl A, Walker S, Stiehl L, Rudolph G, Hofmann WJ, Theilmann L. Effect of ursodeoxycholic acid on liver and bile duct disease in primary sclerosing cholangitis. A 3-year pilot study with a placebo-controlled study period. J Hepatol. 1994;20:57-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 155]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
57.  Metcalf J, James O. The geoepidemiology of primary biliary cirrhosis. Semin Liver Dis. 1997;17:13-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 73]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
58.  Kim WR, Lindor KD, Locke GR 3rd, Therneau TM, Homburger HA, Batts KP, Yawn BP, Petz JL, Melton LJ 3rd, Dickson ER. Epidemiology and natural history of primary biliary cirrhosis in a US community. Gastroenterology. 2000;119:1631-1636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 272]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
59.  Nishio A, Keeffe EB, Gershwin ME. Immunopathogenesis of primary biliary cirrhosis. Semin Liver Dis. 2002;22:291-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
60.  Bergasa NV. Pruritus and fatigue in primary biliary cirrhosis. Clin Liver Dis. 2003;7:879-900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
61.  Sherlock S, Scheuer PJ. The presentation and diagnosis of 100 patients with primary biliary cirrhosis. N Engl J Med. 1973;289:674-678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 369]  [Cited by in F6Publishing: 356]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
62.  Poupon R, Chrétien Y, Poupon RE, Ballet F, Calmus Y, Darnis F. Is ursodeoxycholic acid an effective treatment for primary biliary cirrhosis. Lancet. 1987;1:834-836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 398]  [Cited by in F6Publishing: 385]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
63.  Leuschner U, Fischer H, Kurtz W, Güldütuna S, Hübner K, Hellstern A, Gatzen M, Leuschner M. Ursodeoxycholic acid in primary biliary cirrhosis: results of a controlled double-blind trial. Gastroenterology. 1989;97:1268-1274.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Combes B, Carithers RL Jr, Maddrey WC, Lin D, McDonald MF, Wheeler DE, Eigenbrodt EH, Muñoz SJ, Rubin R, Garcia-Tsao G. A randomized, double-blind, placebo-controlled trial of ursodeoxycholic acid in primary biliary cirrhosis. Hepatology. 1995;22:759-766.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Battezzati PM, Podda M, Bianchi FB, Naccarato R, Orlandi F, Surrenti C, Pagliaro L, Manenti F. Ursodeoxycholic acid for symptomatic primary biliary cirrhosis. Preliminary analysis of a double-blind multicenter trial. Italian Multicenter Group for the Study of UDCA in PBC. J Hepatol. 1993;17:332-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 54]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
66.  Lindor KD, Dickson ER, Baldus WP, Jorgensen RA, Ludwig J, Murtaugh PA, Harrison JM, Wiesner RH, Anderson ML, Lange SM. Ursodeoxycholic acid in the treatment of primary biliary cirrhosis. Gastroenterology. 1994;106:1284-1290.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Shapiro JM, Smith H, Schaffner F. Serum bilirubin: a prognostic factor in primary biliary cirrhosis. Gut. 1979;20:137-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 241]  [Cited by in F6Publishing: 241]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
68.  Corpechot C, Carrat F, Bonnand AM, Poupon RE, Poupon R. The effect of ursodeoxycholic acid therapy on liver fibrosis progression in primary biliary cirrhosis. Hepatology. 2000;32:1196-1199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 239]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
69.  Poupon R, Chazouillères O, Balkau B, Poupon RE. Clinical and biochemical expression of the histopathological lesions of primary biliary cirrhosis. UDCA-PBC Group. J Hepatol. 1999;30:408-412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 66]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
70.  Poupon RE, Lindor KD, Cauch-Dudek K, Dickson ER, Poupon R, Heathcote EJ. Combined analysis of randomized controlled trials of ursodeoxycholic acid in primary biliary cirrhosis. Gastroenterology. 1997;113:884-890.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 477]  [Cited by in F6Publishing: 408]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
71.  Lindor KD, Therneau TM, Jorgensen RA, Malinchoc M, Dickson ER. Effects of ursodeoxycholic acid on survival in patients with primary biliary cirrhosis. Gastroenterology. 1996;110:1515-1518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 174]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
72.  Poupon RE, Bonnand AM, Chrétien Y, Poupon R. Ten-year survival in ursodeoxycholic acid-treated patients with primary biliary cirrhosis. The UDCA-PBC Study Group. Hepatology. 1999;29:1668-1671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 160]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
73.  Kilmurry MR, Heathcote EJ, Cauch-Dudek K, O'Rourke K, Bailey RJ, Blendis LM, Ghent CN, Minuk GY, Pappas SC, Scully LJ. Is the Mayo model for predicting survival useful after the introduction of ursodeoxycholic acid treatment for primary biliary cirrhosis. Hepatology. 1996;23:1148-1153.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Goulis J, Leandro G, Burroughs AK. Randomised controlled trials of ursodeoxycholic-acid therapy for primary biliary cirrhosis: a meta-analysis. Lancet. 1999;354:1053-1060.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 240]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
75.  Gluud C, Christensen E. Ursodeoxycholic acid for primary biliary cirrhosis. Cochrane Database Syst Rev. 2002;CD000551.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Lindor KD, Poupon R, Poupon R, Heathcote EJ, Therneau T. Ursodeoxycholic acid for primary biliary cirrhosis. Lancet. 2000;355:657-658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 47]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
77.  Wiesner RH, Grambsch PM, Dickson ER, Ludwig J, MacCarty RL, Hunter EB, Fleming TR, Fisher LD, Beaver SJ, LaRusso NF. Primary sclerosing cholangitis: natural history, prognostic factors and survival analysis. Hepatology. 1989;10:430-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 483]  [Cited by in F6Publishing: 486]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
78.  Farrant JM, Hayllar KM, Wilkinson ML, Karani J, Portmann BC, Westaby D, Williams R. Natural history and prognostic variables in primary sclerosing cholangitis. Gastroenterology. 1991;100:1710-1717.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Broomé U, Olsson R, Lööf L, Bodemar G, Hultcrantz R, Danielsson A, Prytz H, Sandberg-Gertzén H, Wallerstedt S, Lindberg G. Natural history and prognostic factors in 305 Swedish patients with primary sclerosing cholangitis. Gut. 1996;38:610-615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 596]  [Cited by in F6Publishing: 633]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
80.  Boberg KM, Rocca G, Egeland T, Bergquist A, Broomé U, Caballeria L, Chapman R, Hultcrantz R, Mitchell S, Pares A. Time-dependent Cox regression model is superior in prediction of prognosis in primary sclerosing cholangitis. Hepatology. 2002;35:652-657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 72]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
81.  Boberg KM, Aadland E, Jahnsen J, Raknerud N, Stiris M, Bell H. Incidence and prevalence of primary biliary cirrhosis, primary sclerosing cholangitis, and autoimmune hepatitis in a Norwegian population. Scand J Gastroenterol. 1998;33:99-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 362]  [Cited by in F6Publishing: 315]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
82.  Brandsaeter B, Isoniemi H, Broomé U, Olausson M, Bäckman L, Hansen B, Schrumpf E, Oksanen A, Ericzon BG, Höckerstedt K. Liver transplantation for primary sclerosing cholangitis; predictors and consequences of hepatobiliary malignancy. J Hepatol. 2004;40:815-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 126]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
83.  Lee YM, Kaplan MM. Primary sclerosing cholangitis. N Engl J Med. 1995;332:924-933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 424]  [Cited by in F6Publishing: 419]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
84.  Bergquist A, Ekbom A, Olsson R, Kornfeldt D, Lööf L, Danielsson A, Hultcrantz R, Lindgren S, Prytz H, Sandberg-Gertzén H. Hepatic and extrahepatic malignancies in primary sclerosing cholangitis. J Hepatol. 2002;36:321-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 531]  [Cited by in F6Publishing: 442]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
85.  Portincasa P, Vacca M, Moschetta A, Petruzzelli M, Palasciano G, van Erpecum KJ, van Berge-Henegouwen GP. Primary sclerosing cholangitis: updates in diagnosis and therapy. World J Gastroenterol. 2005;11:7-16.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Broomé U, Löfberg R, Veress B, Eriksson LS. Primary sclerosing cholangitis and ulcerative colitis: evidence for increased neoplastic potential. Hepatology. 1995;22:1404-1408.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Marchesa P, Lashner BA, Lavery IC, Milsom J, Hull TL, Strong SA, Church JM, Navarro G, Fazio VW. The risk of cancer and dysplasia among ulcerative colitis patients with primary sclerosing cholangitis. Am J Gastroenterol. 1997;92:1285-1288.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Stiehl A. Ursodeoxycholic acid therapy in treatment of primary sclerosing cholangitis. Scand J Gastroenterol Suppl. 1994;204:59-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 29]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
89.  O'Brien CB, Senior JR, Arora-Mirchandani R, Batta AK, Salen G. Ursodeoxycholic acid for the treatment of primary sclerosing cholangitis: a 30-month pilot study. Hepatology. 1991;14:838-847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 136]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
90.  Chen W, Gluud C. Bile acids for primary sclerosing cholangitis. Cochrane Database Syst Rev. 2003;CD003626.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Stiehl A, Rudolph G, Klöters-Plachky P, Sauer P, Walker S. Development of dominant bile duct stenoses in patients with primary sclerosing cholangitis treated with ursodeoxycholic acid: outcome after endoscopic treatment. J Hepatol. 2002;36:151-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 168]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
92.  Stiehl A, Rudolph G, Sauer P, Benz C, Stremmel W, Walker S, Theilmann L. Efficacy of ursodeoxycholic acid treatment and endoscopic dilation of major duct stenoses in primary sclerosing cholangitis. An 8-year prospective study. J Hepatol. 1997;26:560-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 136]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
93.  Harnois DM, Angulo P, Jorgensen RA, Larusso NF, Lindor KD. High-dose ursodeoxycholic acid as a therapy for patients with primary sclerosing cholangitis. Am J Gastroenterol. 2001;96:1558-1562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 174]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
94.  Mitchell SA, Bansi DS, Hunt N, Von Bergmann K, Fleming KA, Chapman RW. A preliminary trial of high-dose ursodeoxycholic acid in primary sclerosing cholangitis. Gastroenterology. 2001;121:900-907.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 258]  [Cited by in F6Publishing: 261]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
95.  Olsson R, Boberg KM, de Muckadell OS, Lindgren S, Hultcrantz R, Folvik G, Bell H, Gangsøy-Kristiansen M, Matre J, Rydning A. High-dose ursodeoxycholic acid in primary sclerosing cholangitis: a 5-year multicenter, randomized, controlled study. Gastroenterology. 2005;129:1464-1472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 244]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
96.  Earnest DL, Holubec H, Wali RK, Jolley CS, Bissonette M, Bhattacharyya AK, Roy H, Khare S, Brasitus TA. Chemoprevention of azoxymethane-induced colonic carcinogenesis by supplemental dietary ursodeoxycholic acid. Cancer Res. 1994;54:5071-5074.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Ikegami T, Matsuzaki Y, Shoda J, Kano M, Hirabayashi N, Tanaka N. The chemopreventive role of ursodeoxycholic acid in azoxymethane-treated rats: suppressive effects on enhanced group II phospholipase A2 expression in colonic tissue. Cancer Lett. 1998;134:129-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 53]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
98.  Tung BY, Emond MJ, Haggitt RC, Bronner MP, Kimmey MB, Kowdley KV, Brentnall TA. Ursodiol use is associated with lower prevalence of colonic neoplasia in patients with ulcerative colitis and primary sclerosing cholangitis. Ann Intern Med. 2001;134:89-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in F6Publishing: 339]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
99.  Pardi DS, Loftus EV Jr, Kremers WK, Keach J, Lindor KD. Ursodeoxycholic acid as a chemopreventive agent in patients with ulcerative colitis and primary sclerosing cholangitis. Gastroenterology. 2003;124:889-893.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 423]  [Cited by in F6Publishing: 440]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
100.  Bacq Y, Sapey T, Bréchot MC, Pierre F, Fignon A, Dubois F. Intrahepatic cholestasis of pregnancy: a French prospective study. Hepatology. 1997;26:358-364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 200]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
101.  Alsulyman OM, Ouzounian JG, Ames-Castro M, Goodwin TM. Intrahepatic cholestasis of pregnancy: perinatal outcome associated with expectant management. Am J Obstet Gynecol. 1996;175:957-960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 126]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
102.  Jacquemin E. Role of multidrug resistance 3 deficiency in pediatric and adult liver disease: one gene for three diseases. Semin Liver Dis. 2001;21:551-562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 125]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
103.  Palma J, Reyes H, Ribalta J, Hernández I, Sandoval L, Almuna R, Liepins J, Lira F, Sedano M, Silva O. Ursodeoxycholic acid in the treatment of cholestasis of pregnancy: a randomized, double-blind study controlled with placebo. J Hepatol. 1997;27:1022-1028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 257]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
104.  Jenkins JK, Boothby LA. Treatment of itching associated with intrahepatic cholestasis of pregnancy. Ann Pharmacother. 2002;36:1462-1465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
105.  Burrows RF, Clavisi O, Burrows E. Interventions for treating cholestasis in pregnancy. Cochrane Database Syst Rev. 2001;CD000493.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Kondrackiene J, Beuers U, Kupcinskas L. Efficacy and safety of ursodeoxycholic acid versus cholestyramine in intrahepatic cholestasis of pregnancy. Gastroenterology. 2005;129:894-901.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 179]  [Article Influence: 9.4]  [Reference Citation Analysis (1)]
107.  Fried RH, Murakami CS, Fisher LD, Willson RA, Sullivan KM, McDonald GB. Ursodeoxycholic acid treatment of refractory chronic graft-versus-host disease of the liver. Ann Intern Med. 1992;116:624-629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 68]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
108.  Wulffraat NM, Haddad E, Benkerrou M, Spliet WG, Patey N, Fischer A, de Graeff-Meeder BR. Hepatic GVHD after HLA-haploidentical bone marrow transplantation in children with severe combined immunodeficiency: the effect of ursodeoxycholic acid. Br J Haematol. 1997;96:776-780.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
109.  Essell JH, Schroeder MT, Harman GS, Halvorson R, Lew V, Callander N, Snyder M, Lewis SK, Allerton JP, Thompson JM. Ursodiol prophylaxis against hepatic complications of allogeneic bone marrow transplantation. A randomized, double-blind, placebo-controlled trial. Ann Intern Med. 1998;128:975-981.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 119]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
110.  Feigelson J, Anagnostopoulos C, Poquet M, Pecau Y, Munck A, Navarro J. Liver cirrhosis in cystic fibrosis--therapeutic implications and long term follow up. Arch Dis Child. 1993;68:653-657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 109]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
111.  Colombo C, Castellani MR, Balistreri WF, Seregni E, Assaisso ML, Giunta A. Scintigraphic documentation of an improvement in hepatobiliary excretory function after treatment with ursodeoxycholic acid in patients with cystic fibrosis and associated liver disease. Hepatology. 1992;15:677-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 83]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
112.  Colombo C, Crosignani A, Assaisso M, Battezzati PM, Podda M, Giunta A, Zimmer-Nechemias L, Setchell KD. Ursodeoxycholic acid therapy in cystic fibrosis-associated liver disease: a dose-response study. Hepatology. 1992;16:924-930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 95]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
113.  Cotting J, Lentze MJ, Reichen J. Effects of ursodeoxycholic acid treatment on nutrition and liver function in patients with cystic fibrosis and longstanding cholestasis. Gut. 1990;31:918-921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 101]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
114.  Colombo C, Battezzati PM, Podda M, Bettinardi N, Giunta A. Ursodeoxycholic acid for liver disease associated with cystic fibrosis: a double-blind multicenter trial. The Italian Group for the Study of Ursodeoxycholic Acid in Cystic Fibrosis. Hepatology. 1996;23:1484-1490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 163]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
115.  Lindblad A, Glaumann H, Strandvik B. A two-year prospective study of the effect of ursodeoxycholic acid on urinary bile acid excretion and liver morphology in cystic fibrosis-associated liver disease. Hepatology. 1998;27:166-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 129]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
116.  van de Meeberg PC, Houwen RH, Sinaasappel M, Heijerman HG, Bijleveld CM, Vanberge-Henegouwen GP. Low-dose versus high-dose ursodeoxycholic acid in cystic fibrosis-related cholestatic liver disease. Results of a randomized study with 1-year follow-up. Scand J Gastroenterol. 1997;32:369-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 50]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
117.  Nathanson MH, Burgstahler AD, Masyuk A, Larusso NF. Stimulation of ATP secretion in the liver by therapeutic bile acids. Biochem J. 2001;358:1-5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 45]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]