Guidelines For Basic Science Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Nov 21, 2012; 18(43): 6189-6196
Published online Nov 21, 2012. doi: 10.3748/wjg.v18.i43.6189
MicroRNAs in biliary diseases
Patricia Munoz-Garrido, Elizabeth Hijona, Miguel Carracedo, Luis Bujanda, Jesús M Banales, Division of Hepatology and Gastroenterology, Biodonostia Research Institute, Donostia Universitary Hospital, 20014 San Sebastián, Spain
Maite G Fernandez-Barrena, Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905, United States
Elizabeth Hijona, José J G Marín, Luis Bujanda, Jesús M Banales, Department of Liver and Gastrointestinal Diseases, National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, funded by the Spanish Carlos III Institute), 08036 Barcelona, Spain
José J G Marín, Department of Physiology and Pharmacology, Experimental Hepatology and Drug Targeting (HEVEFARM), Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, 37008 Salamanca, Spain
Jesús M Banales, Ikerbasque, Basque Foundation for Science, 48011 Bilbao, Spain
Author contributions: Munoz-Garrido P and Banales JM performed the manuscript concept and design, drafting of the text and critical revision; Fernandez-Barrena MG, Hijona E, Carracedo M, Marín JJG and Bufanda L performed the drafting of the manuscript and critical revision.
Correspondence to: Jesús M Banales, PhD, Division of Hepatology and Gastroenterology, Biodonostia Research Institute-Donostia Universitary Hospital, Paseo Dr. Begiristain s/n, 20014 San Sebastián, Spain. jesus.banales@biodonostia.org
Telephone: +34-943-006125 Fax: +34-943-006250
Received: May 10, 2012
Revised: July 5, 2012
Accepted: August 14, 2012
Published online: November 21, 2012

Abstract

Cholangiopathies are a group of diseases primarily or secondarily affecting bile duct cells, and result in cholangiocyte proliferation, regression, and/or transformation. Their etiopathogenesis may be associated with a broad variety of causes of different nature, which includes genetic, neoplastic, immune-associated, infectious, vascular, and drug-induced alterations, or being idiopathic. miRNAs, small non-coding endogenous RNAs that post-transcriptionally regulate gene expression, have been associated with pathophysiological processes in different organs and cell types, and are postulated as potential targets for diagnosis and therapy. In the current manuscript, knowledge regarding the role of miRNAs in the development and/or progression of cholangiopathies has been reviewed and the most relevant findings in this promising field of hepatology have been highlighted.

Key Words: miRNAs, Cholangiopathies, Cholangiocarcinoma, Polycystic liver diseases, Primary biliary cirrhosis



INTRODUCTION

Cholangiocytes are the epithelial cells lining the bile ducts, and are key players in normal liver physiology[1]. They participate in fundamental secretory processes by modifying the composition and flow of primary bile generated at the canaliculi of hepatocytes upstream of the biliary tree. Thus, transport of water, ions or solutes between the blood and the bile-duct lumen finally result in fluidization and alkalinization of bile[1-4]. Although cholangiocytes only represent 3%-5% of the total liver cell population, they may account for up to 30% of total bile flow[1]. Cholangiocytes contain a single primary cilium extending from the apical membrane into the bile-duct lumen[5]. This antenna-like organelle possesses sensory properties that detect physical and chemical changes in bile that are transmitted into the cell to activate signaling pathways and finally modify cellular functions. The primary cilium of cholangiocytes functions as a mechano- (detecting changes in bile flow)[6], chemo- (interacting with different molecules[7] and vesicles[8]), and osmo- (identifying changes in osmolarity: hypo- and hyper-tonicity)[9] sensor.

In addition, cholangiocytes are primarily or secondarily affected in a group of diseases termed cholangiopathies, whose etiopathogenesis is of diverse nature (genetic, neoplastic, immune-associated, idiopathic, infectious, vascular or drug-induced)[10]. These biliary diseases may result in cholangiocyte proliferation, regression and/or transformation. Important advances are being achieved in understanding the molecular mechanisms involved in the development and progression of these disorders. In this regard, there is increasing evidence suggesting the role of miRNAs in the etiopathogenesis of cholangiopathies, which are postulated as potential targets for diagnosis and therapy.

miRNAs are small non-coding RNAs (approximately 22 nucleotides) that regulate the expression of multiple genes by binding to complementary sites of targeted mRNAs, causing translational repression (imperfect target duplexes) or degradation (perfect matches)[11,12]. They participate in the regulation of multiple cell types under physiological and pathological conditions, and are fundamental in different cellular processes such as development, proliferation, apoptosis, metabolism, morphogenesis, and in diseases[13,14]. In the current article, knowledge regarding the role of miRNAs in the development and/or progression of cholangiopathies has been highlighted.

CHOLANGIOCARCINOMA

Cholangiocarcinoma (CCA) is a malignant tumor affecting the biliary tree[15,16]. The incidence of CCA (1:50 000) is increasing worldwide and the therapeutic options are very limited because of high chemoresistance. CCA accounts for up to 10%-15% of primary hepatobiliary malignancies and 3% of all gastrointestinal tumors, affecting individuals of both sexes. Owing to slow growth and late metastasis, this cancer is usually diagnosed in patients older than 65 years when it is at an advanced stage, thus reducing the success of surgical procedures. Complications of CCA include bile-duct obstruction, liver failure, metastasis to other organs, infections, and uncontrolled vomiting. Although the pathogenesis of CCA is poorly understood, the presence of primary sclerosing cholangitis, chronic biliary irritation, or choledochal cysts appears to predispose to the development of this cancer.

CCAs can be anatomically classified into extrahepatic or intrahepatic[15,16]. Extrahepatic is more common than intrahepatic CCA, accounting for up to 80%-95% of all CCAs. In addition, depending on the tumor location in the extrahepatic biliary system, extrahepatic CCA can be divided into proximal or perihilar (also frequently referred to as Klatskin tumor), and distal. On the other hand, intrahepatic CCA originates within the liver and accounts for up to 5%-20% of all CCAs.

Increasing evidence suggests the importance of miRNAs in the pathogenesis of CCA. Thus, global changes in the miRNA profile of malignant cholangiocytes has been reported[17-21], which may alter different cholangiocyte features such as cell cycle, proliferation, migration and apoptosis (Table 1).

Table 1 Classification of miRNAs differentially expressed in cholangiocarcinoma regarding their action, expression and targets.
miRNAExpressionDiseaseTargetAltered functionRef.
miR-320DownHuman-ICCMcl-1[17]
miR-29bDownHuman-CCAMcl-1[21,30]
miR-204DownHuman-ICCBcl-2Apoptosis[17]
miR-25UpHuman-ICCDR4[29]
miR-21UpHuman-CCAPDCD4[20,25]
miR-421UpHuman-ICCFXR[22]
miR-34aDownMouse-CCAc-Myc[27]
miR-210UpMouse-CCAMntProliferation[27]
miR-21UpHuman-CCAPTEN[18,21,25]
miR-26aUpHuman-CCAGSK-3β[26]
miR-421UpHuman-ICCFXR[22]
miR-21UpHuman-CCATIMP-3Migration[20]
miR-26aUpHuman-CCAGSK-3β[26]
miR-494DownHuman-CCACDK-6Cell cycle[19]
miR-141UpHuman-CCAGen CLOCKCircadian rhythm[18]
miR-370DownHuman-CCAMAP3K8, DNMT-1[38]
miR-373DownHuman-ECCMBD2Epigenetics[36,37]
miR-148aDownHuman-CCADNMT-1[39]
miR-152DownHuman-CCADNMT-1[39]
miR-let7aUpHuman-CCANF2[41]
miR-21UpHuman-CCAPTENChemoresistance[18]
miR-200bUpHuman-CCAPTPN12[18]
Cell cycle, proliferation and migration

Different miRNAs are able to act as onco-miRNAs by repressing the expression of tumor suppressor genes. In this regard, miR-421, upregulated in human CCA, was reported to modulate the expression of farnesoid X receptor (FXR), an event associated with cell proliferation, colony formation, and migration in vitro[22]. FXR is a member of the nuclear receptor superfamily that plays crucial roles in bile acid, cholesterol, lipid and glucose metabolism[23,24]. Likewise, FXR was shown to act as a tumor suppressor for hepatocellular carcinoma and breast cancer[23,24]. Aberrant bile-acid secretion has been linked to CCA, therefore, further investigations will be needed to test the interplay between bile acids, FXR and miRNAs on biliary tract tumorigenesis.

Another miRNA involved in tumor proliferation is miR-21, which is reported to be overexpressed in human CCA due to upregulation of arsenic resistance protein 2 (Ars2)[25]. This protein plays an important role in miRNA biogenesis, and hence its depletion reduces the levels of different miRNAs, including miR-21. Ars2 knockdown in CCA cells decreases miR-21 levels, inhibits cell proliferation, and prevents tumor formation in immunodeficient mice. It is suggested that miRNA-21 may negatively control the expression of tumor suppressor phosphatase and tension homolog (PTEN)[18,21,25] leading to cell proliferation. Moreover, miR-21 is suggested to regulate the expression of the tissue inhibitor of metalloproteinase (TIMP)-3[20], an inhibitor of cell-matrix TIMP activity downregulated in CCA, and could result in increased migration properties.

In addition, it has been recently reported that miRNA-26a is overexpressed in human CCA promoting cell proliferation and migration in vitro, and tumor growth in vivo[26]. This miRNA was demonstrated to downregulate directly the expression of glycogen synthase kinase (GSK)-3β[26]. This enzyme phosphorylates the serine and threonine residues of β-catenin, leading to its degradation. Thus, miR-26a/GSK-3β targeting results in intracellular accumulation of β-catenin that activates the expression of c-Myc, cyclin D1, and peroxisome proliferator-activated receptor δ; three proteins involved in dedifferentiation, tumor growth, and migration[26].

Induction of chronic cholestasis in a new murine model accelerates the progression of CCA by altering miRNA-34a, let-7a and miRNA-210 expression[27]. Downregulation of miRNA-34a results in overexpression of its target c-Myc that mediates the upregulation of cyclin D1. In this regard, knockdown of c-Myc reduces progression of CCA. Moreover, in this animal model, the suppressor of miRNAs biogenesis LIN28B is found to be upregulated. LIN28B specifically binds to the family of let-7 miRNA precursors, inhibiting their processing, and inducing their degradation[28]. These experimental data suggest that LIN28B upregulation in humans might also be associated with the inhibition of let-7a, a miRNA involved in the development of cystic hyperplasia, cystic atypical hyperplasia, cholangioma and CCA. On the other hand, the hypoxia inducible factor-2α was shown to mediate miRNA-210 upregulation, which further inhibits Mnt (transcriptional repressor and antagonist of c-Myc-dependent transcriptional activation and cell growth)[27].

Among those miRNAs reported to be downregulated in CCA based on miRNA arrays, miR-494 is involved in the control of the cell cycle by direct targeting cyclin-dependent kinase-6[19]. Thus, it is suggested that miRNA-494 could be a potential therapeutic target for CCA because its overexpression decreases the growth of bile-duct cancer cells in vitro and in vivo[19].

Apoptosis

Activation of anti-apoptotic pathways is a general event involved in carcinogenesis. In this regard, CCA is often associated with alterations in the expression profile of miRNAs that regulate apoptosis. Thus, miRNA-25, which is overexpressed in CCA, may protect cholangiocytes from tumour necrosis factor-related apoptosis-inducing ligand-induced apoptosis through the inhibition of death receptor 4[29]. Likewise, the overexpression of miRNA-21 in CCA is also able to downregulate the programmed cell death protein 4[20,25].

On the other hand, several miRNAs that modulate proapoptotic mechanisms are downregulated in CCA. Among those downregulated miRNAs, both miR-320[17] and miR-29b[21,30] play a role in the expression control of the antiapoptotic effector myeloid cell leukemia sequence 1, thus promoting cell survival and proliferation. Moreover, miR-204, which is also downregulated in CCA, targets the antiapoptotic protein B-cell lymphoma 2[17].

Epigenetics

Although genetic alterations have been widely associated with CCA, epigenetic modifications are poorly understood in this type of cancer. However, CCA has been associated with aberrant DNA methylation, which is an essential mechanism for gene expression regulation[31,32]. This mechanism is mostly regulated by at least three DNA methyltransferases (DNMT-1, DNMT-3A and DNMT-3B). Once a DNA sequence becomes methylated, it can repress transcription by blocking the recognition of transcriptional activators to DNA sequences, or by recruiting methyl-CpG-binding domain (MBD) proteins to modify chromatin compaction. MBD proteins are transcription repressors that bind to methylated gene promoters, resulting in gene expression silencing. Interestingly, aberrant DNA methylation has been found in many types of cancers, indicating that hypomethylation or hypermethylation of gene promoter CpG islands may result in tumor cell genomic instability or tumor suppressor gene silencing, respectively[33,34]. Moreover, both miRNAs and methylation are reversible regulators that can interact with each other[35]. In this regard, miRNA-373, which was recently reported to be downregulated in hilar CCA, negatively regulates MBD2 protein expression through specific binding to its 3′ untranslated region (3′-UTR)[36]. As a result of miRNA-373 downregulation in CCA, MBD2 expression is upregulated, leading to hypermethylation and silencing of the tumor suppressor gene RASSF1A. Hence, miRNA-373 downregulation is associated with poor cell differentiation, advanced clinical stage, and shorter survival in hilar CCA. Interestingly, miRNA-373 expression can also be regulated by MBD2 in CCA[37].

In addition, it has been demonstrated that interleukin-6 (IL-6) may epigenetically regulate the expression of selected miRNAs and contribute to CCA growth[38]. IL-6, an inflammation-associated cytokine that induces mitogen and survival features in cholangiocytes, is overexpressed in human CCA and presumably contributes to tumor cell growth. In this regard, IL-6 overexpression in CCA cells stimulates the expression of different methyltransferases (DNMT-1 and HASJ4442) that further downregulate the expression of seven miRNAs; one of these downregulated miRNAs, miRNA-370, directly targets the oncogene mitogen-activated protein kinase mitogen-activated protein kinase 8 in both in vitro and in tumor cell xenografts in vivo[38]. Moreover, IL-6 may also regulate the expression of other miRNAs, such as miR-148a, miRNA-152 and miRNA-301[39]. These three miRNAs are downregulated in CCA cells and possess sequence complementarity to the 3’-UTR region of the DNMT-1 mRNA transcript. However, only miRNA-148a and miRNA-152 directly regulate DNMT-1 expression. Interestingly, DNMT-1 modulates the expression of the tumor suppressor genes Rassf1a and p16INK4a by promoter hypermethylation, leading to malignant cell transformation.

Chemoresistance

CCAs are tumors with a marked multidrug resistance phenotype that includes changes in the expression of genes involved in the apoptosis/survival balance[40].

As mentioned above, miRNA-21 and miRNA-200b are highly overexpressed in malignant cholangiocytes[18]. These two miRNAs are suggested to contribute to chemoresistance in CCA by modulating the chemotherapy-induced apoptosis. Thus, overexpression of oligonucleotides anti-miR21 and anti-miR200b in malignant cholangiocytes cell lines satisfactorily increases gemcitabine-induced cytotoxicity. Moreover, miRNA-21 targets the tumor suppressor PTEN, which through its phosphatase activity inhibits phosphatidylinositol 3-kinase-dependent growth and survival. In addition, miRNA-200b targets the protein tyrosine phosphatase non-receptor type 12 that is involved in cell growth, cell dedifferentiation and oncogenic transformation.

In contrast to the aforementioned data obtained using an experimental animal model of chemically induced CCA, other authors have reported that miRNA-let7a is overexpressed in human CCA[41]. Thus, miRNA-let7a overexpression is able to target the tumor suppressor neurofibromatosis 2 (NF2), which modulates the signal transducer and activator of transcription 3 (STAT3)-activated survival mechanism[41]. Interestingly, intratumor administration of oligonucleotides anti-miRNA-let7a increases NF2 and decreases p-STAT3 expression in CCA xenografts in vivo. Moreover, these effects increase gemcitabine toxicity, resulting in decreased tumor growth[41].

Circadian rhythm

The circadian rhythm controls on a 24-h range many fundamental physiological features such as behavior, metabolism or cell proliferation. The suprachiasmatic nuclei located in the hypothalamus synchronizes the molecular clocks in most mammalian cells through different circadian physiological rhythms including rest-activity, body temperature, feeding patterns, and hormonal secretions. Therefore, circadian alterations have been suggested to affect carcinogenesis risk also in humans[42]. In this regard, miRNA-141, overexpressed in intrahepatic CCA, was predicted by bioinformatics approaches to target CLOCK directly; a tumor suppressor that controls the cellular circadian rhythm[18]. Indeed, CLOCK may inhibit cell cycle, both directly and indirectly, and increases apoptosis. Administration of oligonucleotides anti-miRNA-141 in CCA cells results in CLOCK protein downregulation. These data indicate that circadian rhythm and miRNAs may interact in physiological and pathological conditions.

POLYCYSTIC LIVER DISEASES

Polycystic liver diseases (PCLDs) are genetic disorders characterized by bile-duct dilatation and/or cyst development, which become progressively more severe and require liver transplantation as the only therapeutic option[43]. The large volume of hepatic cysts causes symptoms such as abdominal distension, local pressure with back pain, gastroesophageal reflux, and dyspnea. The estimated prevalence of PCLDs is around 1:1000, and these patients often develop polycystic kidney disease. Different genetic mutations trigger the appearance and growth of cysts in PCLDs, and in some cases this phenomenon is also associated with hepatic fibrosis. Most of the proteins encoded by these genes are located in the primary cilium of cholangiocytes[44]. Many of these proteins interact with each other forming complexes that signal through common pathways. Mutations in genes encoding proteins located in the primary cilium of cholangiocytes result in physical and functional defects of this organelle, and outcome in the development of several forms of PCLD classified as ciliopathies. For this reason, and because the only cells in the liver that have cilia are cholangiocytes, the PCLDs affecting genes encoding proteins which are localized in the cilia are called cholangiociliopathies[44]. It has been recently demonstrated that hepatic cystogenesis in PCLDs is the result of hyperproliferation[45], hypersecretion[46], and alteration in the pattern of miRNAs in the bile duct cells[47], and that these alterations are intracellularly associated with an increase in the levels of cAMP and a decrease in calcium[45,48,49]. In this regard, global changes in the expression pattern of miRNAs were observed between cultured cholangiocytes from normal and PCK rats (animal model of hepatorenal polycystic disease, i.e., autosomal recessive polycystic kidney disease)[47,50]. In total, 121 and 148 miRNAs were detected in normal and PCK rat cholangiocytes, respectively. Twelve miRNAs were expressed in normal rat cholangiocytes and not in PCK rat cholangiocytes, and 39 were present only in PCK rat cholangiocytes. Moreover, there were changes in the expression of 109 common miRNAs between both cell lines. Interestingly, 97 of these common miRNAs (i.e., 87%) were found to be downregulated in PCK rat cholangiocytes compared to normal rat cholangiocytes, and 12 miRNAs (11%) were overexpressed in cystic cholangiocytes. Among those highly downregulated miRNAs in cultured PCK rat cholangiocytes, miRNA-15a is also downregulated in vivo in cholangiocytes from PCK rats and PCLD patients[47]. This miRNA-15a downregulation runs in parallel with the upregulation of its predicted target, the cell-cycle regulator cell division cycle 25A (Cdc25A). Experimental overexpression of miRNA-15a in PCK rat cholangiocytes results in decreased Cdc25A protein levels, inhibition of cell proliferation, and cyst growth reduction. Interestingly, experimental suppression of miRNA-15a in normal rat cholangiocytes accelerates cell proliferation, increases Cdc25A expression, and promotes cyst growth[47]. All these data suggest that the miRNA-15a/Cdc25A interplay could be a potential target to inhibit hepatic cystogenesis (Figure 1). Importantly, inhibition of Cdc25A with its inhibitor vitamin K3 suppresses hepatorenal cystogenesis in rodent models of polycystic kidney and liver disease, being a valuable potential pharmacological approach to test in clinical trials[51] (Figure 1). Moreover, it will be interesting to demonstrate in future studies the role of many other altered miRNAs in the pathogenesis of PCLDs.

Figure 1
Figure 1 miRNA-15a/Cdc25a interplay participates in hepatic cystogenesis of polycystic liver diseases. miRNA-15a directly targets the cell cycle regulator Cdc25a. In polycystic rat and human cholangiocytes, miR15a is downregulated, thus resulting in Cdc25a upregulation, cell proliferation and cystogenesis. The Cdc25a inhibitor vitamin K3 was demonstrated to target Cdc25a, blocking the hepatorenal cystogenesis of several rodent models of polycystic kidney and liver disease. This suggests the therapeutic potential of vitamin K3 for polycystic kidney and liver diseases.
PRIMARY BILIARY CIRRHOSIS

Primary biliary cirrhosis (PBC) is a chronic cholestatic liver disease that mainly affects middle-aged women[52]. It is a rare disease, with an estimated prevalence of 1:8000 in countries such as the United Kingdom. Four stages of this disorder have been characterized by progressive bile-duct loss that runs in parallel with an increase in cholestasis and fibrosis[52]. The etiology of PBC remains unknown, and the pathogenesis is poorly understood. The serological hallmark of PBC is the development of high titers of antimitochondrial autoantibodies (AMAs) in serum associated with an immunological attack on the small intrahepatic bile ducts[52]. Interestingly, PBC patients usually have a poor response to immunosuppressants and the only therapeutic option that improves the outcome of the disease in two thirds of PBC patients is daily administration of ursodeoxycholic acid (UDCA), a hypercholeretic bile acid[52]. In addition, and importantly, PBC patients show alterations in the biliary secretion of bicarbonate. Thus, PBC patients show a failure in the secretin-stimulated biliary secretion of bicarbonate[53], as well as a reduction in the hepatobiliary expression of anion exchanger 2 (AE2/SLC4A2)[54,55]. AE2 is a Cl-/HCO3- exchanger located in the canalicular membrane of hepatocytes and in the apical membrane of cholangiocytes that controls the intracellular pH (pHi) and promotes the alkalinization and fluidization of bile[1,2,56]. An interesting hypothesis suggests that, in PBC patients, long-term maintained alteration of pHi by AE2 downregulation might modify cholangiocyte and lymphocyte function[57]. To test the etiopathogenic role of AE2 downregulation in PBC, an Ae2a,b1,b2-/-mouse knockout was generated. Interestingly, these animals spontaneously reproduce over time many PBC features[57], such as: (1) portal infiltration of T lymphocytes and bile duct damage; (2) increased oxidative stress in cholangiocytes; (3) elevated production of interferon-γ and IL-12; (4) periductular hepatic fibrosis; (5) increased levels of IgM, IgG, and hepatic alkaline phosphatase; and (6) the presence of PBC-specific AMAs. Different agents such as hormones, metabolic influences, infectious agents, xenobiotics, cytokines/inflammatory mediators, and/or miRNAs could be involved in the AE2 downregulation present in PBC patients. Regarding miRNAs, a differentially expressed miRNA profile in livers from PBC patients compared with normal controls has been recently reported[58]. The change consists of 35 differentially expressed miRNAs (11 upregulated and 24 downregulated), which were predicted to target genetic transcripts involved in cell proliferation, apoptosis, inflammation, oxidative stress and metabolism[58]. Regarding those miRNAs that were overexpressed in the miRNA array, we observed by using bioinformatics approaches that miRNA-506 could be a potential direct AE2 regulator. Therefore, we tested if AE2 downregulation in PBC cholangiocytes is dependent on miRNAs, and specifically on miRNA-506. Our data showed that miR-506 is upregulated in cholangiocytes from PBC patients, binds to the 3’-UTR of AE2 mRNA, and prevents protein translation, leading to diminished AE2 activity and impaired biliary secretory functions[59]. These data suggest the ethiopathogenic role of miR-506 in the AE2 downregulation characteristic of PBC cholangiocytes (Figure 2). On the other hand, the reported AE2 downregulation in PBC lymphocytes was not associated with changes in the expression of miRNA-506[59]. This disease preferentially affects women, therefore, it is interesting to remark that the gene encoding miRNA-506 is located in the X chromosome. Further investigations are needed to characterize fully the role of miRNA-506 in the regulation of other targets and cellular features, as well as the analysis of its expression in other tissues. Moreover, the role of UDCA and estrogens on miRNA-506 expression will be valuable information to expand the ethiopathogenic role of this miRNA in PBC.

Figure 2
Figure 2 Role of miRNA-506 in the etiopathogenesis of primary biliary cirrhosis. Different risk factors could induce genetic alterations leading to anion exchange 2 (AE2) downregulation in primary biliary cirrhosis (PBC) cholangiocytes. miRNA-506 is overexpressed in PBC cholangiocytes, thus resulting in the inhibition of both AE2 protein translation and subsequent Cl-/HCO3- activity, and impairing the biliary secretory functions.
CONCLUSION

In summary, the available information clearly indicates that miRNAs represent a new research area in the field of biliary pathophysiology. As stated in this review, they may modify different features in cholangiocytes, such as secretion, apoptosis, proliferation, and migration, and can be regulated by different mechanisms and conditions, such as epigenetics, hypoxia, or circadian rhythms. There is still limited information about the role of miRNAs in biliary diseases, but future investigations will provide more evidence regarding their role in the development and progression of cholangiopathies, as well as in their potential use for diagnosis and therapy. Finally, it is important to remark upon the increasing information about the role of stem cells in the pathophysiology of biliary diseases[60]; in this regard, the potential role of miRNAs in the regulation of biliary stem cells during cholangiocyte injury, as well as the role of miRNAs in the development of cancer stem cells in CCA[61] need to be elucidated.

Footnotes

Peer reviewers: Richard A Kozarek, MD, Executive Director, Digestive Disease Institute, Virginia Mason Medical Center 1100 Ninth Avenue, PO Box 900, Seattle, VA 98111-0900, United States; Dr. Matthias Ocker, MD, Professor, Director, Institute for Surgical Research, Philipps-University Marburg, Baldingerstrasse, 35033 Marburg, Germany

S- Editor Shi ZF L- Editor Kerr C E- Editor Xiong L

References
1.  Banales JM, Prieto J, Medina JF. Cholangiocyte anion exchange and biliary bicarbonate excretion. World J Gastroenterol. 2006;12:3496-3511.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Banales JM, Arenas F, Rodríguez-Ortigosa CM, Sáez E, Uriarte I, Doctor RB, Prieto J, Medina JF. Bicarbonate-rich choleresis induced by secretin in normal rat is taurocholate-dependent and involves AE2 anion exchanger. Hepatology. 2006;43:266-275.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Rodríguez-Ortigosa CM, Banales JM, Olivas I, Uriarte I, Marín JJ, Corrales FJ, Medina JF, Prieto J. Biliary secretion of S-nitrosoglutathione is involved in the hypercholeresis induced by ursodeoxycholic acid in the normal rat. Hepatology. 2010;52:667-677.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Úriz M, Sáez E, Prieto J, Medina JF, Banales JM. Ursodeoxycholic acid is conjugated with taurine to promote secretin-stimulated biliary hydrocholeresis in the normal rat. PLoS One. 2011;6:e28717.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Masyuk AI, Masyuk TV, LaRusso NF. Cholangiocyte primary cilia in liver health and disease. Dev Dyn. 2008;237:2007-2012.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Masyuk AI, Masyuk TV, Splinter PL, Huang BQ, Stroope AJ, LaRusso NF. Cholangiocyte cilia detect changes in luminal fluid flow and transmit them into intracellular Ca2+ and cAMP signaling. Gastroenterology. 2006;131:911-920.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Masyuk AI, Gradilone SA, Banales JM, Huang BQ, Masyuk TV, Lee SO, Splinter PL, Stroope AJ, Larusso NF. Cholangiocyte primary cilia are chemosensory organelles that detect biliary nucleotides via P2Y12 purinergic receptors. Am J Physiol Gastrointest Liver Physiol. 2008;295:G725-G734.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Masyuk AI, Huang BQ, Ward CJ, Gradilone SA, Banales JM, Masyuk TV, Radtke B, Splinter PL, LaRusso NF. Biliary exosomes influence cholangiocyte regulatory mechanisms and proliferation through interaction with primary cilia. Am J Physiol Gastrointest Liver Physiol. 2010;299:G990-G999.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Gradilone SA, Masyuk AI, Splinter PL, Banales JM, Huang BQ, Tietz PS, Masyuk TV, Larusso NF. Cholangiocyte cilia express TRPV4 and detect changes in luminal tonicity inducing bicarbonate secretion. Proc Natl Acad Sci U S A. 2007;104:19138-19143.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Marzioni M, Saccomanno S, Candelaresi C, Rychlicki C, Agostinelli L, Trozzi L, De Minicis S, Benedetti A. Clinical implications of novel aspects of biliary pathophysiology. Dig Liver Dis. 2010;42:238-244.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215-233.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Garzon R, Marcucci G, Croce CM. Targeting microRNAs in cancer: rationale, strategies and challenges. Nat Rev Drug Discov. 2010;9:775-789.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Carthew RW, Sontheimer EJ. Origins and Mechanisms of miRNAs and siRNAs. Cell. 2009;136:642-655.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Iorio MV, Croce CM. microRNA involvement in human cancer. Carcinogenesis. 2012;33:1126-1133.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Blechacz B, Gores GJ. Cholangiocarcinoma: advances in pathogenesis, diagnosis, and treatment. Hepatology. 2008;48:308-321.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Blechacz B, Komuta M, Roskams T, Gores GJ. Clinical diagnosis and staging of cholangiocarcinoma. Nat Rev Gastroenterol Hepatol. 2011;8:512-522.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Chen L, Yan HX, Yang W, Hu L, Yu LX, Liu Q, Li L, Huang DD, Ding J, Shen F. The role of microRNA expression pattern in human intrahepatic cholangiocarcinoma. J Hepatol. 2009;50:358-369.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Meng F, Henson R, Lang M, Wehbe H, Maheshwari S, Mendell JT, Jiang J, Schmittgen TD, Patel T. Involvement of human micro-RNA in growth and response to chemotherapy in human cholangiocarcinoma cell lines. Gastroenterology. 2006;130:2113-2129.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Olaru AV, Ghiaur G, Yamanaka S, Luvsanjav D, An F, Popescu I, Alexandrescu S, Allen S, Pawlik TM, Torbenson M. MicroRNA down-regulated in human cholangiocarcinoma control cell cycle through multiple targets involved in the G1/S checkpoint. Hepatology. 2011;54:2089-2098.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Selaru FM, Olaru AV, Kan T, David S, Cheng Y, Mori Y, Yang J, Paun B, Jin Z, Agarwal R. MicroRNA-21 is overexpressed in human cholangiocarcinoma and regulates programmed cell death 4 and tissue inhibitor of metalloproteinase 3. Hepatology. 2009;49:1595-1601.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Stutes M, Tran S, DeMorrow S. Genetic and epigenetic changes associated with cholangiocarcinoma: from DNA methylation to microRNAs. World J Gastroenterol. 2007;13:6465-6469.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Zhong XY, Yu JH, Zhang WG, Wang ZD, Dong Q, Tai S, Cui YF, Li H. MicroRNA-421 functions as an oncogenic miRNA in biliary tract cancer through down-regulating farnesoid X receptor expression. Gene. 2012;493:44-51.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Kemper JK. Regulation of FXR transcriptional activity in health and disease: Emerging roles of FXR cofactors and post-translational modifications. Biochim Biophys Acta. 2011;1812:842-850.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Teodoro JS, Rolo AP, Palmeira CM. Hepatic FXR: key regulator of whole-body energy metabolism. Trends Endocrinol Metab. 2011;22:458-466.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  He Q, Cai L, Shuai L, Li D, Wang C, Liu Y, Li X, Li Z, Wang S. Ars2 is overexpressed in human cholangiocarcinomas and its depletion increases PTEN and PDCD4 by decreasing MicroRNA-21. Mol Carcinog. 2011;Dec 28 [Epub ahead of print].  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Zhang J, Han C, Wu T. MicroRNA-26a promotes cholangiocarcinoma growth by activating β-catenin. Gastroenterology. 2012;143:246-56.e8.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Yang H, Li TW, Peng J, Tang X, Ko KS, Xia M, Aller MA. A mouse model of cholestasis-associated cholangiocarcinoma and transcription factors involved in progression. Gastroenterology. 2011;141:378-88, 388.e1-4.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Piskounova E, Polytarchou C, Thornton JE, LaPierre RJ, Pothoulakis C, Hagan JP, Iliopoulos D, Gregory RI. Lin28A and Lin28B inhibit let-7 microRNA biogenesis by distinct mechanisms. Cell. 2011;147:1066-1079.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Razumilava N, Bronk SF, Smoot RL, Fingas CD, Werneburg NW, Roberts LR, Mott JL. miR-25 targets TNF-related apoptosis inducing ligand (TRAIL) death receptor-4 and promotes apoptosis resistance in cholangiocarcinoma. Hepatology. 2012;55:465-475.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Mott JL, Kobayashi S, Bronk SF, Gores GJ. mir-29 regulates Mcl-1 protein expression and apoptosis. Oncogene. 2007;26:6133-6140.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Huang L, Frampton G, Liang LJ, Demorrow S. Aberrant DNA methylation profile in cholangiocarcinoma. World J Gastrointest Pathophysiol. 2010;1:23-29.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Mullen PD, Carbonari JP, Glenday MC. Identifying pregnant women who drink alcoholic beverages. Am J Obstet Gynecol. 1991;165:1429-1430.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Sarkies P, Sale JE. Cellular epigenetic stability and cancer. Trends Genet. 2012;28:118-127.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Hoffman AM, Cairns P. Epigenetics of kidney cancer and bladder cancer. Epigenomics. 2011;3:19-34.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Wiklund ED, Kjems J, Clark SJ. Epigenetic architecture and miRNA: reciprocal regulators. Epigenomics. 2010;2:823-840.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Chen Y, Gao W, Luo J, Tian R, Sun H, Zou S. Methyl-CpG binding protein MBD2 is implicated in methylation-mediated suppression of miR-373 in hilar cholangiocarcinoma. Oncol Rep. 2011;25:443-451.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Chen Y, Luo J, Tian R, Sun H, Zou S. miR-373 negatively regulates methyl-CpG-binding domain protein 2 (MBD2) in hilar cholangiocarcinoma. Dig Dis Sci. 2011;56:1693-1701.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Meng F, Wehbe-Janek H, Henson R, Smith H, Patel T. Epigenetic regulation of microRNA-370 by interleukin-6 in malignant human cholangiocytes. Oncogene. 2008;27:378-386.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Braconi C, Huang N, Patel T. MicroRNA-dependent regulation of DNA methyltransferase-1 and tumor suppressor gene expression by interleukin-6 in human malignant cholangiocytes. Hepatology. 2010;51:881-890.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Martinez-Becerra P, Vaquero J, Romero MR, Lozano E, Anadon C, Macias RI, Serrano MA, Grañé-Boladeras N, Muñoz-Bellvis L, Alvarez L. No correlation between the expression of FXR and genes involved in multidrug resistance phenotype of primary liver tumors. Mol Pharm. 2012;9:1693-1704.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Meng F, Henson R, Wehbe-Janek H, Smith H, Ueno Y, Patel T. The MicroRNA let-7a modulates interleukin-6-dependent STAT-3 survival signaling in malignant human cholangiocytes. J Biol Chem. 2007;282:8256-8264.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Khapre RV, Samsa WE, Kondratov RV. Circadian regulation of cell cycle: Molecular connections between aging and the circadian clock. Ann Med. 2010;42:404-415.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Drenth JP, Chrispijn M, Nagorney DM, Kamath PS, Torres VE. Medical and surgical treatment options for polycystic liver disease. Hepatology. 2010;52:2223-2230.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Masyuk T, Masyuk A, LaRusso N. Cholangiociliopathies: genetics, molecular mechanisms and potential therapies. Curr Opin Gastroenterol. 2009;25:265-271.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Banales JM, Masyuk TV, Gradilone SA, Masyuk AI, Medina JF, LaRusso NF. The cAMP effectors Epac and protein kinase a (PKA) are involved in the hepatic cystogenesis of an animal model of autosomal recessive polycystic kidney disease (ARPKD). Hepatology. 2009;49:160-174.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Banales JM, Masyuk TV, Bogert PS, Huang BQ, Gradilone SA, Lee SO, Stroope AJ, Masyuk AI, Medina JF, LaRusso NF. Hepatic cystogenesis is associated with abnormal expression and location of ion transporters and water channels in an animal model of autosomal recessive polycystic kidney disease. Am J Pathol. 2008;173:1637-1646.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Lee SO, Masyuk T, Splinter P, Banales JM, Masyuk A, Stroope A, Larusso N. MicroRNA15a modulates expression of the cell-cycle regulator Cdc25A and affects hepatic cystogenesis in a rat model of polycystic kidney disease. J Clin Invest. 2008;118:3714-3724.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Masyuk TV, Masyuk AI, Torres VE, Harris PC, Larusso NF. Octreotide inhibits hepatic cystogenesis in a rodent model of polycystic liver disease by reducing cholangiocyte adenosine 3',5'-cyclic monophosphate. Gastroenterology. 2007;132:1104-1116.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Gradilone SA, Masyuk TV, Huang BQ, Banales JM, Lehmann GL, Radtke BN, Stroope A, Masyuk AI, Splinter PL, LaRusso NF. Activation of Trpv4 reduces the hyperproliferative phenotype of cystic cholangiocytes from an animal model of ARPKD. Gastroenterology. 2010;139:304-14.e2.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Masyuk T, Masyuk A, LaRusso N. MicroRNAs in cholangiociliopathies. Cell Cycle. 2009;8:1324-1328.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Masyuk TV, Radtke BN, Stroope AJ, Banales JM, Masyuk AI, Gradilone SA, Gajdos GB, Chandok N, Bakeberg JL, Ward CJ. Inhibition of Cdc25A suppresses hepato-renal cystogenesis in rodent models of polycystic kidney and liver disease. Gastroenterology. 2012;142:622-633.e4.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Poupon R. Primary biliary cirrhosis: a 2010 update. J Hepatol. 2010;52:745-758.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Prieto J, García N, Martí-Climent JM, Peñuelas I, Richter JA, Medina JF. Assessment of biliary bicarbonate secretion in humans by positron emission tomography. Gastroenterology. 1999;117:167-172.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  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: ]
55.  Melero S, Spirlì C, Zsembery A, Medina JF, Joplin RE, Duner E, Zuin M, Neuberger JM, Prieto J, Strazzabosco M. Defective regulation of cholangiocyte Cl-/HCO3(-) and Na+/H+ exchanger activities in primary biliary cirrhosis. Hepatology. 2002;35:1513-1521.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Aranda V, Martínez I, Melero S, Lecanda J, Banales JM, Prieto J, Medina JF. Shared apical sorting of anion exchanger isoforms AE2a, AE2b1, and AE2b2 in primary hepatocytes. Biochem Biophys Res Commun. 2004;319:1040-1046.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Salas JT, Banales JM, Sarvide S, Recalde S, Ferrer A, Uriarte I, Oude Elferink RP, Prieto J, Medina JF. Ae2a,b-deficient mice develop antimitochondrial antibodies and other features resembling primary biliary cirrhosis. Gastroenterology. 2008;134:1482-1493.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Padgett KA, Lan RY, Leung PC, Lleo A, Dawson K, Pfeiff J, Mao TK, Coppel RL, Ansari AA, Gershwin ME. Primary biliary cirrhosis is associated with altered hepatic microRNA expression. J Autoimmun. 2009;32:246-253.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Banales JM, Sáez E, Uriz M, Sarvide S, Urribarri AD, Splinter P, Tietz Bogert PS, Bujanda L, Prieto J, Medina JF. Up-regulation of microRNA 506 leads to decreased Cl-/HCO3- anion exchanger 2 expression in biliary epithelium of patients with primary biliary cirrhosis. Hepatology. 2012;56:687-697.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Cardinale V, Wang Y, Carpino G, Mendel G, Alpini G, Gaudio E, Reid LM, Alvaro D. The biliary tree--a reservoir of multipotent stem cells. Nat Rev Gastroenterol Hepatol. 2012;9:231-240.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Cardinale V, Carpino G, Reid L, Gaudio E, Alvaro D. Multiple cells of origin in cholangiocarcinoma underlie biological, epidemiological and clinical heterogeneity. World J Gastrointest Oncol. 2012;4:94-102.  [PubMed]  [DOI]  [Cited in This Article: ]