Topic Highlight Open Access
Copyright ©2010 Baishideng. All rights reserved.
World J Gastrointest Pathophysiol. Jun 15, 2010; 1(2): 38-49
Published online Jun 15, 2010. doi: 10.4291/wjgp.v1.i2.38
Histamine regulation of hyperplastic and neoplastic cell growth in cholangiocytes
Paolo Onori, Experimental Medicine, University of L'Aquila, L'Aquila 67100, Italy
Eugenio Gaudio, Antonio Franchitto, Department of Human Anatomy, University of Rome “La Sapienza”, Rome 00161, Italy
Gianfranco Alpini, Research, Central Texas Veterans Health Care System, TX 76504, United States
Gianfranco Alpini, Heather Francis, Scott and White Digestive Disease Research Center, Scott and White, Temple, TX 76504, United States
Gianfranco Alpini, Heather Francis, Division of Gastroenterology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX 76504, United States
Heather Francis, Division of Research and Education, Scott and White, Temple, TX 76504, United States
Author contributions: Onori P and Gaudio E performed literature searches, contributed conceptually to the writing and reviewing of the article; Franchitto A contributed to the production of technical data and reviewed the article; Alpini G contributed conceptually to the article design and reviewed the article; and Francis H performed literature searches, contributed conceptually to the article design, wrote, reviewed and edited the paper.
Correspondence to: Heather Francis, BS, Graduate Student and Research Associate, Texas A&M College of Medicine and Scott and White Hospital 702 SW H.K. Dodgen Loop, Temple, TX 76504, United States. hfrancis@tamu.edu
Telephone: +1-254-7427055
Received: February 9, 2010
Revised: April 3, 2010
Accepted: April 10, 2010
Published online: June 15, 2010

Abstract

Histamine has long been known to be involved in inflammatory events. The discovery of antihistamines dates back to the first half of the 20th century when a Swiss-Italian pharmacologist, Daniel Bovet began his work. In 1957 he was awarded a Nobel Prize for his production of antihistamines for allergy relief. Since that time, histamine has been found to play a role in other events besides allergic reaction. Possibly unbelievable to Bovet and his peers, histamine has now been marked as playing a role in liver pathologies including hepatobiliary diseases.

Key Words: Histamine, Receptors, Cholangiocytes, Cholestasis, Cholangiocarcinoma



INTRODUCTION

The liver is a dynamic organ that is able to repair and regenerate itself after injury. Numerous resident liver cell types are involved in maintaining a homeostatic state of the liver as well as playing a role during liver injury and disease. Cholangiocytes play a critical role in the regulation and overall function of the liver. These cells are hormone-responsive and much effort has been put forth to delineate the mechanisms behind cholangiocyte function in liver disease. An emerging role for histamine and liver diseases has begun to be investigated.

In this review we will begin with a brief overview of the intrahepatic biliary tree, cholangiocyte function, hepatic vasculature, cholangiocarcinoma and histamine. After, we will discuss the latest findings regarding histaminergic activation in the liver as well as the most recent findings in regards to cholangiocyte regulation by histamine and histamine receptors. Finally, we will conclude with a section on histamine and cholangiocarcinoma and our speculation for the future of histamine in the progression of liver diseases.

INTRAHEPATIC BILIARY TREE

The biliary tree is the common term used for the pathway where bile is secreted from the liver that begins with initiation of bile from the hepatocytes[1]. It starts with numerous small branches that terminate in the common bile duct, also known as the trunk of the biliary tree[2]. It is here that the common duct comes together with branches of the hepatic artery and portal vein forming the central axis of the portal triad[2]. Lining the intrahepatic biliary tree are epithelial cells known as cholangiocytes[1,3,4]. Once defined as “simple” epithelia, cholangiocytes are now regarded as key players in liver pathophysiology[3-7]. Cholangiocytes are responsible for the modification and release of bile from the liver and the transport of bile acids[6,8-12]. These cells are hormone-responsive, and express a multitude of hormone receptor binding sites that enable them to interact with mediators to induce varying effects on liver pathology[1,6,13-18]. These cells, that are cuboidal by nature, line the three-dimensional network of interconnecting ducts[1]. Cholangiocytes are a heterogeneous population of cells[1-4,16,17,19,20] that are derived from small intrahepatic bile ducts (lined by small cholangiocytes)[3,20,21] and large intrahepatic bile ducts (lined by large cholangiocytes)[3,20,21].

Cholangiocyte proliferation

Heterogeneous cholangiocytes have been identified as the target cells for a number of liver diseases (cholangiopathies) that include primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC)[1]. In these diseases there is a destruction of the bile ducts that leads to hyperplastic transformation, termed “typical” or “atypical” proliferation[7,14,22]. “Typical” cholangiocyte proliferation is characterized by an increase in cholangiocyte proliferation that is limited to the portal area, whereas “atypical”[23] is seen in patients with prolonged cholangiopathies (i.e. PBC and PSC) and is noted by irregular proliferation of cholangiocytes (with a well structured lumen) that extend into periportal and parenchymal regions with occasional anastomosing cords with adjacent hepatocytes[7,14,22]. “Oval” cell proliferation is a pre-malignant stage that is seen in early carcinogenesis and characterized by the presence of a disorganized tubular structure without any well-defined lumen that can transform into a full-blown neoplastic condition known as cholangiocarcinoma (CCA)[7,14,22,24].

Models of proliferation

Several animal models of “typical” proliferation have rapidly become an excellent tool for the dissection of the mechanisms behind this event. Bile duct ligation (BDL) is commonly used to mimic “typical” proliferation[19,25-27]. Ligation of the bile duct induces a build-up of bile and increased biliary pressure[8,28] that induce an increase in cholangiocyte proliferation (mostly of large cholangiocytes)[19] and resembles biliary hyperplasia seen in patients with diseases like PBC[7,19,29]. Other models used to mimic “typical” proliferation include 70% partial hepatectomy (PH), acute carbon tetrachloride (CCl4) treatment and chronic feeding of α-naphthylisothiocyanate (ANIT) or bile salts[7,12,14,16,17,30]. All of these models are found to be associated with an upregulation of secretin receptor (SR) gene expression and secretin-stimulated cAMP levels and ductal secretory activity[7,13,14,16,17,30,31]. Chronic treatment of normal rats with forskolin has also been shown to increase biliary hyperplasia (similar to BDL) and secretin-induced responses in cAMP levels as well as downstream signaling components[32]. While BDL targets mainly the large cholangiocytes[3,29], after CCl4 treatment, only small cholangiocytes respond and de novo proliferate[16,17]. This occurs with a parallel loss of large ductal mass[16,17]. After 70% PH, small and large cholangiocytes proliferate during liver regeneration[14,24]. These tools are useful in evaluating the mechanisms of cholangiocyte proliferation during liver cholestasis and disease progression.

CHOLANGIOCYTE FUNCTION

Using the tools described above has enabled researchers to uncover important information regarding cholangiocyte function in relation to secretion, proliferation and apoptosis during liver disease[1,4,6,7,13,18].

Secretory function

In BDL rats, there is increased cholangiocyte proliferation coupled with increased bile flow and bicarbonate secretion after stimulation with secretin[3,8,13,19,25,27,33]. This is an excellent tool to use to evaluate functionality of cholangiocytes in normal and diseased states as well as in response to stimulation by numerous factors. Over the years it has been shown that, in addition to secretin, vasoactive intestinal peptide increases bicarbonate secretion, whereas the hormones somatostatin, insulin and gastrin all decrease bile flow and bicarbonate secretion[33-36]. Other factors have been shown to inhibit the secretin-induced increased cholangiocyte response. These include the alpha 2 adrenergic receptor agonist UK,14304[37] and the phenolic compound, caffeic phenethyl ester (CAPE)[38]. The majority of these responses occur through a cAMP-dependent pathway, however other studies have shown that Ca2+ can also play a role in cholangiocyte secretion. Le Sage, et al[15] have demonstrated that the alpha-1 adrenergic receptor agonist, phenylephrine, increases secretin-stimulated choleresis through activation of Ca2+/protein kinase c (PKC) mechanisms, whereas the hormone gastrin was found to decrease secretin-stimulated ductal secretion in BDL rats via increased expression of Ca2+-dependent PKC isoforms[27].

Proliferative and apoptotic functions

Cholangiocyte proliferation and apoptosis is affected by many hormones and peptides[13]. Recently, Mancinelli et al[31] have shown that the sex hormone, follicle-stimulating hormone (FSH) increases cholangiocyte proliferation through cAMP/PKA-dependent phosphorylation of extracellular signal-regulated kinase (ERK1/2) and Elk-1, a member of the ETS oncogene family. The forkhead box proteins A1 and A1 (Foxa1 and Foxa2) have been descried as “terminators” of bile duct expansion by their ability to inhibit interleukin-6 (IL-6) expression[39]. In BDL Foxl1 (-/-) knockout mice it was found that loss of the winged helix transcription factor Foxl1 induced a decrease in cholangiocyte proliferation and loss of bile ductular mass by activation of the canonical Wnt/beta-catenin pathway, suggesting that this transcription factor plays a critical role in cholangiocyte proliferation during cholestasis[40]. After treating BDL rats with CAPE, Mancinelli et al[38] demonstrated a large decrease in cholangiocyte proliferation coupled with increased cholangiocyte apoptosis that was reversed after chronic feeding with the bile acid taurocholic acid (TC). TC feeding also led to recuperation in the expression of VEGF proteins and receptors (R2 and R3), implicating bile acids and angiogenic factors in the course of cholangiocyte cholestasis[38]. The endocannabinoid, anandamide (AEA) was found to inhibit cholangiocyte proliferation and increase apoptosis through activation of thioredoxin 1/redox factor 1 and activator protein-1 (AP-1), demonstrating the potential role for endocannabinoids in cholestasis[41]. Genetic knockdown of the transcription factors c-Fos and c-Jun ablated the AEA-induced cholangiocyte death[41]. Tumor necrosis factor-alpha (TNF-α) has been shown to induce cholangiocyte apoptosis and decrease proliferation when coupled with a single injection of actinomycin D[42]. Chronic feeding with TC has been found to reverse this ductopenic effect via a phosphatidylinositol-3-kinase (PI3K)-dependent pathway[43]. These studies have provided strong evidence that cholangiocyte function is susceptible to inhibition and stimulation by numerous factors.

Hepatic vasculature

It is pertinent to discuss hepatic vasculature within this article as the microanatomy (including cholangiocytes) of the liver is significantly influenced by blood flow. There is a constant flow of large amounts of blood to and from the liver that is controlled through two separate blood supplies: the portal vein (PV) and the hepatic artery (HA)[44,45]. There are conflicting views regarding HA distribution and where this artery terminates in the liver parenchyma[46-48]. Recently, a detailed study using scanning electron microscopy of vascular corrosion casts demonstrated that the terminal HA branches do not end directly in the sinusoidal beds[49,50]. The terminal HA give rise to capillaries that form the peribiliary plexus, periportal plexus and single capillaries of the portal space; this implies that only venous blood at a lowered pressure reaches the sinusoids and vena portal branches[49,50].

Structurally, the sinusoids are vascular channels lined with fenestrated endothelial cells (sinusoidal endothelial cells or SEC[51]). It is this fenestrated structure that makes the sinusoids very “leaky”, allowing fluid passage to occur[52]. Hepatocytes are found around the sinusoids and are responsible for the initiation, formation and secretion of bile[53]. After secretion of bile by the hepatocytes, bile is modified by cholangiocytes[53]. The hepatic artery is the main supplier of blood to the biliary tract and cholangiocytes within the structure termed peribiliary vascular plexus (PBP)[26,50]. This system has been found to be a source rich in vascular growth factors and other vasoactive substances[50]. Recent work has shown that the PBP has a great influence on cholangiocyte proliferation and/or loss that is mediated by VEGF expression[26]. The bile acid TC has also been shown to influence cholangiocyte growth/loss by regulating VEGF[38]. Please refer to Figure 1 for a graphic representation and diagram of the microcirculation in the liver with regards to blood and bile flow.

Figure 1
Figure 1 Graphic image of blood flow through the liver and the key cells involved.
Effects of blood flow on liver microanatomy

Significant findings have shown that blood flow in the liver directly influences its microanatomy and how it responds to stress and/or disease. In rats with BDL-induced cholestasis, interruption of the hepatic artery blood supply via hepatic artery ligation (HAL) causes a significant loss of the PBP and increases cholangiocyte apoptosis[26]. Ischemic bile duct injury can occur in association with hepatic artery thrombosis during liver transplantation and can induce the development of biliary casts, bile duct death or chronic disease that mimics PSC[54].

CHOLANGIOCARCINOMA
Description of progression and treatment options

Cholangiocyte hyperplasia that becomes uncontrollable or differentiates into a further neoplastic state can lead to cholangiocarcinoma. Cholangiocarcinoma is defined as a tumor from either intrahepatic or extrahepatic origin[55]. Prominent risk factors of CCA are chronic inflammation, congenital abnormalities of the biliary tree and genetic predisposition[56]. Tumors of CCA progress in a slow and undramatic manner with biliary sepsis, malnutrition and liver failure being typical causes of death associated with this disease[57]. CCA is the second most common type of cancer in the liver after hepatocellular carcinoma and the number of incidences of this disease is on the rise worldwide[55]. Early discovery of cholangiocarcinoma is difficult, resulting in limited treatment options. Long-term survival is only reachable by complete surgical resection of the tumor, which is not feasible for some patients and can be highly unsuccessful. Conventional chemotherapy and radiation have not proven to be successful at prolonging survival rates[55,57]. Photodynamic therapy has recently appeared as a possible treatment option to relieve pain and increase survival, however further studies are needed[56]. A most recent review of the latest advances in CCA diagnosis, treatment and patient care can be found in the reference by Aljiffry, et al[56]. Given the current lack of satisfactory treatments, understanding the cellular mechanisms behind the development of CCA will be critical in the development of future curative therapies.

Recent findings

Research to investigate CCA development and mechanism of action has increased in the last decade. A glimpse into the literature reveals thousands of entries including both reviews and original articles. Highlights of some of the latest findings from 2009 are shown here. Most recently, Blechacz et al[58] have shown that Sorafenib, an approved treatment for primary renal cancer, inhibits CCA growth in vitro and in vivo by sensitizing tumor cells to apoptosis. Blocking the production and secretion of dopamine, over-produced in CCA, has also been shown to decrease CCA growth in both cultured cells and an in vivo model of CCA[59]. In liver fluke-associated CCA, inhibition of galectin-3 was found to stimulate apoptosis that was increased by 10 fold by treatment with cisplatin or 5-fluorouracil[60]. Using CCA cell lines, Okada et al[61] demonstrated that rapamycin decreased cell proliferation that was synergistic with treatment with gemcitabine. Suppression of the nuclear factor kappa beta (NF-κB) pathway by treatment with CAPE was found to decrease cholangiocarcinoma growth both in culture and in vivo by increasing apoptosis[62]. Endothelin (ET-1) has been shown in both in vitro and in vivo models to decrease CCA growth by inhibiting VEGF and its receptors[63]. Coupled with decreased angiogenic factors, ET-1 also increased apoptosis and collagen tissue deposition[63]. Tamoxifen, the estrogen receptor antagonist, is showing promise as a possible therapeutic agent in CCA through calmodulin targets AKT [protein kinase B (PKB)] and c-FLIP (cellular-FLICE inhibitory protein)[64]. These studies are just the pinnacle of the multitude of studies that have been performed to investigate potential therapies for this devastating tumor.

HISTAMINE

In 1910 the British scientist, Sir Henry Dale identified histamine as a substance that is released and acts as a “mediator” during an allergic responses. Today we know that histamine is involved in many bodily processes in addition to allergic reaction.

Histamine activation and function

This biogenic amine interacts with four G-protein coupled receptors (GPCRs), H1HR, H2HR, H3HR and H4HR[65-68] that exert their actions on various G-proteins[69]. It has been shown that stimulation of the H1 histamine receptor (HR) activates Gαq, inducing a Ca2+-dependent effect in various cell systems[66,70,71]. In contrast, the H2HR appears to signal mainly through Gαs, stimulating a cAMP- dependent action in a variety of cell types[65,71,72]. Coupling of H3 and H4 HR has been linked to Gαi, inducing a negative regulation of cAMP-dependent signaling[65,73] as well as Gαo, mediating cellular regulation through a phospholipase C/Ca2+-dependent pathway[25,73]. Histamine receptors are also able to induce both inhibitory and stimulatory effects in different cell types. In Leydig cells, the H1HR induces an inhibitory effect, whereas the H2HR has a stimulatory effect on steroidogenesis[74]. In cholangiocytes and cholangiopathies, it has been shown that the H1HR stimulates growth, whereas the H3HR inhibits hyperplastic proliferation[65,66]. Histamine and the histamine receptors have been shown to induce a multitude of effects on various cellular pathologies including the H3HR in Alzheimer’s Disease[75]; histamine and the H1HR in vascular disease[76]; the H4HR in treatment of chronic pruritus disease[77] and many more. There is not yet a proven direct link between histamine and cancer, however growing evidence suggests that histamine and the histamine receptors may be involved in tumor growth and/or depletion[25,78]. Further, dysregulation of the enzymes that are responsible for histamine synthesis, histidine decarboxylase (HDC) and monoamine oxidase-B (MAO-B), have been implicated in certain cancers[79]. The interaction between cells and the four histamine receptors also complicates our understanding of the regulation of liver function in both normal and diseased (including cancer) states.

Histamine and liver disease

Histamine and the histamine receptors play a role in numerous processes including disease progression in the liver. Histamine, via H2HR activation, has been shown to play a protective role in alcohol-induced liver injury by lowering liver enzymes [alanine aminotransferase (ALT) and aspartate aminotransferase (AST)] and decreasing inflammation and necrosis in ethanol treated rats[80]. In patients with cirrhotic-related hepatic encephalopathy, evaluation of brain tissue showed an upregulation of histamine and H1 receptors and decreased H3 receptor density, highlighting the importance of histamine regulation in liver pathologies[81,82]. An H2 receptor antagonist, rantidine[83] and an H3/H4 antagonist, thioperamide[84] have both been shown to suppress ischemic reperfusion liver injury[83-85]. Stimulation with dimaprit, an H2 and H4 agonist showed similar results by inducing a protective effect against ischemic reperfusion injury by decreasing cytokine release[85]. The density of H2 histamine receptors is decreased in hepatic tissues from cirrhotic patients with portal hypertension[86]. Furthermore, in H1HR and H2HR knockout mice it was found that the protective effects of histamine on lipopolysaccharide (LPS)-induced liver injury was partially or completely blocked, suggesting that histamine and its receptors have a protective role in endotoxin-induced hepatic injury[87]. It has also been found that histamine levels in plasma from patients with both PSC and PBC are increased compared to healthy controls[88]. Histamine, via the H2HR, has been shown to protect against fulminant hepatitis[89].

Histamine and liver cancer

There have been numerous studies involving histamine and histamine receptors in liver cancers. In a study using hepatocellular carcinoma cell lines, histamine stimulation was found to induce a differential effect by decreasing the growth of one line while increasing the growth of another[90]. Histamine-induced effects were attenuated by inhibition of either H1 or H2 HRs[90]. The H2HR antagonist, cimetidine is a common drug used for treatment of gastric ulcers and has also been shown to reduce liver metastasis via activation of selectins in liver sinusoids[91]. Both gastrin and histamine H2 receptor antagonists may play a synergistic role in helicobacter-induced gastric cancer[92].

HISTAMINE AND THE BILIARY TREE

The above studies have alluded to the role of histamine in a broad range of liver pathologies from hepatitis to hepatocellular carcinoma. Here we will reflect on the recent studies involving histamine and the histamine receptors in specific cholangiocyte pathologies ranging from cholestasis to cholangiocarcinoma.

Biliary hyperplasia and histamine

To date, only a handful of articles have addressed the direct role of histamine in biliary diseases. Though limited, these studies have provided evidence that there is a strong link between histamine and its receptors in the regulation of cholangiocyte function. Using a BDL-induced model of cholestasis, we have shown that chronic treatment of BDL rats with an H3HR agonist, (R)-(α)-(-)-methylhistamine dihydrobromide (RAMH), results in a significant decrease in biliary hyperplasia[65]. Numerous techniques revealed that liver tissue and RNA from both normal rats and BDL rats express all four of the histamine receptors[65]. Figure 2 depicts expression of the H3 receptor in proliferating cholangiocytes (induced by BDL) by immunohistochemical analysis. This finding is in accordance with other studies showing that cholangiocytes express a wide array of hormone receptors[13]. Biliary hyperplasia was reduced in vivo and also in vitro in freshly isolated cholangiocytes stimulated with RAMH via cAMP/PKA/ERK/Elk1 signaling. By downregulating this signaling pathway, the H3HR is able to decrease and manage biliary hyperplasia that could be a useful therapeutic tool in cholestatic liver diseases like PBC where cholangiocyte proliferation is prevalent.

Figure 2
Figure 2 Immunohistochemistry for the H3 histamine receptor in proliferating cholangiocytes (induced by bile duct ligation). Original magnification × 40.

In contrast, the H1HR agonist, HTMT dimaleate, increases the growth of small murine mouse cholangiocytes, but not large[66]. This study has great implication on the effects of histamine and histamine receptors in the heterogeneous population of cholangiocytes where cholangiopathies are likely to occur. These findings show that the H1HR elicits its effects by activation of Gαq, mobilization of calcium and increased intracellular inositol triphosphate receptor (IP3) levels. This was coupled with increased (1) phosphorylation of calmodulin-dependent protein kinase I (CaMKI) and ERK and (2) activation of the transcription factor CREB (cAMP response element binding protein). It was further shown that knockdown of CaMKI resulted in the loss of CREB activation and H1HR-induced increased proliferation in small cholangiocytes[66]. The importance of calcium-dependent signaling in cholangiocyte function and regulation is demonstrated by this study and is in accordance with other studies[93-95]. Taken together, these studies implicate histamine and the histamine receptors in cholangiocyte regulation.

Other recent studies have demonstrated that treatment with histamine; the H1HR agonist or the H2HR agonist (but not the H3HR agonist) stimulates the proliferation of cholangiocytes in normal rats[96]. Preliminary data suggests that there is a differential effect induced by the H1 and H2 HRs where the H1HR appears to signal predominantly through calcium-dependent mechanisms, whereas the H2HR is more prone to work through a cAMP/PKA-dependent signaling pathway[96]. Histamine is able to utilize both signaling pathways, but may have a preference for the H1HR-mediated path[96]. Figure 3 illustrates the differential signaling induced by histamine and histamine receptors in both normal and proliferating cholangiocytes[5]. It appears that histamine receptors may also mediate the VEGF-regulated cholangiocyte response during liver regeneration after 70% partial hepatectomy[96]. After 3 d post PH, treatment with the H1HR agonist, HTMT, induced a 50x fold increase in cholangiocyte proliferation that was coupled with increased VEGF expression[96]. This study has huge potential to lend a therapeutic “hand” to physicians treating patients after liver transplantation by aiding in the acceleration of liver regeneration and healing. Overall, it is clearly apparent that this biogenic amine and its receptors play numerous, critical roles in liver cholangiopathies and regeneration. Further investigations of these events are currently underway.

Figure 3
Figure 3 Differential signaling of histamine receptors in resting and proliferating cholangiocytes. Reprinted with permission from Demorrow S, Francis H, Alpini G, Biogenic Amine Actions on Cholangiocyte Function. Exp Biol Med (Maywood) 2007; 232: 1005-1013.
Biliary neoplasia and histamine

Little is currently known about the possible interaction between cholangiocarcinoma and histamine. Following our study with the cholestatic rat model and the H3HR agonist, RAMH, we evaluated the role and mechanisms of action by which RAMH regulates CCA growth[25]. Using numerous CCA cell lines and normal cholangiocytes, it was found that all four of the histamine receptors were expressed with an upregulation of H3HR expression compared to normal tissue. Treatment with RAMH induced a decrease in growth in all CCA cell lines, but had no effect in normal cholangiocytes. Interestingly, in this study we demonstrated that RAMH was potentially signaling via Gαo by inducing an increase in intracellular IP3 levels, but having no effect on cAMP signaling, thus ruling out any interaction with this pathway. Inhibitors for [Ca2+]i, PKC and PLC all blocked the RAMH-induced inhibitory action in CCA cells. Because PKC signaling has previously been shown to be important in cholangiocarcinoma[11,97,98], the role of this protein was investigated further. RAMH induced phosphorylation of PKC alpha (PKCα) and also caused translocation of PKCα from the cytosolic region into the membrane of CCA cells. Knockdown of PKCα ablated the suppressive effects induced by RAMH as well as reversing ERK phosphorylation. In final studies, in vivo examination was performed in a nude mouse model[59,62,63], implanted with CCA cells and treated with RAMH over the course of several weeks. Tumor growth was significantly inhibited by RAMH treatment compared to tumor growth from NaCl-treated mice. This was coupled with decreased proliferation and increased apoptosis. Figure 4A shows a representative image for immunohistochemical staining for both PCNA and caspase 3. Total PKCα expression was also increased in RAMH-treated tumors while VEGF and its receptors were decreased as seen by immunohistochemistry images provided in Figure 4B[25]. This is an important finding related to the regulation of PKCα in cholangiocarcinoma growth and also the ability of histamine to mediate VEGF expression, that plays a critical role in tumor development[63,99-102]. In work that is currently underway, we have found a role for histamine secretion and for other histamine receptor agonists and antagonists. Like other cancers, we have found that HDC and MAO-B are dysregulated in CCA and that CCA cell lines secrete an increased amount of histamine compared to normal cholangiocytes[103]. Further investigation has shown that acute histamine treatment has no significant impact on CCA growth, but that long-term stimulation (up to 2 wk) can increase growth that is coupled with increased VEGF gene expression[103]. In vivo treatment with histamine in our nude mouse model also increased tumor growth that was blocked by treatment with an HDC inhibitor[103]. These mechanisms are still being worked out, but it is clear that histamine has a trophic effect in CCA growth. We have also found that the histamine receptors are upregulated in CCA cell lines and have differential effects with regards to changes in proliferation[103]. As stated above, the H3HR agonist decreases CCA growth. However, we have seen that blocking the H2HR with cimetidine has no effect in either the CCA cell lines or in vivo (unpublished observations, Alpini and Francis). Stimulation with an H1HR antagonist also decreases CCA growth and increases apoptosis in vitro (unpublished observations, Alpini and Francis, 2007). Most recently we have found that clobenpropit, a highly potent H4HR agonist and selective H3HR antagonist can decrease CCA growth by integrin-mediated events including decreasing the invasive capacity of CCA cells[104]. These findings are complex and intricately woven together. Please refer to Table 1 for a summary of the effects of histamine and histamine receptors in CCA regulation.

Table 1 Effects of histamine and histamine receptors on cholangiocarcinomagrowth.
Expression levelAgonist effectAntagonist effectIn vitro/in vivo modelKnown or potential signaling mechanism
Histidine decarboxylase (HDC)↑↑↓↓ CCA growthBothInhibits histamine synthesis
Monoamine oxidase-B (MAOB)↓↓UnknownUnknownIn vitroUnknown
Histamine↑↑ secretion by CCA↑↑ CCA growth↓↓ CCA growthBoth↑↓ VEGF expression
H1HR↑↑↑ CCA growth↓↓ CCA growthIn vitroCa2+-mediated signaling
H2HR↑↑BothUnknown
H3HR↑↑↓↓ CCA growthBothPKCa-dependent signaling
H4HR↑↑↓↓ CCA growthIn vitroIntegrin-dependent decreased invasion
Figure 4
Figure 4 Immunohistochemistry images of tumors excised from vehicle- and RAMH-treated mice (× 40). A: Proliferating cholangiocytes (PCNA) and apoptotic cholangiocytes (Caspase 3). RAMH induced a decrease in PCNA-positive cholangiocytes coupled with an increase in apoptotic cholangiocytes compared to vehicle-treated mice; B: VEGF-A and VEGF-R2. RAMH treatment caused a decrease in the expression of both VEGF-A and VEGF-R2.
FUTURE PERSPECTIVES

While the studies reviewed above are relevant and important, it is also critical to remember that there are numerous other factors that may be involved in histamine regulation of liver disease. In vivo, the role of mast cells must be addressed as information regarding the interplay between hepatic mast cells and liver pathologies is increasing. The main source for histamine release is mast cells[105-107] and therefore these cells are likely to play some role in histamine regulation of cholangiocyte function. Bile acids have also been shown to play a major role in cholangiocyte proliferation, secretion and function[9,12,108-113]. We know that bile acids are also able to act on mast cells and can influence the type and amount of a mediator (like histamine) that is being released[114-117]. Not all bile acids induce the same effect. Lipophilic bile acids have been shown to activate mast cells and induce a large amount of histamine release, whereas ursodeoxycholate has almost no effect on histamine secretion from mast cells[118]. Bile acids are currently used as a treatment option for patients with etiologies ranging from gastro-esophageal reflux disease (GERD) to cholestatic liver diseases[119,120]. Histamine receptor agonists and antagonists are also used to treat various digestive diseases. The H2 antagonists, cimetidine and rantidine have both shown promise in treating GERD[121]. Because of the involvement of mast cells in irritable bowel syndrome (IBS), blocking mast cell interaction through either second generation antihistamines or proteinase-activated receptor antagonists could relieve pain in suffering patients[122]. How bile acids interact with hepatic mast cells and possibly cholangiocytes is unknown. However, understanding the interconnecting relationship between bile acidsmast cellscholangiocytes could allow us to take our current understanding of histamine-mediated regulation of cholangiocyte function to a new level and thereby allow us to turn translational science into bedside practice.

CONCLUSION

Taking the evidence discussed in this review we can glimpse into the future and see that histamine and histamine receptor regulation can have a major impact on liver diseases. However this information is only useful if we are able to dissect the mechanisms of histaminergic action that occur during disease progression.

Footnotes

Supported partly by Dr. Nicholas C Hightower Centennial Chair of Gastroenterology from Scott and White, the VA Research Scholar Award, a VA Merit Award and the NIH Grants DK58411, and DK76898 to Dr. Alpini, and by University funds to Dr. Onori and PRIN 2007 and Federate Athenaeum Funds from University of Rome “La Sapienza” to Professor Gaudio

Peer reviewers: Elke Cario, MD, Professor, University Hospital of Essen, Division of Gastroenterology and Hepatology, Institutsgruppe I, Virchowstr. 171, Essen 45147, Germany; Richard A Rippe, PhD, Scientific Review Officer, Office of Extramural Activities, Extramural Project Review Branch, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5635 Fishers Lane, Room 2109, Rockville, MD 20852, United States

S- Editor Zhang HN L- Editor Hughes D E- Editor Liu N

References
1.  Alpini G, Prall RT, LaRusso NF.  The pathobiology of biliary epithelia. The Liver; Biology & Pathobiology, 4E I M Arias, Boyer JL, Chisari FV, Fausto N, Jakoby W, Schachter D, and Shafritz DA, Philadelphia D, PA: Lippincott Williams & Wilkins 2001; 421-435.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Alpini G, Ulrich C, Roberts S, Phillips JO, Ueno Y, Podila PV, Colegio O, LeSage GD, Miller LJ, LaRusso NF. Molecular and functional heterogeneity of cholangiocytes from rat liver after bile duct ligation. Am J Physiol. 1997;272:G289-G297.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Alpini G, Glaser S, Robertson W, Rodgers RE, Phinizy JL, Lasater J, LeSage GD. Large but not small intrahepatic bile ducts are involved in secretin-regulated ductal bile secretion. Am J Physiol. 1997;272:G1064-G1074.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Kanno N, LeSage G, Glaser S, Alvaro D, Alpini G. Functional heterogeneity of the intrahepatic biliary epithelium. Hepatology. 2000;31:555-561.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Demorrow S, Francis H, Alpini G. Biogenic amine actions on cholangiocyte function. Exp Biol Med (Maywood). 2007;232:1005-1013.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Kanno N, LeSage G, Glaser S, Alpini G. Regulation of cholangiocyte bicarbonate secretion. Am J Physiol Gastrointest Liver Physiol. 2001;281:G612-G625.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  LeSage G, Glaser S, Alpini G. Regulation of cholangiocyte proliferation. Liver. 2001;21:73-80.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Alpini G, Lenzi R, Sarkozi L, Tavoloni N. Biliary physiology in rats with bile ductular cell hyperplasia. Evidence for a secretory function of proliferated bile ductules. J Clin Invest. 1988;81:569-578.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Xia X, Francis H, Glaser S, Alpini G, LeSage G. Bile acid interactions with cholangiocytes. World J Gastroenterol. 2006;12:3553-3563.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Alpini G, Glaser SS, Ueno Y, Rodgers R, Phinizy JL, Francis H, Baiocchi L, Holcomb LA, Caligiuri A, LeSage GD. Bile acid feeding induces cholangiocyte proliferation and secretion: evidence for bile acid-regulated ductal secretion. Gastroenterology. 1999;116:179-186.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Alpini G, Kanno N, Phinizy JL, Glaser S, Francis H, Taffetani S, LeSage G. Tauroursodeoxycholate inhibits human cholangiocarcinoma growth via Ca2+-, PKC-, and MAPK-dependent pathways. Am J Physiol Gastrointest Liver Physiol. 2004;286:G973-G982.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Alpini G, Ueno Y, Glaser SS, Marzioni M, Phinizy JL, Francis H, Lesage G. Bile acid feeding increased proliferative activity and apical bile acid transporter expression in both small and large rat cholangiocytes. Hepatology. 2001;34:868-876.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Glaser SS, Gaudio E, Miller T, Alvaro D, Alpini G. Cholangiocyte proliferation and liver fibrosis. Expert Rev Mol Med. 2009;11:e7.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Lesage G, Glaser SS, Gubba S, Robertson WE, Phinizy JL, Lasater J, Rodgers RE, Alpini G. Regrowth of the rat biliary tree after 70% partial hepatectomy is coupled to increased secretin-induced ductal secretion. Gastroenterology. 1996;111:1633-1644.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  LeSage GD, Alvaro D, Glaser S, Francis H, Marucci L, Roskams T, Phinizy JL, Marzioni M, Benedetti A, Taffetani S. Alpha-1 adrenergic receptor agonists modulate ductal secretion of BDL rats via Ca(2+)- and PKC-dependent stimulation of cAMP. Hepatology. 2004;40:1116-1127.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  LeSage GD, Benedetti A, Glaser S, Marucci L, Tretjak Z, Caligiuri A, Rodgers R, Phinizy JL, Baiocchi L, Francis H. Acute carbon tetrachloride feeding selectively damages large, but not small, cholangiocytes from normal rat liver. Hepatology. 1999;29:307-319.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  LeSage GD, Glaser SS, Marucci L, Benedetti A, Phinizy JL, Rodgers R, Caligiuri A, Papa E, Tretjak Z, Jezequel AM. Acute carbon tetrachloride feeding induces damage of large but not small cholangiocytes from BDL rat liver. Am J Physiol. 1999;276:G1289-G1301.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Alvaro D, Mancino MG, Glaser S, Gaudio E, Marzioni M, Francis H, Alpini G. Proliferating cholangiocytes: a neuroendocrine compartment in the diseased liver. Gastroenterology. 2007;132:415-431.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Alpini G, Glaser SS, Ueno Y, Pham L, Podila PV, Caligiuri A, LeSage G, LaRusso NF. Heterogeneity of the proliferative capacity of rat cholangiocytes after bile duct ligation. Am J Physiol. 1998;274:G767-G775.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Alpini G, Roberts S, Kuntz SM, Ueno Y, Gubba S, Podila PV, LeSage G, LaRusso NF. Morphological, molecular, and functional heterogeneity of cholangiocytes from normal rat liver. Gastroenterology. 1996;110:1636-1643.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Glaser SS, Gaudio E, Rao A, Pierce LM, Onori P, Franchitto A, Francis HL, Dostal DE, Venter JK, DeMorrow S. Morphological and functional heterogeneity of the mouse intrahepatic biliary epithelium. Lab Invest. 2009;89:456-469.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  LeSage G, Glaser S, Alpini G. Regulation of cholangiocyte proliferation. Liver. 2001;21:73-80.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Cassiman D, Libbrecht L, Sinelli N, Desmet V, Denef C, Roskams T. The vagal nerve stimulates activation of the hepatic progenitor cell compartment via muscarinic acetylcholine receptor type 3. Am J Pathol. 2002;161:521-530.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Lenzi R, Liu MH, Tarsetti F, Slott PA, Alpini G, Zhai WR, Paronetto F, Lenzen R, Tavoloni N. Histogenesis of bile duct-like cells proliferating during ethionine hepatocarcinogenesis. Evidence for a biliary epithelial nature of oval cells. Lab Invest. 1992;66:390-402.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Francis H, Onori P, Gaudio E, Franchitto A, DeMorrow S, Venter J, Kopriva S, Carpino G, Mancinelli R, White M. H3 histamine receptor-mediated activation of protein kinase Calpha inhibits the growth of cholangiocarcinoma in vitro and in vivo. Mol Cancer Res. 2009;7:1704-1713.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Gaudio E, Barbaro B, Alvaro D, Glaser S, Francis H, Franchitto A, Onori P, Ueno Y, Marzioni M, Fava G. Administration of r-VEGF-A prevents hepatic artery ligation-induced bile duct damage in bile duct ligated rats. Am J Physiol Gastrointest Liver Physiol. 2006;291:G307-G317.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Glaser S, Alvaro D, Ueno Y, Francis H, Marzioni M, Phinizy JL, Baumann B, Mancino MG, Venter J, LeSage G. Gastrin reverses established cholangiocyte proliferation and enhanced secretin-stimulated ductal secretion of BDL rats by activation of apoptosis through increased expression of Ca2+- dependent PKC isoforms. Liver Int. 2003;23:78-88.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Slott PA, Liu MH, Tavoloni N. Origin, pattern, and mechanism of bile duct proliferation following biliary obstruction in the rat. Gastroenterology. 1990;99:466-477.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Alpini G, Glaser S, Robertson W, Rodgers RE, Phinizy JL, Lasater J, LeSage GD. Large but not small intrahepatic bile ducts are involved in secretin-regulated ductal bile secretion. Am J Physiol. 1997;272:G1064-G1074.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Lesage G, Glaser S, Ueno Y, Alvaro D, Baiocchi L, Kanno N, Phinizy JL, Francis H, Alpini G. Regression of cholangiocyte proliferation after cessation of ANIT feeding is coupled with increased apoptosis. Am J Physiol Gastrointest Liver Physiol. 2001;281:G182-G190.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Mancinelli R, Onori P, Gaudio E, DeMorrow S, Franchitto A, Francis H, Glaser S, Carpino G, Venter J, Alvaro D. Follicle-stimulating hormone increases cholangiocyte proliferation by an autocrine mechanism via cAMP-dependent phosphorylation of ERK1/2 and Elk-1. Am J Physiol Gastrointest Liver Physiol. 2009;297:G11-G26.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Francis H, Glaser S, Ueno Y, Lesage G, Marucci L, Benedetti A, Taffetani S, Marzioni M, Alvaro D, Venter J. cAMP stimulates the secretory and proliferative capacity of the rat intrahepatic biliary epithelium through changes in the PKA/Src/MEK/ERK1/2 pathway. J Hepatol. 2004;41:528-537.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Glaser SS, Rodgers RE, Phinizy JL, Robertson WE, Lasater J, Caligiuri A, Tretjak Z, LeSage GD, Alpini G. Gastrin inhibits secretin-induced ductal secretion by interaction with specific receptors on rat cholangiocytes. Am J Physiol. 1997;273:G1061-G1070.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Lesage GD, Marucci L, Alvaro D, Glaser SS, Benedetti A, Marzioni M, Patel T, Francis H, Phinizy JL, Alpini G. Insulin inhibits secretin-induced ductal secretion by activation of PKC alpha and inhibition of PKA activity. Hepatology. 2002;36:641-651.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Cho WK, Boyer JL. Vasoactive intestinal polypeptide is a potent regulator of bile secretion from rat cholangiocytes. Gastroenterology. 1999;117:420-428.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Tietz PS, Alpini G, Pham LD, Larusso NF. Somatostatin inhibits secretin-induced ductal hypercholeresis and exocytosis by cholangiocytes. Am J Physiol. 1995;269:G110-G118.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Francis H, LeSage G, DeMorrow S, Alvaro D, Ueno Y, Venter J, Glaser S, Mancino MG, Marucci L, Benedetti A. The alpha2-adrenergic receptor agonist UK 14,304 inhibits secretin-stimulated ductal secretion by downregulation of the cAMP system in bile duct-ligated rats. Am J Physiol Cell Physiol. 2007;293:C1252-C1262.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Mancinelli R, Onori P, Gaudio E, Franchitto A, Carpino G, Ueno Y, Alvaro D, Annarale LP, Demorrow S, Francis H. Taurocholate feeding to bile duct ligated rats prevents caffeic acid-induced bile duct damage by changes in cholangiocyte VEGF expression. Exp Biol Med (Maywood). 2009;234:462-474.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Li Z, White P, Tuteja G, Rubins N, Sackett S, Kaestner KH. Foxa1 and Foxa2 regulate bile duct development in mice. J Clin Invest. 2009;119:1537-1545.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Sackett SD, Gao Y, Shin S, Esterson YB, Tsingalia A, Hurtt RS, Brondell K, Kaestner KH, Greenbaum LE. Foxl1 promotes liver repair following cholestatic injury in mice. Lab Invest. 2009;89:1387-1396.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  DeMorrow S, Francis H, Gaudio E, Ueno Y, Venter J, Onori P, Franchitto A, Vaculin B, Vaculin S, Alpini G. Anandamide inhibits cholangiocyte hyperplastic proliferation via activation of thioredoxin 1/redox factor 1 and AP-1 activation. Am J Physiol Gastrointest Liver Physiol. 2008;294:G506-G519.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Alpini G, Ueno Y, Tadlock L, Glaser SS, LeSage G, Francis H, Taffetani S, Marzioni M, Alvaro D, Patel T. Increased susceptibility of cholangiocytes to tumor necrosis factor-alpha cytotoxicity after bile duct ligation. Am J Physiol Cell Physiol. 2003;285:C183-C194.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Ueno Y, Francis H, Glaser S, Demorrow S, Venter J, Benedetti A, Fava G, Marzioni M, Alpini G. Taurocholic acid feeding prevents tumor necrosis factor-alpha-induced damage of cholangiocytes by a PI3K-mediated pathway. Exp Biol Med (Maywood). 2007;232:942-949.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Michels NA. Newer anatomy of the liver and its variant blood supply and collateral circulation. Am J Surg. 1966;112:337-347.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Rappaport AM. Hepatic blood flow: morphologic aspects and physiologic regulation. Int Rev Physiol. 1980;21:1-63.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Ekataksin W, Wake K. New concepts in biliary and vascular anatomy of the liver. Prog Liv Dis. 1997;15:1-30.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Kardon RH, Kessel RG. Three-dimensional organization of the hepatic microcirculation in the rodent as observed by scanning electron microscopy of corrosion casts. Gastroenterology. 1980;79:72-81.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Yamamoto K, Sherman I, Phillips MJ, Fisher MM. Three-dimensional observations of the hepatic arterial terminations in rat, hamster and human liver by scanning electron microscopy of microvascular casts. Hepatology. 1985;5:452-456.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Pannarale L, Onori P, Borghese F, Conte D, Gaudio E. Three-dimensional organization of the hepatic artery terminal branches: a scanning electron microscopic study of vascular corrosion casts of rat liver. Ital J Anat Embryol. 2007;112:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Gaudio E, Franchitto A, Pannarale L, Carpino G, Alpini G, Francis H, Glaser S, Alvaro D, Onori P. Cholangiocytes and blood supply. World J Gastroenterol. 2006;12:3546-3552.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Elvevold K, Smedsrød B, Martinez I. The liver sinusoidal endothelial cell: a cell type of controversial and confusing identity. Am J Physiol Gastrointest Liver Physiol. 2008;294:G391-G400.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Vidal-Vanaclocha F, Barberá-Guillem E. Fenestration patterns in endothelial cells of rat liver sinusoids. J Ultrastruct Res. 1985;90:115-123.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Esteller A. Physiology of bile secretion. World J Gastroenterol. 2008;14:5641-5649.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Deltenre P, Valla DC. Ischemic cholangiopathy. Semin Liver Dis. 2008;28:235-246.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Sirica AE. Cholangiocarcinoma: molecular targeting strategies for chemoprevention and therapy. Hepatology. 2005;41:5-15.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Aljiffry M, Walsh MJ, Molinari M. Advances in diagnosis, treatment and palliation of cholangiocarcinoma: 1990-2009. World J Gastroenterol. 2009;15:4240-4262.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Ustundag Y, Bayraktar Y. Cholangiocarcinoma: a compact review of the literature. World J Gastroenterol. 2008;14:6458-6466.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Blechacz BR, Smoot RL, Bronk SF, Werneburg NW, Sirica AE, Gores GJ. Sorafenib inhibits signal transducer and activator of transcription-3 signaling in cholangiocarcinoma cells by activating the phosphatase shatterproof 2. Hepatology. 2009;50:1861-1870.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Coufal M, Invernizzi P, Gaudio E, Bernuzzi F, Frampton GA, Onori P, Franchitto A, Carpino G, Ramirez JC, Alvaro D. Increased local dopamine secretion has growth-promoting effects in cholangiocarcinoma. Int J Cancer. 2010;126:2112-2122.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Wongkham S, Junking M, Wongkham C, Sripa B, Chur-In S, Araki N. Suppression of galectin-3 expression enhances apoptosis and chemosensitivity in liver fluke-associated cholangiocarcinoma. Cancer Sci. 2009;100:2077-2084.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Okada T, Sawada T, Kubota K. Rapamycin inhibits growth of cholangiocarcinoma cells. Hepatogastroenterology. 2009;56:6-10.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Onori P, DeMorrow S, Gaudio E, Franchitto A, Mancinelli R, Venter J, Kopriva S, Ueno Y, Alvaro D, Savage J. Caffeic acid phenethyl ester decreases cholangiocarcinoma growth by inhibition of NF-kappaB and induction of apoptosis. Int J Cancer. 2009;125:565-576.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Fava G, Demorrow S, Gaudio E, Franchitto A, Onori P, Carpino G, Glaser S, Francis H, Coufal M, Marucci L. Endothelin inhibits cholangiocarcinoma growth by a decrease in the vascular endothelial growth factor expression. Liver Int. 2009;29:1031-1042.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Pawar P, Ma L, Byon CH, Liu H, Ahn EY, Jhala N, Arnoletti JP, McDonald JM, Chen Y. Molecular mechanisms of tamoxifen therapy for cholangiocarcinoma: role of calmodulin. Clin Cancer Res. 2009;15:1288-1296.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Francis H, Franchitto A, Ueno Y, Glaser S, DeMorrow S, Venter J, Gaudio E, Alvaro D, Fava G, Marzioni M. H3 histamine receptor agonist inhibits biliary growth of BDL rats by downregulation of the cAMP-dependent PKA/ERK1/2/ELK-1 pathway. Lab Invest. 2007;87:473-487.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Francis H, Glaser S, Demorrow S, Gaudio E, Ueno Y, Venter J, Dostal D, Onori P, Franchitto A, Marzioni M. Small mouse cholangiocytes proliferate in response to H1 histamine receptor stimulation by activation of the IP3/CaMK I/CREB pathway. Am J Physiol Cell Physiol. 2008;295:C499-C513.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Nguyen T, Shapiro DA, George SR, Setola V, Lee DK, Cheng R, Rauser L, Lee SP, Lynch KR, Roth BL. Discovery of a novel member of the histamine receptor family. Mol Pharmacol. 2001;59:427-433.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Repka-Ramirez MS. New concepts of histamine receptors and actions. Curr Allergy Asthma Rep. 2003;3:227-231.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Hill SJ, Ganellin CR, Timmerman H, Schwartz JC, Shankley NP, Young JM, Schunack W, Levi R, Haas HL. International Union of Pharmacology. XIII. Classification of histamine receptors. Pharmacol Rev. 1997;49:253-278.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Fioretti B, Catacuzzeno L, Sforna L, Aiello F, Pagani F, Ragozzino D, Castigli E, Franciolini F. Histamine hyperpolarizes human glioblastoma cells by activating the intermediate-conductance Ca2+-activated K+ channel. Am J Physiol Cell Physiol. 2009;297:C102-C110.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Mondillo C, Patrignani Z, Reche C, Rivera E, Pignataro O. Dual role of histamine in modulation of Leydig cell steroidogenesis via HRH1 and HRH2 receptor subtypes. Biol Reprod. 2005;73:899-907.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Monczor F, Fernandez N, Riveiro E, Mladovan A, Baldi A, Shayo C, Davio C. Histamine H2 receptor overexpression induces U937 cell differentiation despite triggered mechanisms to attenuate cAMP signalling. Biochem Pharmacol. 2006;71:1219-1228.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Lovenberg TW, Pyati J, Chang H, Wilson SJ, Erlander MG. Cloning of rat histamine H(3) receptor reveals distinct species pharmacological profiles. J Pharmacol Exp Ther. 2000;293:771-778.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Khan UW, Rai U. Differential effects of histamine on Leydig cell and testicular macrophage activities in wall lizards: precise role of H1/H2 receptor subtypes. J Endocrinol. 2007;194:441-448.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Medhurst AD, Roberts JC, Lee J, Chen CP, Brown SH, Roman S, Lai MK. Characterization of histamine H3 receptors in Alzheimer’s Disease brain and amyloid over-expressing TASTPM mice. Br J Pharmacol. 2009;157:130-138.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Hao F, Tan M, Xu X, Cui MZ. Histamine induces Egr-1 expression in human aortic endothelial cells via the H1 receptor-mediated protein kinase Cdelta-dependent ERK activation pathway. J Biol Chem. 2008;283:26928-26936.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Dunford PJ, Williams KN, Desai PJ, Karlsson L, McQueen D, Thurmond RL. Histamine H4 receptor antagonists are superior to traditional antihistamines in the attenuation of experimental pruritus. J Allergy Clin Immunol. 2007;119:176-183.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Medina V, Croci M, Crescenti E, Mohamad N, Sanchez-Jiménez F, Massari N, Nuñez M, Cricco G, Martin G, Bergoc R. The role of histamine in human mammary carcinogenesis: H3 and H4 receptors as potential therapeutic targets for breast cancer treatment. Cancer Biol Ther. 2008;7:28-35.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Cianchi F, Vinci MC, Masini E. Histamine in cancer: the dual faces of the coin. Cancer Biol Ther. 2008;7:36-37.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Hornyak SC, Gehlsen KR, Haaparanta T. Histamine dihydrochloride protects against early alcohol-induced liver injury in a rat model. Inflammation. 2003;27:317-327.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Lozeva V, Tuomisto L, Tarhanen J, Butterworth RF. Increased concentrations of histamine and its metabolite, tele-methylhistamine and down-regulation of histamine H3 receptor sites in autopsied brain tissue from cirrhotic patients who died in hepatic coma. J Hepatol. 2003;39:522-527.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Lozeva V, Tuomisto L, Sola D, Plumed C, Hippeläinen M, Butterworth R. Increased density of brain histamine H(1) receptors in rats with portacaval anastomosis and in cirrhotic patients with chronic hepatic encephalopathy. Hepatology. 2001;33:1370-1376.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Okajima K, Harada N, Uchiba M. Ranitidine reduces ischemia/reperfusion-induced liver injury in rats by inhibiting neutrophil activation. J Pharmacol Exp Ther. 2002;301:1157-1165.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Adachi N, Liu K, Motoki A, Nishibori M, Arai T. Suppression of ischemia/reperfusion liver injury by histamine H4 receptor stimulation in rats. Eur J Pharmacol. 2006;544:181-187.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Motoki A, Adachi N, Liu K, Takahashi HK, Nishibori M, Yorozuya T, Arai T, Nagaro T. Suppression of ischaemia-induced cytokine release by dimaprit and amelioration of liver injury in rats. Basic Clin Pharmacol Toxicol. 2008;102:394-398.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Zhang Z, Leng X, Feng H, Ma F, Wang B. [Histamine-1 and -2 receptors in hepatic tissues of cirrhotic patients with portal hypertension]. Zhonghua Wai Ke Za Zhi. 1998;36:348-349.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Masaki T, Chiba S, Tatsukawa H, Noguchi H, Kakuma T, Endo M, Seike M, Watanabe T, Yoshimatsu H. The role of histamine H1 receptor and H2 receptor in LPS-induced liver injury. FASEB J. 2005;19:1245-1252.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Gittlen SD, Schulman ES, Maddrey WC. Raised histamine concentrations in chronic cholestatic liver disease. Gut. 1990;31:96-99.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Mori S, Takahashi HK, Nishibori M. [Inducible histamine protects mice from hepatitis through H2-receptor stimulation]. Yakugaku Zasshi. 2008;128:247-253.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Lampiasi N, Azzolina A, Montalto G, Cervello M. Histamine and spontaneously released mast cell granules affect the cell growth of human hepatocellular carcinoma cells. Exp Mol Med. 2007;39:284-294.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Lefranc F, Yeaton P, Brotchi J, Kiss R. Cimetidine, an unexpected anti-tumor agent, and its potential for the treatment of glioblastoma (review). Int J Oncol. 2006;28:1021-1030.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Takaishi S, Cui G, Frederick DM, Carlson JE, Houghton J, Varro A, Dockray GJ, Ge Z, Whary MT, Rogers AB. Synergistic inhibitory effects of gastrin and histamine receptor antagonists on Helicobacter-induced gastric cancer. Gastroenterology. 2005;128:1965-1083.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Taffetani S, Glaser S, Francis H, DeMorrow S, Ueno Y, Alvaro D, Marucci L, Marzioni M, Fava G, Venter J. Prolactin stimulates the proliferation of normal female cholangiocytes by differential regulation of Ca2+-dependent PKC isoforms. BMC Physiol. 2007;7:6.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Minagawa N, Ehrlich BE, Nathanson MH. Calcium signaling in cholangiocytes. World J Gastroenterol. 2006;12:3466-3470.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Marzioni M, Francis H, Benedetti A, Ueno Y, Fava G, Venter J, Reichenbach R, Mancino MG, Summers R, Alpini G. Ca2+-dependent cytoprotective effects of ursodeoxycholic and tauroursodeoxycholic acid on the biliary epithelium in a rat model of cholestasis and loss of bile ducts. Am J Pathol. 2006;168:398-409.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Francis H, DeMorrow S, Gaudio E, Ueno Y, Venter J, Onori P, Franchitto A, Carpino G, Marzioni M, Mancinelli R. The differential effects of histamine receptor subtypes on cholangiocyte proliferation of normal rats. Gastroenterology. 2009;136:A1578.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Kanno N, Glaser S, Chowdhury U, Phinizy JL, Baiocchi L, Francis H, LeSage G, Alpini G. Gastrin inhibits cholangiocarcinoma growth through increased apoptosis by activation of Ca2+-dependent protein kinase C-alpha. J Hepatol. 2001;34:284-291.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Roman RM, Bodily KO, Wang Y, Raymond JR, Fitz JG. Activation of protein kinase Calpha couples cell volume to membrane Cl- permeability in HTC hepatoma and Mz-ChA-1 cholangiocarcinoma cells. Hepatology. 1998;28:1073-1080.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Mancino A, Mancino MG, Glaser SS, Alpini G, Bolognese A, Izzo L, Francis H, Onori P, Franchitto A, Ginanni-Corradini S. Estrogens stimulate the proliferation of human cholangiocarcinoma by inducing the expression and secretion of vascular endothelial growth factor. Dig Liver Dis. 2009;41:156-163.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Möbius C, Demuth C, Aigner T, Wiedmann M, Wittekind C, Mössner J, Hauss J, Witzigmann H. Evaluation of VEGF A expression and microvascular density as prognostic factors in extrahepatic cholangiocarcinoma. Eur J Surg Oncol. 2007;33:1025-1029.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Park BK, Paik YH, Park JY, Park KH, Bang S, Park SW, Chung JB, Park YN, Song SY. The clinicopathologic significance of the expression of vascular endothelial growth factor-C in intrahepatic cholangiocarcinoma. Am J Clin Oncol. 2006;29:138-142.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Fava G, Marucci L, Glaser S, Francis H, De Morrow S, Benedetti A, Alvaro D, Venter J, Meininger C, Patel T. gamma-Aminobutyric acid inhibits cholangiocarcinoma growth by cyclic AMP-dependent regulation of the protein kinase A/extracellular signal-regulated kinase 1/2 pathway. Cancer Res. 2005;65:11437-11446.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Francis H, DeMorrow H, Venter J, Vaculin S, Vaculin B, Savage J, Alpini G. Evidence for a novel role for histamine regulation of cholangiocarcinoma growth by an autocrine mechanism. Gastroenterology. 2008;134:A58.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Meng F, Alpini G, DeMorrow S, Frampton G, Venter J, White M, Kopriva S, Chiasson V, Francis H. Stimulation of the H4 histamine receptor decreases cholangiocarcinoma growth and invasion via integrin-dependent mechanisms. Hepatology. 2009;50:1071.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Beaven MA. Our perception of the mast cell from Paul Ehrlich to now. Eur J Immunol. 2009;39:11-25.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Vyas H, Krishnaswamy G. Paul Ehrlich’s “Mastzellen”--from aniline dyes to DNA chip arrays: a historical review of developments in mast cell research. Methods Mol Biol. 2006;315:3-11.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Yong LC. The mast cell: origin, morphology, distribution, and function. Exp Toxicol Pathol. 1997;49:409-424.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Xia X, Roundtree M, Merikhi A, Lu X, Shentu S, Lesage G. Degradation of the apical sodium-dependent bile acid transporter by the ubiquitin-proteasome pathway in cholangiocytes. J Biol Chem. 2004;279:44931-44937.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Bijvelds MJ, Jorna H, Verkade HJ, Bot AG, Hofmann F, Agellon LB, Sinaasappel M, de Jonge HR. Activation of CFTR by ASBT-mediated bile salt absorption. Am J Physiol Gastrointest Liver Physiol. 2005;289:G870-G879.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Marzioni M, Francis H, Benedetti A, Ueno Y, Fava G, Venter J, Reichenbach R, Mancino MG, Summers R, Alpini G. Ca2+-dependent cytoprotective effects of ursodeoxycholic and tauroursodeoxycholic acid on the biliary epithelium in a rat model of cholestasis and loss of bile ducts. Am J Pathol. 2006;168:398-409.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Pusl T, Beuers U. Ursodeoxycholic acid treatment of vanishing bile duct syndromes. World J Gastroenterol. 2006;12:3487-3495.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Fiorotto R, Spirlì C, Fabris L, Cadamuro M, Okolicsanyi L, Strazzabosco M. Ursodeoxycholic acid stimulates cholangiocyte fluid secretion in mice via CFTR-dependent ATP secretion. Gastroenterology. 2007;133:1603-1613.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Fava G, Marzioni M, Francis H, Glaser S, Demorrrow S, Ueno Y, Benedetti A, Alpini G. Novel interaction of bile acid and neural signaling in the regulation of cholangiocyte function. Hepatol Res. 2007;37 Suppl 3:S420-S429.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Rees WD, Rhodes J, Williams BC, Owen E, Newcombe RG. Effect of mast cell degranulation on gastric mucosal damage produced by sodium taurocholate in the rat. Gastroenterology. 1978;74:492-495.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Peh KH, Wan BY, Assem ES. Characteristics of deoxycholic acid-induced histamine release from mast cells of guinea-pig rectocolonic mucosa and rat peritoneal cavity. Agents Actions. 1991;33:76-80.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Gelbmann CM, Schteingart CD, Thompson SM, Hofmann AF, Barrett KE. Mast cells and histamine contribute to bile acid-stimulated secretion in the mouse colon. J Clin Invest. 1995;95:2831-2839.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Gui X, Carraway RE. Involvement of mast cells in basal and neurotensin-induced intestinal absorption of taurocholate in rats. Am J Physiol Gastrointest Liver Physiol. 2004;287:G408-G416.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Quist RG, Ton-Nu HT, Lillienau J, Hofmann AF, Barrett KE. Activation of mast cells by bile acids. Gastroenterology. 1991;101:446-456.  [PubMed]  [DOI]  [Cited in This Article: ]
119.  Tack J. Review article: the role of bile and pepsin in the pathophysiology and treatment of gastro-oesophageal reflux disease. Aliment Pharmacol Ther. 2006;24 Suppl 2:10-16.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Hofmann AF. Bile acids: trying to understand their chemistry and biology with the hope of helping patients. Hepatology. 2009;49:1403-1418.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  Schubert ML, Peura DA. Control of gastric acid secretion in health and disease. Gastroenterology. 2008;134:1842-1860.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Barbara G, Stanghellini V, De Giorgio R, Corinaldesi R. Functional gastrointestinal disorders and mast cells: implications for therapy. Neurogastroenterol Motil. 2006;18:6-17.  [PubMed]  [DOI]  [Cited in This Article: ]