Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Hepatol ; 61(6): 1260-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25046848

ABSTRACT

BACKGROUND & AIMS: Hepatic encephalopathy (HE) is a neurologic disorder that develops during liver failure. Few studies exist investigating systemic-central signalling during HE outside of inflammatory signalling. The transcription factor Gli1, which can be modulated by hedgehog signalling or transforming growth factor ß1 (TGFß1) signalling, has been shown to be protective in various neuropathies. We measured Gli1 expression in brain tissues from mice and evaluated how circulating TGFß1 and canonical hedgehog signalling regulate its activation. METHODS: Mice were injected with azoxymethane (AOM) to induce liver failure and HE in the presence of Gli1 vivo-morpholinos, the hedgehog inhibitor cyclopamine, Smoothened vivo-morpholinos, a Smoothened agonist, or TGFß-neutralizing antibodies. Molecular analyses were used to assess Gli1, hedgehog signalling, and TGFß1 signalling in the liver and brain of AOM mice and HE patients. RESULTS: Gli1 expression was increased in brains of AOM mice and in HE patients. Intra-cortical infusion of Gli1 vivo-morpholinos exacerbated the neurologic deficits of AOM mice. Measures to modulate hedgehog signalling had no effect on HE neurological decline. Levels of TGFß1 increased in the liver and serum of mice following AOM administration. TGFß neutralizing antibodies slowed neurologic decline following AOM administration without significantly affecting liver damage. TGFß1 inhibited Gli1 expression via a SMAD3-dependent mechanism. Conversely, inhibiting TGFß1 increased Gli1 expression. CONCLUSIONS: Cortical activation of Gli1 protects mice from induction of HE. TGFß1 suppresses Gli1 in neurons via SMAD3 and promotes the neurologic decline. Strategies to activate Gli1 or inhibit TGFß1 signalling might be developed to treat patients with HE.


Subject(s)
Hepatic Encephalopathy/metabolism , Hepatic Encephalopathy/prevention & control , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/metabolism , Transforming Growth Factor beta1/blood , Animals , Autopsy , Brain/metabolism , Brain/pathology , Case-Control Studies , Disease Models, Animal , Female , Hedgehog Proteins/metabolism , Humans , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Morpholinos/metabolism , Signal Transduction/physiology , Smad3 Protein/metabolism , Transcription Factors/metabolism , Zinc Finger Protein GLI1
2.
Dig Liver Dis ; 46(6): 527-34, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24629820

ABSTRACT

BACKGROUND: The blood brain barrier tightly regulates the passage of molecules into the brain and becomes leaky following obstructive cholestasis. The aim of this study was to determine if increased serum bile acids observed during cholestasis permeabilize the blood brain barrier. METHODS: Rats underwent bile duct ligation or deoxycholic or chenodeoxycholic acid injections and blood brain barrier permeability assessed. In vitro, the permeability of rat brain microvessel endothelial cell monolayers, the expression and phosphorylation of occludin, ZO-1 and ZO-2 as well as the activity of Rac1 was assessed after treatment with plasma from cholestatic rats, or bile acid treatment, in the presence of a Rac1 inhibitor. RESULTS: Blood brain barrier permeability was increased in vivo and in vitro following bile duct ligation or treatment with bile acids. Associated with the bile acid-stimulated increase in endothelial cell monolayer permeability was elevated Rac1 activity and increased phosphorylation of occludin. Pretreatment of endothelial cell monolayers with a Rac1 inhibitor prevented the effects of bile acid treatment on occludin phosphorylation and monolayer permeability. CONCLUSIONS: These data suggest that increased circulating serum bile acids may contribute to the increased permeability of the blood brain barrier seen during obstructive cholestasis via disruption of tight junctions.


Subject(s)
Blood-Brain Barrier/metabolism , Chenodeoxycholic Acid/blood , Cholestasis/physiopathology , Deoxycholic Acid/blood , Microvessels/metabolism , Tight Junctions/metabolism , rac1 GTP-Binding Protein/metabolism , Aminoquinolines/pharmacology , Animals , Bile Ducts/surgery , Chenodeoxycholic Acid/pharmacology , Deoxycholic Acid/pharmacology , Disease Models, Animal , Endothelial Cells , Ligation , Male , Occludin/genetics , Occludin/metabolism , Permeability/drug effects , Phosphorylation/drug effects , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tissue Culture Techniques , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism , Zonula Occludens-2 Protein/genetics , Zonula Occludens-2 Protein/metabolism , rac1 GTP-Binding Protein/antagonists & inhibitors
3.
Am J Physiol Gastrointest Liver Physiol ; 305(3): G250-7, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23703654

ABSTRACT

Neuropeptide Y (NPY) exerts its functions through six subtypes of receptors (Y1-Y6). Biliary homeostasis is regulated by several factors through autocrine/paracrine signaling. NPY inhibits cholangiocarcinoma growth; however, no information exists regarding the autocrine/paracrine role of NPY on biliary hyperplasia during cholestasis. The aims of this study were to determine: 1) the expression of NPY and Y1-Y5 in cholangiocytes and 2) the paracrine/autocrine effects of NPY on cholangiocyte proliferation. Normal or bile duct ligation (BDL) rats were treated with NPY, neutralizing anti-NPY antibody, or vehicle for 7 days. NPY and NPY receptor (NPYR) expression was assessed in liver sections and isolated cholangiocytes. NPY secretion was assessed in serum and bile from normal and BDL rats, as well as supernatants from normal and BDL cholangiocytes and normal rat cholangiocyte cell line [intrahepatic normal cholangiocyte culture (NRICC)]. We evaluated intrahepatic bile ductal mass (IBDM) in liver sections and proliferation in cholangiocytes. With the use of NRICC, the effects of NPY or anti-NPY antibody on cholangiocyte proliferation were determined. The expression of NPY and all NPYR were increased after BDL. NPY levels were lower in serum and cholangiocyte supernatant from BDL compared with normal rats. NPY secretion from NRICC was detected at both the basolateral and apical domains. Chronic NPY treatment decreased proliferating cellular nuclear antigen (PCNA) expression and IBDM in BDL rats. Administration of anti-NPY antibody to BDL rats increased cholangiocyte proliferation and IBDM. NPY treatment of NRICC decreased PCNA expression and increased the cell cycle arrest, whereas treatment with anti-NPY antibody increased proliferation. Therapies targeting NPY-mediated signaling may prove beneficial for the treatment of cholangiopathies.


Subject(s)
Autocrine Communication/physiology , Bile Ducts, Intrahepatic/pathology , Cholestasis/pathology , Neuropeptide Y/pharmacology , Paracrine Communication/physiology , Animals , Antibodies, Neutralizing/pharmacology , Bile Ducts, Intrahepatic/chemistry , Bile Ducts, Intrahepatic/physiopathology , Cell Line , Cell Proliferation/drug effects , Cholestasis/physiopathology , Homeostasis , Hyperplasia , Male , Neuropeptide Y/antagonists & inhibitors , Neuropeptide Y/physiology , Proliferating Cell Nuclear Antigen/analysis , RNA, Messenger/analysis , Rats , Rats, Inbred F344 , Receptors, Neuropeptide Y/analysis , Receptors, Neuropeptide Y/genetics , Signal Transduction/drug effects
4.
Am J Physiol Gastrointest Liver Physiol ; 303(11): G1202-11, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23086914

ABSTRACT

Progranulin (PGRN), a secreted growth factor, regulates the proliferation of various epithelial cells. Its mechanism of action is largely unknown. Sirtuin 1 (Sirt1) is a protein deacetylase that is known to regulate the transcriptional activity of the forkhead receptor FOXO1, thereby modulating the balance between proapoptotic and cell cycle-arresting genes. We have shown that PGRN is overexpressed in cholangiocarcinoma and stimulates proliferation. However, its effects on hyperplastic cholangiocyte proliferation are unknown. In the present study, the expression of PGRN and its downstream targets was determined after bile duct ligation (BDL) in mice and in a mouse cholangiocyte cell line after stimulation with PGRN. The effects of PGRN on cholangiocyte proliferation were assessed in sham-operated (sham) and BDL mice treated with PGRN or by specifically knocking down endogenous PGRN expression using Vivo-Morpholinos or short hairpin RNA. PGRN expression and secretion were upregulated in proliferating cholangiocytes isolated after BDL. Treatment of mice with PGRN increased biliary mass and cholangiocyte proliferation in vivo and in vitro and enhanced cholangiocyte proliferation observed after BDL. PGRN treatment decreased Sirt1 expression and increased the acetylation of FOXO1, resulting in the cytoplasmic accumulation of FOXO1 in cholangiocytes. Overexpression of Sirt1 in vitro prevented the proliferative effects of PGRN. Conversely, knocking down PGRN expression in vitro or in vivo inhibited cholangiocyte proliferation. In conclusion, these data suggest that the upregulation of PGRN may be a key feature stimulating cholangiocyte proliferation. Modulating PGRN levels may be a viable technique for regulating the balance between ductal proliferation and ductopenia observed in a variety of cholangiopathies.


Subject(s)
Cell Proliferation/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Sirtuin 1/physiology , Animals , Bile Ducts/cytology , Bile Ducts/surgery , Cholangiocarcinoma/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/metabolism , Granulins , Ligation , Male , Mice , Mice, Inbred C57BL , Progranulins
5.
Gut ; 61(2): 268-77, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22068162

ABSTRACT

BACKGROUND AND OBJECTIVES: Cholangiocarcinoma is a devastating cancer of biliary origin with limited treatment options. The growth factor, progranulin, is overexpressed in a number of tumours. The study aims were to assess the expression of progranulin in cholangiocarcinoma and to determine its effects on tumour growth. METHODS: The expression and secretion of progranulin were evaluated in multiple cholangiocarcinoma cell lines and in clinical samples from patients with cholangiocarcinoma. The role of interleukin 6 (IL-6)-mediated signalling in the expression of progranulin was assessed using a combination of specific inhibitors and shRNA knockdown techniques. The effect of progranulin on proliferation and Akt activation and subsequent effects of FOXO1 phosphorylation were assessed in vitro. Progranulin knockdown cell lines were established, and the effects on cholangiocarcinoma growth were determined. RESULTS: Progranulin expression and secretion were upregulated in cholangiocarcinoma cell lines and tissue, which were in part via IL-6-mediated activation of the ERK1/2/RSK1/C/EBPß pathway. Blocking any of these signalling molecules, by either pharmacological inhibitors or shRNA, prevented the IL-6-dependent activation of progranulin expression. Treatment of cholangiocarcinoma cells with recombinant progranulin increased cell proliferation in vitro by a mechanism involving Akt phosphorylation leading to phosphorylation and nuclear extrusion of FOXO1. Knockdown of progranulin expression in cholangiocarcinoma cells decreased the expression of proliferating cellular nuclear antigen, a marker of proliferative capacity, and slowed tumour growth in vivo. CONCLUSIONS: Evidence is presented for a role for progranulin as a novel growth factor regulating cholangiocarcinoma growth. Specific targeting of progranulin may represent an alternative for the development of therapeutic strategies.


Subject(s)
Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic , Biomarkers, Tumor/metabolism , Cholangiocarcinoma/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-6/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cholangiocarcinoma/pathology , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Humans , Immunoblotting , Male , Mice , Mice, Inbred BALB C , Progranulins , Real-Time Polymerase Chain Reaction , Signal Transduction , Tissue Array Analysis
6.
Am J Physiol Gastrointest Liver Physiol ; 302(1): G182-93, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21979757

ABSTRACT

Cholestatic patients often present with clinical features suggestive of adrenal insufficiency. In the bile duct-ligated (BDL) model of cholestasis, the hypothalamic-pituitary-adrenal (HPA) axis is suppressed. The consequences of this suppression on cholangiocyte proliferation are unknown. We evaluated 1) HPA axis activity in various rat models of cholestasis and 2) effects of HPA axis modulation on cholangiocyte proliferation. Expression of regulatory molecules of the HPA axis was determined after BDL, partial BDL, and α-naphthylisothiocyanate (ANIT) intoxication. The HPA axis was suppressed by inhibition of hypothalamic corticotropin-releasing hormone (CRH) expression by central administration of CRH-specific Vivo-morpholinos or by adrenalectomy. After BDL, the HPA axis was reactivated by 1) central administration of CRH, 2) systemic ACTH treatment, or 3) treatment with cortisol or corticosterone for 7 days postsurgery. There was decreased expression of 1) hypothalamic CRH, 2) pituitary ACTH, and 3) key glucocorticoid synthesis enzymes in the adrenal glands. Serum corticosterone and cortisol remained low after BDL (but not partial BDL) compared with sham surgery and after 2 wk of ANIT feeding. Experimental suppression of the HPA axis increased cholangiocyte proliferation, shown by increased cytokeratin-19- and proliferating cell nuclear antigen-positive cholangiocytes. Conversely, restoration of HPA axis activity inhibited BDL-induced cholangiocyte proliferation. Suppression of the HPA axis is an early event following BDL and induces cholangiocyte proliferation. Knowledge of the role of the HPA axis during cholestasis may lead to development of innovative treatment paradigms for chronic liver disease.


Subject(s)
Bile Ducts/metabolism , Cholestasis, Extrahepatic/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , 1-Naphthylisothiocyanate/toxicity , Adrenalectomy , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/pharmacology , Animals , Cell Proliferation , Corticosterone/blood , Corticosterone/pharmacology , Corticotropin-Releasing Hormone/blood , Corticotropin-Releasing Hormone/pharmacology , Hydrocortisone/blood , Hydrocortisone/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Male , Morpholinos/pharmacology , Pituitary-Adrenal System/drug effects , Rats , Rats, Sprague-Dawley
7.
Lab Invest ; 91(7): 1007-17, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21464819

ABSTRACT

Cholangiocarcinomas are devastating cancers of biliary origin with limited treatment options. It has previously been shown that the endocannabinoid anandamide exerts antiproliferative effects on cholangiocarcinoma independent of any known cannabinoid receptors, and by the stabilization of lipid rafts, thereby allowing the recruitment and activation of the Fas death receptor complex. Recently, GPR55 was identified as a putative cannabinoid receptor; therefore, the role of GPR55 in the antiproliferative effects of anandamide was evaluated. GPR55 is expressed in all cholangiocarcinoma cells and liver biopsy samples to a similar level as in non-malignant cholangiocytes. Treatment with either anandamide or the GPR55 agonist, O-1602, reduced cholangiocarcinoma cell proliferation in vitro and in vivo. Furthermore, knocking down the expression of GPR55 prevented the antiproliferative effects of anandamide. Coupled to these effects was an increase in JNK activity. The antiproliferative effects of anandamide could be blocked by pretreatment with a JNK inhibitor and the lipid raft disruptors ß-methylcyclodextrin and fillipin III. Activation of GPR55 by anandamide or O-1602 increased the amount of Fas in the lipid raft fractions, which could be blocked by pretreatment with the JNK inhibitor. These data represent the first evidence that GPR55 activation by anandamide can lead to the recruitment and activation of the Fas death receptor complex and that targeting GPR55 activation may be a viable option for the development of therapeutic strategies to treat cholangiocarcinoma.


Subject(s)
Arachidonic Acids/pharmacology , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cannabinoid Receptor Modulators/pharmacology , Cell Proliferation/drug effects , Cholangiocarcinoma/pathology , Polyunsaturated Alkamides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line, Tumor , Endocannabinoids , Humans , Mice , Mice, Nude , Receptors, Cannabinoid
SELECTION OF CITATIONS
SEARCH DETAIL
...