Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Am J Pathol ; 185(6): 1724-39, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25892683

ABSTRACT

Cholangiocarcinomas (CCAs) comprise a mucin-secreting form, intrahepatic or perihilar, and a mixed form located peripherally. We characterized cancer stem cells (CSCs) in CCA subtypes and evaluated their cancerogenic potential. CSC markers were investigated in 25 human CCAs in primary cultures and established cell lines. Tumorigenic potential was evaluated in vitro or in xenografted mice after s.c. or intrahepatic injection in normal and cirrhotic (carbon tetrachloride-induced) mice. CSCs comprised more than 30% of the tumor mass. Although the CSC profile was similar between mucin-intrahepatic and mucin-perihilar subtypes, CD13(+) CSCs characterized mixed-intrahepatic, whereas LGR5(+) characterized mucin-CCA subtypes. Many neoplastic cells expressed epithelial-mesenchymal transition markers and coexpressed mesenchymal and epithelial markers. In primary cultures, epithelial-mesenchymal transition markers, mesenchymal markers (vimentin, CD90), and CD13 largely predominated over epithelial markers (CD133, EpCAM, and LGR5). In vitro, CSCs expressing epithelial markers formed a higher number of spheroids than CD13(+) or CD90(+) CSCs. In s.c. tumor xenografts, tumors dominated by stromal markers were formed primarily by CD90(+) and CD13(+) cells. By contrast, in intrahepatic xenografts in cirrhotic livers, tumors were dominated by epithelial traits reproducing the original human CCAs. In conclusion, CSCs were rich in human CCAs, implicating CCAs as stem cell-based diseases. CSC subpopulations generate different types of cancers depending on the microenvironment. Remarkably, CSCs reproduce the original human CCAs when injected into cirrhotic livers.


Subject(s)
Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Liver/pathology , Neoplastic Stem Cells/pathology , Aged , Aged, 80 and over , Animals , Bile Duct Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cholangiocarcinoma/metabolism , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Liver/metabolism , Male , Mice , Middle Aged , Neoplastic Stem Cells/metabolism , Transplantation, Heterologous
2.
BMC Gastroenterol ; 14: 204, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25471120

ABSTRACT

BACKGROUND: Efforts to identify cell sources and approaches for cell therapy of liver diseases are ongoing, taking into consideration the limits recognized for adult liver tissue and for other forms of stem cells. In the present study, we described the first procedure of via hepatic artery transplantation of human fetal biliary tree stem cells in patients with advanced cirrhosis. METHODS: The cells were immune-sorted from human fetal biliary tree by protocols in accordance with current good manufacturing practice (cGMP) and extensively characterized. Two patients with advanced liver cirrhosis (Child-Pugh C) have been submitted to the procedure and observed through a 12 months follow-up. RESULTS: The resulting procedure was found absolutely safe. Immuno-suppressants were not required, and the patients did not display any adverse effects correlated with cell transplantation or suggestive of immunological complications. From a clinical point of view, both patients showed biochemical and clinical improvement during the 6 month follow-up and the second patient maintained a stable improvement for 12 months. CONCLUSION: This report represents proof of the concept that the human fetal biliary tree stem cells are a suitable and large source for cell therapy of liver cirrhosis. The isolation procedure can be carried out under cGMP conditions and, finally, the infusion procedure is easy and safe for the patients. This represents the basis for forthcoming controlled clinical trials.


Subject(s)
Fetal Tissue Transplantation/methods , Liver Cirrhosis/therapy , Stem Cell Transplantation/methods , Aged , Antigens, Neoplasm/metabolism , Biliary Tract/cytology , Cell Adhesion Molecules/metabolism , Epithelial Cell Adhesion Molecule , Female , Hepatic Artery , Humans , Male
3.
Hepatobiliary Surg Nutr ; 2(5): 272-80, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24570958

ABSTRACT

Cholangiocarcinoma (CCA) is a very heterogeneous cancer from any point of view, including epidemiology, risk factors, morphology, pathology, molecular pathology, modalities of growth and clinical features. Given this heterogeneity, a uniform classification respecting the epidemiologic, pathologic and clinical needs is currently lacking. In this manuscript we discussed the different proposed classifications of CCA in relation with recent advances in pathophysiology and biology of this cancer.

4.
J Hepatol ; 57(5): 987-94, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22820480

ABSTRACT

BACKGROUND & AIMS: Biliary tree, liver, and pancreas share a common embryological origin. We previously demonstrated the presence of stem/progenitor cells of endodermal origin in the adult human extrahepatic biliary tree. This study evaluated the human foetal biliary trees as sources of stem/progenitor cells of multiple endodermal-derived mature fates. METHODS: Human foetal intrahepatic and extrahepatic biliary tree tissues and isolated cells were tested for cytoplasmic and surface markers of stem cells and committed progenitors, as well as endodermal transcription factors requisite for a liver versus pancreatic fate. In vitro and in vivo experiments were conducted to evaluate the potential mature fates of differentiation. RESULTS: Foetal biliary tree cells proliferated clonogenically for more than 1 month on plastic in a serum-free Kubota medium. After culture expansion, cells exhibited multipotency and could be restricted to certain lineages under defined microenvironments, including hepatocytes, cholangiocytes, and pancreatic islet cells. Transplantation of foetal biliary tree cells into the livers of immunodeficient mice resulted in effective engraftment and differentiation into mature hepatocytes and cholangiocytes. CONCLUSIONS: Foetal biliary trees contain multipotent stem/progenitor cells comparable with those in adults. These cells can be easily expanded and induced in vitro to differentiate into liver and pancreatic mature fates, and engrafted and differentiated into mature cells when transplanted in vivo. These findings further characterise the development of these stem/progenitor cell populations from foetuses to adults, which are thought to contribute to liver and pancreas organogenesis throughout life.


Subject(s)
Biliary Tract/cytology , Biliary Tract/embryology , Fetus/cytology , Multipotent Stem Cells/cytology , Animals , Bile Ducts, Extrahepatic/cytology , Bile Ducts, Intrahepatic/cytology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Hepatocytes/cytology , Humans , In Vitro Techniques , Mice , Mice, SCID , Pancreas/cytology , Phenotype
5.
Dig Liver Dis ; 44(2): 134-42, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21782536

ABSTRACT

BACKGROUND: Cholangiocarcinoma cells over-express oestrogen receptor-ß, which displays anti-proliferative and pro-apoptotic effects. AIM: To evaluate the effects of a newly developed and highly selective oestrogen receptor-ß agonist (KB9520) on experimental intrahepatic cholangiocarcinoma. METHODS: In vitro, the effects of KB9520 on apoptosis and proliferation of HuH-28 cells, HuH-28 cells with selective oestrogen receptor-ß silencing (by small interfering RNA), HepG2 cells (oestrogen receptor-α and oestrogen receptor-ß negative) and HepER3 cells (HepG2 cells transformed to stably express oestrogen receptor-α) were evaluated. In vivo, the effects of KB9520 on experimental intrahepatic cholangiocarcinoma, induced by thioacetamide administration were tested. RESULTS: In vitro, KB9520 induced apoptosis and inhibited proliferation of HuH-28 cells. KB9520 effects were absent in cells lacking oestrogen receptor-α and ß (HepG2) and in cells expressing only oestrogen receptor-α (HepER3); its pro-apoptotic effect was impaired in cells where oestrogen receptor-ß expression was decreased by specific small interfering RNA. In vivo, KB9520 inhibited experimental intrahepatic cholangiocarcinoma development in thioacetamide-treated rats and promoted tumour regression in rats where tumour was already established. In treated animals, tumour areas showed reduced proliferation but increased apoptosis. CONCLUSIONS: KB9520 induced apoptosis in cholangiocarcinoma by selectively acting on oestrogen receptor-ß, suggesting that oestrogen receptor-ß selective agonists may be a novel and effective therapeutic option for the medical treatment of intrahepatic cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms/drug therapy , Bile Ducts, Intrahepatic , Cholangiocarcinoma/drug therapy , Estrogen Receptor beta/agonists , Liver Neoplasms/drug therapy , Neoplasms, Experimental/drug therapy , Selective Estrogen Receptor Modulators/therapeutic use , Animals , Apoptosis , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Blotting, Western , Cell Line, Tumor , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Estrogen Receptor beta/biosynthesis , Estrogen Receptor beta/genetics , Gene Expression Regulation, Neoplastic , Humans , In Situ Nick-End Labeling , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , RNA, Neoplasm , Rats , Rats, Wistar , Treatment Outcome
6.
World J Gastrointest Oncol ; 2(11): 407-16, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21160904

ABSTRACT

Cholangiocarcinoma (CCA) is a malignant tumour that arises from biliary epithelium at any portion of the biliary tree. CCA is currently classified as intra-hepatic or extra-hepatic CCA (EH-CCA). Recent evidences suggest that intra-hepatic CCA (IH-CCA) and EH-CCA are biologically different cancers, giving further support to a number of recent epidemiological studies showing large differences in terms of incidence, mortality and risk factors. The purpose of this manuscript is to review recent literature dealing with the descriptive epidemiology and risk factors of CCA with a special effort to compare IH- with EH-CCA.

7.
Am J Pathol ; 177(4): 1779-90, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20724592

ABSTRACT

Prostate apoptosis response-4 (Par-4) is a tumor suppressor protein that sensitizes cells to apoptosis; therefore, Par-4 modulation has therapeutic potential. No data currently exist on Par-4 expression in cholangiocarcinoma (CCA). We evaluated the expression of Par-4 in normal and neoplastic cholangiocytes and the effects of its pharmacological or genetic modulation. The study was performed in human and rat liver, CCA patient biopsies, and two CCA cell lines. PAR-4 was expressed in normal rat and human cholangiocytes, but its expression levels decreased in both human CCA and CCA cell lines. In both intrahepatic and extrahepatic CCA, Par-4 expression (as shown by immunohistochemistry) was inversely correlated with markers of proliferation (eg, proliferating cellular nuclear antigen) and directly correlated with apoptotic markers (eg, Bax and Bax/BCL2 ratio). Par-4 expression was decreased during CCA cell proliferation but was enhanced after apoptosis induction. Pharmacological induction of Par-4 expression in CCA cell lines by diindolymethane or withaferin A promoted activation of apoptosis and inhibition of proliferation. In contrast, specific Par-4 silencing by small-interfering RNA determined activation of CCA cell line proliferation. Par-4 is expressed in rat and human cholangiocytes and is down-regulated in both human CCA and CCA cell lines. Par-4 protein levels decrease during cell proliferation but increase during apoptosis. Pharmacological or genetic induction of Par-4 determines apoptosis of CCA cells, suggesting Par-4 targeting as a CCA treatment strategy.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/drug effects , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Gene Expression Regulation/drug effects , Liver/metabolism , Aged , Animals , Blotting, Western , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cholangiocarcinoma/drug therapy , Down-Regulation , Female , Humans , Immunoenzyme Techniques , Indoles/pharmacology , Male , Middle Aged , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Withanolides/pharmacology
8.
Dig Liver Dis ; 42(4): 253-60, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20097142

ABSTRACT

Cholangiocarcinoma is commonly considered a rare cancer. However, if we consider the hepato-biliary system a single entity, cancers of the gallbladder, intra-hepatic and extra-hepatic biliary tree altogether represent approximately 30% of the total with incidence rates close to that of hepatocellular carcinoma, which is the third most common cause of cancer-related death worldwide. In addition, cholangiocarcinoma is characterized by a very poor prognosis and virtually no response to chemotherapeutics; radical surgery, the only effective treatment, is not frequently applicable because late diagnosis. Biomarkers for screening programs and for follow-up of categories at risk are under investigation, however, currently none of the proposed markers has reached clinical application. For all these considerations, cancers of the biliary tree system should merit much more scientific attention also because a progressive increase in incidence and mortality for these cancers has been reported worldwide. This manuscript deals with the most recent advances in the epidemiology, biology and clinical presentation of cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms , Bile Ducts, Intrahepatic , Cholangiocarcinoma , Bile Duct Neoplasms/diagnosis , Bile Duct Neoplasms/epidemiology , Bile Duct Neoplasms/etiology , Bile Duct Neoplasms/therapy , Biomarkers , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/epidemiology , Cholangiocarcinoma/etiology , Cholangiocarcinoma/therapy , Humans , Risk Factors
9.
Dig Liver Dis ; 42(5): 377-85, 2010 May.
Article in English | MEDLINE | ID: mdl-19897428

ABSTRACT

BACKGROUND: Polycystin-1 and -2 (PC-1 and PC-2) are critical components of primary cilia, which act as mechanosensors and drive cell response to injury. PC-1 activation involves the cleavage/processing of PC-1 cytoplasmic tail, driven by regulated intramembrane proteolysis or ubiquitine/proteasome, translocation in the nucleus and activation of transcription factors. Mutations of PC-1 or PC-2 occur in polycystic liver where cholangiocyte proliferation is enhanced. AIM: We evaluated the involvement of PC-1 and PC-2 in modulating cholangiocyte proliferation. METHODS: We investigated rat cholangiocytes induced to proliferate by 17beta-oestradiol. Proliferation was evaluated by PCNA immunoblotting or [(3)H]-thymidine incorporation into DNA. PC-1 silencing was performed by siRNA, while inhibition of regulated intramembrane proteolysis or proteasome by gamma-secretase inhibitor, leupeptin or MG115. RESULTS: Cholangiocyte proliferation was associated with decreased PC-1 and PC-2 expression, which was inversely correlated with enhanced PCNA. The selective silencing of PC-1 induced activation of cholangiocyte proliferation in association with decreased PC-1 expression. Two different regulated intramembrane proteolysis inhibitors, gamma-secretase-inhibitor and leupeptin, and the proteasome inhibitor, MG115, abolished the 17beta-oestradiol proliferative effect. CONCLUSIONS: PC-1 and PC-2 play a major role as modulators of cholangiocyte proliferation suggesting that primary cilia may act as sensors of cell injury driving, when activated, a proliferative cholangiocyte response to trigger the reparative processes.


Subject(s)
Bile Ducts, Intrahepatic/cytology , Cell Proliferation , TRPP Cation Channels/physiology , Animals , Cells, Cultured , Male , Rats , Rats, Wistar
10.
Lab Invest ; 88(9): 986-94, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18607346

ABSTRACT

A 'locally acting' IGF1 (insulin-like growth factor 1) isoform has been recently identified in the skeletal muscle and neural tissues where it accelerates injury repair. No information exist on the expression and function of IGF1 isoforms in the liver. We investigated IGF1 isoforms in rat hepatocytes and cholangiocytes and evaluated their involvement in cell proliferation or damage induced by experimental cholestasis (bile duct ligation, BDL) or hydrophobic bile salts. IGF1 isoforms were analyzed by real-time PCR by using beta-actin as internal reference. In both hepatocytes and cholangiocytes, the 'locally acting' IGF1 isoform (XO6108) and 'circulating' IGF1 isoform (NM_178866) represented respectively 44 and 52% of the total IGF1. Basal mRNAs for both 'locally acting' and 'circulating' IGF1 isoforms were higher (P<0.05) in hepatocytes than cholangiocytes. After BDL for 3 h, the 'locally acting' IGF1 isoform decreased threefold (P<0.05) in hepatocytes but remained stable in cholangiocytes with respect to sham-controls. After 1 week of BDL, hepatocytes displayed a further fivefold decrease of 'locally acting' IGF1 mRNA. In contrast, cholangiocytes showed an eightfold increase of the 'locally acting' IGF1 mRNA. The effect of 3 h of BDL on IGF1 isoforms was reproduced in vitro by incubation with glycochenodeoxycholate (GCDC). The cytotoxic effects (inhibition of proliferation and induction of apoptosis) of GCDC on isolated cholangiocytes were more pronounced after selective silencing (SiRNA) of 'locally acting' than 'circulating' IGF1 isoform. Rat hepatocytes and cholangiocytes express the 'locally acting' IGF1 isoform, which decreased during cell damage and increased during cell proliferation. The 'locally acting' IGF1 was more active than the 'circulating' isoform in protecting cholangiocytes from GCDC-induced cytotoxicity. These findings indicate that, besides muscle and neural tissues, also in liver cells the 'locally acting' IGF1 isoform is important in modulating response to damage.


Subject(s)
Biliary Tract/metabolism , Cholestasis/prevention & control , Hepatocytes/metabolism , Insulin-Like Growth Factor I/physiology , Isoenzymes/physiology , Animals , Apoptosis , Base Sequence , Biliary Tract/cytology , Blotting, Western , Cell Proliferation , Cells, Cultured , DNA Primers , Gene Silencing , Hepatocytes/cytology , Insulin-Like Growth Factor I/chemistry , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Isoenzymes/chemistry , Isoenzymes/metabolism , Male , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
11.
Am J Pathol ; 172(2): 321-32, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202196

ABSTRACT

We evaluated the morphological and functional features of hepatic cyst epithelium in adult autosomal dominant polycystic kidney disease (ADPKD). In six ADPKD patients, we investigated the morphology of cyst epithelium apical surface by scanning electron microscopy and the expression of estrogen receptors (ERs), insulin-like growth factor 1 (IGF1), IGF1 receptors (IGF1-R), growth hormone receptor, the proliferation marker proliferating cell nuclear antigen, and pAKT by immunohistochemistry and immunofluorescence. Proliferation of liver cyst-derived epithelial cells was evaluated by both MTS proliferation assay and [(3)H]thymidine incorporation into DNA. The hepatic cyst epithelium displayed heterogeneous features, being normal in small cysts (<1 cm), characterized by rare or shortened cilia in 1- to 3-cm cysts, and exhibiting the absence of both primary cilia and microvilli in large cysts (>3 cm). Cyst epithelium showed marked immunohistochemical expression of ER, growth hormone receptor, IGF1, IGF1-R, proliferating cell nuclear antigen, and pAKT. IGF1 was 10-fold more enriched in the hepatic cyst fluid than in serum. Serum-deprived liver cyst-derived epithelial cells proliferated when exposed to 17beta-estradiol and IGF1 and when exposed to human cyst fluid. ER or IGF1-R antagonists inhibited the proliferative effect of serum readmission, cyst fluid, 17beta-estradiol, and IGF1. Our findings could explain the role of estrogens in accelerating the progression of ADPKD and may suggest a potential benefit of therapeutic strategies based on estrogen antagonism.


Subject(s)
Cysts/ultrastructure , Epithelium/ultrastructure , Liver Diseases/pathology , Polycystic Kidney Diseases/complications , Aged , Blotting, Western , Cell Proliferation , Cilia/ultrastructure , Cyst Fluid/metabolism , Cysts/etiology , Epithelium/metabolism , Estradiol/pharmacology , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/pharmacology , Liver Diseases/etiology , Male , Microscopy, Electron, Scanning , Middle Aged , Polycystic Kidney Diseases/metabolism , Receptor, IGF Type 1/antagonists & inhibitors , Receptors, Estrogen/antagonists & inhibitors
12.
Environ Toxicol ; 23(1): 44-51, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18214926

ABSTRACT

On April 1998 a tailing dam of the Aznalcollar pyrite mine partially collapsed and released to the Guadiamar river acidic water and mud containing toxic metals threatening the Doñana National Park, a Spanish wildlife reserve located near the estuary of Guadalquivir river. To assess the possible biological effects on terrestrial ecosystems, biochemical markers were assayed in the kidneys of Algerian mice (Mus spretus) collected in several areas of Doñana and Guadiamar river. Biomarkers assayed are proteins involved in cell cycle regulation, in particular cyclins and their associated kinases, and some cell cycle inhibitors. Moreover Mitogen Activated Protein Kinases (MAPK), a signal transduction system involved in cell division, p53, a protein involved in growth arrest after DNA damage, and HSP70, an early stress-induced protein, were assayed. The kidneys of animals collected one year after the ecological disaster had increased levels of PCNA (proliferating cell nuclear antigen), indicating an increased number of cells in the S phase of cell cycle. This shift of cells from G0 to S phase is due to increased levels of cyclins D1, E, and A, to decreased levels of p21 and p27 cdk inhibitors, and to activation of MAPK cascade. On the other hand, p53 and HSP70 levels are not changed. These data demonstrate that the presence of toxic metals after ecological disaster provoked the induction of kidney cell proliferation interpretable as a compensatory cell growth after tissue damage and apoptosis, and that could lead to the genomic instability characteristic of cancer cell.


Subject(s)
Cell Cycle Proteins/drug effects , Environmental Exposure , Environmental Pollutants/toxicity , Kidney/drug effects , Metals, Heavy/toxicity , Muridae/genetics , Animals , Animals, Wild , Biomarkers , Cell Cycle Proteins/analysis , Environmental Monitoring , Kidney/cytology , Mice , Mining , Rivers , Spain
SELECTION OF CITATIONS
SEARCH DETAIL
...