201
|
Muñoz-Gámez J, Quiles-Pérez R, Ruiz-Extremera A, Martín-Álvarez A, Sanjuan-Nuñez L, Carazo A, León J, Oliver F, Salmerón J. Inhibition of poly (ADP-ribose) polymerase-1 enhances doxorubicin activity against liver cancer cells. Cancer Lett 2011; 301:47-56. [DOI: 10.1016/j.canlet.2010.10.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 10/26/2010] [Accepted: 10/28/2010] [Indexed: 11/30/2022]
|
202
|
Franco DL, Mainez J, Vega S, Sancho P, Murillo MM, de Frutos CA, Del Castillo G, López-Blau C, Fabregat I, Nieto MA. Snail1 suppresses TGF-beta-induced apoptosis and is sufficient to trigger EMT in hepatocytes. J Cell Sci 2011; 123:3467-77. [PMID: 20930141 DOI: 10.1242/jcs.068692] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although TGF-β suppresses early stages of tumour development, it later contributes to tumour progression when cells become resistant to its suppressive effects. In addition to circumventing TGF-β-induced growth arrest and apoptosis, malignant tumour cells become capable of undergoing epithelial-to-mesenchymal transition (EMT), favouring invasion and metastasis. Therefore, defining the mechanisms that allow cancer cells to escape from the suppressive effects of TGF-β is fundamental to understand tumour progression and to design specific therapies. Here, we have examined the role of Snail1 as a suppressor of TGF-β-induced apoptosis in murine non-transformed hepatocytes, rat and human hepatocarcinoma cell lines and transgenic mice. We show that Snail1 confers resistance to TGF-β-induced cell death and that it is sufficient to induce EMT in adult hepatocytes, cells otherwise refractory to this transition upon exposure to TGF-β. Furthermore, we show that Snail1 silencing prevents EMT and restores the cell death response induced by TGF-β. As Snail1 is a known target of TGF-β signalling, our data indicate that Snail1 might transduce the tumour-promoting effects of TGF-β, namely the EMT concomitant with the resistance to cell death.
Collapse
Affiliation(s)
- D Lorena Franco
- Instituto de Neurociencias (CSIC-UMH), 03550 San Juan de Alicante, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Maillard E. [Epidemiology, natural history and pathogenesis of hepatocellular carcinoma]. Cancer Radiother 2011; 15:3-6. [PMID: 21239205 DOI: 10.1016/j.canrad.2010.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 08/04/2010] [Accepted: 11/04/2010] [Indexed: 01/16/2023]
Abstract
Hepatocellular carcinoma (HCC) is the main type of primary liver cancers and the third most common cause of cancer mortality worldwide. In France, rising number between 5000 and 6000 cases are diagnosed each year. The major risk factor for hepatocellular carcinoma is chronic hepatitis: viral hepatitis B, viral hepatitis C, consumption of alcohol, hemochromatosis. Hepatocellular carcinoma is closely associated to liver cirrhosis, which is a true precancerous state. Because hepatocarcinogenesis is a long and heterogeneous process, there is still much to understand. Many genetic and epigenetic alterations are described leading to changes in cellular signalling cascades involved in regulation of growth, differentiation, apoptosis, motility. Hepatitis viruses play a direct oncogenic role through the interaction between viral and cellular proteins, which control cell homeostasis, or by the integration of hepatitis B virus genome into the host genome. Furthermore, hepatitis viruses play an indirect oncogenic role by causing chronic inflammation and hepatocyte regeneration related to viral hepatopathy. In expectation of a better understanding of hepatocarcinogenesis and new treatments, prevention from risk factors and ultrasonographic screening of patients with cirrhosis should increase prognosis.
Collapse
Affiliation(s)
- E Maillard
- Groupement hospitalier Nord, hospices civils de Lyon, hôpital de l'Hôtel-Dieu, 1 place de l'Hôpital, Lyon, France.
| |
Collapse
|
204
|
Waters MD, Jackson M, Lea I. Characterizing and predicting carcinogenicity and mode of action using conventional and toxicogenomics methods. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2010; 705:184-200. [DOI: 10.1016/j.mrrev.2010.04.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2010] [Revised: 04/07/2010] [Accepted: 04/08/2010] [Indexed: 01/10/2023]
|
205
|
Sánchez A, Fabregat I. Growth factor- and cytokine-driven pathways governing liver stemness and differentiation. World J Gastroenterol 2010; 16:5148-61. [PMID: 21049549 PMCID: PMC2975086 DOI: 10.3748/wjg.v16.i41.5148] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Liver is unique in its capacity to regenerate in response to injury or tissue loss. Hepatocytes and other liver cells are able to proliferate and repopulate the liver. However, when this response is impaired, the contribution of hepatic progenitors becomes very relevant. Here, we present an update of recent studies on growth factors and cytokine-driven intracellular pathways that govern liver stem/progenitor cell expansion and differentiation, and the relevance of these signals in liver development, regeneration and carcinogenesis. Tyrosine kinase receptor signaling, in particular, c-Met, epidermal growth factor receptors or fibroblast growth factor receptors, contribute to proliferation, survival and differentiation of liver stem/progenitor cells. Different evidence suggests a dual role for the transforming growth factor (TGF)-β signaling pathway in liver stemness and differentiation. On the one hand, TGF-β mediates progression of differentiation from a progenitor stage, but on the other hand, it contributes to the expansion of liver stem cells. Hedgehog family ligands are necessary to promote hepatoblast proliferation but need to be shut off to permit subsequent hepatoblast differentiation. In the same line, the Wnt family and β-catenin/T-cell factor pathway is clearly involved in the maintenance of liver stemness phenotype, and its repression is necessary for liver differentiation during development. Collectively, data indicate that liver stem/progenitor cells follow their own rules and regulations. The same signals that are essential for their activation, expansion and differentiation are good candidates to contribute, under adequate conditions, to the paradigm of transformation from a pro-regenerative to a pro-tumorigenic role. From a clinical perspective, this is a fundamental issue for liver stem/progenitor cell-based therapies.
Collapse
|
206
|
Hagymási K, Tulassay Z. [New possibilities of targeted therapy in the treatment of hepatocellular carcinoma with the help of molecular biology]. Orv Hetil 2010; 151:1763-1768. [PMID: 20940115 DOI: 10.1556/oh.2010.28984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hepatocellular carcinoma (HCC) has a poor prognosis. Approximately 85% of patients are not candidates for curative treatments at the time of diagnosis; hence palliative modalities (transcatheter arterial chemoembolisation, radiofrequency ablation, systemic chemotherapy) are used. Systemic chemotherapies have disappointing results. The increasing knowledge in the molecular biology of HCC will increase the possibilities of targeted therapy. The multi-tyrosine kinase inhibitor sorafenib is the only drug which has approved. The VEGF-inhibitors (bevacizumab, sunitinib), EGFR-blocker agents (erlotinib), as well as the inhibition of mTOR (rapamycin) are promising. Combination of sorafenib or other anti-angiogenic agents with local ablative procedure (transcatheter arterial chemoembolisation, radiofrequency ablation), or with curative hepatectomy also can be favorable. Alteration of Wnt pathway, retinoid compounds, inhibition of the cell cycle as well as the proteosome, and epigenetic therapy can be other potential promising targets in HCC.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Benzenesulfonates/pharmacology
- Bevacizumab
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/surgery
- Catheter Ablation
- Cell Cycle/drug effects
- Chemoembolization, Therapeutic/methods
- Chemotherapy, Adjuvant
- Epigenesis, Genetic/drug effects
- ErbB Receptors/antagonists & inhibitors
- Erlotinib Hydrochloride
- Gene Expression Regulation, Neoplastic
- Hepatectomy
- Humans
- Indoles/pharmacology
- Intracellular Signaling Peptides and Proteins/antagonists & inhibitors
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/surgery
- Niacinamide/analogs & derivatives
- Phenylurea Compounds
- Proteasome Endopeptidase Complex/drug effects
- Protein Kinase Inhibitors/pharmacology
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Pyridines/pharmacology
- Pyrroles/pharmacology
- Quinazolines/pharmacology
- Signal Transduction/drug effects
- Sirolimus/pharmacology
- Sorafenib
- Sunitinib
- TOR Serine-Threonine Kinases
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Wnt Proteins/metabolism
Collapse
Affiliation(s)
- Krisztina Hagymási
- Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest.
| | | |
Collapse
|
207
|
Li YH, Chen MHC, Gong HY, Hu SY, Li YW, Lin GH, Lin CC, Liu W, Wu JL. Progranulin A-mediated MET signaling is essential for liver morphogenesis in zebrafish. J Biol Chem 2010; 285:41001-9. [PMID: 20961855 DOI: 10.1074/jbc.m110.138743] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The mechanism that regulates embryonic liver morphogenesis remains elusive. Progranulin (PGRN) is postulated to play a critical role in regulating pathological liver growth. Nevertheless, the exact regulatory mechanism of PGRN in relation to its functional role in embryonic liver development remains to be elucidated. In our study, the knockdown of progranulin A (GrnA), an orthologue of mammalian PGRN, using antisense morpholinos resulted in impaired liver morphogenesis in zebrafish (Danio rerio). The vital role of GrnA in hepatic outgrowth and not in liver bud formation was further confirmed using whole-mount in situ hybridization markers. In addition, a GrnA deficiency was also found to be associated with the deregulation of MET-related genes in the neonatal liver using a microarray analysis. In contrast, the decrease in liver size that was observed in grnA morphants was avoided when ectopic MET expression was produced by co-injecting met mRNA and grnA morpholinos. This phenomenon suggests that GrnA might play a role in liver growth regulation via MET signaling. Furthermore, our study has shown that GrnA positively modulates hepatic MET expression both in vivo and in vitro. Therefore, our data have indicated that GrnA plays a vital role in embryonic liver morphogenesis in zebrafish. As a result, a novel link between PGRN and MET signaling is proposed.
Collapse
Affiliation(s)
- Yen-Hsing Li
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung Hsing University and Academia Sinica, Taipei 115, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Rao SK, Pavicevic Z, Du Z, Kim JG, Fan M, Jiao Y, Rosebush M, Samant S, Gu W, Pfeffer LM, Nosrat CA. Pro-inflammatory genes as biomarkers and therapeutic targets in oral squamous cell carcinoma. J Biol Chem 2010; 285:32512-21. [PMID: 20702412 PMCID: PMC2952253 DOI: 10.1074/jbc.m110.150490] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 07/14/2010] [Indexed: 12/20/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a major health problem worldwide, and patients have a particularly poor 5-year survival rate. Thus, identification of the molecular targets in OSCC and subsequent innovative therapies are greatly needed. Prolonged exposure to alcohol, tobacco, and pathogenic agents are known risk factors and have suggested that chronic inflammation may represent a potential common denominator in the development of OSCC. Microarray analysis of gene expression in OSCC cell lines with high basal NF-κB activity and OSCC patient samples identified dysregulation of many genes involved in inflammation, wound healing, angiogenesis, and growth regulation. In particular IL-8, CCL5, STAT1, and VEGF gene expression was up-regulated in OSCC. Moreover, IL-8 protein levels were significantly higher in OSCC cell lines as compared with normal human oral keratinocytes. Targeting IL-8 expression by siRNA significantly reduced the survival of OSCC cells, indicating that it plays an important role in OSCC development and/or progression. Inhibiting the inflammatory pathway by aspirin and the proteasome/NF-κB pathway by bortezomib resulted in marked reduction in cell viability in OSCC lines. Taken together our studies indicate a strong link between inflammation and OSCC development and reveal IL-8 as a potential mediator. Treatment based on prevention of general inflammation and/or the NF-κB pathway shows promise in OSCCs.
Collapse
Affiliation(s)
- Shailaja K. Rao
- From the Departments of Bioscience Research and
- Center for Integrative Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Zoran Pavicevic
- From the Departments of Bioscience Research and
- Center for Integrative Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Ziyun Du
- Departments of Pathology and Laboratory Medicine
- Center for Integrative Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | | | - Meiyun Fan
- Departments of Pathology and Laboratory Medicine
- Center for Integrative Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Yan Jiao
- Orthopedic Surgery, College of Medicine, and
| | | | | | - Weikuan Gu
- Orthopedic Surgery, College of Medicine, and
| | - Lawrence M. Pfeffer
- Departments of Pathology and Laboratory Medicine
- Center for Integrative Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Christopher A. Nosrat
- From the Departments of Bioscience Research and
- Center for Integrative Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| |
Collapse
|
209
|
Abstract
How liver adjusts and stabilizes its size is unsolved so far; the answers to this question may also provide insights into mechanisms of hepatocarcinogenesis. Two recent papers suggest a role for Merlin/Nf2 in control of liver cell turnover, but results appear conflicting at first glance.
Collapse
Affiliation(s)
- Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | | |
Collapse
|
210
|
Abstract
BACKGROUND The hepatocellular carcinoma is one of the most common malignant tumors and carries a poor survival rate. The management of patients at risk for developing HCC remains intricate. METHODS A literature search identified potential markers for hepatocellular carcinoma. These markers were analysed and justification was provided for these factors' inclusion to (or exclusion from) the markers of hepatocellular carcinoma (HCC). A search of the literature was made using cancer literature and the PubMed database for the following keywords: "markers and HCC," "Lens culinaris agglutinin reactive AFP (AFP-L3) and HCC," "Des-γ-carboxy prothrombin (DCP) and HCC," "Glypican-3 and HCC," "Chromogranin A and HCC," "Transforming growth factor β1(TGF) and HCC," "α-l-fucosidase (AFU) and HCC," "Golgi protein-73 (GP73) and HCC," "Hepatocyte growth factor (HGF) and HCC," "Nervous growth factor (NGF) and HCC." CONCLUSIONS Despite the large number of studies devoted to the immunohistochemistry of HCC, at the present time, the absolute positive and negative markers for HCC are still lacking, and even those characterized by very high sensitivity and specificity do not have an universal diagnostic usefulness. Given the poor response to current therapies, a better understanding of the molecular pathways active in this disease could potentially provide new targets for therapy. However, AFP shows a low sensitivity, therefore other biomarkers have been developed to make an early diagnosis and improve patients' prognosis.
Collapse
|
211
|
Charette N, De Saeger C, Lannoy V, Horsmans Y, Leclercq I, Stärkel P. Salirasib inhibits the growth of hepatocarcinoma cell lines in vitro and tumor growth in vivo through ras and mTOR inhibition. Mol Cancer 2010; 9:256. [PMID: 20860815 PMCID: PMC2955616 DOI: 10.1186/1476-4598-9-256] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 09/22/2010] [Indexed: 03/01/2023] Open
Abstract
Background Dysregulation of epidermal growth factor and insulin-like growth factor signaling play important roles in human hepatocellular carcinoma (HCC), leading to frequent activation of their downstream targets, the ras/raf/extracellular signal-regulated kinase (ERK) and the phosphoinositide 3-kinase (PI3K)/Akt/mammalian Target of Rapamycin (mTOR) pathways. Salirasib is an S-prenyl-cysteine analog that has been shown to block ras and/or mTOR activation in several non hepatic tumor cell lines. We investigated in vitro the effect of salirasib on cell growth as well as its mechanism of action in human hepatoma cell lines (HepG2, Huh7, and Hep3B) and its in vivo effect in a subcutaneous xenograft model with HepG2 cells. Results Salirasib induced a time and dose dependent growth inhibition in hepatocarcinoma cells through inhibition of proliferation and partially through induction of apoptosis. A 50 percent reduction in cell growth was obtained in all three cell lines at a dose of 150 μM when they were cultured with serum. By contrast, salirasib was more potent at reducing cell growth after stimulation with EGF or IGF2 under serum-free conditions, with an IC50 ranging from 60 μM to 85 μM. The drug-induced anti-proliferative effect was associated with downregulation of cyclin A and to a lesser extent of cyclin D1, and upregulation of p21 and p27. Apoptosis induction was related to a global pro-apoptotic balance with caspase 3 activation, cytochrome c release, death receptor upregulation, and a reduced mRNA expression of the apoptosis inhibitors cFLIP and survivin. These effects were associated with ras downregulation and mTOR inhibition, without reduction of ERK and Akt activation. In vivo, salirasib reduced tumour growth from day 5 onwards. After 12 days of treatment, mean tumor weight was diminished by 56 percent in the treated animals. Conclusions Our results show for the first time that salirasib inhibits the growth of human hepatoma cell lines through inhibition of proliferation and induction of apoptosis, which is associated with ras and mTOR inhibition. The therapeutic potential of salirasib in human HCC was further confirmed in a subcutaneous xenograft model.
Collapse
Affiliation(s)
- Nicolas Charette
- Laboratory of Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
212
|
Ding W, You H, Dang H, LeBlanc F, Galicia V, Lu SC, Stiles B, Rountree CB. Epithelial-to-mesenchymal transition of murine liver tumor cells promotes invasion. Hepatology 2010; 52:945-53. [PMID: 20564331 PMCID: PMC3032356 DOI: 10.1002/hep.23748] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED Epithelial-to-mesenchymal transition (EMT) is predicted to play a critical role in metastatic disease in hepatocellular carcinoma. In this study, we used a novel murine model of EMT to elucidate a mechanism of tumor progression and metastasis. A total of 2 x 10(6) liver cells isolated from Pten(loxp/loxp)/Alb-Cre(+) mice, expanded from a single CD133(+)CD45(-) cell clone, passage 0 (P0), were sequentially transplanted to obtain two passages of tumor cells, P1 and P2. Cells were analyzed for gene expression using microarray and real-time polymerase chain reaction. Functional analysis included cell proliferation, migration, and invasion in vitro and orthotopic tumor metastasis assays in vivo. Although P0, P1, and P2 each formed tumors consistent with mixed liver epithelium, within the P2 cells, two distinct cell types were clearly visible: cells with epithelial morphology similar to P0 cells and cells with fibroblastoid morphology. These P2 mesenchymal cells demonstrated increased locomotion on wound healing; increased cell invasion on Matrigel basement membrane; increased EMT-associated gene expression of Snail1, Zeb1, and Zeb2; and down-regulated E-cadherin. P2 mesenchymal cells demonstrated significantly faster tumor growth in vivo compared with P2 epithelial counterparts, with invasion of intestine, pancreas, spleen, and lymph nodes. Furthermore, P2 mesenchymal cells secreted high levels of hepatocyte growth factor (HGF), which we propose acts in a paracrine fashion to drive epithelial cells to undergo EMT. In addition, a second murine liver cancer stem cell line with methionine adenosyltransferase 1a deficiency acquired EMT after sequential transplantations, indicating that EMT was not restricted to Pten-deleted tumors. CONCLUSION EMT is associated with a high rate of liver tumor proliferation, invasion, and metastasis in vivo, which is driven by HGF secreted from mesenchymal tumor cells in a feed-forward mechanism.
Collapse
Affiliation(s)
- Wei Ding
- Department of Pediatrics and Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA
| | - Hanning You
- Department of Pediatrics and Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA
| | - Hien Dang
- Department of Pediatrics and Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA
| | - Francis LeBlanc
- Department of Pediatrics and Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA
| | - Vivian Galicia
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA
| | - Shelly C. Lu
- Division of Gastroenterology and Liver Diseases, USC Research Center for Liver Disease, The Southern California Research Center for ALPD and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Bangyan Stiles
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA
| | - C. Bart Rountree
- Department of Pediatrics and Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA
| |
Collapse
|
213
|
Diabetes mellitus impairs the response to intra-arterial chemotherapy in hepatocellular carcinoma. Med Oncol 2010; 28:1080-8. [DOI: 10.1007/s12032-010-9650-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 08/03/2010] [Indexed: 12/15/2022]
|
214
|
Benhamouche S, Curto M, Saotome I, Gladden AB, Liu CH, Giovannini M, McClatchey AI. Nf2/Merlin controls progenitor homeostasis and tumorigenesis in the liver. Genes Dev 2010; 24:1718-30. [PMID: 20675406 PMCID: PMC2922501 DOI: 10.1101/gad.1938710] [Citation(s) in RCA: 219] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2010] [Accepted: 06/18/2010] [Indexed: 12/13/2022]
Abstract
The molecular signals that control the maintenance and activation of liver stem/progenitor cells are poorly understood, and the role of liver progenitor cells in hepatic tumorigenesis is unclear. We report here that liver-specific deletion of the neurofibromatosis type 2 (Nf2) tumor suppressor gene in the developing or adult mouse specifically yields a dramatic, progressive expansion of progenitor cells throughout the liver without affecting differentiated hepatocytes. All surviving mice eventually developed both cholangiocellular and hepatocellular carcinoma, suggesting that Nf2(-/-) progenitors can be a cell of origin for these tumors. Despite the suggested link between Nf2 and the Hpo/Wts/Yki signaling pathway in Drosophila, and recent studies linking the corresponding Mst/Lats/Yap pathway to mammalian liver tumorigenesis, our molecular studies suggest that Merlin is not a major regulator of YAP in liver progenitors, and that the overproliferation of Nf2(-/-) liver progenitors is instead driven by aberrant epidermal growth factor receptor (EGFR) activity. Indeed, pharmacologic inhibition of EGFR blocks the proliferation of Nf2(-/-) liver progenitors in vitro and in vivo, consistent with recent studies indicating that the Nf2-encoded protein Merlin can control the abundance and signaling of membrane receptors such as EGFR. Together, our findings uncover a critical role for Nf2/Merlin in controlling homeostasis of the liver stem cell niche.
Collapse
Affiliation(s)
- Samira Benhamouche
- Department of Pathology, Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Marcello Curto
- Department of Pathology, Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Ichiko Saotome
- Department of Pathology, Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Andrew B. Gladden
- Department of Pathology, Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Ching-Hui Liu
- Department of Pathology, Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Marco Giovannini
- Center for Neural Tumor Research, House Ear Institute, Los Angeles California 90057, USA
| | - Andrea I. McClatchey
- Department of Pathology, Massachusetts General Hospital Center for Cancer Research, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| |
Collapse
|
215
|
Baek JY, Morris SM, Campbell J, Fausto N, Yeh MM, Grady WM. TGF-beta inactivation and TGF-alpha overexpression cooperate in an in vivo mouse model to induce hepatocellular carcinoma that recapitulates molecular features of human liver cancer. Int J Cancer 2010; 127:1060-71. [PMID: 20020490 DOI: 10.1002/ijc.25127] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hepatocellular carcinoma (HCC) results from the cumulative effects of deregulated tumor suppressor genes and oncogenes. The tumor suppressor and oncogenes commonly affected include growth factors, receptors and their downstream signaling pathway components. The overexpression of transforming growth factor alpha (TGF-alpha) and the inhibition of TGF-beta signaling are especially common in human liver cancer. Thus, we assessed whether TGF-alpha overexpression and TGF-beta signaling inactivation cooperate in hepatocarcinogenesis using an in vivo mouse model, MT1/TGFa;AlbCre/Tgfbr2(flx/flx) mice ("TGFa;Tgfbr2(hepko)"), which overexpresses TGF-alpha and lacks a TGF-beta receptor in the liver. TGF-beta signaling inactivation did not alter the frequency or number of cancers in mice with overexpression of TGF-alpha. However, the tumors in the TGFa;Tgfbr2(hepko) mice displayed increased proliferation and increased cdk2, cyclin E and cyclin A expression as well as decreased Cdkn1a/p21 expression compared to normal liver and compared to the cancers arising in the TGF-alpha overexpressing mice with intact TGF-beta receptors. Increased phosphorylated ERK1/2 expression was also present in the tumors from the TGFa;Tgfbr2(hepko) mice and correlated with downregulated Raf kinase inhibitor protein expression, which is a common molecular event in human HCC. Thus, TGF-beta signaling inactivation appears to cooperate with TGF-alpha in vivo to promote the formation of liver cancer that recapitulates molecular features of human HCC.
Collapse
Affiliation(s)
- Ji Yeon Baek
- Center for Colorectal Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | | | | | | | | | | |
Collapse
|
216
|
Involvement of Hepatopoietin Cn in the development of human hepatocellular carcinoma. Clin Exp Metastasis 2010; 27:571-80. [PMID: 20683644 DOI: 10.1007/s10585-010-9346-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 07/15/2010] [Indexed: 01/12/2023]
|
217
|
Molecular targeted therapy for advanced hepatocellular carcinoma: current status and future perspectives. J Gastroenterol 2010; 45:794-807. [PMID: 20567987 DOI: 10.1007/s00535-010-0270-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Accepted: 05/30/2010] [Indexed: 02/06/2023]
Abstract
Sorafenib, a multikinase inhibitor targeting vascular endothelial growth factor (VEGF)-mediated angiogenesis, is the first drug found to prolong survival of patients with advanced hepatocellular carcinoma (HCC). This advance has shifted the paradigm of systemic treatment for HCC toward molecular targeted therapy (MTT). However, the disease-stabilizing effect of VEGF signaling-targeted MTT normally lasts only for a few months, suggesting a rapid emergence of resistance in the majority of patients. To overcome the resistance to VEGF signaling-targeted MTT, strategies incorporating inhibition of either compensatory pro-angiogenic pathways or recruitment of bone marrow-derived circulating endothelial progenitors, as well as suppression of other oncogenic pathways, are currently being investigated. The combination of multiple molecular targeted agents or the use of multi-target agents may enhance the efficacy at the expense of increased toxicities. To facilitate the development of MTT for HCC, current methodologies for pharmacodynamic assessment, patient selection and target identification need to be improved. Patient selection according to the individual molecular signature of the tumor and correlative biomarker studies are encouraged while planning a clinical trial of novel MTT.
Collapse
|
218
|
He AR, Soe K, El Zouhairi M. Current problems with systemic treatment of advanced hepatocellular cancer. Curr Probl Cancer 2010; 34:131-49. [PMID: 20417353 DOI: 10.1016/j.currproblcancer.2010.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Aiwu Ruth He
- Department of Medicine, Lombardi Cancer Center, Georgetown University, Washington, DC, USA
| | | | | |
Collapse
|
219
|
Wörns MA, Galle PR. Novel inhibitors in development for hepatocellular carcinoma. Expert Opin Investig Drugs 2010; 19:615-29. [PMID: 20374038 DOI: 10.1517/13543781003767418] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE OF THE FIELD The multikinase inhibitor sorafenib was the first agent to demonstrate a survival benefit for patients with locally advanced or metastatic hepatocellular carcinoma (HCC). Although sorafenib represents a landmark in the treatment of HCC and proved molecularly targeted therapy to be effective in this disease, it represents just the first step towards an improvement in systemic therapy. Since then, novel inhibitors have been evaluated in early clinical trials, showing potential activity. AREAS COVERED IN THIS REVIEW This article aims to review novel inhibitors emerging in the field of advanced HCC. An Internet-based search was performed to identify abstracts, clinical trials ( www.clinicaltrials.gov , last accessed 30 November 2009), and original research and review articles. WHAT THE READER WILL GAIN Readers will gain a comprehensive survey of current molecularly targeted therapy approaches in advanced HCC. In addition, challenges such as the design of clinical trials, the assessment of radiological response, the role of combination therapy, and future developments in molecularly targeted therapy are discussed. TAKE HOME MESSAGE Sorafenib is the standard of care in patients with advanced HCC. However, promising novel inhibitors are under investigation. Combined molecularly targeted therapies according to an individual genomic and proteomic profiling will probably lead to more personalised medicine in advanced HCC.
Collapse
Affiliation(s)
- Marcus Alexander Wörns
- University Medicine of the Johannes Gutenberg-University Mainz, Department of Internal Medicine, Mainz, Germany.
| | | |
Collapse
|
220
|
Taniguchi CM, Winnay J, Kondo T, Bronson RT, Guimaraes AR, Aleman JO, Luo J, Stephanopoulos G, Weissleder R, Cantley LC, Kahn CR. The phosphoinositide 3-kinase regulatory subunit p85alpha can exert tumor suppressor properties through negative regulation of growth factor signaling. Cancer Res 2010; 70:5305-15. [PMID: 20530665 PMCID: PMC3204358 DOI: 10.1158/0008-5472.can-09-3399] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Phosphoinositide 3-kinase (PI3K) plays a critical role in tumorigenesis, and the PI3K p85 regulatory subunit exerts both positive and negative effects on signaling. Expression of Pik3r1, the gene encoding p85, is decreased in human prostate, lung, ovarian, bladder, and liver cancers, consistent with the possibility that p85 has tumor suppressor properties. We tested this hypothesis by studying mice with a liver-specific deletion of the Pik3r1 gene. These mice exhibited enhanced insulin and growth factor signaling and progressive changes in hepatic pathology, leading to the development of aggressive hepatocellular carcinomas with pulmonary metastases. Liver tumors that arose exhibited markedly elevated levels of phosphatidylinositol (3,4,5)-trisphosphate, along with Akt activation and decreased PTEN expression, at both the mRNA and protein levels. Together, these results substantiate the concept that the p85 subunit of PI3K has a tumor-suppressive role in the liver and possibly other tissues.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Genes, Tumor Suppressor
- Hepatitis, Chronic/genetics
- Hepatitis, Chronic/metabolism
- Hepatitis, Chronic/pathology
- Humans
- Intercellular Signaling Peptides and Proteins/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- PTEN Phosphohydrolase/biosynthesis
- PTEN Phosphohydrolase/genetics
- Phosphatidylinositol 3-Kinases/biosynthesis
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphatidylinositol Phosphates/metabolism
- Protein Subunits
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- Up-Regulation
Collapse
Affiliation(s)
- Cullen M. Taniguchi
- Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Jonathon Winnay
- Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Tatsuya Kondo
- Department of Metabolic Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Alexander R. Guimaraes
- Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital, Boston, MA 02114
| | - José O. Aleman
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Ji Luo
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02215 02115
| | - Gregory Stephanopoulos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Ralph Weissleder
- Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital, Boston, MA 02114
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02215 02115
| | - Lewis C. Cantley
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, MA
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02215 02115
| | - C. Ronald Kahn
- Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, 02215, USA
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
221
|
Zender L, Villanueva A, Tovar V, Sia D, Chiang DY, Llovet JM. Cancer gene discovery in hepatocellular carcinoma. J Hepatol 2010; 52:921-9. [PMID: 20385424 PMCID: PMC2905725 DOI: 10.1016/j.jhep.2009.12.034] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 11/30/2009] [Accepted: 12/31/2009] [Indexed: 12/18/2022]
Abstract
Hepatocellular carcinoma (HCC) is a deadly cancer, whose incidence is increasing worldwide. Albeit the main risk factors for HCC development have been clearly identified, such as hepatitis B and C virus infection and alcohol abuse, there is still preliminary understanding of the key drivers of this malignancy. Recent data suggest that genomic analysis of cirrhotic tissue - the pre-neoplastic carcinogenic field - may provide a read-out to identify at risk populations for cancer development. Given this contextual complexity, it is of utmost importance to characterize the molecular pathogenesis of this disease, and pinpoint the dominant pathways/drivers by integrative oncogenomic approaches and/or sophisticated experimental models. Identification of the dominant proliferative signals and key aberrations will allow for a more personalized therapy. Pathway-based approaches and functional experimental studies have aided in identifying the activation of different signaling cascades in HCC (e.g. epidermal growth factor, insulin-like growth factor, RAS, MTOR, WNT-betacatenin, etc.). However, the introduction of new high-throughput genomic technologies (e.g. microarrays, deep sequencing, etc.), and increased sophistication of computational biology (e.g. bioinformatics, biomodeling, etc.), opens the field to new strategies in oncogene and tumor suppressor discovery. These oncogenomic approaches are framed within emerging new disciplines such as systems biology, which integrates multiple inputs to explain cancer onset and progression. In addition, the consolidation of sophisticated animal models, such as mosaic cancer mouse models or the use of transposons for mutagenesis screens, have been instrumental for the identification of novel tumor drivers. We herein review some classical as well as some recent fast track approaches for oncogene discovery in HCC, and provide a comprehensive landscape of the currently known spectrum of molecular aberrations involved in hepatocarcinogenesis.
Collapse
Affiliation(s)
- Lars Zender
- Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
- Dept. of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover 30625, Germany
| | - Augusto Villanueva
- HCC Translational Research Laboratory, Barcelona-Clinic Liver Cancer Group, Liver Unit. Institut d'Investigacions Biomediques Agusto Pi i Sunyer [IDIBAPS], Centro de Investigación Biomédica en Red de Enferme dades Hepáticas y Digestivas [CIBEREHD], Hospital Clinic, Barcelona, 08036, Spain
| | - Victoria Tovar
- HCC Translational Research Laboratory, Barcelona-Clinic Liver Cancer Group, Liver Unit. Institut d'Investigacions Biomediques Agusto Pi i Sunyer [IDIBAPS], Centro de Investigación Biomédica en Red de Enferme dades Hepáticas y Digestivas [CIBEREHD], Hospital Clinic, Barcelona, 08036, Spain
| | - Daniela Sia
- HCC Translational Research Laboratory, Barcelona-Clinic Liver Cancer Group, Liver Unit. Institut d'Investigacions Biomediques Agusto Pi i Sunyer [IDIBAPS], Centro de Investigación Biomédica en Red de Enferme dades Hepáticas y Digestivas [CIBEREHD], Hospital Clinic, Barcelona, 08036, Spain
| | - Derek Y. Chiang
- Department of Medical Oncology and Center for Cancer Genome Discovery, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Cancer Program, The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Josep M. Llovet
- HCC Translational Research Laboratory, Barcelona-Clinic Liver Cancer Group, Liver Unit. Institut d'Investigacions Biomediques Agusto Pi i Sunyer [IDIBAPS], Centro de Investigación Biomédica en Red de Enferme dades Hepáticas y Digestivas [CIBEREHD], Hospital Clinic, Barcelona, 08036, Spain
- Division of Liver Diseases, Mount Sinai School of Medicine, New York, NY 10029, USA
- Institució Catalana de Recerca i Estudis Avançats, Catalonia, Spain
| |
Collapse
|
222
|
An Overview of Biomarkers and Molecular Signatures in HCC. Cancers (Basel) 2010; 2:809-23. [PMID: 24281095 PMCID: PMC3835106 DOI: 10.3390/cancers2020809] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 04/30/2010] [Accepted: 05/07/2010] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer mortality worldwide. Although most HCCs seem to originate from the accumulation of genetic abnormalities induced by various risk factors, underlying mechanisms of hepatocarcinogenesis remain unclear. Long-term survival of HCC patients is also poor, partly due to HCC recurrence. Although serum alpha-fetoprotein (AFP) level is a useful marker for the detection and monitoring of HCC, AFP levels may remain normal in the patients even with advanced HCC. To identify useful biomarkers for HCC, many studies have been conducted on molecular events such as genetic and epigenetic alterations, and gene expression. This review summarizes recent studies of potential molecular markers for diagnosis and monitoring metastasis or recurrence of HCC.
Collapse
|
223
|
Huynh H. Molecularly targeted therapy in hepatocellular carcinoma. Biochem Pharmacol 2010; 80:550-60. [PMID: 20371362 DOI: 10.1016/j.bcp.2010.03.034] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 03/16/2010] [Accepted: 03/29/2010] [Indexed: 01/02/2023]
Abstract
With an annual incidence of over 660,000 deaths, hepatocellular carcinoma (HCC) is the third leading cause of cancer death globally. This disease is often diagnosed at an advanced stage, when potentially curative therapies are not feasible. HCC is highly resistant to conventional systemic therapies and prognosis for advanced HCC patients remains poor. Given the clear need, clinical development of novel therapeutic agents in HCC has begun in earnest. Our recent knowledge of the molecular mechanisms responsible of tumor initiation and progression has identified several potential molecular targets in HCC. These targets are the receptor tyrosine kinase-activated pathways, which include the Raf/MEK/ERK, PI-3K/Akt/mTOR, and Jak/Stat. Sorafenib is the multikinase inhibitor that has shown modest survival benefits in advanced HCC in two randomized controlled trials, supporting the use of molecularly targeted therapies in treatment of HCC. A number of strategies including monoclonal antibodies and tyrosine kinase inhibitors such as erlotinib, sunitinib, vandetanib, cediranib, brivanib, foretinib, and dovitinib have been developed and tested in various phases of clinical trials. The successful development of these novel targeted agents in the future will be dependent on the selection of patient populations that are most likely to derive clinical benefit, optimization of the dose used and schedules, and investigation of combined therapies. This review describes evolving molecular targeted agents, their common adverse side effects, and its potential use in management of HCC.
Collapse
Affiliation(s)
- Hung Huynh
- Laboratory of Molecular Endocrinology, Division of Molecular and Cellular Research, National Cancer Centre, Level 6, Lab 1, 11 Hospital Drive, Singapore 169610, Singapore.
| |
Collapse
|
224
|
Kotsafti A, Farinati F, Cardin R, Burra P, Bortolami M. Bax inhibitor-1 down-regulation in the progression of chronic liver diseases. BMC Gastroenterol 2010; 10:35. [PMID: 20359348 PMCID: PMC2873598 DOI: 10.1186/1471-230x-10-35] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 04/01/2010] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Bax inhibitor-1 (BI-1) is an evolutionary conserved endoplasmic reticulum protein that, when overexpressed in mammalian cells, suppresses the apoptosis induced by Bax, a pro-apoptotic member of the Bcl-2 family. The aims of this study were: (1) to clarify the role of intrinsic anti- and pro-apoptotic mediators, evaluating Bax and BI-1 mRNA and protein expressions in liver tissues from patients with different degrees of liver damage; (2) to determine whether HCV and HBV infections modulate said expression. METHODS We examined 62 patients: 39 with chronic hepatitis (CH) (31 HCV-related and 8 HBV-related); 7 with cirrhosis (6 HCV-related and 1 HBV-related); 13 with hepatocellular carcinoma (HCC) [7 in viral cirrhosis (6 HCV- and 1 HBV-related), 6 in non-viral cirrhosis]; and 3 controls. Bax and BI-1 mRNAs were quantified by real-time PCR, and BI-1 protein expression by Western blot. RESULTS CH tissues expressed significantly higher BI-1 mRNA levels than cirrhotic tissues surrounding HCC (P < 0.0001) or HCC (P < 0.0001). Significantly higher Bax transcripts were observed in HCV-genotype-1-related than in HCV-genotype-3-related CH (P = 0.033). A positive correlation emerged between BI-1 and Bax transcripts in CH tissues, even when HCV-related CH and HCV-genotype-1-related CH were considered alone (P = 0.0007, P = 0.0005 and P = 0.0017, respectively). CONCLUSIONS BI-1 expression is down-regulated as liver damage progresses. The high BI-1 mRNAs levels observed in early liver disease may protect virus-infected cells against apoptosis, while their progressive downregulation may facilitate hepatocellular carcinogenesis. HCV genotype seems to have a relevant role in Bax transcript expression.
Collapse
Affiliation(s)
- Andromachi Kotsafti
- Department of Surgical and Gastroenterological Sciences, University of Padua, Padua, Italy
| | | | | | | | | |
Collapse
|
225
|
Teranishi M, Toyooka T, Ohura T, Masuda S, Ibuki Y. Benzo[a]pyrene exposed to solar-simulated light inhibits apoptosis and augments carcinogenicity. Chem Biol Interact 2010; 185:4-11. [DOI: 10.1016/j.cbi.2010.02.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Revised: 02/25/2010] [Accepted: 02/25/2010] [Indexed: 11/16/2022]
|
226
|
Neuveut C, Wei Y, Buendia MA. Mechanisms of HBV-related hepatocarcinogenesis. J Hepatol 2010; 52:594-604. [PMID: 20185200 DOI: 10.1016/j.jhep.2009.10.033] [Citation(s) in RCA: 335] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 10/22/2009] [Accepted: 10/23/2009] [Indexed: 12/04/2022]
Abstract
The hepatitis B virus (HBV) is a small enveloped DNA virus, which primarily infects hepatocytes and causes acute and persistent liver disease. Epidemiological studies have provided overwhelming evidence for a causal role of chronic HBV infection in the development of hepatocellular carcinoma, but the molecular mechanisms underlying virally-induced tumourigenesis remain largely debated. In the absence of a dominant oncogene encoded by the HBV genome, indirect roles have been proposed, including insertional activation of cellular cancer-related genes by HBV DNA integration, induction of genetic instability by viral integration or by the regulatory protein HBx, and long-term effects of viral proteins in enhancing immune-mediated liver disease. Recent genetic studies indicate that HBV-related tumours display a distinctive profile with a high rate of chromosomal alterations and low frequency of beta-catenin mutations. This review will discuss the evidence implicating chronic HBV infection as a causal risk factor of primary liver cancer. It will also discuss the molecular mechanisms that are critical for the tumourigenic process due to long lasting infection with HBV.
Collapse
Affiliation(s)
- Christine Neuveut
- Oncogenesis and Molecular Virology Unit, Institut Pasteur, Inserm U579, 28 rue du Dr Roux, Paris cedex 15, France
| | | | | |
Collapse
|
227
|
Tovar V, Alsinet C, Villanueva A, Hoshida Y, Chiang DY, Solé M, Thung S, Moyano S, Toffanin S, Mínguez B, Cabellos L, Peix J, Schwartz M, Mazzaferro V, Bruix J, Llovet JM. IGF activation in a molecular subclass of hepatocellular carcinoma and pre-clinical efficacy of IGF-1R blockage. J Hepatol 2010; 52:550-9. [PMID: 20206398 PMCID: PMC3662876 DOI: 10.1016/j.jhep.2010.01.015] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Revised: 10/01/2009] [Accepted: 10/14/2009] [Indexed: 01/18/2023]
Abstract
BACKGROUND & AIMS IGF signaling has a relevant role in a variety of human malignancies. We analyzed the underlying molecular mechanisms of IGF signaling activation in early hepatocellular carcinoma (HCC; BCLC class 0 or A) and assessed novel targeted therapies blocking this pathway. METHODS An integrative molecular dissection of the axis was conducted in a cohort of 104 HCCs analyzing gene and miRNA expression, structural aberrations, and protein activation. The therapeutic potential of a selective IGF-1R inhibitor, the monoclonal antibody A12, was assessed in vitro and in a xenograft model of HCC. RESULTS Activation of the IGF axis was observed in 21% of early HCCs. Several molecular aberrations were identified, such as overexpression of IGF2 -resulting from reactivation of fetal promoters P3 and P4-, IGFBP3 downregulation and allelic losses of IGF2R (25% of cases). A gene signature defining IGF-1R activation was developed. Overall, activation of IGF signaling in HCC was significantly associated with mTOR signaling (p=0.035) and was clearly enriched in the Proliferation subclass of the molecular classification of HCC (p=0.001). We also found an inverse correlation between IGF activation and miR-100/miR-216 levels (FDR<0.05). In vitro studies showed that A12-induced abrogation of IGF-1R activation and downstream signaling significantly decreased cell viability and proliferation. In vivo, A12 delayed tumor growth and prolonged survival, reducing proliferation rates and inducing apoptosis. CONCLUSIONS Integrative genomic analysis showed enrichment of activation of IGF signaling in the Proliferation subclass of HCC. Effective blockage of IGF signaling with A12 provides the rationale for testing this therapy in clinical trials.
Collapse
Affiliation(s)
- Victoria Tovar
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Clara Alsinet
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Augusto Villanueva
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | | | | | - Manel Solé
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Swan Thung
- Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA
| | - Susana Moyano
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Sara Toffanin
- Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA.,Liver Cancer Group. Gastrointestinal Surgery and Liver Transplantation Unit, National Cancer Institute, Milan, Italy
| | - Beatriz Mínguez
- Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA
| | - Laia Cabellos
- Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA
| | - Judit Peix
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Myron Schwartz
- Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA
| | - Vincenzo Mazzaferro
- Liver Cancer Group. Gastrointestinal Surgery and Liver Transplantation Unit, National Cancer Institute, Milan, Italy
| | - Jordi Bruix
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain
| | - Josep M. Llovet
- BCLC Group. [HCC Translational Lab, Liver Unit and Pathology Department], Hospital Clínic, CIBERehd, IDIBAPS, Barcelona, Catalonia, Spain.,Mount Sinai Liver Cancer Program. Division of Liver Diseases, Mount Sinai School of Medicine, New York, USA.,Institució Catalana de Recerca i Estudis Avançats (ICREA). Generalitat de Catalunya. Barcelona, Catalonia, Spain
| |
Collapse
|
228
|
Porta C, Paglino C. Medical treatment of unresectable hepatocellular carcinoma: Going beyond sorafenib. World J Hepatol 2010; 2:103-13. [PMID: 21160981 PMCID: PMC2998959 DOI: 10.4254/wjh.v2.i3.103] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 01/14/2010] [Accepted: 01/21/2010] [Indexed: 02/06/2023] Open
Abstract
Even though Sorafenib has radically changed the natural history of those hepatocellular carcinoma patients who are not amenable for curative treatments, further therapeutic improvements are badly needed. As it was for Sorafenib, our increasingly refined understanding of the complex mechanisms underlying HCC carcinogenesis are the starting point for the future development of such treatments. Presently, a number of molecularly targeted agents are in different stages of development for this once orphan cancer. Indeed, several pathways are presently being explored to identify potentially active drugs, including epidermal growth factor receptor, vascular endothelial growth factor/vascular endothelial growth factor receptors, mammalian target of rapamycin, phosphatidyl-inositol-3-kinase/Akt, insulin growth factor, Aurora kinase, Wnt/β-catenin, retinoic acid receptor and hepatocyte growth factor/C-Met. This review is aimed at addressing the results obtained so far with these newer drugs, also considering the challenges we shall face in the near future, including the issue of response evaluation and identification of predictive/prognostic biomarkers.
Collapse
Affiliation(s)
- Camillo Porta
- Camillo Porta, Medical Oncology and Laboratory of Preclinical Oncology and Experimental Therapies, IRCCS San Matteo University Hospital Foundation, Pavia I-27100, Italy
| | | |
Collapse
|
229
|
Omata M, Lesmana LA, Tateishi R, Chen PJ, Lin SM, Yoshida H, Kudo M, Lee JM, Choi BI, Poon RTP, Shiina S, Cheng AL, Jia JD, Obi S, Han KH, Jafri W, Chow P, Lim SG, Chawla YK, Budihusodo U, Gani RA, Lesmana CR, Putranto TA, Liaw YF, Sarin SK. Asian Pacific Association for the Study of the Liver consensus recommendations on hepatocellular carcinoma. Hepatol Int 2010; 4:439-474. [PMID: 20827404 PMCID: PMC2900561 DOI: 10.1007/s12072-010-9165-7] [Citation(s) in RCA: 831] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Accepted: 12/09/2009] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The Asian Pacific Association for the Study of the Liver (APASL) convened an international working party on the management of hepatocellular carcinoma (HCC) in December 2008 to develop consensus recommendations. METHODS The working party consisted of expert hepatologist, hepatobiliary surgeon, radiologist, and oncologist from Asian-Pacific region, who were requested to make drafts prior to the consensus meeting held at Bali, Indonesia on 4 December 2008. The quality of existing evidence and strength of recommendations were ranked from 1 (highest) to 5 (lowest) and from A (strongest) to D (weakest), respectively, according to the Oxford system of evidence-based approach for developing the consensus statements. RESULTS Participants of the consensus meeting assessed the quality of cited studies and assigned grades to the recommendation statements. Finalized recommendations were presented at the fourth APASL single topic conference on viral-related HCC at Bali, Indonesia and approved by the participants of the conference.
Collapse
Affiliation(s)
- Masao Omata
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Laurentius A. Lesmana
- Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Ryosuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Pei-Jer Chen
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shi-Ming Lin
- Liver Research Unit, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Haruhiko Yoshida
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Masatoshi Kudo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kinki University School of Medicine, Osaka-Sayama, Japan
| | - Jeong Min Lee
- Abdominal Radiology Section, Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Byung Ihn Choi
- Abdominal Radiology Section, Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Ronnie T. P. Poon
- Department of Surgery, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Shuichiro Shiina
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Ann Lii Cheng
- Department of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ji-Dong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, 100050 Beijing, China
| | - Shuntaro Obi
- Division of Hepatology, Kyoundo Hospital, Tokyo, Japan
| | - Kwang Hyub Han
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Wasim Jafri
- Department of Medicine, The Aga Khan University Hospital, Karachi, Pakistan
| | - Pierce Chow
- Department of General Surgery, Singapore General Hospital, Singapore, Singapore
| | - Seng Gee Lim
- Department of Gastroenterology and Hepatology, National University Hospital, Singapore, Singapore
| | - Yogesh K. Chawla
- Departments of Hepatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Unggul Budihusodo
- Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Rino A. Gani
- Hepatology Division, Internal Medicine Department, RSUPN Cipto Mangunkusumo, Jakarta, Indonesia
| | - C. Rinaldi Lesmana
- Department of Internal Medicine, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | | | - Yun Fan Liaw
- Liver Research Unit, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Shiv Kumar Sarin
- Department of Gastroenterology, G. B. Pant Hospital, University of Delhi, New Delhi, India
| |
Collapse
|
230
|
Omata M, Lesmana LA, Tateishi R, Chen PJ, Lin SM, Yoshida H, Kudo M, Lee JM, Choi BI, Poon RTP, Shiina S, Cheng AL, Jia JD, Obi S, Han KH, Jafri W, Chow P, Lim SG, Chawla YK, Budihusodo U, Gani RA, Lesmana CR, Putranto TA, Liaw YF, Sarin SK. Asian Pacific Association for the Study of the Liver consensus recommendations on hepatocellular carcinoma. Hepatol Int 2010. [PMID: 20827404 DOI: 10.1007/s12072-011-9165-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The Asian Pacific Association for the Study of the Liver (APASL) convened an international working party on the management of hepatocellular carcinoma (HCC) in December 2008 to develop consensus recommendations. METHODS The working party consisted of expert hepatologist, hepatobiliary surgeon, radiologist, and oncologist from Asian-Pacific region, who were requested to make drafts prior to the consensus meeting held at Bali, Indonesia on 4 December 2008. The quality of existing evidence and strength of recommendations were ranked from 1 (highest) to 5 (lowest) and from A (strongest) to D (weakest), respectively, according to the Oxford system of evidence-based approach for developing the consensus statements. RESULTS Participants of the consensus meeting assessed the quality of cited studies and assigned grades to the recommendation statements. Finalized recommendations were presented at the fourth APASL single topic conference on viral-related HCC at Bali, Indonesia and approved by the participants of the conference.
Collapse
|
231
|
Tsiambas E, Manaios L, Papanikolopoulos C, Rigopoulos DN, Tsounis D, Karameris A, Soultati A, Koliopoulou A, Kravvaritis C, Sergentanis T, Patsouris E, Dourakis S. Chromogenic in situ hybridization analysis of Epidermal Growth Factor Receptor gene/chromosome 7 numerical aberrations in hepatocellular carcinoma based on tissue microarrays. Pathol Oncol Res 2010; 15:511-20. [PMID: 19145479 DOI: 10.1007/s12253-008-9146-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Accepted: 12/22/2008] [Indexed: 12/16/2022]
Abstract
Although Epidermal Growth Factor Receptor (EGFR) overexpression is observed frequently in hepatocellular carcinomas (HCC), specific gene deregulation mechanisms remain unknown. Our aim was to investigate the prognostic significance of the combined protein and gene/chromosome 7 numerical alterations. Using tissue microarray technology, thirty-five (n = 35) paraffin embedded histologically confirmed HCCs were cored and re-embedded in a paraffin block. Immunohistochemistry was performed for the determination of EGFR protein levels and evaluated by the performance of digital image analysis. Chromogenic in situ hybridization was also performed based on the use of EGFR gene and chromosome 7 centromeric probes, respectively. EGFR overexpression was observed in 26/35 (74.2%) cases and was correlated to the grade of the tumors and also to the history of the patients (p = 0.013, p = 0.036, respectively). Numerical alterations regarding gene and chromosome 7 were identified in 4/35 (11.4%) and 12/35 (43.2%) cases associated to the grade of the tumors (p = 0.019, p = 0.001, respectively) and to the survival rate of the patients (p = 0.037, p = 0.001, respectively). EGFR overall expression was also correlated to the gene copies (p = 0.020). EGFR gene numerical alterations -although rare- and also chromosome 7 aneuploidy maybe affect prognosis in HCC patients. To our knowledge this is the first chromogenic in situ hybridization analysis based on tissue microarrays in hepatocellular carcinoma.
Collapse
|
232
|
Zittermann SI, Capurro MI, Shi W, Filmus J. Soluble glypican 3 inhibits the growth of hepatocellular carcinoma in vitro and in vivo. Int J Cancer 2010; 126:1291-301. [PMID: 19816934 DOI: 10.1002/ijc.24941] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The heterogeneity of the molecular pathology of HCC poses a formidable obstacle to the development of non-cytotoxic therapies. Several pro-tumorigenic signaling pathways can be aberrantly activated in HCC, including those triggered by Wnts. Glypican-3 (GPC3), a membrane-bound heparan sulfate proteoglycan that is overexpressed in most HCCs, promotes the growth of these tumors by stimulating Wnt signaling. Because GPC3 binds with high affinity to Wnts, and its growth-promoting activity requires attachment to the cell membrane, we have hypothesized that a mutated GPC3 lacking the GPI anchoring domain (sGPC3) will block Wnt signaling and inhibit the growth of Wnt-dependent tumors. In addition, because sGPC3 displays heparan sulfate chains, this secreted glypican could also inhibit HCC growth by blocking the activity of other heparin-binding growth factors. To test this hypothesis, HCC cell lines were infected with an sGPC3-expressing lentivirus or virus control, and the effect of sGPC3 on the in vitro and in vivo growth was investigated. In addition, the signaling pathways targeted by sGPC3 were identified. We observed that sGPC3-expressing cells had lower proliferation rate. In addition, sGPC3 significantly inhibited the in vivo growth of the Huh6, HepG2 and Huh7 HCC cell lines. sGPC3 blocked Wnt signaling in Huh6- and Huh7-derived tumors and Erk1/2 and Akt phosphorylation in tumors generated by Huh7 and HepG2 cells, respectively. An anti-angiogenic effect in Huh7 and HepG2-derived tumors was also observed. We conclude that sGPC3 can inhibit HCC tumorigenicity by blocking the activity of several pro-tumorigenic growth factors.
Collapse
Affiliation(s)
- Sandra I Zittermann
- Division of Molecular and Cellular Biology, Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | | | | | | |
Collapse
|
233
|
van Zijl F, Zulehner G, Petz M, Schneller D, Kornauth C, Hau M, Machat G, Grubinger M, Huber H, Mikulits W. Epithelial-mesenchymal transition in hepatocellular carcinoma. Future Oncol 2010; 5:1169-79. [PMID: 19852728 DOI: 10.2217/fon.09.91] [Citation(s) in RCA: 267] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The transition of epithelial cells to a mesenchymal phenotype is of paramount relevance for embryonic development and adult wound healing. During the past decade, the epithelial-mesenchymal transition (EMT) has been increasingly recognized to occur during the progression of various carcinomas such as hepatocellular carcinoma (HCC). Here, we focus on EMT in both experimental liver models and human HCC, emphasizing the underlying molecular mechanisms which show partial recurrence of embryonic programs such as TGF-beta and Wnt/ beta-catenin signaling, including collaboration with hepatitis viruses. We further discuss the differentiation repertoire of malignant hepatocytes with respect to the potential acquisition of stemness, and the involvement of the mesenchymal to epithelial transition, the reversal of EMT, in cancer dissemination and metastatic colonization. The strong evidence for EMT in HCC patients demands novel strategies in pathological assessments and therapeutic concepts to efficiently combat HCC progression.
Collapse
Affiliation(s)
- Franziska van Zijl
- Department of Medicine I, Division: Institute of Cancer Research, Medical University of Vienna, Borschke-Gasse 8a, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Hoshida Y, Toffanin S, Lachenmayer A, Villanueva A, Minguez B, Llovet JM. Molecular classification and novel targets in hepatocellular carcinoma: recent advancements. Semin Liver Dis 2010; 30:35-51. [PMID: 20175032 PMCID: PMC3668687 DOI: 10.1055/s-0030-1247131] [Citation(s) in RCA: 248] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of most lethal cancers worldwide. Strategic decisions for the advancement of molecular therapies in this neoplasm require a clear understanding of its molecular classification. Studies indicate aberrant activation of signaling pathways involved in cellular proliferation (e.g., epidermal growth factor and RAS/mitogen-activated protein kinase pathways), survival (e.g., Akt/mechanistic target of rapamycin pathway), differentiation (e.g., Wnt and Hedgehog pathways), and angiogenesis (e.g., vascular endothelial growth factor and platelet-derived growth factor), which is heterogeneously presented in each tumor. Integrative analysis of accumulated genomic datasets has revealed a global scheme of molecular classification of HCC tumors observed across diverse etiologic factors and geographic locations. Such a framework will allow systematic understanding of the frequently co-occurring molecular aberrations to design treatment strategy for each specific subclass of tumors. Accompanied by a growing number of clinical trials of molecular targeted drugs, diagnostic and prognostic biomarker development will be facilitated with special attention on study design and with new assay technologies specialized for archived fixed tissues. A new class of genomic information, microRNA dysregulation and epigenetic alterations, will provide insight for more precise understanding of disease mechanism and expand the opportunity of biomarker/therapeutic target discovery. These efforts will eventually enable personalized management of HCC.
Collapse
Affiliation(s)
- Yujin Hoshida
- Cancer Program, Broad Institute, Cambridge, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
235
|
Feng YX, Zhao JS, Li JJ, Wang T, Cheng SQ, Yuan Y, Wang F, Wang XF, Xie D. Liver cancer: EphrinA2 promotes tumorigenicity through Rac1/Akt/NF-kappaB signaling pathway. Hepatology 2010; 51:535-44. [PMID: 19918976 DOI: 10.1002/hep.23313] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
UNLABELLED Eph/Ephrin family, one of the largest receptor tyrosine kinase families, has been extensively studied in morphogenesis and neural development. Recently, growing attention has been paid to its role in the initiation and progression of various cancers. However, the role of Eph/Ephrins in hepatocellular carcinoma (HCC) has been rarely investigated. In this study, we found that the expression of EphrinA2 was significantly up-regulated in both established cell lines and clinical tissue samples of HCC, and the most significant increase was observed in the tumors invading the portal veins. Forced expression of EphrinA2 in HCC cells significantly promoted in vivo tumorigenicity, whereas knockdown of this gene inhibited this oncogenic effect. We further found that suppression of apoptosis, rather than accelerating proliferation, was responsible for EphrinA2-enhanced tumorigenicity. In addition, EphrinA2 endowed cancer cells with resistance to tumor necrosis factor alpha (TNF-alpha)-induced apoptosis, thus facilitating their survival. Furthermore, we disclosed a novel EphrinA2/ras-related c3 botulinum toxin substrate 1 (Rac1)/V-akt murine thymoma viral oncogene homolog (Akt)/nuclear factor-kappa B (NF-kappaB) pathway contributing to the inhibitory effect on apoptosis in HCC cells. CONCLUSION This study revealed that EphrinA2 played an important role in the development and progression of HCC by promoting the survival of cancer cells, indicating its role as a potential therapeutic target in HCC.
Collapse
Affiliation(s)
- Yu-Xiong Feng
- Laboratory of Molecular Oncology, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
236
|
|
237
|
Chang J, Liu Y, Zhang DD, Zhang DJ, Wu CT, Wang LS, Cui CP. Hepatopoietin Cn suppresses apoptosis of human hepatocellular carcinoma cells by up-regulating myeloid cell leukemia-1. World J Gastroenterol 2010; 16:193-200. [PMID: 20066738 PMCID: PMC2806557 DOI: 10.3748/wjg.v16.i2.193] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of hepatopoietin Cn (HPPCn) in apoptosis of hepatocellular carcinoma (HCC) cells and its mechanism.
METHODS: Two human HCC cell lines, SMMC7721 and HepG2, were used in this study. Immunostaining, Western blotting and enzyme linked immunosorbent assay were conducted to identify the expression of HPPCn and the existence of an autocrine loop of HPPCn/HPPCn receptor in SMMC7721 and HepG2. Apoptotic cells were detected using fluorescein isothiocyanate (FITC)-conjugated Annexin V and propidium iodide.
RESULTS: The HPPCn was highly expressed in human HCC cells and secreted into culture medium (CM). FITC-labeled recombinant human protein (rhHPPCn) could specifically bind to its receptor on HepaG2 cells. Treatment with 400 ng/mL rhHPPCn dramatically increased the viability of HCC-derived cells from 48.1% and 36.9% to 85.6% and 88.4%, respectively (P < 0.05). HPPCn silenced by small-interfering RNA reduced the expression and secretion of HPPCn and increased the apoptosis induced by trichostatin A. Additionally, HPPCn could up-regulate the expression of myeloid cell leukemia-1 (Mcl-1) in HCC cells via mitogen-activated protein kinase (MAPK) and sphingosine kinase-1.
CONCLUSION: HPPCn is a novel hepatic growth factor that can be secreted to CM and suppresses apoptosis of HCC cells by up-regulating Mcl-1 expression.
Collapse
|
238
|
Ubagai T, Kikuchi T, Fukusato T, Ono Y. Aflatoxin B1 modulates the insulin-like growth factor-2 dependent signaling axis. Toxicol In Vitro 2009; 24:783-9. [PMID: 20036727 DOI: 10.1016/j.tiv.2009.12.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Revised: 11/17/2009] [Accepted: 12/21/2009] [Indexed: 12/16/2022]
Abstract
Although aflatoxin B(1) (AFB(1)) is known as a mycotoxin that induces hepatocellular carcinoma (HCC), its effects on HCC cells have not been sufficiently investigated. The HCC cell lines HepG2, Huh-6, Huh-7, and PLC were cultured (5 x 10(5)cells/ml) and various concentrations of AFB(1) were added. The expression levels of the alpha-fetoprotein (AFP), insulin-like growth factor-2 (IGF-2), and insulin-like growth factor-1 receptor (IGF-1R) genes in each sample were determined by real-time PCR, with the following results: (1) The level of AFP expression in HepG2 increased at 5-50 ng/ml of AFB(1) in a dose-dependent manner. The AFP expression level in Huh-6 increased at 0.01-5 ng/ml of AFB(1) in a dose-dependent manner and decreased to half controls level at 50 ng/ml of AFB(1). The AFP expression level in Huh-7 decreased to one-third the original level at 0.5-50 ng/ml of AFB(1). The AFP expression level in PLC decreased at 0-0.5 ng/ml of AFB(1) in a dose-dependent manner, and decreased to one-third at concentrations of AFB(1) between 0.5 and 50 ng/ml. (2) The IGF-2 and IGF-1R expression levels in Huh-6 increased more than 10-fold at 0.5-5 ng/ml of AFB(1), but decreased to half at 50 ng/ml of AFB(1). The IGF-2 and IGF-1R expression levels in other cell lines increased in a dose-dependent manner. AFB(1) induced translations of IGF-2 and IGF-1R and cell proliferation: When 50 ng/ml AFB(1) was administrated, cell numbers were 2.0-, 1.7-, and 1.5-fold higher than those of controls after 3 days of culture in HepG2, Huh-7, and PLC, respectively. Particularly, in Huh-6, it increased 2.5-fold higher than those of controls following 5 ng/ml AFB(1) administration. The ratio of fold-change phospho-IGF-1R in all cell lines that were treated with AFB(1), increased 1.1-1.5-fold. These results indicate that AFB(1) may enhance HCC cell proliferation through an IGF-2-dependent signal axis, although it remains to be investigated whether those effects are associated with human hepatocarcinogenesis resulting from AFB(1) exposure.
Collapse
Affiliation(s)
- Tsuneyuki Ubagai
- Department of Microbiology and Immunology, Teikyo University School of Medicine, Tokyo 173-8605, Japan
| | | | | | | |
Collapse
|
239
|
Zulehner G, Mikula M, Schneller D, van Zijl F, Huber H, Sieghart W, Grasl-Kraupp B, Waldhör T, Peck-Radosavljevic M, Beug H, Mikulits W. Nuclear beta-catenin induces an early liver progenitor phenotype in hepatocellular carcinoma and promotes tumor recurrence. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:472-81. [PMID: 20008139 DOI: 10.2353/ajpath.2010.090300] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Transforming growth factor-beta cooperates with oncogenic Ras to activate nuclear beta-catenin during the epithelial to mesenchymal transition of hepatocytes, a process relevant in the progression of hepatocellular carcinoma (HCC). In this study we investigated the role of beta-catenin in the differentiation of murine, oncogene-targeted hepatocytes and in 133 human HCC patients scheduled for orthotopic liver transplantation. Transforming growth factor-beta caused dissociation of plasma membrane E-cadherin/beta-catenin complexes and accumulation of nuclear beta-catenin in Ras-transformed, but otherwise normal hepatocytes in p19(ARF)-/- mice. Both processes were inhibited by Smad7-mediated disruption of transforming growth factor-beta signaling. Overexpression of constitutively active beta-catenin resulted in high levels of CK19 and M2-PK, whereas ablation of beta-catenin by axin overexpression caused strong expression of CK8 and CK18. Therefore, nuclear beta-catenin resulted in dedifferentiation of neoplastic hepatocytes to immature progenitor cells, whereas loss of nuclear beta-catenin led to a differentiated HCC phenotype. Poorly differentiated human HCC showed cytoplasmic redistribution or even loss of E-cadherin, suggesting epithelial to mesenchymal transition. Analysis of 133 HCC patient samples revealed that 58.6% of human HCC exhibited strong nuclear beta-catenin accumulation, which correlated with clinical features such as vascular invasion and recurrence of disease after orthotopic liver transplantation. These data suggest that activation of beta-catenin signaling causes dedifferentiation to malignant, immature hepatocyte progenitors and facilitates recurrence of human HCC after orthotopic liver transplantation.
Collapse
Affiliation(s)
- Gudrun Zulehner
- Department of Internal Medicine I, Centre of Public Health, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Abstract
There have been innumerable studies published in the attempt to identify gene expression signatures in hepatocellular carcinoma (HCC). When all the regulators and targets of the differentially expressed genes are analyzed from larger studies, the most striking theme is upregulation of mitosis-promoting and cell proliferation genes in HCC compared with 'liver-specific gene clusters' in non-tumorous tissue. A major limitation of expression profiling is that it only provides a 'snapshot' of what is an evolving process and thus cannot distinguish the differences in gene expression that are primary effectors of dysregulated growth from those that represent downstream consequences. The development of HCC in a chronically diseased liver, often referred to as hepatocarcinogenesis, is a multistep process characterized by the progressive accumulation and interplay of genetic alterations causing aberrant growth, malignant transformation of liver parenchymal cells, followed by vascular invasion and metastasis. This review will discuss HCC precursor lesions, draw on the 'proliferation cluster' genes highlighted from HCC expression profiling studies, relate them to a selection of regulatory networks important in liver regeneration, cell cycle control and their potential significance in the pathogenesis of HCC or primary liver cancer.
Collapse
Affiliation(s)
- Narci C Teoh
- Gastroenterology and Hepatology Laboratory, Australian National University Medical School, The Canberra Hospital, Australian Capital Territory, Australia.
| |
Collapse
|
241
|
Differential roles of insulin-like growth factor receptor- and insulin receptor-mediated signaling in the phenotypes of hepatocellular carcinoma cells. Neoplasia 2009; 11:835-45. [PMID: 19724677 DOI: 10.1593/neo.09476] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 05/29/2009] [Accepted: 06/01/2009] [Indexed: 01/23/2023] Open
Abstract
Intrahepatic and extrahepatic metastases are common findings in hepatocellular carcinoma (HCC). Insulin-like growth factor 2 (IGF2) expression is frequently induced in HCC, and serum IGF2 levels correlate with the presence of extrahepatic metastases. Yet, the role of IGF-induced signaling in the dissemination of HCC remains unclear. We have previously observed elevated IGF2 levels in tumors with metastatic potential in an HCC mouse model. Here, we demonstrate that inhibition of IGF2, or its receptor IGF1R, impairs the migration and invasion activities of murine HCC cells. Furthermore, inhibition of IGF1R also impairs the ability of HCC cells to colonize the lungs after introduction into the circulation through the tail vein but does not impair subcutaneous tumor growth. Collectively, these findings suggest that IGF1R-mediated signaling plays a causative role in tumor dissemination but is not required for tumor growth per se. Although previous studies indicate that IGF ligands can signal through IGF1R/insulin receptor (IR) heterodimers, and IR-A homodimers, we demonstrate that the IR is not required for invasion and metastasis by HCC cells. Finally, we identify matrix metalloproteinase 2 as a mediator of the invasive phenotype downstream of IGF1R-induced signaling. Thus, our studies demonstrate the importance of IGF2-induced signaling in the dissemination of HCC cells.
Collapse
|
242
|
Malz M, Weber A, Singer S, Riehmer V, Bissinger M, Riener MO, Longerich T, Soll C, Vogel A, Angel P, Schirmacher P, Breuhahn K. Overexpression of far upstream element binding proteins: a mechanism regulating proliferation and migration in liver cancer cells. Hepatology 2009; 50:1130-9. [PMID: 19585652 DOI: 10.1002/hep.23051] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
UNLABELLED Microtubule-dependent effects are partly regulated by factors that coordinate polymer dynamics such as the microtubule-destabilizing protein stathmin (oncoprotein 18). In cancer cells, increased microtubule turnover affects cell morphology and cellular processes that rely on microtubule dynamics such as mitosis and migration. However, the molecular mechanisms deregulating modifiers of microtubule activity in human hepatocarcinogenesis are poorly understood. Based on profiling data of human hepatocellular carcinoma (HCC), we identified far upstream element binding proteins (FBPs) as significantly coregulated with stathmin. Coordinated overexpression of two FBP family members (FBP-1 and FBP-2) in >70% of all analyzed human HCCs significantly correlated with poor patient survival. In vitro, FBP-1 predominantly induced tumor cell proliferation, while FBP-2 primarily supported migration in different HCC cell lines. Surprisingly, reduction of FBP-2 levels was associated with elevated FBP-1 expression, suggesting a regulatory interplay of FBP family members that functionally discriminate between cell division and mobility. Expression of FBP-1 correlated with stathmin expression in HCC tissues and inhibition of FBP-1 but not of FBP-2 drastically reduced stathmin at the transcript and protein levels. In contrast, further overexpression of FBP-1 did not affect stathmin bioavailability. Accordingly, analyzing nuclear and cytoplasmic areas of HCC cells revealed that reduced FBP-1 levels affected cell morphology and were associated with a less malignant phenotype. CONCLUSION The coordinated activation of FBP-1 and FBP-2 represents a novel and frequent pro-tumorigenic mechanism promoting proliferation (tumor growth) and motility (dissemination) of human liver cancer cells. FBPs promote tumor-relevant functions by at least partly employing the microtubule-destabilizing factor stathmin and represent a new potential target structure for HCC treatment.
Collapse
Affiliation(s)
- Mona Malz
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Abstract
Although there is strong evidence that hepatitis C virus (HCV) is one of the leading causes of hepatocellular carcinoma (HCC), there is still much to understand regarding the mechanism of HCV-induced transformation. While liver fibrosis resulting from long-lasting chronic inflammation and liver regeneration resulting from immune-mediated cell death are likely factors that contribute to the development of HCC, the direct role of HCV proteins remains to be determined. In vitro studies have shown that HCV expression may interfere with cellular functions that are important for cell differentiation and cell growth. However, most studies were performed in artificial models which can only give clues for potential mechanisms that need to be confirmed in more relevant models. Furthermore, the difficulty to identify HCV proteins and infected liver cells in patients, contributes to the complexity of our current understanding. For these reasons, there is currently very little experimental evidence for a direct oncogenic role of HCV. Further studies are warranted to clarify these issues.
Collapse
Affiliation(s)
- Birke Bartosch
- INSERM, U871, 151 Cours Albert Thomas, 69003 Lyon, France
| | | | | | | |
Collapse
|
244
|
Affiliation(s)
- Tadashi Ikegami
- Division of Gastroenterology and Hepatology, Tokyo Medical University, Ibaraki Medical Center, Ibaraki, Japan
| |
Collapse
|
245
|
van Zijl F, Mair M, Csiszar A, Schneller D, Zulehner G, Huber H, Eferl R, Beug H, Dolznig H, Mikulits W. Hepatic tumor-stroma crosstalk guides epithelial to mesenchymal transition at the tumor edge. Oncogene 2009; 28:4022-33. [PMID: 19718050 DOI: 10.1038/onc.2009.253] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The tumor-stroma crosstalk is a dynamic process fundamental in tumor development. In hepatocellular carcinoma (HCC), the progression of malignant hepatocytes frequently depends on transforming growth factor (TGF)-beta provided by stromal cells. TGF-beta induces an epithelial to mesenchymal transition (EMT) of oncogenic Ras-transformed hepatocytes and an upregulation of platelet-derived growth factor (PDGF) signaling. To analyse the influence of the hepatic tumor-stroma crosstalk onto tumor growth and progression, we co-injected malignant hepatocytes and myofibroblasts (MFBs). For this, we either used in vitro-activated p19(ARF) MFBs or in vivo-activated MFBs derived from physiologically inflamed livers of Mdr2/p19(ARF) double-null mice. We show that co-transplantation of MFBs with Ras-transformed hepatocytes strongly enhances tumor growth. Genetic interference with the PDGF signaling decreases tumor cell growth and maintains plasma membrane-located E-cadherin and beta-catenin at the tumor-host border, indicating a blockade of hepatocellular EMT. We further generated a collagen gel-based three dimensional HCC model in vitro to monitor the MFB-induced invasion of micro-organoid HCC spheroids. This invasion was diminished after inhibition of TGF-beta or PDGF signaling. These data suggest that the TGF-beta/PDGF axis is crucial during hepatic tumor-stroma crosstalk, regulating both tumor growth and cancer progression.
Collapse
Affiliation(s)
- F van Zijl
- Department of Medicine I, Division: Institute of Cancer Research, Medical University of Vienna, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Desbois-Mouthon C, Baron A, Blivet-Van Eggelpoël MJ, Fartoux L, Venot C, Bladt F, Housset C, Rosmorduc O. Insulin-like growth factor-1 receptor inhibition induces a resistance mechanism via the epidermal growth factor receptor/HER3/AKT signaling pathway: rational basis for cotargeting insulin-like growth factor-1 receptor and epidermal growth factor receptor in hepatocellular carcinoma. Clin Cancer Res 2009; 15:5445-56. [PMID: 19706799 DOI: 10.1158/1078-0432.ccr-08-2980] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The insulin-like growth factor (IGF) signaling axis is frequently dysregulated in hepatocellular carcinoma (HCC). Therefore, we investigated whether the specific targeting of the IGF type 1 receptor (IGF-1R) might represent a new therapeutic approach for this tumor. EXPERIMENTAL DESIGN Total and phosphorylated levels of IGF-1R were measured in 21 paired samples of human HCCs and adjacent nontumoral livers using ELISA. The antineoplastic potency of a novel anti-IGF-1R antibody, AVE1642, was examined in five human hepatoma cell lines. RESULTS Overexpression of IGF-1R was detected in 33% of HCCs and increased activation of IGF-1R was observed in 52% of tumors. AVE1642 alone had moderate inhibitory effects on cell viability. However, its combination with gefitinib, an epidermal growth factor receptor (EGFR) inhibitor, induced supra-additive effects in all cell lines that were associated with cell cycle blockage and inhibition of AKT phosphorylation. The combination of AVE1642 with rapamycin also induced a synergistic reduction of viability and of AKT phosphorylation. Of marked interest, AVE1642 alone up-regulated the phosphorylated and total levels of HER3, the main partner of EGFR, and AVE1642-induced phosphorylation of HER3 was prevented by gefitinib. Moreover, the down-regulation of HER3 expression with siRNA reduced AKT phosphorylation and increased cell sensitivity to AVE1642. CONCLUSIONS These findings indicate that hepatoma cells overcome IGF-1R inhibition through HER3 activation in an EGFR-dependent mechanism, and that HER3 represents a critical mediator in acquired resistance to anti-IGF-1R therapy. These results provide a strong rational for targeting simultaneously EGFR and IGF-1R in clinical trials for HCC].
Collapse
|
247
|
Gramantieri L, Fornari F, Ferracin M, Veronese A, Sabbioni S, Calin GA, Grazi GL, Croce CM, Bolondi L, Negrini M. MicroRNA-221 targets Bmf in hepatocellular carcinoma and correlates with tumor multifocality. Clin Cancer Res 2009; 15:5073-81. [PMID: 19671867 DOI: 10.1158/1078-0432.ccr-09-0092] [Citation(s) in RCA: 249] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED Deregulated cell proliferation and apoptosis play a major role in hepatocellular carcinoma (HCC). MicroRNAs participate in the modulation of key molecules linked to hepatocarcinogenesis. PURPOSE This study aims to investigate the role of miR-221 in the modulation of Bmf, a proapoptotic BH3-only protein, and to characterize miR-221 contribution to hepatocarcinogenesis through modulation of apoptosis. EXPERIMENTAL DESIGN Transfection of miR-221 and anti-miR-221 in HCC-derived cell lines and luciferase reporter assay were used to assess Bmf as a target of miR-221. Modulation of miR-221 and Bmf expression contributed to characterize their role in anoikis. Primary HCC tissues were analyzed to assess the clinical relevance of in vitro findings. RESULTS Enforced miR-221 expression caused Bmf down-regulation, whereas anti-miR-221 induced its up-regulation. A luciferase reporter assay confirmed Bmf as a target of miR-221. Following matrix detachment, miR-221 silencing led to increased apoptotic cell death. The analysis of HCC tissues revealed an inverse correlation between miR-221 and Bmf expression and a direct correlation between Bmf and activated caspase-3, as a marker of apoptosis. High miR-221 levels were associated with tumor multifocality and reduced time to recurrence after surgery. CONCLUSIONS Our results indicate that miR-221, by targeting Bmf, inhibits apoptosis. Moreover, in HCC, miR-221 overexpression is associated with a more aggressive phenotype. These findings, together with the previously reported modulation of CDKN1B/p27 and CDKN1C/p57, show that miR-221 simultaneously affects multiple pro-oncogenic pathways and suggest miR-221 as a potential target for nonconventional treatment against HCC.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Aged
- Aged, 80 and over
- Apoptosis/drug effects
- Apoptosis/genetics
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Gene Targeting
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Male
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/physiology
- Middle Aged
- Neoplasm Recurrence, Local
- Neoplasms, Multiple Primary/genetics
- Neoplasms, Multiple Primary/mortality
- Neoplasms, Multiple Primary/pathology
- RNA, Small Interfering/pharmacology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Laura Gramantieri
- Dipartimento di Medicina Interna e Gastroenterologia e Centro di Ricerca Biomedica Applicata, Università di Bologna, Bologna, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Piscaglia AC, Shupe TD, Pani G, Tesori V, Gasbarrini A, Petersen BE. Establishment of cancer cell lines from rat hepatocholangiocarcinoma and assessment of the role of granulocyte-colony stimulating factor and hepatocyte growth factor in their growth, motility and survival. J Hepatol 2009; 51:77-92. [PMID: 19446912 PMCID: PMC2694236 DOI: 10.1016/j.jhep.2009.02.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 01/27/2009] [Accepted: 02/14/2009] [Indexed: 01/16/2023]
Abstract
BACKGROUND/AIMS Oval cells (OCs), putative hepatic stem cells, may give rise to liver cancers. We developed a carcinogenesis regimen, based upon induction of OC proliferation prior to carcinogen exposure. In our model, rats subjected to 2-acetylaminofluorene/ partial-hepatectomy followed by aflatoxin injection (APA regimen) developed well-differentiated hepatocholangiocarcinomas. The aim of this study was to establish and characterize cancer cell lines from this animal model. METHODS Cancer cells were cultured from animals sacrificed eight months after treatment, and single clones were selected. The established cell lines, named LCSCs, were characterized, and their tumorigenicity was assessed in vivo. The roles of granulocyte-colony stimulating factor (G-CSF) and hepatocyte growth factor (HGF) in LCSC growth, survival and motility were also investigated. RESULTS From primary tumors, six cell lines were developed. LCSCs shared with the primary tumors the expression of various OC-associated markers, including cMet and G-CSF receptor. In vitro, HGF conferred protection from death by serum withdrawal. Stimulation with G-CSF increased LCSC growth and motility, while the blockage of its receptor inhibited LCSC proliferation and migration. CONCLUSIONS Six cancer cell lines were established from our model of hepatocholangiocarcinoma. HGF modulated LCSC resistance to apoptosis, while G-CSF acted on LCSCs as a proliferative and chemotactic agent.
Collapse
Affiliation(s)
- Anna C Piscaglia
- Department of Internal Medicine, GI & Liver Stem Cell Research Group, Catholic University of Rome - Gemelli Hospital, Largo A. Gemelli 8, 00168 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
249
|
Kaufmann R, Oettel C, Horn A, Halbhuber KJ, Eitner A, Krieg R, Katenkamp K, Henklein P, Westermann M, Böhmer FD, Ramachandran R, Saifeddine M, Hollenberg MD, Settmacher U. Met receptor tyrosine kinase transactivation is involved in proteinase-activated receptor-2-mediated hepatocellular carcinoma cell invasion. Carcinogenesis 2009; 30:1487-96. [PMID: 19546160 DOI: 10.1093/carcin/bgp153] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The expression of proteinase-activated receptor (PAR)(2) in human hepatocellular carcinoma (HCC) was established by reverse transcription-polymerase chain reaction, confocal immunofluorescence and electron microscopy in permanent cell lines, primary HCC cell cultures and HCC tumor tissue. Stimulation of HCC cells with trypsin and the PAR(2)-selective activating peptide, 2-furoyl-LIGRLO-NH(2), increased cell invasion across Matrigel. Both effects were blocked by a PAR(2)-selective pepducin antagonist peptide (pal-PAR(2)) and by PAR(2) silencing with specific small interfering RNA (siRNA). PAR(2)-initiated HCC cell invasion was also blocked by inhibiting the hepatocyte growth factor receptor (Met receptor tyrosine kinase) with the receptor-targeted kinase inhibitors, SU 11274 and PHA 665752, or by downregulation of Met with specific siRNA. The involvement of Met in PAR(2)-mediated HCC invasive signaling was further supported by the finding that treatment of HCC cells with trypsin or the PAR(2)-selective agonist peptide, 2-furoyl-LIGRLO-NH(2), stimulated Met activation-phosphorylation. In addition, Met-dependent stimulation of p42/p44 mitogen-activated protein Kinases was found to be critical for the PAR(2)-Met receptor tyrosine kinase-invasive signaling axis in HCC cells. Our study establishes an important link between the PAR(2) and Met receptor tyrosine kinase signaling in promoting HCC cell invasion.
Collapse
Affiliation(s)
- Roland Kaufmann
- Department of General, Visceral and Vascular Surgery, Research Center Lobeda Medical Faculty, Friedrich Schiller University Jena, D-07747 Jena, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Zakaria Y, Rahmat A, Pihie AHL, Abdullah NR, Houghton PJ. Eurycomanone induce apoptosis in HepG2 cells via up-regulation of p53. Cancer Cell Int 2009; 9:16. [PMID: 19508737 PMCID: PMC2700790 DOI: 10.1186/1475-2867-9-16] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Accepted: 06/10/2009] [Indexed: 11/10/2022] Open
Abstract
Background Eurycomanone is a cytotoxic compound found in Eurycoma longifolia Jack. Previous studies had noted the cytotoxic effect against various cancer cell lines. The aim of this study is to investigate the cytotoxicity against human hepato carcinoma cell in vitro and the mode of action. The cytotoxicity of eurycomanone was evaluated using MTT assay and the mode of cell death was detected by Hoechst 33258 nuclear staining and flow cytometry with Annexin-V/propidium iodide double staining. The protein expression Bax, Bcl-2, p53 and cytochrome C were studied by flow cytometry using a spesific antibody conjugated fluorescent dye to confirm the up-regulation of p53 and Bax in cancer cells. Results The findings suggested that eurycomanone was cytotoxic on cancerous liver cell, HepG2 and less toxic on normal cells Chang's liver and WLR-68. Furthermore, various methods proved that apoptosis was the mode of death in eurycomanone-treated HepG2 cells. The characteristics of apoptosis including chromatin condensation, DNA fragmentation and apoptotic bodies were found following eurycomanone treatment. This study also found that apoptotic process triggered by eurycomanone involved the up-regulation of p53 tumor suppressor protein. The up-regulation of p53 was followed by the increasing of pro-apoptotic Bax and decreasing of anti-apoptotic Bcl-2. The increased of cytochrome C levels in cytosol also results in induction of apoptosis. Conclusion The data suggest that eurycomanone was cytotoxic on HepG2 cells by inducing apoptosis through the up-regulation of p53 and Bax, and down-regulation of Bcl-2.
Collapse
Affiliation(s)
- Yusmazura Zakaria
- Institute of Biosciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
| | | | | | | | | |
Collapse
|