51
|
Passman AM, Low J, London R, Tirnitz-Parker JEE, Miyajima A, Tanaka M, Strick-Marchand H, Darlington GJ, Finch-Edmondson M, Ochsner S, Zhu C, Whelan J, Callus BA, Yeoh GCT. A Transcriptomic Signature of Mouse Liver Progenitor Cells. Stem Cells Int 2016; 2016:5702873. [PMID: 27777588 PMCID: PMC5061959 DOI: 10.1155/2016/5702873] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 08/04/2016] [Accepted: 08/14/2016] [Indexed: 01/07/2023] Open
Abstract
Liver progenitor cells (LPCs) can proliferate extensively, are able to differentiate into hepatocytes and cholangiocytes, and contribute to liver regeneration. The presence of LPCs, however, often accompanies liver disease and hepatocellular carcinoma (HCC), indicating that they may be a cancer stem cell. Understanding LPC biology and establishing a sensitive, rapid, and reliable method to detect their presence in the liver will assist diagnosis and facilitate monitoring of treatment outcomes in patients with liver pathologies. A transcriptomic meta-analysis of over 400 microarrays was undertaken to compare LPC lines against datasets of muscle and embryonic stem cell lines, embryonic and developed liver (DL), and HCC. Three gene clusters distinguishing LPCs from other liver cell types were identified. Pathways overrepresented in these clusters denote the proliferative nature of LPCs and their association with HCC. Our analysis also revealed 26 novel markers, LPC markers, including Mcm2 and Ltbp3, and eight known LPC markers, including M2pk and Ncam. These markers specified the presence of LPCs in pathological liver tissue by qPCR and correlated with LPC abundance determined using immunohistochemistry. These results showcase the value of global transcript profiling to identify pathways and markers that may be used to detect LPCs in injured or diseased liver.
Collapse
Affiliation(s)
- Adam M. Passman
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
- The Centre for Medical Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia
| | - Jasmine Low
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
- ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Crawley, WA 6009, Australia
| | - Roslyn London
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
| | - Janina E. E. Tirnitz-Parker
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
- School of Medicine and Pharmacology, The University of Western Australia, Fremantle, WA 6160, Australia
| | - Atsushi Miyajima
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-8654, Japan
| | - Minoru Tanaka
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-8654, Japan
| | | | | | - Megan Finch-Edmondson
- Department of Physiology, NUS Yong Loo Lin School of Medicine, Singapore 117411
- Mechanobiology Institute (MBI), National University of Singapore, Singapore 117411
| | - Scott Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cornelia Zhu
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
- The Centre for Medical Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia
| | - James Whelan
- ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Crawley, WA 6009, Australia
- Department of Animal, Plant and Soil Sciences, La Trobe University, Melbourne, VIC 3086, Australia
| | - Bernard A. Callus
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
- The Centre for Medical Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia
- School of Health Sciences, The University of Notre Dame Australia, Fremantle, WA 6959, Australia
| | - George C. T. Yeoh
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA 6009, Australia
- The Centre for Medical Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia
| |
Collapse
|
52
|
Weber SN, Bohner A, Dapito DH, Schwabe RF, Lammert F. TLR4 Deficiency Protects against Hepatic Fibrosis and Diethylnitrosamine-Induced Pre-Carcinogenic Liver Injury in Fibrotic Liver. PLoS One 2016; 11:e0158819. [PMID: 27391331 PMCID: PMC4938399 DOI: 10.1371/journal.pone.0158819] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/22/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The development of hepatocellular carcinoma (HCC) is a common consequence of advanced liver fibrosis but the interactions between fibrogenesis and carcinogenesis are still poorly understood. Recently it has been shown that HCC promotion depends on Toll-like receptor (TLR) 4. Pre-cancerogenous events can be modelled in mice by the administration of a single dose of diethylnitrosamine (DEN), with HCC formation depending amongst others on interleukin (IL) 6 production. Mice lacking the hepatocanalicular phosphatidylcholine transporter ABCB4 develop liver fibrosis spontaneously, resemble patients with sclerosing cholangitis due to mutations of the orthologous human gene, and represent a valid model to study tumour formation in pre-injured cholestatic liver. The aim of this study was to investigate DEN-induced liver injury in TLR4-deficient mice with biliary fibrosis. METHODS ABCB4-deficient mice on the FVB/NJ genetic background were crossed to two distinct genetic backgrounds (TLR4-sufficient C3H/HeN and TLR4-deficient C3H/HeJ) for more than 10 generations. The two congenic knockout and the two corresponding wild-type mouse lines were treated with a single dose of DEN for 48 hours. Phenotypic differences were assessed by measuring hepatic collagen contents, inflammatory markers (ALT, CRP, IL6) as well as hepatic apoptosis (TUNEL) and proliferation (Ki67) rates. RESULTS Hepatic collagen accumulation is significantly reduced in ABCB4-/-:TLR4-/-double-deficient mice. After DEN challenge, apoptosis, proliferation and inflammatory markers are decreased in TLR4-deficient in comparison to TLR4-sufficient mice. When combining ABCB4 and TLR4 deficiency with DEN treatment, hepatic IL6 expression and proliferation rates are lowest in fibrotic livers from the double-deficient line. Consistent with these effects, selective digestive tract decontamination in ABCB4-/- mice also led to reduced tumor size and number after DEN. CONCLUSION This study demonstrates that liver injury upon DEN challenge depends on pre-existing fibrosis and genetic background. The generation of ABCB4-/: TLR4-/- double-deficient mice illustrates that TLR4-deficiency protects against hepatic injury in a preclinical mouse model of chronic liver disease.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- Animals
- Carcinoma, Hepatocellular/chemically induced
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Diethylnitrosamine/toxicity
- Interleukin-6/genetics
- Interleukin-6/metabolism
- Liver Cirrhosis/chemically induced
- Liver Cirrhosis/genetics
- Liver Cirrhosis/metabolism
- Liver Cirrhosis/pathology
- Liver Neoplasms, Experimental/chemically induced
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/pathology
- Mice, Knockout
- Neoplasm Proteins/deficiency
- Neoplasm Proteins/metabolism
- Precancerous Conditions/chemically induced
- Precancerous Conditions/genetics
- Precancerous Conditions/metabolism
- Precancerous Conditions/pathology
- Toll-Like Receptor 4/deficiency
- ATP-Binding Cassette Sub-Family B Member 4
Collapse
Affiliation(s)
| | - Annika Bohner
- Department of Medicine II, Saarland University Medical Center, Homburg, Germany
| | - Dianne H. Dapito
- Department of Medicine, Columbia University, New York, NY, United States of America
| | - Robert F. Schwabe
- Department of Medicine, Columbia University, New York, NY, United States of America
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
53
|
Tagaram HRS, Desai D, Li G, Liu D, Rountree CB, Gowda K, Berg A, Amin S, Staveley-O'Carroll KF, Kimchi ET. A Selenium Containing Inhibitor for the Treatment of Hepatocellular Cancer. Pharmaceuticals (Basel) 2016; 9:E18. [PMID: 27023566 PMCID: PMC4932536 DOI: 10.3390/ph9020018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/11/2016] [Accepted: 03/16/2016] [Indexed: 11/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most deadly cancer in the world. New treatment strategies are desperately needed due to limited standard therapies. Activation of the Erk, Akt, and STAT3pathways is implicated in the prognosis of HCC. The Se,Se'-1,4-phenylenebis(1,2-ethanediyl) bisisoselenourea (PBISe), is a selenium-containing MAPK and PI3 kinase inhibitor, effectively inhibit tumorigenesis in a variety of experimental models. The aim of our study is to demonstrate the potential role of PBISe in the treatment of HCC. The anti-proliferative and pro-apoptotic ability of PBISe is studied in vitro in four human HCC cell lines and in vivo in a spontaneous murine HCC model. Inhibition of cancer growth was performed by cell viability assay and apoptosis by caspase 3/7, PARP cleavage, annexin-V, and TUNEL assays. Role of PBISe on PI3 kinase, MAPK and STAT3 signaling is determined by Western blotting. In vivo effects of PBISe on tumor sizes were monitored using MRI in a spontaneous murine HCC. Liver tissues from the PBISe-treated mice are analyzed for angiogenesis, proliferation, and signaling pathway markers. Overall, PBISe activated caspase-3/7 and increased DNA fragmentation, which is positively correlated with the increased PARP cleavage. PBISe promoted apoptosis by inhibiting PI3K, MAPK, and STAT3 signaling with significant reduction in the tumor sizes (p < 0.007). PBISe-treated tumors reduced survival marker PCNA, and angiogenesis markers Vegf-A, Vegf-R3 and CD34. These results demonstrate the chemotherapeutic effects of PBISe, by inhibiting tumor growth and facilitating tumor apoptosis for HCC treatment.
Collapse
Affiliation(s)
| | - Dhimant Desai
- Department of Pharmacology, Pennsylvania State University, Hershey, PA 17033, USA.
| | - Guangfu Li
- Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Dai Liu
- Medical University of South Carolina, Charleston, SC 29425, USA.
| | - C Bart Rountree
- Bon Secours Pediatric Associates, 5875 Bremo Road, Richmond, VA 23226, USA.
| | - Kavitha Gowda
- Department of Surgery, Pennsylvania State University, Hershey, PA 17033, USA.
| | - Arthur Berg
- Department of Public Health Sciences, Pennsylvania State University, Hershey, PA 17033, USA.
| | - Shantu Amin
- Department of Pharmacology, Pennsylvania State University, Hershey, PA 17033, USA
| | | | - Eric T Kimchi
- Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
54
|
Bulvik BE, Rozenblum N, Gourevich S, Ahmed M, Andriyanov AV, Galun E, Goldberg SN. Irreversible Electroporation versus Radiofrequency Ablation: A Comparison of Local and Systemic Effects in a Small-Animal Model. Radiology 2016; 280:413-24. [PMID: 27429143 DOI: 10.1148/radiol.2015151166] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Purpose To compare both periablational and systemic effects of two mechanistically different types of ablation: thermal radiofrequency (RF) ablation and electroporative ablation with irreversible electroporation (IRE) in appropriately selected animal models. Materials and Methods Animal experiments were performed according to a protocol approved by the Animal Care Committee of Hebrew University. Female C57BL/6 mice (n = 165) were randomized to undergo either RF or IRE ablation of noncancerous normal liver. The inflammatory response, cell proliferation, interleukin 6 (IL-6) levels, and intactness of vessels in the liver were assessed at 6, 12, and 24 hours and at 3, 7, and 14 days after ablation (n = 122 for mechanistic experiments). Systemic effects were then assessed by comparing tumor formation in an Mdr2-knockout (KO) mouse model (n = 15) and tumor growth in a remote BNL 1ME hepatoma xenograft tumor (n = 28). Results were averaged and evaluated by using two-tailed t tests. Results Although RF ablation was associated with a well-defined periablational inflammatory rim, for IRE, the infiltrate penetrated the ablation zone, largely along persistently patent vessels. Peak IL-6 levels (6 hours after ablation) were 10 and three times higher than at baseline for IRE and RF, respectively (P < .03). Mdr2-KO mice that were treated with IRE ablation had more tumors that were 3 mm or larger than mice treated with RF ablation or sham operation (mean, 3.6 ± 1.3 [standard deviation] vs 2.4 ± 1.1 and 2.2 ± 0.8, respectively; P < .05 for IRE vs both RF ablation and sham operation). For BNL 1ME tumors, both RF and IRE liver ablation reduced tumor growth, with a greater effect noted for IRE (1329 mm(3) ± 586 and 819 mm(3) ± 327 vs 2241 mm(3) ± 548 for sham operation; P < .05) that was accompanied by more infiltrating lymphocytes compared with sham operation (7.6 cells per frame ± 1.9 vs 11.2 ± 2.1 vs 0.3 ± 0.1; P < .05). Conclusion Persistent patency of vasculature within the coagulated zone from IRE increases the area and accumulation of infiltrative cells that is associated with a higher serum IL-6 level than RF ablation. These local changes of IRE induce more robust systemic effects, including both tumorigenic and immunogenic effects. (©) RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Baruch E Bulvik
- From the Goldyne Savad Institute of Gene Therapy (B.E.B., N.R., S.G., E.G., S.N.G.), Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research Israel-Canada (A.V.A.), and Department of Radiology (S.N.G.), Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; and Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A., S.N.G.)
| | - Nir Rozenblum
- From the Goldyne Savad Institute of Gene Therapy (B.E.B., N.R., S.G., E.G., S.N.G.), Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research Israel-Canada (A.V.A.), and Department of Radiology (S.N.G.), Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; and Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A., S.N.G.)
| | - Svetlana Gourevich
- From the Goldyne Savad Institute of Gene Therapy (B.E.B., N.R., S.G., E.G., S.N.G.), Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research Israel-Canada (A.V.A.), and Department of Radiology (S.N.G.), Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; and Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A., S.N.G.)
| | - Muneeb Ahmed
- From the Goldyne Savad Institute of Gene Therapy (B.E.B., N.R., S.G., E.G., S.N.G.), Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research Israel-Canada (A.V.A.), and Department of Radiology (S.N.G.), Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; and Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A., S.N.G.)
| | - Alexander V Andriyanov
- From the Goldyne Savad Institute of Gene Therapy (B.E.B., N.R., S.G., E.G., S.N.G.), Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research Israel-Canada (A.V.A.), and Department of Radiology (S.N.G.), Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; and Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A., S.N.G.)
| | - Eithan Galun
- From the Goldyne Savad Institute of Gene Therapy (B.E.B., N.R., S.G., E.G., S.N.G.), Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research Israel-Canada (A.V.A.), and Department of Radiology (S.N.G.), Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; and Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A., S.N.G.)
| | - S Nahum Goldberg
- From the Goldyne Savad Institute of Gene Therapy (B.E.B., N.R., S.G., E.G., S.N.G.), Laboratory of Membrane and Liposome Research, Department of Biochemistry, Institute for Medical Research Israel-Canada (A.V.A.), and Department of Radiology (S.N.G.), Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; and Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass (M.A., S.N.G.)
| |
Collapse
|
55
|
Leikin-Frenkel A, Shomonov-Wagner L, Juknat A, Pasmanik-Chor M. Maternal Diet Enriched with α-Linolenic or Saturated Fatty Acids Differentially Regulates Gene Expression in the Liver of Mouse Offspring. JOURNAL OF NUTRIGENETICS AND NUTRIGENOMICS 2016; 8:185-94. [DOI: 10.1159/000442945] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 11/30/2015] [Indexed: 11/19/2022]
|
56
|
Hashimoto K, Suzuki AM, Dos Santos A, Desterke C, Collino A, Ghisletti S, Braun E, Bonetti A, Fort A, Qin XY, Radaelli E, Kaczkowski B, Forrest ARR, Kojima S, Samuel D, Natoli G, Buendia MA, Faivre J, Carninci P. CAGE profiling of ncRNAs in hepatocellular carcinoma reveals widespread activation of retroviral LTR promoters in virus-induced tumors. Genome Res 2015; 25:1812-24. [PMID: 26510915 PMCID: PMC4665003 DOI: 10.1101/gr.191031.115] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 09/30/2015] [Indexed: 12/20/2022]
Abstract
An increasing number of noncoding RNAs (ncRNAs) have been implicated in various human diseases including cancer; however, the ncRNA transcriptome of hepatocellular carcinoma (HCC) is largely unexplored. We used CAGE to map transcription start sites across various types of human and mouse HCCs with emphasis on ncRNAs distant from protein-coding genes. Here, we report that retroviral LTR promoters, expressed in healthy tissues such as testis and placenta but not liver, are widely activated in liver tumors. Despite HCC heterogeneity, a subset of LTR-derived ncRNAs were more than 10-fold up-regulated in the vast majority of samples. HCCs with a high LTR activity mostly had a viral etiology, were less differentiated, and showed higher risk of recurrence. ChIP-seq data show that MYC and MAX are associated with ncRNA deregulation. Globally, CAGE enabled us to build a mammalian promoter map for HCC, which uncovers a new layer of complexity in HCC genomics.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Animals
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Viral
- Computational Biology/methods
- Disease Models, Animal
- Disease Progression
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic
- Humans
- Liver Neoplasms/etiology
- Liver Neoplasms/pathology
- Mice
- Mice, Knockout
- Promoter Regions, Genetic
- Protein Binding
- RNA, Untranslated/genetics
- Terminal Repeat Sequences
- Transcription Factors/metabolism
- Transcription Initiation Site
- Transcriptome
- ATP-Binding Cassette Sub-Family B Member 4
Collapse
Affiliation(s)
- Kosuke Hashimoto
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa, 230-0045 Japan
| | - Ana Maria Suzuki
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa, 230-0045 Japan
| | - Alexandre Dos Santos
- INSERM, U1193, Paul-Brousse Hospital, Hepatobiliary Centre, 94800 Villejuif, France; Université Paris Saclay, Faculté de Médecine Le Kremlin Bicêtre, 94800 Villejuif, France
| | - Christophe Desterke
- INSERM, U1193, Paul-Brousse Hospital, Hepatobiliary Centre, 94800 Villejuif, France; Université Paris Saclay, Faculté de Médecine Le Kremlin Bicêtre, 94800 Villejuif, France
| | - Agnese Collino
- European Institute of Oncology (IEO), Department of Experimental Oncology, IFOM-IEO Campus, 20139 Milan, Italy
| | - Serena Ghisletti
- European Institute of Oncology (IEO), Department of Experimental Oncology, IFOM-IEO Campus, 20139 Milan, Italy
| | - Emilie Braun
- INSERM, U1193, Paul-Brousse Hospital, Hepatobiliary Centre, 94800 Villejuif, France; Université Paris Saclay, Faculté de Médecine Le Kremlin Bicêtre, 94800 Villejuif, France
| | - Alessandro Bonetti
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa, 230-0045 Japan
| | - Alexandre Fort
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa, 230-0045 Japan
| | - Xian-Yang Qin
- RIKEN Center for Life Science Technologies, Division of Bio-function Dynamics Imaging, Wako, Saitama, 351-0198, Japan
| | - Enrico Radaelli
- VIB Center for the Biology of Disease, KU Leuven Center for Human Genetics, B-3000 Leuven, Belgium
| | - Bogumil Kaczkowski
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa, 230-0045 Japan
| | - Alistair R R Forrest
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa, 230-0045 Japan
| | - Soichi Kojima
- RIKEN Center for Life Science Technologies, Division of Bio-function Dynamics Imaging, Wako, Saitama, 351-0198, Japan
| | - Didier Samuel
- INSERM, U1193, Paul-Brousse Hospital, Hepatobiliary Centre, 94800 Villejuif, France; Université Paris Saclay, Faculté de Médecine Le Kremlin Bicêtre, 94800 Villejuif, France
| | - Gioacchino Natoli
- European Institute of Oncology (IEO), Department of Experimental Oncology, IFOM-IEO Campus, 20139 Milan, Italy
| | - Marie Annick Buendia
- INSERM, U1193, Paul-Brousse Hospital, Hepatobiliary Centre, 94800 Villejuif, France; Université Paris Saclay, Faculté de Médecine Le Kremlin Bicêtre, 94800 Villejuif, France
| | - Jamila Faivre
- INSERM, U1193, Paul-Brousse Hospital, Hepatobiliary Centre, 94800 Villejuif, France; Université Paris Saclay, Faculté de Médecine Le Kremlin Bicêtre, 94800 Villejuif, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Pôle de Biologie Médicale, Paul-Brousse Hospital, 94800 Villejuif, France
| | - Piero Carninci
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa, 230-0045 Japan
| |
Collapse
|
57
|
Lehman HL, Stairs DB. Single and Multiple Gene Manipulations in Mouse Models of Human Cancer. CANCER GROWTH AND METASTASIS 2015; 8:1-15. [PMID: 26380553 PMCID: PMC4558888 DOI: 10.4137/cgm.s21217] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/17/2015] [Accepted: 06/20/2015] [Indexed: 12/14/2022]
Abstract
Mouse models of human cancer play a critical role in understanding the molecular and cellular mechanisms of tumorigenesis. Advances continue to be made in modeling human disease in a mouse, though the relevance of a mouse model often relies on how closely it is able to mimic the histologic, molecular, and physiologic characteristics of the respective human cancer. A classic use of a genetically engineered mouse in studying cancer is through the overexpression or deletion of a gene. However, the manipulation of a single gene often falls short of mimicking all the characteristics of the carcinoma in humans; thus a multiple gene approach is needed. Here we review genetic mouse models of cancers and their abilities to recapitulate human carcinoma with single versus combinatorial approaches with genes commonly involved in cancer.
Collapse
Affiliation(s)
- Heather L Lehman
- Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Douglas B Stairs
- Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
58
|
Li T, Apte U. Bile Acid Metabolism and Signaling in Cholestasis, Inflammation, and Cancer. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 74:263-302. [PMID: 26233910 DOI: 10.1016/bs.apha.2015.04.003] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bile acids are synthesized from cholesterol in the liver. Some cytochrome P450 (CYP) enzymes play key roles in bile acid synthesis. Bile acids are physiological detergent molecules, so are highly cytotoxic. They undergo enterohepatic circulation and play important roles in generating bile flow and facilitating biliary secretion of endogenous metabolites and xenobiotics and intestinal absorption of dietary fats and lipid-soluble vitamins. Bile acid synthesis, transport, and pool size are therefore tightly regulated under physiological conditions. In cholestasis, impaired bile flow leads to accumulation of bile acids in the liver, causing hepatocyte and biliary injury and inflammation. Chronic cholestasis is associated with fibrosis, cirrhosis, and eventually liver failure. Chronic cholestasis also increases the risk of developing hepatocellular or cholangiocellular carcinomas. Extensive research in the last two decades has shown that bile acids act as signaling molecules that regulate various cellular processes. The bile acid-activated nuclear receptors are ligand-activated transcriptional factors that play critical roles in the regulation of bile acid, drug, and xenobiotic metabolism. In cholestasis, these bile acid-activated receptors regulate a network of genes involved in bile acid synthesis, conjugation, transport, and metabolism to alleviate bile acid-induced inflammation and injury. Additionally, bile acids are known to regulate cell growth and proliferation, and altered bile acid levels in diseased conditions have been implicated in liver injury/regeneration and tumorigenesis. We will cover the mechanisms that regulate bile acid homeostasis and detoxification during cholestasis, and the roles of bile acids in the initiation and regulation of hepatic inflammation, regeneration, and carcinogenesis.
Collapse
Affiliation(s)
- Tiangang Li
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, USA.
| | - Udayan Apte
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
59
|
Territo PR, Maluccio M, Riley AA, McCarthy BP, Fletcher J, Tann M, Saxena R, Skill NJ. Evaluation of 11C-acetate and 18F-FDG PET/CT in mouse multidrug resistance gene-2 deficient mouse model of hepatocellular carcinoma. BMC Med Imaging 2015; 15:15. [PMID: 25981587 PMCID: PMC4493966 DOI: 10.1186/s12880-015-0058-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 05/08/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a global health problem with unique diagnostic and therapeutic challenges, including difficulties in identifying the highest risk patients. Previous work from our lab has established the murine multidrug resistance-2 mouse (MDR2) model of HCC as a reasonable preclinical model that parallels the changes seen in human inflammatory associated HCC. The purpose of this study is to evaluate modalities of PET/CT in MDR2(-/-) mice in order to facilitate therapeutic translational studies from bench to bedside. METHODS 18F-FDG and 11C-acetate PET/CT was performed on 12 m MDR2(-/-) mice (n = 3/tracer) with HCC and 12 m MDR2(-/+) control mice (n = 3/tracer) without HCC. To compare PET/CT to biological markers of HCC and cellular function, serum alpha-fetoprotein (AFP), lysophosphatidic acid (LPA), cAMP and hepatic tumor necrosis factor α (TNFα) were quantified in 3-12 m MDR2(-/-) (n = 10) mice using commercially available ELISA analysis. To translate results in mice to patients 11C-acetate PET/CT was also performed in 8 patents suspected of HCC recurrence following treatment and currently on the liver transplant wait list. RESULTS Hepatic18F-FDG metabolism was not significantly increased in MDR2(-/-) mice. In contrast, hepatic 11C-acetate metabolism was significantly elevated in MDR2(-/-) mice when compared to MDR2(-/+) controls. Serum AFP and LPA levels increased in MDR2(-/-) mice contemporaneous with the emergence of HCC. This was accompanied by a significant decrease in serum cAMP levels and an increase in hepatic TNFα. In patients suspected of HCC recurrence there were 5 true positives, 2 true negatives and 1 suspected false 11C-acetate negative. CONCLUSIONS Hepatic 11C-acetate PET/CT tracks well with HCC in MDR2(-/-) mice and patients with underlying liver disease. Consequently 11C-acetate PET/CT is well suited to study (1) HCC emergence/progression in patients and (2) reduce animal numbers required to study new chemotherapeutics in murine models of HCC.
Collapse
Affiliation(s)
- Paul R Territo
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - Mary Maluccio
- Department of Surgery, Indiana University School of Medicine, C519 Walthur Cancer Research Building (R3), 980 W Walnut Street, Indianapolis, IN, 46077, USA.
| | - Amanda A Riley
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - Brian P McCarthy
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - James Fletcher
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - Mark Tann
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - Romil Saxena
- Department of Surgery, Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA.
| | - Nicholas J Skill
- Department of Surgery, Indiana University School of Medicine, C519 Walthur Cancer Research Building (R3), 980 W Walnut Street, Indianapolis, IN, 46077, USA.
| |
Collapse
|
60
|
Stoyanov E, Ludwig G, Mizrahi L, Olam D, Schnitzer-Perlman T, Tasika E, Sass G, Tiegs G, Jiang Y, Nie T, Kohler J, Schinazi RF, Vertino PM, Cedar H, Galun E, Goldenberg D. Chronic liver inflammation modifies DNA methylation at the precancerous stage of murine hepatocarcinogenesis. Oncotarget 2015; 6:11047-60. [PMID: 25918251 PMCID: PMC4484438 DOI: 10.18632/oncotarget.3567] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/26/2015] [Indexed: 02/07/2023] Open
Abstract
Chronic liver inflammation precedes the majority of hepatocellular carcinomas (HCC). Here, we explore the connection between chronic inflammation and DNA methylation in the liver at the late precancerous stages of HCC development in Mdr2(-/-) (Mdr2/Abcb4-knockout) mice, a model of inflammation-mediated HCC. Using methylated DNA immunoprecipitation followed by hybridization with "CpG islands" (CGIs) microarrays, we found specific CGIs in 76 genes which were hypermethylated in the Mdr2(-/-) liver compared to age-matched healthy controls. The observed hypermethylation resulted mainly from an age-dependent decrease of methylation of the specific CGIs in control livers with no decrease in mutant mice. Chronic inflammation did not change global levels of DNA methylation in Mdr2(-/-) liver, but caused a 2-fold decrease of the global 5-hydroxymethylcytosine level in mutants compared to controls. Liver cell fractionation revealed, that the relative hypermethylation of specific CGIs in Mdr2(-/-) livers affected either hepatocyte, or non-hepatocyte, or both fractions without a correlation between changes of gene methylation and expression. Our findings demonstrate that chronic liver inflammation causes hypermethylation of specific CGIs, which may affect both hepatocytes and non-hepatocyte liver cells. These changes may serve as useful markers of an increased regenerative activity and of a late precancerous stage in the chronically inflamed liver.
Collapse
Affiliation(s)
- Evgeniy Stoyanov
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Guy Ludwig
- Department of Developmental Biology and Cancer Research, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Lina Mizrahi
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Devorah Olam
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Temima Schnitzer-Perlman
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Elena Tasika
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Sass
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yong Jiang
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Veterans Affairs Medical Center, Decatur, GA, USA
| | - Ting Nie
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Veterans Affairs Medical Center, Decatur, GA, USA
| | - James Kohler
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Veterans Affairs Medical Center, Decatur, GA, USA
| | - Raymond F. Schinazi
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Veterans Affairs Medical Center, Decatur, GA, USA
| | - Paula M. Vertino
- Department of Radiation Oncology and the Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Howard Cedar
- Department of Developmental Biology and Cancer Research, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Eithan Galun
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Daniel Goldenberg
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
61
|
Han J, Liu Y, Wang R, Yang J, Ling V, Borchers CH. Metabolic profiling of bile acids in human and mouse blood by LC-MS/MS in combination with phospholipid-depletion solid-phase extraction. Anal Chem 2015; 87:1127-36. [PMID: 25496250 DOI: 10.1021/ac503816u] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
To obtain a more comprehensive profile of bile acids (BAs) in blood, we developed an ultrahigh performance liquid chromatography/multiple-reaction monitoring-mass spectrometry (UPLC-MRM-MS) method for the separation and detection of 50 known BAs. This method utilizes phospholipid-depletion solid-phase extraction as a new high-efficiency sample preparation procedure for BA assay. UPLC/scheduled MRM-MS with negative ion electrospray ionization enabled targeted quantitation of 43 and 44 BAs, respectively, in serum samples from seven individuals with and without fasting, as well as in plasma samples from six cholestatic gene knockout mice and six age- and gender-matched wild-type (FVB/NJ) animals. Many minor BAs were identified and quantitated in the blood for the first time. Method validation indicated good quantitation precision with intraday and interday relative standard deviations of ≤9.3% and ≤10.8%, respectively. Using a pooled human serum sample and a pooled mouse plasma sample as the two representative test samples, the quantitation accuracy was measured to be 80% to 120% for most of the BAs, using two standard-substance spiking approaches. To profile other potential BAs not included in the 50 known targets from the knockout versus wild-type mouse plasma, class-specific precursor/fragment ion transitions were used to perform UPLC-MRM-MS for untargeted detection of the structural isomers of glycine- and taurine-conjugated BAs and unconjugated tetra-hydroxy BAs. As a result, as many as 36 such compounds were detected. In summary, this UPLC-MRM-MS method has enabled the quantitation of the largest number of BAs in the blood thus far, and the results presented have revealed an unexpectedly complex BA profile in mouse plasma.
Collapse
Affiliation(s)
- Jun Han
- University of Victoria-Genome BC Proteomics Centre, University of Victoria , Vancouver Island Technology Park, 3101-4464 Markham Street, Victoria, British Columbia V8Z 7X8, Canada
| | | | | | | | | | | |
Collapse
|
62
|
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide, and the third leading cause of cancer mortality. The great majority of patients are not eligible for curative therapies, and therapeutic approaches for advanced disease show only limited efficacy. Difficulties to treat HCC are due to the heterogenous genetic alterations of HCC, profound alterations in the hepatic microenvironment, and incomplete understanding of HCC biology. Mouse models of HCC will be helpful to improve our understanding of HCC biology, the contributions of the specific pathways and genetic alterations to carcinogenesis. In addition, mouse models of HCC may contribute to elucidate the role of the tumor microenvironment, and serve as models for preclinical studies. As no single mouse model is appropriate to study all of the above, we discuss key features and limitations of commonly used models. Furthermore, we provide detailed protocols for select models, in which HCC is induced genetically, chemically or by transplantation of tumor cells.
Collapse
Affiliation(s)
- Jorge Matias Caviglia
- Department of Medicine, Columbia University, Russ Berrie Pavilion, Room 415, 1150 St. Nicholas Ave, New York, NY, 10032, USA
| | | |
Collapse
|
63
|
Ella E, Heim D, Stoyanov E, Harari-Steinfeld R, Steinfeld I, Pappo O, Perlman TS, Nachmansson N, Rivkin L, Olam D, Abramovitch R, Wege H, Galun E, Goldenberg D. Specific genomic and transcriptomic aberrations in tumors induced by partial hepatectomy of a chronically inflamed murine liver. Oncotarget 2014; 5:10318-31. [PMID: 25401338 PMCID: PMC4279375 DOI: 10.18632/oncotarget.2515] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 09/24/2014] [Indexed: 11/25/2022] Open
Abstract
Resection of hepatocellular carcinoma (HCC) tumors by partial hepatectomy (PHx) is associated with promoting hepatocarcinogenesis. We have previously reported that PHx promotes hepatocarcinogenesis in the Mdr2-knockout (Mdr2-KO) mouse, a model for inflammation-mediated HCC. Now, to explore the molecular mechanisms underlying the tumor-promoting effect of PHx, we compared genomic and transcriptomic profiles of HCC tumors developing in the Mdr2-KO mice either spontaneously or following PHx. PHx accelerated HCC development in these mice by four months. PHx-induced tumors had major chromosomal aberrations: all were amplifications affecting multiple chromosomes. Most of these amplifications were located near the acrocentric centromeres of murine chromosomes. Four different chromosomal regions were amplified each in at least three tumors. The human orthologs of these common amplified regions are known to be amplified in HCC. All tumors of untreated mice had chromosomal aberrations, including both deletions and amplifications. Amplifications in spontaneous tumors affected fewer chromosomes and were not located preferentially at the chromosomal edges. Comparison of gene expression profiles revealed a significantly enriched expression of oncogenes, chromosomal instability markers and E2F1 targets in the post-PHx compared to spontaneous tumors. Both tumor groups shared the same frequent amplification at chromosome 18. Here, we revealed that one of the regulatory genes encoded by this amplified region, Crem, was over-expressed in the nuclei of murine and human HCC cells in vivo, and that it stimulated proliferation of human HCC cells in vitro. Our results demonstrate that PHx of a chronically inflamed liver directed tumor development to a discrete pathway characterized by amplification of specific chromosomal regions and expression of specific tumor-promoting genes. Crem is a new candidate HCC oncogene frequently amplified in this model and frequently over-expressed in human HCC.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Animals
- Carcinogenesis/genetics
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/surgery
- Cell Line, Tumor
- Chromosome Aberrations
- Chromosomes, Human, Pair 18/genetics
- Cyclic AMP Response Element Modulator/genetics
- Cyclic AMP Response Element Modulator/metabolism
- Disease Models, Animal
- E2F1 Transcription Factor/genetics
- E2F1 Transcription Factor/metabolism
- Gene Amplification
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Hepatectomy
- Hepatitis, Chronic/genetics
- Hepatitis, Chronic/surgery
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/surgery
- Mice
- Mice, Knockout
- Postoperative Complications/genetics
- Up-Regulation
- ATP-Binding Cassette Sub-Family B Member 4
Collapse
Affiliation(s)
- Ezra Ella
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Denise Heim
- Department of Gastroenterology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Evgeniy Stoyanov
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Rona Harari-Steinfeld
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Israel Steinfeld
- Computer Science Department, Technion-Israel Institute of Technology, Haifa, Israel
| | - Orit Pappo
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Temima Schnitzer Perlman
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Natalie Nachmansson
- Magnetic Resonance Imaging/Magnetic Resonance Spectroscopy Laboratory, Human Biology Research Center, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ludmila Rivkin
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Devorah Olam
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Rinat Abramovitch
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- Magnetic Resonance Imaging/Magnetic Resonance Spectroscopy Laboratory, Human Biology Research Center, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Henning Wege
- Department of Gastroenterology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eithan Galun
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Daniel Goldenberg
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
64
|
Shibasaki Y, Sakaguchi T, Hiraide T, Morita Y, Suzuki A, Baba S, Setou M, Konno H. Expression of indocyanine green-related transporters in hepatocellular carcinoma. J Surg Res 2014; 193:567-76. [PMID: 25173835 DOI: 10.1016/j.jss.2014.07.055] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 07/09/2014] [Accepted: 07/23/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND Indocyanine green (ICG), an organic anion used in liver function tests, is known to accumulate in hepatocellular carcinoma (HCC) tissues after an intravenous injection. Because the intratumoral expression of transporters for chemical agents influences the behaviors of some malignant tumors, we investigated whether the expression of ICG-related transporters influenced the clinicopathologic features of HCC. MATERIALS AND METHODS ICG accumulation patterns were examined using near-infrared spectroscopy and the protein expression of ICG-related transporters was assessed using immunohistochemistry and immunoblotting in 40 resected HCC specimens. We also determined whether the intratumor expression of these transporters affected intratumor lipid composition by imaging mass spectrometry. RESULTS Immunoblot analysis revealed that the expression of organic anion transporting polypeptide 1B3 (OATP1B3) and multidrug resistance p-glycoprotein (MDR)-3, as influx and efflux transporters, respectively, were significantly higher in ICG-accumulated HCC (ICG-high HCC) than in ICG-low HCC. ICG was fluorescently observed in the pseudoglands and bile canaliculi abundantly expressing MDR3. An immunohistochemical examination revealed significantly worse disease-free and overall survival rates in patients with MDR3-negative HCC, in which the intratumoral accumulation of some phosphatidylcholine species was observed under imaging mass spectrometry. CONCLUSIONS The intratumoral expression of MDR3, a key efflux transporter of ICG, affected the prognosis of patients with HCC, presumably by altering the lipid composition of the lipid bilayers.
Collapse
Affiliation(s)
- Yasushi Shibasaki
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Takanori Sakaguchi
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takanori Hiraide
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yoshifumi Morita
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Atsushi Suzuki
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Satoshi Baba
- Faculty of Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mitsutoshi Setou
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroyuki Konno
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
65
|
Iannelli F, Collino A, Sinha S, Radaelli E, Nicoli P, D'Antiga L, Sonzogni A, Faivre J, Buendia MA, Sturm E, Thompson RJ, Knisely AS, Natoli G, Ghisletti S, Ciccarelli FD. Massive gene amplification drives paediatric hepatocellular carcinoma caused by bile salt export pump deficiency. Nat Commun 2014; 5:3850. [PMID: 24819516 DOI: 10.1038/ncomms4850] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 04/10/2014] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is almost invariably associated with an underlying inflammatory state, whose direct contribution to the acquisition of critical genomic changes is unclear. Here we map acquired genomic alterations in human and mouse HCCs induced by defects in hepatocyte biliary transporters, which expose hepatocytes to bile salts and cause chronic inflammation that develops into cancer. In both human and mouse cancer genomes, we find few somatic point mutations with no impairment of cancer genes, but massive gene amplification and rearrangements. This genomic landscape differs from that of virus- and alcohol-associated liver cancer. Copy-number gains preferentially occur at late stages of cancer development and frequently target the MAPK signalling pathway, and in particular direct regulators of JNK. The pharmacological inhibition of JNK retards cancer progression in the mouse. Our study demonstrates that intrahepatic cholestasis leading to hepatocyte exposure to bile acids and inflammation promotes cancer through genomic modifications that can be distinguished from those determined by other aetiological factors.
Collapse
Affiliation(s)
- Fabio Iannelli
- 1] European Institute of Oncology (IEO), Department of Experimental Oncology, IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy [2]
| | - Agnese Collino
- 1] European Institute of Oncology (IEO), Department of Experimental Oncology, IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy [2]
| | - Shruti Sinha
- 1] European Institute of Oncology (IEO), Department of Experimental Oncology, IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy [2] Division of Cancer Studies, King's College London, London SE1 1UL, UK [3]
| | - Enrico Radaelli
- VIB Center for the Biology of Disease, KU Leuven Center for Human Genetics, O&N4 Herestraat 49 box 602, B-3000 Leuven, Belgium
| | - Paola Nicoli
- European Institute of Oncology (IEO), Department of Experimental Oncology, IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy
| | - Lorenzo D'Antiga
- Paediatric Hepatology, Gastroenterology and Transplantation, Ospedale Papa Giovanni XXIII, Piazza OMS - Organizzazione Mondiale della Sanità 1, 24128 Bergamo, Italy
| | - Aurelio Sonzogni
- Department of Pathology, Ospedale Papa Giovanni XXIII, Piazza OMS - Organizzazione Mondiale della Sanità 1, 24128 Bergamo, Italy
| | - Jamila Faivre
- Institut National de la Santé et de la Recherche Médicale (INSERM) U785, University Paris-Sud, France, Centre Hépatobiliaire, Hôpital Paul Brousse, Villejuif F94800, France
| | - Marie Annick Buendia
- Institut National de la Santé et de la Recherche Médicale (INSERM) U785, University Paris-Sud, France, Centre Hépatobiliaire, Hôpital Paul Brousse, Villejuif F94800, France
| | - Ekkehard Sturm
- University Hospital for Children and Adolescents, University of Tuebingen, 72076 Tuebingen, Germany
| | | | - A S Knisely
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, UK
| | - Gioacchino Natoli
- European Institute of Oncology (IEO), Department of Experimental Oncology, IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy
| | - Serena Ghisletti
- European Institute of Oncology (IEO), Department of Experimental Oncology, IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy
| | - Francesca D Ciccarelli
- 1] European Institute of Oncology (IEO), Department of Experimental Oncology, IFOM-IEO Campus, Via Adamello 16, 20139 Milan, Italy [2] Division of Cancer Studies, King's College London, London SE1 1UL, UK
| |
Collapse
|
66
|
Cuperus FJC, Claudel T, Gautherot J, Halilbasic E, Trauner M. The role of canalicular ABC transporters in cholestasis. Drug Metab Dispos 2014; 42:546-60. [PMID: 24474736 DOI: 10.1124/dmd.113.056358] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cholestasis, a hallmark feature of hepatobiliary disease, is characterized by the retention of biliary constituents. Some of these constituents, such as bile acids, inflict damage to hepatocytes and bile duct cells. This damage may lead to inflammation, fibrosis, cirrhosis, and eventually carcinogenesis, sequelae that aggravate the underlying disease and deteriorate clinical outcome. Canalicular ATP-binding cassette (ABC) transporters, which mediate the excretion of individual bile constituents, play a key role in bile formation and cholestasis. The study of these transporters and their regulatory nuclear receptors has revolutionized our understanding of cholestatic disease. This knowledge has served as a template to develop novel treatment strategies, some of which are currently already undergoing phase III clinical trials. In this review we aim to provide an overview of the structure, function, and regulation of canalicular ABC transporters. In addition, we will focus on the role of these transporters in the pathogenesis and treatment of cholestatic bile duct and liver diseases.
Collapse
Affiliation(s)
- Frans J C Cuperus
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | | | | | | | | |
Collapse
|
67
|
Condiotti R, Goldenberg D, Giladi H, Schnitzer-Perlman T, Waddington SN, Buckley SM, Heim D, Cheung W, Themis M, Coutelle C, Simerzin A, Osejindu E, Wege H, Themis M, Galun E. Transduction of fetal mice with a feline lentiviral vector induces liver tumors which exhibit an E2F activation signature. Mol Ther 2013; 22:59-68. [PMID: 23982166 DOI: 10.1038/mt.2013.193] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 08/12/2013] [Indexed: 01/10/2023] Open
Abstract
Lentiviral vectors are widely used in basic research and clinical applications for gene transfer and long-term expression; however, safety issues have not yet been completely resolved. In this study, we characterized hepatocarcinomas that developed in mice 1 year after in utero administration of a feline-derived lentiviral vector. Mapped viral integration sites differed among tumors and did not coincide with the regions of chromosomal aberrations. Furthermore, gene expression profiling revealed that no known cancer-associated genes were deregulated in the vicinity of viral integrations. Nevertheless, five of the six tumors exhibited highly significant upregulation of E2F target genes, of which a majority are associated with oncogenesis, DNA damage response, and chromosomal instability. We further show in vivo and in vitro that E2F activation occurs early on following transduction of both fetal mice and cultured human hepatocytes. On the basis of the similarities in E2F target gene expression patterns among tumors and the lack of evidence implicating insertional mutagenesis, we propose that transduction of fetal mice with a feline lentiviral vector induces E2F-mediated major cellular processes that drive hepatocytes toward uncontrolled proliferation culminating in tumorigenesis.
Collapse
Affiliation(s)
- Reba Condiotti
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Daniel Goldenberg
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Hilla Giladi
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Temima Schnitzer-Perlman
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Simon N Waddington
- 1] Institute for Women's Health, University College London, London, UK [2] School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Suzanne Mk Buckley
- Department of Haematology, University College Medical School, London, UK
| | - Denise Heim
- Department of Gastroenterology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wing Cheung
- Institute for Women's Health, University College London, London, UK
| | - Matthew Themis
- Division of Biosciences, Brunel University, Middlesex, UK
| | | | - Alina Simerzin
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Emma Osejindu
- Division of Biosciences, Brunel University, Middlesex, UK
| | - Henning Wege
- Department of Gastroenterology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Themis
- 1] Division of Biosciences, Brunel University, Middlesex, UK [2] National Heart and Lung Institute, Imperial College, London, UK
| | - Eithan Galun
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
68
|
Potikha T, Stoyanov E, Pappo O, Frolov A, Mizrahi L, Olam D, Shnitzer-Perlman T, Weiss I, Barashi N, Peled A, Sass G, Tiegs G, Poirier F, Rabinovich GA, Galun E, Goldenberg D. Interstrain differences in chronic hepatitis and tumor development in a murine model of inflammation-mediated hepatocarcinogenesis. Hepatology 2013; 58:192-204. [PMID: 23423643 DOI: 10.1002/hep.26335] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 02/10/2013] [Indexed: 02/06/2023]
Abstract
UNLABELLED Chronic inflammation is strongly associated with an increased risk for hepatocellular carcinoma (HCC) development. The multidrug resistance 2 (Mdr2)-knockout (KO) mouse (adenosine triphosphate-binding cassette b4(-/-) ), a model of inflammation-mediated HCC, develops chronic cholestatic hepatitis at an early age and HCC at an adult age. To delineate factors contributing to hepatocarcinogenesis, we compared the severity of early chronic hepatitis and late HCC development in two Mdr2-KO strains: Friend virus B-type/N (FVB) and C57 black 6 (B6). We demonstrated that hepatocarcinogenesis was significantly less efficient in the Mdr2-KO/B6 mice versus the Mdr2-KO/FVB mice; this difference was more prominent in males. Chronic hepatitis in the Mdr2-KO/B6 males was more severe at 1 month of age but was less severe at 3 months of age in comparison with age-matched Mdr2-KO/FVB males. A comparative genome-scale gene expression analysis of male livers of both strains at 3 months of age revealed both common and strain-specific aberrantly expressed genes, including genes associated with the regulation of inflammation, the response to oxidative stress, and lipid metabolism. One of these regulators, galectin-1 (Gal-1), possesses both anti-inflammatory and protumorigenic activities. To study its regulatory role in the liver, we transferred the Gal-1-KO mutation (lectin galactoside-binding soluble 1(-/-) ) from the B6 strain to the FVB strain, and we demonstrated that endogenous Gal-1 protected the liver against concanavalin A-induced hepatitis with the B6 genetic background but not the FVB genetic background. CONCLUSION Decreased chronic hepatitis in Mdr2-KO/B6 mice at the age of 3 months correlated with a significant retardation of liver tumor development in this strain versus the Mdr2-KO/FVB strain. We found candidate factors that may determine strain-specific differences in the course of chronic hepatitis and HCC development in the Mdr2-KO model, including inefficient anti-inflammatory activity of the endogenous lectin Gal-1 in the FVB strain.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/deficiency
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Animals
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/pathology
- Cell Transformation, Neoplastic/pathology
- Chemical and Drug Induced Liver Injury/prevention & control
- Concanavalin A
- Galectin 1/physiology
- Hepatitis, Chronic/complications
- Hepatitis, Chronic/etiology
- Hepatitis, Chronic/pathology
- Liver/metabolism
- Liver Neoplasms/etiology
- Liver Neoplasms/pathology
- Male
- Methionine Adenosyltransferase/biosynthesis
- Mice
- Mice, Inbred Strains/genetics
- Mice, Knockout
- ATP-Binding Cassette Sub-Family B Member 4
Collapse
Affiliation(s)
- Tamara Potikha
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Liu Y, Meyer C, Xu C, Weng H, Hellerbrand C, ten Dijke P, Dooley S. Animal models of chronic liver diseases. Am J Physiol Gastrointest Liver Physiol 2013; 304:G449-68. [PMID: 23275613 DOI: 10.1152/ajpgi.00199.2012] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Chronic liver diseases are frequent and potentially life threatening for humans. The underlying etiologies are diverse, ranging from viral infections, autoimmune disorders, and intoxications (including alcohol abuse) to imbalanced diets. Although at early stages of disease the liver regenerates in the absence of the insult, advanced stages cannot be healed and may require organ transplantation. A better understanding of underlying mechanisms is mandatory for the design of new drugs to be used in clinic. Therefore, rodent models are being developed to mimic human liver disease. However, no model to date can completely recapitulate the "corresponding" human disorder. Limiting factors are the time frame required in humans to establish a certain liver disease and the fact that rodents possess a distinct immune system compared with humans and have different metabolic rates affecting liver homeostasis. These features account for the difficulties in developing adequate rodent models for studying disease progression and for testing new pharmaceuticals to be translated into the clinic. Nevertheless, traditional and new promising animal models that mimic certain attributes of chronic liver diseases are established and being used to deepen our understanding in the underlying mechanisms of distinct liver diseases. This review aims at providing a comprehensive overview of recent advances in animal models recapitulating different features and etiologies of human liver diseases.
Collapse
Affiliation(s)
- Yan Liu
- Department of Medicine II, Section Molecular Hepatology-Alcohol Associated Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | | | | | | | | | | |
Collapse
|
70
|
Hernandez-Gea V, Toffanin S, Friedman SL, Llovet JM. Role of the microenvironment in the pathogenesis and treatment of hepatocellular carcinoma. Gastroenterology 2013; 144:512-27. [PMID: 23313965 PMCID: PMC3578068 DOI: 10.1053/j.gastro.2013.01.002] [Citation(s) in RCA: 574] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 12/03/2012] [Accepted: 01/07/2013] [Indexed: 12/02/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver tumor and the third greatest cause of cancer-related death worldwide, and its incidence is increasing. Despite the significant improvement in management of HCC over the past 30 years, there are no effective chemoprevention strategies, and only one systemic therapy has been approved for patients with advanced tumors. This drug, sorafenib, acts on tumor cells and the stroma. HCC develops from chronically damaged tissue that contains large amounts of inflammation and fibrosis, which also promote tumor progression and resistance to therapy. Increasing our understanding of how stromal components interact with cancer cells and the signaling pathways involved could help identify new therapeutic and chemopreventive targets.
Collapse
Affiliation(s)
| | - Sara Toffanin
- Division of Liver Diseases, Mount Sinai School of Medicine, New York, New York, USA
- Gastrointestinal Surgery and Liver Transplantation Unit, National Cancer Institute, IRCSS Foundation, Milan, Italy
| | - Scott L. Friedman
- Division of Liver Diseases, Mount Sinai School of Medicine, New York, New York, USA
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases, Tisch Cancer Institute), Mount Sinai School of Medicine, New York, New York, USA
| | - Josep M. Llovet
- Division of Liver Diseases, Mount Sinai School of Medicine, New York, New York, USA
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases, Tisch Cancer Institute), Mount Sinai School of Medicine, New York, New York, USA
- HCC Translational Research Laboratory, Barcelona-Clínic Liver Cancer Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Liver Unit and Pathology Department. Hospital Clinic, Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Catalonia, Spain
- University of Barcelona, Catalonia, Spain
| |
Collapse
|
71
|
Abstract
Hepatocellular carcinoma (HCC), the most common form of primary liver cancer is the third leading cause of cancer-related cell death in human and the fifth in women worldwide. The incidence of HCC is increasing despite progress in identifying risk factors, understanding disease etiology and developing anti-viral strategies. Therapeutic options are limited and survival after diagnosis is poor. Therefore, better preventive, diagnostic and therapeutic tools are urgently needed, in particular given the increased contribution from systemic metabolic disease to HCC incidence worldwide. In the last three decades, technological advances have facilitated the generation of genetically engineered mouse models (GEMMs) to mimic the alterations frequently observed in human cancers or to conduct intervention studies and assess the relevance of candidate gene networks in tumor establishment, progression and maintenance. Because these studies allow molecular and cellular manipulations impossible to perform in patients, GEMMs have improved our understanding of this complex disease and represent a source of great potential for mechanism-based therapy development. In this review, we provide an overview of the current state of HCC modeling in the mouse, highlighting successes, current challenges and future opportunities.
Collapse
|
72
|
Ehlken H, Kondylis V, Heinrichsdorff J, Ochoa-Callejero L, Roskams T, Pasparakis M. Hepatocyte IKK2 protects Mdr2-/- mice from chronic liver failure. PLoS One 2011; 6:e25942. [PMID: 22022477 PMCID: PMC3195080 DOI: 10.1371/journal.pone.0025942] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 09/14/2011] [Indexed: 11/23/2022] Open
Abstract
UNLABELLED Mice lacking the Abc4 protein encoded by the multidrug resistance-2 gene (Mdr2(-/-)) develop chronic periductular inflammation and cholestatic liver disease resulting in the development of hepatocellular carcinoma (HCC). Inhibition of NF-κB by expression of an IκBα super-repressor (IκBαSR) transgene in hepatocytes was shown to prevent HCC development in Mdr2(-/-) mice, suggesting that NF-κB acts as a tumour promoter in this model of inflammation-associated carcinogenesis. On the other hand, inhibition of NF-κB by hepatocyte specific ablation of IKK2 resulted in increased liver tumour development induced by the chemical carcinogen DEN. To address the role of IKK2-mediated NF-κB activation in hepatocytes in the pathogenesis of liver disease and HCC in Mdr2(-/-) mice, we generated Mdr2-deficient animals lacking IKK2 specifically in hepatocytes using the Cre-loxP system. Mdr2(-/-) mice lacking IKK2 in hepatocytes developed spontaneously a severe liver disease characterized by cholestasis, major hyperbilirubinemia and severe to end-stage fibrosis, which caused muscle wasting, loss of body weight, lethargy and early spontaneous death. Cell culture experiments showed that primary hepatocytes lacking IKK2 were more sensitive to bile acid induced death, suggesting that hepatocyte-specific IKK2 deficiency sensitized hepatocytes to the toxicity of bile acids under conditions of cholestasis resulting in greatly exacerbated liver damage. Mdr2(-/-)IKK2(Hep-KO) mice remarkably recapitulate chronic liver failure in humans and might be of special importance for the study of the mechanisms contributing to the pathogenesis of end-stage chronic liver disease or its implications on other organs. CONCLUSION IKK2-mediated signaling in hepatocytes protects the liver from damage under conditions of chronic inflammatory cholestasis and prevents the development of severe fibrosis and liver failure.
Collapse
Affiliation(s)
- Hanno Ehlken
- Institute for Genetics, Centre for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Vangelis Kondylis
- Institute for Genetics, Centre for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jan Heinrichsdorff
- Institute for Genetics, Centre for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Laura Ochoa-Callejero
- Institute for Genetics, Centre for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Tania Roskams
- Department of Pathology, Catholic University of Leuven, Leuven, Belgium
| | - Manolis Pasparakis
- Institute for Genetics, Centre for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
73
|
Philips GM, Chan IS, Swiderska M, Schroder VT, Guy C, Karaca GF, Moylan C, Venkatraman T, Feuerlein S, Syn WK, Jung Y, Witek RP, Choi S, Michelotti GA, Rangwala F, Merkle E, Lascola C, Diehl AM. Hedgehog signaling antagonist promotes regression of both liver fibrosis and hepatocellular carcinoma in a murine model of primary liver cancer. PLoS One 2011; 6:e23943. [PMID: 21912653 PMCID: PMC3166282 DOI: 10.1371/journal.pone.0023943] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 07/27/2011] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Chronic fibrosing liver injury is a major risk factor for hepatocarcinogenesis in humans. Mice with targeted deletion of Mdr2 (the murine ortholog of MDR3) develop chronic fibrosing liver injury. Hepatocellular carcinoma (HCC) emerges spontaneously in such mice by 50-60 weeks of age, providing a model of fibrosis-associated hepatocarcinogenesis. We used Mdr2(-/-) mice to investigate the hypothesis that activation of the hedgehog (Hh) signaling pathway promotes development of both liver fibrosis and HCC. METHODS Hepatic injury and fibrosis, Hh pathway activation, and liver progenitor populations were compared in Mdr2(-/-) mice and age-matched wild type controls. A dose finding experiment with the Hh signaling antagonist GDC-0449 was performed to optimize Hh pathway inhibition. Mice were then treated with GDC-0449 or vehicle for 9 days, and effects on liver fibrosis and tumor burden were assessed by immunohistochemistry, qRT-PCR, Western blot, and magnetic resonance imaging. RESULTS Unlike controls, Mdr2(-/-) mice consistently expressed Hh ligands and progressively accumulated Hh-responsive liver myofibroblasts and progenitors with age. Treatment of aged Mdr2-deficient mice with GDC-0449 significantly inhibited hepatic Hh activity, decreased liver myofibroblasts and progenitors, reduced liver fibrosis, promoted regression of intra-hepatic HCCs, and decreased the number of metastatic HCC without increasing mortality. CONCLUSIONS Hh pathway activation promotes liver fibrosis and hepatocarcinogenesis, and inhibiting Hh signaling safely reverses both processes even when fibrosis and HCC are advanced.
Collapse
Affiliation(s)
- George M. Philips
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Isaac S. Chan
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Genetics, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Marzena Swiderska
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Vanessa T. Schroder
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Cynthia Guy
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Gamze F. Karaca
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Cynthia Moylan
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Talaignair Venkatraman
- Department of Radiology, Duke University, Durham, North Carolina, United States of America
| | - Sebastian Feuerlein
- Department of Radiology, Duke University, Durham, North Carolina, United States of America
| | - Wing-Kin Syn
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Youngmi Jung
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Biological Science, Pusan National University, Pusan, Korea
| | - Rafal P. Witek
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Steve Choi
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Gregory A. Michelotti
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Fatima Rangwala
- Divisions of Cell Therapy, Hematology and Medical Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Elmar Merkle
- Department of Radiology, Duke University, Durham, North Carolina, United States of America
| | - Christopher Lascola
- Department of Radiology, Duke University, Durham, North Carolina, United States of America
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
74
|
Latasa MU, Gil-Puig C, Fernández-Barrena MG, Rodríguez-Ortigosa CM, Banales JM, Urtasun R, Goñi S, Méndez M, Arcelus S, Juanarena N, Recio JA, Lotersztajn S, Prieto J, Berasain C, Corrales FJ, Lecanda J, Ávila MA. Oral methylthioadenosine administration attenuates fibrosis and chronic liver disease progression in Mdr2-/- mice. PLoS One 2010; 5:e15690. [PMID: 21209952 PMCID: PMC3012093 DOI: 10.1371/journal.pone.0015690] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 11/21/2010] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Inflammation and fibrogenesis are directly related to chronic liver disease progression, including hepatocellular carcinoma (HCC) development. Currently there are few therapeutic options available to inhibit liver fibrosis. We have evaluated the hepatoprotective and anti-fibrotic potential of orally-administered 5'-methylthioadenosine (MTA) in Mdr2(-/-) mice, a clinically relevant model of sclerosing cholangitis and spontaneous biliary fibrosis, followed at later stages by HCC development. METHODOLOGY MTA was administered daily by gavage to wild type and Mdr2(-/-) mice for three weeks. MTA anti-inflammatory and anti-fibrotic effects and potential mechanisms of action were examined in the liver of Mdr2(-/-) mice with ongoing fibrogenesis and in cultured liver fibrogenic cells (myofibroblasts). PRINCIPAL FINDINGS MTA treatment reduced hepatomegaly and liver injury. α-Smooth muscle actin immunoreactivity and collagen deposition were also significantly decreased. Inflammatory infiltrate, the expression of the cytokines IL6 and Mcp-1, pro-fibrogenic factors like TGFβ2 and tenascin-C, as well as pro-fibrogenic intracellular signalling pathways were reduced by MTA in vivo. MTA inhibited the activation and proliferation of isolated myofibroblasts and down-regulated cyclin D1 gene expression at the transcriptional level. The expression of JunD, a key transcription factor in liver fibrogenesis, was also reduced by MTA in activated myofibroblasts. CONCLUSIONS/SIGNIFICANCE Oral MTA administration was well tolerated and proved its efficacy in reducing liver inflammation and fibrosis. MTA may have multiple molecular and cellular targets. These include the inhibition of inflammatory and pro-fibrogenic cytokines, as well as the attenuation of myofibroblast activation and proliferation. Downregulation of JunD and cyclin D1 expression in myofibroblasts may be important regarding the mechanism of action of MTA. This compound could be a good candidate to be tested for the treatment of (biliary) liver fibrosis.
Collapse
Affiliation(s)
- M. Ujue Latasa
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Carmen Gil-Puig
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- Digna Biotech, Madrid, Spain
| | - Maite G. Fernández-Barrena
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- CIBERehd, University Clinic, University of Navarra, Pamplona, Spain
| | - Carlos M. Rodríguez-Ortigosa
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- CIBERehd, University Clinic, University of Navarra, Pamplona, Spain
| | - Jesús M. Banales
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- CIBERehd, University Clinic, University of Navarra, Pamplona, Spain
| | - Raquel Urtasun
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Saioa Goñi
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Miriam Méndez
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Sara Arcelus
- CIBERehd, University Clinic, University of Navarra, Pamplona, Spain
| | - Nerea Juanarena
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Juan A. Recio
- Vall d'Hebron Research Institute, Institute of Oncology and Hospital, Barcelona, Spain
| | - Sophie Lotersztajn
- Inserm, U955, Créteil, France
- Université Paris-Est, Faculté de Médecine, UMR-S955, Créteil, France
| | - Jesús Prieto
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- CIBERehd, University Clinic, University of Navarra, Pamplona, Spain
| | - Carmen Berasain
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Fernando J. Corrales
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| | - Jon Lecanda
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
- Digna Biotech, Madrid, Spain
| | - Matías A. Ávila
- Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona, Spain
| |
Collapse
|
75
|
Weber SN, Wasmuth HE. Liver fibrosis: from animal models to mapping of human risk variants. Best Pract Res Clin Gastroenterol 2010; 24:635-46. [PMID: 20955966 DOI: 10.1016/j.bpg.2010.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 07/25/2010] [Accepted: 07/28/2010] [Indexed: 02/06/2023]
Abstract
Liver fibrosis is the sequel of chronic liver diseases and the main reason for increased mortality in affected patients. The extent of liver fibrosis displays great interindividual variation, even after controlling for exogenous factors. Thus, host genetic factors are considered to play an important role in the process of liver scarring. From a genetic perspective, liver fibrosis is a complex trait with many genes contributing to the expression of the phenotype. In genetically manipulated and inbred animals several risk loci for liver fibrosis have been identified. Some of these loci have been replicated in case-control studies of patients with hepatitis C infection. In humans, genetic risk loci were identified by single marker studies, haplotype studies or the combination of single markers. Recently, the first genome-wide association studies have also been performed in patients with liver diseases. Some of the identified gene variants have been functionally characterized in vitro, thereby opening the potential for novel therapeutic approaches and risk stratification.
Collapse
Affiliation(s)
- Susanne N Weber
- Department of Medicine II, Saarland University Hospital, Homburg, Germany
| | | |
Collapse
|
76
|
Barash H, R. Gross E, Edrei Y, Ella E, Israel A, Cohen I, Corchia N, Ben-Moshe T, Pappo O, Pikarsky E, Goldenberg D, Shiloh Y, Galun E, Abramovitch R. Accelerated carcinogenesis following liver regeneration is associated with chronic inflammation-induced double-strand DNA breaks. Proc Natl Acad Sci U S A 2010; 107:2207-12. [PMID: 20133864 PMCID: PMC2836653 DOI: 10.1073/pnas.0908867107] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer mortality worldwide and is considered to be the outcome of chronic liver inflammation. Currently, the main treatment for HCC is surgical resection. However, survival rates are suboptimal partially because of tumor recurrence in the remaining liver. Our aim was to understand the molecular mechanisms linking liver regeneration under chronic inflammation to hepatic tumorigenesis. Mdr2-KO mice, a model of inflammation-associated cancer, underwent partial hepatectomy (PHx), which led to enhanced hepatocarcinogenesis. Moreover, liver regeneration in these mice was severely attenuated. We demonstrate the activation of the DNA damage-response machinery and increased genomic instability during early liver inflammatory stages resulting in hepatocyte apoptosis, cell-cycle arrest, and senescence and suggest their involvement in tumor growth acceleration subsequent to PHx. We propose that under the regenerative proliferative stress induced by liver resection, the genomic unstable hepatocytes generated during chronic inflammation escape senescence and apoptosis and reenter the cell cycle, triggering the enhanced tumorigenesis. Thus, we clarify the immediate and long-term contributions of the DNA damage response to HCC development and recurrence.
Collapse
Affiliation(s)
- Hila Barash
- The Goldyne Savad Institute of Gene Therapy
- Magnetic Resonance Imaging/Magnetic Resonance Spectroscopy Laboratory, Human Biology Research Center, and
| | | | - Yifat Edrei
- The Goldyne Savad Institute of Gene Therapy
- Magnetic Resonance Imaging/Magnetic Resonance Spectroscopy Laboratory, Human Biology Research Center, and
| | - Ezra Ella
- The Goldyne Savad Institute of Gene Therapy
| | | | - Irit Cohen
- The Goldyne Savad Institute of Gene Therapy
| | - Nathalie Corchia
- The Goldyne Savad Institute of Gene Therapy
- Magnetic Resonance Imaging/Magnetic Resonance Spectroscopy Laboratory, Human Biology Research Center, and
| | | | - Orit Pappo
- Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel; and
| | - Eli Pikarsky
- Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel; and
| | | | - Yosef Shiloh
- The David and Inez Myers Laboratory for Cancer Research, Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel
| | | | - Rinat Abramovitch
- The Goldyne Savad Institute of Gene Therapy
- Magnetic Resonance Imaging/Magnetic Resonance Spectroscopy Laboratory, Human Biology Research Center, and
| |
Collapse
|
77
|
Lohse AW, Weiler-Normann C, Tiegs G. Immune-mediated liver injury. J Hepatol 2010; 52:136-44. [PMID: 19913936 DOI: 10.1016/j.jhep.2009.10.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2009] [Revised: 09/14/2009] [Accepted: 09/16/2009] [Indexed: 02/06/2023]
Affiliation(s)
- Ansgar W Lohse
- Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
| | | | | |
Collapse
|
78
|
Sánchez A, Fabregat I. Genetically modified animal models recapitulating molecular events altered in human hepatocarcinogenesis. Clin Transl Oncol 2009; 11:208-14. [DOI: 10.1007/s12094-009-0342-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
79
|
Klopstock N, Katzenellenbogen M, Pappo O, Sklair-Levy M, Olam D, Mizrahi L, Potikha T, Galun E, Goldenberg D. HCV tumor promoting effect is dependent on host genetic background. PLoS One 2009; 4:e5025. [PMID: 19340302 PMCID: PMC2660413 DOI: 10.1371/journal.pone.0005025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2008] [Accepted: 02/27/2009] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The hepatitis C virus (HCV) is one of the major risk factors for the development of hepatocellular carcinoma (HCC). Nevertheless, transgenic mice which express the whole HCV polyprotein (HCV-Tg) do not develop HCC. Whereas chronic HCV infection causes inflammation in patients, in HCV-Tg mice, the host immune reaction against viral proteins is lacking. We aimed to test the role of HCV proteins in HCC development on the background of chronic inflammation in vivo. METHODOLOGY/PRINCIPAL FINDINGS We crossed HCV-Tg mice that do not develop HCC with the Mdr2-knockout (Mdr2-KO) mice which develop inflammation-associated HCC, to generate Mdr2-KO/HCV-Tg mice. We studied the effect of the HCV transgene on tumor incidence, hepatocyte mitosis and apoptosis, and investigated the potential contributing factors for the generated phenotype by gene expression and protein analyses. The Mdr2-KO/HCV-Tg females from the N2 generation of this breeding (having 75% of the FVB/N genome and 25% of the C57BL/6 genome) produced significantly larger tumors in comparison with Mdr2-KO mice. In parallel, the Mdr2-KO/HCV-Tg females had an enhanced inflammatory gene expression signature. However, in the N7 generation (having 99.2% of the FVB/N genome and 0.8% of the C57BL/6 genome) there was no difference in tumor development between Mdr2-KO/HCV-Tg and Mdr2-KO animals of both sexes. The HCV transgene was similarly expressed in the livers of Mdr2-KO/HCV-Tg females of both generations, as revealed by detection of the HCV transcript and the core protein. CONCLUSION These findings suggest that the HCV transgene accelerated inflammation-associated hepatocarcinogenesis in a host genetic background-dependent manner.
Collapse
Affiliation(s)
- Naama Klopstock
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Mark Katzenellenbogen
- Bioinformatics and Microarray Unit, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Orit Pappo
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Devorah Olam
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Lina Mizrahi
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tamara Potikha
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Eithan Galun
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Daniel Goldenberg
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
80
|
Trauner M, Fickert P, Halilbasic E, Moustafa T. Lessons from the toxic bile concept for the pathogenesis and treatment of cholestatic liver diseases. Wien Med Wochenschr 2008; 158:542-8. [PMID: 18998069 DOI: 10.1007/s10354-008-0592-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 01/07/2008] [Indexed: 01/03/2023]
Abstract
Alterations in bile secretion at the hepatocellular and cholangiocellular levels may cause cholestasis. Formation of 'toxic bile' may be the consequence of abnormal bile composition and can result in hepatocellular and/or bile duct injury. The canalicular phospholipid flippase (Mdr2/MDR3) normally mediates biliary excretion of phospholipids, which normally form mixed micelles with bile acids and cholesterol to protect the bile duct epithelium from the detergent properties of bile acids. Mdr2 knockout mice are not capable of excreting phospholipids into bile and spontaneously develop bile duct injury with macroscopic and microscopic features closely resembling human sclerosing cholangitis. MDR3 mutations have been linked to a broad spectrum of hepatobiliary disorders in humans ranging from progressive familial intrahepatic cholestasis in neonates to intrahepatic cholestasis of pregnancy, drug-induced cholestasis, intrahepatic cholelithiasis, sclerosing cholangitis and biliary cirrhosis in adults. Other examples for bile injury due to the formation of toxic bile include the cholangiopathy seen in cystic fibrosis, after lithocholate feeding (in mice) and vanishing bile duct syndromes induced by drugs and xenobiotics. Therapeutic strategies for cholangiopathies may target bile composition/toxicity and the affected bile duct epithelium itself, and ideally should also have anti-cholestatic, anti-fibrotic and anti-neoplastic properties. Ursodeoxycholic acid (UDCA) shows some of these properties, but is of limited efficacy in the treatment of human cholangiopathies. By contrast to UDCA, its side chain-shortened homologue norUDCA undergoes cholehepatic shunting leading to a bicarbonate-rich hypercholeresis. Moreover, norUDCA has anti-inflammatory, anti-fibrotic and anti-proliferative effects, and stimulates bile acid detoxification. Upcoming clinical trials will have to demonstrate whether norUDCA or other side chain-modified bile acids are also clinically effective in humans. Finally, drugs for the treatment of cholangiopathies may target bile toxicity via nuclear receptors (FXR, PPARalpha) regulating biliary phospholipid and bile acid excretion.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Animals
- Bile/physiology
- Bile Acids and Salts/toxicity
- Bile Canaliculi/physiopathology
- Bile Duct Neoplasms/genetics
- Bile Duct Neoplasms/physiopathology
- Bile Ducts, Intrahepatic/physiopathology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/physiopathology
- Cell Transformation, Neoplastic/genetics
- Cholangiocarcinoma/genetics
- Cholangiocarcinoma/physiopathology
- Cholangitis, Sclerosing/genetics
- Cholangitis, Sclerosing/physiopathology
- Cholestasis, Intrahepatic/genetics
- Cholestasis, Intrahepatic/physiopathology
- Cholic Acids
- Disease Models, Animal
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/physiopathology
- Mice
- Mice, Knockout
- Mutation/genetics
- Norsteroids
- Ursodeoxycholic Acid/physiology
- ATP-Binding Cassette Sub-Family B Member 4
Collapse
Affiliation(s)
- Michael Trauner
- Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
| | | | | | | |
Collapse
|