51
|
Brivio S, Cadamuro M, Fabris L, Strazzabosco M. Molecular Mechanisms Driving Cholangiocarcinoma Invasiveness: An Overview. Gene Expr 2018; 18:31-50. [PMID: 29070148 PMCID: PMC5860940 DOI: 10.3727/105221617x15088670121925] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The acquisition of invasive functions by tumor cells is a first and crucial step toward the development of metastasis, which nowadays represents the main cause of cancer-related death. Cholangiocarcinoma (CCA), a primary liver cancer originating from the biliary epithelium, typically develops intrahepatic or lymph node metastases at early stages, thus preventing the majority of patients from undergoing curative treatments, consistent with their very poor prognosis. As in most carcinomas, CCA cells gradually adopt a motile, mesenchymal-like phenotype, enabling them to cross the basement membrane, detach from the primary tumor, and invade the surrounding stroma. Unfortunately, little is known about the molecular mechanisms that synergistically orchestrate this proinvasive phenotypic switch. Autocrine and paracrine signals (cyto/chemokines, growth factors, and morphogens) permeating the tumor microenvironment undoubtedly play a prominent role in this context. Moreover, a number of recently identified signaling systems are currently drawing attention as putative mechanistic determinants of CCA cell invasion. They encompass transcription factors, protein kinases and phosphatases, ubiquitin ligases, adaptor proteins, and miRNAs, whose aberrant expression may result from either stochastic mutations or the abnormal activation of upstream pro-oncogenic pathways. Herein we sought to summarize the most relevant molecules in this field and to discuss their mechanism of action and potential prognostic relevance in CCA. Hopefully, a deeper knowledge of the molecular determinants of CCA invasiveness will help to identify clinically useful biomarkers and novel druggable targets, with the ultimate goal to develop innovative approaches to the management of this devastating malignancy.
Collapse
Affiliation(s)
- Simone Brivio
- *School of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Massimiliano Cadamuro
- *School of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- †International Center for Digestive Health, University of Milan-Bicocca, Monza, Italy
| | - Luca Fabris
- †International Center for Digestive Health, University of Milan-Bicocca, Monza, Italy
- ‡Department of Molecular Medicine, University of Padua, Padua, Italy
- §Liver Center, School of Medicine Section of Digestive Diseases, Yale University, New Haven, CT, USA
| | - Mario Strazzabosco
- *School of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- †International Center for Digestive Health, University of Milan-Bicocca, Monza, Italy
- §Liver Center, School of Medicine Section of Digestive Diseases, Yale University, New Haven, CT, USA
| |
Collapse
|
52
|
Ren FH, Yang H, He RQ, Lu JN, Lin XG, Liang HW, Dang YW, Feng ZB, Chen G, Luo DZ. Analysis of microarrays of miR-34a and its identification of prospective target gene signature in hepatocellular carcinoma. BMC Cancer 2018; 18:12. [PMID: 29298665 PMCID: PMC5753510 DOI: 10.1186/s12885-017-3941-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 12/19/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Currently, some studies have demonstrated that miR-34a could serve as a suppressor of several cancers including hepatocellular carcinoma (HCC). Previously, we discovered that miR-34a was downregulated in HCC and involved in the tumorigenesis and progression of HCC; however, the mechanism remains unclear. The purpose of this study was to estimate the expression of miR-34a in HCC by applying the microarray profiles and analyzing the predicted targets of miR-34a and their related biological pathways of HCC. METHODS Gene expression omnibus (GEO) datasets were conducted to identify the difference of miR-34a expression between HCC and corresponding normal tissues and to explore its relationship with HCC clinicopathologic features. The natural language processing (NLP), gene ontology (GO), pathway and network analyses were performed to analyze the genes associated with the carcinogenesis and progression of HCC and the targets of miR-34a predicted in silico. In addition, the integrative analysis was performed to explore the targets of miR-34a which were also relevant to HCC. RESULTS The analysis of GEO datasets demonstrated that miR-34a was downregulated in HCC tissues, and no heterogeneity was observed (Std. Mean Difference(SMD) = 0.63, 95% confidence intervals(95%CI):[0.38, 0.88], P < 0.00001; Pheterogeneity = 0.08 I2 = 41%). However, no association was found between the expression value of miR-34a and any clinicopathologic characteristics. In the NLP analysis of HCC, we obtained 25 significant HCC-associated signaling pathways. Besides, we explored 1000 miR-34a-related genes and 5 significant signaling pathways in which CCND1 and Bcl-2 served as necessary hub genes. In the integrative analysis, we found 61 hub genes and 5 significant pathways, including cell cycle, cytokine-cytokine receptor interaction, notching pathway, p53 pathway and focal adhesion, which proposed the relevant functions of miR-34a in HCC. CONCLUSION Our results may lead researchers to understand the molecular mechanism of miR-34a in the diagnosis, prognosis and therapy of HCC. Therefore, the interaction between miR-34a and its targets may promise better prediction and treatment for HCC. And the experiments in vivo and vitro will be conducted by our group to identify the specific mechanism of miR-34a in the progress and deterioration of HCC.
Collapse
Affiliation(s)
- Fang-Hui Ren
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Hong Yang
- Department of Ultrasonography, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Rong-Quan He
- Center for Genomic and Personalized Medicine, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Jing-Ning Lu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Xing-Gu Lin
- Center for Genomic and Personalized Medicine, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Hai-Wei Liang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Yi-Wu Dang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Zhen-Bo Feng
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China.
| | - Dian-Zhong Luo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China.
| |
Collapse
|
53
|
Pan Y, Mao Y, Jin R, Jiang L. Crosstalk between the Notch signaling pathway and non-coding RNAs in gastrointestinal cancers. Oncol Lett 2018; 15:31-40. [PMID: 29285185 PMCID: PMC5738678 DOI: 10.3892/ol.2017.7294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 09/07/2017] [Indexed: 12/14/2022] Open
Abstract
The Notch signaling pathway is one of the main signaling pathways that mediates direct contact between cells, and is essential for normal development. It regulates various cellular processes, including cell proliferation, apoptosis, migration, invasion, angiogenesis and metastasis. It additionally serves an important function in tumor progression. Non-coding RNAs mainly include small microRNAs, long non-coding RNAs and circular RNAs. At present, a large body of literature supports the biological significance of non-coding RNAs in tumor progression. It is also becoming increasingly evident that cross-talk exists between Notch signaling and non-coding RNAs. The present review summarizes the current knowledge of Notch-mediated gastrointestinal cancer cell processes, and the effect of the crosstalk between the three major types of non-coding RNAs and the Notch signaling pathway on the fate of gastrointestinal cancer cells.
Collapse
Affiliation(s)
- Yangyang Pan
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuyan Mao
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Rong Jin
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Lei Jiang
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
54
|
Xu L, Gu L, Tao X, Xu Y, Qi Y, Yin L, Han X, Peng J. Effect of dioscin on promoting liver regeneration via activating Notch1/Jagged1 signal pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 38:107-117. [PMID: 29425642 DOI: 10.1016/j.phymed.2017.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 10/20/2017] [Accepted: 11/12/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND Development of novel candidates to promote liver regeneration is critical important after partial hepatectomy (PH). Dioscin, a natural product, shows potent effect on liver protection in our previous works. PURPOSE This work aimed to investigate the effect and underlying mechanisms of dioscin on liver regeneration. METHODS The promoting proliferation effects of dioscin on mouse hepatocytem AML12 cells, rat primary hepatocytes, rats and mice after 70% PH were evaluated. RESULTS Dioscin significantly promoted proliferation of rat primary hepatocytes and AML12 cells through MTT, BrdU and PCNA staining assays. Meanwhile, dioscin rapidly recovered the liver to body weight ratios, declined ALT and AST levels, and relieved hepatocytes necrosis compared with 70% PH operation groups in rats and mice. Mechanistic test showed that dioscin significantly increased Notch1 and Jagged1 levels, and accelerated γ-secretase activity by up-regulating PS1 expression, leading to nuclear translocation of Notch1 intracellular domain (NICD1). Subsequently, the significant activation of Notch-dependent target genes (Hey1, Hes1, EGFR, VEGF), and cell-cycle regulatory proteins (CyclinD1, CyclinE1, CDK4 and CDK2) were all recognized. In addition, these results were further confirmed by Notch1 siRNA silencing and inhibition of γ-secretase by DAPT (a well-characterized γ-secretase inhibitor) in vitro. CONCLUSIONS Dioscin, as a novel efficient γ-secretase activator, NICD1 nucleus translocation promoter and cell cycle regulator, markedly activated Notch1/Jagged1 pathway to promote hepato-proliferation. Our findings provide novel insights into dioscin as a natural product with facilitating liver regeneration after PH.
Collapse
Affiliation(s)
- Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Gu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xufeng Tao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| |
Collapse
|
55
|
Morell CM, Fiorotto R, Meroni M, Raizner A, Torsello B, Cadamuro M, Spagnuolo G, Kaffe E, Sutti S, Albano E, Strazzabosco M. Notch signaling and progenitor/ductular reaction in steatohepatitis. PLoS One 2017; 12:e0187384. [PMID: 29140985 PMCID: PMC5687773 DOI: 10.1371/journal.pone.0187384] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 10/18/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Persistent hepatic progenitor cells (HPC) activation resulting in ductular reaction (DR) is responsible for pathologic liver repair in cholangiopathies. Also, HPC/DR expansion correlates with fibrosis in several chronic liver diseases, including steatohepatitis. Increasing evidence indicates Notch signaling as a key regulator of HPC/DR response in biliary and more in general liver injuries. Therefore, we aimed to investigate the role of Notch during HPC/DR activation in a mouse model of steatohepatitis. METHODS Steatohepatitis was generated using methionine-choline deficient (MCD) diet. For hepatocyte lineage tracing, R26R-YFP mice were infected with AAV8-TBG-Cre. RESULTS MCD diet promoted a strong HPC/DR response that progressively diffused in the lobule, and correlated with increased fibrosis and TGF-β1 expression. Notch signaling was unchanged in laser-capture microdissected HPC/DR, whereas Notch receptors were down regulated in hepatocytes. However, in-vivo lineage tracing experiments identified discrete hepatocytes showing Notch-1 activation and expressing (the Notch-dependent) Sox9. Stimulation of AML-12 hepatocyte-cell line with immobilized Jag1 induced Sox9 and down-regulated albumin and BSEP expression. TGF-β1 treatment in primary hepatic stellate cells (HSC) induced Jag1 expression. In MCD diet-fed mice, αSMA-positive HSC were localized around Sox9 expressing hepatocytes, suggesting that Notch activation in hepatocytes was promoted by TGF-β1 stimulated HSC. In-vivo Notch inhibition reduced HPC response and fibrosis progression. CONCLUSION Our data suggest that Notch signaling is an important regulator of DR and that in steatohepatitis, hepatocytes exposed to Jag1-positive HSC, contribute to pathologic DR by undergoing Notch-mediated differentiation towards an HPC-like phenotype. Given the roles of Notch in fibrosis and liver cancer, these data suggest mesenchymal expression of Jag1 as an alternative therapeutic target.
Collapse
Affiliation(s)
- Carola M. Morell
- International Center for Digestive Health, Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Romina Fiorotto
- International Center for Digestive Health, Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Liver Center, Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, Connecticut, United States of America
| | - Marica Meroni
- International Center for Digestive Health, Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Aileen Raizner
- Liver Center, Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, Connecticut, United States of America
| | - Barbara Torsello
- International Center for Digestive Health, Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Massimiliano Cadamuro
- International Center for Digestive Health, Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Gaia Spagnuolo
- International Center for Digestive Health, Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Eleanna Kaffe
- Liver Center, Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, Connecticut, United States of America
| | - Salvatore Sutti
- Department of Health Sciences, University “A. Avogadro” of East Piedmont, Novara, Italy
| | - Emanuele Albano
- Department of Health Sciences, University “A. Avogadro” of East Piedmont, Novara, Italy
| | - Mario Strazzabosco
- International Center for Digestive Health, Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Liver Center, Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
56
|
Notch inhibitors and their role in the treatment of triple negative breast cancer: promises and failures. Curr Opin Oncol 2017; 29:411-427. [DOI: 10.1097/cco.0000000000000406] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
57
|
Kwon H, Song K, Han C, Zhang J, Lu L, Chen W, Wu T. Epigenetic Silencing of miRNA-34a in Human Cholangiocarcinoma via EZH2 and DNA Methylation: Impact on Regulation of Notch Pathway. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2288-2299. [PMID: 28923203 DOI: 10.1016/j.ajpath.2017.06.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 06/19/2017] [Indexed: 12/12/2022]
Abstract
Aberrant expression and regulation of miRNAs have been implicated in multiple stages of tumorigenic processes. The current study was designed to explore the biological function and epigenetic regulation of miR-34a in human cholangiocarcinoma (CCA). Our data show that the expression of miR-34a is decreased significantly in CCA cells compared with non-neoplastic biliary epithelial cells. Forced overexpression of miR-34a in CCA cells inhibited their proliferation and clonogenic capacity in vitro, and suppressed tumor xenograft growth in severe combined immunodeficiency mice. We identified three key components of the Notch pathway, Notch1, Notch2, and Jagged 1, as direct targets of miR-34a. Our further studies show that down-regulation of miR-34a is caused by Enhancer of zeste homolog 2 (EZH2)-mediated H3 lysine 27 trimethylation as well as DNA methylation. Accordingly, treatment with the EZH2 inhibitor, selective S-adenosyl-methionine-competitive small-molecule (GSK126), or the DNA methylation inhibitor, 5-Aza-2'-deoxycytidine, partially restored miR-34a levels in human CCA cells. Immunohistochemical staining and Western blot analyses showed increased EZH2 expression in human CCA tissues and cell lines. We observed that GSK126 significantly reduced CCA cell growth in vitro and intrahepatic metastasis in vivo. Our findings provide novel evidence that miR-34a expression is silenced epigenetically by EZH2 and DNA methylation, which promotes CCA cell growth through activation of the Notch pathway. Consequently, these signaling cascades may represent potential therapeutic targets for effective treatment of human CCA.
Collapse
Affiliation(s)
- Hyunjoo Kwon
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Kyoungsub Song
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Chang Han
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Jinqiang Zhang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Lu Lu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Weina Chen
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
58
|
Siebel C, Lendahl U. Notch Signaling in Development, Tissue Homeostasis, and Disease. Physiol Rev 2017; 97:1235-1294. [PMID: 28794168 DOI: 10.1152/physrev.00005.2017] [Citation(s) in RCA: 685] [Impact Index Per Article: 85.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/19/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Notch signaling is an evolutionarily highly conserved signaling mechanism, but in contrast to signaling pathways such as Wnt, Sonic Hedgehog, and BMP/TGF-β, Notch signaling occurs via cell-cell communication, where transmembrane ligands on one cell activate transmembrane receptors on a juxtaposed cell. Originally discovered through mutations in Drosophila more than 100 yr ago, and with the first Notch gene cloned more than 30 yr ago, we are still gaining new insights into the broad effects of Notch signaling in organisms across the metazoan spectrum and its requirement for normal development of most organs in the body. In this review, we provide an overview of the Notch signaling mechanism at the molecular level and discuss how the pathway, which is architecturally quite simple, is able to engage in the control of cell fates in a broad variety of cell types. We discuss the current understanding of how Notch signaling can become derailed, either by direct mutations or by aberrant regulation, and the expanding spectrum of diseases and cancers that is a consequence of Notch dysregulation. Finally, we explore the emerging field of Notch in the control of tissue homeostasis, with examples from skin, liver, lung, intestine, and the vasculature.
Collapse
Affiliation(s)
- Chris Siebel
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Urban Lendahl
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
59
|
Cervello M, Augello G, Cusimano A, Emma MR, Balasus D, Azzolina A, McCubrey JA, Montalto G. Pivotal roles of glycogen synthase-3 in hepatocellular carcinoma. Adv Biol Regul 2017; 65:59-76. [PMID: 28619606 DOI: 10.1016/j.jbior.2017.06.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/24/2017] [Accepted: 06/04/2017] [Indexed: 06/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers in the world, and represents the second most frequently cancer and third most common cause of death from cancer worldwide. At advanced stage, HCC is a highly aggressive tumor with a poor prognosis and with very limited response to common therapies. Therefore, there is still the need for new effective and well-tolerated therapeutic strategies. Molecular-targeted therapies hold promise for HCC treatment. One promising molecular target is the multifunctional serine/threonine kinase glycogen synthase kinase 3 (GSK-3). The roles of GSK-3β in HCC remain controversial, several studies suggested a possible role of GSK-3β as a tumor suppressor gene in HCC, whereas, other studies indicate that GSK-3β is a potential therapeutic target for this neoplasia. In this review, we will focus on the different roles that GSK-3 plays in HCC and its interaction with signaling pathways implicated in the pathogenesis of HCC, such as Insulin-like Growth Factor (IGF), Notch, Wnt/β-catenin, Hedgehog (HH), and TGF-β pathways. In addition, the pivotal roles of GSK3 in epithelial-mesenchymal transition (EMT), invasion and metastasis will be also discussed.
Collapse
Affiliation(s)
- Melchiorre Cervello
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy", National Research Council (CNR), Palermo, Italy.
| | - Giuseppa Augello
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy", National Research Council (CNR), Palermo, Italy
| | - Antonella Cusimano
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy", National Research Council (CNR), Palermo, Italy
| | - Maria Rita Emma
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy", National Research Council (CNR), Palermo, Italy
| | - Daniele Balasus
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy", National Research Council (CNR), Palermo, Italy
| | - Antonina Azzolina
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy", National Research Council (CNR), Palermo, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Giuseppe Montalto
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy", National Research Council (CNR), Palermo, Italy; Biomedic Department of Internal Medicine and Specialties (DiBiMIS), University of Palermo, Palermo, Italy
| |
Collapse
|
60
|
Wu CX, Xu A, Zhang CC, Olson P, Chen L, Lee TK, Cheung TT, Lo CM, Wang XQ. Notch Inhibitor PF-03084014 Inhibits Hepatocellular Carcinoma Growth and Metastasis via Suppression of Cancer Stemness due to Reduced Activation of Notch1-Stat3. Mol Cancer Ther 2017; 16:1531-1543. [PMID: 28522590 DOI: 10.1158/1535-7163.mct-17-0001] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/29/2017] [Accepted: 05/01/2017] [Indexed: 01/12/2023]
Abstract
Aberrant activation of the Notch signaling pathway is implicated in many solid tumors, including hepatocellular carcinoma, indicating a potential use of Notch inhibitors for treatment. In this study, we investigated the antitumor and antimetastasis efficacy of the novel Notch inhibitor (γ-secretase inhibitor) PF-03084014 in hepatocellular carcinoma. Hepatocellular carcinoma spherical cells (stem-like cancer cells), a sphere-derived orthotopic tumor model and one patient-derived xenograft (PDX) model were used in our experiment. We demonstrated that PF-03084014 inhibited the self-renewal and proliferation of cancer stem cells. PF-03084014 reduced the hepatocellular carcinoma sphere-derived orthotopic tumor and blocked the hepatocellular carcinoma tumor liver to lung metastasis. We further tested the PF-03084014 in PDX models and confirmed the inhibition tumor growth effect. In addition, a low dose of PF-03084014 induced hepatocellular carcinoma sphere differentiation, resulting in chemosensitization. Antitumor activity was associated with PF-03084014-induced suppression of Notch1 activity, decreased Stat3 activation and phosphorylation of the Akt signaling pathway, and reduced epithelial-mesenchymal transition. These are the key contributors to the maintenance of cancer stemness and the promotion of cancer metastasis. Moreover, the Notch-Stat3 association was implicated in the clinical hepatocellular carcinoma prognosis. Collectively, PF-03084014 revealed antitumor and antimetastatic effects in hepatocellular carcinoma, providing evidence for the potential use of gamma-secretase inhibitors as a therapeutic option for the treatment of hepatocellular carcinoma. Mol Cancer Ther; 16(8); 1531-43. ©2017 AACR.
Collapse
Affiliation(s)
- Chuan Xing Wu
- Department of Surgery, The University of Hong Kong, Hong Kong
| | - Aimin Xu
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Cathy C Zhang
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, California, USA
| | - Peter Olson
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla, California, USA
| | - Lin Chen
- Department of Surgery, The University of Hong Kong, Hong Kong
| | - Terence K Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong
| | - Tan To Cheung
- Department of Surgery, The University of Hong Kong, Hong Kong
| | - Chung Mau Lo
- Department of Surgery, The University of Hong Kong, Hong Kong
| | - Xiao Qi Wang
- Department of Surgery, The University of Hong Kong, Hong Kong. .,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong
| |
Collapse
|
61
|
Öztürk Akcora B, Storm G, Prakash J, Bansal R. Tyrosine kinase inhibitor BIBF1120 ameliorates inflammation, angiogenesis and fibrosis in CCl 4-induced liver fibrogenesis mouse model. Sci Rep 2017; 7:44545. [PMID: 28291245 PMCID: PMC5349608 DOI: 10.1038/srep44545] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/10/2017] [Indexed: 12/15/2022] Open
Abstract
Hepatic fibrosis, a progressive chronic disease mainly caused by hepatitis viral infections, alcohol abuse or metabolic syndrome leading to liver dysfunction and is the growing cause of mortality worldwide. Tyrosine kinase inhibitor BIBF1120 (Nintedanib) has been evaluated in clinical trials for idiopathic pulmonary fibrosis and advanced Hepatocellular carcinoma, but has not been explored for liver fibrosis yet. In this study, we aimed to investigate the therapeutic effects and mechanism of BIBF1120 in liver fibrogenesis. The effects of BIBF1120 were evaluated in TGFβ-activated mouse 3T3 fibroblasts, LX2 cells, primary human hepatic stellate cells (HSCs) and CCl4-induced liver fibrogenesis mouse model. Fibroblasts-conditioned medium studies were performed to assess the paracrine effects on macrophages and endothelial cells. In-vitro in TGFβ-activated fibroblasts, BIBF1120 significantly inhibited expression of major fibrotic parameters, wound-healing and contractility. In vivo in CCl4-induced acute liver injury model, post-disease BIBF1120 administration significantly attenuated collagen accumulation and HSC activation. Interestingly, BIBF1120 drastically inhibited intrahepatic inflammation and angiogenesis. To further elucidate the mechanism of action, 3T3-conditioned medium studies demonstrated increased 3T3-mediated macrophage chemotaxis and endothelial cells tube formation and activation, which was significantly decreased by BIBF1120. These results suggests that BIBF1120 can be a potential therapeutic approach for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Büsra Öztürk Akcora
- Targeted Therapeutics, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Gert Storm
- Targeted Therapeutics, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.,Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Jai Prakash
- Targeted Therapeutics, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Targeted Therapeutics, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
62
|
NOR1 promotes hepatocellular carcinoma cell proliferation and migration through modulating the Notch signaling pathway. Exp Cell Res 2017; 352:375-381. [DOI: 10.1016/j.yexcr.2017.02.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/16/2017] [Accepted: 02/19/2017] [Indexed: 12/19/2022]
|
63
|
Potter EA, Dolgova EV, Proskurina AS, Efremov YR, Minkevich AM, Rozanov AS, Peltek SE, Nikolin VP, Popova NA, Seledtsov IA, Molodtsov VV, Zavyalov EL, Taranov OS, Baiborodin SI, Ostanin AA, Chernykh ER, Kolchanov NA, Bogachev SS. Gene expression profiling of tumor-initiating stem cells from mouse Krebs-2 carcinoma using a novel marker of poorly differentiated cells. Oncotarget 2017; 8:9425-9441. [PMID: 28031533 PMCID: PMC5354742 DOI: 10.18632/oncotarget.14116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 12/15/2016] [Indexed: 12/18/2022] Open
Abstract
Using the ability of poorly differentiated cells to natively internalize fragments of extracellular double-stranded DNA as a marker, we isolated a tumorigenic subpopulation present in Krebs-2 ascites that demonstrated the features of tumor-inducing cancer stem cells. Having combined TAMRA-labeled DNA probe and the power of RNA-seq technology, we identified a set of 168 genes specifically expressed in TAMRA-positive cells (tumor-initiating stem cells), these genes remaining silent in TAMRA-negative cancer cells. TAMRA+ cells displayed gene expression signatures characteristic of both stem cells and cancer cells. The observed expression differences between TAMRA+ and TAMRA- cells were validated by Real Time PCR. The results obtained corroborated the biological data that TAMRA+ murine Krebs-2 tumor cells are tumor-initiating stem cells. The approach developed can be applied to profile any poorly differentiated cell types that are capable of immanent internalization of double-stranded DNA.
Collapse
Affiliation(s)
- Ekaterina A. Potter
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Evgenia V. Dolgova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Anastasia S. Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Yaroslav R. Efremov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
- Novosibirsk State University, Novosibirsk 630090, Russia
| | - Alexandra M. Minkevich
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Aleksey S. Rozanov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Sergey E. Peltek
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Valeriy P. Nikolin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Nelly A. Popova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
- Novosibirsk State University, Novosibirsk 630090, Russia
| | | | - Vladimir V. Molodtsov
- Novosibirsk State University, Novosibirsk 630090, Russia
- Softberry Inc., New York 10549, USA
| | - Evgeniy L Zavyalov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Oleg S. Taranov
- The State Research Center of Virology and Biotechnology VECTOR, Koltsovo, Novosibirsk 630559, Russia
| | - Sergey I. Baiborodin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Alexander A. Ostanin
- Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Medical Sciences, Novosibirsk 630099, Russia
| | - Elena R. Chernykh
- Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Medical Sciences, Novosibirsk 630099, Russia
| | - Nikolay A. Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Sergey S. Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| |
Collapse
|
64
|
Ferrarotto R, Mitani Y, Diao L, Guijarro I, Wang J, Zweidler-McKay P, Bell D, William WN, Glisson BS, Wick MJ, Kapoun AM, Patnaik A, Eckhardt G, Munster P, Faoro L, Dupont J, Lee JJ, Futreal A, El-Naggar AK, Heymach JV. Activating NOTCH1 Mutations Define a Distinct Subgroup of Patients With Adenoid Cystic Carcinoma Who Have Poor Prognosis, Propensity to Bone and Liver Metastasis, and Potential Responsiveness to Notch1 Inhibitors. J Clin Oncol 2017. [DOI: 10.1200/jco.2016.67.5264 pmid:278705702017-01-20]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Purpose Adenoid cystic carcinomas (ACCs) represent a heterogeneous group of chemotherapy refractory tumors, with a subset demonstrating an aggressive phenotype. We investigated the molecular underpinnings of this phenotype and assessed the Notch1 pathway as a potential therapeutic target. Methods We genotyped 102 ACCs that had available pathologic and clinical data. Notch1 activation was assessed by immunohistochemistry for Notch1 intracellular domain. Luciferase reporter assays were used to confirm Notch1 target gene expression in vitro. The Notch1 inhibitor brontictuzumab was tested in patient-derived xenografts from patients with ACC and in a patient with ACC who was enrolled in a phase I study. Results NOTCH1 mutations occurred predominantly (14 of 15 patients) in the negative regulatory region and Pro-Glu-Ser-Thr–rich domains, the same two hotspots seen in T-cell acute lymphoblastic leukemias, and led to pathway activation in vitro. NOTCH1-mutant tumors demonstrated significantly higher levels of Notch1 pathway activation than wild-type tumors on the basis of Notch1 intracellular domain staining ( P = .004). NOTCH1 mutations define a distinct aggressive ACC subgroup with a significantly higher likelihood of solid subtype ( P < .001), advanced-stage disease at diagnosis ( P = .02), higher rate of liver and bone metastasis ( P ≤ .02), shorter relapse-free survival (median, 13 v 34 months; P = .01), and shorter overall survival (median 30 v 122 months; P = .001) when compared with NOTCH1 wild-type tumors. Significant tumor growth inhibition with brontictuzumab was observed exclusively in the ACC patient-derived xenograft model that harbored a NOTCH1 activating mutation. Furthermore, an index patient with NOTCH1-mutant ACC had a partial response to brontictuzumab. Conclusion NOTCH1 mutations define a distinct disease phenotype characterized by solid histology, liver and bone metastasis, poor prognosis, and potential responsiveness to Notch1 inhibitors. Clinical studies targeting Notch1 in a genotype-defined ACC subgroup are warranted.
Collapse
Affiliation(s)
- Renata Ferrarotto
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Yoshitsugu Mitani
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Lixia Diao
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Irene Guijarro
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Jing Wang
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Patrick Zweidler-McKay
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Diana Bell
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - William N. William
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Bonnie S. Glisson
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Michael J. Wick
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Ann M. Kapoun
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Amita Patnaik
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Gail Eckhardt
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Pamela Munster
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Leonardo Faoro
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Jakob Dupont
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - J. Jack Lee
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Andrew Futreal
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - Adel K. El-Naggar
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| | - John V. Heymach
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California
| |
Collapse
|
65
|
Kim W, Khan SK, Gvozdenovic-Jeremic J, Kim Y, Dahlman J, Kim H, Park O, Ishitani T, Jho EH, Gao B, Yang Y. Hippo signaling interactions with Wnt/β-catenin and Notch signaling repress liver tumorigenesis. J Clin Invest 2017. [PMID: 27869648 DOI: 10.1172/jci88486.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Malignant tumors develop through multiple steps of initiation and progression, and tumor initiation is of singular importance in tumor prevention, diagnosis, and treatment. However, the molecular mechanism whereby a signaling network of interacting pathways restrains proliferation in normal cells and prevents tumor initiation is still poorly understood. Here, we have reported that the Hippo, Wnt/β-catenin, and Notch pathways form an interacting network to maintain liver size and suppress hepatocellular carcinoma (HCC). Ablation of the mammalian Hippo kinases Mst1 and Mst2 in liver led to rapid HCC formation and activated Yes-associated protein/WW domain containing transcription regulator 1 (YAP/TAZ), STAT3, Wnt/β-catenin, and Notch signaling. Previous work has shown that abnormal activation of these downstream pathways can lead to HCC. Rigorous genetic experiments revealed that Notch signaling forms a positive feedback loop with the Hippo signaling effector YAP/TAZ to promote severe hepatomegaly and rapid HCC initiation and progression. Surprisingly, we found that Wnt/β-catenin signaling activation suppressed HCC formation by inhibiting the positive feedback loop between YAP/TAZ and Notch signaling. Furthermore, we found that STAT3 in hepatocytes is dispensable for HCC formation when mammalian sterile 20-like kinase 1 and 2 (Mst1 and Mst2) were removed. The molecular network we have identified provides insights into HCC molecular classifications and therapeutic developments for the treatment of liver tumors caused by distinct genetic mutations.
Collapse
|
66
|
Fiore E, Malvicini M, Bayo J, Peixoto E, Atorrasagasti C, Sierra R, Rodríguez M, Gómez Bustillo S, García MG, Aquino JB, Mazzolini G. Involvement of hepatic macrophages in the antifibrotic effect of IGF-I-overexpressing mesenchymal stromal cells. Stem Cell Res Ther 2016; 7:172. [PMID: 27876093 PMCID: PMC5120504 DOI: 10.1186/s13287-016-0424-y] [Citation(s) in RCA: 18] [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: 08/22/2016] [Revised: 10/07/2016] [Accepted: 10/18/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Cirrhosis is a major health problem worldwide and new therapies are needed. Hepatic macrophages (hMø) have a pivotal role in liver fibrosis, being able to act in both its promotion and its resolution. It is well-known that mesenchymal stromal cells (MSCs) can modulate the immune/inflammatory cells. However, the effects of MSCs over hMø in the context of liver fibrosis remain unclear. We previously described evidence of the antifibrotic effects of in vivo applying MSCs, which were enhanced by forced overexpression of insulin-like growth factor 1 (AdIGF-I-MSCs). The aim of this work was to analyze the effect of MSCs on hMø behavior in the context of liver fibrosis resolution. METHODS Fibrosis was induced in BALB/c mice by chronic administration of thioacetamide (8 weeks). In vivo gene expression analyses, in vitro experiments using hMø isolated from the nonparenchymal liver cells fraction, and in vivo experiments with depletion of Mø were performed. RESULTS One day after treatment, hMø from fibrotic livers of MSCs-treated animals showed reduced pro-inflammatory and pro-fibrogenic gene expression profiles. These shifts were more pronounced in AdIGF-I-MSCs condition. This group showed a significant upregulation in the expression of arginase-1 and a higher downregulation of iNOS expression thus suggesting decreased levels of oxidative stress. An upregulation in IGF-I and HGF expression was observed in hMø from AdIGF-I-MSCs-treated mice suggesting a restorative phenotype in these cells. Factors secreted by hMø, preconditioned with MSCs supernatant, caused a reduction in the expression levels of hepatic stellate cells pro-fibrogenic and activation markers. Interestingly, hMø depletion abrogated the therapeutic effect achieved with AdIGF-I-MSCs therapy. Expression profile analyses for cell cycle markers were performed on fibrotic livers after treatment with AdIGF-I-MSCs and showed a significant regulation in genes related to DNA synthesis and repair quality control, cell cycle progression, and DNA damage/cellular stress compatible with early induction of pro-regenerative and hepatoprotective mechanisms. Moreover, depletion of hMø abrogated such effects on the expression of the most highly regulated genes. CONCLUSIONS Our results indicate that AdIGF-I-MSCs are able to induce a pro-fibrotic to resolutive phenotype shift on hepatic macrophages, which is a key early event driving liver fibrosis amelioration.
Collapse
Affiliation(s)
- Esteban Fiore
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Mariana Malvicini
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas) Godoy Cruz 2290, Buenos Aires, Argentina
| | - Juan Bayo
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Estanislao Peixoto
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Catalina Atorrasagasti
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas) Godoy Cruz 2290, Buenos Aires, Argentina
| | - Romina Sierra
- Developmental Biology and Regenerative Medicine Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Marcelo Rodríguez
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Sofia Gómez Bustillo
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Mariana G. García
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas) Godoy Cruz 2290, Buenos Aires, Argentina
| | - Jorge B. Aquino
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas) Godoy Cruz 2290, Buenos Aires, Argentina
- Developmental Biology and Regenerative Medicine Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
| | - Guillermo Mazzolini
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Av. Pte. Peron 1500, Derqui-Pilar, Buenos Aires B1629AHJ Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas) Godoy Cruz 2290, Buenos Aires, Argentina
| |
Collapse
|
67
|
Ferrarotto R, Mitani Y, Diao L, Guijarro I, Wang J, Zweidler-McKay P, Bell D, William WN, Glisson BS, Wick MJ, Kapoun AM, Patnaik A, Eckhardt G, Munster P, Faoro L, Dupont J, Lee JJ, Futreal A, El-Naggar AK, Heymach JV. Activating NOTCH1 Mutations Define a Distinct Subgroup of Patients With Adenoid Cystic Carcinoma Who Have Poor Prognosis, Propensity to Bone and Liver Metastasis, and Potential Responsiveness to Notch1 Inhibitors. J Clin Oncol 2016; 35:352-360. [PMID: 27870570 PMCID: PMC5456373 DOI: 10.1200/jco.2016.67.5264] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Purpose Adenoid cystic carcinomas (ACCs) represent a heterogeneous group of chemotherapy refractory tumors, with a subset demonstrating an aggressive phenotype. We investigated the molecular underpinnings of this phenotype and assessed the Notch1 pathway as a potential therapeutic target. Methods We genotyped 102 ACCs that had available pathologic and clinical data. Notch1 activation was assessed by immunohistochemistry for Notch1 intracellular domain. Luciferase reporter assays were used to confirm Notch1 target gene expression in vitro. The Notch1 inhibitor brontictuzumab was tested in patient-derived xenografts from patients with ACC and in a patient with ACC who was enrolled in a phase I study. Results NOTCH1 mutations occurred predominantly (14 of 15 patients) in the negative regulatory region and Pro-Glu-Ser-Thr-rich domains, the same two hotspots seen in T-cell acute lymphoblastic leukemias, and led to pathway activation in vitro. NOTCH1-mutant tumors demonstrated significantly higher levels of Notch1 pathway activation than wild-type tumors on the basis of Notch1 intracellular domain staining ( P = .004). NOTCH1 mutations define a distinct aggressive ACC subgroup with a significantly higher likelihood of solid subtype ( P < .001), advanced-stage disease at diagnosis ( P = .02), higher rate of liver and bone metastasis ( P ≤ .02), shorter relapse-free survival (median, 13 v 34 months; P = .01), and shorter overall survival (median 30 v 122 months; P = .001) when compared with NOTCH1 wild-type tumors. Significant tumor growth inhibition with brontictuzumab was observed exclusively in the ACC patient-derived xenograft model that harbored a NOTCH1 activating mutation. Furthermore, an index patient with NOTCH1-mutant ACC had a partial response to brontictuzumab. Conclusion NOTCH1 mutations define a distinct disease phenotype characterized by solid histology, liver and bone metastasis, poor prognosis, and potential responsiveness to Notch1 inhibitors. Clinical studies targeting Notch1 in a genotype-defined ACC subgroup are warranted.
Collapse
Affiliation(s)
- Renata Ferrarotto
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Yoshitsugu Mitani
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Lixia Diao
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Irene Guijarro
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Jing Wang
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Patrick Zweidler-McKay
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Diana Bell
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - William N William
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Bonnie S Glisson
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Michael J Wick
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Ann M Kapoun
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Amita Patnaik
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Gail Eckhardt
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Pamela Munster
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Leonardo Faoro
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Jakob Dupont
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - J Jack Lee
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Andrew Futreal
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - Adel K El-Naggar
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| | - John V Heymach
- Renata Ferrarotto, Yoshitsugu Mitani, Lixia Diao, Irene Guijarro, Jing Wang, Patrick Zweidler-McKay, Diana Bell, William N. William Jr, Bonnie S. Glisson, J. Jack Lee, Andrew Futreal, Adel K. El-Naggar, and John V. Heymach, University of Texas MD Anderson Cancer Center, Houston; Michael J. Wick and Amita Patnaik, South Texas Accelerated Research Therapeutics, San Antonio, TX; Ann M. Kapoun, Leonardo Faoro, and Jakob Dupont, OncoMed Pharmaceuticals, Redwood City; Pamela Munster, University of California San Francisco, San Francisco, CA; and Gail Eckhardt, University of Colorado Denver School of Medicine, Denver, CO
| |
Collapse
|
68
|
Kim W, Khan SK, Gvozdenovic-Jeremic J, Kim Y, Dahlman J, Kim H, Park O, Ishitani T, Jho EH, Gao B, Yang Y. Hippo signaling interactions with Wnt/β-catenin and Notch signaling repress liver tumorigenesis. J Clin Invest 2016; 127:137-152. [PMID: 27869648 DOI: 10.1172/jci88486] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 10/13/2016] [Indexed: 12/14/2022] Open
Abstract
Malignant tumors develop through multiple steps of initiation and progression, and tumor initiation is of singular importance in tumor prevention, diagnosis, and treatment. However, the molecular mechanism whereby a signaling network of interacting pathways restrains proliferation in normal cells and prevents tumor initiation is still poorly understood. Here, we have reported that the Hippo, Wnt/β-catenin, and Notch pathways form an interacting network to maintain liver size and suppress hepatocellular carcinoma (HCC). Ablation of the mammalian Hippo kinases Mst1 and Mst2 in liver led to rapid HCC formation and activated Yes-associated protein/WW domain containing transcription regulator 1 (YAP/TAZ), STAT3, Wnt/β-catenin, and Notch signaling. Previous work has shown that abnormal activation of these downstream pathways can lead to HCC. Rigorous genetic experiments revealed that Notch signaling forms a positive feedback loop with the Hippo signaling effector YAP/TAZ to promote severe hepatomegaly and rapid HCC initiation and progression. Surprisingly, we found that Wnt/β-catenin signaling activation suppressed HCC formation by inhibiting the positive feedback loop between YAP/TAZ and Notch signaling. Furthermore, we found that STAT3 in hepatocytes is dispensable for HCC formation when mammalian sterile 20-like kinase 1 and 2 (Mst1 and Mst2) were removed. The molecular network we have identified provides insights into HCC molecular classifications and therapeutic developments for the treatment of liver tumors caused by distinct genetic mutations.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Cell Cycle Proteins
- Hepatocyte Growth Factor/genetics
- Hepatocyte Growth Factor/metabolism
- Hippo Signaling Pathway
- Liver Neoplasms, Experimental/genetics
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/pathology
- Mice
- Mice, Knockout
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Serine-Threonine Kinase 3
- Wnt Signaling Pathway
- YAP-Signaling Proteins
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
|
69
|
Zeng J, Jing Y, Shi R, Pan X, Lai F, Liu W, Li R, Gao L, Hou X, Wu M, Wei L. Autophagy regulates biliary differentiation of hepatic progenitor cells through Notch1 signaling pathway. Cell Cycle 2016; 15:1602-10. [PMID: 27259983 DOI: 10.1080/15384101.2016.1181234] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Autophagy plays important roles in self-renewal and differentiation of stem cells. Hepatic progenitor cells (HPCs) are thought to have the ability of self-renewal as well as possess a bipotential capacity, which allows them to differentiate into both hepatocytes and bile ductular cells. However, how autophagy contributes to self-renewal and differentiation of hepatic progenitor cells is not well understood. In this study, we use a well-established rat hepatic progenitor cell lines called WB-F344, which is treated with 3.75 mM sodium butyrate (SB) to promote the differentiation of WB-F344 along the biliary phenotype. We found that autophagy was decreased in the early stage of biliary differentiation, and maintained a low level at the late stage. Activation of autophagy by rapamycin or starvation suppressed the biliary differentiation of WB-F344. Further study reported that autophagy inhibited Notch1 signaling pathway, which contributed to biliary differentiation and morphogenesis. In conclusions, autophagy regulates biliary differentiation of hepatic progenitor cells through Notch1 signaling pathway.
Collapse
Affiliation(s)
- Jianxing Zeng
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China.,c Fujian Medical University , Fuzhou , Fujian , China
| | - Yingying Jing
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Rongyu Shi
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China.,c Fujian Medical University , Fuzhou , Fujian , China
| | - Xiaorong Pan
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China.,c Fujian Medical University , Fuzhou , Fujian , China
| | - Fobao Lai
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China.,c Fujian Medical University , Fuzhou , Fujian , China
| | - Wenting Liu
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Rong Li
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Lu Gao
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Xiaojuan Hou
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Mengchao Wu
- b Department of Comprehensive Treatment , Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| | - Lixin Wei
- a Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University , Shanghai , China
| |
Collapse
|
70
|
Horst AK, Neumann K, Diehl L, Tiegs G. Modulation of liver tolerance by conventional and nonconventional antigen-presenting cells and regulatory immune cells. Cell Mol Immunol 2016; 13:277-92. [PMID: 27041638 PMCID: PMC4856800 DOI: 10.1038/cmi.2015.112] [Citation(s) in RCA: 199] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 12/18/2015] [Accepted: 12/18/2015] [Indexed: 12/11/2022] Open
Abstract
The liver is a tolerogenic organ with exquisite mechanisms of immune regulation that ensure upkeep of local and systemic immune tolerance to self and foreign antigens, but that is also able to mount effective immune responses against pathogens. The immune privilege of liver allografts was recognized first in pigs in spite of major histo-compatibility complex mismatch, and termed the "liver tolerance effect". Furthermore, liver transplants are spontaneously accepted with only low-dose immunosuppression, and induce tolerance for non-hepatic co-transplanted allografts of the same donor. Although this immunotolerogenic environment is favorable in the setting of organ transplantation, it is detrimental in chronic infectious liver diseases like hepatitis B or C, malaria, schistosomiasis or tumorigenesis, leading to pathogen persistence and weak anti-tumor effects. The liver is a primary site of T-cell activation, but it elicits poor or incomplete activation of T cells, leading to their abortive activation, exhaustion, suppression of their effector function and early death. This is exploited by pathogens and can impair pathogen control and clearance or allow tumor growth. Hepatic priming of T cells is mediated by a number of local conventional and nonconventional antigen-presenting cells (APCs), which promote tolerance by immune deviation, induction of T-cell anergy or apoptosis, and generating and expanding regulatory T cells. This review will focus on the communication between classical and nonclassical APCs and lymphocytes in the liver in tolerance induction and will discuss recent insights into the role of innate lymphocytes in this process.
Collapse
Affiliation(s)
- Andrea Kristina Horst
- Institute of Experimental Immunology and Hepatology Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg D-20246, Germany
| | - Katrin Neumann
- Institute of Experimental Immunology and Hepatology Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg D-20246, Germany
| | - Linda Diehl
- Institute of Experimental Immunology and Hepatology Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg D-20246, Germany
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg D-20246, Germany
| |
Collapse
|
71
|
Li T, Guo B, Gao Y, Yu QH, Li JJ, Xian WJ, Jiang S, Zheng QC, Zhang Y. Tumor necrosis factor-alpha up-regulates expression of Jagged-1 and induces epithelial-mesenchymal transition in rat cholangiocytes in vitro. Shijie Huaren Xiaohua Zazhi 2016; 24:1806-1811. [DOI: 10.11569/wcjd.v24.i12.1806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of tumor necrosis factor-alpha (TNF-α) in the epithelial-mesenchymal transition (EMT) process in rat cholangiocytes in vitro.
METHODS: Primary rat cholangiocytes were treated with TNF-α (10 ng/mL) alone, TNF-α plus nuclear factor kappa B (NF-κB) inhibitor PDTC (50 µmol/L), or PDTC alone for 72 h. The expression of Jagged-1, mesenchymal markers [fibroblast-specific protein-1 (FSP-1), Vimentin and α-SMA] as well as epithelial marker CK19 was detected by Western blot. NF-κB binding activity was measured by EMSA. Migration ability and morphological changes of cholangiocytes were also examined.
RESULTS: In the TNF-α alone group, the protein levels of Jagged-1, FSP-1, Vimentin and α-SMA were up-regulated compared to control cells, whereas the expression of CK19 was down-regulated. The migration ability of cholangiocytes was increased and their shape changed from stone-like to fiber-like. For the TNF-α plus PTCD group and the PTCD alone group, no significant changes in EMT markers as well as migration ability were observed compared to control cells.
CONCLUSION: TNF-α is able to increase the expression of Jagged-1 and induce EMT in rat cholangiocytes in vitro possibly through activation of NF-κB signaling.
Collapse
|
72
|
Sun JH, Luo Q, Liu LL, Song GB. Liver cancer stem cell markers: Progression and therapeutic implications. World J Gastroenterol 2016; 22:3547-3557. [PMID: 27053846 PMCID: PMC4814640 DOI: 10.3748/wjg.v22.i13.3547] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/12/2016] [Accepted: 03/02/2016] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) are a small subpopulation in cancer, have been proposed to be cancer-initiating cells, and have been shown to be responsible for chemotherapy resistance and cancer recurrence. The identification of CSC subpopulations inside a tumor presents a new understanding of cancer development because it implies that tumors can only be eradicated by targeting CSCs. Although advances in liver cancer detection and treatment have increased the possibility of curing the disease at early stages, unfortunately, most patients will relapse and succumb to their disease. Strategies aimed at efficiently targeting liver CSCs are becoming important for monitoring the progress of liver cancer therapy and for evaluating new therapeutic approaches. Herein, we provide a critical discussion of biological markers described in the literature regarding liver cancer stem cells and the potential of these markers to serve as therapeutic targets.
Collapse
|
73
|
Xiao S, Chang R, Yang M, Lei X, Liu X, Gao W, Xiao J, Yang L. Actin-like 6A predicts poor prognosis of hepatocellular carcinoma and promotes metastasis and epithelial-mesenchymal transition. Hepatology 2016; 63:1256-71. [PMID: 26698646 PMCID: PMC4834727 DOI: 10.1002/hep.28417] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/20/2015] [Indexed: 12/15/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) is one of the most lethal cancers worldwide because of metastasis. Epithelial-mesenchymal transition (EMT) is widely considered to be crucial to the invasion-metastasis cascade during cancer progression. Actin-like 6A (ACTL6A) is initially verified important for cell proliferation, differentiation, and migration. In this study, we find that ACTL6A plays an essential role in metastasis and EMT of HCC. ACTL6A expression is up-regulated in HCC cells and tissues. A high level of ACTL6A in HCCs is correlated with aggressive clinicopathological features and is an independent poor prognostic factor for overall and disease-free survival of HCC patients. Ectopic expression of ACTL6A markedly promotes HCC cells migration, invasion, as well as EMT in vitro and promotes tumor growth and metastasis in the mouse xenograft model. Opposite results are observed when ACTL6A is knocked down. Mechanistically, ACTL6A promotes metastasis and EMT through activating Notch signaling. ACTL6A knockdown has the equal blockage effect as the Notch signaling inhibitor, N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butylester, in HCC cells. Further studies indicate that ACTL6A might manipulate SRY (sex determining region Y)-box 2 (SOX2) expression and then activate Notch1 signaling. CONCLUSIONS ACTL6A promotes metastasis and EMT by SOX2/Notch1 signaling, indicating a prognostic biomarker candidate and a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Shuai Xiao
- Liver Cancer Laboratory, Department of Surgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Rui‐Min Chang
- Liver Cancer Laboratory, Department of Surgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Ming‐Yang Yang
- Liver Cancer Laboratory, Department of Surgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Xiong Lei
- Liver Cancer Laboratory, Department of Surgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Xiao Liu
- Liver Cancer Laboratory, Department of Surgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Wen‐Bin Gao
- Liver Cancer Laboratory, Department of Surgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Jing‐Lei Xiao
- Liver Cancer Laboratory, Department of Surgery, Xiangya HospitalCentral South UniversityChangshaChina
| | - Lian‐Yue Yang
- Liver Cancer Laboratory, Department of Surgery, Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
74
|
Abstract
MicroRNAs are involved in almost all biological processes and have emerged as regulators of signaling pathways. We show that miRNA target genes and pathway genes are not uniformly expressed across human tissues. To capture tissue specific effects, we developed a novel methodology for tissue specific pathway analysis of miRNAs. We incorporated the most recent and highest quality miRNA targeting data (TargetScan and StarBase), RNA-seq based gene expression data (EBI Expression Atlas) and multiple new pathway data sources to increase the biological relevance of the predicted miRNA-pathway associations. We identified new potential roles of miR-199a-3p, miR-199b-3p and the miR-200 family in hepatocellular carcinoma, involving the regulation of metastasis through MAPK and Wnt signaling. Also, an association of miR-571 and Notch signaling in liver fibrosis was proposed. To facilitate data update and future extensions of our tool, we developed a flexible database backend using the graph database neo4j. The new backend as well as the novel methodology were included in the updated miTALOS v2, a tool that provides insights into tissue specific miRNA regulation of biological pathways. miTALOS v2 is available at http://mips.helmholtz-muenchen.de/mitalos.
Collapse
|
75
|
El-Hachem N, Grossmann P, Blanchet-Cohen A, Bateman AR, Bouchard N, Archambault J, Aerts HJ, Haibe-Kains B. Characterization of Conserved Toxicogenomic Responses in Chemically Exposed Hepatocytes across Species and Platforms. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:313-20. [PMID: 26173225 PMCID: PMC4786983 DOI: 10.1289/ehp.1409157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 07/09/2015] [Indexed: 05/03/2023]
Abstract
BACKGROUND Genome-wide expression profiling is increasingly being used to identify transcriptional changes induced by drugs and environmental stressors. In this context, the Toxicogenomics Project-Genomics Assisted Toxicity Evaluation system (TG-GATEs) project generated transcriptional profiles from rat liver samples and human/rat cultured primary hepatocytes exposed to more than 100 different chemicals. OBJECTIVES To assess the capacity of the cell culture models to recapitulate pathways induced by chemicals in vivo, we leveraged the TG-GATEs data set to compare the early transcriptional responses observed in the liver of rats treated with a large set of chemicals with those of cultured rat and human primary hepatocytes challenged with the same compounds in vitro. METHODS We developed a new pathway-based computational pipeline that efficiently combines gene set enrichment analysis (GSEA) using pathways from the Reactome database with biclustering to identify common modules of pathways that are modulated by several chemicals in vivo and in vitro across species. RESULTS We found that some chemicals induced conserved patterns of early transcriptional responses in in vitro and in vivo settings, and across human and rat genomes. These responses involved pathways of cell survival, inflammation, xenobiotic metabolism, oxidative stress, and apoptosis. Moreover, our results support the transforming growth factor beta receptor (TGF-βR) signaling pathway as a candidate biomarker associated with exposure to environmental toxicants in primary human hepatocytes. CONCLUSIONS Our integrative analysis of toxicogenomics data provides a comprehensive overview of biochemical perturbations affected by a large panel of chemicals. Furthermore, we show that the early toxicological response occurring in animals is recapitulated in human and rat primary hepatocyte cultures at the molecular level, indicating that these models reproduce key pathways in response to chemical stress. These findings expand our understanding and interpretation of toxicogenomics data from human hepatocytes exposed to environmental toxicants.
Collapse
Affiliation(s)
- Nehme El-Hachem
- Integrative systems biology, Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Montréal, Quebec, Canada
| | - Patrick Grossmann
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Alain R. Bateman
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Nicolas Bouchard
- Department of Medicine, University of Montreal, Montréal, Quebec, Canada
- Molecular Biology of Neural Development, Institut de Recherches Cliniques de Montréal, Montreal, Canada
| | - Jacques Archambault
- Laboratory of Molecular Virology, Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
| | - Hugo J.W.L. Aerts
- Department of Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Address correspondence to B. Haibe-Kains, Princess Margaret Cancer Centre, University Health Network, 101 College St., Toronto, ON, M5G 1L7, Canada. Telephone: 1 (416) 581-7628. E-mail: , or to H.J.W.L. Aerts, Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA. E-mail:
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Medical Biophysics Department, University of Toronto, Toronto, Ontario, Canada
- Address correspondence to B. Haibe-Kains, Princess Margaret Cancer Centre, University Health Network, 101 College St., Toronto, ON, M5G 1L7, Canada. Telephone: 1 (416) 581-7628. E-mail: , or to H.J.W.L. Aerts, Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA. E-mail:
| |
Collapse
|
76
|
Weiskirchen R. Hepatoprotective and Anti-fibrotic Agents: It's Time to Take the Next Step. Front Pharmacol 2016; 6:303. [PMID: 26779021 PMCID: PMC4703795 DOI: 10.3389/fphar.2015.00303] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 12/11/2015] [Indexed: 12/21/2022] Open
Abstract
Hepatic fibrosis and cirrhosis cause strong human suffering and necessitate a monetary burden worldwide. Therefore, there is an urgent need for the development of therapies. Pre-clinical animal models are indispensable in the drug discovery and development of new anti-fibrotic compounds and are immensely valuable for understanding and proofing the mode of their proposed action. In fibrosis research, inbreed mice and rats are by far the most used species for testing drug efficacy. During the last decades, several hundred or even a thousand different drugs that reproducibly evolve beneficial effects on liver health in respective disease models were identified. However, there are only a few compounds (e.g., GR-MD-02, GM-CT-01) that were translated from bench to bedside. In contrast, the large number of drugs successfully tested in animal studies is repeatedly tested over and over engender findings with similar or identical outcome. This circumstance undermines the 3R (Replacement, Refinement, Reduction) principle of Russell and Burch that was introduced to minimize the suffering of laboratory animals. This ethical framework, however, represents the basis of the new animal welfare regulations in the member states of the European Union. Consequently, the legal authorities in the different countries are halted to foreclose testing of drugs in animals that were successfully tested before. This review provides a synopsis on anti-fibrotic compounds that were tested in classical rodent models. Their mode of action, potential sources and the observed beneficial effects on liver health are discussed. This review attempts to provide a reference compilation for all those involved in the testing of drugs or in the design of new clinical trials targeting hepatic fibrosis.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy, and Clinical Chemistry, RWTH University Hospital Aachen Aachen, Germany
| |
Collapse
|
77
|
Gil-García B, Baladrón V. The complex role of NOTCH receptors and their ligands in the development of hepatoblastoma, cholangiocarcinoma and hepatocellular carcinoma. Biol Cell 2015; 108:29-40. [DOI: 10.1111/boc.201500029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 11/24/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Borja Gil-García
- Laboratory of Biochemistry and Molecular Biology; Department of Inorganic and Organic Chemistry and Biochemistry; Medical School/CRIB/Biomedicine Unit; University of Castilla-La Mancha (UCLM)/CSIC; 02008, Albacete Spain
| | - Victoriano Baladrón
- Laboratory of Biochemistry and Molecular Biology; Department of Inorganic and Organic Chemistry and Biochemistry; Medical School/CRIB/Biomedicine Unit; University of Castilla-La Mancha (UCLM)/CSIC; 02008, Albacete Spain
| |
Collapse
|
78
|
Hepatic metastatic niche: from normal to pre-metastatic and metastatic niche. Tumour Biol 2015; 37:1493-503. [DOI: 10.1007/s13277-015-4557-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/01/2015] [Indexed: 12/15/2022] Open
|
79
|
Sun G, Mackey LV, Coy DH, Yu CY, Sun L. The Histone Deacetylase Inhibitor Vaproic Acid Induces Cell Growth Arrest in Hepatocellular Carcinoma Cells via Suppressing Notch Signaling. J Cancer 2015; 6:996-1004. [PMID: 26366213 PMCID: PMC4565849 DOI: 10.7150/jca.12135] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 07/19/2015] [Indexed: 12/29/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a type of malignant cancer. Notch signaling is aberrantly expressed in HCC tissues with more evidence showing that this signaling plays a critical role in HCCs. In the present study, we investigate the effects of the anti-convulsant drug valproic acid (VPA) in HCC cells and its involvement in modulating Notch signaling. We found that VPA, acting as a histone deacetylase (HDAC) inhibitor, induced a decrease in HDAC4 and an increase in acetylated histone 4 (AcH4) and suppressed HCC cell growth. VPA also induced down-regulation of Notch signaling via suppressing the expression of Notch1 and its target gene HES1, with an increase of tumor suppressor p21 and p63. Furthermore, Notch1 activation via overexpressing Notch1 active form ICN1 induced HCC cell proliferation and anti-apoptosis, indicating Notch signaling played an oncogenic role in HCC cells. Meanwhile, VPA could reverse Notch1-induced increase of cell proliferation. Interestingly, VPA was also observed to stimulate the expression of G protein-coupled somatostatin receptor type 2 (SSTR2) that has been used in receptor-targeting therapies. This discovery supports a combination therapy of VPA with the SSTR2-targeting agents. Our in vitro assay did show that the combination of VPA and the peptide-drug conjugate camptothecin-somatostatin (CPT-SST) displayed more potent anti-proliferative effects on HCC cells than did each alone. VPA may be a potential drug candidate in the development of anti-HCC drugs via targeting Notch signaling, especially in combination with receptor-targeting cytotoxic agents.
Collapse
Affiliation(s)
- Guangchun Sun
- 1. Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University; 801 He-Qing Rd., Shanghai 200240, China
| | - Lily V Mackey
- 3. Department of Medicine, School of Medicine, Tulane Health Sciences Center, New Orleans, LA 70112-2699, USA
| | - David H Coy
- 3. Department of Medicine, School of Medicine, Tulane Health Sciences Center, New Orleans, LA 70112-2699, USA
| | - Cui-Yun Yu
- 2. Institute of Pharmacy & Pharmacology, Department of Pharmacy, University of South China, Hengyang 421001, China ; 3. Department of Medicine, School of Medicine, Tulane Health Sciences Center, New Orleans, LA 70112-2699, USA
| | - Lichun Sun
- 1. Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University; 801 He-Qing Rd., Shanghai 200240, China ; 2. Institute of Pharmacy & Pharmacology, Department of Pharmacy, University of South China, Hengyang 421001, China ; 3. Department of Medicine, School of Medicine, Tulane Health Sciences Center, New Orleans, LA 70112-2699, USA
| |
Collapse
|
80
|
Karsdal MA, Manon-Jensen T, Genovese F, Kristensen JH, Nielsen MJ, Sand JMB, Hansen NUB, Bay-Jensen AC, Bager CL, Krag A, Blanchard A, Krarup H, Leeming DJ, Schuppan D. Novel insights into the function and dynamics of extracellular matrix in liver fibrosis. Am J Physiol Gastrointest Liver Physiol 2015; 308:G807-30. [PMID: 25767261 PMCID: PMC4437019 DOI: 10.1152/ajpgi.00447.2014] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/04/2015] [Indexed: 02/06/2023]
Abstract
Emerging evidence suggests that altered components and posttranslational modifications of proteins in the extracellular matrix (ECM) may both initiate and drive disease progression. The ECM is a complex grid consisting of multiple proteins, most of which play a vital role in containing the essential information needed for maintenance of a sophisticated structure anchoring the cells and sustaining normal function of tissues. Therefore, the matrix itself may be considered as a paracrine/endocrine entity, with more complex functions than previously appreciated. The aims of this review are to 1) explore key structural and functional components of the ECM as exemplified by monogenetic disorders leading to severe pathologies, 2) discuss selected pathological posttranslational modifications of ECM proteins resulting in altered functional (signaling) properties from the original structural proteins, and 3) discuss how these findings support the novel concept that an increasing number of components of the ECM harbor signaling functions that can modulate fibrotic liver disease. The ECM entails functions in addition to anchoring cells and modulating their migratory behavior. Key ECM components and their posttranslational modifications often harbor multiple domains with different signaling potential, in particular when modified during inflammation or wound healing. This signaling by the ECM should be considered a paracrine/endocrine function, as it affects cell phenotype, function, fate, and finally tissue homeostasis. These properties should be exploited to establish novel biochemical markers and antifibrotic treatment strategies for liver fibrosis as well as other fibrotic diseases.
Collapse
Affiliation(s)
- Morten A. Karsdal
- 1Nordic Bioscience A/S, Herlev Hovedgade, Herlev, Denmark; ,2University of Southern Denmark, SDU, Odense, Denmark;
| | | | | | | | | | | | | | | | | | - Aleksander Krag
- 3Department of Gastroenterology and Hepatology, Odense University Hospital, University of Southern Denmark, Odense, Denmark;
| | - Andy Blanchard
- 4GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, United Kingdom;
| | - Henrik Krarup
- 5Section of Molecular Biology, Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark;
| | | | - Detlef Schuppan
- 6Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany; ,7Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
81
|
Bi P, Kuang S. Notch signaling as a novel regulator of metabolism. Trends Endocrinol Metab 2015; 26:248-55. [PMID: 25805408 PMCID: PMC4435535 DOI: 10.1016/j.tem.2015.02.006] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 12/11/2022]
Abstract
Evolutionarily unprepared for modern high-calorie diets and sedentary lifestyles, humans are now unprecedentedly susceptible to metabolic disorders such as obesity, type 2 diabetes (T2D), nonalcoholic fatty liver, and cardiovascular disease. These metabolic conditions are intertwined, together known as metabolic syndrome, and compromise human life quality as well as lives. Notch signaling, a fundamental signal transduction pathway critical for cell-cell communication and development, has recently been recognized as a key player in metabolism. This review summarizes the emerging roles of Notch signaling in regulating the metabolism of various cell and tissue types, with emphasis on the underlying molecular mechanisms and the potential of targeting this signal axis to treat metabolic diseases.
Collapse
Affiliation(s)
- Pengpeng Bi
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
82
|
Raggi C, Invernizzi P, Andersen JB. Impact of microenvironment and stem-like plasticity in cholangiocarcinoma: molecular networks and biological concepts. J Hepatol 2015; 62:198-207. [PMID: 25220250 DOI: 10.1016/j.jhep.2014.09.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 08/30/2014] [Accepted: 09/03/2014] [Indexed: 12/13/2022]
Abstract
Clinical complexity, anatomic diversity and molecular heterogeneity of cholangiocarcinoma (CCA) represent a major challenge in the assessment of effective targeted therapies. Molecular and cellular mechanisms underlying the diversity of CCA growth patterns remain a key issue of clinical concern. Crucial questions comprise the nature of the CCA-origin, the initial target for cellular transformation as well as the relationship with the cancer stem cells (CSC) concept. Additionally, since CCA often develops in the context of an inflammatory milieu (cirrhosis and cholangitis), the stromal compartment or tumour microenvironment (TME) likely promotes initiation and progression of this malignancy, contributing to its heterogeneity. This review will emphasize the dynamic interplay between stem-like intrinsic and TME-extrinsic pathways, which may represent novel options for multi-targeted therapies in CCA.
Collapse
Affiliation(s)
- Chiara Raggi
- Liver Unit and Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy.
| | - Pietro Invernizzi
- Liver Unit and Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Jesper B Andersen
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
83
|
Geisler F, Strazzabosco M. Emerging roles of Notch signaling in liver disease. Hepatology 2015; 61:382-92. [PMID: 24930574 PMCID: PMC4268103 DOI: 10.1002/hep.27268] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/11/2014] [Accepted: 06/12/2014] [Indexed: 12/13/2022]
Abstract
This review critically discusses the most recent advances in the role of Notch signaling in liver development, homeostasis, and disease. It is now clear that the significance of Notch in determining mammalian cell fates and functions extends beyond development, and Notch is a major regular of organ homeostasis. Moreover, Notch signaling is reactivated upon injury and regulates the complex interactions between the distinct liver cell types involved in the repair process. Notch is also involved in the regulation of liver metabolism, inflammation, and cancer. The net effects of Notch signaling are highly variable and finely regulated at multiple levels, but also depend on the specific cellular context in which Notch is activated. Persistent activation of Notch signaling is associated with liver malignancies, such as hepatocellular carcinoma with stem cell features and intrahepatic cholangiocarcinoma. The complexity of the pathway provides several possible targets for agents able to inhibit Notch. However, further cell- and context-specific in-depth understanding of Notch signaling in liver homeostasis and disease will be essential to translate these concepts into clinical practice and be able to predict benefits and risks of evolving therapies.
Collapse
Affiliation(s)
- Fabian Geisler
- 2nd Department of Internal Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Mario Strazzabosco
- Liver Center & Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA,Department of Surgery and Interdisciplinary Medicine, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
84
|
Abstract
Cholangiocarcinoma (CCA) is an aggressive biliary tract malignancy with limited treatment options and low survival rates. Currently, there are no curative medical therapies for CCA. Recent advances have enhanced our understanding of the genetic basis of this disease, and elucidated therapeutically relevant targets. Therapeutic efforts in development are directed at several key pathways due to genetic aberrations including receptor tyrosine kinase pathways, mutant IDH enzymes, the PI3K-AKT-mTOR pathway, and chromatin remodeling networks. A highly desmoplastic, hypovascular stroma is characteristic of CCAs and recent work has highlighted the importance of targeting this pathway via stromal myofibroblast depletion. Future efforts should concentrate on combination therapies with action against the cancer cell and the surrounding tumor stroma. As the mutational landscape of CCA is being illuminated, molecular profiling of patient tumors will enable identification of specific mutations and the opportunity to offer directed, personalized treatment options.
Collapse
Affiliation(s)
- Sumera Rizvi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Mitesh J. Borad
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, Arizona
| | - Tushar Patel
- Departments of Internal Medicine, Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
85
|
Maemura K, Natsugoe S, Takao S. Molecular mechanism of cholangiocarcinoma carcinogenesis. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2014; 21:754-760. [DOI: 10.1002/jhbp.126] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Kosei Maemura
- Department of Digestive Surgery, Breast and Thyroid Surgery; Kagoshima University Graduate School of Medical and Dental Sciences; Kagoshima Japan
| | - Shoji Natsugoe
- Department of Digestive Surgery, Breast and Thyroid Surgery; Kagoshima University Graduate School of Medical and Dental Sciences; Kagoshima Japan
| | - Sonshin Takao
- Center for Biomedical Science and Swine Research; Kagoshima University; 8-35-1 Sakuragaoka Kagoshima 890-8520 Japan
| |
Collapse
|