101
|
A hepatocyte differentiation model reveals two subtypes of liver cancer with different oncofetal properties and therapeutic targets. Proc Natl Acad Sci U S A 2020; 117:6103-6113. [PMID: 32123069 DOI: 10.1073/pnas.1912146117] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Clinical observation of the association between cancer aggressiveness and embryonic development stage implies the importance of developmental signals in cancer initiation and therapeutic resistance. However, the dynamic gene expression during organogenesis and the master oncofetal drivers are still unclear, which impeded the efficient elimination of poor prognostic tumors, including human hepatocellular carcinoma (HCC). In this study, human embryonic stem cells were induced to differentiate into adult hepatocytes along hepatic lineages to mimic liver development in vitro. Combining transcriptomic data from liver cancer patients with the hepatocyte differentiation model, the active genes derived from different hepatic developmental stages and the tumor tissues were selected. Bioinformatic analysis followed by experimental assays was used to validate the tumor subtype-specific oncofetal signatures and potential therapeutic values. Hierarchical clustering analysis revealed the existence of two subtypes of liver cancer with different oncofetal properties. The gene signatures and their clinical significance were further validated in an independent clinical cohort and The Cancer Genome Atlas database. Upstream activator analysis and functional screening further identified E2F1 and SMAD3 as master transcriptional regulators. Small-molecule inhibitors specifically targeting the oncofetal drivers extensively down-regulated subtype-specific developmental signaling and inhibited tumorigenicity. Liver cancer cells and primary HCC tumors with different oncofetal properties also showed selective vulnerability to their specific inhibitors. Further precise targeting of the tumor initiating steps and driving events according to subtype-specific biomarkers might eliminate tumor progression and provide novel therapeutic strategy.
Collapse
|
102
|
Abstract
Hepatocellular carcinoma (HCC), the most common form of primary liver cancer, typically develops on the background of chronic liver disease and is an aggressive disease with dismal prognosis. Studies using next-generation sequencing of multiple regions of the same tumour nodule suggest different patterns of HCC evolution and confirm the high molecular heterogeneity in a subset of patients. Different hypotheses have been proposed to explain tumour evolution, including clonal selection or neutral and punctuated acquisition of genetic alterations. In parallel, data indicate a fundamental contribution of nonmalignant cells of the tumour microenvironment to cancer clonal evolution. Delineating these dynamics is crucial to improve the treatment of patients with HCC, and particularly to help understand how HCC evolution drives resistance to systemic therapies. A number of new minimally invasive techniques, such as liquid biopsies, could help track cancer evolution in HCC. These tools might improve our understanding of how systemic therapies affect tumour clonal composition and could facilitate implementation of real-time molecular monitoring of patients with HCC.
Collapse
|
103
|
Roos E, Strijker M, Franken LC, Busch OR, van Hooft JE, Klümpen HJ, van Laarhoven HW, Wilmink JW, Verheij J, van Gulik TM, Besselink MG. Comparison of short- and long-term outcomes between anatomical subtypes of resected biliary tract cancer in a Western high-volume center. HPB (Oxford) 2020; 22:405-414. [PMID: 31494056 DOI: 10.1016/j.hpb.2019.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/25/2019] [Accepted: 07/19/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Outcomes for the four anatomical subtypes of biliary tract carcinoma (BTC) - intrahepatic, perihilar and distal cholangiocarcinoma (ICC, PHCC, DCC) and gallbladder carcinoma (GBC) - are often combined. However, large cohorts comparing short- and long-term outcomes for the anatomical subtypes of BTC are lacking. METHODS All patients who underwent resection for pathology proven ICC, PHCC, DCC or GBC (2000-2016) from a single Western high-volume center were retrospectively selected. Clinicopathological characteristics, short- and long-term outcomes were compared between the four anatomical subtypes. RESULTS Overall, 361 patients with resected BTC were included (33 ICC, 135 PHCC, 148 DCC, 45 GBC). Clavien-Dindo grade III or higher complications were 48%, 51%, 36% and 8% (p < 0.001) and 90-day mortality was 9%, 15%, 3%, 4% (p < 0.001), for ICC, PHCC, DCC, GBC. Median overall survival was 37, 42, 29 and 41 months (p = 0.722), for ICC, PHCC, DCC, GBC. Five-year survival ranged between 29% and 37%. Anatomical subtype was not an independent predictor for overall survival. CONCLUSION In this large single-center cohort of resected BTC, major morbidity and 90-day mortality varied between the four anatomical subtypes of BTC, mainly due to differences in surgical approach However, a significant difference in overall survival was not detected.
Collapse
Affiliation(s)
- Eva Roos
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands.
| | - Marin Strijker
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Lotte C Franken
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Olivier R Busch
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Jeanin E van Hooft
- Department of Gastroenterology & Hepatology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Heinz-Josef Klümpen
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Hanneke W van Laarhoven
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Johanna W Wilmink
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Joanne Verheij
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Thomas M van Gulik
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Marc G Besselink
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, the Netherlands.
| |
Collapse
|
104
|
Wang H, Wang H, Li K, Li S, Sun B. IGFBP-3 Is the Key Target of Sanguinarine in Promoting Apoptosis in Hepatocellular Carcinoma. Cancer Manag Res 2020; 12:1007-1015. [PMID: 32104082 PMCID: PMC7023858 DOI: 10.2147/cmar.s234291] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/11/2020] [Indexed: 12/14/2022] Open
Abstract
Introduction Chemotherapeutic treatment of hepatocellular carcinoma (HCC) has always been plagued by nonspecific and side effects. Plant extracts have potential anticancer capabilities with low cytotoxicity and few side effects, but their detailed mechanisms are still unclear, thus limiting their clinical applications. Methods In this study, five plant extracts were chosen, their inhibition on HCC cell viability was compared by CCK-8 assay and sanguinarine (SAN) was selected. Then, wound healing assay, transwell assay, and apoptosis assay were carried out in Hep3B cells. Bioinformatics methods were performed and IGFBP-3 was predicted the targets of SAN in HCC. The mechanism of SAN regulating IGFBP-3 was explored using qRT-PCR, Western blotting, cell viability assay and apoptosis assay. Meanwhile, knockdown of IGFBP-3 were used by small interfering RNA (siRNA). Results In five plant extracts, SAN inhibited the proliferation of HCC cell lines most considerably. In addition, apoptosis was promoted, and invasion and migration were inhibited in the Hep3B cell line by treatment with SAN at 2 μM. Bioinformatics indicated that SAN could affect HCC apoptosis through the TP53/IGFBP-3 pathway, and further verification experiments showed that SAN upregulated the expression of insulin-like growth factor binding protein-3 (IGFBP-3) in the Hep3B cell line; SAN also inhibited the expression of Bcl-2 and promoted the expression of BAX and caspase-3. After using siRNA to inhibit the expression of IGFBP-3, the effect of SAN was blocked. Conclusion Our study further reveals a novel mechanism that IGFBP-3 is an important target of SAN, by upregulating expression of IGFBP-3, SAN promotes apoptosis in HCC.
Collapse
Affiliation(s)
- Huiwen Wang
- Department of Interventional, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, People's Republic of China
| | - He Wang
- Department of Interventional, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, People's Republic of China
| | - Kai Li
- Department of Interventional, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, People's Republic of China
| | - Shijie Li
- Department of Interventional, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, People's Republic of China
| | - Bingyi Sun
- Department of General Surgery, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang Province, People's Republic of China
| |
Collapse
|
105
|
Crosstalk between NLRP12 and JNK during Hepatocellular Carcinoma. Int J Mol Sci 2020; 21:ijms21020496. [PMID: 31941025 PMCID: PMC7013925 DOI: 10.3390/ijms21020496] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 12/31/2019] [Accepted: 01/08/2020] [Indexed: 12/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC), a leading cause of cancer-related death, is initiated and promoted by chronic inflammation. Inflammatory mediators are transcriptionally regulated by several inflammatory signaling pathways, including nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK). cJun N-terminal kinase (JNK), a member of the MAPK family, plays a central role in HCC pathogenesis. Pathogen-associated molecular patterns (PAMPs) activate JNK and other MAPK upon recognition by toll-like receptors (TLRs). Apart from TLRs, PAMPs are sensed by several other pattern recognition receptors, including cytosolic NOD-like receptors (NLRs). In a recent study, we demonstrated that the NLR member NLRP12 plays a critical role in suppressing HCC via negative regulation of the JNK pathway. This article briefly reviews the crosstalk between NLRP12 and JNK that occurs during HCC.
Collapse
|
106
|
Kwan SY, Sheel A, Song CQ, Zhang XO, Jiang T, Dang H, Cao Y, Ozata DM, Mou H, Yin H, Weng Z, Wang XW, Xue W. Depletion of TRRAP Induces p53-Independent Senescence in Liver Cancer by Down-Regulating Mitotic Genes. Hepatology 2020; 71:275-290. [PMID: 31188495 PMCID: PMC6906267 DOI: 10.1002/hep.30807] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 05/27/2019] [Indexed: 01/10/2023]
Abstract
Hepatocellular carcinoma (HCC) is an aggressive subtype of liver cancer with few effective treatments, and the underlying mechanisms that drive HCC pathogenesis remain poorly characterized. Identifying genes and pathways essential for HCC cell growth will aid the development of new targeted therapies for HCC. Using a kinome CRISPR screen in three human HCC cell lines, we identified transformation/transcription domain-associated protein (TRRAP) as an essential gene for HCC cell proliferation. TRRAP has been implicated in oncogenic transformation, but how it functions in cancer cell proliferation is not established. Here, we show that depletion of TRRAP or its co-factor, histone acetyltransferase KAT5, inhibits HCC cell growth through induction of p53-independent and p21-independent senescence. Integrated cancer genomics analyses using patient data and RNA sequencing identified mitotic genes as key TRRAP/KAT5 targets in HCC, and subsequent cell cycle analyses revealed that TRRAP-depleted and KAT5-depleted cells are arrested at the G2/M phase. Depletion of topoisomerase II alpha (TOP2A), a mitotic gene and TRRAP/KAT5 target, was sufficient to recapitulate the senescent phenotype of TRRAP/KAT5 knockdown. Conclusion: Our results uncover a role for TRRAP/KAT5 in promoting HCC cell proliferation by activating mitotic genes. Targeting the TRRAP/KAT5 complex is a potential therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Suet-Yan Kwan
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| | - Ankur Sheel
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| | - Chun-Qing Song
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| | - Xiao-Ou Zhang
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Tingting Jiang
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| | - Hien Dang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Yueying Cao
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| | - Deniz M. Ozata
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| | - Haiwei Mou
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
| | - Hao Yin
- Medical research institute, Wuhan University, Wuhan, PR China
| | - Zhiping Weng
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605
- Department of Bioinformatics, School of Life Science and Technology, Tongji University, Shanghai, P. R. China
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Wen Xue
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605
- Program in Molecular Medicine, Department of Molecular, Cell and Cancer Biology, and Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605
| |
Collapse
|
107
|
Marquardt JU, Edlich F. Predisposition to Apoptosis in Hepatocellular Carcinoma: From Mechanistic Insights to Therapeutic Strategies. Front Oncol 2019; 9:1421. [PMID: 31921676 PMCID: PMC6923252 DOI: 10.3389/fonc.2019.01421] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/29/2019] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the most rapidly evolving cancers in the Western world. The majority of HCCs develop on the basis of a chronic inflammatory liver damage that predisposes liver cancer development and leads to deregulation of multiple cellular signaling pathways. The resulting dysbalance between uncontrolled proliferation and impaired predisposition to cell death with consecutive failure to clear inflammatory damage is a key driver of malignant transformation. Therefore, resistance to death signaling accompanied by metabolic changes as well as failed immunological clearance of damaged pre-neoplastic hepatocytes are considered hallmarks of hepatocarcinogenesis. Hereby, the underlying liver disease, the type of liver damage and individual predisposition to apoptosis determines the natural course of the disease as well as the therapeutic response. Here, we will review common and individual aspects of cell death pathways in hepatocarcinogenesis with a particular emphasis on regulatory networks and key molecular alterations. We will further delineate the potential of targeting cell death-related signaling as a viable therapeutic strategy to improve the outcome of HCC patients.
Collapse
Affiliation(s)
- Jens U Marquardt
- Department of Medicine I, University Medical Center Schleswig-Holstein, Lübeck, Germany.,Department of Medicine, Lichtenberg Research Group, University Mainz, Mainz, Germany
| | - Frank Edlich
- Heisenberg Research Group "Regulation von Bcl-2-Proteinen Durch Konformationelle Flexibilität," Institute for Biochemistry and Molecular Biology, University of Freiburg, Freiburg, Germany.,CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
108
|
Nuciforo S, Fofana I, Matter MS, Blumer T, Calabrese D, Boldanova T, Piscuoglio S, Wieland S, Ringnalda F, Schwank G, Terracciano LM, Ng CKY, Heim MH. Organoid Models of Human Liver Cancers Derived from Tumor Needle Biopsies. Cell Rep 2019; 24:1363-1376. [PMID: 30067989 PMCID: PMC6088153 DOI: 10.1016/j.celrep.2018.07.001] [Citation(s) in RCA: 277] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/24/2018] [Accepted: 06/29/2018] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and the second most frequent cause of cancer-related mortality worldwide. The multikinase inhibitor sorafenib is the only treatment option for advanced HCC. Due to tumor heterogeneity, its efficacy greatly varies between patients and is limited due to adverse effects and drug resistance. Current in vitro models fail to recapitulate key features of HCCs. We report the generation of long-term organoid cultures from tumor needle biopsies of HCC patients with various etiologies and tumor stages. HCC organoids retain the morphology as well as the expression pattern of HCC tumor markers and preserve the genetic heterogeneity of the originating tumors. In a proof-of-principle study, we show that liver cancer organoids can be used to test sensitivity to sorafenib. In conclusion, organoid models can be derived from needle biopsies of liver cancers and provide a tool for developing tailored therapies. Organoids can be derived from tumor needle biopsies of liver cancers Organoids retain the morphology and tumor marker expression of the original tumors Tumor organoids preserve the genetic heterogeneity of the originating tumors Tumor organoids provide a tool for developing tailored therapies
Collapse
Affiliation(s)
- Sandro Nuciforo
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Isabel Fofana
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Matthias S Matter
- Institute of Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Tanja Blumer
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Diego Calabrese
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Tujana Boldanova
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; Clinic of Gastroenterology and Hepatology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Salvatore Piscuoglio
- Institute of Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Stefan Wieland
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Femke Ringnalda
- Institute of Molecular Health Sciences, ETH Zurich, Otto-Stern-Weg 7, HPL, 8093 Zürich, Switzerland
| | - Gerald Schwank
- Institute of Molecular Health Sciences, ETH Zurich, Otto-Stern-Weg 7, HPL, 8093 Zürich, Switzerland
| | - Luigi M Terracciano
- Institute of Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Charlotte K Y Ng
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; Institute of Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Markus H Heim
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; Clinic of Gastroenterology and Hepatology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland.
| |
Collapse
|
109
|
Wang D, Zhang Q, Li F, Wang C, Yang C, Yu H. β-TrCP-mediated ubiquitination and degradation of Dlg5 regulates hepatocellular carcinoma cell proliferation. Cancer Cell Int 2019; 19:298. [PMID: 31787846 PMCID: PMC6858669 DOI: 10.1186/s12935-019-1029-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
Background Discs large homolog 5 (Dlg5) is a member of the membrane-associated guanylate kinase (MAGUK) adaptor family of proteins and its deregulation has been implicated in the malignancy of several cancer types. Dlg5 was down-regulated in hepatocellular carcinoma (HCC) and lower Dlg5 expression was associated with poor survival of HCC patients. However, how to regulate Dlg5 remains largely unknown. Methods The co-immunoprecipitation assay was used to determine the interaction between Dlg5 and β-TrCP. The in vivo ubiquitination assay was performed to determine the regulation of Dlg5 by β-TrCP. CCK-8 and colony formation assay were implemented to detect the biological effect of Dlg5 on the growth of HCC cells in vitro. The effect of Dlg5 on HCC tumor growth in vivo was studied in a tumor xenograft model in mice. Results Here we report that Dlg5 is regulated by the ubiquitin proteasome system and depletion of either Cullin 1 or β-TrCP led to increased levels of Dlg5. β-TrCP regulated Dlg5 protein stability by targeting it for ubiquitination and subsequent destruction in a phosphorylation-dependent manner. We further demonstrated a crucial role of Ser730 in the non-canonical phosphodegron of Dlg5 in governing β-TrCP-mediated Dlg5 degradation. Importantly, failure to degrade Dlg5 significantly inhibited HCC cells proliferation both in vitro and in vivo. Conclusion Collectively, our finding provides a novel molecular mechanism for the negative regulation of Dlg5 by β-TRCP in HCC cells. It further suggests that preventing Dlg5 degradation could be a possible novel strategy for clinical treatment of HCC.
Collapse
Affiliation(s)
- Dongping Wang
- Department of Anesthesiology, The First People's of Hospital of Jingmen City, Jingmen, Hubei 448000 China
| | - Qi Zhang
- Department of Operation Room, The First People's of Hospital of Jingmen City, Jingmen, Hubei 448000 China
| | - Fenfen Li
- Department of Nursing, The First People's of Hospital of Jingmen City, Jingmen, Hubei 448000 China
| | - Chan Wang
- Department of Anesthesiology, The First People's of Hospital of Jingmen City, Jingmen, Hubei 448000 China
| | - Changming Yang
- Department of Anesthesiology, The First People's of Hospital of Jingmen City, Jingmen, Hubei 448000 China
| | - Hong Yu
- Department of Neonatology, The First People's of Hospital of Jingmen City, Jingmen, Hubei 448000 China
| |
Collapse
|
110
|
Kroh A, Walter J, Schüler H, Nolting J, Eickhoff R, Heise D, Neumann UP, Cramer T, Ulmer TF, Fragoulis A. A Newly Established Murine Cell Line as a Model for Hepatocellular Cancer in Non-Alcoholic Steatohepatitis. Int J Mol Sci 2019; 20:ijms20225658. [PMID: 31726709 PMCID: PMC6888677 DOI: 10.3390/ijms20225658] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) has become a major risk factor for hepatocellular cancer (HCC) due to the worldwide increasing prevalence of obesity. However, the pathophysiology of NASH and its progression to HCC is incompletely understood. Thus, the aim of this study was to generate a model specific NASH-derived HCC cell line. A murine NASH-HCC model was conducted and the obtained cancer cells (N-HCC25) were investigated towards chromosomal aberrations, the expression of cell type-specific markers, dependency on nutrients, and functional importance of mTOR. N-HCC25 exhibited several chromosomal aberrations as compared to healthy hepatocytes. Hepatocytic (HNF4), EMT (Twist, Snail), and cancer stem cell markers (CD44, EpCAM, CK19, Sox9) were simultaneously expressed in these cells. Proliferation highly depended on the supply of glucose and FBS, but not glutamine. Treatment with a second generation mTOR inhibitor (KU-0063794) resulted in a strong decrease of cell growth in a dose-dependent manner. In contrast, a first generation mTOR inhibitor (Everolimus) only slightly reduced cell proliferation. Cell cycle analyses revealed that the observed growth reduction was most likely due to G1/G0 cell cycle arrest. These results indicate that N-HCC25 is a highly proliferative HCC cell line from a NASH background, which might serve as a suitable in vitro model for future investigations of NASH-derived HCC.
Collapse
Affiliation(s)
- Andreas Kroh
- Department of General, Visceral and Transplantation Surgery, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (J.W.); (J.N.); (R.E.); (D.H.); (U.P.N.); (T.C.); (T.F.U.); (A.F.)
- Correspondence: ; Tel.: +49-241-80-89-501
| | - Jeanette Walter
- Department of General, Visceral and Transplantation Surgery, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (J.W.); (J.N.); (R.E.); (D.H.); (U.P.N.); (T.C.); (T.F.U.); (A.F.)
| | - Herdit Schüler
- Institute of Human Genetics, Uniklinik RWTH Aachen, 52074 Aachen, Germany;
| | - Jochen Nolting
- Department of General, Visceral and Transplantation Surgery, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (J.W.); (J.N.); (R.E.); (D.H.); (U.P.N.); (T.C.); (T.F.U.); (A.F.)
| | - Roman Eickhoff
- Department of General, Visceral and Transplantation Surgery, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (J.W.); (J.N.); (R.E.); (D.H.); (U.P.N.); (T.C.); (T.F.U.); (A.F.)
| | - Daniel Heise
- Department of General, Visceral and Transplantation Surgery, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (J.W.); (J.N.); (R.E.); (D.H.); (U.P.N.); (T.C.); (T.F.U.); (A.F.)
| | - Ulf Peter Neumann
- Department of General, Visceral and Transplantation Surgery, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (J.W.); (J.N.); (R.E.); (D.H.); (U.P.N.); (T.C.); (T.F.U.); (A.F.)
- Department of Surgery, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
- ESCAM—European Surgery Center Aachen Maastricht, 52074 Aachen, Germany
- ESCAM—European Surgery Center Aachen Maastricht, 6200 MD Maastricht, The Netherlands
| | - Thorsten Cramer
- Department of General, Visceral and Transplantation Surgery, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (J.W.); (J.N.); (R.E.); (D.H.); (U.P.N.); (T.C.); (T.F.U.); (A.F.)
- Department of Surgery, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
- ESCAM—European Surgery Center Aachen Maastricht, 52074 Aachen, Germany
- ESCAM—European Surgery Center Aachen Maastricht, 6200 MD Maastricht, The Netherlands
| | - Tom Florian Ulmer
- Department of General, Visceral and Transplantation Surgery, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (J.W.); (J.N.); (R.E.); (D.H.); (U.P.N.); (T.C.); (T.F.U.); (A.F.)
- Department of Surgery, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
| | - Athanassios Fragoulis
- Department of General, Visceral and Transplantation Surgery, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (J.W.); (J.N.); (R.E.); (D.H.); (U.P.N.); (T.C.); (T.F.U.); (A.F.)
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, 52074 Aachen, Germany
| |
Collapse
|
111
|
van Tienderen GS, Groot Koerkamp B, IJzermans JNM, van der Laan LJW, Verstegen MMA. Recreating Tumour Complexity in a Dish: Organoid Models to Study Liver Cancer Cells and their Extracellular Environment. Cancers (Basel) 2019; 11:E1706. [PMID: 31683901 PMCID: PMC6896153 DOI: 10.3390/cancers11111706] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
Primary liver cancer, consisting predominantly of hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), remains one of the most lethal malignancies worldwide. This high malignancy is related to the complex and dynamic interactions between tumour cells, stromal cells and the extracellular environment. Novel in vitro models that can recapitulate the tumour are essential in increasing our understanding of liver cancer. Herein, primary liver cancer-derived organoids have opened up new avenues due to their patient-specificity, self-organizing ability and potential recapitulation of many of the tumour properties. Organoids are solely of epithelial origin, but incorporation into co-culture models can enable the investigation of the cellular component of the tumour microenvironment. However, the extracellular component also plays a vital role in cancer progression and representation is lacking within current in vitro models. In this review, organoid technology is discussed in the context of liver cancer models through comparisons to other cell culture systems. In addition, the role of the tumour extracellular environment in primary liver cancer will be highlighted with an emphasis on its importance in in vitro modelling. Converging novel organoid-based models with models incorporating the native tumour microenvironment could lead to experimental models that can better recapitulate liver tumours in vivo.
Collapse
Affiliation(s)
- Gilles S van Tienderen
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands.
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands.
| | - Jan N M IJzermans
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands.
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands.
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands.
| |
Collapse
|
112
|
Contextual Regulation of TGF-β Signaling in Liver Cancer. Cells 2019; 8:cells8101235. [PMID: 31614569 PMCID: PMC6829617 DOI: 10.3390/cells8101235] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023] Open
Abstract
Primary liver cancer is one of the leading causes for cancer-related death worldwide. Transforming growth factor beta (TGF-β) is a pleiotropic cytokine that signals through membrane receptors and intracellular Smad proteins, which enter the nucleus upon receptor activation and act as transcription factors. TGF-β inhibits liver tumorigenesis in the early stage by inducing cytostasis and apoptosis, but promotes malignant progression in more advanced stages by enhancing cancer cell survival, EMT, migration, invasion and finally metastasis. Understanding the molecular mechanisms underpinning the multi-faceted roles of TGF-β in liver cancer has become a persistent pursuit during the last two decades. Contextual regulation fine-tunes the robustness, duration and plasticity of TGF-β signaling, yielding versatile albeit specific responses. This involves multiple feedback and feed-forward regulatory loops and also the interplay between Smad signaling and non-Smad pathways. This review summarizes the known regulatory mechanisms of TGF-β signaling in liver cancer, and how they channel, skew and even switch the actions of TGF-β during cancer progression.
Collapse
|
113
|
Lv X, Li Y, Li Y, Li H, Zhou L, Wang B, Zhi Z, Tang W. FAL1: A critical oncogenic long non-coding RNA in human cancers. Life Sci 2019; 236:116918. [PMID: 31610208 DOI: 10.1016/j.lfs.2019.116918] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/26/2019] [Accepted: 09/26/2019] [Indexed: 12/29/2022]
Abstract
Long noncoding RNAs (lncRNAs) are characterized as a group of endogenous RNAs that are more than 200 nucleotides in length and have no protein-encoding function. More and more evidence indicates that lncRNAs play vital roles in various human diseases, especially in tumorigenesis. Focally amplified lncRNA on chromosome 1 (FAL1), a novel lncRNA with enhancer-like activity, has been identified as an oncogene in multiple cancers and high expression level of FAL1 is usually associated with poor prognosis. Dysregulation of FAL1 has been shown to promote the proliferation and metastasis of cancer cells. In the present review, we summarized and illustrated the functions and underlying molecular mechanisms of FAL1 in the occurrence and development of different cancers and other diseases. FAL1 has the potential to appear as a feasible diagnostic and prognostic tool and new therapeutic target for cancer patients though further investigation is needed so as to accelerate clinical application.
Collapse
Affiliation(s)
- Xiurui Lv
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuhan Li
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hongxing Li
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Lingling Zhou
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Binyu Wang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengke Zhi
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Weibing Tang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
114
|
Fezza M, Moussa M, Aoun R, Haber R, Hilal G. DKK1 promotes hepatocellular carcinoma inflammation, migration and invasion: Implication of TGF-β1. PLoS One 2019; 14:e0223252. [PMID: 31568519 PMCID: PMC6768474 DOI: 10.1371/journal.pone.0223252] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 09/17/2019] [Indexed: 12/24/2022] Open
Abstract
Dickkopf-1 (DKK1), an inhibitor of the most frequently impaired signaling pathway in hepatocellular carcinoma (HCC), the Wnt/beta-catenin pathway, seems to fulfill contradictory functions in the process of tumorigenesis, acting either as an oncogenic promoter of metastasis or as a tumor suppressor. Elevated serum levels of DKK1 have been reported in HCC; however, little is known about its functional significance. In the current study, we treated HepG2/C3A and PLC/PRF/5 with the recombinant protein DKK1. Cytotoxicity was first determined by the WST-8 assay. AFP expression was measured at both the mRNA and protein levels. Expression of the oncogenes MYC, CCND1, hTERT, and MDM2 and the tumor suppressor genes TP53, P21 and RB was assessed. Western blot analysis of non-phosphorylated ẞ-catenin and Sanger sequencing were performed to explain the functional differences between the two cell lines. Subsequently, inflammation, migration and invasion were evaluated by qPCR, ELISA, the Boyden chamber assay, zymography, and MMP-2 and MMP-9 western blot analysis. Knockdown of DKK1 and TGF-β1 were also performed. Our results suggest that DKK1 exerts an oncogenic effect on HepG2/C3A cell line by upregulating the expression of oncogenes and downregulating that of tumor suppressor genes, whereas the opposite effect was demonstrated in PLC/PRF/5 cells. This differential impact of DKK1 can be explained by the mutations that affect the canonical Wnt pathway that were detected in exon 3 of the CTNNB1 gene in the HepG2 cell line. We further confirmed that DKK1 promotes inflammation, tumor invasion and migration in both cell types. The canonical pathway was not responsible for the DKK1 proinvasive effect, as indicated by the active ẞ-catenin levels in the two cell lines upon DKK1 treatment. Interestingly, knockdown of TGF-β1 negatively affected the DKK1 proinvasive effect. Taken together, DKK1 appears to facilitate tumor invasion and migration through TGF- β1 by remodeling the tumor microenvironment and inducing inflammation. This finding endorses the relevance of TGF-β1 as a therapeutic target.
Collapse
Affiliation(s)
- Maha Fezza
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Mayssam Moussa
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Rita Aoun
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Rita Haber
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - George Hilal
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
- * E-mail:
| |
Collapse
|
115
|
Li Y, Lu J, Chen Q, Han S, Shao H, Chen P, Jin Q, Yang M, Shangguan F, Fei M, Wang L, Liu Y, Liu N, Lu B. Artemisinin suppresses hepatocellular carcinoma cell growth, migration and invasion by targeting cellular bioenergetics and Hippo-YAP signaling. Arch Toxicol 2019; 93:3367-3383. [PMID: 31563988 DOI: 10.1007/s00204-019-02579-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/17/2019] [Indexed: 01/17/2023]
Abstract
The primary liver cancer (PLC) is one of the leading causes of cancer-related death worldwide. The predominant form of PLC is hepatocellular carcinoma (HCC), which accounts for about 85% of all PLC. Artemisinin (ART) was clinically used as anti-malarial agents. Recently, it was demonstrated to inhibit cell growth and migration in multiple cancer types. However, the molecular mechanism underlying these anti-cancer activity remains largely unknown. Herein, it is discovered that ART dramatically suppresses HCC cell growth in vitro through arresting cell cycle progression, and represses cell migration and invasion via regulating N-cadherin-Snail-E-cadherin axis. In addition, the disruption of cellular bioenergetics contributed to ART-caused cell growth, migration and invasion inhibition. Moreover, ART (100 mg/kg, intraperitoneally) substantially inhibits HCC xenograft growth in vivo. Importantly, Hippo-YAP signal transduction is remarkably inactivated in HCC cells upon ART administration. Collectively, these data reveal a novel mechanism of ART in regulating HCC cell growth, migration, and invasion, which indicates that ART could be considered as a potential drug for the treatment of HCC.
Collapse
Affiliation(s)
- Yujie Li
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China.,Department of Intensive Care, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jing Lu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Qin Chen
- Department of Intensive Care, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Shengnan Han
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Hua Shao
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Pingyi Chen
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Qiumei Jin
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Mingyue Yang
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Fugen Shangguan
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Mingming Fei
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Lu Wang
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China
| | - Yongzhang Liu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China. .,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Naxin Liu
- Department of Pancreatitis Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China. .,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Bin Lu
- Protein Quality Control and Diseases Laboratory, Key Laboratory of Medical Genetics of Zhejiang Province, Key Laboratory of Laboratory Medicine, Ministry of Education of China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, University-Town, Wenzhou, Zhejiang, 325035, China. .,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
116
|
Comparison of anesthetic effects of dexmedetomidine and tramadol, respectively, combined with propofol in percutaneous microwave coagulation therapy for hepatocellular carcinoma. Oncol Lett 2019; 18:3599-3604. [PMID: 31516574 PMCID: PMC6733012 DOI: 10.3892/ol.2019.10717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 07/03/2019] [Indexed: 11/21/2022] Open
Abstract
Anesthetic effects and safety of dexmedetomidine and tramadol, respectively, combined with propofol in ultrasound-guided percutaneous microwave coagulation therapy (PMCT) for hepatocellular carcinoma (HCC) were compared. One hundred and seventy-six patients with HCC, treated by ultrasound-guided PMCT in The Affiliated Hospital of Qingdao University from January 2014 to December 2016, were retrospectively analyzed and divided into two groups: dexmedetomidine group (anesthetized with dexmedetomidine combined with propofol, n=91) and tramadol group (anesthetized with tramadol combined with propofol, n=85). Changes in heart rate (HR), mean arterial pressure (MAP), and oxygen saturation (SpO2) were recorded before oxygen inhalation (T1), intraoperationally (T2), and at 30 min postoperatively (T3), and the recovery time (recorded from the moment the use of anesthetic drugs stopped), hospital stay, visual analogue scale (VAS) score at 48 h after surgery, as well as the adverse reactions in the perioperative period were compared between the two groups. HR and SpO2 in the dexmedetomidine group at T2 and T3 were significantly lower than those in the tramadol group (P<0.05). HR and SpO2 at T2 were significantly lower than those at T1 and T3, and HR at T3 was lower than that at T1 (P<0.05). MAP in the dexmedetomidine group at T2 was significantly lower than that in the tramadol group (t=3.836, P<0.001). MAP at T2 was significantly lower than those at T1 and T3, and MAP at T3 was lower than that at T1 (P<0.05). The number of patients with shivering in the dexmedetomidine group was significantly higher than that in the tramadol group (P<0.05). Both tramadol and dexmedetomidine, respectively combined with propofol in PMCT for HCC surgery can achieve satisfactory anesthetic effects. However, tramadol combined with propofol is more effective in stabilizing the vital signs with less side-effects, and is more suitable for PMCT in patients with HCC than dexmedetomidine combined with propofol, which is worth popularizing and applying in clinic.
Collapse
|
117
|
Liu J, Chen S, Zou Z, Tan D, Liu X, Wang X. Pathological Pattern of Intrahepatic HBV in HCC is Phenocopied by PDX-Derived Mice: a Novel Model for Antiviral Treatment. Transl Oncol 2019; 12:1138-1146. [PMID: 31202090 PMCID: PMC6581976 DOI: 10.1016/j.tranon.2019.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/08/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) is one of the most prominent risk factors for hepatocellular carcinoma (HCC) development and virus-mediated cases represents more than 80% of HCC in East Asia, where it is endemic. Currently, the HBV status of pathological HCC is not fully clarified, especially by comparison to nontumorous tissues. Lack of clinicopathological animal models of HCC impedes clinical application of antiviral treatment in the field. MATERIALS AND METHODS A cohort sample of 14 HCC and corresponding stroma tissues were analyzed for pathological patterns of HBV antigens using immunohistochemistry; 10 fresh primary tumor tissues were inoculated into NOD/SCID mice and risk factors for patient-derived xenograft (PDX) model were identified by the univariate F test. Consistency of HBV features and cellular biomarkers between patient tissues and tumor grafts were examined. RESULTS In HCC, HBV surface antigen (HBsAg) was mainly absent. Only 9.9% of samples showed HBsAg positivity in the tumor tissue that was limited to benign hepatocytes. In contrast, HBV core antigen (HBcAg) exhibited positive staining in all HCC tissues, located mainly in the cytoplasm of tumor cells. Of 14 HCC cases, three were diagnosed as occult infection of HBV based on HBcAg expression. The successful rate for the PDX model was 20% (2/10). Tumor lesions on hepatic lobes of V and VI, severe liver dysfunction and higher CA125 showed p-values of 0.01, 0.035, and 0.01, respectively. HBsAg absence in original tumors of #6 and 8 patients were faithfully reproduced by engraftments. Mixed distribution of HBcAg in cellular compartments of original tumor cells was also observed in mice. ki67 was dramatically increased in tumor grafts. CONCLUSION We delineated pathological HBV profiles of HCC specimens and perilesional areas, which provided evidence for virus-based therapy in the future. PDX mice may phenocopy virological and cellular features of patient tissues, which is novel in the virus-related hepatocarcinogenesis field.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Endoscope, the General Hospital of Shenyang Military Region, Shenyang 110000, PR China.
| | - Siyuan Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Army Medical University, Chongqing 400007, PR China.
| | - Zhe Zou
- Department of Gastroenterology, the Second Affiliated Hospital of Army Medical University, Chongqing 400007, PR China.
| | - Dehong Tan
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Army Medical University, Chongqing 400007, PR China.
| | - Xiangde Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Army Medical University, Chongqing 400007, PR China.
| | - Xing Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Army Medical University, Chongqing 400007, PR China.
| |
Collapse
|
118
|
Granat LM, Kambhampati O, Klosek S, Niedzwecki B, Parsa K, Zhang D. The promises and challenges of patient-derived tumor organoids in drug development and precision oncology. Animal Model Exp Med 2019; 2:150-161. [PMID: 31773090 PMCID: PMC6762043 DOI: 10.1002/ame2.12077] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/25/2019] [Accepted: 07/27/2019] [Indexed: 12/14/2022] Open
Abstract
In the era of precision medicine, cancer researchers and oncologists are eagerly searching for more realistic, cost effective, and timely tumor models to aid drug development and precision oncology. Tumor models that can faithfully recapitulate the histological and molecular characteristics of various human tumors will be extremely valuable in increasing the successful rate of oncology drug development and discovering the most efficacious treatment regimen for cancer patients. Two-dimensional (2D) cultured cancer cell lines, genetically engineered mouse tumor (GEMT) models, and patient-derived tumor xenograft (PDTX) models have been widely used to investigate the biology of various types of cancers and test the efficacy of oncology drug candidates. However, due to either the failure to faithfully recapitulate the complexity of patient tumors in the case of 2D cultured cancer cells, or high cost and untimely for drug screening and testing in the case of GEMT and PDTX, new tumor models are urgently needed. The recently developed patient-derived tumor organoids (PDTO) offer great potentials in uncovering novel biology of cancer development, accelerating the discovery of oncology drugs, and individualizing the treatment of cancers. In this review, we will summarize the recent progress in utilizing PDTO for oncology drug discovery. In addition, we will discuss the potentials and limitations of the current PDTO tumor models.
Collapse
Affiliation(s)
- Lauren M. Granat
- Department of Biomedical Sciences, College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew York
| | - Ooha Kambhampati
- Department of Biomedical Sciences, College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew York
| | - Stephanie Klosek
- Department of Biomedical Sciences, College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew York
| | - Brian Niedzwecki
- Department of Biomedical Sciences, College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew York
| | - Kian Parsa
- Department of Biomedical Sciences, College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew York
| | - Dong Zhang
- Department of Biomedical Sciences, College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew York
| |
Collapse
|
119
|
Development of an oncogenic dedifferentiation SOX signature with prognostic significance in hepatocellular carcinoma. BMC Cancer 2019; 19:851. [PMID: 31462277 PMCID: PMC6714407 DOI: 10.1186/s12885-019-6041-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 08/14/2019] [Indexed: 12/25/2022] Open
Abstract
Background Gradual loss of terminal differentiation markers and gain of stem cell-like properties is a major hall mark of cancer malignant progression. The stem cell pluripotent transcriptional factor SOX family play critical roles in governing tumor plasticity and lineage specification. This study aims to establish a novel SOX signature to monitor the extent of tumor dedifferentiation and predict prognostic significance in hepatocellular carcinoma (HCC). Methods The RNA-seq data from The Cancer Genome Atlas (TCGA) LIHC project were chronologically divided into the training (n = 188) and testing cohort (n = 189). LIRI-JP project from International Cancer Genome Consortium (ICGC) data portal was used as an independent validation cohort (n = 232). Kaplan-Meier and multivariable Cox analyses were used to examine the clinical significance and prognostic value of the signature genes. Results The SOX gene family members were found to be aberrantly expressed in clinical HCC patients. A five-gene SOX signature with prognostic value was established in the training cohort. The SOX signature genes were found to be closely associated with tumor grade and tumor stage. Liver cancer dedifferentiation markers (AFP, CD133, EPCAM, and KRT19) were found to be progressively increased while hepatocyte terminal differentiation markers (ALB, G6PC, CYP3A4, and HNF4A) were progressively decreased from HCC patients with low SOX signature scores to patients with high SOX signature scores. Kaplan-Meier survival analysis further indicated that the newly established SOX signature could robustly predict patient overall survival in both training, testing, and independent validation cohort. Conclusions An oncogenic dedifferentiation SOX signature presents a great potential in predicting prognostic significance in HCC, and might provide novel biomarkers for precision oncology further in the clinic. Electronic supplementary material The online version of this article (10.1186/s12885-019-6041-2) contains supplementary material, which is available to authorized users.
Collapse
|
120
|
Johnson RL. Hippo signaling and epithelial cell plasticity in mammalian liver development, homeostasis, injury and disease. SCIENCE CHINA-LIFE SCIENCES 2019; 62:1609-1616. [PMID: 31463737 DOI: 10.1007/s11427-018-9510-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 06/15/2019] [Indexed: 12/11/2022]
Abstract
A traditional view of cellular differentiation is unidirectional: progenitor cells adopt specific fates in response to environmental cues resulting in deployment of cell-specific gene expression programs and acquisition of unique differentiated cellular properties such as production of structural and functional proteins that define individual cell types. In both development and in tissue repair stem and progenitor cells are thought to both self-renew to maintain the pool of precursors and to expand to give rise to transient amplifying and differentiated cell types. Recently, however, it has become appreciated that differentiated cell types can be reprogrammed to adopt progenitor and stem cell properties. In the case of epithelial cells in the mammalian liver, hepatocytes and biliary epithelial cells there is a significant degree of plasticity between these lineages that has been implicated in mechanisms of tissue repair and in liver pathologies such as cancer. Recent studies have highlighted the role of Hippo signaling, an emerging growth control and tumor suppressor pathway, in regulating epithelial cell plasticity in the mammalian liver and in this review, the role of cellular plasticity and Hippo signaling in regulating normal and abnormal tissue responses in the mammalian liver will be discussed.
Collapse
Affiliation(s)
- Randy L Johnson
- Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, 6767 Bertner Ave, Houston, TX, 77030, USA.
| |
Collapse
|
121
|
Di Blasi D, Boldanova T, Mori L, Terracciano L, Heim MH, De Libero G. Unique T-Cell Populations Define Immune-Inflamed Hepatocellular Carcinoma. Cell Mol Gastroenterol Hepatol 2019; 9:195-218. [PMID: 31445190 PMCID: PMC6957799 DOI: 10.1016/j.jcmgh.2019.08.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS The characterization of T cells infiltrating hepatocellular carcinoma (HCC) provides information on cancer immunity and also on selection of patients with precise indication of immunotherapy. The aim of the study was to characterize T-cell populations within tumor tissue and compare them with non-neoplastic liver tissue as well as circulating cells of the same patients. METHODS The presence of unique cell populations was investigated in 36 HCC patients by multidimensional flow cytometry followed by t-distributed stochastic neighbor embedding analysis. Functional activity of tumor-infiltrating T cells was determined after activation by phorbol 12-myristate 13-acetate and ionomycin. RESULTS Within the tumor there were more cells expressing CD137 and ICOS than in non-neoplastic liver tissue, possibly after recent antigenic activation. These cells contained several populations, including the following: (1) functionally impaired, proliferating CD4+ cells co-expressing Inducible T-cell costimulator (ICOS) and T cell immunoreceptor with Ig and ITIM domains (TIGIT); (2) functionally active CD8+ cells co-expressing CD38 and Programmed cell-death protein 1 (PD1); and (3) CD4-CD8 double-negative T-cell receptor αβ and γδ cells (both non-major histocompatibility complex-restricted T cells). When the identified clusters were compared with histologic classification performed on the same samples, an accumulation of activated T cells was observed in immune-inflamed HCC. The same analyses performed in 7 patients receiving nivolumab treatment showed a remarkable reduction in the functionally impaired CD4+ cells, which returned to almost normal activity over time. CONCLUSIONS Unique populations of activated T cells are present in HCC tissue, whose antigen specificity remains to be investigated. Some of these cell populations are functionally impaired and nivolumab treatment restores their responsiveness. The finding of ongoing immune response within the tumor shows which lymphocyte populations are impaired within the HCC and identifies the patients who might take benefit from immunotherapy.
Collapse
Affiliation(s)
- Daniela Di Blasi
- Experimental Immunology, Department of Biomedicine, University of Basel, Switzerland,Hepatology Laboratory, Department of Biomedicine, University of Basel, Switzerland
| | - Tujana Boldanova
- Hepatology Laboratory, Department of Biomedicine, University of Basel, Switzerland,Division of Gastroenterology and Hepatology, University Hospital Basel, Basel, Switzerland
| | - Lucia Mori
- Experimental Immunology, Department of Biomedicine, University of Basel, Switzerland
| | - Luigi Terracciano
- Institute of Pathology, Division of Molecular Pathology, University Hospital Basel, Basel, Switzerland
| | - Markus H. Heim
- Hepatology Laboratory, Department of Biomedicine, University of Basel, Switzerland,Division of Gastroenterology and Hepatology, University Hospital Basel, Basel, Switzerland,Correspondence Address correspondence to: Gennaro De Libero, MD, or Markus H. Heim, MD, Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland. fax: +41 61 265 23 50.
| | - Gennaro De Libero
- Experimental Immunology, Department of Biomedicine, University of Basel, Switzerland,Correspondence Address correspondence to: Gennaro De Libero, MD, or Markus H. Heim, MD, Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland. fax: +41 61 265 23 50.
| |
Collapse
|
122
|
Czauderna C, Castven D, Mahn FL, Marquardt JU. Context-Dependent Role of NF-κB Signaling in Primary Liver Cancer-from Tumor Development to Therapeutic Implications. Cancers (Basel) 2019; 11:cancers11081053. [PMID: 31349670 PMCID: PMC6721782 DOI: 10.3390/cancers11081053] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammatory cell death is a major risk factor for the development of diverse cancers including liver cancer. Herein, disruption of the hepatic microenvironment as well as the immune cell composition are major determinants of malignant transformation and progression in hepatocellular carcinomas (HCC). Considerable research efforts have focused on the identification of predisposing factors that promote induction of an oncogenic field effect within the inflammatory liver microenvironment. Among the most prominent factors involved in this so-called inflammation-fibrosis-cancer axis is the NF-κB pathway. The dominant role of this pathway for malignant transformation and progression in HCC is well documented. Pathway activation is significantly linked to poor prognostic traits as well as stemness characteristics, which places modulation of NF-κB signaling in the focus of therapeutic interventions. However, it is well recognized that the mechanistic importance of the pathway for HCC is highly context and cell type dependent. While constitutive pathway activation in an inflammatory etiological background can significantly promote HCC development and progression, absence of NF-κB signaling in differentiated liver cells also significantly enhances liver cancer development. Thus, therapeutic targeting of NF-κB as well as associated family members may not only exert beneficial effects but also negatively impact viability of healthy hepatocytes and/or cholangiocytes, respectively. The review presented here aims to decipher the complexity and paradoxical functions of NF-κB signaling in primary liver and non-parenchymal cells, as well as the induced molecular alterations that drive HCC development and progression with a particular focus on (immune-) therapeutic interventions.
Collapse
Affiliation(s)
- Carolin Czauderna
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Darko Castven
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Friederike L Mahn
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Jens U Marquardt
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany.
| |
Collapse
|
123
|
Wang Y, Zhu P, Luo J, Wang J, Liu Z, Wu W, Du Y, Ye B, Wang D, He L, Ren W, Wang J, Sun X, Chen R, Tian Y, Fan Z. LncRNA HAND2-AS1 promotes liver cancer stem cell self-renewal via BMP signaling. EMBO J 2019; 38:e101110. [PMID: 31334575 DOI: 10.15252/embj.2018101110] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 06/07/2019] [Accepted: 06/13/2019] [Indexed: 12/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent liver cancer, characterized by a high rate of recurrence and heterogeneity. Liver cancer stem cells (CSCs) may well contribute to both of these pathological properties, but the mechanism underlying their self-renewal maintenance is poorly understood. Here, we identified a long noncoding RNA (lncRNA) termed HAND2-AS1 that is highly expressed in liver CSCs. Human HAND2-AS1 and its mouse ortholog lncHand2 display a high level of conservation. HAND2-AS1 is required for the self-renewal maintenance of liver CSCs to initiate HCC development. Mechanistically, HAND2-AS1 recruits the INO80 chromatin-remodeling complex to the promoter of BMPR1A, thereby inducing its expression and leading to the activation of BMP signaling. Importantly, interfering with expression of HAND2-AS1 by antisense oligonucleotides (ASOs) and BMPR1A by siRNAs has synergistic anti-tumorigenic effects on humanized HCC models. Moreover, knockout of lncHand2 or Bmpr1a in mouse hepatocytes impairs BMP signaling and suppresses the initiation of liver cancer. Our findings reveal that HAND2-AS1 promotes the self-renewal of liver CSCs and drives liver oncogenesis, offering a potential new target for HCC therapy.
Collapse
Affiliation(s)
- Yanying Wang
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Pingping Zhu
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jianjun Luo
- CAS Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jing Wang
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhiwei Liu
- Department of Hepatobiliary Surgery, PLA General Hospital, Beijing, China
| | - Wei Wu
- CAS Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Ying Du
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Buqing Ye
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Dongpeng Wang
- CAS Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lei He
- Department of Hepatobiliary Surgery, PLA General Hospital, Beijing, China
| | - Weizheng Ren
- Department of Hepatobiliary Surgery, PLA General Hospital, Beijing, China
| | - Jianyi Wang
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xianhui Sun
- CAS Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Runsheng Chen
- CAS Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yong Tian
- CAS Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zusen Fan
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
124
|
Modelling liver cancer initiation with organoids derived from directly reprogrammed human hepatocytes. Nat Cell Biol 2019; 21:1015-1026. [PMID: 31332348 DOI: 10.1038/s41556-019-0359-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 06/11/2019] [Indexed: 12/24/2022]
Abstract
Human liver cancers, including hepatocellular carcinomas and intra-hepatic cholangiocarcinomas, are often diagnosed late with poor prognosis. A better understanding of cancer initiation could provide potential preventive therapies and increase survival. Models for studying human liver cancer initiation are largely missing. Here, using directly reprogrammed human hepatocytes (hiHeps) and inactivation of p53 and RB, we established organoids possessing liver architecture and function. HiHep organoids were genetically engineered to model the initial alterations in human liver cancers. Bona fide hepatocellular carcinomas were developed by overexpressing c-Myc. Excessive mitochondrion-endoplasmic reticulum coupling induced by c-Myc facilitated hepatocellular carcinoma initiation and seemed to be a target of preventive treatment. Furthermore, through the analysis of human intra-hepatic cholangiocarcinoma-enriched mutations, we demonstrate that the RAS-induced lineage conversion from hepatocytes to intra-hepatic cholangiocarcinoma cells can be prevented by the combined inhibition of Notch and JAK-STAT. Together, hiHep organoids represent a system that can be genetically manipulated to model cancer initiation and identify potential preventive therapies.
Collapse
|
125
|
Chan LC, Li CW, Xia W, Hsu JM, Lee HH, Cha JH, Wang HL, Yang WH, Yen EY, Chang WC, Zha Z, Lim SO, Lai YJ, Liu C, Liu J, Dong Q, Yang Y, Sun L, Wei Y, Nie L, Hsu JL, Li H, Ye Q, Hassan MM, Amin HM, Kaseb AO, Lin X, Wang SC, Hung MC. IL-6/JAK1 pathway drives PD-L1 Y112 phosphorylation to promote cancer immune evasion. J Clin Invest 2019; 129:3324-3338. [PMID: 31305264 PMCID: PMC6668668 DOI: 10.1172/jci126022] [Citation(s) in RCA: 233] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 05/21/2019] [Indexed: 02/06/2023] Open
Abstract
Glycosylation of immune receptors and ligands, such as T cell receptor and coinhibitory molecules, regulates immune signaling activation and immune surveillance. However, how oncogenic signaling initiates glycosylation of coinhibitory molecules to induce immunosuppression remains unclear. Here we show that IL-6-activated JAK1 phosphorylates programmed death-ligand 1 (PD-L1) Tyr112, which recruits the endoplasmic reticulum-associated N-glycosyltransferase STT3A to catalyze PD-L1 glycosylation and maintain PD-L1 stability. Targeting of IL-6 by IL-6 antibody induced synergistic T cell killing effects when combined with anti-T cell immunoglobulin mucin-3 (anti-Tim-3) therapy in animal models. A positive correlation between IL-6 and PD-L1 expression was also observed in hepatocellular carcinoma patient tumor tissues. These results identify a mechanism regulating PD-L1 glycosylation initiation and suggest the combination of anti-IL-6 and anti-Tim-3 as an effective marker-guided therapeutic strategy.
Collapse
Affiliation(s)
- Li-Chuan Chan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Chia-Wei Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Weiya Xia
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jung-Mao Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Heng-Huan Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jong-Ho Cha
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Hung-Ling Wang
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Hao Yang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Er-Yen Yen
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Wei-Chao Chang
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Zhengyu Zha
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Seung-Oe Lim
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yun-Ju Lai
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Chunxiao Liu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jielin Liu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Qiongzhu Dong
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of General Surgery, Huashan Hospital and Cancer Metastasis Institute and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yi Yang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Linlin Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yongkun Wei
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lei Nie
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer L. Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Hui Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Qinghai Ye
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Manal M. Hassan
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hesham M. Amin
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ahmed O. Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xin Lin
- Institute for Immunology, Tsinghua University School of Medicine, Beijing, China
| | - Shao-Chun Wang
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
126
|
Sharma S, Mehak, Chhimwal J, Patial V, Sk UH. Dendrimer-conjugated podophyllotoxin suppresses DENA-induced HCC progression by modulation of inflammatory and fibrogenic factors. Toxicol Res (Camb) 2019; 8:560-567. [PMID: 31367338 PMCID: PMC6621132 DOI: 10.1039/c9tx00103d] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 05/21/2019] [Indexed: 12/13/2022] Open
Abstract
Podophyllotoxin has been explored as an anticancer, antiviral, and antibacterial agent; however, its low water solubility and toxicity limit its use. In this study, the efficacy of a more soluble and less toxic polyamidoamine (PAMAM) dendrimer-conjugated podophyllotoxin (DPODO) was evaluated against chemically induced hepatocellular carcinoma (HCC) in mice. HCC was induced by giving 0.01% diethylnitrosamine (DENA) in drinking water for 16 weeks. The HCC-induced mice were treated with 10 or 20 mg per kg body weight DPODO. The DENA administration led to HCC development, characterized by anisocytosis, karyomegaly, inflammation and degenerative changes in the liver. The DPODO treatment at 10 mg and 20 mg doses significantly reduced the histopathological changes in liver tissue. The DPODO treatment also significantly lowered the levels of inflammatory markers IL-6 and NF-κB in serum and tissue, respectively. Further, the treatment also significantly reduced fibrous tissue deposition in the liver, which was further confirmed by the reduced mRNA levels and tissue expression of fibrogenic markers TGF-β and α-SMA in the liver. The results of the present study indicate that DPODO treatment suppresses the progression of HCC by modulating the inflammatory and fibrogenic factors, which play important roles in HCC development.
Collapse
Affiliation(s)
- Supriya Sharma
- Pharmacology and Toxicology Laboratory , Institute of Himalayan Bioresource Technology , Palampur , H.P. , India . ;
- Academy of Scientific & Innovative Research , Institute of Himalayan Bioresource Technology , Palampur , H.P. , India
| | - Mehak
- Natural Products Chemistry and Process Development Division , Institute of Himalayan Bioresource Technology , Palampur , H.P. , India
| | - Jyoti Chhimwal
- Pharmacology and Toxicology Laboratory , Institute of Himalayan Bioresource Technology , Palampur , H.P. , India . ;
- Academy of Scientific & Innovative Research , Institute of Himalayan Bioresource Technology , Palampur , H.P. , India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory , Institute of Himalayan Bioresource Technology , Palampur , H.P. , India . ;
- Academy of Scientific & Innovative Research , Institute of Himalayan Bioresource Technology , Palampur , H.P. , India
| | - Ugir Hossain Sk
- Natural Products Chemistry and Process Development Division , Institute of Himalayan Bioresource Technology , Palampur , H.P. , India
- Clinical and Translational Research , Chittaranjan National Cancer Institute , Kolkata 700026 , India .
| |
Collapse
|
127
|
Pollicino T, Musolino C, Saitta C, Tripodi G, Lanza M, Raffa G, Tocco FCD, Raggi C, Bragazzi MC, Barbera A, Navarra G, Invernizzi P, Alvaro D, Raimondo G. Free episomal and integrated HBV DNA in HBsAg-negative patients with intrahepatic cholangiocarcinoma. Oncotarget 2019; 10:3931-3938. [PMID: 31231470 PMCID: PMC6570464 DOI: 10.18632/oncotarget.27002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/20/2019] [Indexed: 02/07/2023] Open
Abstract
There is evidence that chronic hepatitis B virus (HBV) infection is associated with an increased risk of intrahepatic cholangiocarcinoma (ICC) development, and it has been hypothesized an etiological role of HBV in the development of this tumor. Very little is known about occult HBV infection (OBI) in ICC. Aims of the study were to investigate the OBI prevalence and to characterize the HBV molecular status at intrahepatic level in OBI-positive cases with ICC. Frozen liver tumor specimens from 47 HBV surface-antigen-negative patients with ICC and 41 paired non-tumor liver tissues were tested for OBI by 4 different HBV-specific nested PCR. Covalently closed circular HBV DNA (HBV cccDNA) and viral integrations were investigated in OBI-positive cases. HBV DNA was detected in tumor and/or non-tumor specimens from 29/47 (61.7%) ICC patients. HBV cccDNA was found in tissues from 5/17 (34.5%) cases examined. HBV integration was detected in 4/10 (40%) tumor tissues tested and involved HBx and HBV-core gene sequences in 3 and 1 cases, respectively. Viral integration occurred: (a) 9,367 nucleotides upstream of the cat-eye-syndrome critical region protein-5-isoform coding sequence; (b) within the cystinosin isoform-1-precursor gene; (c) within the thromboxane-A-synthase-1 gene; (d) within the ATPase phospholipid transporting 9B gene. Occult HBV infection is highly prevalent in patients with ICC. Both free viral genomes and integrated HBV DNA can be present in these cases. These results suggest an involvement of HBV in the carcinogenic process leading to ICC development even in cases with occult infection.
Collapse
Affiliation(s)
- Teresa Pollicino
- Division of Clinical and Molecular Hepatology, University Hospital of Messina, Italy.,Department of Human Pathology, University of Messina, Italy
| | - Cristina Musolino
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Carlo Saitta
- Division of Clinical and Molecular Hepatology, University Hospital of Messina, Italy
| | - Gianluca Tripodi
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Marika Lanza
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Giuseppina Raffa
- Division of Clinical and Molecular Hepatology, University Hospital of Messina, Italy.,Department of Clinical and Experimental Medicine, University of Messina, Italy
| | | | - Chiara Raggi
- Humanitas Research and Clinical Center, Rozzano, Milan, Italy.,Present address: Department of Experimental and Clinical Medicine, University of Florence, Italy
| | | | - Adalberto Barbera
- Department of Human Pathology, University of Messina, Italy.,Division of Surgical Oncology, University Hospital of Messina, Italy
| | - Giuseppe Navarra
- Department of Human Pathology, University of Messina, Italy.,Division of Surgical Oncology, University Hospital of Messina, Italy
| | - Pietro Invernizzi
- Humanitas Research and Clinical Center, Rozzano, Milan, Italy.,Present address: Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano - Bicocca, Milan, Italy
| | - Domenico Alvaro
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Giovanni Raimondo
- Division of Clinical and Molecular Hepatology, University Hospital of Messina, Italy.,Department of Clinical and Experimental Medicine, University of Messina, Italy
| |
Collapse
|
128
|
Samie HAA, Saeed M, Faisal SM, Kausar MA, Kamal MA. Recent Findings on Nanotechnology-based Therapeutic Strategies Against Hepatocellular Carcinoma. Curr Drug Metab 2019; 20:283-291. [DOI: 10.2174/1389200220666190308134351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 12/14/2018] [Accepted: 02/14/2019] [Indexed: 02/07/2023]
Abstract
Background:
Nanotechnology-based therapies are emerging as a promising new anticancer approach.
Early clinical studies suggest that nanoparticle-based therapeutics can show enhanced efficacy while reducing side
effects minimal, owing to targeted delivery and active intracellular uptake.
Methods:
To overcome the problems of gene and drug delivery, nanotechnology based delivery system gained interest
in the last two decades. Encouraging results from Nano formulation based drug delivery systems revealed that
these emerging restoratives can efficiently lead to more effective, targeted, selective and efficacious delivery of chemotherapeutic
agents to the affected target cells.
Results:
Nanotechnology not only inhibits targeted gene products in patients with cancer, but also taught us valuable
lessons regarding appropriate dosages and route of administrations. Besides, nanotechnology based therapeutics
holds remarkable potential as an effective drug delivery system. We critically highlight the recent findings on
nanotechnology mediated therapeutics strategies to combat hepatocellular carcinoma and discuss how nanotechnology
platform can have enhanced anticancer effects compared with the parent therapeutic agents they contain.
Conclusion:
In this review, we discussed the key challenges, recent findings and future perspective in the development
of effective nanotechnology-based cancer therapeutics. The emphasis here is focused on nanotechnology-based
therapies that are likely to affect clinical investigations and their implications for advancing the treatment of patients
with hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hany A. Abdel Samie
- Department of Zoology, Faculty of Science, Menoufia University, Al Minufya, Egypt
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail, Saudi Arabia
| | - Syed Mohd Faisal
- Molecular Immunology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh-202002, India
| | - Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Hail, Saudi Arabia
| | - Mohammad A. Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
129
|
Abstract
Prognosis of combined hepatocellular carcinoma-intrahepatic cholangiocarcinoma, a type of primary liver cancer comprising areas with HCC and ICC histopathology, is dismal, and it is unclear if such tumors develop clonally and how they should be treated. In this issue of Cancer Cell, Xue et al. (2019) provide answers to these questions.
Collapse
Affiliation(s)
- Marco Seehawer
- Department of Medical Oncology & Pneumology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Luana D'Artista
- Department of Medical Oncology & Pneumology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Lars Zender
- Department of Medical Oncology & Pneumology, University Hospital Tübingen, 72076 Tübingen, Germany; German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany.
| |
Collapse
|
130
|
Xue R, Chen L, Zhang C, Fujita M, Li R, Yan SM, Ong CK, Liao X, Gao Q, Sasagawa S, Li Y, Wang J, Guo H, Huang QT, Zhong Q, Tan J, Qi L, Gong W, Hong Z, Li M, Zhao J, Peng T, Lu Y, Lim KHT, Boot A, Ono A, Chayama K, Zhang Z, Rozen SG, Teh BT, Wang XW, Nakagawa H, Zeng MS, Bai F, Zhang N. Genomic and Transcriptomic Profiling of Combined Hepatocellular and Intrahepatic Cholangiocarcinoma Reveals Distinct Molecular Subtypes. Cancer Cell 2019; 35:932-947.e8. [PMID: 31130341 PMCID: PMC8317046 DOI: 10.1016/j.ccell.2019.04.007] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 03/22/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023]
Abstract
We performed genomic and transcriptomic sequencing of 133 combined hepatocellular and intrahepatic cholangiocarcinoma (cHCC-ICC) cases, including separate, combined, and mixed subtypes. Integrative comparison of cHCC-ICC with hepatocellular carcinoma and intrahepatic cholangiocarcinoma revealed that combined and mixed type cHCC-ICCs are distinct subtypes with different clinical and molecular features. Integrating laser microdissection, cancer cell fraction analysis, and single nucleus sequencing, we revealed both mono- and multiclonal origins in the separate type cHCC-ICCs, whereas combined and mixed type cHCC-ICCs were all monoclonal origin. Notably, cHCC-ICCs showed significantly higher expression of Nestin, suggesting Nestin may serve as a biomarker for diagnosing cHCC-ICC. Our results provide important biological and clinical insights into cHCC-ICC.
Collapse
MESH Headings
- Asia
- Bile Duct Neoplasms/chemistry
- Bile Duct Neoplasms/classification
- Bile Duct Neoplasms/genetics
- Bile Duct Neoplasms/pathology
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Carcinoma, Hepatocellular/chemistry
- Carcinoma, Hepatocellular/classification
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Cholangiocarcinoma/chemistry
- Cholangiocarcinoma/classification
- Cholangiocarcinoma/genetics
- Cholangiocarcinoma/pathology
- Databases, Genetic
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Gene Regulatory Networks
- Humans
- Immunohistochemistry
- Liver Neoplasms/chemistry
- Liver Neoplasms/classification
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Male
- Neoplasms, Complex and Mixed/chemistry
- Neoplasms, Complex and Mixed/classification
- Neoplasms, Complex and Mixed/genetics
- Neoplasms, Complex and Mixed/pathology
- Nestin/analysis
- Nestin/genetics
- Predictive Value of Tests
- Prognosis
- Transcriptome
- Up-Regulation
Collapse
Affiliation(s)
- Ruidong Xue
- Biomedical Pioneering Innovation Center (BIOPIC) and Translational Cancer Research Center, School of Life Sciences, First Hospital, Peking University, Beijing 100871, China
| | - Lu Chen
- Department of Hepatobiliary Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Chong Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and Translational Cancer Research Center, School of Life Sciences, First Hospital, Peking University, Beijing 100871, China
| | - Masashi Fujita
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Ruoyan Li
- Biomedical Pioneering Innovation Center (BIOPIC) and Translational Cancer Research Center, School of Life Sciences, First Hospital, Peking University, Beijing 100871, China
| | - Shu-Mei Yan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Choon Kiat Ong
- Lymphoma Genomic Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai 200032, China
| | - Shota Sasagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Yanmeng Li
- Biomedical Pioneering Innovation Center (BIOPIC) and Translational Cancer Research Center, School of Life Sciences, First Hospital, Peking University, Beijing 100871, China
| | - Jincheng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and Translational Cancer Research Center, School of Life Sciences, First Hospital, Peking University, Beijing 100871, China
| | - Hua Guo
- Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Qi-Tao Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Qian Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Jing Tan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Wenchen Gong
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Zhixian Hong
- Department of Hepatobiliary Surgery, Beijing 302 Hospital, Beijing 100039, China
| | - Meng Li
- Department of Ultrasonography, Beijing 302 Hospital, Beijing 100039, China
| | - Jingmin Zhao
- Department of Pathology and Hepatology, Beijing 302 Hospital, Beijing 100039, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yinying Lu
- Comprehensive Liver Cancer Center, Beijing 302 Hospital, Beijing 100039, China
| | - Kiat Hon Tony Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Arnoud Boot
- Centre for Computational Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Atushi Ono
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and Translational Cancer Research Center, School of Life Sciences, First Hospital, Peking University, Beijing 100871, China
| | - Steve George Rozen
- Centre for Computational Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Hidewaki Nakagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan.
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China.
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC) and Translational Cancer Research Center, School of Life Sciences, First Hospital, Peking University, Beijing 100871, China.
| | - Ning Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and Translational Cancer Research Center, School of Life Sciences, First Hospital, Peking University, Beijing 100871, China; Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.
| |
Collapse
|
131
|
O'Rourke CJ, Lafuente-Barquero J, Andersen JB. Epigenome Remodeling in Cholangiocarcinoma. Trends Cancer 2019; 5:335-350. [PMID: 31208696 DOI: 10.1016/j.trecan.2019.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 12/22/2022]
Abstract
Cholangiocarcinoma (CCA) comprises a heterogeneous collection of malignancies arising within the biliary tract, characterized by late diagnosis, innate chemoresistance, and abysmal prognosis. Sequencing data have uncovered recurrent mutations in diverse epigenetic regulators, implicating epigenetic destabilization at the root of these tumors. However, few studies have characterized biliary tumor epigenomes. In this Opinion article, we argue that an epigenome-oriented approach to CCA could establish diverse interconnections between many key aspects of research on this disease, including molecular heterogeneity, diverse cells of origin, and prominent tumor microenvironments. Moreover, we discuss plausible causes of epigenome dysregulation in biliary tumors, including genetic, epigenetic, metabolic, microenvironmental, and physiological factors. Lastly, we assess the translational potential of epigenomics in CCA to uncover robust biomarkers and therapeutic opportunities for this growing group of patients with limited treatment options.
Collapse
Affiliation(s)
- Colm J O'Rourke
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Juan Lafuente-Barquero
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark.
| |
Collapse
|
132
|
Zhu YJ, Zheng B, Luo GJ, Ma XK, Lu XY, Lin XM, Yang S, Zhao Q, Wu T, Li ZX, Liu XL, Wu R, Liu JF, Ge Y, Yang L, Wang HY, Chen L. Circular RNAs negatively regulate cancer stem cells by physically binding FMRP against CCAR1 complex in hepatocellular carcinoma. Theranostics 2019; 9:3526-3540. [PMID: 31281495 PMCID: PMC6587157 DOI: 10.7150/thno.32796] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/17/2019] [Indexed: 12/17/2022] Open
Abstract
Circular RNA (circRNA) possesses great pre-clinical diagnostic and therapeutic potentials in multiple cancers. It has been reported playing roles in multiple malignant behaviors including proliferation, migration, metastasis and chemoresistance. However, the underlying correlation between circRNAs and cancer stem cells (CSCs) has not been reported yet. Methods: circZKSCAN1 level was detected in HCC tissue microarrays to clarify its prognostic values. Gain and loss function experiments were applied to investigate the role of circZKSCAN1 in HCC stemness. Bioinformatic analysis was used to predict the possible downstream RNA binding protein and further RNA immunoprecipitation sequencing was carried out to identify the RBP-regulated genes. Results: The absence of circZKSCAN1 endowed several malignant properties including cancer stemness and tightly correlated with worse overall and recurrence-free survival rate in HCC. Bioinformatics analysis and RNA immunoprecipitation-sequencing (RIP-seq) results revealed that circZKSCAN1 exerted its inhibitive role by competitively binding FMRP, therefore, block the binding between FMRP and β-catenin-binding protein-cell cycle and apoptosis regulator 1 (CCAR1) mRNA, and subsequently restrain the transcriptional activity of Wnt signaling. In addition, RNA-splicing protein Quaking 5 was found downregulated in HCC tissues and responsible for the reduction of circZKSCAN1. Conclusion: Collectively, this study revealed the mechanisms underlying the regulatory role of circZKSCAN1 in HCC CSCs and identified the newly discovered Qki5-circZKSCAN1-FMRP-CCAR1-Wnt signaling axis as a potentially important therapeutic target for HCC treatment.
Collapse
|
133
|
Goeppert B, Toth R, Singer S, Albrecht T, Lipka DB, Lutsik P, Brocks D, Baehr M, Muecke O, Assenov Y, Gu L, Endris V, Stenzinger A, Mehrabi A, Schirmacher P, Plass C, Weichenhan D, Roessler S. Integrative Analysis Defines Distinct Prognostic Subgroups of Intrahepatic Cholangiocarcinoma. Hepatology 2019; 69:2091-2106. [PMID: 30615206 PMCID: PMC6594081 DOI: 10.1002/hep.30493] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 01/03/2019] [Indexed: 12/11/2022]
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is the second most common primary liver cancer. It is defined by cholangiocytic differentiation and has poor prognosis. Recently, epigenetic processes have been shown to play an important role in cholangiocarcinogenesis. We performed an integrative analysis on 52 iCCAs using both genetic and epigenetic data with a specific focus on DNA methylation components. We found recurrent isocitrate dehydrogenase 1 (IDH1) and IDH2 (28%) gene mutations, recurrent arm-length copy number alterations (CNAs), and focal alterations such as deletion of 3p21 or amplification of 12q15, which affect BRCA1 Associated Protein 1, polybromo 1, and mouse double minute 2 homolog. DNA methylome analysis revealed excessive hypermethylation of iCCA, affecting primarily the bivalent genomic regions marked with both active and repressive histone modifications. Integrative clustering of genetic and epigenetic data identified four iCCA subgroups with prognostic relevance further designated as IDH, high (H), medium (M), and low (L) alteration groups. The IDH group consisted of all samples with IDH1 or IDH2 mutations and showed, together with the H group, a highly disrupted genome, characterized by frequent deletions of chromosome arms 3p and 6q. Both groups showed excessive hypermethylation with distinct patterns. The M group showed intermediate characteristics regarding both genetic and epigenetic marks, whereas the L group exhibited few methylation changes and mutations and a lack of CNAs. Methylation-based latent component analysis of cell-type composition identified differences among these four groups. Prognosis of the H and M groups was significantly worse than that of the L group. Conclusion: Using an integrative genomic and epigenomic analysis approach, we identified four major iCCA subgroups with widespread genomic and epigenomic differences and prognostic implications. Furthermore, our data suggest differences in the cell-of-origin of the iCCA subtypes.
Collapse
Affiliation(s)
- Benjamin Goeppert
- Institute of PathologyUniversity Clinic of HeidelbergHeidelbergGermany,Liver Cancer Center HeidelbergHeidelbergGermany
| | - Reka Toth
- Division of Cancer EpigenomicsGerman Cancer Research CenterHeidelbergGermany
| | - Stephan Singer
- Institute of PathologyUniversity Clinic of HeidelbergHeidelbergGermany,Institute of PathologyErnst‐Moritz‐Arndt UniversityGreifswaldGermany
| | - Thomas Albrecht
- Institute of PathologyUniversity Clinic of HeidelbergHeidelbergGermany
| | - Daniel B. Lipka
- Division of Cancer EpigenomicsGerman Cancer Research CenterHeidelbergGermany
| | - Pavlo Lutsik
- Division of Cancer EpigenomicsGerman Cancer Research CenterHeidelbergGermany
| | - David Brocks
- Division of Cancer EpigenomicsGerman Cancer Research CenterHeidelbergGermany
| | - Marion Baehr
- Division of Cancer EpigenomicsGerman Cancer Research CenterHeidelbergGermany
| | - Oliver Muecke
- Division of Cancer EpigenomicsGerman Cancer Research CenterHeidelbergGermany
| | - Yassen Assenov
- Division of Cancer EpigenomicsGerman Cancer Research CenterHeidelbergGermany
| | - Lei Gu
- Division of Cancer EpigenomicsGerman Cancer Research CenterHeidelbergGermany,Boston Children's HospitalBostonMA
| | - Volker Endris
- Institute of PathologyUniversity Clinic of HeidelbergHeidelbergGermany
| | | | - Arianeb Mehrabi
- Liver Cancer Center HeidelbergHeidelbergGermany,Department of General Visceral and Transplantation SurgeryUniversity Hospital HeidelbergHeidelbergGermany
| | - Peter Schirmacher
- Institute of PathologyUniversity Clinic of HeidelbergHeidelbergGermany,Liver Cancer Center HeidelbergHeidelbergGermany,German Consortium for Translational Cancer ResearchHeidelbergGermany
| | - Christoph Plass
- Division of Cancer EpigenomicsGerman Cancer Research CenterHeidelbergGermany,German Consortium for Translational Cancer ResearchHeidelbergGermany
| | - Dieter Weichenhan
- Division of Cancer EpigenomicsGerman Cancer Research CenterHeidelbergGermany
| | - Stephanie Roessler
- Institute of PathologyUniversity Clinic of HeidelbergHeidelbergGermany,Liver Cancer Center HeidelbergHeidelbergGermany
| |
Collapse
|
134
|
Mou H, Ozata DM, Smith JL, Sheel A, Kwan SY, Hough S, Kucukural A, Kennedy Z, Cao Y, Xue W. CRISPR-SONIC: targeted somatic oncogene knock-in enables rapid in vivo cancer modeling. Genome Med 2019; 11:21. [PMID: 30987660 PMCID: PMC6466773 DOI: 10.1186/s13073-019-0627-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 03/08/2019] [Indexed: 12/26/2022] Open
Abstract
CRISPR/Cas9 has revolutionized cancer mouse models. Although loss-of-function genetics by CRISPR/Cas9 is well-established, generating gain-of-function alleles in somatic cancer models is still challenging because of the low efficiency of gene knock-in. Here we developed CRISPR-based Somatic Oncogene kNock-In for Cancer Modeling (CRISPR-SONIC), a method for rapid in vivo cancer modeling using homology-independent repair to integrate oncogenes at a targeted genomic locus. Using a dual guide RNA strategy, we integrated a plasmid donor in the 3'-UTR of mouse β-actin, allowing co-expression of reporter genes or oncogenes from the β-actin promoter. We showed that knock-in of oncogenic Ras and loss of p53 efficiently induced intrahepatic cholangiocarcinoma in mice. Further, our strategy can generate bioluminescent liver cancer to facilitate tumor imaging. This method simplifies in vivo gain-of-function genetics by facilitating targeted integration of oncogenes.
Collapse
Affiliation(s)
- Haiwei Mou
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Deniz M Ozata
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Jordan L Smith
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Ankur Sheel
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Suet-Yan Kwan
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Soren Hough
- The Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Alper Kucukural
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Zachary Kennedy
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Yueying Cao
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Wen Xue
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
- Program in Molecular Medicine, Department of Molecular, Cell and Cancer Biology, and Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
135
|
Udden SN, Kwak YT, Godfrey V, Khan MAW, Khan S, Loof N, Peng L, Zhu H, Zaki H. NLRP12 suppresses hepatocellular carcinoma via downregulation of cJun N-terminal kinase activation in the hepatocyte. eLife 2019; 8:40396. [PMID: 30990169 PMCID: PMC6483596 DOI: 10.7554/elife.40396] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 03/25/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a deadly human cancer associated with chronic inflammation. The cytosolic pathogen sensor NLRP12 has emerged as a negative regulator of inflammation, but its role in HCC is unknown. Here we investigated the role of NLRP12 in HCC using mouse models of HCC induced by carcinogen diethylnitrosamine (DEN). Nlrp12-/- mice were highly susceptible to DEN-induced HCC with increased inflammation, hepatocyte proliferation, and tumor burden. Consistently, Nlrp12-/- tumors showed higher expression of proto-oncogenes cJun and cMyc and downregulation of tumor suppressor p21. Interestingly, antibiotics treatment dramatically diminished tumorigenesis in Nlrp12-/- mouse livers. Signaling analyses demonstrated higher JNK activation in Nlrp12-/- HCC and cultured hepatocytes during stimulation with microbial pattern molecules. JNK inhibition or NLRP12 overexpression reduced proliferative and inflammatory responses of Nlrp12-/- hepatocytes. In summary, NLRP12 negatively regulates HCC pathogenesis via downregulation of JNK-dependent inflammation and proliferation of hepatocytes.
Collapse
Affiliation(s)
- Sm Nashir Udden
- Department of Pathology, UT Southwestern Medical Center, Dallas, United States
| | - Youn-Tae Kwak
- Department of Pathology, UT Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, UT Southwestern Medical Center, Dallas, United States
| | - Victoria Godfrey
- Department of Pathology, UT Southwestern Medical Center, Dallas, United States
| | - Md Abdul Wadud Khan
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, United States
| | - Shahanshah Khan
- Department of Pathology, UT Southwestern Medical Center, Dallas, United States
| | - Nicolas Loof
- Children's Research Institute, UT Southwestern Medical Center, Dallas, United States
| | - Lan Peng
- Department of Pathology, UT Southwestern Medical Center, Dallas, United States
| | - Hao Zhu
- Children's Research Institute, UT Southwestern Medical Center, Dallas, United States.,Department of Pediatrics, UT Southwestern Medical Center, Dallas, United States.,Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, United States.,Department of Internal Medicine, UT Southwestern Medical Center, Dallas, United States
| | - Hasan Zaki
- Department of Pathology, UT Southwestern Medical Center, Dallas, United States
| |
Collapse
|
136
|
Manieri E, Herrera-Melle L, Mora A, Tomás-Loba A, Leiva-Vega L, Fernández DI, Rodríguez E, Morán L, Hernández-Cosido L, Torres JL, Seoane LM, Cubero FJ, Marcos M, Sabio G. Adiponectin accounts for gender differences in hepatocellular carcinoma incidence. J Exp Med 2019; 216:1108-1119. [PMID: 30944152 PMCID: PMC6504215 DOI: 10.1084/jem.20181288] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 01/11/2019] [Accepted: 02/08/2019] [Indexed: 12/24/2022] Open
Abstract
Gender disparity in liver cancer incidence is a relevant feature of this malignancy. Manieri et al. show that testosterone-induced JNK1 activation in adipose tissue results in decreased levels of circulating adiponectin, which is responsible for higher incidence of hepatocellular carcinoma in males. Hepatocellular carcinoma (HCC) is the sixth most common cancer type and the fourth leading cause of cancer-related death. This cancer appears with higher incidence in men and during obesity; however, the specific mechanisms underlying this correlation are unknown. Adipose tissue, a key organ in metabolic syndrome, shows evident gender disparities in the production of adipokines. Levels of the important adipokine adiponectin decrease in men during puberty, as well as in the obese state. Here, we show that this decrease in adiponectin levels is responsible for the increased liver cancer risk in males. We found that testosterone activates the protein JNK in mouse and human adipocytes. JNK-mediated inhibition of adiponectin secretion increases liver cancer cell proliferation, since adiponectin protects against liver cancer development through the activation of AMP-activated protein kinase (AMPK) and p38α. This study provides insight into adipose tissue to liver crosstalk and its gender relation during cancer development, having the potential to guide strategies for new cancer therapeutics.
Collapse
Affiliation(s)
- Elisa Manieri
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro Nacional de Biotecnología, Madrid, Spain
| | | | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Antonia Tomás-Loba
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Delia I Fernández
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Elena Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Laura Morán
- Department of Immunology, Ophthalmology and Otorhinolaryngology, Complutense University School of Medicine, Madrid, Spain.,12 de Octubre Health Research Institute, Madrid, Spain
| | - Lourdes Hernández-Cosido
- University of Salamanca, University Hospital of Salamanca-Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Jorge L Torres
- University of Salamanca, University Hospital of Salamanca-Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Luisa M Seoane
- Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago, Hospital Clínico Universitario de Santiago de Compostela Servicio Gallego de Salud, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red (CIBER), Fisiopatología Obesidad y Nutrición, Instituto Salud Carlos III, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and Otorhinolaryngology, Complutense University School of Medicine, Madrid, Spain.,12 de Octubre Health Research Institute, Madrid, Spain
| | - Miguel Marcos
- University of Salamanca, University Hospital of Salamanca-Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
137
|
Zhang J, Han C, Ungerleider N, Chen W, Song K, Wang Y, Kwon H, Ma W, Wu T. A Transforming Growth Factor-β and H19 Signaling Axis in Tumor-Initiating Hepatocytes That Regulates Hepatic Carcinogenesis. Hepatology 2019; 69:1549-1563. [PMID: 30014520 PMCID: PMC6335184 DOI: 10.1002/hep.30153] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 06/23/2018] [Indexed: 02/06/2023]
Abstract
Functions of transforming growth factor-β (TGF-β) in the liver vary depending on specific cell types and their temporal response to TGF-β during different stages of hepatocarcinogenesis (HCG). Through analysis of tumor tissues from hepatocellular carcinoma (HCC) patients, we were able to cluster hepatic epithelial cell-derived TGF-β gene signatures in association with distinct clinical prognoses. To delineate the role of hepatic epithelial TGF-β signaling in HCC development, we used an experimental system in which tumor-initiating hepatocytes (TICs) were isolated from TGF-β receptor II floxed mice (Tgfbr2fl/fl ) and transplanted into syngeneic C57BL/6J mice by splenic injection. Recipient mice were then administered Cre-expressing adenovirus (Ad-Cre) to inactivate Tgfbr2 in transplanted TICs. After latency, Tgfbr2-inactivated TICs formed larger and more tumor nodules in recipient livers compared to TICs without Tgfbr2 inactivation. In vitro analyses revealed that treatment of cultured TICs with TGF-β inhibited expression of progenitor cell factors (including SRY (sex determining region Y)-box 2 [Sox2]). RNA sequencing (RNA-seq) analysis identified H19 as one of the most up-regulated long noncoding RNA (lncRNA) in association with Tgfbr2 inactivation in TICs. Tgfbr2 inactivation by Ad-Cre led to a 5-fold increase of H19 expression in TICs. Accordingly, TGF-β treatment reduced H19 expression. We observed that forced overexpression of Sox2 in TICs increased transcription of H19, whereas knockdown of Sox2 decreased it. Furthermore, depletion of H19 reduced the progenitor property of TICs in vitro and decreased their tumorigenic potential in vivo. Finally, we observed a low level of H19 mRNA expression in human HCC tissues from patients with the epithelial TGF-β gene signature in association with favorable prognosis. Conclusion: Our findings describe a TGF-β and H19 signaling axis by Sox2 in TICs that importantly regulates HCG.
Collapse
Affiliation(s)
- Jinqiang Zhang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Chang Han
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Nathan Ungerleider
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Weina Chen
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Kyoungsub Song
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Ying Wang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Hyunjoo Kwon
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Wenbo Ma
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
138
|
Miethe C, Zamora M, Torres L, Raign KG, Groll CJ, Price RS. Characterizing the differential physiological effects of adipocytokines visfatin and resistin in liver cancer cells. Horm Mol Biol Clin Investig 2019; 38:/j/hmbci.ahead-of-print/hmbci-2018-0068/hmbci-2018-0068.xml. [PMID: 30917102 DOI: 10.1515/hmbci-2018-0068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/14/2019] [Indexed: 02/06/2023]
Abstract
Background Obesity, a major public health concern, increases the risk of developing liver cancer which is the leading cause of cancer-related deaths worldwide. Obesity is associated with increased adiposity and macrophage infiltration both of which promote secretion of adipokines and cytokines in the tumor microenvironment. Specifically, visfatin and resistin have been detected at higher levels in the serum of obese individuals and liver tumors. However, the contribution of these adipocytokines in the progression of liver cancer remains unclear. Materials and methods The objective of this study was to characterize the effects of visfatin and resistin on HepG2, SNU-449 and HuH7 liver cancer cells. Cells exposed to visfatin and resistin were analyzed for fatty acid synthase protein, and phosphorylation of Akt and ERK tumorigenic signaling pathways, cell viability, lipogenesis, reactive oxygen species (ROS), matrix metallopeptidase 9 (MMP-9) enzyme activity and invasion. Results HepG2, SNU-449, and HuH7 liver cancer cells treated with visfatin and resistin increased cell viability, invasion, FASN protein, and Akt and ERK phosphorylation. Visfatin and resistin selectively increased ROS production in HepG2 and SNU-449 cells while there was no statistical difference in HuH7 cells. Visfatin and resistin stimulated lipogenesis in HepG2 cells while visfatin increased lipogenesis in SNU-449 cells, and visfatin nor resistin had an effect on lipogenesis in HuH7 cells. Lastly, visfatin and resistin increased MMP-9 enzyme activity in HepG2 and HuH-7 cells but only visfatin increased MMP-9 activity in SNU-449 cells. Conclusions Future studies are needed to determine if inhibition of ERK and Akt suppresses the visfatin and resistin-induced invasive liver cancer phenotype.
Collapse
Affiliation(s)
- Candace Miethe
- Texas State University, Family and Consumer Sciences, Nutrition, San Marcos, TX, USA
| | - Megan Zamora
- Texas State University, Family and Consumer Sciences, Nutrition, San Marcos, TX, USA
| | - Linda Torres
- Texas State University, Family and Consumer Sciences, Nutrition, San Marcos, TX, USA
| | - Kelsie G Raign
- Texas State University, Family and Consumer Sciences, Nutrition, San Marcos, TX, USA
| | - Curissa J Groll
- Texas State University, Family and Consumer Sciences, Nutrition, San Marcos, TX, USA
| | - Ramona S Price
- Texas State University, Family and Consumer Sciences, Nutrition, San Marcos, TX, USA.,Ramona Salcedo Price, 601 University Dr, San Marcos, TX 78666, USA, Phone: +512-245-6202
| |
Collapse
|
139
|
Muto Y, Moroishi T, Ichihara K, Nishiyama M, Shimizu H, Eguchi H, Moriya K, Koike K, Mimori K, Mori M, Katayama Y, Nakayama KI. Disruption of FBXL5-mediated cellular iron homeostasis promotes liver carcinogenesis. J Exp Med 2019; 216:950-965. [PMID: 30877170 PMCID: PMC6446870 DOI: 10.1084/jem.20180900] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 01/15/2019] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular iron overload elicited by ablation of the iron-sensing ubiquitin ligase FBXL5 promotes liver carcinogenesis induced by exposure to a chemical carcinogen or hepatitis virus, suggesting that FBXL5 is a previously unrecognized oncosuppressor in liver carcinogenesis in mice. Hepatic iron overload is a risk factor for progression of hepatocellular carcinoma (HCC), although the molecular mechanisms underlying this association have remained unclear. We now show that the iron-sensing ubiquitin ligase FBXL5 is a previously unrecognized oncosuppressor in liver carcinogenesis in mice. Hepatocellular iron overload elicited by FBXL5 ablation gave rise to oxidative stress, tissue damage, inflammation, and compensatory proliferation of hepatocytes and to consequent promotion of liver carcinogenesis induced by exposure to a chemical carcinogen. The tumor-promoting outcome of FBXL5 deficiency in the liver was also found to be effective in a model of virus-induced HCC. FBXL5-deficient mice thus constitute the first genetically engineered mouse model of liver carcinogenesis promoted by iron overload. In addition, dysregulation of FBXL5-mediated cellular iron homeostasis was found to be associated with poor prognosis in human HCC, suggesting that FBXL5 plays a key role in defense against hepatocarcinogenesis.
Collapse
Affiliation(s)
- Yoshiharu Muto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Toshiro Moroishi
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Kazuya Ichihara
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Masaaki Nishiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hideyuki Shimizu
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kyoji Moriya
- Department of Infection Control and Prevention, The University of Tokyo Hospital, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University, Beppu Hospital, Beppu, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuta Katayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
140
|
Fekir K, Dubois-Pot-Schneider H, Désert R, Daniel Y, Glaise D, Rauch C, Morel F, Fromenty B, Musso O, Cabillic F, Corlu A. Retrodifferentiation of Human Tumor Hepatocytes to Stem Cells Leads to Metabolic Reprogramming and Chemoresistance. Cancer Res 2019; 79:1869-1883. [DOI: 10.1158/0008-5472.can-18-2110] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 01/04/2019] [Accepted: 02/26/2019] [Indexed: 11/16/2022]
|
141
|
Zhang Y, Li Y, Jiang W, Li Q, Lan Y. The clinical significance of microRNA-122 in predicting the prognosis of patients with hepatocellular carcinoma: A meta-analysis validated by the Cancer Genome Atlas dataset. Medicine (Baltimore) 2019; 98:e14810. [PMID: 30921182 PMCID: PMC6456118 DOI: 10.1097/md.0000000000014810] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Although the prognostic value of microRNA-122 (miR-122) for hepatocellular carcinoma (HCC) patients have been evaluated by numerous studies, the results of them were not completely consistent. The present study aims to comprehensively evaluate the predicting value of miR-122 on the prognosis of patients with HCC based on all eligible literatures. METHODS Numerous electronic databases (MEDLINE, Embase, Pubmed, Google Scholar, and China Biology Medicine disc) were applied to retrieve relevant studies. Overall survival (OS) and progression-free survival (PFS) were used as primary endpoints. All statistical analyses were performed by RevMan software version 5.3.5 and STATA software version 14.1. In addition, the results of this meta-analysis were validated by an independent dataset from the Cancer Genome Atlas (TCGA). RESULTS A total of 11 studies containing 1124 patients were included in this meta-analysis. The pooled results showed that low miR-122 expression in HCC tissues significantly associated with unfavorable OS (hazard ratio [HR] = 1.48, 95% confidence interval [CI] 1.22-1.80, P < .001) and PFS (HR = 1.54, 95% CI 1.28-1.85, P < .001) in patients with HCC. However, the expression level of miR-122 in blood did not have the ability in predicting OS (HR = 0.75, 95% CI 0.44-1.28, P = .29) and PFS (HR = 0.84, 95% CI 0.58-1.20, P = .33) of HCC. Subgroup analysis further indicated that low expression of miR-122 in tumor tissues predicted poor OS in HCC patients who received curative liver resection (HR = 2.00, 95% CI 1.08-3.70, P = .03). Analysis using TCGA dataset suggested that low miR-122 expression in HCC tissues was significantly associated with OS (HR = 1.61, 95% CI 1.13-2.27, P = .008) other than PFS (HR = 1.30, 95% CI 0.96-1.75, P = .09). CONCLUSION Low miR-122 expression in HCC tissues was a reliable indicator for predicting the OS of HCC patients who underwent curative resection. Owing to the disagreement between this meta-analysis and the TCGA dataset, the predictive value of miR-122 in tissues for PFS needs to be verified by future well-designed studies with large sample size.
Collapse
|
142
|
Ferretti AC, Hidalgo F, Tonucci FM, Almada E, Pariani A, Larocca MC, Favre C. Metformin and glucose starvation decrease the migratory ability of hepatocellular carcinoma cells: targeting AMPK activation to control migration. Sci Rep 2019; 9:2815. [PMID: 30809021 PMCID: PMC6391381 DOI: 10.1038/s41598-019-39556-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 01/17/2019] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly metastatic cancer with very poor prognosis. AMP activated kinase (AMPK) constitutes a candidate to inhibit HCC progression. First, AMPK is downregulated in HCC. Second, glucose starvation induces apoptosis in HCC cells via AMPK. Correspondingly, metformin activates AMPK and inhibits HCC cell proliferation. Nevertheless, the effect of AMPK activation on HCC cell invasiveness remains elusive. Here, migration/invasion was studied in HCC cells exposed to metformin and glucose starvation. Cell viability, proliferation and differentiation, as well as AMPK and PKA activation were analyzed. In addition, invasiveness in mutants of the AMPKα activation loop was assessed. Metformin decreased cell migration, invasion and epithelial-mesenchymal transition, and interference with AMPKα expression avoided metformin actions. Those antitumor effects were potentiated by glucose deprivation. Metformin activated AMPK at the same time that inhibited PKA, and both effects were enhanced by glucose starvation. Given that AMPKα(S173) phosphorylation by PKA decreases AMPK activation, we hypothesized that the reduction of PKA inhibitory effect by metformin could explain the increased antitumor effects observed. Supporting this, in AMPK activating conditions, cell migration/invasion was further impaired in AMPKα(S173C) mutant cells. Metformin emerges as a strong inhibitor of migration/invasion in HCC cells, and glucose restriction potentiates this effect.
Collapse
Affiliation(s)
- Anabela C Ferretti
- Institute of Experimental Physiology, CONICET, School of Biochemical Sciences, University of Rosario, Rosario, Argentina
| | - Florencia Hidalgo
- Institute of Experimental Physiology, CONICET, School of Biochemical Sciences, University of Rosario, Rosario, Argentina
| | - Facundo M Tonucci
- Institute of Experimental Physiology, CONICET, School of Biochemical Sciences, University of Rosario, Rosario, Argentina
| | - Evangelina Almada
- Institute of Experimental Physiology, CONICET, School of Biochemical Sciences, University of Rosario, Rosario, Argentina
| | - Alejandro Pariani
- Institute of Experimental Physiology, CONICET, School of Biochemical Sciences, University of Rosario, Rosario, Argentina
| | - María C Larocca
- Institute of Experimental Physiology, CONICET, School of Biochemical Sciences, University of Rosario, Rosario, Argentina
| | - Cristián Favre
- Institute of Experimental Physiology, CONICET, School of Biochemical Sciences, University of Rosario, Rosario, Argentina.
| |
Collapse
|
143
|
Dreval K, Tryndyak V, Kindrat I, Twaddle NC, Orisakwe OE, Mudalige TK, Beland FA, Doerge DR, Pogribny IP. Cellular and Molecular Effects of Prolonged Low-Level Sodium Arsenite Exposure on Human Hepatic HepaRG Cells. Toxicol Sci 2019; 162:676-687. [PMID: 29301061 DOI: 10.1093/toxsci/kfx290] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Inorganic arsenic is a human carcinogen associated with several types of cancers, including liver cancer. Inorganic arsenic has been postulated to target stem cells, causing their oncogenic transformation. This is proposed to be one of the key events in arsenic-associated carcinogenesis; however, the underlying mechanisms for this process remain largely unknown. To address this question, human hepatic HepaRG cells, at progenitor and differentiated states, were continuously treated with a noncytotoxic concentration of 1 μM sodium arsenite (NaAsO2). The HepaRG cells demonstrated active intracellular arsenite metabolism that shared important characteristic with primary human hepatocytes. Treatment of proliferating progenitor-like HepaRG cells with NaAsO2 inhibited their differentiation into mature hepatocyte-like cells, up-regulated genes involved in cell growth, proliferation, and survival, and down-regulated genes involved in cell death. In contrast, treatment of differentiated hepatocyte-like HepaRG cells with NaAsO2 resulted in enhanced cell death of mature hepatocyte-like cells, overexpression of cell death-related genes, and down-regulation of genes in the cell proliferation pathway, while biliary-like cells remained largely unaffected. Mechanistically, the cytotoxic effect of arsenic on mature hepatocyte-like HepaRG cells may be attributed to arsenic-induced dysregulation of cellular iron metabolism. The inhibitory effect of NaAsO2 on the differentiation of progenitor cells, the resistance of biliary-like cells to cell death, and the enhanced cell death of functional hepatocyte-like cells resulted in stem-cell activation. These effects favored the proliferation of liver progenitor cells that can serve as a source of initiation and driving force of arsenic-mediated liver carcinogenesis.
Collapse
Affiliation(s)
- Kostiantyn Dreval
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Volodymyr Tryndyak
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Iryna Kindrat
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079.,Department of Biological and Medical Chemistry, Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine
| | - Nathan C Twaddle
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Orish Ebere Orisakwe
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079.,Department of Experimental Pharmacology and Toxicology, University of Port-Harcourt, Rivers State, Nigeria
| | - Thilak K Mudalige
- Office of Regulatory Affairs, Arkansas Regional Laboratory, U.S. Food and Drug Administration, Jefferson, Arkansas 72079
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Daniel R Doerge
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Igor P Pogribny
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, Arkansas 72079
| |
Collapse
|
144
|
Xu L, Feng X, Hao X, Wang P, Zhang Y, Zheng X, Li L, Ren S, Zhang M, Xu M. CircSETD3 (Hsa_circ_0000567) acts as a sponge for microRNA-421 inhibiting hepatocellular carcinoma growth. J Exp Clin Cancer Res 2019; 38:98. [PMID: 30795787 PMCID: PMC6385474 DOI: 10.1186/s13046-019-1041-2] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/15/2019] [Indexed: 02/05/2023] Open
Abstract
Background Circular RNAs (circRNAs) play important roles in tumourigenesis and tumour progression. However, the expression profiles and functions of circRNAs in hepatocellular carcinoma (HCC) are largely unclear. Methods The expression profiles of circRNAs in HCC were identified through microarray analysis and were validated through quantitative reverse transcription polymerase chain reaction (qRT-PCR). Survival curves were plotted using the Kaplan-Meier method and compared using the log-rank test. The circular structure of candidate circRNA was confirmed through Sanger sequencing, divergent primer PCR, and RNase R treatments. Proliferation of HCC cells was evaluated in vitro and in vivo. The microRNA (miRNA) sponge mechanism of circRNAs was demonstrated using dual-luciferase reporter and RNA immunoprecipitation assays. Results CircSETD3 (hsa_circRNA_0000567/hsa_circRNA_101436) was significantly downregulated in HCC tissues and cell lines. Low expression of circSETD3 in HCC tissues significantly predicted an unfavourable prognosis and was correlated with larger tumour size and poor differentiation of HCC in patients. In vitro experiments showed that circSETD3 inhibited the proliferation of HCC cells and induced G1/S arrest in HCC cells. In vivo studies revealed that circSETD3 was stably overexpressed in a xenograft mouse model and inhibited the growth of HCC. Furthermore, we demonstrated that circSETD3 acts as a sponge for miR-421 and verified that mitogen-activated protein kinase (MAPK)14 is a novel target of miR-421. Conclusion CircSETD3 is a novel tumour suppressor of HCC and is a valuable prognostic biomarker. Moreover, circSETD3 inhibits the growth of HCC partly through the circSETD3/miR-421/MAPK14 pathway. Electronic supplementary material The online version of this article (10.1186/s13046-019-1041-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liangliang Xu
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, China
| | - Xinfu Feng
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, China.,Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, 550000, Guizhou Province, China
| | - Xiangyong Hao
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, China.,Department of General Surgery, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China
| | - Peng Wang
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, China
| | - Yanfang Zhang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, China
| | - Xiaobo Zheng
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, China
| | - Lian Li
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, China
| | - Shengsheng Ren
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, China
| | - Ming Zhang
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, China. .,Department of General surgery, Mianzhu hospital of West China hospital, Sichuan University, Mianzhu City, Sichuan Province, China.
| | - Mingqing Xu
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan Province, Chengdu, 610041, China.
| |
Collapse
|
145
|
Remodelling and Improvements in Organoid Technology to Study Liver Carcinogenesis in a Dish. Stem Cells Int 2019; 2019:3831213. [PMID: 30915124 PMCID: PMC6399527 DOI: 10.1155/2019/3831213] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/24/2019] [Indexed: 02/07/2023] Open
Abstract
Primary liver cancer (PLC) is the sixth most common tumour disease and one of the leading causes of cancer-related death worldwide. The two most common types of PLC are hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA). Diverse subgroups are described and a manifold number of gene mutations are known. Asymptomatic disease progression and limited therapeutic options are the reasons for the high mortality rate in PLC. Up to date, the multikinase inhibitors sorafenib and lenvatinib are the only FDA-approved first-line treatments for advanced HCC. One of the major drawbacks in the preclinical drug development is the lack of suitable model systems. In recent years, 3D organoid cultures were established from several organs and tumour subtypes, thereby opening new avenues in tumour research. 3D organoid cultures are used to describe the tumour diversity, for cancer modelling in a dish and for therapy responsiveness. The establishment of living biobanks and the development of next-generation matrices are promising approaches to overcome drug resistance and to improve the quality of personalised anticancer strategies for patients with PLC. In this review, we summarise the current knowledge of 3D cultures generated from healthy liver and primary liver cancer.
Collapse
|
146
|
Lee JH, Suh JH, Choi SY, Kang HJ, Lee HH, Ye BJ, Lee GR, Jung SW, Kim CJ, Lee-Kwon W, Park J, Myung K, Park NH, Kwon HM. Tonicity-responsive enhancer-binding protein promotes hepatocellular carcinogenesis, recurrence and metastasis. Gut 2019; 68:347-358. [PMID: 29420225 PMCID: PMC6352413 DOI: 10.1136/gutjnl-2017-315348] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Hepatocellular carcinoma (HCC) is a common cancer with high rate of recurrence and mortality. Diverse aetiological agents and wide heterogeneity in individual tumours impede effective and personalised treatment. Tonicity-responsive enhancer-binding protein (TonEBP) is a transcriptional cofactor for the expression of proinflammatory genes. Although inflammation is intimately associated with the pathogenesis of HCC, the role of TonEBP is unknown. We aimed to identify function of TonEBP in HCC. DESIGN Tumours with surrounding hepatic tissues were obtained from 296 patients with HCC who received completion resection. TonEBP expression was analysed by quantitative reverse transcription-quantitative real-time PCR (RT-PCR) and immunohfistochemical analyses of tissue microarrays. Mice with TonEBP haplodeficiency, and hepatocyte-specific and myeloid-specific TonEBP deletion were used along with HCC and hepatocyte cell lines. RESULTS TonEBP expression is higher in tumours than in adjacent non-tumour tissues in 92.6% of patients with HCC regardless of aetiology associated. The TonEBP expression in tumours and adjacent non-tumour tissues predicts recurrence, metastasis and death in multivariate analyses. TonEBP drives the expression of cyclo-oxygenase-2 (COX-2) by stimulating the promoter. In mouse models of HCC, three common sites of TonEBP action in response to diverse aetiological agents leading to tumourigenesis and tumour growth were found: cell injury and inflammation, induction by oxidative stress and stimulation of the COX-2 promoter. CONCLUSIONS TonEBP is a key component of the common pathway in tumourigenesis and tumour progression of HCC in response to diverse aetiological insults. TonEBP is involved in multiple steps along the pathway, rendering it an attractive therapeutic target as well as a prognostic biomarker.
Collapse
Affiliation(s)
- Jun Ho Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jae Hee Suh
- Department of Pathology, University of Ulsan College of Medicine, Ulsan University Hospital, Ulsan, Republic of Korea
| | - Soo Youn Choi
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Hyun Je Kang
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Hwan Hee Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Byeong Jin Ye
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Gap Ryol Lee
- Department of Life Science, Sogang University, Seoul, Republic of Korea
| | - Seok Won Jung
- Department of Internal Medicine, University of Ulsan College of Medicine, Ulsan University Hospital, Ulsan, Republic of Korea
| | - Chang Jae Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Whaseon Lee-Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jiyoung Park
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Kyungjae Myung
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea,Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| | - Neung Hwa Park
- Department of Internal Medicine, University of Ulsan College of Medicine, Ulsan University Hospital, Ulsan, Republic of Korea
| | - Hyug Moo Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea,Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| |
Collapse
|
147
|
Li Y, Zhang X, Yang X, Liu J, Li L, Ma W, Chen M. Differential effects of ginkgol C17:1 on cisplatin-induced cytotoxicity: Protecting human normal L02 hepatocytes versus sensitizing human hepatoma HepG2 cells. Oncol Lett 2019; 17:3181-3190. [PMID: 30867748 PMCID: PMC6396107 DOI: 10.3892/ol.2019.9974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022] Open
Abstract
Liver cancer is a major healthcare problem and one of the leading causes of cancer-associated mortality in the world. To date, chemotherapy remains a common method for treating cancer and cisplatin is one of the most widely used chemotherapeutics. However, owing to drug resistance and side effects, it is imperative to identify a novel approach to improve the anticancer effect of cisplatin. Auxiliary chemotherapy drugs with minor toxicity to normal cells may represent a novel strategy for cancer therapy. Previous studies have indicated that ginkgol C17:1 exhibits anticancer effects in liver cancer cells in vitro and in vivo. The antitumor activity of ginkgol C17:1 has been reported in combination with cisplatin in human liver cancer cells. Owing to the route of systemic administration, liver cancer cells and normal hepatocytes were exposed to chemotherapeutics and auxiliary chemotherapy drugs. However, the effects of ginkgol C17:1 in normal hepatocytes remain unclear. In the present study, the biological effects of ginkgol C17:1 alone and as co-treatment with cisplatin were compared in human hepatoma cells and normal hepatocytes. Consistently, the results confirmed that in human hepatoma HepG2 cells, ginkgol C17:1 or cisplatin alone induced autophagy and apoptosis. The co-treatment increased cisplatin-induced apoptosis and inhibited cisplatin-induced autophagy. In comparison, the treatments in human normal L02 hepatocytes indicated that ginkgol C17:1 alone induced autophagy, whereas cisplatin alone induced apoptosis. The co-treatment inhibited cisplatin-induced apoptosis, but enhanced autophagy in L02 cells. Further investigation revealed that the AMP-activated protein kinase/serine/threonine protein kinase ULK1 and phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin signaling pathways were involved in the underlying regulatory mechanisms. Taken together, the results of the present study provide the first evidence that ginkgol C17:1 protects normal hepatocytes against cisplatin-induced cytotoxicity while potentiating the anticancer effect of cisplatin chemotherapy. The differential effects on normal and cancer cells suggest that ginkgol C17:1 is a promising candidate for auxiliary chemotherapy.
Collapse
Affiliation(s)
- Yueying Li
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xinchi Zhang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiaoming Yang
- Department of Food Science and Engineering, School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jun Liu
- Department of Biology, Institute of Life Science, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Linjie Li
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenbin Ma
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Min Chen
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
148
|
Qiu L, Huang Y, Li Z, Dong X, Chen G, Xu H, Zeng Y, Cai Z, Liu X, Liu J. Circular RNA profiling identifies circADAMTS13 as a miR-484 sponge which suppresses cell proliferation in hepatocellular carcinoma. Mol Oncol 2019; 13:441-455. [PMID: 30537115 PMCID: PMC6360375 DOI: 10.1002/1878-0261.12424] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 11/11/2018] [Accepted: 11/22/2018] [Indexed: 12/31/2022] Open
Abstract
Circular RNA (circRNA) can participate in various biological processes, including tumorigenesis, through their microRNA response elements. Alterations in circRNA profiles during hepatocellular carcinoma (HCC) progression and their clinical significance remain unclear. Here, we present extensive analysis of circRNA profiles in tumor and matched peritumor tissues collected from 10 HCC patients, conducted to identify circRNA related to HCC progression. A total of 42 dysregulated circRNA (38 down‐regulated and 4 up‐regulated) were identified in HCC tumor tissues compared with matched peritumor tissues, revealing the heterogeneity of circRNA profiles in HCC. CircADAMTS13, derived from Exon 13–14 of the ADAMTS13 gene, was significantly downregulated in HCC tumor tissues. Furthermore, clinicopathological analysis revealed that up‐regulation of circADAMTS13 was negatively associated with tumor size but positively associated with prognosis. In addition, overexpression of circADAMTS13 could markedly inhibit HCC cell proliferation in vitro. Bioinformatic analysis and luciferase reporter assays further revealed that circADAMTS13 directly interacts with microRNA (miR)‐484. Rescue experiments showed that miR‐484 mimics can reverse the tumor‐suppressing roles of circADAMTS13 in HCC. Therefore, our results demonstrated that circADAMTS13 can serve as a tumor suppressor during HCC progression via the functional pathway of sponging miR‐484.
Collapse
Affiliation(s)
- Liman Qiu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Yanbing Huang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China.,Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhenli Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Xiuqing Dong
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Geng Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China.,School of Life Science and Technology, Xi'an Jiaotong University, China
| | - Haipo Xu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China.,Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China.,School of Life Science and Technology, Xi'an Jiaotong University, China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China.,School of Life Science and Technology, Xi'an Jiaotong University, China
| | - Jingfeng Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, China.,Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
149
|
Molecular Subtypes and Genomic Signatures of Hepatocellular Carcinoma for Prognostication and Therapeutic Decision-Making. MOLECULAR AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/978-3-030-21540-8_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
150
|
Yamamoto M, Xin B, Nishikawa Y. Mouse Model for Hepatocellular Carcinoma and Cholangiocarcinoma Originated from Mature Hepatocytes. Methods Mol Biol 2019; 1905:221-236. [PMID: 30536104 DOI: 10.1007/978-1-4939-8961-4_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Liver cancer consists of two main histological subtypes, hepatocellular carcinoma and cholangiocarcinoma, both of which have poor prognosis. Therefore, in searching for new therapeutic targets, adequate mouse models to develop and validate therapeutic strategies are urgently needed. Although there are mouse models of liver cancer, each model has shortcomings. To overcome these shortcomings, a mouse model using a hydrodynamic tail vein injection and the Sleeping Beauty transposon was developed. By inducing stable expression of oncogenes in mouse hepatocytes in vivo, the model can easily induce liver cancer with specific characteristics that depend on the oncogenes used to induce carcinogenesis. Here, we describe the details of the methods to induce hepatocellular carcinoma or cholangiocarcinoma from mouse hepatocytes.
Collapse
Affiliation(s)
- Masahiro Yamamoto
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan.
| | - Bing Xin
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| |
Collapse
|