1301
|
E2F-Family Members Engage the PIDDosome to Limit Hepatocyte Ploidy in Liver Development and Regeneration. Dev Cell 2020; 52:335-349.e7. [PMID: 31983631 DOI: 10.1016/j.devcel.2019.12.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 10/27/2019] [Accepted: 12/24/2019] [Indexed: 01/12/2023]
Abstract
E2F transcription factors control the cytokinesis machinery and thereby ploidy in hepatocytes. If or how these proteins limit proliferation of polyploid cells with extra centrosomes remains unknown. Here, we show that the PIDDosome, a signaling platform essential for caspase-2-activation, limits hepatocyte ploidy and is instructed by the E2F network to control p53 in the developing as well as regenerating liver. Casp2 and Pidd1 act as direct transcriptional targets of E2F1 and its antagonists, E2F7 and E2F8, that together co-regulate PIDDosome expression during juvenile liver growth and regeneration. Of note, whereas hepatocyte aneuploidy correlates with the basal ploidy state, the degree of aneuploidy itself is not limited by PIDDosome-dependent p53 activation. Finally, we provide evidence that the same signaling network is engaged to control ploidy in the human liver after resection. Our study defines the PIDDosome as a primary target to manipulate hepatocyte ploidy and proliferation rates in the regenerating liver.
Collapse
|
1302
|
Study on the anti-tumor mechanism related to immune microenvironment of Bombyx Batryticatus on viral and non-viral infections of hepatocellular carcinoma. Biomed Pharmacother 2020; 124:109838. [PMID: 31981943 DOI: 10.1016/j.biopha.2020.109838] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/24/2019] [Accepted: 12/31/2019] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant primary liver cancer with poor prognosis. Most previous studies on anti-HCC effects of traditional Chinese medicines (TCM) have focused on the mechanism of direct action and few researchers considered that TCM can inhibit tumor progression and improve prognosis of HCC patients through regulating tumor microenvironment (TME). In this study, network pharmacology combined bioinformatics methods were employed to analysis mechanism of Bombyx batryticatus (B. batryticatus, one of the most frequently used traditional Chinese animal medicines, has been used in some Asian countries for centuries as an anticancer agent, anti-inflammatory agent, and antioxidant.) in regulating TME of HCC. The results showed that 24 core targets and 2 compounds were identified from overlapping between differential expression genes related to HCC in the cancer genome atlas (TCGA) database and targets of B. batryticatus in TCMSP database. For further analyzing the role of TME heterogeneity of HCC on anti-HCC mechanism of B. batryticatus, the correlation of core targets related with overall survival of HCC with TME cells in hepatitis C or hepatitis B virus-associated hepatocellular carcinoma (VIR) and non-hepatitis C or hepatitis B virus-associated hepatocellular carcinoma (NVIR) were calculated, respectively. The results showed that AKR1C3, SPP1 were significantly related with macrophages in VIR and other targets including NR1I2, CYP1A2 and CYP3A4 were significantly associated with macrophages in NVIR; the target protein AKR1C3 was significantly negative correlated with macrophages M1 in VIR (cor=-0.35, P-value<0.001) and the correlation between AKR1C3 and macrophages M1 was poor in NVIR group (cor = 0.064, P-value = 0.36). Additionally, survival curve of AKR1C3 showed that poor prognosis in VIR group can be related to high level of AKR1C3 (HR = 2.32, 95 % CI: 1.18-4.56, P-value = 0.012), and no signified gene can be found in NVIR group (P-value>0.05). In conclusion, the molecular mechanism of anti-HCC of B. batryticatus can be related to the tumor microenvironment to some extent. B. batryticatus may exert its anti-cancer effects and improve prognosis of patients by regulating macrophages M1 in VIR and NVIR through acting on different targets.
Collapse
|
1303
|
Rashed WM, Kandeil MAM, Mahmoud MO, Ezzat S. Hepatocellular Carcinoma (HCC) in Egypt: A comprehensive overview. J Egypt Natl Canc Inst 2020; 32:5. [PMID: 32372179 DOI: 10.1186/s43046-020-0016-x] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Worldwide, hepatocellular carcinoma (HCC) is a universal problem and its epidemiological data showed variation from place to place. Hepatocellular carcinoma (HCC) is the sixth and fourth common cancer in worldwide and Egypt, respectively. Egypt ranks the third and 15th most populous country in Africa and worldwide, respectively. The aim of this review is to compare the status of HCC in Egypt to that in the worldwide from different issues; risk factors, screening and surveillance, diagnosis and treatment, prevention, as well as research strategy. MAIN BODY The risk factors for HCC in Egypt are of great importance to be reported. The risk factor for HCC are either environmental- or host/genetic-related risk factors. In the last years, there is a tangible improvement of both screening and surveillance strategies of HCC in Egypt. The unprecedented national screening campaign launched by the end of 2018 is a mirror image of this improvement. While the improvement of the HCC prevention requires the governmental health administration to implement health policies. Although the diagnosis of Egyptian HCC patients follows the international guidelines but HCC treatment options are limited in terms of cost. In addition, there are limited Egyptian reports about HCC survival and relapse. Both basic and clinical HCC research in Egypt are still limited compared to worldwide. SHORT CONCLUSION Deep analysis and understanding of factors affecting HCC burden variation worldwide help in customization of efforts exerted to face HCC in different countries especially large country like Egypt. Overall, the presence of a research strategy to fight HCC in Egyptian patients will help in the optimum allocation of available resources to reduce the numbers of HCC cases and deaths and to improve the quality of life.
Collapse
Affiliation(s)
- Wafaa M Rashed
- Department of Research, Children's Cancer Hospital-57357, Cairo, Egypt.
| | | | - Mohamed O Mahmoud
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Sameera Ezzat
- Department of Epidemiology and Prevention Medicine, National Liver Institute, Menoufia University, Menoufia, Egypt
| |
Collapse
|
1304
|
Losic B, Craig AJ, Villacorta-Martin C, Martins-Filho SN, Akers N, Chen X, Ahsen ME, von Felden J, Labgaa I, DʹAvola D, Allette K, Lira SA, Furtado GC, Garcia-Lezana T, Restrepo P, Stueck A, Ward SC, Fiel MI, Hiotis SP, Gunasekaran G, Sia D, Schadt EE, Sebra R, Schwartz M, Llovet JM, Thung S, Stolovitzky G, Villanueva A. Intratumoral heterogeneity and clonal evolution in liver cancer. Nat Commun 2020; 11:291. [PMID: 31941899 PMCID: PMC6962317 DOI: 10.1038/s41467-019-14050-z] [Citation(s) in RCA: 252] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 12/13/2019] [Indexed: 02/08/2023] Open
Abstract
Clonal evolution of a tumor ecosystem depends on different selection pressures that are principally immune and treatment mediated. We integrate RNA-seq, DNA sequencing, TCR-seq and SNP array data across multiple regions of liver cancer specimens to map spatio-temporal interactions between cancer and immune cells. We investigate how these interactions reflect intra-tumor heterogeneity (ITH) by correlating regional neo-epitope and viral antigen burden with the regional adaptive immune response. Regional expression of passenger mutations dominantly recruits adaptive responses as opposed to hepatitis B virus and cancer-testis antigens. We detect different clonal expansion of the adaptive immune system in distant regions of the same tumor. An ITH-based gene signature improves single-biopsy patient survival predictions and an expression survey of 38,553 single cells across 7 regions of 2 patients further reveals heterogeneity in liver cancer. These data quantify transcriptomic ITH and how the different components of the HCC ecosystem interact during cancer evolution. Immune-mediated selection pressures impact the clonal evolution of tumours. Here, in hepatocellular carcinoma the authors map spatio-temporal interactions between tumor and immune cells, highlighting the regulatory substrate of intra-tumoural heterogeneity that correlates with regional adaptive immune responses.
Collapse
Affiliation(s)
- Bojan Losic
- Department of Genetics and Genomic Sciences, Cancer Immunology Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda J Craig
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carlos Villacorta-Martin
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sebastiao N Martins-Filho
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Pathology, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Nicholas Akers
- Department of Genetics and Genomic Sciences, Cancer Immunology Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Adaptive Biotechnologies, Seattle, WA, USA
| | - Xintong Chen
- Department of Genetics and Genomic Sciences, Cancer Immunology Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mehmet E Ahsen
- Department of Genetics and Genomic Sciences, Cancer Immunology Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Johann von Felden
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ismail Labgaa
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Visceral Surgery, Lausanne University Hospital CHUV, Lausanne, Switzerland
| | - Delia DʹAvola
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Liver Unit and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Clínica Universidad de Navarra, Pamplona, Spain
| | - Kimaada Allette
- Department of Genetics and Genomic Sciences, Cancer Immunology Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sergio A Lira
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Glaucia C Furtado
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Teresa Garcia-Lezana
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paula Restrepo
- Department of Genetics and Genomic Sciences, Cancer Immunology Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashley Stueck
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Stephen C Ward
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria I Fiel
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Spiros P Hiotis
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ganesh Gunasekaran
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniela Sia
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric E Schadt
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Sema4, a Mount Sinai venture, Stamford, CT, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Cancer Immunology Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Sema4, a Mount Sinai venture, Stamford, CT, USA
| | - Myron Schwartz
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josep M Llovet
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Liver Cancer Translational Research Laboratory, BCLC Group, IDIBAPS, Hospital Clinic, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Catalonia, Spain
| | - Swan Thung
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gustavo Stolovitzky
- Department of Genetics and Genomic Sciences, Cancer Immunology Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,IBM T. J. Watson Research Center, Yorktown Heights, New York, NY, USA
| | - Augusto Villanueva
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
1305
|
Franch-Expósito S, Bassaganyas L, Vila-Casadesús M, Hernández-Illán E, Esteban-Fabró R, Díaz-Gay M, Lozano JJ, Castells A, Llovet JM, Castellví-Bel S, Camps J. CNApp, a tool for the quantification of copy number alterations and integrative analysis revealing clinical implications. eLife 2020; 9:e50267. [PMID: 31939734 PMCID: PMC7010409 DOI: 10.7554/elife.50267] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/14/2020] [Indexed: 12/18/2022] Open
Abstract
Somatic copy number alterations (CNAs) are a hallmark of cancer, but their role in tumorigenesis and clinical relevance remain largely unclear. Here, we developed CNApp, a web-based tool that allows a comprehensive exploration of CNAs by using purity-corrected segmented data from multiple genomic platforms. CNApp generates genome-wide profiles, computes CNA scores for broad, focal and global CNA burdens, and uses machine learning-based predictions to classify samples. We applied CNApp to the TCGA pan-cancer dataset of 10,635 genomes showing that CNAs classify cancer types according to their tissue-of-origin, and that each cancer type shows specific ranges of broad and focal CNA scores. Moreover, CNApp reproduces recurrent CNAs in hepatocellular carcinoma and predicts colon cancer molecular subtypes and microsatellite instability based on broad CNA scores and discrete genomic imbalances. In summary, CNApp facilitates CNA-driven research by providing a unique framework to identify relevant clinical implications. CNApp is hosted at https://tools.idibaps.org/CNApp/.
Collapse
Affiliation(s)
- Sebastià Franch-Expósito
- Gastrointestinal and Pancreatic Oncology TeamInstitut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Universitat de BarcelonaBarcelonaSpain
| | - Laia Bassaganyas
- Liver Cancer Translational Research Group, Liver UnitInstitut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Universitat de BarcelonaBarcelonaSpain
| | | | - Eva Hernández-Illán
- Gastrointestinal and Pancreatic Oncology TeamInstitut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Universitat de BarcelonaBarcelonaSpain
| | - Roger Esteban-Fabró
- Liver Cancer Translational Research Group, Liver UnitInstitut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Universitat de BarcelonaBarcelonaSpain
| | - Marcos Díaz-Gay
- Gastrointestinal and Pancreatic Oncology TeamInstitut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Universitat de BarcelonaBarcelonaSpain
| | | | - Antoni Castells
- Gastrointestinal and Pancreatic Oncology TeamInstitut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Universitat de BarcelonaBarcelonaSpain
| | - Josep Maria Llovet
- Liver Cancer Translational Research Group, Liver UnitInstitut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Universitat de BarcelonaBarcelonaSpain
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkUnited States
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
| | - Sergi Castellví-Bel
- Gastrointestinal and Pancreatic Oncology TeamInstitut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Universitat de BarcelonaBarcelonaSpain
| | - Jordi Camps
- Gastrointestinal and Pancreatic Oncology TeamInstitut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Universitat de BarcelonaBarcelonaSpain
- Unitat de Biologia Cel·lular i Genètica Mèdica, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de MedicinaUniversitat Autònoma de BarcelonaBellaterraSpain
| |
Collapse
|
1306
|
Folschette M, Legagneux V, Poret A, Chebouba L, Guziolowski C, Théret N. A pipeline to create predictive functional networks: application to the tumor progression of hepatocellular carcinoma. BMC Bioinformatics 2020; 21:18. [PMID: 31937236 PMCID: PMC6958715 DOI: 10.1186/s12859-019-3316-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Integrating genome-wide gene expression patient profiles with regulatory knowledge is a challenging task because of the inherent heterogeneity, noise and incompleteness of biological data. From the computational side, several solvers for logic programs are able to perform extremely well in decision problems for combinatorial search domains. The challenge then is how to process the biological knowledge in order to feed these solvers to gain insights in a biological study. It requires formalizing the biological knowledge to give a precise interpretation of this information; currently, very few pathway databases offer this possibility. RESULTS The presented work proposes an automatic pipeline to extract automatically regulatory knowledge from pathway databases and generate novel computational predictions related to the state of expression or activity of biological molecules. We applied it in the context of hepatocellular carcinoma (HCC) progression, and evaluate the precision and the stability of these computational predictions. Our working base is a graph of 3383 nodes and 13,771 edges extracted from the KEGG database, in which we integrate 209 differentially expressed genes between low and high aggressive HCC across 294 patients. Our computational model predicts the shifts of expression of 146 initially non-observed biological components. Our predictions were validated at 88% using a larger experimental dataset and cross-validation techniques. In particular, we focus on the protein complexes predictions and show for the first time that NFKB1/BCL-3 complexes are activated in aggressive HCC. In spite of the large dimension of the reconstructed models, our analyses over the computational predictions discover a well constrained region where KEGG regulatory knowledge constrains gene expression of several biomolecules. These regions can offer interesting windows to perturb experimentally such complex systems. CONCLUSION This new pipeline allows biologists to develop their own predictive models based on a list of genes. It facilitates the identification of new regulatory biomolecules using knowledge graphs and predictive computational methods. Our workflow is implemented in an automatic python pipeline which is publicly available at https://github.com/LokmaneChebouba/key-pipeand contains as testing data all the data used in this paper.
Collapse
Affiliation(s)
- Maxime Folschette
- Univ Rennes, Inria, CNRS, IRISA, UMR 6074, Rennes, France
- Univ Rennes, Inserm, EHESP, Irset, UMR S1085, Rennes, France
- IFB-CORE, Institut Français de Bioinformatique, UMS CNRS 3601, Évry, France
- LS2N, Laboratoire des Sciences du Numérique de Nantes, UMR 6004, Nantes, France
- Univ. Lille, CNRS, Centrale Lille, CRIStAL, Centre de Recherche en Informatique Signal et Automatique de Lille, UMR 9189, F-59000, Lille, France
| | | | - Arnaud Poret
- LS2N, Laboratoire des Sciences du Numérique de Nantes, UMR 6004, Nantes, France
| | - Lokmane Chebouba
- LS2N, Laboratoire des Sciences du Numérique de Nantes, UMR 6004, Nantes, France
- École centrale de Nantes, Nantes, France
- Department of Computer Science, LRIA Laboratory, Electrical Engineering and Computer Science Faculty, University of Science and Technology Houari Boumediene (USTHB), Algiers, Algeria
| | - Carito Guziolowski
- LS2N, Laboratoire des Sciences du Numérique de Nantes, UMR 6004, Nantes, France.
- École centrale de Nantes, Nantes, France.
| | - Nathalie Théret
- Univ Rennes, Inria, CNRS, IRISA, UMR 6074, Rennes, France.
- Univ Rennes, Inserm, EHESP, Irset, UMR S1085, Rennes, France.
| |
Collapse
|
1307
|
Li W, Lu J, Ma Z, Zhao J, Liu J. An Integrated Model Based on a Six-Gene Signature Predicts Overall Survival in Patients With Hepatocellular Carcinoma. Front Genet 2020; 10:1323. [PMID: 32010188 PMCID: PMC6971204 DOI: 10.3389/fgene.2019.01323] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/05/2019] [Indexed: 02/05/2023] Open
Abstract
Background: Nowadays, clinical treatment outcomes of patients with hepatocellular carcinoma (HCC) have been improved. However, due to the complexity of the molecular mechanisms, the recurrence rate and mortality in HCC inpatients are still at a high level. Therefore, there is an urgent need in screening biomarkers of HCC to show therapeutic effects and improve the prognosis. Methods: In this study, we aim to establish a gene signature that can predict the prognosis of HCC patients by downloading and analyzing RNA sequencing data and clinical information from three independent public databases. Firstly, we applied the limma R package to analyze biomarkers by the genetic data and clinical information downloaded from the Gene Expression Omnibus database (GEO), and then used the least absolute shrinkage and selection operator (LASSO) Cox regression and survival analysis to establish a gene signature and a prediction model by data from the Cancer Genome Atlas (TCGA). Besides, messenger RNA (mRNA) and protein expressions of the six-gene signature were explored using Oncomine, Human Protein Atlas (HPA) and the International Cancer Genome Consortium (ICGC). Results: A total of 8,306 differentially expressed genes (DEGs) were obtained between HCC (n = 115) and normal tissues (n = 52). Top 5,000 significant genes were selected and subjected to the weighted correlation network analysis (WGCNA), which constructed nine gene co-expression modules that assign these genes to different modules by cluster dendrogram trees. By analyzing the most significant module (red module), six genes (SQSTM1, AHSA1, VNN2, SMG5, SRXN1, and GLS) were screened by univariate, LASSO, and multivariate Cox regression analysis. By a survival analysis with the HCC data in TCGA, we established a nomogram based on the six-gene signature and multiple clinicopathological features. The six-gene signature was then validated as an independent prognostic factor in independent HCC cohort from ICGC. Receiver operating characteristic (ROC) curve analysis confirmed the predictive capacity of the six-gene signature and nomogram. Besides, overexpression of the six genes at the mRNA and protein levels was validated using Oncomine and HPA, respectively. Conclusion: The predictive six-gene signature and nomograms established in this study can assist clinicians in selecting personalized treatment for patients with HCC.
Collapse
Affiliation(s)
- Wenli Li
- Department of Clinical Laboratory, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
- Department of Reproductive Medicine Center, The Affiliated Yue Bei People's Hospital of Shantou University Medical College, Shaoguan, China
| | - Jianjun Lu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Medical Services, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Morning Star Academic Cooperation, Shanghai, China
| | - Zhanzhong Ma
- Department of Clinical Laboratory, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
| | - Jiafeng Zhao
- Department of Hepatobiliary Surgery, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
| | - Jun Liu
- Department of Clinical Laboratory, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, China
- Morning Star Academic Cooperation, Shanghai, China
| |
Collapse
|
1308
|
Yamamoto S, Midorikawa Y, Nagae G, Tatsuno K, Ueda H, Moriyama M, Takayama T, Aburatani H. Spatial and temporal expansion of intrahepatic metastasis by molecularly-defined clonality in multiple liver cancers. Cancer Sci 2020; 111:601-609. [PMID: 31845427 PMCID: PMC7004543 DOI: 10.1111/cas.14282] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/17/2022] Open
Abstract
Multiple hepatocellular carcinoma (HCC) is divided into two categories: intrahepatic metastasis (IM), which is a true relapse of HCC, and multicentric origin (MO), which is a second primary tumor. Clinical diagnosis of multiple HCC is usually made based on tumor location and/or time to recurrence; however, it is often difficult to distinguish the two types of multiple HCC. Using 41 matched pairs of multiple HCC specimens, we confirmed the accuracy of clinical diagnoses using exome sequence data and investigated the importance of discriminating the type of multiple HCC. Genomic analysis revealed that 18 (43.9%) patients diagnosed as having genomic IM had common mutations in a pair of HCC tumors with the main tumor of these patients being more progressive compared to those with genomic MO. The accuracy of clinical diagnosis based on lobe (Definition 1) and segment (Definition 2) were 68.3% and 78.0%, respectively. Intriguingly, recurrence ≥2 years after initial surgery for 3 patients was IM. The survival of patients with clinical IM was significantly shorter than for those with clinical MO based on both Definition 1 (P = 0.045) and Definition 2 (P = 0.043). However, mean survival was not different between the patients with genomic IM and those with MO (P = 0.364). Taken together, genomic analysis elucidated that liver cancer may spread more extensively and more slowly than previously thought. In addition, distinguishing multiple HCC as IM or MC may have provided biological information but was not of clinical importance with respect to patient prognosis.
Collapse
Affiliation(s)
- Shogo Yamamoto
- Genome Science Division, RCAST, University of Tokyo, Tokyo, Japan
| | - Yutaka Midorikawa
- Genome Science Division, RCAST, University of Tokyo, Tokyo, Japan.,Department of Digestive Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Genta Nagae
- Genome Science Division, RCAST, University of Tokyo, Tokyo, Japan
| | - Kenji Tatsuno
- Genome Science Division, RCAST, University of Tokyo, Tokyo, Japan
| | - Hiroki Ueda
- Genome Science Division, RCAST, University of Tokyo, Tokyo, Japan
| | - Mitsuhiko Moriyama
- Gastroenterology and Hepatology, Nihon University School of Medicine, Tokyo, Japan
| | - Tadatoshi Takayama
- Department of Digestive Surgery, Nihon University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
1309
|
Dumenci OE, U AM, Khan SA, Holmes E, Taylor-Robinson SD. Exploring Metabolic Consequences of CPS1 and CAD Dysregulation in Hepatocellular Carcinoma by Network Reconstruction. J Hepatocell Carcinoma 2020; 7:1-9. [PMID: 32021853 PMCID: PMC6955626 DOI: 10.2147/jhc.s239039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 12/19/2019] [Indexed: 01/05/2023] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) is the fourth commonest cause of cancer-related mortality; it is associated with various genetic alterations, some involved in metabolic reprogramming. This study aimed to explore the potential metabolic impact of Carbamoyl Phosphate Synthase I (CPS1) and carbamoyl phosphate synthetase/aspartate transcarbamoylase/dihydroorotase (CAD) dysregulation through the reconstruction of a network that integrates information from the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, Human Metabolome Database (HMDB) and Human Protein Atlas (HPA). Methods and results Existing literature was used to determine the roles of CPS1 and CAD in HCC. CPS1 downregulation is thought to play a role in hepatocarcinogenesis through an increased glutamine availability for de novo pyrimidine biosynthesis, which CAD catalyzes the first three steps for. KEGG, HMDB and HPA were used to reconstruct a network of relevant pathways, demonstrating the relationships between genes and metabolites using the MetaboSignal package in R. The network was filtered to exclude any duplicates, and those greater than three steps away from CPS1 or CAD. Consequently, a network of 18 metabolites, 28 metabolic genes and 1 signaling gene was obtained, which indicated expression profiles and prognostic information of each gene in the network. Conclusion Information from different databases was collated to form an informative network that integrated different “-omics” approaches, demonstrating the relationships between genetic and metabolic components of urea cycle and the de novo pyrimidine biosynthesis pathway. This study paves the way for further research by acting as a template to investigate the relationships between genes and metabolites, explore their potential roles in various diseases and aid the development of new screening and treatment methods through network reconstruction.
Collapse
|
1310
|
PRAS40 hyperexpression promotes hepatocarcinogenesis. EBioMedicine 2020; 51:102604. [PMID: 31901857 PMCID: PMC6950779 DOI: 10.1016/j.ebiom.2019.102604] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/03/2019] [Accepted: 12/12/2019] [Indexed: 12/16/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most common cancers, whereas the molecular mechanism remains largely unknown. PRAS40 (encoded by AKT1S1) phosphorylation was increased in human melanoma, prostate cancer and lung cancer specimens, which was considered as the results of Akt activation. However the mechanism in detail and its role in HCC stay elusive. Methods PRAS40 expression and phosphorylation were analyzed in HCC specimens, and the survival rates of patients were investigated. Functional analyses of PRAS40 in HCC were performed in vivo and in vitro. The miR-124-3p binding sites in PRAS40 were investigated using luciferase assay. MiR-124-3p expression in HCC specimens was examined by In Situ hybridization, and the correlation to PRAS40 level was evaluated. Findings The phosphorylation, protein and mRNA levels of PRAS40 were increased significantly in HCC specimens from our cohorts and TCGA database, which was positively correlated to the poor prognosis of HCC patients. Compared to Akt1s1+/+ mice, hepatocarcinogenesis was suppressed in Akt1s1−/− mice, and the activation of Akt was impaired. PRAS40 depletion resulted in the inhibition of HCC cellular proliferation. Tumor suppressor miR-124-3p was found to downregulate PRAS40 expression by targeting its 3′UTR. MiR-124-3p levels were inversely correlated to PRAS40 protein and phosphorylation levels in HCC specimens. The proliferation inhibition by miR-124-3p mimics was partially reversed by exogenous PRAS40 introduction in HCC cells. Interpretation PRAS40 hyperexpression induced by loss of miR-124-3p contributes to PRAS40 hyperphosphorylation and hepatocarcinogenesis. These results could be expected to offer novel clues for understanding hepatocarcinogenesis and developing approaches.
Collapse
|
1311
|
TAZ target gene ITGAV regulates invasion and feeds back positively on YAP and TAZ in liver cancer cells. Cancer Lett 2020; 473:164-175. [PMID: 31904487 DOI: 10.1016/j.canlet.2019.12.044] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/13/2019] [Accepted: 12/25/2019] [Indexed: 12/13/2022]
Abstract
The Hippo pathway effectors yes-associated protein (YAP) and WW domain containing transcription regulator 1 (TAZ/WWTR1) support tumor initiation and progression in various cancer entities including hepatocellular carcinoma (HCC). However, to which extent YAP and TAZ contribute to liver tumorigenesis via common and exclusive molecular mechanisms is poorly understood. RNAinterference (RNAi) experiments illustrate that YAP and TAZ individually support HCC cell viability and migration, while for invasion additive effects were observed. Comprehensive expression profiling revealed partly overlapping YAP/TAZ target genes as well as exclusively regulated genes. Integrin-αV (ITGAV) is a novel TAZ-specific target gene, whose overexpression in human HCC patients correlates with poor clinical outcome, TAZ expression in HCCs, and the abundance of YAP/TAZ target genes. Functionally, ITGAV contributes to actin stress fiber assembly, tumor cell migration and invasion. Perturbation of ITGAV diminishes actin fiber formation and nuclear YAP/TAZ protein levels. We describe a novel Hippo downstream mechanism in HCC cells, which is regulated by TAZ and ITGAV and that feedbacks on YAP/TAZ activity. This mechanism may represent a therapeutic target structure since it contributes to signal amplification of oncogenic YAP/TAZ in hepatocarcinogenesis.
Collapse
|
1312
|
Chen W, Peng J, Ye J, Dai W, Li G, He Y. Aberrant AFP expression characterizes a subset of hepatocellular carcinoma with distinct gene expression patterns and inferior prognosis. J Cancer 2020; 11:403-413. [PMID: 31897235 PMCID: PMC6930420 DOI: 10.7150/jca.31435] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 08/26/2019] [Indexed: 12/20/2022] Open
Abstract
Background Serum tumor markers are ubiquitously used in the clinic for cancer screening. However, the mechanisms accounting for the elevated levels of the serum tumor markers remain to be explored. Methods We performed a pan-cancer analysis of serum alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA) and prostate-specific antigen (PSA). The relation between concentration of serum tumor markers and the expression of their coding genes was assessed. Then the expression of AFP and its genomic background in hepatocellular carcinoma (liver cancer) was studied. Results High expression of AFP mRNA was found mainly in liver cancer. In gastric cancer, breast cancer and lung cancer, high AFP mRNA expression was also discovered occasionally. In liver cancer patients, serum AFP levels correlated significantly with AFP mRNA expression in cancer tissues (r = 0.72, p = 1.6e-45). Whole transcriptome analysis revealed that serum AFP levels clearly separated liver cancer into two classes with distinct expression profiles according to PCA analysis. Gene co-expression analysis revealed that AFP expression was connected to a module enriched with genes accounting for cell cycle and cell proliferation regulation. In addition, high AFP expression was associated with the molecular classification of liver cancer, including iCluster (Chi-square: 16.86, P = 0.0002). Methylation analysis revealed de-methylation of AFP promoter occurred in some liver cancer tissues, which was significantly related to AFP mRNA expression. Survival analysis indicated high serum AFP levels was prognostic of poorer survival of the liver cancer patients (Log-rank test: p = 0.046). This was confirmed by an independent dataset in which liver cancer patients with high serum AFP also had poorer survival (Log-rank test: p = 0.024). Conclusion High expression of AFP defined a subtype of liver cancer with distinct gene expression profiles and clinical features. De-methylation of cytosine from CpG di-nucleotides in AFP promoter may be the cause of AFP re-expression in adult human liver cancer tissue.
Collapse
Affiliation(s)
- Wei Chen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, 510080 Guangzhou, Guangdong province, People's Republic of China
| | - Jianjun Peng
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, 510080 Guangzhou, Guangdong province, People's Republic of China
| | - Jinning Ye
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, 510080 Guangzhou, Guangdong province, People's Republic of China
| | - Weigang Dai
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, 510080 Guangzhou, Guangdong province, People's Republic of China
| | - Guanghua Li
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, 510080 Guangzhou, Guangdong province, People's Republic of China
| | - Yulong He
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, 510080 Guangzhou, Guangdong province, People's Republic of China
| |
Collapse
|
1313
|
Mansouri V, Razzaghi M, Nikzamir A, Ahmadzadeh A, Iranshahi M, Haghazali M, Hamdieh M. Assessment of liver cancer biomarkers. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2020; 13:S29-S39. [PMID: 33585001 PMCID: PMC7881406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Liver cancer is the third cause of cancer-related deaths in the world. It is primarily divides into two main types, namely hepatocellular carcinoma (HC) and cholangiocarcinoma (IC). Due to the increasing number of patients with liver cancer and the high mortality rate, early diagnosis of the disease can be helpful in treatment, but most patients are diagnosed atlate stages of HC. The aim of this study is to screen and provide an overview on candidate biomarkers related to primary liver cancer to introduce the critical ones. In this study, various biomarkers related to the diagnosis of primary liver cancer have been studied. Accordingly, biomarkers are divided into different groups as blood biomarkers classified as serum and plasma biomarkers, tissue biomarkers, microRNA biomarkers, proteomic biomarkers and altered genes. Previous researches have focused on liver cells and bile ducts, the surround cellular environment, how cells differentiate, and the types of genes expressed in liver cancer. Some even have focused on the origin of tumor cells and how they differentiate and develop. In all these studies, the expression of specific proteins and genes in liver cancer has been considered. Based on available sources, biomarkers can be considered as candidates to diagnose and prognosis of various types of primary liver cancer, from sources such as blood, tissue, mic-RNA, proteome and genes. However, more investigations are required to introduce a biomarker for precise detection of early liver cancer.
Collapse
Affiliation(s)
- Vahid Mansouri
- Proteomics Research Center, School of Rehabilitation, ShahidBeheshti University of Medical Sciences, Tehran, Iran
| | - Mohhamadreza Razzaghi
- Laser Application in Medical Sciences Research Center, ShahidBeheshti University of Medical Sciences, Tehran, Iran
| | - Abdolrahim Nikzamir
- Faculty of Medicine, ShahidBeheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Ahmadzadeh
- Proteomics Research Center, Faculty of Paramedical Sciences, ShahidBeheshti University of Medical Sciences, Tehran, Iran
| | - Majid Iranshahi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Haghazali
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mostafa Hamdieh
- Department of Psychosomatic, Taleghani Hospital, Faculty of Medicine, ShahidBeheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
1314
|
Verhelst X, Dias AM, Colombel JF, Vermeire S, Van Vlierberghe H, Callewaert N, Pinho SS. Protein Glycosylation as a Diagnostic and Prognostic Marker of Chronic Inflammatory Gastrointestinal and Liver Diseases. Gastroenterology 2020; 158:95-110. [PMID: 31626754 DOI: 10.1053/j.gastro.2019.08.060] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/03/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022]
Abstract
Glycans are sequences of carbohydrates that are added to proteins or lipids to modulate their structure and function. Glycans modify proteins required for regulation of immune cells, and alterations have been associated with inflammatory conditions. For example, specific glycans regulate T-cell activation, structures, and functions of immunoglobulins; interactions between microbes and immune and epithelial cells; and malignant transformation in the intestine and liver. We review the effects of protein glycosylation in regulation of gastrointestinal and liver functions, and how alterations in glycosylation serve as diagnostic or prognostic factors, or as targets for therapy.
Collapse
Affiliation(s)
- Xavier Verhelst
- Department of Gastroenterology and Hepatology, Ghent University Hospital, Ghent, Belgium
| | - Ana M Dias
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal; Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
| | | | - Severine Vermeire
- Translational Research in Gastrointestinal Disorders, Department of Clinical and Experimental Medicine, Katholieke Universiteit Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Hans Van Vlierberghe
- Department of Gastroenterology and Hepatology, Ghent University Hospital, Ghent, Belgium
| | - Nico Callewaert
- Vlaams Instituut voor Biotechnologie-UGent Center for Medical Biotechnology, Gent, Belgium
| | - Salomé S Pinho
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal; Institute for Research and Innovation in Health, University of Porto, Porto, Portugal; Medical Faculty, University of Porto, Porto, Portugal.
| |
Collapse
|
1315
|
Zhang CH, Li M, Lin YP, Gao Q. Systemic Therapy for Hepatocellular Carcinoma: Advances and Hopes. Curr Gene Ther 2020; 20:84-99. [PMID: 32600231 DOI: 10.2174/1566523220666200628014530] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/24/2022]
Abstract
The majority of patients with hepatocellular carcinoma (HCC) are diagnosed at an advanced stage that can only benefit from systemic treatments. Although HCC is highly treatmentresistant, significant achievements have been made in the molecular targeted therapy and immunotherapy of HCC. In addition to regorafenib, cabozantinib and ramucirumab were approved for the second- line targeted treatment by the FDA after disease progression on sorafenib. Nivolumab failed to demonstrate remarkable benefit in overall survival (OS) as first-line therapy, while pembrolizumab did not achieve pre-specified statistical significance in both OS and progression-free survival (PFS) as second-line treatment. Combinations of targeted agents, immune checkpoint inhibitors and other interventions showed favorable results. In this review, we summarized the progress of systemic therapy in HCC and discussed the future directions of the treatment of HCC.
Collapse
Affiliation(s)
- Chen-Hao Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Ming Li
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - You-Pei Lin
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| |
Collapse
|
1316
|
Nault JC, Martin Y, Caruso S, Hirsch TZ, Bayard Q, Calderaro J, Charpy C, Copie-Bergman C, Ziol M, Bioulac-Sage P, Couchy G, Blanc JF, Nahon P, Amaddeo G, Ganne-Carrie N, Morcrette G, Chiche L, Duvoux C, Faivre S, Laurent A, Imbeaud S, Rebouissou S, Llovet JM, Seror O, Letouzé E, Zucman-Rossi J. Clinical Impact of Genomic Diversity From Early to Advanced Hepatocellular Carcinoma. Hepatology 2020; 71:164-182. [PMID: 31206197 DOI: 10.1002/hep.30811] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/18/2019] [Indexed: 12/24/2022]
Abstract
To date, genomic analyses of hepatocellular carcinoma (HCC) have been limited to early stages obtained from liver resection. We aim to describe the genomic profiling of HCC from early to advanced stages. We analyzed 801 HCC from 720 patients (410 resections, 137 transplantations, 122 percutaneous ablations, and 52 noncurative) for 190 gene expressions and for 31 gene mutations. Forty-one advanced HCC and 156 whole exome of Barcelona Clinic Liver Cancer (BCLC) 0/A were analyzed by whole-exome sequencing. Genomic profiling was correlated with tumor stages, clinical features, and survival. Our cohort included patients classified in BCLC stage 0 (9.4%), A (59.5%), B (16.2%), and C (14.9%). Among the overall 801 HCC, the most frequently mutated genes were telomerase reverse transcriptase (TERT) (58.1%), catenin beta 1 (CTNNB1) (30.7%), tumor protein 53 (TP53; 18.7%), AT-rich interaction domain 1A (ARID1A) (13%), albumin (11.4%), apolipoprotein B (APOB) (9.4%), and AXIN1 (9.2%). Advanced-stage HCC (BCLC B/C) showed higher frequencies of splicing factor 3b subunit 1 (SF3B1) (P = 0.0003), TP53 (P = 0.0006), and RB Transcriptional Corepressor 1 mutations (P = 0.03). G1-G6 transcriptomic classification and the molecular prognostic 5-gene score showed different distributions according to the stage of the disease and the type of treatment with an enrichment of G3 (P < 0.0001), poor prognostic score (P < 0.0001), and increased proliferation and dedifferentiation at the transcriptomic level in advanced HCC. The 5-gene score predicted survival in patients treated by resection (P < 0.0001) and ablation (P = 0.01) and in advanced HCC (P = 0.04). Twenty-two percent of advanced HCC harbored potentially druggable genetic alterations, and MET amplification was associated with complete tumor response in patients with advanced HCC treated by a specific MET inhibitor. Conclusion: Genomic analysis across the different stages of HCC revealed the mechanisms of tumor progression and helped to identify biomarkers of response to targeted therapies.
Collapse
Affiliation(s)
- Jean-Charles Nault
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France.,Service d'Hépatologie, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance-Publique Hôpitaux de Paris, Bondy, France.,Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d'Universités et Etablissements Sorbonne Paris Cité, Paris, France
| | - Yoann Martin
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Stefano Caruso
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Théo Z Hirsch
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Quentin Bayard
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Julien Calderaro
- Service d'Anatomopathologie, Hôpital Henri Mondor, Créteil, France.,Université Paris Est Créteil, Inserm U955, Team 18, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Cecile Charpy
- Service d'Anatomopathologie, Hôpital Henri Mondor, Créteil, France.,Université Paris Est Créteil, Inserm U955, Team 18, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Christiane Copie-Bergman
- Service d'Anatomopathologie, Hôpital Henri Mondor, Créteil, France.,Université Paris Est Créteil, Inserm U955, Team 18, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Marianne Ziol
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France.,Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d'Universités et Etablissements Sorbonne Paris Cité, Paris, France.,Service d'Anatomopathologie, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance-Publique Hôpitaux de Paris, Bondy, France.,Centre de Ressources Biologiques (BB-0033-00027) Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance-Publique Hôpitaux de Paris, Bondy, France
| | - Paulette Bioulac-Sage
- University of Bordeaux, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France.,Service de Pathologie, Hôpital Pellegrin, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Gabrielle Couchy
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Jean-Frédéric Blanc
- Service Hépato-Gastroentérologie et Oncologie Digestive, Centre Medico-Chirurgical Magellan, Hôpital Haut-Lévêque, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Pierre Nahon
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France.,Service d'Hépatologie, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance-Publique Hôpitaux de Paris, Bondy, France.,Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d'Universités et Etablissements Sorbonne Paris Cité, Paris, France
| | - Giuliana Amaddeo
- Service d'Hépatogastroentérologie, Hôpital Henri Mondor, Université Paris Est Créteil, Créteil, France
| | - Nathalie Ganne-Carrie
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France.,Service d'Hépatologie, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance-Publique Hôpitaux de Paris, Bondy, France.,Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d'Universités et Etablissements Sorbonne Paris Cité, Paris, France
| | - Guillaume Morcrette
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Laurence Chiche
- Service de Chirurgie Digestive, Centre Medico-Chirurgical Magellan, Hôpital Haut-Lévêque, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Christophe Duvoux
- Service d'Hépatogastroentérologie, Hôpital Henri Mondor, Université Paris Est Créteil, Créteil, France
| | - Sandrine Faivre
- Service d'Oncologie Médicale, Hôpital Beaujon, Assistance-Publique Hôpitaux de Paris, Clichy, France
| | - Alexis Laurent
- Service de Chirurgie Digestive, Hôpital Henri Mondor, Créteil, France.,Université Paris Est Créteil, Créteil, France
| | - Sandrine Imbeaud
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Sandra Rebouissou
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Josep M Llovet
- Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY.,Liver Cancer Translational Research Laboratory, BCLC Group, IDIBAPS, CIBEREHD, Hospital Clinic, Universitat de Barcelona, Catalonia, Spain.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Olivier Seror
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France.,Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d'Universités et Etablissements Sorbonne Paris Cité, Paris, France.,Service de Radiologie, Hôpital Jean Verdier, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance-Publique Hôpitaux de Paris, Bondy, France
| | - Eric Letouzé
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Functional Genomics of Solid Tumors Laboratory, Paris, France.,Hôpital Europeen Georges Pompidou, Assistance-Publique Hôpitaux de Paris, Paris, France
| |
Collapse
|
1317
|
Abstract
Cullin 3 (Cul3) family of ubiquitin ligases comprises three components, the RING finger protein RBX1, the Cul3 scaffold, and a Bric-a-brac/Tramtrack/Broad complex (BTB) protein. The BTB protein serves as a bridge to connect Cul3 to substrate and is functionally equivalent to the combination of substrate adaptor and linker in other Cullin complexes. Human genome encodes for ~180 BTB proteins, implying a broad spectrum of ubiquitination signals and substrate repertoire. Accordingly, Cul3 ubiquitin ligases are involved in diverse cellular processes, including cell division, differentiation, cytoskeleton remodeling, stress responses, and nerve cell functions. Emerging evidence has pointed to the prominent role of Cul3 ubiquitin ligases in cancer. This chapter will describe recent advances on the roles of Cul3 E3 ligase complexes in regulating various cancer hallmarks and therapeutic responses and the mutation/dysregulation of Cul3 substrate adaptors in cancer. In particular, we will focus on several extensively studied substrate adaptors, such as Keap1, SPOP, KLHL20, and LZTR1, and will also discuss other recently identified Cul3 adaptors with oncogenic or tumor-suppressive functions. We conclude that Cul3 ubiquitin ligases represent master regulators of human malignancies and highlight the importance of developing modulating agents for oncogenic/tumor-suppressive Cul3 E3 ligase complexes to prevent or intervene tumorigenesis.
Collapse
Affiliation(s)
- Ruey-Hwa Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
1318
|
Wang R, Song S, Harada K, Ghazanfari Amlashi F, Badgwell B, Pizzi MP, Xu Y, Zhao W, Dong X, Jin J, Wang Y, Scott A, Ma L, Huo L, Vicente D, Blum Murphy M, Shanbhag N, Tatlonghari G, Thomas I, Rogers J, Kobayashi M, Vykoukal J, Estrella JS, Roy-Chowdhuri S, Han G, Zhang S, Mao X, Song X, Zhang J, Gu J, Johnson RL, Calin GA, Peng G, Lee JS, Hanash SM, Futreal A, Wang Z, Wang L, Ajani JA. Multiplex profiling of peritoneal metastases from gastric adenocarcinoma identified novel targets and molecular subtypes that predict treatment response. Gut 2020; 69:18-31. [PMID: 31171626 PMCID: PMC6943252 DOI: 10.1136/gutjnl-2018-318070] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/14/2019] [Accepted: 04/04/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Peritoneal carcinomatosis (PC) occurs frequently in patients with gastric adenocarcinoma (GAC) and confers a poor prognosis. Multiplex profiling of primary GACs has been insightful but the underpinnings of PC's development/progression remain largely unknown. We characterised exome/transcriptome/immune landscapes of PC cells from patients with GAC aiming to identify novel therapeutic targets. DESIGN We performed whole-exome sequencing (WES) and whole transcriptome sequencing (RNA-seq) on 44 PC specimens (43 patients with PC) including an integrative analysis of WES, RNA-seq, immune profile, clinical and pathological phenotypes to dissect the molecular pathogenesis, identifying actionable targets and/or biomarkers and comparison with TCGA primary GACs. RESULTS We identified distinct alterations in PC versus primary GACs, such as more frequent CDH1 and TAF1 mutations, 6q loss and chr19 gain. Alterations associated with aggressive PC phenotypes emerged with increased mutations in TP53, CDH1, TAF1 and KMT2C, higher level of 'clock-like' mutational signature, increase in whole-genome doublings, chromosomal instability (particularly, copy number losses), reprogrammed microenvironment, enriched cell cycle pathways, MYC activation and impaired immune response. Integrated analysis identified two main molecular subtypes: 'mesenchymal-like' and 'epithelial-like' with discriminating response to chemotherapy (31% vs 71%). Patients with the less responsive 'mesenchymal-like' subtype had high expression of immune checkpoint T-Cell Immunoglobulin And Mucin Domain-Containing Protein 3 (TIM-3), its ligand galectin-9, V-domain Ig suppressor of T cell activation (VISTA) and transforming growth factor-β as potential therapeutic immune targets. CONCLUSIONS We have uncovered the unique mutational landscape, copy number alteration and gene expression profile of PC cells and defined PC molecular subtypes, which correlated with PC therapy resistance/response. Novel targets and immune checkpoint proteins have been identified with a potential to be translated into clinics.
Collapse
Affiliation(s)
| | - Shumei Song
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Kazuto Harada
- GI Medical Oncology, UT MDACC, Houston, Texas, USA,Gastroenterological Surgery, Kumamoto University, Kumamoto, Japan
| | | | | | | | - Yan Xu
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Wei Zhao
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | | | | | - Ying Wang
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Ailing Scott
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Lang Ma
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Longfei Huo
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | | | | | | | | | - Irene Thomas
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Jane Rogers
- Pharmacy Clinical Programs, UT MDACC, Houston, TX, USA
| | | | - Jody Vykoukal
- Clinical Cancer Prevention, UT MDACC, Houston, Texas, USA
| | | | | | | | | | - Xizeng Mao
- Genomic Medicine, UT MDACC, Houston, Texas, USA
| | | | | | - Jian Gu
- Epidemiology, UT MDACC, Houston, Texas, USA
| | | | | | - Guang Peng
- Clinical Cancer Prevention, UT MDACC, Houston, Texas, USA
| | - Ju-Seog Lee
- Systems Biology, UT MDACC, Houston, Texas, USA
| | - Samir M Hanash
- Clinical Cancer Prevention, UT MDACC, Houston, Texas, USA
| | | | - Zhenning Wang
- Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, China
| | | | | |
Collapse
|
1319
|
Chen H, He Y, Jia W. Precise hepatectomy in the intelligent digital era. Int J Biol Sci 2020; 16:365-373. [PMID: 32015674 PMCID: PMC6990894 DOI: 10.7150/ijbs.39387] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
In the past 20 years, the concept of surgery has undergone profound changes. Surgical practice has shifted from emphasizing the complete elimination of lesions to achieving optimal rehabilitation in patients. Collaborative optimization of surgery consists of three core elements, removal of lesions, organ protection and injury close monitoring, and controlled surgical intervention. As a result, the traditional surgical paradigm has quietly transformed into a modern precision surgical paradigm. In this review, we summarized the latest breakthroughs and applications of precision medicine in liver surgery. In addition, we also outlined the progresses that have been made in precision liver surgery, the opportunities and challenges that may encountered in the future.
Collapse
Affiliation(s)
- Hao Chen
- Department of Hepatic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, HeFei, 230001, China.,Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, HeFei, 230001, China
| | - Yuchen He
- Xiangya School of Medicine, Central South University, ChangSha, 410008, China
| | - Weidong Jia
- Department of Hepatic Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, HeFei, 230001, China.,Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, HeFei, 230001, China
| |
Collapse
|
1320
|
Li X, Wu Y, Suo P, Liu G, Li L, Zhang X, Chen S, Xu M, Song L. Identification of a novel germline frameshift mutation p.D300fs of PMS1 in a patient with hepatocellular carcinoma: A case report and literature review. Medicine (Baltimore) 2020; 99:e19076. [PMID: 32000458 PMCID: PMC7004782 DOI: 10.1097/md.0000000000019076] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 12/03/2019] [Accepted: 01/07/2020] [Indexed: 12/22/2022] Open
Abstract
RATIONALE PMS1 is one of the mismatch repair (MMR) genes with potential crucial roles in carcinogenesis. Very few reports have been identified on germline PMS1 mutations with definite disease phenotype. Here we report a case of hepatocellular carcinoma (HCC) with a novel potential pathogenic germline PMS1 mutation. PATIENT CONCERNS A 46-year-old Chinese male with Hepatitis B infection history presented a single cancerous nodule (10×12×10 mm) at the left lobe of liver. The nodule was considered malignant by type-B ultrasonic and computed tomography (CT) examinations. DIAGNOSIS AND INTERVENTION Liver lobectomy was performed to remove the liver cancerous nodule and postoperative TACE was performed for recurrence prevention. Pathological examination on resected tumor tissue confirmed the diagnosis of HCC. Whole-exome sequencing (WES) identified the c.900delT (p.D300fs) heterozygous germline mutation of PMS1, along with 253 nonsynonymous single nucleotide variations (SNVs), 14 Insertion or deletion mutations (INDELs) and 21 genes with copy number variations (CNVs). Three-dimensional prediction of protein tertiary structure suggested that the conformation of the enzyme active site and the ligand binding site might be changed due to the protein truncation. OUTCOMES The patient was still alive in good condition with no sign of recurrence in 12 months follow-up period. LESSONS The affected pathways in this case were unique from previously reported HCC patients with no PMS1 germline mutations. The novel PMS1 germline mutation may increase cancer risk. The roles of PMS1 germline mutations in carcinogenesis need further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Lele Song
- HaploX Biotechnology, Co., Ltd
- Department of Radiotherapy, the Eighth Medical Center of the Chinese PLA General Hospital, PR China
| |
Collapse
|
1321
|
Deng X, Bi Q, Chen S, Chen X, Li S, Zhong Z, Guo W, Li X, Deng Y, Yang Y. Identification of a Five-Autophagy-Related-lncRNA Signature as a Novel Prognostic Biomarker for Hepatocellular Carcinoma. Front Mol Biosci 2020; 7:611626. [PMID: 33505990 PMCID: PMC7831610 DOI: 10.3389/fmolb.2020.611626] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022] Open
Abstract
Although great progresses have been made in the diagnosis and treatment of hepatocellular carcinoma (HCC), its prognostic marker remains controversial. In this current study, weighted correlation network analysis and Cox regression analysis showed significant prognostic value of five autophagy-related long non-coding RNAs (AR-lncRNAs) (including TMCC1-AS1, PLBD1-AS1, MKLN1-AS, LINC01063, and CYTOR) for HCC patients from data in The Cancer Genome Atlas. By using them, we constructed a five-AR-lncRNA prognostic signature, which accurately distinguished the high- and low-risk groups of HCC patients. All of the five AR lncRNAs were highly expressed in the high-risk group of HCC patients. This five-AR-lncRNA prognostic signature showed good area under the curve (AUC) value (AUC = 0.751) for the overall survival (OS) prediction in either all HCC patients or HCC patients stratified according to several clinical traits. A prognostic nomogram with this five-AR-lncRNA signature predicted the 3- and 5-year OS outcomes of HCC patients intuitively and accurately (concordance index = 0.745). By parallel comparison, this five-AR-lncRNA signature has better prognosis accuracy than the other three recently published signatures. Furthermore, we discovered the prediction ability of the signature on therapeutic outcomes of HCC patients, including chemotherapy and immunotherapeutic responses. Gene set enrichment analysis and gene mutation analysis revealed that dysregulated cell cycle pathway, purine metabolism, and TP53 mutation may play an important role in determining the OS outcomes of HCC patients in the high-risk group. Collectively, our study suggests a new five-AR-lncRNA prognostic signature for HCC patients.
Collapse
Affiliation(s)
- Xiaoyu Deng
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qinghua Bi
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yao Yang
| | - Shihan Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Xianhua Chen
- Diagosis and Treatment Center for Servicemen, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuhui Li
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhaoyang Zhong
- Cancer Center, Daping Hospital and Research Institute of Surgery, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wei Guo
- Department of Pharmacy, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiaohui Li
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
- Youcai Deng
| | - Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
- Xiaohui Li
| | - Yao Yang
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
- Qinghua Bi
| |
Collapse
|
1322
|
Che L, Chi W, Qiao Y, Zhang J, Song X, Liu Y, Li L, Jia J, Pilo MG, Wang J, Cigliano A, Ma Z, Kuang W, Tang Z, Zhang Z, Shui G, Ribback S, Dombrowski F, Evert M, Pascale RM, Cossu C, Pes GM, Osborne TF, Calvisi DF, Chen X, Chen L. Cholesterol biosynthesis supports the growth of hepatocarcinoma lesions depleted of fatty acid synthase in mice and humans. Gut 2020; 69:177-186. [PMID: 30954949 PMCID: PMC6943247 DOI: 10.1136/gutjnl-2018-317581] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Increased de novo fatty acid (FA) synthesis and cholesterol biosynthesis have been independently described in many tumour types, including hepatocellular carcinoma (HCC). DESIGN We investigated the functional contribution of fatty acid synthase (Fasn)-mediated de novo FA synthesis in a murine HCC model induced by loss of Pten and overexpression of c-Met (sgPten/c-Met) using liver-specific Fasn knockout mice. Expression arrays and lipidomic analysis were performed to characterise the global gene expression and lipid profiles, respectively, of sgPten/c-Met HCC from wild-type and Fasn knockout mice. Human HCC cell lines were used for in vitro studies. RESULTS Ablation of Fasn significantly delayed sgPten/c-Met-driven hepatocarcinogenesis in mice. However, eventually, HCC emerged in Fasn knockout mice. Comparative genomic and lipidomic analyses revealed the upregulation of genes involved in cholesterol biosynthesis, as well as decreased triglyceride levels and increased cholesterol esters, in HCC from these mice. Mechanistically, loss of Fasn promoted nuclear localisation and activation of sterol regulatory element binding protein 2 (Srebp2), which triggered cholesterogenesis. Blocking cholesterol synthesis via the dominant negative form of Srebp2 (dnSrebp2) completely prevented sgPten/c-Met-driven hepatocarcinogenesis in Fasn knockout mice. Similarly, silencing of FASN resulted in increased SREBP2 activation and hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase (HMGCR) expression in human HCC cell lines. Concomitant inhibition of FASN-mediated FA synthesis and HMGCR-driven cholesterol production was highly detrimental for HCC cell growth in culture. CONCLUSION Our study uncovers a novel functional crosstalk between aberrant lipogenesis and cholesterol biosynthesis pathways in hepatocarcinogenesis, whose concomitant inhibition might represent a therapeutic option for HCC.
Collapse
Affiliation(s)
- Li Che
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Wenna Chi
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, China,Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Yu Qiao
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,Department of Oncology, National Center of Gerontology, Beijing Hospital, Beijing, China
| | - Jie Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital, Beijing, China,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Xinhua Song
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Ye Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, China
| | - Lei Li
- School of Pharmacy, Huazhong University of Science and Technology Tongji Medical College, Wuhan, Hubei, China
| | - Jiaoyuan Jia
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,Department of Oncology and Hematology, The Second Hospital, Jilin University, Changchun, Jilin, China
| | - Maria G Pilo
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Sardegna, Italy
| | - Jingxiao Wang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA,Beijing University of Chinese Medicine, Beijing, China
| | - Antonio Cigliano
- Institute of Pathology, University Medicine of Greifswald, Greifswald, Germany
| | - Zhilong Ma
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, China
| | - Wenhua Kuang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, China
| | - Zefang Tang
- BIOPIC, Beijing Advanced Innovation Center for Genomics, and School of Life Sciences, Peking University, Beijing, China,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Zemin Zhang
- BIOPIC, Beijing Advanced Innovation Center for Genomics, and School of Life Sciences, Peking University, Beijing, China,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology Chinese Academy of Sciences, Beijing, China
| | - Silvia Ribback
- Institute of Pathology, University Medicine of Greifswald, Greifswald, Germany
| | - Frank Dombrowski
- Institute of Pathology, University Medicine of Greifswald, Greifswald, Germany
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Rosa Maria Pascale
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Sardegna, Italy
| | - Carla Cossu
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Sardegna, Italy
| | - Giovanni Mario Pes
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Sardegna, Italy
| | - Timothy F Osborne
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Diego F Calvisi
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Sardegna, Italy
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Ligong Chen
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Tsinghua University, Beijing, China,Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
1323
|
Takahashi SS, Sou YS, Saito T, Kuma A, Yabe T, Sugiura Y, Lee HC, Suematsu M, Yokomizo T, Koike M, Terai S, Mizushima N, Waguri S, Komatsu M. Loss of autophagy impairs physiological steatosis by accumulation of NCoR1. Life Sci Alliance 2020; 3:e201900513. [PMID: 31879337 PMCID: PMC6932742 DOI: 10.26508/lsa.201900513] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 02/02/2023] Open
Abstract
Lipid droplets (LDs) are dynamic organelles that store neutral lipids during times of energy excess, such as after a meal. LDs serve as an energy reservoir during fasting and have a buffering capacity that prevents lipotoxicity. Autophagy and the autophagic machinery have been proposed to play a role in LD biogenesis, but the underlying molecular mechanism remains unclear. Here, we show that when nuclear receptor co-repressor 1 (NCoR1), which inhibits the transactivation of nuclear receptors, accumulates because of autophagy suppression, LDs decrease in size and number. Ablation of ATG7, a gene essential for autophagy, suppressed the expression of gene targets of liver X receptor α, a nuclear receptor responsible for fatty acid and triglyceride synthesis in an NCoR1-dependent manner. LD accumulation in response to fasting and after hepatectomy was hampered by the suppression of autophagy. These results suggest that autophagy controls physiological hepatosteatosis by fine-tuning NCoR1 protein levels.
Collapse
Affiliation(s)
- Shun-Saku Takahashi
- Division of Gastroenterology and Hepatology, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, Japan
| | - Yu-Shin Sou
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Tetsuya Saito
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Akiko Kuma
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takayuki Yabe
- Department of Anatomy and Histology, Fukushima Medical University School of Medicine, Hikarigaoka, Fukushima, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Hyeon-Cheol Lee
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, Japan
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoshi Waguri
- Department of Anatomy and Histology, Fukushima Medical University School of Medicine, Hikarigaoka, Fukushima, Japan
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
1324
|
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: 50] [Impact Index Per Article: 10.0] [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
|
1325
|
Hilmi M, Vienot A, Rousseau B, Neuzillet C. Immune Therapy for Liver Cancers. Cancers (Basel) 2019; 12:E77. [PMID: 31892230 PMCID: PMC7016834 DOI: 10.3390/cancers12010077] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/24/2019] [Accepted: 12/25/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) and biliary tract cancers (BTC) display a poor prognosis with 5-year overall survival rates around 15%, all stages taken together. These primary liver malignancies are often diagnosed at advanced stages where therapeutic options are limited. Recently, immune therapy has opened new opportunities in oncology. Based on their high programmed death-ligand 1 expression and tumor-infiltrating lymphocytes, HCC and BTC are theoretically good candidates for immune checkpoint blockade. However, clinical activity of single agent immunotherapy appears limited to a subset of patients, which is still ill-defined, and combinations are under investigation. In this review, we provide an overview of (i) the biological rationale for immunotherapies in HCC and BTC, (ii) the current state of their clinical development, and (iii) the predictive value of immune signatures for both clinical outcome and response to these therapies.
Collapse
Affiliation(s)
- Marc Hilmi
- Department of Medical Oncology, Curie Institute, University of Versailles Saint-Quentin, 35 rue Dailly, 92210 Saint-Cloud, France;
- GERCOR Group, 151 rue du Faubourg Saint-Antoine, 75011 Paris, France; (A.V.); (B.R.)
| | - Angélique Vienot
- GERCOR Group, 151 rue du Faubourg Saint-Antoine, 75011 Paris, France; (A.V.); (B.R.)
- Department of Medical Oncology, Besançon University Hospital, 3 Boulevard Alexandre Fleming, 25030 Besançon, France
| | - Benoît Rousseau
- GERCOR Group, 151 rue du Faubourg Saint-Antoine, 75011 Paris, France; (A.V.); (B.R.)
- Department of Medicine, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Cindy Neuzillet
- Department of Medical Oncology, Curie Institute, University of Versailles Saint-Quentin, 35 rue Dailly, 92210 Saint-Cloud, France;
- GERCOR Group, 151 rue du Faubourg Saint-Antoine, 75011 Paris, France; (A.V.); (B.R.)
| |
Collapse
|
1326
|
Li Z, Lou Y, Tian G, Wu J, Lu A, Chen J, Xu B, Shi J, Yang J. Discovering master regulators in hepatocellular carcinoma: one novel MR, SEC14L2 inhibits cancer cells. Aging (Albany NY) 2019; 11:12375-12411. [PMID: 31851620 PMCID: PMC6949064 DOI: 10.18632/aging.102579] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022]
Abstract
Identification of master regulator (MR) genes offers a relatively rapid and efficient way to characterize disease-specific molecular programs. Since strong consensus regarding commonly altered MRs in hepatocellular carcinoma (HCC) is lacking, we generated a compendium of HCC datasets from 21 studies and identified a comprehensive signature consisting of 483 genes commonly deregulated in HCC. We then used reverse engineering of transcriptional networks to identify the MRs that underpin the development and progression of HCC. After cross-validation in different HCC datasets, systematic assessment using patient-derived data confirmed prognostic predictive capacities for most HCC MRs and their corresponding regulons. Our HCC signature covered well-established liver cancer hallmarks, and network analyses revealed coordinated interaction between several MRs. One novel MR, SEC14L2, exerted an anti-proliferative effect in HCC cells and strongly suppressed tumor growth in a mouse model. This study advances our knowledge of transcriptional MRs potentially involved in HCC development and progression that may be targeted by specific interventions.
Collapse
Affiliation(s)
- Zhihui Li
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Yi Lou
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China.,Department of Occupational Medicine, Zhejiang Provincial Integrated Chinese and Western Medicine Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Guoyan Tian
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Jianyue Wu
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Anqian Lu
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Jin Chen
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Beibei Xu
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Junping Shi
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Jin Yang
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| |
Collapse
|
1327
|
Tang F, Gao R, Jeevan-Raj B, Wyss CB, Kalathur RKR, Piscuoglio S, Ng CKY, Hindupur SK, Nuciforo S, Dazert E, Bock T, Song S, Buechel D, Morini MF, Hergovich A, Matthias P, Lim DS, Terracciano LM, Heim MH, Hall MN, Christofori G. LATS1 but not LATS2 represses autophagy by a kinase-independent scaffold function. Nat Commun 2019; 10:5755. [PMID: 31848340 PMCID: PMC6917744 DOI: 10.1038/s41467-019-13591-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 11/14/2019] [Indexed: 12/27/2022] Open
Abstract
Autophagy perturbation represents an emerging therapeutic strategy in cancer. Although LATS1 and LATS2 kinases, core components of the mammalian Hippo pathway, have been shown to exert tumor suppressive activities, here we report a pro-survival role of LATS1 but not LATS2 in hepatocellular carcinoma (HCC) cells. Specifically, LATS1 restricts lethal autophagy in HCC cells induced by sorafenib, the standard of care for advanced HCC patients. Notably, autophagy regulation by LATS1 is independent of its kinase activity. Instead, LATS1 stabilizes the autophagy core-machinery component Beclin-1 by promoting K27-linked ubiquitination at lysine residues K32 and K263 on Beclin-1. Consequently, ubiquitination of Beclin-1 negatively regulates autophagy by promoting inactive dimer formation of Beclin-1. Our study highlights a functional diversity between LATS1 and LATS2, and uncovers a scaffolding role of LATS1 in mediating a cross-talk between the Hippo signaling pathway and autophagy.
Collapse
Affiliation(s)
- Fengyuan Tang
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Ruize Gao
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Beena Jeevan-Raj
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Christof B Wyss
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | | - Charlotte K Y Ng
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | | | - Sandro Nuciforo
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Eva Dazert
- Biozentrum, University of Basel, Basel, Switzerland
| | - Thomas Bock
- Biozentrum, University of Basel, Basel, Switzerland
| | - Shuang Song
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - David Buechel
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marco F Morini
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Patrick Matthias
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Dae-Sik Lim
- Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | | | - Markus H Heim
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | |
Collapse
|
1328
|
Kim H, Lee AJ, Lee J, Chun H, Ju YS, Hong D. FIREVAT: finding reliable variants without artifacts in human cancer samples using etiologically relevant mutational signatures. Genome Med 2019; 11:81. [PMID: 31847917 PMCID: PMC6916105 DOI: 10.1186/s13073-019-0695-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/18/2019] [Indexed: 12/22/2022] Open
Abstract
Background Accurate identification of real somatic variants is a primary part of cancer genome studies and precision oncology. However, artifacts introduced in various steps of sequencing obfuscate confidence in variant calling. Current computational approaches to variant filtering involve intensive interrogation of Binary Alignment Map (BAM) files and require massive computing power, data storage, and manual labor. Recently, mutational signatures associated with sequencing artifacts have been extracted by the Pan-cancer Analysis of Whole Genomes (PCAWG) study. These spectrums can be used to evaluate refinement quality of a given set of somatic mutations. Results Here we introduce a novel variant refinement software, FIREVAT (FInding REliable Variants without ArTifacts), which uses known spectrums of sequencing artifacts extracted from one of the largest publicly available catalogs of human tumor samples. FIREVAT performs a quick and efficient variant refinement that accurately removes artifacts and greatly improves the precision and specificity of somatic calls. We validated FIREVAT refinement performance using orthogonal sequencing datasets totaling 384 tumor samples with respect to ground truth. Our novel method achieved the highest level of performance compared to existing filtering approaches. Application of FIREVAT on additional 308 The Cancer Genome Atlas (TCGA) samples demonstrated that FIREVAT refinement leads to identification of more biologically and clinically relevant mutational signatures as well as enrichment of sequence contexts associated with experimental errors. FIREVAT only requires a Variant Call Format file (VCF) and generates a comprehensive report of the variant refinement processes and outcomes for the user. Conclusions In summary, FIREVAT facilitates a novel refinement strategy using mutational signatures to distinguish artifactual point mutations called in human cancer samples. We anticipate that FIREVAT results will further contribute to precision oncology efforts that rely on accurate identification of variants, especially in the context of analyzing mutational signatures that bear prognostic and therapeutic significance. FIREVAT is freely available at https://github.com/cgab-ncc/FIREVAT
Collapse
Affiliation(s)
- Hyunbin Kim
- Bioinformatics Analysis Team, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Andy Jinseok Lee
- Bioinformatics Analysis Team, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Jongkeun Lee
- Bioinformatics Analysis Team, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Hyonho Chun
- Department of Mathematics and Statistics, Boston University, Boston, MA, 02215, USA
| | - Young Seok Ju
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Dongwan Hong
- Bioinformatics Analysis Team, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10408, Republic of Korea.
| |
Collapse
|
1329
|
A tumorigenic index for quantitative analysis of liver cancer initiation and progression. Proc Natl Acad Sci U S A 2019; 116:26873-26880. [PMID: 31843886 DOI: 10.1073/pnas.1911193116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Primary liver cancer develops from multifactorial etiologies, resulting in extensive genomic heterogeneity. To probe the common mechanism of hepatocarcinogenesis, we interrogated temporal gene expression profiles in a group of mouse models with hepatic steatosis, fibrosis, inflammation, and, consequently, tumorigenesis. Instead of anticipated progressive changes, we observed a sudden molecular switch at a critical precancer stage, by developing analytical platform that focuses on transcription factor (TF) clusters. Coarse-grained network modeling demonstrated that an abrupt transcriptomic transition occurred once changes were accumulated to reach a threshold. Based on the experimental and bioinformatic data analyses as well as mathematical modeling, we derived a tumorigenic index (TI) to quantify tumorigenic signal strengths. The TI is powerful in predicting the disease status of patients with metabolic disorders and also the tumor stages and prognosis of liver cancer patients with diverse backgrounds. This work establishes a quantitative tool for triage of liver cancer patients and also for cancer risk assessment of chronic liver disease patients.
Collapse
|
1330
|
Qin Y, Wu CW, Taylor WR, Sawas T, Burger KN, Mahoney DW, Sun Z, Yab TC, Lidgard GP, Allawi HT, Buttar NS, Smyrk TC, Iyer PG, Katzka DA, Ahlquist DA, Kisiel JB. Discovery, Validation, and Application of Novel Methylated DNA Markers for Detection of Esophageal Cancer in Plasma. Clin Cancer Res 2019; 25:7396-7404. [PMID: 31527170 PMCID: PMC6911634 DOI: 10.1158/1078-0432.ccr-19-0740] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/20/2019] [Accepted: 09/11/2019] [Indexed: 12/29/2022]
Abstract
PURPOSE The burden of esophageal cancer continues to rise, and noninvasive screening tools are needed. Methylated DNA markers (MDM) assayed from plasma show promise in detection of other cancers. For esophageal cancer detection, we aimed to discover and validate MDMs in tissue, and determine their feasibility when assayed from plasma. EXPERIMENTAL DESIGN Whole-methylome sequencing was performed on DNA extracted from 37 tissues (28 EC; 9 normal esophagus) and 8 buffy coat samples. Top MDMs were validated by methylation specific PCR on tissue from 76 EC (41 adeno, 35 squamous cell) and 17 normal esophagus. Quantitative allele-specific real-time target and signal amplification was used to assay MDMs in plasma from 183 patients (85 EC, 98 controls). Recursive partitioning (rPART) identified MDM combinations predictive of esophageal cancer. Validation was performed in silico by bootstrapping. RESULTS From discovery, 23 candidate MDMs were selected for independent tissue validation; median area under the receiver operating curve (AUC) for individual MDMs was 0.93. Among 12 MDMs advanced to plasma testing, rPART modeling selected a 5 MDM panel (FER1L4, ZNF671, ST8SIA1, TBX15, ARHGEF4) which achieved an AUC of 0.93 (95% CI, 0.89-0.96) on best-fit and 0.81 (95% CI, 0.75-0.88) on cross-validation. At 91% specificity, the panel detected 74% of esophageal cancer overall, and 43%, 64%, 77%, and 92% of stages I, II, III, and IV, respectively. Discrimination was not affected by age, sex, smoking, or body mass index. CONCLUSIONS Novel MDMs assayed from plasma detect esophageal cancer with moderate accuracy. Further optimization and clinical testing are warranted.
Collapse
Affiliation(s)
- Yi Qin
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Chung W Wu
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - William R Taylor
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Tarek Sawas
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Kelli N Burger
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Douglas W Mahoney
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Zhifu Sun
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Tracy C Yab
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Navtej S Buttar
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Thomas C Smyrk
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Prasad G Iyer
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - David A Katzka
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - David A Ahlquist
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - John B Kisiel
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
1331
|
Zhang X, Li J, Ghoshal K, Fernandez S, Li L. Identification of a Subtype of Hepatocellular Carcinoma with Poor Prognosis Based on Expression of Genes within the Glucose Metabolic Pathway. Cancers (Basel) 2019; 11:E2023. [PMID: 31847435 PMCID: PMC6966574 DOI: 10.3390/cancers11122023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/11/2019] [Accepted: 12/11/2019] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent primary cancer and a highly aggressive liver malignancy. Liver cancer cells reprogram their metabolism to meet their needs for rapid proliferation and tumor growth. In the present study, we investigated the alterations in the expression of the genes involved in glucose metabolic pathways as well as their association with the clinical stage and survival of HCC patients. We found that the expressions of around 30% of genes involved in the glucose metabolic pathway are consistently dysregulated with a predominant down-regulation in HCC tumors. Moreover, the differentially expressed genes are associated with an advanced clinical stage and a poor prognosis. More importantly, unsupervised clustering analysis with the differentially expressed genes that were also associated with overall survival (OS) revealed a subgroup of patients with a worse prognosis including reduced OS, disease specific survival, and recurrence-free survival. This aggressive subtype had significantly increased expression of stemness-related genes and down-regulated metabolic genes, as well as increased immune infiltrates that contribute to a poor prognosis. Collectively, this integrative study indicates that expressions of the glucose metabolic genes could be used as potential prognostic markers and/or therapeutic targets, which might be helpful in developing precise treatment for patients with HCC.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43201, USA; (J.L.); (S.F.); (L.L.)
| | - Jin Li
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43201, USA; (J.L.); (S.F.); (L.L.)
| | - Kalpana Ghoshal
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, OH 43201, USA;
| | - Soledad Fernandez
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43201, USA; (J.L.); (S.F.); (L.L.)
| | - Lang Li
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43201, USA; (J.L.); (S.F.); (L.L.)
| |
Collapse
|
1332
|
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: 320] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [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
|
1333
|
Survival correlation of immune response in human cancers. Oncotarget 2019; 10:6885-6897. [PMID: 31839882 PMCID: PMC6901338 DOI: 10.18632/oncotarget.27360] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/17/2019] [Indexed: 12/16/2022] Open
Abstract
Background: The clinical benefit of immune response is largely unknown. We systematically explored the correlation of immune response with patient outcome in human cancers. Results: The global immune gene signature was primarily located on the plasma membrane with a high gene density at 6p21 and 1q23-1q24. Immune responses varied with a wide range in human cancers. A total of 11 cancer types exhibited significant correlation of immune response with overall survival. Higher immune response was significantly associated with longer overall survival in 7 types and with shorter overall survival in 4 types. In addition, 11 cancer types exhibited significant correlation of immune response with progression-free interval. Higher immune response was significantly associated with longer progression-free interval in 7 types and with shorter progression-free interval in 4 types. Methods: The Ingenuity Knowledge Base and human genome assembly GRCh38 were used to annotate the immune gene signature by cellular components and genomic coordinates, respectively. We devised an mRNA-based metric of pre-existing immune conditions by using the gene signature, and calculated the metric for 10,062 The Cancer Genome Atlas tumor samples across 32 different cancer types. The Kaplan-Meier method was used to evaluate the overall survival and progression-free interval differences between dichotomic groups stratified by the median metric for each cancer type. Conclusions: Immune responses have different impacts on patient outcome in different human cancers. Prospective verification is needed before the findings can be applied for clinical trial development.
Collapse
|
1334
|
Heidor R, Affonso JM, Ong TP, Moreno FS. Nutrition and Liver Cancer Prevention. NUTRITION AND CANCER PREVENTION 2019:339-367. [DOI: 10.1039/9781788016506-00339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Liver cancer represents a major public health problem. Hepatocarcinogenesis is a complex process that comprises several stages and is caused by multiple factors. Both progressive genetic and epigenetic alterations are described in liver cancer development. The most effective strategy to reduce the impact of this disease is through prevention. In addition to vaccination against HBV and treatment of HCV infection, other preventive measures include avoiding ingesting aflatoxin-contaminated foods and drinking alcoholic beverages, as well as maintaining healthy body weight and practicing physical exercise. Bioactive compounds from fruits and vegetables present great potential for liver cancer chemoprevention. Among them, tea catechins, carotenoids, retinoids, β-ionone, geranylgeraniol and folic acid can be highlighted. In addition, butyric acid, tributyrin and structured lipids based on butyric acid and other fatty acids represent additional promising chemopreventive agents. These bioactive food compounds have been shown to modulate key cellular and molecular processes that are deregulated in hepatocarcinogenesis. Furthermore, combinations of different classes of bioactive food compounds or of bioactive food compounds with synthetic drugs could lead to synergistic liver cancer chemopreventive effects.
Collapse
Affiliation(s)
- R. Heidor
- University of São Paulo, Faculty of Pharmaceutical Sciences, Department of Food and Experimental Nutrition, Food Research Center (FoRC) São Paulo 05508-000 Brazil
| | - J. M. Affonso
- University of São Paulo, Faculty of Pharmaceutical Sciences, Department of Food and Experimental Nutrition, Food Research Center (FoRC) São Paulo 05508-000 Brazil
| | - T. P. Ong
- University of São Paulo, Faculty of Pharmaceutical Sciences, Department of Food and Experimental Nutrition, Food Research Center (FoRC) São Paulo 05508-000 Brazil
| | - F. S. Moreno
- University of São Paulo, Faculty of Pharmaceutical Sciences, Department of Food and Experimental Nutrition, Food Research Center (FoRC) São Paulo 05508-000 Brazil
| |
Collapse
|
1335
|
Qiao Y, Wang J, Karagoz E, Liang B, Song X, Shang R, Evert K, Xu M, Che L, Evert M, Calvisi DF, Tao J, Wang B, Monga SP, Chen X. Axis inhibition protein 1 (Axin1) Deletion-Induced Hepatocarcinogenesis Requires Intact β-Catenin but Not Notch Cascade in Mice. Hepatology 2019; 70:2003-2017. [PMID: 30737831 PMCID: PMC7206928 DOI: 10.1002/hep.30556] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/02/2019] [Indexed: 12/12/2022]
Abstract
Inactivating mutations of axis inhibition protein 1 (AXIN1), a negative regulator of the Wnt/β-Catenin cascade, are among the common genetic events in human hepatocellular carcinoma (HCC), affecting approximately 10% of cases. In the present manuscript, we sought to define the genetic crosstalk between Axin1 mutants and Wnt/β-catenin as well as Notch signaling cascades along hepatocarcinogenesis. We discovered that c-MET activation and AXIN1 mutations occur concomitantly in ~3%-5% of human HCC samples. Subsequently, we generated a murine HCC model by means of CRISPR/Cas9-based gene deletion of Axin1 (sgAxin1) in combination with transposon-based expression of c-Met in the mouse liver (c-Met/sgAxin1). Global gene expression analysis of mouse normal liver, HCCs induced by c-Met/sgAxin1, and HCCs induced by c-Met/∆N90-β-Catenin revealed activation of the Wnt/β-Catenin and Notch signaling in c-Met/sgAxin1 HCCs. However, only a few of the canonical Wnt/β-Catenin target genes were induced in c-Met/sgAxin1 HCC when compared with corresponding lesions from c-Met/∆N90-β-Catenin mice. To study whether endogenous β-Catenin is required for c-Met/sgAxin1-driven HCC development, we expressed c-Met/sgAxin1 in liver-specific Ctnnb1 null mice, which completely prevented HCC development. Consistently, in AXIN1 mutant or null human HCC cell lines, silencing of β-Catenin strongly inhibited cell proliferation. In striking contrast, blocking the Notch cascade through expression of either the dominant negative form of the recombinant signal-binding protein for immunoglobulin kappa J region (RBP-J) or the ablation of Notch2 did not significantly affect c-Met/sgAxin1-driven hepatocarcinogenesis. Conclusion: We demonstrated here that loss of Axin1 cooperates with c-Met to induce HCC in mice, in a β-Catenin signaling-dependent but Notch cascade-independent way.
Collapse
Affiliation(s)
- Yu Qiao
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China;,Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA
| | - Jingxiao Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Eylul Karagoz
- Department of Medicine and Liver Center, University of California, San Francisco, CA;,School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Binyong Liang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinhua Song
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Runze Shang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Military Medical University, Xi’an, China
| | - Katja Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Meng Xu
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Diego F. Calvisi
- Institute of Pathology, University Medicine Greifswald, Greifswald, Germany
| | - Junyan Tao
- Department of Pathology, University of Pittsburgh School of Medicine, and Pittsburgh Liver Research Center, Pittsburgh, PA
| | - Bruce Wang
- Department of Medicine and Liver Center, University of California, San Francisco, CA
| | - Satdarshan P. Monga
- Department of Pathology, University of Pittsburgh School of Medicine, and Pittsburgh Liver Research Center, Pittsburgh, PA
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA
| |
Collapse
|
1336
|
Detecting viral sequences in NGS data. Curr Opin Virol 2019; 39:41-48. [DOI: 10.1016/j.coviro.2019.07.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 01/03/2023]
|
1337
|
Ding X, He M, Chan AWH, Song QX, Sze SC, Chen H, Man MKH, Man K, Chan SL, Lai PBS, Wang X, Wong N. Genomic and Epigenomic Features of Primary and Recurrent Hepatocellular Carcinomas. Gastroenterology 2019; 157:1630-1645.e6. [PMID: 31560893 DOI: 10.1053/j.gastro.2019.09.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Intratumor heterogeneity and divergent clonal lineages within and among primary and recurrent hepatocellular carcinomas (HCCs) produce challenges to patient management. We investigated genetic and epigenetic variations within liver tumors, among hepatic lesions, and between primary and relapsing tumors. METHODS Tumor and matched nontumor liver specimens were collected from 113 patients who underwent partial hepatectomy for primary or recurrent HCC at 2 hospitals in Hong Kong. We performed whole-genome, whole-exome, or targeted capture sequencing analyses of 356 HCC specimens collected from multiple tumor regions and matched initial and recurrent tumors. We performed parallel DNA methylation profiling analyses of 95 specimens. Genomes and epigenomes of nontumor tissues that contained areas of cirrhosis or fibrosis were analyzed. We developed liver cancer cell lines that endogenously expressed a mutant form of TP53 (R249S) or overexpressed mutant forms of STAT3 (D170Y, K348E, and Y640F) or JAK1 (S703I and L910P) and tested the abilities of pharmacologic agents to reduce activity. Cells were analyzed by immunoblotting and chromatin immunoprecipitation with quantitative polymerase chain reaction. RESULTS We determined the monoclonal origins of individual tumors using a single sample collection approach that captured more than 90% of mutations that are detected in all regions of tumors. Phylogenetic and phylo-epigenetic analyses revealed interactions and codependence between the genomic and epigenomic features of HCCs. Methylation analysis revealed a field effect in cirrhotic liver tissues that predisposes them to tumor development. Comparisons of genetic features revealed that 52% of recurrent HCCs derive from the clonal lineage of the initial tumor. The clonal origin if recurrent HCCs allowed construction of a temporal map of genetic alterations that associated with tumor recurrence. Activation of JAK signaling to STAT was a characteristic of HCC progression via mutations that associate with response to drug sensitivity. The combination of a mutation that increases the function of TP53 and the 17p chromosome deletion might provide liver cancer cells with a replicative advantage. Chromatin immunoprecipitation analysis of TP53 with the R249S substitution revealed its interaction with genes that encode chromatin regulators (MLL1 and MLL2). We validated MLL1 and MLL2 as direct targets of TP53R249S and affirmed their association in the Cancer Genome Atlas dataset. The MLL-complex antagonists MI-2-2 (inhibitor of protein interaction) and OICR-9492 (inhibitor of activity) specifically inhibited proliferation of HCC cells that express TP53R249S at nanomolar concentrations. CONCLUSIONS We performed a systematic evaluation of intra- and intertumor genetic heterogeneity in HCC samples and identified genetic and epigenetic changes that associate with tumor progression and recurrence. We identified chromatin regulators that are upregulated by mutant TP53 in HCC cells and inhibitors that reduce proliferation of these cells. DNA methylation patterns in cirrhotic or fibrotic liver tissues might be used to identify those at risk of HCC development.
Collapse
Affiliation(s)
- Xiaofan Ding
- Department of Anatomical and Cellular Pathology at Sir Y.K. Pao Centre for Cancer, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Mian He
- Department of Anatomical and Cellular Pathology at Sir Y.K. Pao Centre for Cancer, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Anthony W H Chan
- Department of Anatomical and Cellular Pathology at Sir Y.K. Pao Centre for Cancer, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Qi Xiu Song
- Department of Anatomical and Cellular Pathology at Sir Y.K. Pao Centre for Cancer, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Siu Ching Sze
- Department of Anatomical and Cellular Pathology at Sir Y.K. Pao Centre for Cancer, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Hui Chen
- Department of Anatomical and Cellular Pathology at Sir Y.K. Pao Centre for Cancer, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Matthew K H Man
- Department of Anatomical and Cellular Pathology at Sir Y.K. Pao Centre for Cancer, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Kwan Man
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Stephen L Chan
- Department of Clinical Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Paul B S Lai
- Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Nathalie Wong
- Department of Anatomical and Cellular Pathology at Sir Y.K. Pao Centre for Cancer, The Chinese University of Hong Kong, Shatin, Hong Kong, China; Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong, China; State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, Hong Kong, China; State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
1338
|
Satriano L, Lewinska M, Rodrigues PM, Banales JM, Andersen JB. Metabolic rearrangements in primary liver cancers: cause and consequences. Nat Rev Gastroenterol Hepatol 2019; 16:748-766. [PMID: 31666728 DOI: 10.1038/s41575-019-0217-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
Primary liver cancer (PLC) is the fourth most frequent cause of cancer-related death. The high mortality rates arise from late diagnosis and the limited accuracy of diagnostic and prognostic biomarkers. The liver is a major regulator, orchestrating the clearance of toxins, balancing glucose, lipid and amino acid uptake, managing whole-body metabolism and maintaining metabolic homeostasis. Tumour onset and progression is frequently accompanied by rearrangements of metabolic pathways, leading to dysregulation of metabolism. The limitation of current therapies targeting PLCs, such as hepatocellular carcinoma and cholangiocarcinoma, points towards the importance of deciphering this metabolic complexity. In this Review, we discuss the role of metabolic liver disruptions and the implications of these processes in PLCs, emphasizing their clinical relevance and value in early diagnosis and prognosis and as putative therapeutic targets. We also describe system biology approaches able to reconstruct the metabolic complexity of liver diseases. We also discuss whether metabolic rearrangements are a cause or consequence of PLCs, emphasizing the opportunity to clinically exploit the rewired metabolism. In line with this idea, we discuss circulating metabolites as promising biomarkers for PLCs.
Collapse
Affiliation(s)
- Letizia Satriano
- Biotech Research and Innovation Centre (BRIC) Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Monika Lewinska
- Biotech Research and Innovation Centre (BRIC) Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pedro M Rodrigues
- Biodonostia Health Research Institute, Donostia University Hospital, San Sebastian, Spain
| | - Jesus M Banales
- Biodonostia Health Research Institute, Donostia University Hospital, San Sebastian, Spain.,National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC) Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
1339
|
Hilmi M, Neuzillet C, Calderaro J, Lafdil F, Pawlotsky JM, Rousseau B. Angiogenesis and immune checkpoint inhibitors as therapies for hepatocellular carcinoma: current knowledge and future research directions. J Immunother Cancer 2019; 7:333. [PMID: 31783782 PMCID: PMC6884868 DOI: 10.1186/s40425-019-0824-5] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/13/2019] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second deadliest cancer worldwide, due to its high incidence and poor prognosis. Frequent initial presentation at advanced stages along with impaired liver function limit the use of a broad therapeutic arsenal in patients with HCC. Although main HCC oncogenic drivers have been deciphered in recent years (TERT, TP53, CTNNB1 mutations, miR122 and CDKN2A silencing), therapeutic applications derived from this molecular knowledge are still limited. Given its high vascularization and immunogenicity, antiangiogenics and immune checkpoint inhibitors (ICI), respectively, are two therapeutic approaches that have shown efficacy in HCC. Depending on HCC immune profile, combinations of these therapies aim to modify the protumoral/antitumoral immune balance, and to reactivate and favor the intratumoral trafficking of cytotoxic T cells. Combination therapies involving antiangiogenics and ICI may be synergistic, because vascular endothelial growth factor A inhibition increases intratumoral infiltration and survival of cytotoxic T lymphocytes and decreases regulatory T lymphocyte recruitment, resulting in a more favorable immune microenvironment for ICI antitumoral activity. First results from clinical trials evaluating combinations of these therapies are encouraging with response rates never observed before in patients with HCC. A better understanding of the balance and interactions between protumoral and antitumoral immune cells will help to ensure the success of future therapeutic trials. Here, we present an overview of the current state of clinical development of antitumoral therapies in HCC and the biological rationale for their use. Moreover, translational studies on tumor tissue and blood, prior to and during treatment, will help to identify biomarkers and immune signatures with predictive value for both clinical outcome and response to combination therapies.
Collapse
MESH Headings
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/adverse effects
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/antagonists & inhibitors
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/pathology
- Clinical Trials as Topic
- Combined Modality Therapy
- Disease Susceptibility
- Humans
- Immunomodulation/drug effects
- Liver Neoplasms/drug therapy
- Liver Neoplasms/etiology
- Liver Neoplasms/pathology
- Molecular Targeted Therapy
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/metabolism
- Treatment Outcome
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Marc Hilmi
- Department of Medical Oncology, Curie Institute, University of Versailles Saint-Quentin, Paris, France
| | - Cindy Neuzillet
- Department of Medical Oncology, Curie Institute, University of Versailles Saint-Quentin, Paris, France
| | - Julien Calderaro
- Department of Pathology, Henri Mondor Hospital, Créteil, France
- IMRB-INSERM U955 Team 18, Créteil, France
- Université Paris-Est-Créteil, Créteil, France
| | - Fouad Lafdil
- IMRB-INSERM U955 Team 18, Créteil, France
- Université Paris-Est-Créteil, Créteil, France
- Institut Universitaire de France, Paris, France
| | - Jean-Michel Pawlotsky
- IMRB-INSERM U955 Team 18, Créteil, France
- Université Paris-Est-Créteil, Créteil, France
- National Reference Center for Viral Hepatitis B, C and D, Department of Virology, Henri Mondor Hospital, Créteil, France
| | - Benoit Rousseau
- IMRB-INSERM U955 Team 18, Créteil, France.
- Department of Medicine, Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
1340
|
Hausser J, Szekely P, Bar N, Zimmer A, Sheftel H, Caldas C, Alon U. Tumor diversity and the trade-off between universal cancer tasks. Nat Commun 2019; 10:5423. [PMID: 31780652 PMCID: PMC6882839 DOI: 10.1038/s41467-019-13195-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/11/2019] [Indexed: 02/06/2023] Open
Abstract
Recent advances have enabled powerful methods to sort tumors into prognosis and treatment groups. We are still missing, however, a general theoretical framework to understand the vast diversity of tumor gene expression and mutations. Here we present a framework based on multi-task evolution theory, using the fact that tumors need to perform multiple tasks that contribute to their fitness. We find that trade-offs between tasks constrain tumor gene-expression to a continuum bounded by a polyhedron whose vertices are gene-expression profiles, each specializing in one task. We find five universal cancer tasks across tissue-types: cell-division, biomass and energy, lipogenesis, immune-interaction and invasion and tissue-remodeling. Tumors that specialize in a task are sensitive to drugs that interfere with this task. Driver, but not passenger, mutations tune gene-expression towards specialization in specific tasks. This approach can integrate additional types of molecular data into a framework of tumor diversity grounded in evolutionary theory.
Collapse
Affiliation(s)
- Jean Hausser
- Department of Molecular Cell Biology, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Pablo Szekely
- Department of Molecular Cell Biology, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Noam Bar
- Department of Molecular Cell Biology, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Anat Zimmer
- Department of Molecular Cell Biology, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Hila Sheftel
- Department of Molecular Cell Biology, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Carlos Caldas
- Department of Oncology and Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK.
- Breast Cancer Programme, Cancer Research UK Cambridge Cancer Centre, Cambridge, CB2 0RE, UK.
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
1341
|
Sathyanarayanan A, Gupta R, Thompson EW, Nyholt DR, Bauer DC, Nagaraj SH. A comparative study of multi-omics integration tools for cancer driver gene identification and tumour subtyping. Brief Bioinform 2019; 21:1920-1936. [PMID: 31774481 PMCID: PMC7711266 DOI: 10.1093/bib/bbz121] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 09/09/2019] [Accepted: 09/13/2019] [Indexed: 12/11/2022] Open
Abstract
Oncogenesis and cancer can arise as a consequence of a wide range of genomic aberrations including mutations, copy number alterations, expression changes and epigenetic modifications encompassing multiple omics layers. Integrating genomic, transcriptomic, proteomic and epigenomic datasets via multi-omics analysis provides the opportunity to derive a deeper and holistic understanding of the development and progression of cancer. There are two primary approaches to integrating multi-omics data: multi-staged (focused on identifying genes driving cancer) and meta-dimensional (focused on establishing clinically relevant tumour or sample classifications). A number of ready-to-use bioinformatics tools are available to perform both multi-staged and meta-dimensional integration of multi-omics data. In this study, we compared nine different integration tools using real and simulated cancer datasets. The performance of the multi-staged integration tools were assessed at the gene, function and pathway levels, while meta-dimensional integration tools were assessed based on the sample classification performance. Additionally, we discuss the influence of factors such as data representation, sample size, signal and noise on multi-omics data integration. Our results provide current and much needed guidance regarding selection and use of the most appropriate and best performing multi-omics integration tools.
Collapse
Affiliation(s)
- Anita Sathyanarayanan
- School of Biomedical Sciences, Faculty of Health, and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Rohit Gupta
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India.,Department of Computational Biology, Indraprastha Institute of Information Technology, Delhi, India
| | - Erik W Thompson
- School of Biomedical Sciences, Faculty of Health, and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Brisbane, Australia
| | - Dale R Nyholt
- School of Biomedical Sciences, Faculty of Health, and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | | | - Shivashankar H Nagaraj
- School of Biomedical Sciences, Faculty of Health, and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Brisbane, Australia
| |
Collapse
|
1342
|
Kamimura K, Yokoo T, Abe H, Terai S. Gene Therapy for Liver Cancers: Current Status from Basic to Clinics. Cancers (Basel) 2019; 11:1865. [PMID: 31769427 PMCID: PMC6966544 DOI: 10.3390/cancers11121865] [Citation(s) in RCA: 23] [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: 10/30/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023] Open
Abstract
The liver is a key organ for metabolism, protein synthesis, detoxification, and endocrine function, and among liver diseases, including hepatitis, cirrhosis, malignant tumors, and congenital disease, liver cancer is one of the leading causes of cancer-related deaths worldwide. Conventional therapeutic options such as embolization and chemotherapy are not effective against advanced-stage liver cancer; therefore, continuous efforts focus on the development of novel therapeutic options, including molecular targeted agents and gene therapy. In this review, we will summarize the progress toward the development of gene therapies for liver cancer, with an emphasis on recent clinical trials and preclinical studies.
Collapse
Affiliation(s)
- Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Aasahimachi-Dori, Chuo-Ku, Niigata 9518510, Japan; (T.Y.); (H.A.); (S.T.)
| | | | | | | |
Collapse
|
1343
|
Zhou B, Ho SS, Greer SU, Spies N, Bell JM, Zhang X, Zhu X, Arthur JG, Byeon S, Pattni R, Saha I, Huang Y, Song G, Perrin D, Wong WH, Ji HP, Abyzov A, Urban AE. Haplotype-resolved and integrated genome analysis of the cancer cell line HepG2. Nucleic Acids Res 2019; 47:3846-3861. [PMID: 30864654 PMCID: PMC6486628 DOI: 10.1093/nar/gkz169] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/19/2019] [Accepted: 03/01/2019] [Indexed: 12/19/2022] Open
Abstract
HepG2 is one of the most widely used human cancer cell lines in biomedical research and one of the main cell lines of ENCODE. Although the functional genomic and epigenomic characteristics of HepG2 are extensively studied, its genome sequence has never been comprehensively analyzed and higher order genomic structural features are largely unknown. The high degree of aneuploidy in HepG2 renders traditional genome variant analysis methods challenging and partially ineffective. Correct and complete interpretation of the extensive functional genomics data from HepG2 requires an understanding of the cell line’s genome sequence and genome structure. Using a variety of sequencing and analysis methods, we identified a wide spectrum of genome characteristics in HepG2: copy numbers of chromosomal segments at high resolution, SNVs and Indels (corrected for aneuploidy), regions with loss of heterozygosity, phased haplotypes extending to entire chromosome arms, retrotransposon insertions and structural variants (SVs) including complex and somatic genomic rearrangements. A large number of SVs were phased, sequence assembled and experimentally validated. We re-analyzed published HepG2 datasets for allele-specific expression and DNA methylation and assembled an allele-specific CRISPR/Cas9 targeting map. We demonstrate how deeper insights into genomic regulatory complexity are gained by adopting a genome-integrated framework.
Collapse
Affiliation(s)
- Bo Zhou
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Steve S Ho
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stephanie U Greer
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Noah Spies
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.,Genome-scale Measurements Group, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - John M Bell
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA 94304, USA
| | - Xianglong Zhang
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xiaowei Zhu
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph G Arthur
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Seunggyu Byeon
- School of Computer Science and Engineering, College of Engineering, Pusan National University, Busan 46241, South Korea
| | - Reenal Pattni
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ishan Saha
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yiling Huang
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Giltae Song
- School of Computer Science and Engineering, College of Engineering, Pusan National University, Busan 46241, South Korea
| | - Dimitri Perrin
- Science and Engineering Faculty, Queensland University of Technology, Brisbane, QLD 4001, Australia
| | - Wing H Wong
- Department of Statistics, Stanford University, Stanford, CA 94305, USA.,Department of Biomedical Data Science, Bio-X Program, Stanford University, Stanford, CA 94305, USA
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Stanford Genome Technology Center, Stanford University, Palo Alto, CA 94304, USA
| | - Alexej Abyzov
- Department of Health Sciences Research, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Alexander E Urban
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.,Tashia and John Morgridge Faculty Scholar, Stanford Child Health Research Institute, Stanford, CA 94305, USA
| |
Collapse
|
1344
|
Wang H, Liao P, Zeng SX, Lu H. Co-targeting p53-R249S and CDK4 synergistically suppresses survival of hepatocellular carcinoma cells. Cancer Biol Ther 2019; 21:269-277. [PMID: 31747859 PMCID: PMC7012101 DOI: 10.1080/15384047.2019.1685289] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 10/10/2019] [Accepted: 10/22/2019] [Indexed: 12/31/2022] Open
Abstract
p53-R249S (p53-RS) is frequently detected in human hepatocellular carcinoma (HCC) that is highly associated with hepatitis B infection and aflatoxin B1 exposure. Our previous study showed that CDK4/Cyclin D1 phosphorylates p53-RS at the cancer-derived Ser249 and promotes its interaction with c-Myc in the nucleus, consequently enhancing c-Myc-dependent ribosomal biogenesis and HCC cell proliferation. Here we explored the possibility of co-targeting CDK4 and p53-RS with available small molecule inhibitors as a potential combined therapy for HCC that harbor p53-RS. Indeed, co-treatment of p53-RS-containing, but not wild-type p53 or p53-null, HCC cells with PD-0332991 (PD), a CDK4/6 inhibitor, and CP-31398 (CP), a compound that can restore the intrinsic conformation and transcriptional activity of mutant p53, drastically repressed the c-Myc activation function of p53-RS. This combination of PD with CP exhibited a synergistic effect on the inhibition of HCC cell growth in a p53-RS dependent manner, especially at a lower dose. These results suggest that co-targeting CDK4 and p53-RS can serve as a potential approach for the development of an effective therapy for HCC that harbor p53-RS.
Collapse
Affiliation(s)
- Huai Wang
- Department of Biochemistry and Molecular Biology, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
- School of Public Health, Nanchang University, Nanchang, Jiangxi, P.R. China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Peng Liao
- Department of Biochemistry and Molecular Biology, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Shelya X. Zeng
- Department of Biochemistry and Molecular Biology, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Hua Lu
- Department of Biochemistry and Molecular Biology, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
1345
|
Gnoni A, Licchetta A, Memeo R, Argentiero A, Solimando AG, Longo V, Delcuratolo S, Brunetti O. Role of BRAF in Hepatocellular Carcinoma: A Rationale for Future Targeted Cancer Therapies. ACTA ACUST UNITED AC 2019; 55:medicina55120754. [PMID: 31766556 PMCID: PMC6956203 DOI: 10.3390/medicina55120754] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/11/2019] [Accepted: 11/15/2019] [Indexed: 01/18/2023]
Abstract
The few therapeutic strategies for advance hepatocellular carcinoma (HCC) on poor knowledge of its biology. For several years, sorafenib, a tyrosine kinase inhibitors (TKI) inhibitor, has been the approved treatment option, to date, for advanced HCC patients. Its activity is the inhibition of the retrovirus-associated DNA sequences protein (RAS)/Rapidly Accelerated Fibrosarcoma protein (RAF)/mitogen-activated and extracellular-signal regulated kinase (MEK)/extracellular-signal regulated kinases (ERK) signaling pathway. However, the efficacy of sorafenib is limited by the development of drug resistance, and the major neuronal isoform of RAF, BRAF and MEK pathways play a critical and central role in HCC escape from TKIs activity. Advanced HCC patients with a BRAF mutation display a multifocal and/or more aggressive behavior with resistance to TKI. Moreover, also long non-coding RNA (lnc-RNA) have been studied in epigenetic studies for BRAF aggressiveness in HCC. So far, lnc-RNA of BRAF could be another mechanism of cancer proliferation and TKI escape in HCC and the inhibition could become a possible strategy treatment for HCC. Moreover, recent preclinical studies and clinical trials evidence that combined treatments, involving alternative pathways, have an important role of therapy for HCC and they could bypass resistance to the following TKIs: MEK, ERKs/ribosomal protein S6 kinase 2 (RSK2), and phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR). These initial data must be confirmed in clinical studies, which are currently ongoing. Translational research discoveries could create new strategies of targeted therapy combinations, including BRAF pathway, and they could eventually bring light in new treatment of HCC.
Collapse
Affiliation(s)
- Antonio Gnoni
- Medical Oncology Unit, “S. Cuore di Gesù” Hospital, 73014 Gallipoli, Italy;
- Correspondence: (A.G.); (O.B.); Tel.: +39-338-118-5854 (A.G.)
| | | | - Riccardo Memeo
- Department of Surgery and Liver Transplantation, Policlinico di Bari, 70124 Bari, Italy;
| | - Antonella Argentiero
- Medical Oncology Unit, National Cancer Research Centre, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Antonio G. Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy;
| | - Vito Longo
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco, 65, 70124 Bari, Italy;
| | - Sabina Delcuratolo
- Scientific direction, National Cancer Research Centre, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Oronzo Brunetti
- Medical Oncology Unit, National Cancer Research Centre, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
- Correspondence: (A.G.); (O.B.); Tel.: +39-338-118-5854 (A.G.)
| |
Collapse
|
1346
|
Morton AR, Dogan-Artun N, Faber ZJ, MacLeod G, Bartels CF, Piazza MS, Allan KC, Mack SC, Wang X, Gimple RC, Wu Q, Rubin BP, Shetty S, Angers S, Dirks PB, Sallari RC, Lupien M, Rich JN, Scacheri PC. Functional Enhancers Shape Extrachromosomal Oncogene Amplifications. Cell 2019; 179:1330-1341.e13. [PMID: 31761532 DOI: 10.1016/j.cell.2019.10.039] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 09/20/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022]
Abstract
Non-coding regions amplified beyond oncogene borders have largely been ignored. Using a computational approach, we find signatures of significant co-amplification of non-coding DNA beyond the boundaries of amplified oncogenes across five cancer types. In glioblastoma, EGFR is preferentially co-amplified with its two endogenous enhancer elements active in the cell type of origin. These regulatory elements, their contacts, and their contribution to cell fitness are preserved on high-level circular extrachromosomal DNA amplifications. Interrogating the locus with a CRISPR interference screening approach reveals a diversity of additional elements that impact cell fitness. The pattern of fitness dependencies mirrors the rearrangement of regulatory elements and accompanying rewiring of the chromatin topology on the extrachromosomal amplicon. Our studies indicate that oncogene amplifications are shaped by regulatory dependencies in the non-coding genome.
Collapse
Affiliation(s)
- Andrew R Morton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Nergiz Dogan-Artun
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Zachary J Faber
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Graham MacLeod
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Cynthia F Bartels
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Megan S Piazza
- Center for Human Genetics Laboratory, University Hospitals, Cleveland, OH 44106, USA
| | - Kevin C Allan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Stephen C Mack
- Department of Pediatrics, Division of Hematology and Oncology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Xiuxing Wang
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Ryan C Gimple
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH 44120, USA
| | - Qiulian Wu
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Brian P Rubin
- Departments of Anatomic Pathology and Molecular Genetics, Cleveland Clinic, Lerner Research Institute and Taussig Cancer Center, Cleveland, OH 44195, USA
| | - Shashirekha Shetty
- Center for Human Genetics Laboratory, University Hospitals, Cleveland, OH 44106, USA
| | - Stephane Angers
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Peter B Dirks
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | | | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jeremy N Rich
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92037, USA.
| | - Peter C Scacheri
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA.
| |
Collapse
|
1347
|
Li X, Wang H, Li T, Wang L, Wu X, Liu J, Xu Y, Wei W. Circulating tumor DNA/circulating tumor cells and the applicability in different causes induced hepatocellular carcinoma. Curr Probl Cancer 2019; 44:100516. [PMID: 31836136 DOI: 10.1016/j.currproblcancer.2019.100516] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/31/2019] [Accepted: 09/30/2019] [Indexed: 12/25/2022]
Abstract
In 2015, liquid biopsy was rated one of the top 10 breakthrough technologies of the year by MIT Technology Review. Liquid biopsy is a type of in vitro diagnostic method involving a noninvasive blood test. It is also a breakthrough technology used to detect tumors and cancers and assist in therapeutic strategies. The most widely used markers are circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). Primary carcinoma of the liver is a malignancy of hepatocytes or intrahepatic biliary epithelial cells. The most common type of liver cancer is hepatocellular carcinoma (HCC), the causes of which mainly include infection with hepatitis B virus (HBV) and/or hepatitis C virus (HCV), alcohol abuse, aflatoxicosis, and nonalcoholic fatty liver disease/ nonalcoholic steatohepatitis. As there are few typical clinical characteristics during the early stage of the disease, early diagnosis of HCC is very challenging. However, CTCs and ctDNA carry tumor-specific information. Therefore, the detection and analysis of CTCs and ctDNA can provide evidence for the early diagnosis of HCC and guide treatment. Furthermore, several studies have indicated that different inducers of HCC cause different DNA mutations, and accordingly, detection of specific mutations in ctDNA will facilitate the determination of the HCC type and help physicians provide distinctive therapies.
Collapse
Affiliation(s)
- Xuemei Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huihui Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tao Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Anti-inflammatory and Immune Medicine innovation team, Hefei, China.
| | - Lianzi Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xian Wu
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiaqing Liu
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuanhong Xu
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Anti-inflammatory and Immune Medicine innovation team, Hefei, China.
| |
Collapse
|
1348
|
Kwon SM, Budhu A, Woo HG, Chaisaingmongkol J, Dang H, Forgues M, Harris CC, Zhang G, Auslander N, Ruppin E, Mahidol C, Ruchirawat M, Wang XW. Functional Genomic Complexity Defines Intratumor Heterogeneity and Tumor Aggressiveness in Liver Cancer. Sci Rep 2019; 9:16930. [PMID: 31729408 PMCID: PMC6858353 DOI: 10.1038/s41598-019-52578-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation and chromosome aneuploidy are major traits of primary liver cancer (PLC), which represent the second most common cause of cancer-related death worldwide. Increased cancer fitness and aggressiveness of PLC may be achieved by enhancing tumoral genomic complexity that alters tumor biology. Here, we developed a scoring method, namely functional genomic complexity (FGC), to determine the degree of molecular heterogeneity among 580 liver tumors with diverse ethnicities and etiologies by assessing integrated genomic and transcriptomic data. We found that tumors with higher FGC scores are associated with chromosome instability and TP53 mutations, and a worse prognosis, while tumors with lower FGC scores have elevated infiltrating lymphocytes and a better prognosis. These results indicate that FGC scores may serve as a surrogate to define genomic heterogeneity of PLC linked to chromosomal instability and evasion of immune surveillance. Our findings demonstrate an ability to define genomic heterogeneity and corresponding tumor biology of liver cancer based only on bulk genomic and transcriptomic data. Our data also provide a rationale for applying this approach to survey liver tumor immunity and to stratify patients for immune-based therapy.
Collapse
Affiliation(s)
- So Mee Kwon
- Laboratory of Human Carcinogenesis and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892, USA
- Department of Physiology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Anuradha Budhu
- Laboratory of Human Carcinogenesis and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Hyun Goo Woo
- Laboratory of Human Carcinogenesis and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892, USA
- Department of Biomedical Science, Graduate School, Ajou University, Suwon, 16499, Republic of Korea
| | - Jittiporn Chaisaingmongkol
- Laboratory of Human Carcinogenesis and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892, USA
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok, 10210, Thailand
- Center of Excellence on Environmental Health and Toxicology, Office of the Higher Education Commission, Ministry of Education, Bangkok, 10400, Thailand
| | - Hien Dang
- Laboratory of Human Carcinogenesis and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Marshonna Forgues
- Laboratory of Human Carcinogenesis and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892, USA
| | - Gao Zhang
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Noam Auslander
- Cancer Data Science Lab, National Cancer Institute, National Institute of health, MD, 20892, USA
| | - Eytan Ruppin
- Cancer Data Science Lab, National Cancer Institute, National Institute of health, MD, 20892, USA
| | - Chulabhorn Mahidol
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Mathuros Ruchirawat
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok, 10210, Thailand
- Center of Excellence on Environmental Health and Toxicology, Office of the Higher Education Commission, Ministry of Education, Bangkok, 10400, Thailand
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, 20892, USA.
| |
Collapse
|
1349
|
Wang D, Zhu Y, Tang J, Lian Q, Luo G, Wen W, Zhang MQ, Wang H, Chen L, Gu J. Integrative molecular analysis of metastatic hepatocellular carcinoma. BMC Med Genomics 2019; 12:164. [PMID: 31722693 PMCID: PMC6854708 DOI: 10.1186/s12920-019-0586-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the major type of primary liver cancer. Intrahepatic metastasis, such as portal vein tumor thrombosis (PVTT), strongly indicates poor prognosis of HCC. But now, there are limited understandings of the molecular features and mechanisms of those metastatic HCCs. Methods To characterize the molecular alterations of the metastatic HCCs, we implemented an integrative analysis of the copy number variations (CNVs), DNA methylations and transcriptomes of matched adjacent normal, primary tumor and PVTT samples from 19 HCC patients. Results CNV analysis identified a frequently amplified focal region chr11q13.3 and a novel deletion peak chr19q13.41 containing three miRNAs. The integrative analysis with RNA-seq data suggests that CNVs and differential promoter methylations regulate distinct oncogenic processes. Then, we used individualized differential analysis to identify the differentially expressed genes between matched primary tumor and PVTT of each patient. Results show that 5 out of 19 studied patients acquire evidential progressive alterations of gene expressions (more than 1000 differentially expressed genes were identified in each patient). While, another subset of eight patients have nearly identical gene expressions between the corresponding matched primary tumor and PVTT. Twenty genes were found to be recurrently and progressively differentially expressed in multiple patients. These genes are mainly associated with focal adhesion, xenobiotics metabolism by cytochrome P450 and amino acid metabolism. For several differentially expressed genes in metabolic pathways, their expressions are significantly associated with overall survivals and vascular invasions of HCC patients. The following transwell assay experiments validate that they can regulate invasive phenotypes of HCC cells. Conclusions The metastatic HCCs with PVTTs have significant molecular alterations comparing with adjacent normal tissues. The recurrent alteration patterns are similar to several previously published general HCC cohorts, but usually with higher severity. By an individualized differential analysis strategy, the progressively differentially expressed genes between the primary tumor and PVTT were identified for each patient. A few patients aquire evidential progressive alterations of gene expressions. And, experiments show that several recurrently differentially expressed genes can strongly regulate HCC cell invasions.
Collapse
Affiliation(s)
- Dongfang Wang
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Yanjing Zhu
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, 200438, China
| | - Jing Tang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, 200438, China.,Department of Neurosurgery, Wuhan General Hospital, 627 Wuluo Road, Wuhan, 430070, China
| | - Qiuyu Lian
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Guijuan Luo
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, 200438, China
| | - Wen Wen
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, 200438, China
| | - Michael Q Zhang
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, 100084, China.,Department of Biological Sciences, Center for Systems Biology, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Hongyang Wang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, 200438, China.
| | - Lei Chen
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, 200438, China.
| | - Jin Gu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
1350
|
Desterke C, Chiappini F. Lipid Related Genes Altered in NASH Connect Inflammation in Liver Pathogenesis Progression to HCC: A Canonical Pathway. Int J Mol Sci 2019; 20:ijms20225594. [PMID: 31717414 PMCID: PMC6888337 DOI: 10.3390/ijms20225594] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/03/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is becoming a public health problem worldwide. While the number of research studies on NASH progression rises every year, sometime their findings are controversial. To identify the most important and commonly described findings related to NASH progression, we used an original bioinformatics, integrative, text-mining approach that combines PubMed database querying and available gene expression omnibus dataset. We have identified a signature of 25 genes that are commonly found to be dysregulated during steatosis progression to NASH and cancer. These genes are implicated in lipid metabolism, insulin resistance, inflammation, and cancer. They are functionally connected, forming the basis necessary for steatosis progression to NASH and further progression to hepatocellular carcinoma (HCC). We also show that five of the identified genes have genome alterations present in HCC patients. The patients with these genes associated to genome alteration are associated with a poor prognosis. In conclusion, using an integrative literature- and data-mining approach, we have identified and described a canonical pathway underlying progression of NASH. Other parameters (e.g., polymorphisms) can be added to this pathway that also contribute to the progression of the disease to cancer. This work improved our understanding of the molecular basis of NASH progression and will help to develop new therapeutic approaches.
Collapse
Affiliation(s)
| | - Franck Chiappini
- Laboratoire Croissance, Régénération, Réparation et Régénération Tissulaires (CRRET)/ EAC CNRS 7149, Univ Paris-Est Créteil (UPEC), F-94010 Créteil, France
- Correspondence: ; Tel.: +33-(0)1-45177080; Fax: +33-(0)1-45171816
| |
Collapse
|