1
|
Airola C, Pallozzi M, Cesari E, Cerrito L, Stella L, Sette C, Giuliante F, Gasbarrini A, Ponziani FR. Hepatocellular-Carcinoma-Derived Organoids: Innovation in Cancer Research. Cells 2024; 13:1726. [PMID: 39451244 PMCID: PMC11505656 DOI: 10.3390/cells13201726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Hepatocellular carcinomas (HCCs) are highly heterogeneous malignancies. They are characterized by a peculiar tumor microenvironment and dense vascularization. The importance of signaling between immune cells, endothelial cells, and tumor cells leads to the difficult recapitulation of a reliable in vitro HCC model using the conventional two-dimensional cell cultures. The advent of three-dimensional organoid tumor technology has revolutionized our understanding of the pathogenesis and progression of several malignancies by faithfully replicating the original cancer genomic, epigenomic, and microenvironmental landscape. Organoids more closely mimic the in vivo environment and cell interactions, replicating factors such as the spatial organization of cell surface receptors and gene expression, and will probably become an important tool in the choice of therapies and the evaluation of tumor response to treatments. This review aimed to describe the ongoing and potential applications of organoids as an in vitro model for the study of HCC development, its interaction with the host's immunity, the analysis of drug sensitivity tests, and the current limits in this field.
Collapse
Affiliation(s)
- Carlo Airola
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Maria Pallozzi
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Eleonora Cesari
- GSTeP Organoids Research Core Facility, Fondazione Policlinico A. Gemelli, 00168 Rome, Italy; (E.C.); (C.S.)
| | - Lucia Cerrito
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Leonardo Stella
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
| | - Claudio Sette
- GSTeP Organoids Research Core Facility, Fondazione Policlinico A. Gemelli, 00168 Rome, Italy; (E.C.); (C.S.)
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Felice Giuliante
- Department of Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
2
|
Ladd AD, Duarte S, Sahin I, Zarrinpar A. Mechanisms of drug resistance in HCC. Hepatology 2024; 79:926-940. [PMID: 36680397 DOI: 10.1097/hep.0000000000000237] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 11/21/2022] [Indexed: 01/22/2023]
Abstract
HCC comprises ∼80% of primary liver cancer. HCC is the only major cancer for which death rates have not improved over the last 10 years. Most patients are diagnosed with advanced disease when surgical and locoregional treatments are not feasible or effective. Sorafenib, a multikinase inhibitor targeting cell growth and angiogenesis, was approved for advanced unresectable HCC in 2007. Since then, other multikinase inhibitors have been approved. Lenvatinib was found to be noninferior to sorafenib as a first-line agent. Regorafenib, cabozantinib, and ramucirumab were shown to prolong survival as second-line agents. Advances in immunotherapy for HCC have also added hope for patients, but their efficacy remains limited. A large proportion of patients with advanced HCC gain no long-term benefit from systemic therapy due to primary and acquired drug resistance, which, combined with its rising incidence, keeps HCC a highly fatal disease. This review summarizes mechanisms of primary and acquired resistance to therapy and includes methods for bypassing resistance. It addresses recent advancements in immunotherapy, provides new perspectives on the linkage between drug resistance and molecular etiology of HCC, and evaluates the role of the microbiome in drug resistance. It also discusses alterations in signaling pathways, dysregulation of apoptosis, modulations in the tumor microenvironment, involvement of cancer stem cells, changes in drug metabolism/transport, tumor hypoxia, DNA repair, and the role of microRNAs in drug resistance. Understanding the interplay among these factors will provide guidance on the development of new therapeutic strategies capable of improving patient outcomes.
Collapse
Affiliation(s)
- Alexandra D Ladd
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Sergio Duarte
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ilyas Sahin
- Division of Hematology/Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ali Zarrinpar
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
3
|
Desoteux M, Maillot B, Bévant K, Ferlier T, Leroux R, Angenard G, Louis C, Sulpice L, Boudjema K, Coulouarn C. Transcriptomic evidence for tumor-specific beneficial or adverse effects of TGFβ pathway inhibition on the prognosis of patients with liver cancer. FEBS Open Bio 2023; 13:1278-1290. [PMID: 37195148 PMCID: PMC10315808 DOI: 10.1002/2211-5463.13647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/03/2023] [Accepted: 05/16/2023] [Indexed: 05/18/2023] Open
Abstract
Therapeutic targeting of the transforming growth factor beta (TGFβ) pathway in cancer represents a clinical challenge since TGFβ exhibits either tumor suppressive or tumor promoting properties, depending on the tumor stage. Thus, treatment with galunisertib, a small molecule inhibitor of TGFβ receptor type 1, demonstrated clinical benefits only in subsets of patients. Due to the functional duality of TGFβ in cancer, one can hypothesize that inhibiting this pathway could result in beneficial or adverse effects depending on tumor subtypes. Here, we report distinct gene expression signatures in response to galunisertib in PLC/PRF/5 and SNU-449, two cell lines that recapitulate human hepatocellular carcinoma (HCC) with good and poor prognosis, respectively. More importantly, integrative transcriptomics using independent cohorts of patients with HCC demonstrates that galunisertib-induced transcriptional reprogramming in SNU-449 is associated with human HCC with a better clinical outcome (i.e., increased overall survival), while galunisertib-induced transcriptional reprogramming in PLC/PRF/5 is associated with human HCC with a worse clinical outcome (i.e., reduced overall survival), demonstrating that galunisertib could indeed be beneficial or detrimental depending on HCC subtypes. Collectively, our study highlights the importance of patient selection to demonstrate a clinical benefit of TGFβ pathway inhibition and identifies Serpin Family F Member 2 (SERPINF2) as a putative companion biomarker for galunisertib in HCC.
Collapse
Affiliation(s)
- Matthis Desoteux
- Inserm, UMR_S 1242, OSS (Oncogenesis Stress Signaling)Centre de Lutte contre le Cancer Eugène Marquis, Univ RennesFrance
| | - Betty Maillot
- Department of Hepatobiliary and Digestive SurgeryRennes University HospitalFrance
| | - Kevin Bévant
- Inserm, UMR_S 1242, OSS (Oncogenesis Stress Signaling)Centre de Lutte contre le Cancer Eugène Marquis, Univ RennesFrance
| | - Tanguy Ferlier
- Inserm, UMR_S 1242, OSS (Oncogenesis Stress Signaling)Centre de Lutte contre le Cancer Eugène Marquis, Univ RennesFrance
| | - Raffaële Leroux
- Inserm, UMR_S 1242, OSS (Oncogenesis Stress Signaling)Centre de Lutte contre le Cancer Eugène Marquis, Univ RennesFrance
| | - Gaëlle Angenard
- Inserm, Inrae, UMR_S 1317NuMeCan (Nutrition, Metabolisms and Cancer), Univ RennesFrance
| | - Corentin Louis
- Inserm, UMR_S 1242, OSS (Oncogenesis Stress Signaling)Centre de Lutte contre le Cancer Eugène Marquis, Univ RennesFrance
| | - Laurent Sulpice
- Inserm, UMR_S 1242, OSS (Oncogenesis Stress Signaling)Centre de Lutte contre le Cancer Eugène Marquis, Univ RennesFrance
- Department of Hepatobiliary and Digestive SurgeryRennes University HospitalFrance
- Clinical Investigation Center 1414CHU de RennesFrance
| | - Karim Boudjema
- Department of Hepatobiliary and Digestive SurgeryRennes University HospitalFrance
- Clinical Investigation Center 1414CHU de RennesFrance
| | - Cédric Coulouarn
- Inserm, UMR_S 1242, OSS (Oncogenesis Stress Signaling)Centre de Lutte contre le Cancer Eugène Marquis, Univ RennesFrance
| |
Collapse
|
4
|
Sharma S, Rawal P, Kaur S, Puria R. Liver organoids as a primary human model to study HBV-mediated Hepatocellular carcinoma. A review. Exp Cell Res 2023; 428:113618. [PMID: 37142202 DOI: 10.1016/j.yexcr.2023.113618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
Hepatitis B Virus (HBV) is the prevailing cause of chronic liver disease, which progresses to Hepatocellular carcinoma (HCC) in 75% of cases. It represents a serious health concern being the fourth leading cause of cancer-related mortality worldwide. Treatments available to date fail to provide a complete cure with high chances of recurrence and related side effects. The lack of reliable, reproducible, and scalable in vitro modeling systems that could recapitulate the viral life cycle and represent virus-host interactions has hindered the development of effective treatments so far. The present review provides insights into the current in-vivo and in-vitro models used for studying HBV and their major limitations. We highlight the use of three-dimensional liver organoids as a novel and suitable platform for modeling HBV infection and HBV-mediated HCC. HBV organoids can be expanded, genetically altered, patient-derived, tested for drug discovery, and biobanked. This review also provides the general guidelines for culturing HBV organoids and highlights their several prospects for HBV drug discovery and screening.
Collapse
Affiliation(s)
- Simran Sharma
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | - Preety Rawal
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | - Savneet Kaur
- Institute of Liver and Biliary Sciences, Delhi, India.
| | - Rekha Puria
- School of Biotechnology, Gautam Buddha University, Greater Noida, India.
| |
Collapse
|
5
|
Wu Y, Hao X, Wei H, Sun R, Chen Y, Tian Z. Blockade of T-cell receptor with Ig and ITIM domains elicits potent antitumor immunity in naturally occurring HBV-related HCC in mice. Hepatology 2023; 77:965-981. [PMID: 35938354 DOI: 10.1002/hep.32715] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 07/28/2022] [Accepted: 08/05/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Chronic HBV infection is the leading cause of HCC and a serious health problem in China, East Asia, and North African countries. Effective treatment of HBV-related HCC is currently unavailable. This study evaluated the therapeutic potential of T-cell immunoreceptor with Ig and ITIM domains (TIGIT) blockade in HBV-related HCC. APPROACH AND RESULTS A mouse model of spontaneous HBV-related HCC was generated by replacing wild-type hepatocytes with HBsAg + hepatocytes (namely HBs-HepR mice). The tumors in HBs-HepR mice were inflammation-associated HCC, similar to HBV-related HCC in patients, which was distinguished from other HCC mouse models, such as diethylnitrosamine-induced HCC, TGF-β-activated kinase 1 knockout-induced HCC, HCC in a stelic animal model, or NASH-induced HCC. HCC in HBs-HepR mice was characterized by an increased number of CD8 + T cells, whereas the production of IL-2, TNF-α, and interferon-gamma (IFN-γ) by intrahepatic CD8 + T cells was decreased. Increased expression of TIGIT on CD8 + T cells was responsible for functional exhaustion. The therapeutic effect of TIGIT blockade was investigated at the early and middle stages of HCC progression in HBs-HepR mice. TIGIT blockade reinvigorated intrahepatic CD8 + T cells with increased TNF-α and IFN-γ production and an increased number of CD8 + T cells in tumors, thereby slowing the development of HCC in HBs-HepR mice. Blocking PD-L1 did not show direct therapeutic effects or synergize with TIGIT blockade. CONCLUSIONS Blockade of TIGIT alone enhanced the antitumor activity of CD8 + T cells during the progression of HBV-related HCC in a spontaneous HCC mouse model.
Collapse
Affiliation(s)
- Yuwei Wu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine , University of Science and Technology of China , Hefei , China.,Institute of Immunology, University of Science and Technology of China , Hefei , China
| | - Xiaolei Hao
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine , University of Science and Technology of China , Hefei , China.,Institute of Immunology, University of Science and Technology of China , Hefei , China
| | - Haiming Wei
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine , University of Science and Technology of China , Hefei , China.,Institute of Immunology, University of Science and Technology of China , Hefei , China
| | - Rui Sun
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine , University of Science and Technology of China , Hefei , China.,Institute of Immunology, University of Science and Technology of China , Hefei , China
| | - Yongyan Chen
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine , University of Science and Technology of China , Hefei , China.,Institute of Immunology, University of Science and Technology of China , Hefei , China
| | - Zhigang Tian
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine , University of Science and Technology of China , Hefei , China.,Institute of Immunology, University of Science and Technology of China , Hefei , China
| |
Collapse
|
6
|
Zhang X, Zhang C, Zhao Q, Wang S, Wang L, Si Y, Su Q, Cheng S, Ding W. Inhibition of Annexin A10 Contributes to ZNF281 Mediated Aggressiveness of Hepatocellular Carcinoma. J Hepatocell Carcinoma 2023; 10:553-571. [PMID: 37041757 PMCID: PMC10083037 DOI: 10.2147/jhc.s400989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 03/15/2023] [Indexed: 04/13/2023] Open
Abstract
Objective To investigate the involvement and transcriptional targets of zinc finger protein 281 (ZNF281) in the progression of hepatocellular carcinoma (HCC). Methods The expression of ZNF281 in HCC was detected in tissue microarray and cell lines. The role of ZNF281 in aggressiveness of HCC was examined using wound healing, matrigel transwell, pulmonary metastasis model and assays for expression of EMT markers. RNA-seq was used to find potential target gene of ZNF281. Chromatin immunoprecipitation (ChIP) assay and co-immunoprecipitation (Co-IP) were employed to uncover the mechanism of the transcriptional regulation of ZNF281 on the target gene. Results ZNF281 was increased in tumor tissues and positively correlated with vascular invasion in HCC. Knockdown of ZNF281 suppressed the migration and invasion with significant alteration of EMT marker expression in HLE and Huh7 HCC cell lines. RNA-seq screening showed that the tumor suppressor gene Annexin A10 (ANXA10) was a most up-regulated gene in response to ZNF281 depletion and responsible for the attenuation of aggressiveness. Mechanistically, ZNF281 interacted with the ANXA10 promoter region harboring ZNF281 recognition sites, and recruited components of nucleosome remodeling and deacetylation (NuRD) complex. By knocking down such components like HDAC1 or MTA1, ANXA10 was released from transcriptional repression by ZNF281/NuRD, and in turn reversed the EMT, invasion and metastasis driven by ZNF281. Conclusion ZNF281 drives invasion and metastasis of HCC partially through transcriptional repression of tumor suppressor gene ANXA10 by recruiting NuRD complex.
Collapse
Affiliation(s)
- Xialu Zhang
- School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Chenguang Zhang
- School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Key Laboratory for Cancer Invasion and Metastasis Mechanism Research, Capital Medical University, Beijing, People’s Republic of China
- Correspondence: Chenguang Zhang; Wei Ding, Email ;
| | - Qingfang Zhao
- School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Shanshan Wang
- Beijing Institute of Hepatology, Beijing You’An Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Liyong Wang
- Core Facilities for Molecular Biology, Capital Medical University, Beijing, People’s Republic of China
| | - Yang Si
- School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Qiang Su
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Shan Cheng
- School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Wei Ding
- School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
7
|
Pu XY, Zheng DF, Lv T, Zhou YJ, Yang JY, Jiang L. Overexpression of transcription factor 3 drives hepatocarcinoma development by enhancing cell proliferation via activating Wnt signaling pathway. Hepatobiliary Pancreat Dis Int 2022; 21:378-386. [PMID: 35033448 DOI: 10.1016/j.hbpd.2022.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/02/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Transcription factor 3 (TCF3) plays pivotal roles in embryonic development, stem cell maintenance and carcinogenesis. However, its role in hepatocellular carcinoma (HCC) remains largely unknown. This study aimed to analyze the correlation between TCF3 expression and clinicopathological features of HCC, and further explore the underlying mechanism in HCC progression. METHODS The expression of TCF3 was collected from the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) HCC datasets, and further confirmed by immunostaining and Western blotting assays. The correlation between TCF3 expression and the clinicopathological features was evaluated. Bioinformatical analysis and in vitro experiments were conducted to explore the potential role of TCF3 in HCC development. RESULTS Both the mRNA and protein levels of TCF3 were significantly higher in HCC tumor tissues compared to tumor adjacent tissues (P < 0.001 and P < 0.01). Analysis based on TCGA datasets showed that TCF3 was positively correlated with tumor clinical stage and grade, and patients with high TCF3 expression had shorter overall survival (P = 0.012), disease-specific survival (P = 0.022) and progression-free survival (P = 0.013). Similarly, the immunostaining results revealed that the high expression of TCF3 was closely correlated with tumor size (P = 0.001) and TNM stage (P = 0.002), and TCF3 was an independent risk factor of HCC. In vitro study exhibited that TCF3 knockdown dramatically suppressed cancer cell proliferation, and the underlying mechanism might be that the silencing of TCF3 reduced the expression of critical regulating proteins towards cell cycle and proteins involved in Wnt signaling pathways. CONCLUSIONS TCF3 expression is significantly elevated in HCC and positively associated with the tumor size and TNM stage, as well as poor prognosis of HCC patients. The mechanism might be that TCF3 promotes cancer cell proliferation via activating Wnt signaling pathway.
Collapse
Affiliation(s)
- Xing-Yu Pu
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dao-Feng Zheng
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Lv
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong-Jie Zhou
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jia-Yin Yang
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Jiang
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
8
|
Bévant K, Desoteux M, Angenard G, Pineau R, Caruso S, Louis C, Papoutsoglou P, Sulpice L, Gilot D, Zucman‐Rossi J, Coulouarn C. TGFβ-induced FOXS1 controls epithelial-mesenchymal transition and predicts a poor prognosis in liver cancer. Hepatol Commun 2022; 6:1157-1171. [PMID: 34825776 PMCID: PMC9035581 DOI: 10.1002/hep4.1866] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/12/2021] [Accepted: 10/26/2021] [Indexed: 01/13/2023] Open
Abstract
Transforming growth factor beta (TGF-β) plays a key role in tumor progression, notably as a potent inducer of epithelial-mesenchymal transition (EMT). However, all of the molecular effectors driving TGFβ-induced EMT are not fully characterized. Here, we report that forkhead box S1 (FOXS1) is a SMAD (mothers against decapentaplegic)-dependent TGFβ-induced transcription factor, which regulates the expression of genes required for the initial steps of EMT (e.g., snail family transcription repressor 1) and to maintain a mesenchymal phenotype in hepatocellular carcinoma (HCC) cells. In human HCC, we report that FOXS1 is a biomarker of poorly differentiated and aggressive tumor subtypes. Importantly, FOXS1 expression level and activity are associated with a poor prognosis (e.g., reduced patient survival), not only in HCC but also in colon, stomach, and kidney cancers. Conclusion: FOXS1 constitutes a clinically relevant biomarker for tumors in which the pro-metastatic arm of TGF-β is active (i.e., patients who may benefit from targeted therapies using inhibitors of the TGF-β pathway).
Collapse
Affiliation(s)
- Kevin Bévant
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance
- InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - Matthis Desoteux
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance
- InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - Gaëlle Angenard
- InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - Raphaël Pineau
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance
| | - Stefano Caruso
- Centre de Recherche des CordeliersInsermSorbonne UniversitéUniversité de ParisUniversité Paris 13Functional Genomics of Solid Tumors LaboratoryParisFrance
| | - Corentin Louis
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance
- InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - Panagiotis Papoutsoglou
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance
- InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - Laurent Sulpice
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance
- InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - David Gilot
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance
| | - Jessica Zucman‐Rossi
- Centre de Recherche des CordeliersInsermSorbonne UniversitéUniversité de ParisUniversité Paris 13Functional Genomics of Solid Tumors LaboratoryParisFrance
- European Hospital Georges PompidouAP‐HPParisFrance
| | - Cédric Coulouarn
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance
- InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| |
Collapse
|
9
|
Data mining of key genes expression in hepatocellular carcinoma: novel potential biomarkers of diagnosis prognosis or progression. Clin Exp Metastasis 2022; 39:589-602. [PMID: 35429302 PMCID: PMC9338913 DOI: 10.1007/s10585-022-10164-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/22/2022] [Indexed: 11/30/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the main cancer-related causes of death worldwide. The study aimed to perform a data mining analysis of the expression and regulatory role of key genes in HCC to reveal novel potential biomarkers of diagnosis prognosis, or progression since their availability is still almost lacking. Starting from data of our cohort of patients (HCV-positive HCC pts undergoing liver transplantation (LR, n = 10) and donors (LD, n = 14), deeply analyzed previously, in which apelin, osteopontin, osteoprotegerin, NOTCH-1, CASP-3, Bcl-2, BAX, PTX3, and NPTX2 were analyzed, we applied statistical analysis and in-silico tools (Gene Expression Profiling Interactive Analysis, HCCDB database and GeneMania, UALCAN) to screen and identify the key genes. Firstly, we performed a stepwise regression analysis using our mRNA-datasets which revealed that higher expression levels of apelin and osteopontin were positively associated with the HCC and identified that the most consistently differentially expressed gene across multiple HCC expression datasets was only OPN. This comprehensive strategy of data mining evidenced that OPN might have a potential function as an important tumor marker-driven oncogenesis being associated with poor prognosis of HCC patients.
Collapse
|
10
|
Ho CM, Lin KT, Shen R, Gu DL, Lee SS, Su WH, Jou YS. Prognostic comparative genes predict targets for sorafenib combination therapies in hepatocellular carcinoma. Comput Struct Biotechnol J 2022; 20:1752-1763. [PMID: 35495118 PMCID: PMC9024375 DOI: 10.1016/j.csbj.2022.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 11/17/2022] Open
Abstract
Large-scale comparative transcriptomics analysis of hepatocellular carcinoma reveals 664 prognostic comparative HCC (pcHCC) genes. pcHCC genes included novel targets with potential utility in sorafenib combination therapies. Knockdown of the selective pcHCC genes NCAPG and CENPW downregulated the p38/STAT3 axis to enhance sorafenib combination treatments.
With the increasing incidence and mortality of human hepatocellular carcinoma (HCC) worldwide, revealing innovative targets to improve therapeutic strategies is crucial for prolonging the lives of patients. To identify innovative targets, we conducted a comprehensive comparative transcriptome analysis of 5,410 human HCCs and 974 mouse liver cancers to identify concordantly expressed genes associated with patient survival. Among the 664 identified prognostic comparative HCC (pcHCC) genes, upregulated pcHCC genes were associated with prognostic clinical features, including large tumor size, vascular invasion and late HCC stages. Interestingly, after validating HCC patient prognoses in multiple independent datasets, we matched the 664 aberrant pcHCC genes with the sorafenib-altered genes in TCGA_LIHC patients and found these 664 pcHCC genes were enriched in sorafenib-related functions, such as downregulated xenobiotic and lipid metabolism and upregulated cell proliferation. Therapeutic agents targeting aberrant pcHCC genes presented divergent molecular mechanisms, including suppression of sorafenib-unrelated oncogenic pathways, induction of sorafenib-unrelated ferroptosis, and modulation of sorafenib transportation and metabolism, to potentiate sorafenib therapeutic effects in HCC combination therapy. Moreover, the pcHCC genes NCAPG and CENPW, which have not been targeted in combination with sorafenib treatment, were knocked down and combined with sorafenib treatment, which reduced HCC cell viability based on disruption to the p38/STAT3 axis, thereby hypersensitizing HCC cells. Together, our results provide important resources and reveal that 664 pcHCC genes represent innovative targets suitable for developing therapeutic strategies in combination with sorafenib based on the divergent synergistic mechanisms for HCC tumor suppression.
Collapse
|
11
|
Aoun R, El Hadi C, Tahtouh R, El Habre R, Hilal G. Microarray analysis of breast cancer gene expression profiling in response to 2-deoxyglucose, metformin, and glucose starvation. Cancer Cell Int 2022; 22:123. [PMID: 35305635 PMCID: PMC8933915 DOI: 10.1186/s12935-022-02542-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is the most frequently diagnosed cancer in women. Altering glucose metabolism and its effects on cancer progression and treatment resistance is an emerging interest in BC research. For instance, combining chemotherapy with glucose-lowering drugs (2-deoxyglucose (2-DG), metformin (MET)) or glucose starvation (GS) has shown better outcomes than with chemotherapy alone. However, the genes and molecular mechanisms that govern the action of these glucose deprivation conditions have not been fully elucidated. Here, we investigated the differentially expressed genes in MCF-7 and MDA-MB-231 BC cell lines upon treatment with glucose-lowering drugs (2-DG, MET) and GS using microarray analysis to study the difference in biological functions between the glucose challenges and their effect on the vulnerability of BC cells. METHODS MDA-MB-231 and MCF-7 cells were treated with 20 mM MET or 4 mM 2-DG for 48 h. GS was performed by gradually decreasing the glucose concentration in the culture medium to 0 g/L, in which the cells remained with fetal bovine serum for one week. Expression profiling was carried out using Affymetrix Human Clariom S microarrays. Differentially expressed genes were obtained from the Transcriptome Analysis Console and enriched using DAVID and R packages. RESULTS Our results showed that MDA-MB-231 cells were more responsive to glucose deprivation than MCF-7 cells. Endoplasmic reticulum stress response and cell cycle inhibition were detected after all three glucose deprivations in MDA-MB-231 cells and only under the metformin and GS conditions in MCF-7 cells. Induction of apoptosis and inhibition of DNA replication were observed with all three treatments in MDA-MB-231 cells and metformin-treated MCF-7 cells. Upregulation of cellular response to reactive oxygen species and inhibition of DNA repair mechanisms resulted after metformin and GS administration in MDA-MB-231 cell lines and metformin-treated MCF-7 cells. Autophagy was induced after 2-DG treatment in MDA-MB-231 cells and after metformin in MCF-7 cells. Finally, inhibition of DNA methylation were observed only with GS in MDA-MB-231 cells. CONCLUSION The procedure used to process cancer cells and analyze their expression data distinguishes our study from others. GS had the greatest effect on breast cancer cells compared to 2-DG and MET. Combining MET and GS could restrain both cell lines, making them more vulnerable to conventional chemotherapy.
Collapse
Affiliation(s)
- Rita Aoun
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | | | - Roula Tahtouh
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Rita El Habre
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - George Hilal
- Cancer and Metabolism Laboratory, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon.
| |
Collapse
|
12
|
Louis C, Leclerc D, Coulouarn C. Emerging roles of circular RNAs in liver cancer. JHEP Rep 2022; 4:100413. [PMID: 35036887 PMCID: PMC8749337 DOI: 10.1016/j.jhepr.2021.100413] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma and cholangiocarcinoma are the most common primary liver tumours, whose incidence and associated mortality have increased over recent decades. Liver cancer is often diagnosed late when curative treatments are no longer an option. Characterising new molecular determinants of liver carcinogenesis is crucial for the development of innovative treatments and clinically relevant biomarkers. Recently, circular RNAs (circRNAs) emerged as promising regulatory molecules involved in cancer onset and progression. Mechanistically, circRNAs are mainly known for their ability to sponge and regulate the activity of microRNAs and RNA-binding proteins, although other functions are emerging (e.g. transcriptional and post-transcriptional regulation, protein scaffolding). In liver cancer, circRNAs have been shown to regulate tumour cell proliferation, migration, invasion and cell death resistance. Their roles in regulating angiogenesis, genome instability, immune surveillance and metabolic switching are emerging. Importantly, circRNAs are detected in body fluids. Due to their circular structure, circRNAs are often more stable than mRNAs or miRNAs and could therefore serve as promising biomarkers - quantifiable with high specificity and sensitivity through minimally invasive methods. This review focuses on the role and the clinical relevance of circRNAs in liver cancer, including the development of innovative biomarkers and therapeutic strategies.
Collapse
Key Words
- ASO, antisense oligonucleotide
- CCA, cholangiocarcinoma
- CLIP, cross-linking immunoprecipitation
- EMT, epithelial-to-mesenchymal transition
- EVs, extracellular vesicles
- HCC, hepatocellular carcinoma
- HN1, haematopoietic- and neurologic-expressed sequence 1
- IRES, internal ribosome entry sites
- NGS, next-generation sequencing
- QKI, Quaking
- RBP, RNA-binding protein
- RISC, RNA-induced silencing complex
- TAM, tumour-associated macrophage
- TSB, target site blockers
- biomarker
- cancer hallmarks
- cholangiocarcinoma
- circRNA
- circRNA, circular RNA
- hepatocellular carcinoma
- miRNA, microRNA
- shRNA, small-hairpin RNA
- snRNP, small nuclear ribonuclear proteins
Collapse
Affiliation(s)
- Corentin Louis
- Inserm, Univ Rennes 1, COSS (Chemistry, Oncogenesis Stress Signaling), UMR_S 1242, Centre de Lutte contre le Cancer Eugène Marquis, F-35042, Rennes, France
| | - Delphine Leclerc
- Inserm, Univ Rennes 1, COSS (Chemistry, Oncogenesis Stress Signaling), UMR_S 1242, Centre de Lutte contre le Cancer Eugène Marquis, F-35042, Rennes, France
| | - Cédric Coulouarn
- Inserm, Univ Rennes 1, COSS (Chemistry, Oncogenesis Stress Signaling), UMR_S 1242, Centre de Lutte contre le Cancer Eugène Marquis, F-35042, Rennes, France
| |
Collapse
|
13
|
Desoteux M, Louis C, Bévant K, Glaise D, Coulouarn C. A Minimal Subset of Seven Genes Associated with Tumor Hepatocyte Differentiation Predicts a Poor Prognosis in Human Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:5624. [PMID: 34830779 PMCID: PMC8616205 DOI: 10.3390/cancers13225624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a deadly cancer worldwide as a result of a frequent late diagnosis which limits the therapeutic options. Tumor progression in HCC is closely correlated with the dedifferentiation of hepatocytes, the main parenchymal cells in the liver. Here, we hypothesized that the expression level of genes reflecting the differentiation status of tumor hepatocytes could be clinically relevant in defining subsets of patients with different clinical outcomes. To test this hypothesis, an integrative transcriptomics approach was used to stratify a cohort of 139 HCC patients based on a gene expression signature established in vitro in the HepaRG cell line using well-controlled culture conditions recapitulating tumor hepatocyte differentiation. The HepaRG model was first validated by identifying a robust gene expression signature associated with hepatocyte differentiation and liver metabolism. In addition, the signature was able to distinguish specific developmental stages in mice. More importantly, the signature identified a subset of human HCC associated with a poor prognosis and cancer stem cell features. By using an independent HCC dataset (TCGA consortium), a minimal subset of seven differentiation-related genes was shown to predict a reduced overall survival, not only in patients with HCC but also in other types of cancers (e.g., kidney, pancreas, skin). In conclusion, the study identified a minimal subset of seven genes reflecting the differentiation status of tumor hepatocytes and clinically relevant for predicting the prognosis of HCC patients.
Collapse
Affiliation(s)
- Matthis Desoteux
- Inserm, Univ. Rennes, UMR1242, Chemistry Oncogenesis Stress Signaling (COSS), 35042 Rennes, France; (M.D.); (C.L.); (K.B.)
| | - Corentin Louis
- Inserm, Univ. Rennes, UMR1242, Chemistry Oncogenesis Stress Signaling (COSS), 35042 Rennes, France; (M.D.); (C.L.); (K.B.)
| | - Kevin Bévant
- Inserm, Univ. Rennes, UMR1242, Chemistry Oncogenesis Stress Signaling (COSS), 35042 Rennes, France; (M.D.); (C.L.); (K.B.)
| | - Denise Glaise
- Inserm, Univ. Rennes, UMR991, Liver Metabolisms and Cancer, 35043 Rennes, France;
| | - Cédric Coulouarn
- Inserm, Univ. Rennes, UMR1242, Chemistry Oncogenesis Stress Signaling (COSS), 35042 Rennes, France; (M.D.); (C.L.); (K.B.)
| |
Collapse
|
14
|
Al-Harazi O, Kaya IH, Al-Eid M, Alfantoukh L, Al Zahrani AS, Al Sebayel M, Kaya N, Colak D. Identification of Gene Signature as Diagnostic and Prognostic Blood Biomarker for Early Hepatocellular Carcinoma Using Integrated Cross-Species Transcriptomic and Network Analyses. Front Genet 2021; 12:710049. [PMID: 34659334 PMCID: PMC8511318 DOI: 10.3389/fgene.2021.710049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/09/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is considered the most common type of liver cancer and the fourth leading cause of cancer-related deaths in the world. Since the disease is usually diagnosed at advanced stages, it has poor prognosis. Therefore, reliable biomarkers are urgently needed for early diagnosis and prognostic assessment. Methods: We used genome-wide gene expression profiling datasets from human and rat early HCC (eHCC) samples to perform integrated genomic and network-based analyses, and discovered gene markers that are expressed in blood and conserved in both species. We then used independent gene expression profiling datasets for peripheral blood mononuclear cells (PBMCs) for eHCC patients and from The Cancer Genome Atlas (TCGA) database to estimate the diagnostic and prognostic performance of the identified gene signature. Furthermore, we performed functional enrichment, interaction networks and pathway analyses. Results: We identified 41 significant genes that are expressed in blood and conserved across species in eHCC. We used comprehensive clinical data from over 600 patients with HCC to verify the diagnostic and prognostic value of 41-gene-signature. We developed a prognostic model and a risk score using the 41-geneset that showed that a high prognostic index is linked to a worse disease outcome. Furthermore, our 41-gene signature predicted disease outcome independently of other clinical factors in multivariate regression analysis. Our data reveals a number of cancer-related pathways and hub genes, including EIF4E, H2AFX, CREB1, GSK3B, TGFBR1, and CCNA2, that may be essential for eHCC progression and confirm our gene signature's ability to detect the disease in its early stages in patients' biological fluids instead of invasive procedures and its prognostic potential. Conclusion: Our findings indicate that integrated cross-species genomic and network analysis may provide reliable markers that are associated with eHCC that may lead to better diagnosis, prognosis, and treatment options.
Collapse
Affiliation(s)
- Olfat Al-Harazi
- Department of Biostatistics, Epidemiology, and Scientific Computing, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ibrahim H Kaya
- AlFaisal University, College of Medicine, Riyadh, Saudi Arabia
| | - Maha Al-Eid
- Department of Biostatistics, Epidemiology, and Scientific Computing, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Lina Alfantoukh
- Department of Biostatistics, Epidemiology, and Scientific Computing, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ali Saeed Al Zahrani
- Gulf Centre for Cancer Control and Prevention, King Faisal Special Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mohammed Al Sebayel
- Liver and Small Bowel Transplantation and Hepatobiliary-Pancreatic Surgery Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Department of Surgery, University of Almaarefa, Riyadh, Saudi Arabia
| | - Namik Kaya
- Translational Genomics Department, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Dilek Colak
- Department of Biostatistics, Epidemiology, and Scientific Computing, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Piorońska W, Nwosu ZC, Han M, Büttner M, Ebert MP, Dooley S, Meyer C. Dysregulated paired related homeobox 1 impacts on hepatocellular carcinoma phenotypes. BMC Cancer 2021; 21:1006. [PMID: 34496784 PMCID: PMC8424914 DOI: 10.1186/s12885-021-08637-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 07/27/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a major cause of cancer-related death. Paired related homeobox 1 (PRRX1) is a transcription factor that regulates cell growth and differentiation, but its importance in HCC is unclear. METHODS We examined the expression pattern of PRRX1 in nine microarray datasets of human HCC tumour samples (n > 1100) and analyzed its function in HCC cell lines. In addition, we performed gene set enrichment, Kaplan-Meier overall survival analysis, metabolomics and functional assays. RESULTS PRRX1 is frequently upregulated in human HCC. Pathway enrichment analysis predicted a direct correlation between PRRX1 and focal adhesion and epithelial-mesenchymal transition. High expression of PRRX1 and low ZEB1 or high ZEB2 significantly predicted better overall survival in HCC patients. In contrast, metabolic processes correlated inversely and transcriptional analyses revealed that glycolysis, TCA cycle and amino acid metabolism were affected. These findings were confirmed by metabolomics analysis. At the phenotypic level, PRRX1 knockdown accelerated proliferation and clonogenicity in HCC cell lines. CONCLUSIONS Our results suggest that PRRX1 controls metabolism, has a tumour suppressive role, and may function in cooperation with ZEB1/2. These findings have functional relevance in HCC, including in understanding transcriptional control of distinct cancer hallmarks.
Collapse
Affiliation(s)
- Weronika Piorońska
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Zeribe Chike Nwosu
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,Current address: Department of Molecular and Integrative Physiology, Rogel Cancer Centre, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mei Han
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,Current address: Department of Internal Medicine, the Second Hospital of Dalian Medical University, Shahekou District, Dalian City, Liaoning Province, China
| | - Michael Büttner
- The Metabolomics Core Technology Platform of the University of Heidelberg, Im Neuenheimer Feld 360, 69120, Heidelberg, Germany
| | - Matthias Philip Ebert
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Christoph Meyer
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| |
Collapse
|
16
|
Bévant K, Desoteux M, Abdel Wahab AHA, Abdel Wahab SA, Metwally AM, Coulouarn C. DNA Methylation of TGFβ Target Genes: Epigenetic Control of TGFβ Functional Duality in Liver Cancer. Cells 2021; 10:2207. [PMID: 34571856 PMCID: PMC8468746 DOI: 10.3390/cells10092207] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor beta (TGFβ) plays a key role in liver carcinogenesis. However, its action is complex, since TGFβ exhibits tumor-suppressive or oncogenic properties, depending on the tumor stage. At an early stage TGFβ exhibits cytostatic features, but at a later stage it promotes cell growth and metastasis, as a potent inducer of epithelial to mesenchymal transition (EMT). Here, we evaluated DNA methylation as a possible molecular mechanism switching TGFβ activity toward tumor progression in hepatocellular carcinoma (HCC). We report that decitabine, a demethylating agent already used in the clinic for the treatment of several cancers, greatly impairs the transcriptional response of SNU449 HCC cells to TGFβ. Importantly, decitabine was shown to induce the expression of EMT-related transcription factors (e.g., SNAI1/2, ZEB1/2). We also report that the promoter of SNAI1 was hypomethylated in poor-prognosis human HCC, i.e., associated with high grade, high AFP level, metastasis and recurrence. Altogether, the data highlight an epigenetic control of several effectors of the TGFβ pathway in human HCC possibly involved in switching its action toward EMT and tumor progression. Thus, we conclude that epidrugs should be carefully evaluated for the treatment of HCC, as they may activate tumor promoting pathways.
Collapse
Affiliation(s)
- Kevin Bévant
- Centre de Lutte Contre le Cancer Eugène Marquis, Inserm, University of Rennes 1, UMR_S 1242, COSS (Chemistry, Oncogenesis Stress Signaling), 35042 Rennes, France; (K.B.); (M.D.)
| | - Matthis Desoteux
- Centre de Lutte Contre le Cancer Eugène Marquis, Inserm, University of Rennes 1, UMR_S 1242, COSS (Chemistry, Oncogenesis Stress Signaling), 35042 Rennes, France; (K.B.); (M.D.)
| | | | - Sabrin A. Abdel Wahab
- Medical Laboratory Department, Students Hospital, Cairo University, Cairo 11796, Egypt;
| | - Ayman Mohamed Metwally
- Medical Laboratory Technology Department, College of Applied Health Science Technology, Misr University for Science and Technology (MUST), Al-Motamayez District, 6th of October P.O. Box 77, Egypt
| | - Cédric Coulouarn
- Centre de Lutte Contre le Cancer Eugène Marquis, Inserm, University of Rennes 1, UMR_S 1242, COSS (Chemistry, Oncogenesis Stress Signaling), 35042 Rennes, France; (K.B.); (M.D.)
| |
Collapse
|
17
|
De Crignis E, Hossain T, Romal S, Carofiglio F, Moulos P, Khalid MM, Rao S, Bazrafshan A, Verstegen MM, Pourfarzad F, Koutsothanassis C, Gehart H, Kan TW, Palstra RJ, Boucher C, IJzermans JN, Huch M, Boj SF, Vries R, Clevers H, van der Laan LJ, Hatzis P, Mahmoudi T. Application of human liver organoids as a patient-derived primary model for HBV infection and related hepatocellular carcinoma. eLife 2021; 10:e60747. [PMID: 34328417 PMCID: PMC8384419 DOI: 10.7554/elife.60747] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
The molecular events that drive hepatitis B virus (HBV)-mediated transformation and tumorigenesis have remained largely unclear, due to the absence of a relevant primary model system. Here we propose the use of human liver organoids as a platform for modeling HBV infection and related tumorigenesis. We first describe a primary ex vivo HBV-infection model derived from healthy donor liver organoids after challenge with recombinant virus or HBV-infected patient serum. HBV-infected organoids produced covalently closed circular DNA (cccDNA) and HBV early antigen (HBeAg), expressed intracellular HBV RNA and proteins, and produced infectious HBV. This ex vivo HBV-infected primary differentiated hepatocyte organoid platform was amenable to drug screening for both anti-HBV activity and drug-induced toxicity. We also studied HBV replication in transgenically modified organoids; liver organoids exogenously overexpressing the HBV receptor sodium taurocholate co-transporting polypeptide (NTCP) after lentiviral transduction were not more susceptible to HBV, suggesting the necessity for additional host factors for efficient infection. We also generated transgenic organoids harboring integrated HBV, representing a long-term culture system also suitable for viral production and the study of HBV transcription. Finally, we generated HBV-infected patient-derived liver organoids from non-tumor cirrhotic tissue of explants from liver transplant patients. Interestingly, transcriptomic analysis of patient-derived liver organoids indicated the presence of an aberrant early cancer gene signature, which clustered with the hepatocellular carcinoma (HCC) cohort on The Cancer Genome Atlas Liver Hepatocellular Carcinoma dataset and away from healthy liver tissue, and may provide invaluable novel biomarkers for the development of HCC and surveillance in HBV-infected patients.
Collapse
Affiliation(s)
- Elisa De Crignis
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Tanvir Hossain
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Shahla Romal
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Fabrizia Carofiglio
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Panagiotis Moulos
- Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
| | - Mir Mubashir Khalid
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Shringar Rao
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ameneh Bazrafshan
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Monique Ma Verstegen
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | | | - Helmuth Gehart
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Tsung Wai Kan
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Robert-Jan Palstra
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Charles Boucher
- Department of Viroscience, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Jan Nm IJzermans
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Meritxell Huch
- Max Plank Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Sylvia F Boj
- Foundation Hubrecht Organoid Technology (HUB), Utrecht, Netherlands
| | - Robert Vries
- Foundation Hubrecht Organoid Technology (HUB), Utrecht, Netherlands
| | - Hans Clevers
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Luc Jw van der Laan
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Pantelis Hatzis
- Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Urology, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
18
|
Rybakova Y, Gonzalez JT, Bogorad R, Chauhan VP, Dong YL, Whittaker CA, Zatsepin T, Koteliansky V, Anderson DG. Identification of a long non-coding RNA regulator of liver carcinoma cell survival. Cell Death Dis 2021; 12:178. [PMID: 33589614 PMCID: PMC7884843 DOI: 10.1038/s41419-021-03453-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023]
Abstract
Genomic studies have significantly improved our understanding of hepatocellular carcinoma (HCC) biology and have led to the discovery of multiple protein-coding genes driving hepatocarcinogenesis. In addition, these studies have identified thousands of new non-coding transcripts deregulated in HCC. We hypothesize that some of these transcripts may be involved in disease progression. Long non-coding RNAs are a large class of non-coding transcripts which participate in the regulation of virtually all cellular functions. However, a majority of lncRNAs remain dramatically understudied. Here, we applied a pooled shRNA-based screen to identify lncRNAs essential for HCC cell survival. We validated our screening results using RNAi, CRISPRi, and antisense oligonucleotides. We found a lncRNA, termed ASTILCS, that is critical for HCC cell growth and is overexpressed in tumors from HCC patients. We demonstrated that HCC cell death upon ASTILCS knockdown is associated with apoptosis induction and downregulation of a neighboring gene, protein tyrosine kinase 2 (PTK2), a crucial protein for HCC cell survival. Taken together, our study describes a new, non-coding RNA regulator of HCC.
Collapse
Affiliation(s)
- Yulia Rybakova
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Skolkovo Institute of Science and Technology, Moscow, 121205, Russia
| | - John T Gonzalez
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Roman Bogorad
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Vikash P Chauhan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Yize L Dong
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Charles A Whittaker
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Timofei Zatsepin
- Skolkovo Institute of Science and Technology, Moscow, 121205, Russia
| | | | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
19
|
Niu Y, Chen L, Wu M, Huang W, Wu X, Huang D, Xie Y, Shi G. Partial abrogation of FXR-KNG1 signaling by carboxyl-terminal truncated HBx-C30 in hepatitis B virus-associated hepatocellular carcinoma. Virus Res 2021; 293:198264. [PMID: 33359549 DOI: 10.1016/j.virusres.2020.198264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Hepatitis B virus (HBV) X protein (HBx) is a key regulator of HBV-associated hepatocarcinogenesis. C-terminal-truncated HBx is frequently detected in hepatocellular carcinoma (HCC). The role of HBx, especially C-terminal-truncated HBx, in HCC pathogenesis has been controversial. To elucidate the biological role of C-terminal-truncated HBx underlying HBV-associated hepato-oncogenesis, we constructed a vector expressing HBx-C30 (deletion of 30 aa from the C terminus of HBx) and functionally analyzed its regulation on farnesoid X receptor (FXR) signaling, which is known to inhibit hepatocarcinogenesis. In the present study, we found full-length HBx and HBx C-terminal truncation coexist in HCC, and both full length HBx and HBx-C30 can activate FXR signaling. Moreover, HBx-C30 weakly coactivates FXR-KNG1 signaling compared to full-length HBx.
Collapse
Affiliation(s)
- Yongdong Niu
- Department of Pharmacology, Shantou University Medical College, Guangdong, China.
| | - Liming Chen
- Department of Oncology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Manpeng Wu
- The Second People's Hospital of Shantou, Shantou, China
| | - Weiyi Huang
- Department of Pharmacology, Shantou University Medical College, Guangdong, China
| | - Xuejun Wu
- Department of Pharmacology, Shantou University Medical College, Guangdong, China
| | - Danmei Huang
- Department of Pharmacology, Shantou University Medical College, Guangdong, China
| | - Yangmin Xie
- Department of Experimental Animal Center, Medical College of Shantou University, Guangdong, China
| | - Ganggang Shi
- Department of Pharmacology, Shantou University Medical College, Guangdong, China.
| |
Collapse
|
20
|
Abstract
The incidence and mortality related to hepatocellular carcinoma (HCC) continue to increase in the United States, with most patients presenting at advanced stages where curative therapy and long-term survival is unlikely. Fortunately, significant progress has been made in identifying the landscape of HCC mutations due to advances in whole genome expression profiling. Drug development has also evolved in recent years with multiple first- and second-line systemic chemotherapy agents approved for HCC for the first time in over a decade. However, despite advances in molecular profiling, mutations that were identified at the highest frequency were not amenable to drug development, and curative therapy for advanced HCC remains elusive. This review focuses on the current understanding of HCC genomics and the limitations of translating the current HCC molecular profiling into clinical practice and examines the effectiveness and benefits of approved systemic therapies for advanced HCC.
Collapse
|
21
|
Gurses SA, Banskar S, Stewart C, Trimoski B, Dziarski R, Gupta D. Nod2 protects mice from inflammation and obesity-dependent liver cancer. Sci Rep 2020; 10:20519. [PMID: 33239685 PMCID: PMC7688964 DOI: 10.1038/s41598-020-77463-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Nod2 is a pattern recognition receptor that modulates host innate immune responses and protects from inflammation, steatosis, and obesity. Obesity and inflammation are risk factors for hepatocellular carcinoma, however, the role of Nod2 in obesity-dependent hepatic tumorigenesis is not known. Here we tested the hypothesis that Nod2 protects from high fat diet (HFD)-dependent hepatic cancer. We used an obesity-dependent hepatic tumor model. WT and Nod2−/− mice were treated with the carcinogen dimethylbenz[a]anthracene (DMBA) and maintained on HFD. Nod2−/− mice treated with DMBA and maintained on HFD gain significantly more weight and develop more liver tumors than similarly treated WT mice. Livers of Nod2−/− tumorigenic mice had increased expression of genes involved in cell proliferation, immune responses, and cholesterol biosynthesis, increased infiltration of neutrophils, inflammatory monocytes, and T cells, and increased activation of STAT3 and ERK during the later stages of tumorigenesis. Bioinformatic analyses of genes with differential expression predicted an increase in cancer, immune, and cholesterol biosynthesis pathways. In summary, we have identified a novel role for Nod2 and demonstrate that Nod2 protects from HFD-dependent liver malignancy and this protection is accompanied by decreased cell proliferation, inflammation, steroid biosynthesis, neutrophils and macrophages infiltration, and STAT3 and MAPK signaling in the liver.
Collapse
Affiliation(s)
- Serdar A Gurses
- Indiana University School of Medicine-Northwest, Gary, IN, 46408, USA
| | - Sunil Banskar
- Indiana University School of Medicine-Northwest, Gary, IN, 46408, USA
| | - Cody Stewart
- Indiana University School of Medicine-Northwest, Gary, IN, 46408, USA
| | - Bill Trimoski
- Indiana University School of Medicine-Northwest, Gary, IN, 46408, USA
| | - Roman Dziarski
- Indiana University School of Medicine-Northwest, Gary, IN, 46408, USA
| | - Dipika Gupta
- Indiana University School of Medicine-Northwest, Gary, IN, 46408, USA.
| |
Collapse
|
22
|
Pang Y, Liu Z, Han H, Wang B, Li W, Mao C, Liu S. Peptide SMIM30 promotes HCC development by inducing SRC/YES1 membrane anchoring and MAPK pathway activation. J Hepatol 2020; 73:1155-1169. [PMID: 32461121 DOI: 10.1016/j.jhep.2020.05.028] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Growing evidence shows that some non-coding RNAs (ncRNAs) contain small open reading frames (smORFs) that are translated into short peptides. Herein, we aimed to determine where and how these short peptides might promote hepatocellular carcinoma (HCC) development. METHODS We performed an RNA-immunoprecipitation followed by high-throughput sequencing (RIP-seq) assay with an antibody against ribosomal protein S6 (RPS6) on 4 cancer cell lines. Focusing on 1 long non-coding RNA (lncRNA), LINC00998, we used qPCR and public databases to evaluate its expression level in patients with HCC. Special vectors were constructed to confirm its coding potential. We also explored the function and mechanism of LINC00998-encoded peptide in tumor growth and metastasis. RESULTS We discovered that many lncRNAs bind to RPS6 in cancer cells. One of these lncRNAs, LINC00998, encoded a small endogenous peptide, termed SMIM30. SMIM30, rather than the RNA itself, promoted HCC tumorigenesis by modulating cell proliferation and migration, and its level was correlated with poor survival in patients with HCC. Furthermore, SMIM30 was transcribed by c-Myc and then drove the membrane anchoring of the non-receptor tyrosine kinases SRC/YES1. Moreover, the downstream MAPK signaling pathway was activated by SRC/YES1. CONCLUSIONS Our results not only unravel a new mechanism of HCC tumorigenesis promoted by ncRNA-encoded peptides, but also suggest that these peptides can serve as a new target for HCC cancer therapy and a new biomarker for HCC diagnosis and prognosis. LAY SUMMARY Very little is known about how peptides activate signaling pathways that play a crucial role in diseases such as cancer. Specifically, we reported on a conserved peptide encoded by LINC00998, SMIM30. This peptide promoted the tumorigenesis of hepatocellular carcinoma (HCC) by modulating cell proliferation and migration. Of note, it bound the non-receptor tyrosine kinases, SRC/YES1, to drive their membrane anchoring and phosphorylation, activating the downstream MAPK signaling pathway. Our work not only unravels a new mechanism of HCC tumorigenesis promoted by peptides, but also demonstrates how the peptide works to activate a signaling pathway.
Collapse
Affiliation(s)
- Yanan Pang
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Zhiyong Liu
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Huan Han
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Beilei Wang
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Wei Li
- Changzheng Hospital, Second Military Medical University, Shanghai 200433, China
| | - Chuanbin Mao
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019-5300, USA; School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, PR China.
| | - Shanrong Liu
- Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
23
|
Yim SY, Kang SH, Shin JH, Jeong YS, Sohn BH, Um SH, Lee JS. Low ARID1A Expression is Associated with Poor Prognosis in Hepatocellular Carcinoma. Cells 2020; 9:E2002. [PMID: 32878261 PMCID: PMC7564185 DOI: 10.3390/cells9092002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/26/2020] [Accepted: 07/29/2020] [Indexed: 01/01/2023] Open
Abstract
AT-rich interactive domain 1A (ARID1A) is one of the most frequently mutated genes in hepatocellular carcinoma (HCC), but its clinical significance is not clarified. We aimed to evaluate the clinical significance of low ARID1A expression in HCC. By analyzing the gene expression data of liver from Arid1a-knockout mice, hepatic Arid1a-specific gene expression signature was identified (p < 0.05 and 0.5-fold difference). From this signature, a prediction model was developed to identify tissues lacking Arid1a activity and was applied to gene expression data from three independent cohorts of HCC patients to stratify patients according to ARID1A activity. The molecular features associated with loss of ARID1A were analyzed using The Cancer Genome Atlas (TCGA) multi-platform data, and Ingenuity Pathway Analysis (IPA) was done to uncover potential signaling pathways associated with ARID1A loss. ARID1A inactivation was clinically associated with poor prognosis in all three independent cohorts and was consistently related to poor prognosis subtypes of previously reported gene signatures (highly proliferative, hepatic stem cell, silence of Hippo pathway, and high recurrence signatures). Immune activity, indicated by significantly lower IFNG6 and cytolytic activity scores and enrichment of regulatory T-cell composition, was lower in the ARID1A-low subtype than ARID1A-high subtype. Ingenuity pathway analysis revealed that direct upstream transcription regulators of the ARID1A signature were genes associated with cell cycle, including E2F group, CCND1, and MYC, while tumor suppressors such as TP53, SMAD3, and CTNNB1 were significantly inhibited. ARID1A plays an important role in immune activity and regulating multiple genes involved in HCC development. Low-ARID1A subtype was associated with poor clinical outcome and suggests the possibility of ARID1A as a prognostic biomarker in HCC patients.
Collapse
Affiliation(s)
- Sun Young Yim
- Department of Internal Medicine, Korea University College of Medicine, Seoul 136-701, Korea; (S.Y.Y.); (S.H.U.)
| | - Sang Hee Kang
- Department of Surgery, Korea University College of Medicine, Seoul 136-701, Korea;
| | - Ji-Hyun Shin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.-H.S.); (Y.S.J.); (B.H.S.)
| | - Yun Seong Jeong
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.-H.S.); (Y.S.J.); (B.H.S.)
| | - Bo Hwa Sohn
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.-H.S.); (Y.S.J.); (B.H.S.)
| | - Soon Ho Um
- Department of Internal Medicine, Korea University College of Medicine, Seoul 136-701, Korea; (S.Y.Y.); (S.H.U.)
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.-H.S.); (Y.S.J.); (B.H.S.)
| |
Collapse
|
24
|
Robin F, Angenard G, Cano L, Courtin-Tanguy L, Gaignard E, Khene ZE, Bergeat D, Clément B, Boudjema K, Coulouarn C, Sulpice L. Molecular profiling of stroma highlights stratifin as a novel biomarker of poor prognosis in pancreatic ductal adenocarcinoma. Br J Cancer 2020; 123:72-80. [PMID: 32376891 PMCID: PMC7341840 DOI: 10.1038/s41416-020-0863-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/22/2020] [Accepted: 04/09/2020] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a deadly cancer worldwide, as a result of a late diagnosis and limited therapeutic options. Tumour microenvironment (or stroma) plays a key role in cancer onset and progression and constitutes an intrinsic histological hallmark of PDAC. Thus we hypothesised that relevant prognostic biomarkers and therapeutic targets can be identified in the stroma. METHODS Laser microdissection of the stroma from freshly frozen PDAC was combined to gene expression profiling. Protein expression of candidate biomarkers was evaluated by immunohistochemistry on tissue microarrays (n = 80 tumours) and by ELISA in plasma samples (n = 51 patients). RESULTS A signature made of 1256 genes that significantly discriminate the stroma from the non-tumour fibrous tissue was identified. Upregulated genes were associated with inflammation and metastasis processes and linked to NF-Kappa B and TGFβ pathways. TMA analysis validated an increased expression of SFN, ADAMTS12 and CXCL3 proteins in the stroma of PDAC. Stromal expression of SFN was further identified as an independent prognostic factor of overall (p = 0.003) and disease-free survival (DFS) (p = 0.034). SFN plasma expression was significantly associated with reduced DFS (p = 0.006). CONCLUSIONS We demonstrated that gene expression changes within the stroma of PDAC correlate with tumour progression, and we identified Stratifin as a novel independent prognostic biomarker.
Collapse
Affiliation(s)
- Fabien Robin
- Univ Rennes, Inserm, Inra, CHU Rennes, Service de Chirurgie Hépatobiliaire et Digestive, Institut NuMeCan (Nutrition Metabolisms and Cancer), UMR_S 1241, Rennes, France
| | - Gaëlle Angenard
- Univ Rennes, Inserm, Inra, CHU Rennes, Service de Chirurgie Hépatobiliaire et Digestive, Institut NuMeCan (Nutrition Metabolisms and Cancer), UMR_S 1241, Rennes, France
| | - Luis Cano
- Univ Rennes, Inserm, Inra, CHU Rennes, Service de Chirurgie Hépatobiliaire et Digestive, Institut NuMeCan (Nutrition Metabolisms and Cancer), UMR_S 1241, Rennes, France
| | - Laetitia Courtin-Tanguy
- Univ Rennes, Inserm, Inra, CHU Rennes, Service de Chirurgie Hépatobiliaire et Digestive, Institut NuMeCan (Nutrition Metabolisms and Cancer), UMR_S 1241, Rennes, France
| | - Elodie Gaignard
- Univ Rennes, Inserm, Inra, CHU Rennes, Service de Chirurgie Hépatobiliaire et Digestive, Institut NuMeCan (Nutrition Metabolisms and Cancer), UMR_S 1241, Rennes, France
| | - Zine-Eddine Khene
- Univ Rennes, Inserm, Inra, CHU Rennes, Service de Chirurgie Hépatobiliaire et Digestive, Institut NuMeCan (Nutrition Metabolisms and Cancer), UMR_S 1241, Rennes, France
| | - Damien Bergeat
- Univ Rennes, Inserm, Inra, CHU Rennes, Service de Chirurgie Hépatobiliaire et Digestive, Institut NuMeCan (Nutrition Metabolisms and Cancer), UMR_S 1241, Rennes, France
| | - Bruno Clément
- Univ Rennes, Inserm, Inra, CHU Rennes, Service de Chirurgie Hépatobiliaire et Digestive, Institut NuMeCan (Nutrition Metabolisms and Cancer), UMR_S 1241, Rennes, France
| | - Karim Boudjema
- Univ Rennes, Inserm, Inra, CHU Rennes, Service de Chirurgie Hépatobiliaire et Digestive, Institut NuMeCan (Nutrition Metabolisms and Cancer), UMR_S 1241, Rennes, France
| | - Cédric Coulouarn
- Univ Rennes, Inserm, Inra, CHU Rennes, Service de Chirurgie Hépatobiliaire et Digestive, Institut NuMeCan (Nutrition Metabolisms and Cancer), UMR_S 1241, Rennes, France.
| | - Laurent Sulpice
- Univ Rennes, Inserm, Inra, CHU Rennes, Service de Chirurgie Hépatobiliaire et Digestive, Institut NuMeCan (Nutrition Metabolisms and Cancer), UMR_S 1241, Rennes, France.
| |
Collapse
|
25
|
Coulouarn C. Artificial intelligence and omics in cancer. Artif Intell Cancer 2020; 1:1-7. [DOI: 10.35713/aic.v1.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023] Open
Affiliation(s)
- Cédric Coulouarn
- Institut National de la Sante et de la Recherche Medicale (Inserm), Université de Rennes 1, Rennes F-35000, France
| |
Collapse
|
26
|
Chen Z, Tian R, She Z, Cai J, Li H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radic Biol Med 2020; 152:116-141. [PMID: 32156524 DOI: 10.1016/j.freeradbiomed.2020.02.025] [Citation(s) in RCA: 784] [Impact Index Per Article: 156.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide and is strongly associated with the presence of oxidative stress. Disturbances in lipid metabolism lead to hepatic lipid accumulation, which affects different reactive oxygen species (ROS) generators, including mitochondria, endoplasmic reticulum, and NADPH oxidase. Mitochondrial function adapts to NAFLD mainly through the downregulation of the electron transport chain (ETC) and the preserved or enhanced capacity of mitochondrial fatty acid oxidation, which stimulates ROS overproduction within different ETC components upstream of cytochrome c oxidase. However, non-ETC sources of ROS, in particular, fatty acid β-oxidation, appear to produce more ROS in hepatic metabolic diseases. Endoplasmic reticulum stress and NADPH oxidase alterations are also associated with NAFLD, but the degree of their contribution to oxidative stress in NAFLD remains unclear. Increased ROS generation induces changes in insulin sensitivity and in the expression and activity of key enzymes involved in lipid metabolism. Moreover, the interaction between redox signaling and innate immune signaling forms a complex network that regulates inflammatory responses. Based on the mechanistic view described above, this review summarizes the mechanisms that may account for the excessive production of ROS, the potential mechanistic roles of ROS that drive NAFLD progression, and therapeutic interventions that are related to oxidative stress.
Collapse
Affiliation(s)
- Ze Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Ruifeng Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Zhigang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China; Basic Medical School, Wuhan University, Wuhan, 430071, PR China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, PR China
| | - Jingjing Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China; Basic Medical School, Wuhan University, Wuhan, 430071, PR China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, PR China.
| |
Collapse
|
27
|
Schriml LM, Mitraka E, Munro J, Tauber B, Schor M, Nickle L, Felix V, Jeng L, Bearer C, Lichenstein R, Bisordi K, Campion N, Hyman B, Kurland D, Oates CP, Kibbey S, Sreekumar P, Le C, Giglio M, Greene C. Human Disease Ontology 2018 update: classification, content and workflow expansion. Nucleic Acids Res 2020; 47:D955-D962. [PMID: 30407550 PMCID: PMC6323977 DOI: 10.1093/nar/gky1032] [Citation(s) in RCA: 278] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/22/2018] [Indexed: 12/22/2022] Open
Abstract
The Human Disease Ontology (DO) (http://www.disease-ontology.org), database has undergone significant expansion in the past three years. The DO disease classification includes specific formal semantic rules to express meaningful disease models and has expanded from a single asserted classification to include multiple-inferred mechanistic disease classifications, thus providing novel perspectives on related diseases. Expansion of disease terms, alternative anatomy, cell type and genetic disease classifications and workflow automation highlight the updates for the DO since 2015. The enhanced breadth and depth of the DO’s knowledgebase has expanded the DO’s utility for exploring the multi-etiology of human disease, thus improving the capture and communication of health-related data across biomedical databases, bioinformatics tools, genomic and cancer resources and demonstrated by a 6.6× growth in DO’s user community since 2015. The DO’s continual integration of human disease knowledge, evidenced by the more than 200 SVN/GitHub releases/revisions, since previously reported in our DO 2015 NAR paper, includes the addition of 2650 new disease terms, a 30% increase of textual definitions, and an expanding suite of disease classification hierarchies constructed through defined logical axioms.
Collapse
Affiliation(s)
- Lynn M Schriml
- University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD, USA
| | | | - James Munro
- University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD, USA
| | - Becky Tauber
- University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD, USA
| | - Mike Schor
- University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD, USA
| | - Lance Nickle
- University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD, USA
| | - Victor Felix
- University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD, USA
| | - Linda Jeng
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cynthia Bearer
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | - Nicole Campion
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brooke Hyman
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - David Kurland
- New York University Langone Medical Center, Department of Neurosurgery, New York, NY, USA
| | - Connor Patrick Oates
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Siobhan Kibbey
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Chris Le
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michelle Giglio
- University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD, USA
| | - Carol Greene
- University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Nwosu ZC, Piorońska W, Battello N, Zimmer AD, Dewidar B, Han M, Pereira S, Blagojevic B, Castven D, Charlestin V, Holenya P, Lochead J, De La Torre C, Gretz N, Sajjakulnukit P, Zhang L, Ward MH, Marquardt JU, di Magliano MP, Lyssiotis CA, Sleeman J, Wölfl S, Ebert MP, Meyer C, Hofmann U, Dooley S. Severe metabolic alterations in liver cancer lead to ERK pathway activation and drug resistance. EBioMedicine 2020; 54:102699. [PMID: 32330875 PMCID: PMC7182727 DOI: 10.1016/j.ebiom.2020.102699] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/16/2020] [Accepted: 02/18/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The extracellular signal-regulated kinase (ERK) pathway regulates cell growth, and is hyper-activated and associated with drug resistance in hepatocellular carcinoma (HCC). Metabolic pathways are profoundly dysregulated in HCC. Whether an altered metabolic state is linked to activated ERK pathway and drug response in HCC is unaddressed. METHODS We deprived HCC cells of glutamine to induce metabolic alterations and performed various assays, including metabolomics (with 13C-glucose isotope tracing), microarray analysis, and cell proliferation assays. Glutamine-deprived cells were also treated with kinase inhibitors (e.g. Sorafenib, Erlotinib, U0126 amongst other MEK inhibitors). We performed bioinformatics analysis and stratification of HCC tumour microarrays to determine upregulated ERK gene signatures in patients. FINDINGS In a subset of HCC cells, the withdrawal of glutamine triggers a severe metabolic alteration and ERK phosphorylation (pERK). This is accompanied by resistance to the anti-proliferative effect of kinase inhibitors, despite pERK inhibition. High intracellular serine is a consistent feature of an altered metabolic state and contributes to pERK induction and the kinase inhibitor resistance. Blocking the ERK pathway facilitates cell proliferation by reprogramming metabolism, notably enhancing aerobic glycolysis. We have identified 24 highly expressed ERK gene signatures that their combined expression strongly indicates a dysregulated metabolic gene network in human HCC tissues. INTERPRETATION A severely compromised metabolism lead to ERK pathway induction, and primes some HCC cells to pro-survival phenotypes upon ERK pathway blockade. Our findings offer novel insights for understanding, predicting and overcoming drug resistance in liver cancer patients. FUND: DFG, BMBF and Sino-German Cooperation Project.
Collapse
Affiliation(s)
- Zeribe Chike Nwosu
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany.
| | - Weronika Piorońska
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Nadia Battello
- Luxembourg Science Center, L-4620 Differdange, Luxembourg, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-Belval, Luxembourg
| | - Andreas David Zimmer
- Signal Transduction Laboratory, Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Bedair Dewidar
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, 31527 Tanta, Egypt
| | - Mei Han
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Sharon Pereira
- Department of Medicine I, Lichtenberg Research Group, Johannes Gutenberg University, Mainz, Germany
| | - Biljana Blagojevic
- Department of Biology, Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Darko Castven
- Department of Medicine I, Lichtenberg Research Group, Johannes Gutenberg University, Mainz, Germany
| | - Verodia Charlestin
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, 46556 IN, United States
| | - Pavlo Holenya
- Department of Biology, Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Julia Lochead
- Department of Biology, Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Carolina De La Torre
- Medical Research Center, Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Research Center, Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
| | - Peter Sajjakulnukit
- Rogel Cancer Center, University of Michigan, Ann Arbor 48109 MI, United States
| | - Li Zhang
- Rogel Cancer Center, University of Michigan, Ann Arbor 48109 MI, United States
| | - Matthew H Ward
- Rogel Cancer Center, University of Michigan, Ann Arbor 48109 MI, United States
| | - Jens U Marquardt
- Department of Medicine I, Lichtenberg Research Group, Johannes Gutenberg University, Mainz, Germany
| | | | - Costas A Lyssiotis
- Rogel Cancer Center, University of Michigan, Ann Arbor 48109 MI, United States
| | - Jonathan Sleeman
- Medical Faculty Mannheim, ECAS TRIDOMUS-Gebäude Haus C, University of Heidelberg, 68167 Mannheim, Germany; IBCS-BIP, Campus Nord, Karlsruhe Institute for Technology (KIT), 76021 Karlsruhe, Germany
| | - Stefan Wölfl
- Department of Biology, Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Matthias Philip Ebert
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Christoph Meyer
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, 70376 Stuttgart, Germany
| | - Steven Dooley
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany.
| |
Collapse
|
29
|
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
|
30
|
Liu X, Xie C, Li A, Zhang Y, Liu X, Zhou S, Shen J, Huo Z, Cao W, Ma Y, Xu R, Xing Y, Xie Y, Cai S, Tang X. BEZ235 enhances chemosensitivity of paclitaxel in hepatocellular carcinoma through inhibiting the PI3K/Akt/mTOR pathway. Am J Transl Res 2019; 11:7255-7271. [PMID: 31934276 PMCID: PMC6943462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 11/27/2019] [Indexed: 06/10/2023]
Abstract
Desensitization of hepatocellular carcinoma (HCC) to paclitaxel chemotherapy is a major deterrent to successful treatment of the cancer. Abnormal activation of the PI3K/Akt/mTOR, pathway is a common outcome of chemotherapy for HCC. Therefore, we investigated whether BEZ235, a dual PI3K and mTOR inhibitor, could increase the sensitivity of HCC to paclitaxel. In vitro results showed that paclitaxel, combined with BEZ235, inhibited HCC cell proliferation and migration, arrested the cell cycle in the G2/M phase, and promoted cell apoptosis by decreasing PI3K/Akt/mTOR activity. In vivo experiments confirmed that BEZ235 enhances the anti-tumor effect of paclitaxel by reducing PI3K/Akt/mTOR activity. Immunohistochemical staining showed that paclitaxel combined with BEZ235 reduced the numbers of Ki-67- and GPC3-positive HepG2 cells in tumor tissues. We conclude that BEZ235 enhanced the sensitivity of HCC to paclitaxel, and inhibition of PI3K/Akt/mTOR signaling might be a therapeutic strategy against paclitaxel-resistant HCC.
Collapse
Affiliation(s)
- Xueke Liu
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and TechnologyHuainan 232001, China
| | - Chunmei Xie
- Department of Blood Transfusion, Guangzhou 8th People’s Hospital, Guangzhou Medical UniversityGuangzhou 510100, China
| | - Amin Li
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and TechnologyHuainan 232001, China
| | - Yinci Zhang
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and TechnologyHuainan 232001, China
| | - Xinkuang Liu
- First Affiliated Hospital of Medical College, Anhui University of Science and TechnologyHuainan 232001, China
| | - Shuping Zhou
- First Affiliated Hospital of Medical College, Anhui University of Science and TechnologyHuainan 232001, China
| | - Jing Shen
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and TechnologyHuainan 232001, China
| | - Zhen Huo
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and TechnologyHuainan 232001, China
| | - Weiya Cao
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and TechnologyHuainan 232001, China
| | - Yongfang Ma
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and TechnologyHuainan 232001, China
| | - Ruyue Xu
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and TechnologyHuainan 232001, China
| | - Yingru Xing
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and TechnologyHuainan 232001, China
| | - Yinghai Xie
- First Affiliated Hospital of Medical College, Anhui University of Science and TechnologyHuainan 232001, China
| | - Shuyu Cai
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and TechnologyHuainan 232001, China
| | - Xiaolong Tang
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health of The Ministry of Education, Anhui University of Science and TechnologyHuainan 232001, China
| |
Collapse
|
31
|
Zahid KR, Yao S, Khan ARR, Raza U, Gou D. mTOR/HDAC1 Crosstalk Mediated Suppression of ADH1A and ALDH2 Links Alcohol Metabolism to Hepatocellular Carcinoma Onset and Progression in silico. Front Oncol 2019; 9:1000. [PMID: 31637215 PMCID: PMC6787164 DOI: 10.3389/fonc.2019.01000] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is ranked the third deadliest cancer worldwide whose molecular pathogenesis is not fully understood. Although deregulated metabolic pathways have been implicated in HCC onset and progression, the mechanisms triggering this metabolic imbalance are yet to be explored. Here, we identified a gene signature coding catabolic enzymes (Cat-GS) involved in key metabolic pathways like amino acid, lipid, carbohydrate, drug, and retinol metabolism as suppressed in HCC. A higher expression of deregulated Cat-GS is associated with good survival and less aggressive disease state in HCC patients. On the other hand, we identified mTOR signaling as a key determinant in HCC onset and progression, whose hyperactivation is found associated with poor survival and aggressive disease state in HCC patients. Next, out of Cat-GS, we established two key regulators of alcohol metabolism, alcohol dehydrogenase 1A (ADH1A) and aldehyde dehydrogenase 2 (ALDH2), as being transcriptionally suppressed by histone deacetylase 1 (HDAC1) at the downstream of mTORC1 signaling. Suppressed ADH1A and ALDH2 expression aligns well with HCC-specific molecular profile and can efficiently predict disease onset and progression, whereas higher ADH1A and ALDH2 expression is associated with good survival and less aggressive disease state in HCC patients. Overall, our in silico findings suggest that transcriptional suppression of alcohol metabolism regulators, ADH1A and ALDH2, at the downstream of mTOR signaling is, in part, responsible for triggering oncogenic transformation of hepatocytes resulting in disease onset and progression in HCC.
Collapse
Affiliation(s)
- Kashif Rafiq Zahid
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Shenzhen, China.,Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering Shenzhen University, Shenzhen, China
| | - Shun Yao
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Abdur Rehman Raza Khan
- Military College of Signals, National University of Science and Technology, Rawalpindi, Pakistan
| | - Umar Raza
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
32
|
Angenard G, Merdrignac A, Louis C, Edeline J, Coulouarn C. Expression of long non-coding RNA ANRIL predicts a poor prognosis in intrahepatic cholangiocarcinoma. Dig Liver Dis 2019; 51:1337-1343. [PMID: 31040073 DOI: 10.1016/j.dld.2019.03.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/05/2019] [Accepted: 03/22/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (iCCA) is a deadly cancer worldwide associated with an increased incidence, limited therapeutic options and absence of reliable prognostic biomarkers. Long non-coding RNAs (lncRNA) emerge as relevant biomarkers in cancer being associated with tumor progression. However, lncRNA have been poorly investigated in iCCA. AIM To identify lncRNA significantly associated with the survival of patients with iCCA after tumor resection for curative intent. METHODS Gene expression profiling and Q-RT-PCR were performed from a cohort of 39 clinically well-annotated iCCA. Univariate Cox proportional hazards model with Wald Statistic was used to identify lncRNA significantly associated with overall (OS) and/or disease-free (DFS) survival. RESULTS A signature made of 9 lncRNA was identified to be significantly (P < 0.05) associated with OS and DFS, including 4 lncRNA (lnc-CDK9-1, XLOC_l2_009441, CDKN2B-AS1, HOXC13-AS) highly expressed in poor prognosis iCCA and 5 lncRNA (lnc-CCHCR1-1, lnc-AF131215.3.1, lnc-CBLB-5, COL18A1-AS2, lnc-RELL2-1) highly expressed in better prognosis iCCA. We further validated CDKN2B-AS1 (ANRIL) as a poor prognosis biomarker, not only in iCCA, but also in hepatocellular carcinoma, kidney renal clear cell carcinoma and uterine corpus endometrial carcinoma. CONCLUSIONS We report a prognosis lncRNA signature in iCCA and the clinical relevance of CDKN2B-AS1 (ANRIL) overexpression in several cancers.
Collapse
Affiliation(s)
- Gaëlle Angenard
- Inserm, Univ Rennes, Inra, Institut NuMeCan (Nutrition Metabolisms and Cancer), CHU Rennes, Centre de Lutte contre le Cancer Eugène Marquis, Rennes, France
| | - Aude Merdrignac
- Inserm, Univ Rennes, Inra, Institut NuMeCan (Nutrition Metabolisms and Cancer), CHU Rennes, Centre de Lutte contre le Cancer Eugène Marquis, Rennes, France
| | - Corentin Louis
- Inserm, Univ Rennes, Inra, Institut NuMeCan (Nutrition Metabolisms and Cancer), CHU Rennes, Centre de Lutte contre le Cancer Eugène Marquis, Rennes, France
| | - Julien Edeline
- Inserm, Univ Rennes, Inra, Institut NuMeCan (Nutrition Metabolisms and Cancer), CHU Rennes, Centre de Lutte contre le Cancer Eugène Marquis, Rennes, France
| | - Cédric Coulouarn
- Inserm, Univ Rennes, Inra, Institut NuMeCan (Nutrition Metabolisms and Cancer), CHU Rennes, Centre de Lutte contre le Cancer Eugène Marquis, Rennes, France.
| |
Collapse
|
33
|
Zhang Y, Bharathi SS, Beck ME, Goetzman ES. The fatty acid oxidation enzyme long-chain acyl-CoA dehydrogenase can be a source of mitochondrial hydrogen peroxide. Redox Biol 2019; 26:101253. [PMID: 31234015 PMCID: PMC6597861 DOI: 10.1016/j.redox.2019.101253] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/31/2019] [Accepted: 06/10/2019] [Indexed: 11/29/2022] Open
Abstract
Fatty acid oxidation (FAO)-driven H2O2 has been shown to be a major source of oxidative stress in several tissues and disease states. Here, we established that the mitochondrial flavoprotein long-chain acyl-CoA dehydrogenase (LCAD), which catalyzes a key step in mitochondrial FAO, directly produces H2O2in vitro by leaking electrons to oxygen. Kinetic analysis of recombinant human LCAD showed that it produces H2O2 15-fold faster than the related mitochondrial enzyme very long-chain acyl-CoA dehydrogenase (VLCAD), but 50-fold slower than a bona fide peroxisomal acyl-CoA oxidase. The rate of H2O2 formation by human LCAD is slow compared to its activity as a dehydrogenase (about 1%). However, expression of hLCAD in HepG2 cells is sufficient to significantly increase H2O2 in the presence of fatty acids. Liver mitochondria from LCAD−/− mice, but not VLCAD−/− mice, produce significantly less H2O2 during incubation with fatty acids. Finally, we observe highest LCAD expression in human liver, followed by kidney, lung, and pancreas. Based on our data, we propose that the presence of LCAD drives H2O2 formation in response to fatty acids in these tissues.
Collapse
Affiliation(s)
- Yuxun Zhang
- Department of Pediatrics, Division of Medical Genetics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Sivakama S Bharathi
- Department of Pediatrics, Division of Medical Genetics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Megan E Beck
- Department of Pediatrics, Division of Medical Genetics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Eric S Goetzman
- Department of Pediatrics, Division of Medical Genetics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
34
|
Huminiecki L, Horbańczuk J. The functional genomic studies of resveratrol in respect to its anti-cancer effects. Biotechnol Adv 2018; 36:1699-1708. [DOI: 10.1016/j.biotechadv.2018.02.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/25/2018] [Accepted: 02/20/2018] [Indexed: 12/24/2022]
|
35
|
Liu M, Zhang L, Li H, Hinoue T, Zhou W, Ohtani H, El-Khoueiry A, Daniels J, O’Connell C, Dorff TB, Lu Q, Weisenberger DJ, Liang G. Integrative Epigenetic Analysis Reveals Therapeutic Targets to the DNA Methyltransferase Inhibitor Guadecitabine (SGI-110) in Hepatocellular Carcinoma. Hepatology 2018; 68:1412-1428. [PMID: 29774579 PMCID: PMC6173644 DOI: 10.1002/hep.30091] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 04/28/2018] [Indexed: 12/14/2022]
Abstract
UNLABELLED There is an urgent need to develop more effective therapies for hepatocellular carcinoma (HCC) because of its aggressiveness. Guadecitabine (SGI-110) is a second-generation DNA methyltransferase inhibitor (DNMTi), which is currently in clinical trials for HCC and shows greater stability and performance over first-generation DNMTis. In order to identify potential therapeutic targets of SGI-110 for clinical trials, HCC cell lines (SNU398, HepG2, and SNU475) were used to evaluate the effects of transient SGI-110 treatment by an integrative analysis of DNA methylation, nucleosome accessibility, gene expression profiles, and its clinical relevance by comparison to The Cancer Genome Atlas (TCGA) HCC clinical data. Each HCC cell line represents a different DNA methylation subtype of primary HCC tumors based on TCGA data. After SGI-110 treatment, all cell lines were sensitive to SGI-110 with prolonged antiproliferation effects. Expression of up-regulated genes, including tumor suppressors, was positively correlated with nucleosome accessibility and negatively correlated with gene promoter DNA methylation. Alternatively, expression of down-regulated genes, such as oncogenes, was negatively correlated with nucleosome accessibility and positively correlated with gene body DNA methylation. SGI-110 can also act as a dual inhibitor to down-regulate polycomb repressive complex 2 (PRC2) genes by demethylating their gene bodies, resulting in reactivation of PRC2 repressed genes without involvement of DNA methylation. Furthermore, it can up-regulate endogenous retroviruses to reactivate immune pathways. Finally, about 48% of frequently altered genes in primary HCC tumors can be reversed by SGI-110 treatment. CONCLUSION Our integrative analysis has successfully linked the antitumor effects of SGI-110 to detailed epigenetic alterations in HCC cells, identified potential therapeutic targets, and provided a rationale for combination treatments of SGI-110 with immune checkpoint therapies.
Collapse
Affiliation(s)
- Minmin Liu
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA,Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Lian Zhang
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA,Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongtao Li
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | | | - Wanding Zhou
- Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Hitoshi Ohtani
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA,Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Anthony El-Khoueiry
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - John Daniels
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Casey O’Connell
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Tanya B. Dorff
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Daniel J. Weisenberger
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Gangning Liang
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA,Corresponding author: Gangning Liang, , 1441 Eastlake Ave. NOR7344, Los Angeles, CA 90089, Tel: 323-865-0470
| |
Collapse
|
36
|
Morse MA, Sun W, Kim R, He AR, Abada PB, Mynderse M, Finn RS. The Role of Angiogenesis in Hepatocellular Carcinoma. Clin Cancer Res 2018; 25:912-920. [PMID: 30274981 DOI: 10.1158/1078-0432.ccr-18-1254] [Citation(s) in RCA: 385] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/17/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) accounts for about 90% of all primary liver cancers and is the second leading cause of cancer-related deaths worldwide. The hypervascular nature of most HCC tumors underlines the importance of angiogenesis in the pathobiology of these tumors. Several angiogenic pathways have been identified as being dysregulated in HCC, suggesting they may be involved in the development and pathogenesis of HCC. These data provide practical targets for systemic treatments such as those targeting the vascular endothelial growth factor receptor and its ligand. However, the clinical relevance of other more recently identified angiogenic pathways in HCC pathogenesis or treatment remains unclear. Research into molecular profiles and validation of prognostic or predictive biomarkers will be required to identify the patient subsets most likely to experience meaningful benefit from this important class of agents.
Collapse
Affiliation(s)
- Michael A Morse
- Department of Medicine, Division of Medical Oncology, Duke University Health System, Durham, North Carolina.
| | - Weijing Sun
- Division of Medical Oncology, University of Kansas School of Medicine, Kansas City, Kansas
| | - Richard Kim
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aiwu Ruth He
- Department of Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | | | | | - Richard S Finn
- Department of Medicine, Division of Hematology/Oncology, Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
37
|
Lian Q, Wang S, Zhang G, Wang D, Luo G, Tang J, Chen L, Gu J. HCCDB: A Database of Hepatocellular Carcinoma Expression Atlas. GENOMICS PROTEOMICS & BIOINFORMATICS 2018; 16:269-275. [PMID: 30266410 PMCID: PMC6205074 DOI: 10.1016/j.gpb.2018.07.003] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is highly heterogeneous in nature and has been one of the most common cancer types worldwide. To ensure repeatability of identified gene expression patterns and comprehensively annotate the transcriptomes of HCC, we carefully curated 15 public HCC expression datasets that cover around 4000 clinical samples and developed the database HCCDB to serve as a one-stop online resource for exploring HCC gene expression with user-friendly interfaces. The global differential gene expression landscape of HCC was established by analyzing the consistently differentially expressed genes across multiple datasets. Moreover, a 4D metric was proposed to fully characterize the expression pattern of each gene by integrating data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx). To facilitate a comprehensive understanding of gene expression patterns in HCC, HCCDB also provides links to third-party databases on drug, proteomics, and literatures, and graphically displays the results from computational analyses, including differential expression analysis, tissue-specific and tumor-specific expression analysis, survival analysis, and co-expression analysis. HCCDB is freely accessible at http://lifeome.net/database/hccdb.
Collapse
Affiliation(s)
- Qiuyu Lian
- MOE Key Laboratory of Bioinformatics, Beijing National Research Center for Information Science and Technology, Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Shicheng Wang
- MOE Key Laboratory of Bioinformatics, Beijing National Research Center for Information Science and Technology, Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Guchao Zhang
- MOE Key Laboratory of Bioinformatics, Beijing National Research Center for Information Science and Technology, Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Dongfang Wang
- MOE Key Laboratory of Bioinformatics, Beijing National Research Center for Information Science and Technology, 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
| | - Jing Tang
- 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, Beijing National Research Center for Information Science and Technology, Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
38
|
Nwosu ZC, Battello N, Rothley M, Piorońska W, Sitek B, Ebert MP, Hofmann U, Sleeman J, Wölfl S, Meyer C, Megger DA, Dooley S. Liver cancer cell lines distinctly mimic the metabolic gene expression pattern of the corresponding human tumours. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:211. [PMID: 30176945 PMCID: PMC6122702 DOI: 10.1186/s13046-018-0872-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/31/2018] [Indexed: 02/06/2023]
Abstract
Background Although metabolism is profoundly altered in human liver cancer, the extent to which experimental models, e.g. cell lines, mimic those alterations is unresolved. Here, we aimed to determine the resemblance of hepatocellular carcinoma (HCC) cell lines to human liver tumours, specifically in the expression of deregulated metabolic targets in clinical tissue samples. Methods We compared the overall gene expression profile of poorly-differentiated (HLE, HLF, SNU-449) to well-differentiated (HUH7, HEPG2, HEP3B) HCC cell lines in three publicly available microarray datasets. Three thousand and eighty-five differentially expressed genes in ≥2 datasets (P < 0.05) were used for pathway enrichment and gene ontology (GO) analyses. Further, we compared the topmost gene expression, pathways, and GO from poorly differentiated cell lines to the pattern from four human HCC datasets (623 tumour tissues). In well- versus poorly differentiated cell lines, and in representative models HLE and HUH7 cells, we specifically assessed the expression pattern of 634 consistently deregulated metabolic genes in human HCC. These data were complemented by quantitative PCR, proteomics, metabolomics and assessment of response to thirteen metabolism-targeting compounds in HLE versus HUH7 cells. Results We found that poorly-differentiated HCC cells display upregulated MAPK/RAS/NFkB signaling, focal adhesion, and downregulated complement/coagulation cascade, PPAR-signaling, among pathway alterations seen in clinical tumour datasets. In HLE cells, 148 downregulated metabolic genes in liver tumours also showed low gene/protein expression – notably in fatty acid β-oxidation (e.g. ACAA1/2, ACADSB, HADH), urea cycle (e.g. CPS1, ARG1, ASL), molecule transport (e.g. SLC2A2, SLC7A1, SLC25A15/20), and amino acid metabolism (e.g. PHGDH, PSAT1, GOT1, GLUD1). In contrast, HUH7 cells showed a higher expression of 98 metabolic targets upregulated in tumours (e.g. HK2, PKM, PSPH, GLUL, ASNS, and fatty acid synthesis enzymes ACLY, FASN). Metabolomics revealed that the genomic portrait of HLE cells co-exist with profound reliance on glutamine to fuel tricarboxylic acid cycle, whereas HUH7 cells use both glucose and glutamine. Targeting glutamine pathway selectively suppressed the proliferation of HLE cells. Conclusions We report a yet unappreciated distinct expression pattern of clinically-relevant metabolic genes in HCC cell lines, which could enable the identification and therapeutic targeting of metabolic vulnerabilities at various liver cancer stages. Electronic supplementary material The online version of this article (10.1186/s13046-018-0872-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zeribe C Nwosu
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3 (H42, Floor 4), 68167, Mannheim, Germany.,Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3 (H42, Floor 4), 68167, Mannheim, Germany
| | - Nadia Battello
- Luxembourg Science Center, 50 rue Emile Mark, L-4620, Differdange, Luxembourg
| | - Melanie Rothley
- Institut für Toxikologie und Genetik, Campus Nord, Karlsruhe Institute for Technology (KIT), Postfach 3640, 76021, Karlsruhe, Germany.,Medical Faculty Mannheim, CBTM TRIDOMUS-Gebäude Haus C, University of Heidelberg, 68167, Mannheim, Germany
| | - Weronika Piorońska
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3 (H42, Floor 4), 68167, Mannheim, Germany.,Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3 (H42, Floor 4), 68167, Mannheim, Germany
| | - Barbara Sitek
- Medizinisches Proteom-Center, Department of Clinical Proteomics, Ruhr-Universität Bochum, Bochum, Germany
| | - Matthias P Ebert
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3 (H42, Floor 4), 68167, Mannheim, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, 70376, Stuttgart, Germany
| | - Jonathan Sleeman
- Institut für Toxikologie und Genetik, Campus Nord, Karlsruhe Institute for Technology (KIT), Postfach 3640, 76021, Karlsruhe, Germany.,Medical Faculty Mannheim, CBTM TRIDOMUS-Gebäude Haus C, University of Heidelberg, 68167, Mannheim, Germany
| | - Stefan Wölfl
- Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, University of Heidelberg, 69120, Heidelberg, Germany
| | - Christoph Meyer
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3 (H42, Floor 4), 68167, Mannheim, Germany.,Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3 (H42, Floor 4), 68167, Mannheim, Germany
| | - Dominik A Megger
- Institut für Toxikologie und Genetik, Campus Nord, Karlsruhe Institute for Technology (KIT), Postfach 3640, 76021, Karlsruhe, Germany.,Institute of Virology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Steven Dooley
- Department of Medicine II, Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3 (H42, Floor 4), 68167, Mannheim, Germany. .,Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3 (H42, Floor 4), 68167, Mannheim, Germany.
| |
Collapse
|
39
|
Vaquero J, Lobe C, Tahraoui S, Clapéron A, Mergey M, Merabtene F, Wendum D, Coulouarn C, Housset C, Desbois-Mouthon C, Praz F, Fouassier L. The IGF2/IR/IGF1R Pathway in Tumor Cells and Myofibroblasts Mediates Resistance to EGFR Inhibition in Cholangiocarcinoma. Clin Cancer Res 2018; 24:4282-4296. [PMID: 29716918 DOI: 10.1158/1078-0432.ccr-17-3725] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/03/2018] [Accepted: 04/27/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Cholangiocarcinoma (CCA) is a desmoplastic tumor of the biliary tree in which epidermal growth factor receptor (EGFR) is overexpressed and contributes to cancer progression. Although EGFR has been envisaged as a target for therapy, treatment with tyrosine kinase inhibitors (TKI) such as erlotinib did not provide therapeutic benefit in patients with CCA, emphasizing the need to investigate resistance mechanisms against EGFR inhibition.Experimental Design: Resistant CCA cells to EGFR inhibition were obtained upon long-time exposure of cells with erlotinib. Cell signaling, viability, migration, and spheroid growth were determined in vitro, and tumor growth was evaluated in CCA xenograft models.Results: Erlotinib-resistant CCA cells displayed metastasis-associated signatures that correlated with a marked change in cell plasticity associated with an epithelial-mesenchymal transition (EMT) and a cancer stem cell (CSC)-like phenotype. Resistant cells exhibited an upregulation of insulin receptor (IR) and insulin-like growth factor (IGF) 1 receptor (IGF1R), along with an increase in IGF2 expression. IR/IGF1R inhibition reduced EMT and CSC-like traits in resistant cells. In vivo, tumors developed from resistant CCA cells were larger and exhibited a more prominent stromal compartment, enriched in cancer-associated fibroblasts (CAF). Pharmacological coinhibition of EGFR and IR/IGF1R reduced tumor growth and stromal compartment in resistant tumors. Modeling of CCA-CAF crosstalk showed that IGF2 expressed by fibroblasts boosted IR/IGF1R signaling in resistant cells. Furthermore, IR/IGF1R signaling positively regulated fibroblast proliferation and activation.Conclusions: To escape EGFR-TKI treatment, CCA tumor cells develop an adaptive mechanism by undergoing an IR/IGF1R-dependent phenotypic switch, involving a contribution of stromal cells. Clin Cancer Res; 24(17); 4282-96. ©2018 AACR.
Collapse
Affiliation(s)
- Javier Vaquero
- Sorbonne University, INSERM, Saint-Antoine Research Center (CRSA), Paris, France
- Fondation ARC, Villejuif, France
- LPP, CNRS, Ecole Polytech., Univ. Paris-Sud, Observatoire de Paris, Univ. Paris-Saclay, Sorbonne University, PSL Research University, Paris, France
| | - Cindy Lobe
- Sorbonne University, INSERM, Saint-Antoine Research Center (CRSA), Paris, France
| | - Sylvana Tahraoui
- Sorbonne University, INSERM, Saint-Antoine Research Center (CRSA), Paris, France
| | - Audrey Clapéron
- Sorbonne University, INSERM, Saint-Antoine Research Center (CRSA), Paris, France
| | - Martine Mergey
- Sorbonne University, INSERM, Saint-Antoine Research Center (CRSA), Paris, France
| | - Fatiha Merabtene
- Sorbonne University, INSERM, Saint-Antoine Research Center (CRSA), Paris, France
| | - Dominique Wendum
- Sorbonne University, INSERM, Saint-Antoine Research Center (CRSA), Paris, France
- Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Department of Pathology, Paris, France
| | - Cédric Coulouarn
- INSERM, INRA, Univ Rennes 1, Univ Bretagne Loire, Nutrition Metabolisms and Cancer (NuMeCan), Rennes, France
| | - Chantal Housset
- Sorbonne University, INSERM, Saint-Antoine Research Center (CRSA), Paris, France
- Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Hepatology Department, Paris, France
| | | | - Françoise Praz
- Sorbonne University, INSERM, Saint-Antoine Research Center (CRSA), Paris, France
| | - Laura Fouassier
- Sorbonne University, INSERM, Saint-Antoine Research Center (CRSA), Paris, France.
| |
Collapse
|
40
|
Merdrignac A, Angenard G, Allain C, Petitjean K, Bergeat D, Bellaud P, Fautrel A, Turlin B, Clément B, Dooley S, Sulpice L, Boudjema K, Coulouarn C. A novel transforming growth factor beta-induced long noncoding RNA promotes an inflammatory microenvironment in human intrahepatic cholangiocarcinoma. Hepatol Commun 2018; 2:254-269. [PMID: 29507901 PMCID: PMC5831019 DOI: 10.1002/hep4.1142] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 11/20/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is a deadly liver primary cancer associated with poor prognosis and limited therapeutic opportunities. Active transforming growth factor beta (TGFβ) signaling is a hallmark of the iCCA microenvironment. However, the impact of TGFβ on the transcriptome of iCCA tumor cells has been poorly investigated. Here, we have identified a specific TGFβ signature of genes commonly deregulated in iCCA cell lines, namely HuCCT1 and Huh28. Novel coding and noncoding TGFβ targets were identified, including a TGFβ-induced long noncoding RNA (TLINC), formerly known as cancer susceptibility candidate 15 (CASC15). TLINC is a general target induced by TGFβ in hepatic and nonhepatic cell types. In iCCA cell lines, the expression of a long and short TLINC isoform was associated with an epithelial or mesenchymal phenotype, respectively. Both isoforms were detected in the nucleus and cytoplasm. The long isoform of TLINC was associated with a migratory phenotype in iCCA cell lines and with the induction of proinflammatory cytokines, including interleukin 8, both in vitro and in resected human iCCA. TLINC was also identified as a tumor marker expressed in both epithelial and stroma cells. In nontumor livers, TLINC was only expressed in specific portal areas with signs of ductular reaction and inflammation. Finally, we provide experimental evidence of circular isoforms of TLINC, both in iCCA cells treated with TGFβ and in resected human iCCA. Conclusion: We identify a novel TGFβ-induced long noncoding RNA up-regulated in human iCCA and associated with an inflammatory microenvironment. (Hepatology Communications 2018;2:254-269).
Collapse
Affiliation(s)
- Aude Merdrignac
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| | - Gaëlle Angenard
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| | - Coralie Allain
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| | - Kilian Petitjean
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| | - Damien Bergeat
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| | - Pascale Bellaud
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| | - Allain Fautrel
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| | - Bruno Turlin
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| | - Bruno Clément
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| | - Steven Dooley
- Department of Medicine II, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Laurent Sulpice
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| | - Karim Boudjema
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| | - Cédric Coulouarn
- Institut National de la Santé et de la Recherche Médicale, INRA, Université de Rennes, CHU Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Service de Chirurgie Hépatobiliaire et Digestive, Biosit, Biogenouest, Core Facility H2P2 and CRB SantéRennesFrance
| |
Collapse
|
41
|
Bévant K, Coulouarn C. Landscape of genomic alterations in hepatocellular carcinoma: current knowledge and perspectives for targeted therapies. Hepatobiliary Surg Nutr 2017; 6:404-407. [PMID: 29312976 DOI: 10.21037/hbsn.2017.10.02] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Kévin Bévant
- Inserm, Inra, Univ Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Rennes, France
| | - Cédric Coulouarn
- Inserm, Inra, Univ Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Rennes, France
| |
Collapse
|
42
|
Soukupova J, Malfettone A, Hyroššová P, Hernández-Alvarez MI, Peñuelas-Haro I, Bertran E, Junza A, Capellades J, Giannelli G, Yanes O, Zorzano A, Perales JC, Fabregat I. Role of the Transforming Growth Factor-β in regulating hepatocellular carcinoma oxidative metabolism. Sci Rep 2017; 7:12486. [PMID: 28970582 PMCID: PMC5624948 DOI: 10.1038/s41598-017-12837-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 09/14/2017] [Indexed: 02/07/2023] Open
Abstract
Transforming Growth Factor beta (TGF-β) induces tumor cell migration and invasion. However, its role in inducing metabolic reprogramming is poorly understood. Here we analyzed the metabolic profile of hepatocellular carcinoma (HCC) cells that show differences in TGF-β expression. Oxygen consumption rate (OCR), extracellular acidification rate (ECAR), metabolomics and transcriptomics were performed. Results indicated that the switch from an epithelial to a mesenchymal/migratory phenotype in HCC cells is characterized by reduced mitochondrial respiration, without significant differences in glycolytic activity. Concomitantly, enhanced glutamine anaplerosis and biosynthetic use of TCA metabolites were proved through analysis of metabolite levels, as well as metabolic fluxes from U-13C6-Glucose and U-13C5-Glutamine. This correlated with increase in glutaminase 1 (GLS1) expression, whose inhibition reduced cell migration. Experiments where TGF-β function was activated with extracellular TGF-β1 or inhibited through TGF-β receptor I silencing showed that TGF-β induces a switch from oxidative metabolism, coincident with a decrease in OCR and the upregulation of glutamine transporter Solute Carrier Family 7 Member 5 (SLC7A5) and GLS1. TGF-β also regulated the expression of key genes involved in the flux of glycolytic intermediates and fatty acid metabolism. Together, these results indicate that autocrine activation of the TGF-β pathway regulates oxidative metabolism in HCC cells.
Collapse
Affiliation(s)
- Jitka Soukupova
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Andrea Malfettone
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Petra Hyroššová
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
- Department of Physiological Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - María-Isabel Hernández-Alvarez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Irene Peñuelas-Haro
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Esther Bertran
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Alexandra Junza
- Metabolomics Platform, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, Tarragona, Spain
- Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Jordi Capellades
- Metabolomics Platform, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, Tarragona, Spain
- Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Gianluigi Giannelli
- National Institute of Gastroenterology IRCCS "S. De Bellis", Castellana Grotte Bari, Italy
| | - Oscar Yanes
- Metabolomics Platform, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, Tarragona, Spain
- Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - José Carlos Perales
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
- Department of Physiological Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Isabel Fabregat
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain.
- Department of Physiological Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
43
|
Dolezal JM, Wang H, Kulkarni S, Jackson L, Lu J, Ranganathan S, Goetzman ES, Bharathi SS, Beezhold K, Byersdorfer CA, Prochownik EV. Sequential adaptive changes in a c-Myc-driven model of hepatocellular carcinoma. J Biol Chem 2017; 292:10068-10086. [PMID: 28432125 DOI: 10.1074/jbc.m117.782052] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/12/2017] [Indexed: 01/09/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common cancer that frequently overexpresses the c-Myc (Myc) oncoprotein. Using a mouse model of Myc-induced HCC, we studied the metabolic, biochemical, and molecular changes accompanying HCC progression, regression, and recurrence. These involved altered rates of pyruvate and fatty acid β-oxidation and the likely re-directing of glutamine into biosynthetic rather than energy-generating pathways. Initial tumors also showed reduced mitochondrial mass and differential contributions of electron transport chain complexes I and II to respiration. The uncoupling of complex II's electron transport function from its succinate dehydrogenase activity also suggested a mechanism by which Myc generates reactive oxygen species. RNA sequence studies revealed an orderly progression of transcriptional changes involving pathways pertinent to DNA damage repair, cell cycle progression, insulin-like growth factor signaling, innate immunity, and further metabolic re-programming. Only a subset of functions deregulated in initial tumors was similarly deregulated in recurrent tumors thereby indicating that the latter can "normalize" some behaviors to suit their needs. An interactive and freely available software tool was developed to allow continued analyses of these and other transcriptional profiles. Collectively, these studies define the metabolic, biochemical, and molecular events accompanyingHCCevolution, regression, and recurrence in the absence of any potentially confounding therapies.
Collapse
Affiliation(s)
| | - Huabo Wang
- From the Divisions of Hematology/Oncology and
| | | | | | - Jie Lu
- From the Divisions of Hematology/Oncology and
| | | | | | | | - Kevin Beezhold
- Bone Marrow and Stem Cell Transplantation, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania 15224
| | - Craig A Byersdorfer
- Bone Marrow and Stem Cell Transplantation, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania 15224
| | - Edward V Prochownik
- From the Divisions of Hematology/Oncology and .,the Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, and.,the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232
| |
Collapse
|