1
|
Dwyer BJ, Tirnitz-Parker JEE. Patient-derived organoid models to decode liver pathophysiology. Trends Endocrinol Metab 2025; 36:235-248. [PMID: 39191607 DOI: 10.1016/j.tem.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Liver diseases represent a growing global health challenge, and the increasing prevalence of obesity and metabolic disorders is set to exacerbate this crisis. To meet evolving regulatory demands, patient-specific in vitro liver models are essential for understanding disease mechanisms and developing new therapeutic approaches. Organoid models, which faithfully recapitulate liver biology, can be established from both non-malignant and malignant liver tissues, offering insight into various liver conditions, from acute injuries to chronic diseases and cancer. Improved understanding of liver microenvironments, innovative biomaterials, and advanced imaging techniques now facilitate comprehensive and unbiased data analysis, paving the way for personalised medicine. In this review, we discuss state-of-the-art patient-derived liver organoid models, recent technological advancements, and strategies to enhance their clinical impact.
Collapse
Affiliation(s)
- Benjamin J Dwyer
- Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia; Liver Cancer Collaborative, Perth, WA, Australia; www.livercancercollaborative.au.
| | - Janina E E Tirnitz-Parker
- Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia; Liver Cancer Collaborative, Perth, WA, Australia; www.livercancercollaborative.au.
| |
Collapse
|
2
|
Zhu H, Chen H, Zhu X, Zhang B, Jin C. Diagnostic lncRNA high expression for liver patients prognosis and medication guidance: a systematic review and meta-analysis. Front Pharmacol 2024; 15:1462512. [PMID: 39211780 PMCID: PMC11357924 DOI: 10.3389/fphar.2024.1462512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The study of long non-coding RNAs (lncRNAs) has gained significant attention due to their roles in regulating gene expression and their potential as diagnostic biomarkers. This systematic review and meta-analysis aimed to evaluate the diagnostic value of high-expression lncRNAs in liver disease patients, including those with hepatitis, cirrhosis, and hepatocellular carcinoma (HCC). METHODS A comprehensive literature search was conducted across multiple electronic databases, including PubMed, Embase, Web of Science, and Cochrane Library, up to July 2024. Studies were included if they investigated the expression of lncRNAs in liver disease patients and evaluated their diagnostic performance. The Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) tool was used to assess the quality of included studies. Pooled sensitivity, specificity, diagnostic odds ratios (DOR), and summary receiver operating characteristic (SROC) curves were calculated using a bivariate random-effects model. RESULTS Nine studies involving 888 samples were included in the meta-analysis. The pooled hazard ratio (HR) for overall survival (OS) was 2.01 (95% CI: 1.71-2.36), indicating a significant association between high lncRNA expression and poor liver disease outcomes. Subgroup analyses revealed a pooled odds ratio (OR) of 1.99 (95% CI: 1.53-2.60) for tissue samples and 8.62 (95% CI: 1.16-63.71) for blood samples, suggesting a stronger diagnostic value for blood-based lncRNAs. The funnel plots indicated minimal publication bias, and sensitivity analyses confirmed the robustness of the findings. CONCLUSION High-expression lncRNAs show significant potential as diagnostic biomarkers for liver diseases, offering non-invasive, accurate, and timely diagnostic information. Despite the promising results, further research is needed to standardize detection methods, elucidate the biological functions of lncRNAs, and validate their clinical utility in diverse patient populations. Integrating lncRNA biomarkers with traditional diagnostic approaches could enhance diagnostic accuracy and improve patient management and outcomes in liver disease.
Collapse
Affiliation(s)
- Hengzhou Zhu
- Department of Oncology, Wuxi Hospital of Triditional Chinese Medicine, Wuxi, China
| | - Haoyan Chen
- Department of Respiratory, Wuxi Hospital of Triditional Chinese Medicine, Wuxi, China
| | - Xiaodan Zhu
- Department of Oncology, Wuxi Hospital of Triditional Chinese Medicine, Wuxi, China
| | - Baonan Zhang
- Department of Oncology, Wuxi Hospital of Triditional Chinese Medicine, Wuxi, China
| | - Chunhui Jin
- Department of Oncology, Wuxi Hospital of Triditional Chinese Medicine, Wuxi, China
| |
Collapse
|
3
|
Lu Z, Wang X, Feng J, Chai W, Wang W, Wang Q, Yang S, Yang W, Su Y, Mou W, Peng Y, Wang H, Gui J. Intratumoral CXCR4 hi neutrophils display ferroptotic and immunosuppressive signatures in hepatoblastoma. Front Immunol 2024; 15:1363454. [PMID: 38487536 PMCID: PMC10937446 DOI: 10.3389/fimmu.2024.1363454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 02/07/2024] [Indexed: 03/17/2024] Open
Abstract
Pediatric hepatoblastoma (HB) is the most common primary liver malignancy in infants and children. With great diversity and plasticity, tumor-infiltrating neutrophils were one of the most determining factors for poor prognosis in many malignant tumors. In this study, through bulk RNA sequencing for sorted blood and tumor-infiltrated neutrophils and comparison of neutrophils in tumor and para-tumor tissue by single-cell sequencing, we found that intratumoral neutrophils were composed of heterogenous functional populations at different development stages. Our study showed that terminally differentiated neutrophils with active ferroptosis prevailed in tumor tissue, whereas, in para-tumor, pre-fate naïve neutrophils were dominant and ferroptotic neutrophils dispersed in a broad spectrum of cell maturation. Gene profiling and in vitro T-cell coculture experiment confirmed that one of main functional intratumoral neutrophils was mainly immunosuppressive, which relied on the activation of ferroptosis. Combining the bulk RNA-seq, scRNA-seq data, and immunochemistry staining of tumor samples, CXCL12/CXCR4 chemotaxis pathway was suggested to mediate the migration of neutrophils in tumors as CXCR4 highly expressed by intratumoral neutrophils and its ligand CXCL12 expressed much higher level in tumor than that in para-tumor. Moreover, our study pinpointed that infiltrated CXCR4hi neutrophils, regardless of their differential distribution of cell maturation status in HB tumor and para-tumor regions, were the genuine perpetrators for immune suppression. Our data characterized the ferroptosis-dependent immunosuppression energized by intratumoral CXCR4 expression neutrophils and suggest a potential cell target for cancer immunotherapies.
Collapse
Affiliation(s)
- Zhengjing Lu
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xiaolin Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jun Feng
- Department of Surgical Oncology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Wenjia Chai
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Wei Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Qixin Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Shen Yang
- Department of Surgical Oncology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Wei Yang
- Department of Surgical Oncology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Yan Su
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Ministry of Education, Key Laboratory of Major Diseases in Children, Hematology Oncology Center, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Wenjun Mou
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Yun Peng
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Huanmin Wang
- Department of Surgical Oncology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jingang Gui
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| |
Collapse
|
4
|
Huth T, Dreher EC, Lemke S, Fritzsche S, Sugiyanto RN, Castven D, Ibberson D, Sticht C, Eiteneuer E, Jauch A, Pusch S, Albrecht T, Goeppert B, Candia J, Wang XW, Ji J, Marquardt JU, Nahnsen S, Schirmacher P, Roessler S. Chromosome 8p engineering reveals increased metastatic potential targetable by patient-specific synthetic lethality in liver cancer. SCIENCE ADVANCES 2023; 9:eadh1442. [PMID: 38134284 PMCID: PMC10745716 DOI: 10.1126/sciadv.adh1442] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
Large-scale chromosomal aberrations are prevalent in human cancer, but their function remains poorly understood. We established chromosome-engineered hepatocellular carcinoma cell lines using CRISPR-Cas9 genome editing. A 33-mega-base pair region on chromosome 8p (chr8p) was heterozygously deleted, mimicking a frequently observed chromosomal deletion. Using this isogenic model system, we delineated the functional consequences of chr8p loss and its impact on metastatic behavior and patient survival. We found that metastasis-associated genes on chr8p act in concert to induce an aggressive and invasive phenotype characteristic for chr8p-deleted tumors. Genome-wide CRISPR-Cas9 viability screening in isogenic chr8p-deleted cells served as a powerful tool to find previously unidentified synthetic lethal targets and vulnerabilities accompanying patient-specific chromosomal alterations. Using this target identification strategy, we showed that chr8p deletion sensitizes tumor cells to targeting of the reactive oxygen sanitizing enzyme Nudix hydrolase 17. Thus, chromosomal engineering allowed for the identification of novel synthetic lethalities specific to chr8p loss of heterozygosity.
Collapse
Affiliation(s)
- Thorben Huth
- Heidelberg University, Medical Faculty, Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Emely C. Dreher
- Heidelberg University, Medical Faculty, Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Steffen Lemke
- Quantitative Biology Center (QBiC), University of Tübingen, 72076 Tübingen, Germany
- Department of Peptide-based Immunotherapy, University and University Hospital Tübingen, 72076 Tübingen, Germany
- Institute for Cell Biology, Department of Immunology, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
| | - Sarah Fritzsche
- Heidelberg University, Medical Faculty, Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Raisatun N. Sugiyanto
- Heidelberg University, Medical Faculty, Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Darko Castven
- Department of Medicine I, University Medical Center Schleswig Holstein, 23538 Lübeck, Germany
| | - David Ibberson
- Deep Sequencing Core Facility, CellNetworks Excellence Cluster, Heidelberg University, 69120 Heidelberg, Germany
| | - Carsten Sticht
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Eva Eiteneuer
- Heidelberg University, Medical Faculty, Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Anna Jauch
- Institute of Human Genetics, Heidelberg University, 69120 Heidelberg, Germany
| | - Stefan Pusch
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Thomas Albrecht
- Heidelberg University, Medical Faculty, Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Benjamin Goeppert
- Heidelberg University, Medical Faculty, Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
- Institute of Pathology and Neuropathology, RKH Klinikum Ludwigsburg, 71640 Ludwigsburg, Germany
| | - Julián Candia
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis and Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Junfang Ji
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Jens U. Marquardt
- Department of Medicine I, University Medical Center Schleswig Holstein, 23538 Lübeck, Germany
| | - Sven Nahnsen
- Quantitative Biology Center (QBiC), University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- Biomedical Data Science, Department of Computer Science, University of Tübingen, 72076 Tübingen, Germany
- The M3 Research Center, University of Tübingen, 72076 Tübingen, Germany
| | - Peter Schirmacher
- Heidelberg University, Medical Faculty, Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Stephanie Roessler
- Heidelberg University, Medical Faculty, Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
5
|
Duwe L, Munoz-Garrido P, Lewinska M, Lafuente-Barquero J, Satriano L, Høgdall D, Taranta A, Nielsen BS, Ghazal A, Matter MS, Banales JM, Aldana BI, Gao YT, Marquardt JU, Roberts LR, Oliveira RC, Koshiol J, O'Rourke CJ, Andersen JB. MicroRNA-27a-3p targets FoxO signalling to induce tumour-like phenotypes in bile duct cells. J Hepatol 2023; 78:364-375. [PMID: 36848245 DOI: 10.1016/j.jhep.2022.10.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND & AIMS Cholangiocarcinoma (CCA) is a heterogeneous and lethal malignancy, the molecular origins of which remain poorly understood. MicroRNAs (miRs) target diverse signalling pathways, functioning as potent epigenetic regulators of transcriptional output. We aimed to characterise miRNome dysregulation in CCA, including its impact on transcriptome homeostasis and cell behaviour. METHODS Small RNA sequencing was performed on 119 resected CCAs, 63 surrounding liver tissues, and 22 normal livers. High-throughput miR mimic screens were performed in three primary human cholangiocyte cultures. Integration of patient transcriptomes and miRseq together with miR screening data identified an oncogenic miR for characterization. MiR-mRNA interactions were investigated by a luciferase assay. MiR-CRISPR knockout cells were generated and phenotypically characterized in vitro (proliferation, migration, colony, mitochondrial function, glycolysis) and in vivo using subcutaneous xenografts. RESULTS In total, 13% (140/1,049) of detected miRs were differentially expressed between CCA and surrounding liver tissues, including 135 that were upregulated in tumours. CCA tissues were characterised by higher miRNome heterogeneity and miR biogenesis pathway expression. Unsupervised hierarchical clustering of tumour miRNomes identified three subgroups, including distal CCA-enriched and IDH1 mutant-enriched subgroups. High-throughput screening of miR mimics uncovered 71 miRs that consistently increased proliferation of three primary cholangiocyte models and were upregulated in CCA tissues regardless of anatomical location, among which only miR-27a-3p had consistently increased expression and activity in several cohorts. FoxO signalling was predominantly downregulated by miR-27a-3p in CCA, partially through targeting of FOXO1. MiR-27a knockout increased FOXO1 levels in vitro and in vivo, impeding tumour behaviour and growth. CONCLUSIONS The miRNomes of CCA tissues are highly remodelled, impacting transcriptome homeostasis in part through regulation of transcription factors like FOXO1. MiR-27a-3p arises as an oncogenic vulnerability in CCA. IMPACT AND IMPLICATIONS Cholangiocarcinogenesis entails extensive cellular reprogramming driven by genetic and non-genetic alterations, but the functional roles of these non-genetic events remain poorly understood. By unveiling global miRNA upregulation in patient tumours and their functional ability to increase proliferation of cholangiocytes, these small non-coding RNAs are implicated as critical non-genetic alterations promoting biliary tumour initiation. These findings identify possible mechanisms for transcriptome rewiring during transformation, with potential implications for patient stratification.
Collapse
Affiliation(s)
- Lea Duwe
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Patricia Munoz-Garrido
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Monika Lewinska
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Juan Lafuente-Barquero
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Letizia Satriano
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Dan Høgdall
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark; Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Andrzej Taranta
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | | | - Awaisa Ghazal
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Matthias S Matter
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, China
| | - Jens U Marquardt
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, 23558 Lübeck, Germany
| | - Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Rui C Oliveira
- Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Institute of Biophysics, Faculty of Medicine, University of Coimbra, Portugal; Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Jill Koshiol
- Division of Cancer Epidemiology and Genetics, NIH, USA
| | - Colm J O'Rourke
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark
| | - Jesper B Andersen
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Ole Maaløes Vej 5, 2200, Copenhagen N, Denmark.
| |
Collapse
|
6
|
Chulkova SV, Loginov VI, Podluzhnyi DV, Egorova AV, Syskova AY, Semichev DG, Gladilina IA, Kudashkin NE. [The role of molecular genetic factors in the development of cholangiocellular carcinoma]. Arkh Patol 2022; 84:76-83. [PMID: 35639847 DOI: 10.17116/patol20228403176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The article lists the main inducers of cholangiocarcinogenesis. The main inflammatory mediators (IL-6, nitric oxide, COX2) have been considered. Data on the study of gene mutations in cholangiocarcinomas are presented. The spectrum of genetic mutations depends on the biliary cancer origin (FGFR2 with intrahepatic cholangiocarcinoma, PRKACA, PRKACB with extrahepatic cholangiocarcinoma). Mutations in the KRAS, TP53, ARIAD1A genes are common in extrahepatic bile duct cancer. The role of epigenetic changes such as DNA hypermethylation, histone modifications, chromatin remodeling, as well as disturbances in miRNA expression is presented. A number of epigenetic features, such as the presence of a TP53 mutations with hypermethylation of p14ARF, DAPK, and/or ASC, correlate with a more aggressive course of the disease. The role of the SOX17 gene in the development of drug resistance is highlighted. The study of the molecular genetic features of extrahepatic bile duct cancer can help to better understand the pathogenesis of this type of tumor, to establish new prognostic and diagnostic markers of the disease.
Collapse
Affiliation(s)
- S V Chulkova
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia.,N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - V I Loginov
- Scientific Research Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - D V Podluzhnyi
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - A V Egorova
- N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - A Yu Syskova
- N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - D G Semichev
- N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - I A Gladilina
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia.,N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| | - N E Kudashkin
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia.,N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
7
|
Abstract
The evolutionary history of hepatobiliary cancers is embedded in their genomes. By analysing their catalogue of somatic mutations and the DNA sequence context in which they occur, it is possible to infer the mechanisms underpinning tumorigenesis. These mutational signatures reflect the exogenous and endogenous origins of genetic damage as well as the capacity of hepatobiliary cells to repair and replicate DNA. Genomic analysis of thousands of patients with hepatobiliary cancers has highlighted the diversity of mutagenic processes active in these malignancies, highlighting a prominent source of the inter-cancer-type, inter-patient, intertumour and intratumoural heterogeneity that is observed clinically. However, a substantial proportion of mutational signatures detected in hepatocellular carcinoma and biliary tract cancer remain of unknown cause, emphasizing the important contribution of processes yet to be identified. Exploiting mutational signatures to retrospectively understand hepatobiliary carcinogenesis could advance preventative management of these aggressive tumours as well as potentially predict treatment response and guide the development of therapies targeting tumour evolution.
Collapse
|
8
|
Simoni-Nieves A, Salas-Silva S, Chávez-Rodríguez L, Escobedo-Calvario A, Desoteux M, Bucio L, Souza V, Miranda-Labra RU, Muñoz-Espinosa LE, Coulouarn C, Gutiérrez-Ruiz MC, Marquardt JU, Gomez-Quiroz LE. The Consumption of Cholesterol-Enriched Diets Conditions the Development of a Subtype of HCC with High Aggressiveness and Poor Prognosis. Cancers (Basel) 2021; 13:1721. [PMID: 33917315 PMCID: PMC8038696 DOI: 10.3390/cancers13071721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/19/2021] [Accepted: 03/09/2021] [Indexed: 12/16/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and progression to non-alcoholic steatohepatitis (NASH) result as a consequence of diverse conditions, mainly unbalanced diets. Particularly, high-fat and cholesterol content, as well as carbohydrates, such as those commonly ingested in Western countries, frequently drive adverse metabolic alterations in the liver and promote NAFLD development. Lipid liver overload is also one of the main risk factors for initiation and progression of hepatocellular carcinoma (HCC), but detailed knowledge on the relevance of high nutritional cholesterol remains elusive. We were aimed to characterize HCC development in mice fed with a Western diet (high in lipids and cholesterol) and to identify molecular alterations that define a subtype of liver cancer induced by lipid overload. Mice under western or high cholesterol diets more frequently developed tumors with a more aggressive phenotype than animals fed with a chow diet. Associated changes involved macrophage infiltration, angiogenesis, and stemness features. RNA-seq revealed a specific gene expression signature (Slc41a; Fabp5; Igdcc4 and Mthfd1l) resembling the adverse phenotypic features and poor clinical outcomes seen in patients with HCC. In conclusion; consumption of lipid enriched diets; particularly cholesterol; could accelerate HCC development with an aggressive phenotype and poor prognosis.
Collapse
Affiliation(s)
- Arturo Simoni-Nieves
- Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Mexico City 09340, Mexico; (A.S.-N.); (S.S.-S.); (L.C.-R.); (A.E.-C.)
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico; (L.B.); (V.S.); (R.U.M.-L.); (M.C.G.-R.)
| | - Soraya Salas-Silva
- Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Mexico City 09340, Mexico; (A.S.-N.); (S.S.-S.); (L.C.-R.); (A.E.-C.)
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico; (L.B.); (V.S.); (R.U.M.-L.); (M.C.G.-R.)
| | - Lisette Chávez-Rodríguez
- Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Mexico City 09340, Mexico; (A.S.-N.); (S.S.-S.); (L.C.-R.); (A.E.-C.)
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico; (L.B.); (V.S.); (R.U.M.-L.); (M.C.G.-R.)
| | - Alejandro Escobedo-Calvario
- Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Mexico City 09340, Mexico; (A.S.-N.); (S.S.-S.); (L.C.-R.); (A.E.-C.)
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico; (L.B.); (V.S.); (R.U.M.-L.); (M.C.G.-R.)
| | - Matthis Desoteux
- Centre de Lutte contre le Cancer Eugène Marquis, Inserm, Univ Rennes, COSS (Chemistry, Oncogenesis Stress Signaling), UMR_S 1242, 35042 Rennes, France; (M.D.); (C.C.)
| | - Leticia Bucio
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico; (L.B.); (V.S.); (R.U.M.-L.); (M.C.G.-R.)
- Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional, IIB, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City 14080, Mexico
| | - Verónica Souza
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico; (L.B.); (V.S.); (R.U.M.-L.); (M.C.G.-R.)
- Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional, IIB, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City 14080, Mexico
| | - Roxana U. Miranda-Labra
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico; (L.B.); (V.S.); (R.U.M.-L.); (M.C.G.-R.)
- Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional, IIB, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City 14080, Mexico
| | - Linda E. Muñoz-Espinosa
- Liver Unit, Department of Internal Medicine, “Dr. José E. González” University Hospital, Universidad Autónoma de Nuevo León, Monterrey 64460, NL, Mexico;
| | - Cédric Coulouarn
- Centre de Lutte contre le Cancer Eugène Marquis, Inserm, Univ Rennes, COSS (Chemistry, Oncogenesis Stress Signaling), UMR_S 1242, 35042 Rennes, France; (M.D.); (C.C.)
| | - María Concepción Gutiérrez-Ruiz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico; (L.B.); (V.S.); (R.U.M.-L.); (M.C.G.-R.)
- Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional, IIB, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City 14080, Mexico
| | - Jens U. Marquardt
- Department of Medicine I, University Hospital Schleswig-Holstein, 23562 Lübeck, Germany;
| | - Luis E. Gomez-Quiroz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico; (L.B.); (V.S.); (R.U.M.-L.); (M.C.G.-R.)
- Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional, IIB, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City 14080, Mexico
| |
Collapse
|
9
|
Verduin M, Hoeben A, De Ruysscher D, Vooijs M. Patient-Derived Cancer Organoids as Predictors of Treatment Response. Front Oncol 2021; 11:641980. [PMID: 33816288 PMCID: PMC8012903 DOI: 10.3389/fonc.2021.641980] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Patient-derived cancer organoids have taken a prominent role in pre-clinical and translational research and have been generated for most common solid tumors. Cancer organoids have been shown to retain key genetic and phenotypic characteristics of their tissue of origin, tumor subtype and maintain intratumoral heterogeneity and therefore have the potential to be used as predictors for individualized treatment response. In this review, we highlight studies that have used cancer organoids to compare the efficacy of standard-of-care and targeted combination treatments with clinical patient response. Furthermore, we review studies using cancer organoids to identify new anti-cancer treatments using drug screening. Finally, we discuss the current limitations and improvements needed to understand the full potential of cancer organoids as avatars for clinical management of cancer therapy.
Collapse
Affiliation(s)
- Maikel Verduin
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ann Hoeben
- Department of Medical Oncology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Marc Vooijs
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
10
|
O'Rourke CJ, Munoz-Garrido P, Andersen JB. Molecular Targets in Cholangiocarcinoma. Hepatology 2021; 73 Suppl 1:62-74. [PMID: 32304327 DOI: 10.1002/hep.31278] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 12/18/2022]
Abstract
Cholangiocarcinoma (CCA) encompasses a heterogeneous collection of malignancies for which diagnostic biomarkers are lacking and population screening is infeasible because of its status as a rare disease. Coupled with high postsurgical recurrence rates among the minority of patients diagnosed at resectable stages, systemic clinical management will inevitably be required for the majority of patients with CCA with recurrent and advanced disease. In this review, we discuss the therapeutic potential of different classes of molecular targets at various stages of development in CCA, including those targeted to the tumor epithelia (oncogenic, developmental, metabolic, epigenomic) and tumor microenvironment (angiogenesis, checkpoint regulation). Furthermore, we discuss the successes and failures of CCA-targeted therapies, emphasizing key lessons learned that should pave the way for future molecular target evaluation in this uncommon yet bona fide target-rich disease.
Collapse
Affiliation(s)
- Colm J O'Rourke
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Patricia Munoz-Garrido
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Hernandez S, Simoni-Nieves A, Gerardo-Ramírez M, Torres S, Fucho R, Gonzalez J, Castellanos-Tapia L, Hernández-Pando R, Tejero-Barrera E, Bucio L, Souza V, Miranda-Labra R, Fernández-Checa JC, Marquardt JU, Gomez-Quiroz LE, García-Ruiz C, Gutiérrez-Ruiz MC. GDF11 restricts aberrant lipogenesis and changes in mitochondrial structure and function in human hepatocellular carcinoma cells. J Cell Physiol 2020; 236:4076-4090. [PMID: 33174245 DOI: 10.1002/jcp.30151] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/18/2022]
Abstract
Growth differentiation factor 11 (GDF11) has been characterized as a key regulator of differentiation in cells that retain stemness features. Recently, it has been reported that GDF11 exerts tumor-suppressive properties in hepatocellular carcinoma cells, decreasing clonogenicity, proliferation, spheroid formation, and cellular function, all associated with a decrement in stemness features, resulting in mesenchymal to epithelial transition and loss of aggressiveness. The aim of the present work was to investigate the mechanism associated with the tumor-suppressive properties displayed by GDF11 in liver cancer cells. Hepatocellular carcinoma-derived cell lines were exposed to GDF11 (50 ng/ml), RNA-seq analysis in Huh7 cell line revealed that GDF11 exerted profound transcriptomic impact, which involved regulation of cholesterol metabolic process, steroid metabolic process as well as key signaling pathways, resembling endoplasmic reticulum-related functions. Cholesterol and triglycerides determination in Huh7 and Hep3B cells treated with GDF11 exhibited a significant decrement in the content of these lipids. The mTOR signaling pathway was downregulated, and this was associated with a reduction in key proteins involved in the mevalonate pathway. In addition, real-time metabolism assessed by Seahorse technology showed abridged glycolysis as well as glycolytic capacity, closely related to an impaired oxygen consumption rate and decrement in adenosine triphosphate production. Finally, transmission electron microscopy revealed mitochondrial abnormalities, such as cristae disarrangement, consistent with metabolic changes. Results provide evidence that GDF11 impairs cancer cell metabolism targeting lipid homeostasis, glycolysis, and mitochondria function and morphology.
Collapse
Affiliation(s)
- Sharik Hernandez
- Posgrado en Biología Experimental, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Arturo Simoni-Nieves
- Posgrado en Biología Experimental, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Monserrat Gerardo-Ramírez
- Posgrado en Biología Experimental, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | - Sandra Torres
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Liver Unit Hospital Clinic, IDIBAPS and CIBERehd, Barcelona, Spain, Southern California Research Center for ALPD and Cirrhosis, KECK School of Medicine, USC, Los Angeles, USA.,Southern California Research Center for ALPD and Cirrhosis, KECK School of Medicine, USC, Los Angeles, USA
| | - Raquel Fucho
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Liver Unit Hospital Clinic, IDIBAPS and CIBERehd, Barcelona, Spain, Southern California Research Center for ALPD and Cirrhosis, KECK School of Medicine, USC, Los Angeles, USA.,Southern California Research Center for ALPD and Cirrhosis, KECK School of Medicine, USC, Los Angeles, USA
| | - Jonathan Gonzalez
- Posgrado en Biología Experimental, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico
| | | | | | | | - Leticia Bucio
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Translacional, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
| | - Verónica Souza
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Translacional, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
| | - Roxana Miranda-Labra
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Translacional, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
| | - José C Fernández-Checa
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Liver Unit Hospital Clinic, IDIBAPS and CIBERehd, Barcelona, Spain, Southern California Research Center for ALPD and Cirrhosis, KECK School of Medicine, USC, Los Angeles, USA.,Southern California Research Center for ALPD and Cirrhosis, KECK School of Medicine, USC, Los Angeles, USA
| | - Jens U Marquardt
- Department of Medicine I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Luis E Gomez-Quiroz
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Translacional, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
| | - Carmen García-Ruiz
- Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, CSIC, Liver Unit Hospital Clinic, IDIBAPS and CIBERehd, Barcelona, Spain, Southern California Research Center for ALPD and Cirrhosis, KECK School of Medicine, USC, Los Angeles, USA.,Southern California Research Center for ALPD and Cirrhosis, KECK School of Medicine, USC, Los Angeles, USA
| | - María C Gutiérrez-Ruiz
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Translacional, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cardiología Ignacio Chavez, Mexico City, Mexico
| |
Collapse
|
12
|
Oh S, Jo Y, Jung S, Yoon S, Yoo KH. From genome sequencing to the discovery of potential biomarkers in liver disease. BMB Rep 2020. [PMID: 32475383 PMCID: PMC7330805 DOI: 10.5483/bmbrep.2020.53.6.074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Chronic liver disease progresses through several stages, fatty liver, steatohepatitis, cirrhosis, and eventually, it leads to hepatocellular carcinoma (HCC) over a long period of time. Since a large proportion of patients with HCC are accompanied by cirrhosis, it is considered to be an important factor in the diagnosis of liver cancer. This is because cirrhosis leads to an irreversible harmful effect, but the early stages of chronic liver disease could be reversed to a healthy state. Therefore, the discovery of biomarkers that could identify the early stages of chronic liver disease is important to prevent serious liver damage. Biomarker discovery at liver cancer and cirrhosis has enhanced the development of sequencing technology. Next generation sequencing (NGS) is one of the representative technical innovations in the biological field in the recent decades and it is the most important thing to design for research on what type of sequencing methods are suitable and how to handle the analysis steps for data integration. In this review, we comprehensively summarized NGS techniques for identifying genome, transcriptome, DNA methylome and 3D/4D chromatin structure, and introduced framework of processing data set and integrating multi-omics data for uncovering biomarkers.
Collapse
Affiliation(s)
- Sumin Oh
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
- Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Yeeun Jo
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| | - Sungju Jung
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| | - Sumin Yoon
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| | - Kyung Hyun Yoo
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
- Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| |
Collapse
|
13
|
Funk K, Czauderna C, Klesse R, Becker D, Hajduk J, Oelgeklaus A, Reichenbach F, Fimm-Todt F, Lauterwasser J, Galle PR, Marquardt JU, Edlich F. BAX Redistribution Induces Apoptosis Resistance and Selective Stress Sensitivity in Human HCC. Cancers (Basel) 2020; 12:cancers12061437. [PMID: 32486514 PMCID: PMC7352885 DOI: 10.3390/cancers12061437] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 05/27/2020] [Indexed: 12/23/2022] Open
Abstract
Cancer therapies induce differential cell responses, ranging from efficient cell death to complete stress resistance. The BCL-2 proteins BAX and BAK govern the cellular decision between survival and mitochondrial apoptosis. Therefore, the status of BAX/BAK regulation can predict the cellular apoptosis predisposition. Relative BAX/BAK localization was analyzed in tumor and corresponding non-tumor samples from 34 hepatocellular carcinoma (HCC) patients. Key transcriptome changes and gene expression profiles related to the status of BAX regulation were applied to two independent cohorts including over 500 HCC patients. The prediction of apoptotic response was tested using cell lines and polyclonal tumor isolates. Cellular protection from BAX was confirmed by challenging cells with mitochondrial BAX. We discovered a subgroup of HCC with selective protection from BAX-dependent apoptosis. BAX-protected tumors showed enrichment of signaling pathways associated with oxidative stress response and DNA repair as well as increased genetic heterogeneity. Gene expression profiles characteristic to BAX-specific protection are enriched in poorly differentiated HCCs and show significant association to the overall survival of HCC patients. Consistently, addiction to DNA repair of BAX-protected cancer cells caused selective sensitivity to PARP inhibition. Molecular characteristics of BAX-protected HCC were enriched in cells challenged with mitochondrial BAX. Our results demonstrate that predisposition to BAX activation impairs tumor biology in HCC. Selective BAX inhibition or lack thereof delineates distinct subgroups of HCC patients with molecular features and differential response pattern to apoptotic stimuli and inhibition of DNA repair mechanisms.
Collapse
Affiliation(s)
- Kathrin Funk
- Institute for Biochemistry and Molecular Biology, University of Freiburg, 79104 Freiburg, Germany; (K.F.); (R.K.); (A.O.); (F.R.); (F.F.-T.); (J.L.)
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Carolin Czauderna
- Department of Medicine, Lichtenberg Research Group, University Mainz, 55116 Mainz, Germany; (C.C.); (D.B.); (J.H.); (P.R.G.)
| | - Ramona Klesse
- Institute for Biochemistry and Molecular Biology, University of Freiburg, 79104 Freiburg, Germany; (K.F.); (R.K.); (A.O.); (F.R.); (F.F.-T.); (J.L.)
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Diana Becker
- Department of Medicine, Lichtenberg Research Group, University Mainz, 55116 Mainz, Germany; (C.C.); (D.B.); (J.H.); (P.R.G.)
| | - Jovana Hajduk
- Department of Medicine, Lichtenberg Research Group, University Mainz, 55116 Mainz, Germany; (C.C.); (D.B.); (J.H.); (P.R.G.)
| | - Aline Oelgeklaus
- Institute for Biochemistry and Molecular Biology, University of Freiburg, 79104 Freiburg, Germany; (K.F.); (R.K.); (A.O.); (F.R.); (F.F.-T.); (J.L.)
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Frank Reichenbach
- Institute for Biochemistry and Molecular Biology, University of Freiburg, 79104 Freiburg, Germany; (K.F.); (R.K.); (A.O.); (F.R.); (F.F.-T.); (J.L.)
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Franziska Fimm-Todt
- Institute for Biochemistry and Molecular Biology, University of Freiburg, 79104 Freiburg, Germany; (K.F.); (R.K.); (A.O.); (F.R.); (F.F.-T.); (J.L.)
| | - Joachim Lauterwasser
- Institute for Biochemistry and Molecular Biology, University of Freiburg, 79104 Freiburg, Germany; (K.F.); (R.K.); (A.O.); (F.R.); (F.F.-T.); (J.L.)
| | - Peter R. Galle
- Department of Medicine, Lichtenberg Research Group, University Mainz, 55116 Mainz, Germany; (C.C.); (D.B.); (J.H.); (P.R.G.)
| | - Jens U. Marquardt
- Department of Medicine I, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23562 Lübeck, Germany
- Correspondence: (J.U.M.); (F.E.)
| | - Frank Edlich
- Institute for Biochemistry and Molecular Biology, University of Freiburg, 79104 Freiburg, Germany; (K.F.); (R.K.); (A.O.); (F.R.); (F.F.-T.); (J.L.)
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
- Correspondence: (J.U.M.); (F.E.)
| |
Collapse
|
14
|
Zarrinpar A, Kim UB, Boominathan V. Phenotypic Response and Personalized Medicine in Liver Cancer and Transplantation: Approaches to Complex Systems. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ali Zarrinpar
- Department of Surgery, College of MedicineUniversity of Florida Gainesville FL 32610 USA
- Department of Biochemistry and Molecular Biology, College of MedicineUniversity of Florida Gainesville FL 32610 USA
- Department of Bioengineering, Herbert Wertheim College of EngineeringUniversity of Florida Gainesville FL 32610 USA
| | - Un Bi Kim
- Department of Surgery, College of MedicineUniversity of Florida Gainesville FL 32610 USA
| | - Vijay Boominathan
- Department of Surgery, College of MedicineUniversity of Florida Gainesville FL 32610 USA
| |
Collapse
|
15
|
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
|
16
|
van Tienderen GS, Groot Koerkamp B, IJzermans JNM, van der Laan LJW, Verstegen MMA. Recreating Tumour Complexity in a Dish: Organoid Models to Study Liver Cancer Cells and their Extracellular Environment. Cancers (Basel) 2019; 11:E1706. [PMID: 31683901 PMCID: PMC6896153 DOI: 10.3390/cancers11111706] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
Primary liver cancer, consisting predominantly of hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), remains one of the most lethal malignancies worldwide. This high malignancy is related to the complex and dynamic interactions between tumour cells, stromal cells and the extracellular environment. Novel in vitro models that can recapitulate the tumour are essential in increasing our understanding of liver cancer. Herein, primary liver cancer-derived organoids have opened up new avenues due to their patient-specificity, self-organizing ability and potential recapitulation of many of the tumour properties. Organoids are solely of epithelial origin, but incorporation into co-culture models can enable the investigation of the cellular component of the tumour microenvironment. However, the extracellular component also plays a vital role in cancer progression and representation is lacking within current in vitro models. In this review, organoid technology is discussed in the context of liver cancer models through comparisons to other cell culture systems. In addition, the role of the tumour extracellular environment in primary liver cancer will be highlighted with an emphasis on its importance in in vitro modelling. Converging novel organoid-based models with models incorporating the native tumour microenvironment could lead to experimental models that can better recapitulate liver tumours in vivo.
Collapse
Affiliation(s)
- Gilles S van Tienderen
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands.
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands.
| | - Jan N M IJzermans
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands.
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands.
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC-University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands.
| |
Collapse
|
17
|
Models for Understanding Resistance to Chemotherapy in Liver Cancer. Cancers (Basel) 2019; 11:cancers11111677. [PMID: 31671735 PMCID: PMC6896032 DOI: 10.3390/cancers11111677] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/19/2022] Open
Abstract
The lack of response to pharmacological treatment constitutes a substantial limitation in the handling of patients with primary liver cancers (PLCs). The existence of active mechanisms of chemoresistance (MOCs) in hepatocellular carcinoma, cholangiocarcinoma, and hepatoblastoma hampers the usefulness of chemotherapy. A better understanding of MOCs is needed to develop strategies able to overcome drug refractoriness in PLCs. With this aim, several experimental models are commonly used. These include in vitro cell-free assays using subcellular systems; studies with primary cell cultures; cancer cell lines or heterologous expression systems; multicellular models, such as spheroids and organoids; and a variety of in vivo models in rodents, such as subcutaneous and orthotopic tumor xenografts or chemically or genetically induced liver carcinogenesis. Novel methods to perform programmed genomic edition and more efficient techniques to isolate circulating microvesicles offer new opportunities for establishing useful experimental tools for understanding the resistance to chemotherapy in PLCs. In the present review, using three criteria for information organization: (1) level of research; (2) type of MOC; and (3) type of PLC, we have summarized the advantages and limitations of the armamentarium available in the field of pharmacological investigation of PLC chemoresistance.
Collapse
|
18
|
Gérard C, Di-Luoffo M, Gonay L, Caruso S, Couchy G, Loriot A, Castven D, Tao J, Konobrocka K, Cordi S, Monga SP, Hanert E, Marquardt JU, Zucman-Rossi J, Lemaigre FP. Dynamics and predicted drug response of a gene network linking dedifferentiation with beta-catenin dysfunction in hepatocellular carcinoma. J Hepatol 2019; 71:323-332. [PMID: 30953666 DOI: 10.1016/j.jhep.2019.03.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 03/15/2019] [Accepted: 03/21/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Alterations of individual genes variably affect the development of hepatocellular carcinoma (HCC). Thus, we aimed to characterize the function of tumor-promoting genes in the context of gene regulatory networks (GRNs). METHODS Using data from The Cancer Genome Atlas, from the LIRI-JP (Liver Cancer - RIKEN, JP project), and from our transcriptomic, transfection and mouse transgenic experiments, we identify a GRN which functionally links LIN28B-dependent dedifferentiation with dysfunction of β-catenin (CTNNB1). We further generated and validated a quantitative mathematical model of the GRN using human cell lines and in vivo expression data. RESULTS We found that LIN28B and CTNNB1 form a GRN with SMARCA4, Let-7b (MIRLET7B), SOX9, TP53 and MYC. GRN functionality is detected in HCC and gastrointestinal cancers, but not in other cancer types. GRN status negatively correlates with HCC prognosis, and positively correlates with hyperproliferation, dedifferentiation and HGF/MET pathway activation, suggesting that it contributes to a transcriptomic profile typical of the proliferative class of HCC. The mathematical model predicts how the expression of GRN components changes when the expression of another GRN member varies or is inhibited by a pharmacological drug. The dynamics of GRN component expression reveal distinct cell states that can switch reversibly in normal conditions, and irreversibly in HCC. The mathematical model is available via a web-based tool which can evaluate the GRN status of HCC samples and predict the impact of therapeutic agents on the GRN. CONCLUSIONS We conclude that identification and modelling of the GRN provide insights into the prognosis of HCC and the mechanisms by which tumor-promoting genes impact on HCC development. LAY SUMMARY Hepatocellular carcinoma (HCC) is a heterogeneous disease driven by the concomitant deregulation of several genes functionally organized as networks. Here, we identified a gene regulatory network involved in a subset of HCCs. This subset is characterized by increased proliferation and poor prognosis. We developed a mathematical model which uncovers the dynamics of the network and allows us to predict the impact of a therapeutic agent, not only on its specific target but on all the genes belonging to the network.
Collapse
Affiliation(s)
- Claude Gérard
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Mickaël Di-Luoffo
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Léolo Gonay
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium; Earth and Life Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Stefano Caruso
- Centre de Recherche des Cordeliers, Sorbonne Universités, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Gabrielle Couchy
- Centre de Recherche des Cordeliers, Sorbonne Universités, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Axelle Loriot
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Darko Castven
- Department of Medicine I, Johannes Gutenberg University, Mainz, Germany
| | - Junyan Tao
- Department of Pathology, Medicine and the Pittsburgh Liver Research Center, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Sabine Cordi
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Satdarshan P Monga
- Department of Pathology, Medicine and the Pittsburgh Liver Research Center, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Emmanuel Hanert
- Earth and Life Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Jens U Marquardt
- Department of Medicine I, Johannes Gutenberg University, Mainz, Germany
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Sorbonne Universités, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | | |
Collapse
|
19
|
O'Rourke CJ, Lafuente-Barquero J, Andersen JB. Epigenome Remodeling in Cholangiocarcinoma. Trends Cancer 2019; 5:335-350. [PMID: 31208696 DOI: 10.1016/j.trecan.2019.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 12/22/2022]
Abstract
Cholangiocarcinoma (CCA) comprises a heterogeneous collection of malignancies arising within the biliary tract, characterized by late diagnosis, innate chemoresistance, and abysmal prognosis. Sequencing data have uncovered recurrent mutations in diverse epigenetic regulators, implicating epigenetic destabilization at the root of these tumors. However, few studies have characterized biliary tumor epigenomes. In this Opinion article, we argue that an epigenome-oriented approach to CCA could establish diverse interconnections between many key aspects of research on this disease, including molecular heterogeneity, diverse cells of origin, and prominent tumor microenvironments. Moreover, we discuss plausible causes of epigenome dysregulation in biliary tumors, including genetic, epigenetic, metabolic, microenvironmental, and physiological factors. Lastly, we assess the translational potential of epigenomics in CCA to uncover robust biomarkers and therapeutic opportunities for this growing group of patients with limited treatment options.
Collapse
Affiliation(s)
- Colm J O'Rourke
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Juan Lafuente-Barquero
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark.
| |
Collapse
|