1
|
Arechederra M, Bik E, Rojo C, Elurbide J, Elizalde M, Kruk B, Krasnodębski M, Pertkiewicz J, Kozieł S, Grąt M, Raszeja‐Wyszomirska J, Rullan M, Alkorta‐Aranburu G, Oyón D, Fernández‐Barrena MG, Candels LS, Białek A, Krupa Ł, Schneider KM, Urman J, Strnad P, Trautwein C, Milkiewicz P, Krawczyk M, Ávila MA, Berasain C. Mutational Analysis of Bile Cell-Free DNA in Primary Sclerosing Cholangitis: A Pilot Study. Liver Int 2025; 45:e70049. [PMID: 40029142 PMCID: PMC11874897 DOI: 10.1111/liv.70049] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Primary sclerosing cholangitis (PSC) is a chronic liver disease characterised by inflammation and fibrosis of the bile ducts, conferring an increased risk of cholangiocarcinoma (CCA). However, detecting CCA early in PSC patients remains challenging due to the limited sensitivity of conventional diagnostic methods, including imaging or bile duct brush cytology during endoscopic retrograde cholangiopancreatography (ERCP). This study aims to evaluate the potential of bile cell-free DNA (cfDNA) mutational analysis, termed the Bilemut assay, as a tool for CCA detection in PSC patients. METHODS Sixty-three PSC patients undergoing ERCP due to biliary strictures were prospectively recruited. Bile samples were collected, and cfDNA was extracted and analysed using the Oncomine Pan-Cancer Cell-Free assay. Twenty healthy liver donors were included for comparison. Samples with a mutant allele frequency (MAF) ≥ 0.1% were considered positive. Correlations between mutational status and clinical characteristics were assessed. RESULTS cfDNA mutational analysis was successful in all bile samples. Mutations predominantly in KRAS, GNAS, and TP53 were detected in 36.5% (23/63) of PSC patients, compared to 10% (2/20) of healthy donors (p = 0.0269). The clinical characteristics of Bilemut-positive and -negative patients were comparable, though there was a trend towards a lower prevalence of inflammatory bowel disease in the Bilemut-positive group. Among PSC patients diagnosed with CCA during follow-up, 75% were Bilemut-positive, suggesting an association between mutational status and malignancy risk. CONCLUSIONS Mutational analysis of cfDNA obtained from bile collected from PSC patients undergoing ERCP is feasible. Implementing the Bilemut assay may help identify patients needing closer surveillance and further imaging studies.
Collapse
Affiliation(s)
- Maria Arechederra
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUNUniversity of NavarraPamplonaSpain
- IdiSNANavarra Institute for Health ResearchPamplonaSpain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute)MadridSpain
| | - Emil Bik
- Liver and Internal Medicine Unit, Department of General, Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
| | - Carla Rojo
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUNUniversity of NavarraPamplonaSpain
| | - Jasmin Elurbide
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUNUniversity of NavarraPamplonaSpain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute)MadridSpain
| | - María Elizalde
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUNUniversity of NavarraPamplonaSpain
| | - Beata Kruk
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
| | - Maciej Krasnodębski
- Department of General Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
| | - Jan Pertkiewicz
- Department of General Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
| | - Sławomir Kozieł
- Department of General Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
| | - Michał Grąt
- Department of General Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
| | - Joanna Raszeja‐Wyszomirska
- Liver and Internal Medicine Unit, Department of General, Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
| | - Maria Rullan
- IdiSNANavarra Institute for Health ResearchPamplonaSpain
- Department of Gastroenterology and HepatologyNavarra University HospitalPamplonaSpain
| | | | - Daniel Oyón
- Department of Gastroenterology and HepatologyHospital General Universitario Gregorio MarañónMadridSpain
| | - Maite G. Fernández‐Barrena
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUNUniversity of NavarraPamplonaSpain
- IdiSNANavarra Institute for Health ResearchPamplonaSpain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute)MadridSpain
| | - Lena S. Candels
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH Aachen, Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE LIVER)AachenGermany
| | - Andrzej Białek
- Department of GastroenterologyPomeranian Medical UniversitySzczecinPoland
| | - Łukasz Krupa
- Department of Gastroenterology and Hepatology With Internal Disease UnitTeaching Hospital No 1 in RzeszówRzeszówPoland
- Medical DepartmentUniversity of RzeszówRzeszówPoland
| | - Kai M. Schneider
- Department of Medicine 1University Hospital Carl Gustav Carus Dresden, Technische Universität (TU)DresdenGermany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU)DresdenGermany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of TechnologyDresdenGermany
- Department of Medicine IIIUniversity Hospital RWTH AachenAachenGermany
| | - Jesús Urman
- IdiSNANavarra Institute for Health ResearchPamplonaSpain
- Department of Gastroenterology and HepatologyNavarra University HospitalPamplonaSpain
| | - Pavel Strnad
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH Aachen, Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE LIVER)AachenGermany
| | - Christian Trautwein
- Department of ToxicologyLeibniz Research Centre for Working Environment and Human Factors (IfADo)DortmundGermany
| | - Piotr Milkiewicz
- Liver and Internal Medicine Unit, Department of General, Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
- Translational Medicine Group, Pomeranian Medical UniversitySzczecinPoland
| | - Marcin Krawczyk
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver SurgeryMedical University of WarsawWarsawPoland
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical FacultyUniversity of Duisburg‐EssenEssenGermany
| | - Matías A. Ávila
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUNUniversity of NavarraPamplonaSpain
- IdiSNANavarra Institute for Health ResearchPamplonaSpain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute)MadridSpain
| | - Carmen Berasain
- Hepatology Laboratory, Solid Tumors Program, CIMA, CCUNUniversity of NavarraPamplonaSpain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Carlos III Health Institute)MadridSpain
| |
Collapse
|
2
|
Liang J, Deng X, Zhang Y, Fei T, Ouyang M, Yu C, Xiang Y, Jia D, Duan F. Bioinformatics insights into the role of GFPT1 in breast invasive carcinoma: implications for tumor prognosis, immune modulation, and therapeutic applications. Front Genet 2024; 15:1482929. [PMID: 39664728 PMCID: PMC11633769 DOI: 10.3389/fgene.2024.1482929] [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: 08/19/2024] [Accepted: 10/31/2024] [Indexed: 12/13/2024] Open
Abstract
Background Metabolic reprogramming is a hallmark of cancer, including alterations in the hexosamine biosynthesis pathway (HBP). Glutamine-fructose-6-phosphate transaminase 1 (GFPT1) is the key regulatory enzyme in the HBP; however, its role in invasive breast carcinoma remains underexplored. Methods This study utilized integrated data from The Cancer Genome Atlas (TCGA) to assess GFPT1 expression in breast cancer (BRCA) patients. Functional enrichment and mutational landscape analyses were performed, along with chemosensitivity predictions. In vitro experiments were conducted by silencing GFPT1 in malignant breast epithelial cells to evaluate changes in proliferation, migration, and apoptosis. Results Elevated GFPT1 expression was linked to advanced-stage breast cancer and identified as an independent prognostic marker for overall survival (OS). High GFPT1 levels were associated with increased cytoplasmic translation, activation of oncogenic pathways, and infiltration of M2 macrophages. The GFPT1-High group also showed a higher mutational burden, with frequent TP53 mutations. Chemosensitivity analysis revealed increased IC50 values for chemotherapy drugs in this group. GFPT1 silencing led to reduced cell proliferation and migration, along with enhanced apoptosis. Conclusion These findings indicate that GFPT1 is a novel prognostic biomarker and a predictive indicator of chemotherapy response in invasive breast carcinoma. GFPT1 influences mRNA translation, cell cycle regulation, and M2 macrophage infiltration, thereby promoting cancer cell proliferation and metastasis.
Collapse
Affiliation(s)
- Jianghui Liang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Xiaolian Deng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Yingyi Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Tianchi Fei
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Muzi Ouyang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Chengjie Yu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Yang Xiang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Dongwei Jia
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fangfang Duan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
- Department of Pharmacology, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
3
|
Ng AS, Chan DKH. Commonalities and differences in the mutational signature and somatic driver mutation landscape across solid and hollow viscus organs. Oncogene 2023; 42:2713-2724. [PMID: 37573406 PMCID: PMC10491491 DOI: 10.1038/s41388-023-02802-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/14/2023]
Abstract
Advances in sequencing have revealed a highly variegated landscape of mutational signatures and somatic driver mutations in a range of normal tissues. Normal tissues accumulate mutations at varying rates ranging from 11 per cell per year in the liver, to 1879 per cell per year in the bladder. In addition, some normal tissues are also comprised of a large proportion of cells which possess driver mutations while appearing phenotypically normal, as in the oesophagus where a majority of cells harbour driver mutations. Individual tissue proliferation and mutation rate, unique mutagenic stimuli, and local tissue architecture contribute to this highly variegated landscape which confounds the functional characterization of driver mutations found in normal tissue. In particular, our understanding of the relationship between normal tissue somatic mutations and tumour initiation or future cancer risk remains poor. Here, we describe the mutational signatures and somatic driver mutations in solid and hollow viscus organs, highlighting unique characteristics in a tissue-specific manner, while simultaneously seeking to describe commonalities which can bring forward a basic unified theory on the role of these driver mutations in tumour initiation. We discuss novel findings which can be used to inform future research in this field.
Collapse
Affiliation(s)
- Aik Seng Ng
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Dedrick Kok Hong Chan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Division of Colorectal Surgery, University Surgical Cluster, National University Hospital, Singapore, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
4
|
Shared genetic and epigenetic changes link aging and cancer. Trends Cell Biol 2022; 32:338-350. [PMID: 35144882 DOI: 10.1016/j.tcb.2022.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/28/2021] [Accepted: 01/07/2022] [Indexed: 12/12/2022]
Abstract
Aging is a universal biological process that increases the risk of multiple diseases including cancer. Growing evidence shows that alterations in the genome and epigenome, driven by similar mechanisms, are found in both aged cells and cancer cells. In this review, we detail the genetic and epigenetic changes associated with normal aging and the mechanisms responsible for these changes. By highlighting genetic and epigenetic alterations in the context of tumorigenesis, cancer progression, and the aging tumor microenvironment, we examine the possible impacts of the normal aging process on malignant transformation. Finally, we examine the implications of age-related genetic and epigenetic alterations in both tumors and patients for the treatment of cancer.
Collapse
|
5
|
Borden ES, Buetow KH, Wilson MA, Hastings KT. Cancer Neoantigens: Challenges and Future Directions for Prediction, Prioritization, and Validation. Front Oncol 2022; 12:836821. [PMID: 35311072 PMCID: PMC8929516 DOI: 10.3389/fonc.2022.836821] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/07/2022] [Indexed: 12/16/2022] Open
Abstract
Prioritization of immunogenic neoantigens is key to enhancing cancer immunotherapy through the development of personalized vaccines, adoptive T cell therapy, and the prediction of response to immune checkpoint inhibition. Neoantigens are tumor-specific proteins that allow the immune system to recognize and destroy a tumor. Cancer immunotherapies, such as personalized cancer vaccines, adoptive T cell therapy, and immune checkpoint inhibition, rely on an understanding of the patient-specific neoantigen profile in order to guide personalized therapeutic strategies. Genomic approaches to predicting and prioritizing immunogenic neoantigens are rapidly expanding, raising new opportunities to advance these tools and enhance their clinical relevance. Predicting neoantigens requires acquisition of high-quality samples and sequencing data, followed by variant calling and variant annotation. Subsequently, prioritizing which of these neoantigens may elicit a tumor-specific immune response requires application and integration of tools to predict the expression, processing, binding, and recognition potentials of the neoantigen. Finally, improvement of the computational tools is held in constant tension with the availability of datasets with validated immunogenic neoantigens. The goal of this review article is to summarize the current knowledge and limitations in neoantigen prediction, prioritization, and validation and propose future directions that will improve personalized cancer treatment.
Collapse
Affiliation(s)
- Elizabeth S Borden
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, United States.,Department of Research and Internal Medicine (Dermatology), Phoenix Veterans Affairs Health Care System, Phoenix, AZ, United States
| | - Kenneth H Buetow
- School of Life Sciences, Arizona State University, Tempe, AZ, United States.,Center for Evolution and Medicine, Arizona State University, Tempe, AZ, United States
| | - Melissa A Wilson
- School of Life Sciences, Arizona State University, Tempe, AZ, United States.,Center for Evolution and Medicine, Arizona State University, Tempe, AZ, United States
| | - Karen Taraszka Hastings
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, United States.,Department of Research and Internal Medicine (Dermatology), Phoenix Veterans Affairs Health Care System, Phoenix, AZ, United States
| |
Collapse
|
6
|
Birkou M, Raptis V, Marousis KD, Tsevis A, Bourikas K, Bentrop D, Episkopou V, Spyroulias GA. Impact of a Single Nucleotide Polymorphism on the 3D Protein Structure and Ubiquitination Activity of E3 Ubiquitin Ligase Arkadia. Front Mol Biosci 2022; 9:844129. [PMID: 35281275 PMCID: PMC8905620 DOI: 10.3389/fmolb.2022.844129] [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: 12/27/2021] [Accepted: 01/31/2022] [Indexed: 11/16/2022] Open
Abstract
Single nucleotide polymorphisms (SNPs) are genetic variations which can play a vital role in the study of human health. SNP studies are often used to identify point mutations that are associated with diseases. Arkadia (RNF111) is an E3 ubiquitin ligase that enhances transforming growth factor-beta (TGF-β) signaling by targeting negative regulators for degradation. Dysregulation of the TGF-β pathway is implicated in cancer because it exhibits tumor suppressive activity in normal cells while in tumor cells it promotes invasiveness and metastasis. Τhe SNP CGT > TGT generated an amino-acid (aa) substitution of Arginine 957 to Cysteine on the enzymatic RING domain of Arkadia. This was more prevalent in a tumor than in a normal tissue sample of a patient with colorectal cancer. This prompted us to investigate the effect of this mutation in the structure and activity of Arkadia RING. We used nuclear magnetic resonance (NMR) to analyze at an atomic-level the structural and dynamic properties of the R957C Arkadia RING domain, while ubiquitination and luciferase assays provided information about its enzymatic functionality. Our study showed that the R957C mutation changed the electrostatic properties of the RING domain however, without significant effects on the structure of its core region. However, the functional studies revealed that the R957C Arkadia exhibits significantly increased enzymatic activity supporting literature data that Arkadia within tumor cells promotes aggressive and metastatic behavior.
Collapse
Affiliation(s)
- Maria Birkou
- Department of Pharmacy, University of Patras, Patras, Greece
| | - Vasilios Raptis
- Department of Pharmacy, University of Patras, Patras, Greece
| | | | - Athanasios Tsevis
- School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Kyriakos Bourikas
- School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Detlef Bentrop
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Vasso Episkopou
- Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes, London, United Kingdom
| | | |
Collapse
|
7
|
Cho EJ, Kim M, Jo D, Kim J, Oh JH, Chung HC, Lee SH, Kim D, Chun SM, Kim J, Lee H, Kim TW, Yu CS, Sung CO, Jang SJ. Immuno-genomic classification of colorectal cancer organoids reveals cancer cells with intrinsic immunogenic properties associated with patient survival. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:230. [PMID: 34256801 PMCID: PMC8276416 DOI: 10.1186/s13046-021-02034-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/30/2021] [Indexed: 12/28/2022]
Abstract
Background The intrinsic immuno-ge7nomic characteristics of colorectal cancer cells that affect tumor biology and shape the tumor immune microenvironment (TIM) are unclear. Methods We developed a patient-derived colorectal cancer organoid (CCO) model and performed pairwise analysis of 87 CCOs and their matched primary tumors. The TIM type of the primary tumor was classified as immuno-active, immuno-exhausted, or immuno-desert. Results The gene expression profiles, signaling pathways, major oncogenic mutations, and histology of the CCOs recapitulated those of the primary tumors, but not the TIM of primary tumors. Two distinct intrinsic molecular subgroups of highly proliferative and mesenchymal phenotypes with clinical significance were identified in CCOs with various cancer signaling pathways. CCOs showed variable expression of cancer-specific immune-related genes such as those encoding HLA-I and HLA-II, and molecules involved in immune checkpoint activation/inhibition. Among these genes, the expression of HLA-II in CCOs was associated with favorable patient survival. K-means clustering analysis based on HLA-II expression in CCOs revealed a subgroup of patients, in whom cancer cells exhibited Intrinsically Immunogenic Properties (Ca-IIP), and were characterized by high expression of signatures associated with HLA-I, HLA-II, antigen presentation, and immune stimulation. Patients with the Ca-IIP phenotype had an excellent prognosis, irrespective of age, disease stage, intrinsic molecular type, or TIM status. Ca-IIP was negatively correlated with intrinsic E2F/MYC signaling. Analysis of the correlation between CCO immuno-genotype and TIM phenotype revealed that the TIM phenotype was associated with microsatellite instability, Wnt/β-catenin signaling, APC/KRAS mutations, and the unfolded protein response pathway linked to the FBXW7 mutation in cancer cells. However, Ca-IIP was not associated with the TIM phenotype. Conclusions We identified a Ca-IIP phenotype from a large set of CCOs. Our findings may provide an unprecedented opportunity to develop new strategies for optimal patient stratification in this era of immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02034-1.
Collapse
Affiliation(s)
- Eun Jeong Cho
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.,Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Minsuh Kim
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Daum Jo
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Jihye Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Ji-Hye Oh
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.,Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee Chul Chung
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Sun-Hye Lee
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Deokhoon Kim
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Sung-Min Chun
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jihun Kim
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hyeonjin Lee
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Tae Won Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chang Sik Yu
- Department of Colon and Rectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chang Ohk Sung
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea. .,Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea. .,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Se Jin Jang
- Asan Center for Cancer Genome Discovery, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea. .,Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea. .,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea. .,OncoClew Life Science Co., Ltd, Songpa-gu, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Lee H, Kim EN, Lee JY, Kim JH, Oh JH, Kim WK, Cho EJ, Lim J, Chun SM, Sung CO. Characterization of early postzygotic somatic mutations through multi-organ analysis. J Hum Genet 2021; 66:777-784. [PMID: 33611337 DOI: 10.1038/s10038-021-00908-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/14/2021] [Accepted: 01/29/2021] [Indexed: 11/09/2022]
Abstract
Mosaicisms caused by postzygotic mutational events are of increasing interest because of their potential association with various human diseases. Postzygotic somatic mutations have not been well characterized however in terms of their developmental lineage in humans. We conducted whole-genome sequencing (WGS) and targeted deep sequencing in 15 organs across three developmental lineages from a single male fetus with polycystic kidney disease (PKD) of 21 weeks gestational age. This fetus had no detectable neurological abnormalities at autopsy but germline mutations in the PKHD1 gene were identified that may have been associated with the PKD. Eight early embryonic mosaic variants with no alteration of protein function were detected. These variants were thought to have occurred at the two or four cell stages after fertilization with a mutational pattern involving frequent C>T and T>C transitions. In our current analyses, no tendency toward organ-specific mutation occurrences was found as the eight variants were detected in all 15 organs. However different allele fractions of these variants were found in different organs, suggesting a tissue-specific asymmetric growth of cells that reflected the developmental germ layer of each organ. This indicated that somatic mutation occurrences, even in early embryogenesis, can affect specific organ development or disease. Our current analyses demonstrate that multi-organ analysis is helpful for understanding genomic mosaicism. Our results also provide insights into the biological role of mosaicism in embryonic development and disease.
Collapse
Affiliation(s)
- Hyeonjin Lee
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Na Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji-Young Lee
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Hun Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ji-Hye Oh
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Won-Kyung Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Jeong Cho
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jinyeong Lim
- Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Min Chun
- Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea. .,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Chang Ohk Sung
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea. .,Center for Cancer Genome Discovery, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea. .,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|