1
|
Ooki A, Osumi H, Yoshino K, Yamaguchi K. Potent therapeutic strategy in gastric cancer with microsatellite instability-high and/or deficient mismatch repair. Gastric Cancer 2024; 27:907-931. [PMID: 38922524 PMCID: PMC11335850 DOI: 10.1007/s10120-024-01523-4] [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: 04/17/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024]
Abstract
Gastric cancer (GC) is a common malignancy that presents challenges in patient care worldwide. The mismatch repair (MMR) system is a highly conserved DNA repair mechanism that protects genome integrity during replication. Deficient MMR (dMMR) results in an increased accumulation of genetic errors in microsatellite sequences, leading to the development of a microsatellite instability-high (MSI-H) phenotype. Most MSI-H/dMMR GCs arise sporadically, mainly due to MutL homolog 1 (MLH1) epigenetic silencing. Unlike microsatellite-stable (MSS)/proficient MMR (pMMR) GCs, MSI-H/dMMR GCs are relatively rare and represent a distinct subtype with genomic instability, a high somatic mutational burden, favorable immunogenicity, different responses to treatment, and prognosis. dMMR/MSI-H status is a robust predictive biomarker for treatment with immune checkpoint inhibitors (ICIs) due to high neoantigen load, prominent tumor-infiltrating lymphocytes, and programmed cell death ligand 1 (PD-L1) overexpression. However, a subset of MSI-H/dMMR GC patients does not benefit from immunotherapy, highlighting the need for further research into predictive biomarkers and resistance mechanisms. This review provides a comprehensive overview of the clinical, molecular, immunogenic, and therapeutic aspects of MSI-H/dMMR GC, with a focus on the impact of ICIs in immunotherapy and their potential as neoadjuvant therapies. Understanding the complexity and diversity of the molecular and immunological profiles of MSI-H/dMMR GC will drive the development of more effective therapeutic strategies and molecular targets for future precision medicine.
Collapse
Affiliation(s)
- Akira Ooki
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan.
| | - Hiroki Osumi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Koichiro Yoshino
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| |
Collapse
|
2
|
Hwang I, Cho Y, Kang SY, Kim DG, Ahn S, Lee J, Kim KM. Comparative analysis of ARID1A mutations with mRNA levels and protein expression in gastric carcinoma. Pathol Res Pract 2024; 255:155063. [PMID: 38324965 DOI: 10.1016/j.prp.2023.155063] [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: 11/27/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 02/09/2024]
Abstract
The ARID1A gene is pivotal in chromatin remodeling and genomic integrity and is frequently mutated in various cancer types. ARID1A mutation is the second most frequently mutated tumor suppressor gene and has been suggested as a predictor of immunotherapeutic responsiveness in gastric carcinoma (GC). Despite its significance, the relationship among ARID1A somatic mutations, RNA expression levels, and protein expression remains unclear, particularly in GC. For this purpose, we performed comparative study in two cohorts. Cohort 1 used next-generation sequencing (NGS) to identify 112 GC cases with ARID1A mutations. These cases were compared with ARID1A immunohistochemistry (IHC) results. Cohort 2 employed microarray gene expression data to assess ARID1A RNA levels and compare them with ARID1A IHC results. In Cohort 1, 38.4% of ARID1A-mutated GC exhibited a complete loss of ARID1A protein when assessed by IHC, whereas the remaining 61.6% displayed intact ARID1A. Discordance between NGS and IHC results was not associated with specific mutation sites, variant classifications, or variant allele frequencies. In Cohort 2, 24.1% of the patients demonstrated a loss of ARID1A protein, and there was no significant difference in mRNA levels between the ARID1A protein-intact and -loss groups. Our study revealed a substantial discrepancy between ARID1A mutations detected using NGS and protein expression assessed using IHC in GC. Moreover, ARID1A mRNA expression levels did not correlate well with protein expression. These findings highlighted the complexity of ARID1A expression in GC.
Collapse
Affiliation(s)
- Inwoo Hwang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yunjoo Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - So Young Kang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Deok Geun Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Soomin Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeeyun Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
3
|
Zhang Z, Li Q, Sun S, Ye J, Li Z, Cui Z, Liu Q, Zhang Y, Xiong S, Zhang S. Prognostic and immune infiltration significance of ARID1A in TCGA molecular subtypes of gastric adenocarcinoma. Cancer Med 2023; 12:16716-16733. [PMID: 37366273 PMCID: PMC10501255 DOI: 10.1002/cam4.6294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/19/2023] [Accepted: 06/18/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND AT-rich interaction domain 1A (ARID1A) is an essential subunit of the switch/sucrose non-fermentable chromatin remodeling complex and is considered to be a tumor suppressor. The Cancer Genome Atlas (TCGA) molecular classification has deepened our understanding of gastric cancer at the molecular level. This study explored the significance of ARID1A expression in TCGA subtypes of gastric adenocarcinoma. METHODS We collected 1248 postoperative patients with gastric adenocarcinoma, constructed tissue microarrays, performed immunohistochemistry for ARID1A, and obtained correlations between ARID1A and clinicopathological variables. We then carried out the prognostic analysis of ARID1A in TCGA subtypes. Finally, we screened patients by random sampling and propensity score matching method and performed multiplex immunofluorescence to explore the effects of ARID1A on CD4, CD8, and PD-L1 expression in TCGA subtypes. RESULTS Seven variables independently associated with ARID1A were screened out: mismatch repair proteins, PD-L1, T stage, differentiation status, p53, E-cadherin, and EBER. The independent prognostic variables in the genomically stable (GS) subtype were N stage, M stage, T stage, chemotherapy, size, and ARID1A. PD-L1 expression was higher in the ARID1A negative group than in the ARID1A positive group in all TCGA subgroups. CD4 showed higher expression in the ARID1A negative group in most subtypes, while CD8 did not show the difference in most subtypes. When ARID1A was negative, PD-L1 expression was positively correlated with CD4/CD8 expression; while when ARID1A was positive, this correlation disappeared. CONCLUSIONS The negative expression of ARID1A occurred more frequently in the Epstein-Barr virus and microsatellite instability subtypes and was an independent adverse prognostic factor in the GS subtype. In the TCGA subtypes, ARID1A negative expression caused increased CD4 and PD-L1 expression, whereas CD8 expression appeared independent of ARID1A. The expression of CD4/CD8 induced by ARID1A negativity was accompanied by an increase in PD-L1 expression.
Collapse
Affiliation(s)
- Zhenkun Zhang
- Weihai Municipal HospitalShandong UniversityWeihaiChina
- Department of OncologyShouguang People's HospitalWeifangChina
| | - Qiujing Li
- Department of Pathology, Weihai Municipal HospitalShandong UniversityWeihaiChina
| | - Shanshan Sun
- Department of Oncology, Weihai Municipal HospitalShandong UniversityWeihaiChina
| | - Jing Ye
- Binzhou Medical UniversityYantaiChina
| | - Zhe Li
- Weifang Medical CollegeWeifangChina
| | - Zhengguo Cui
- Department of Environmental HealthUniversity of Fukui School of Medical SciencesFukuiJapan
| | - Qian Liu
- Department of Pathology, Weihai Municipal HospitalShandong UniversityWeihaiChina
| | - Yujie Zhang
- Department of Pathology, Weihai Municipal HospitalShandong UniversityWeihaiChina
| | | | - Shukun Zhang
- Department of Pathology, Weihai Municipal HospitalShandong UniversityWeihaiChina
| |
Collapse
|
4
|
Yang D, Yu J, Han B, Sun Y, Mo S, Hu J. Long Non-coding RNA Expression Patterns in Stomach Adenocarcinoma Serve as an Indicator of Tumor Mutation Burden and Are Associated With Tumor-Infiltrating Lymphocytes and Microsatellite Instability. Front Cell Dev Biol 2021; 9:618313. [PMID: 33644056 PMCID: PMC7907456 DOI: 10.3389/fcell.2021.618313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/21/2021] [Indexed: 01/10/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are crucial in controlling important aspects of tumor immunity. However, whether the expression pattern of lncRNAs in stomach adenocarcinoma (STAD) reflects tumor immunity is not fully understood. We screened differentially expressed lncRNAs (DElncRNAs) between high and low tumor mutation burden (TMB) STAD samples. Using the least absolute shrinkage and selection operator method, 33 DElncRNAs were chosen to establish a lncRNA-based signature classifier for predicting TMB levels. The accuracy of the 33-lncRNA-based signature classifier was 0.970 in the training set and 0.950 in the test set, suggesting the expression patterns of the 33 lncRNAs may be an indicator of TMB in STAD. Survival analysis showed that a lower classifier index reflected better prognosis for STAD patients, and the index showed correlation with expression of immune checkpoint molecules (PD1, PDL1, and CTLA4), tumor-infiltrating lymphocytes, and microsatellite instability. In conclusion, STAD samples with different tumor mutation burdens have different lncRNA expression patterns. The 33-lncRNA-based signature classifier index may be an indicator of TMB and is associated expression of immune checkpoints, tumor-infiltrating lymphocytes, and microsatellite instability.
Collapse
Affiliation(s)
- Dongdong Yang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinling Yu
- Department of General Practice, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bing Han
- Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yue Sun
- Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Steven Mo
- YDILife Academy of Sciences, Nanning, China.,Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China
| | - Jing Hu
- Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
5
|
Migliori AD, Patel LA, Neale C. The RIT1 C-terminus associates with lipid bilayers via charge complementarity. Comput Biol Chem 2021; 91:107437. [PMID: 33517146 DOI: 10.1016/j.compbiolchem.2021.107437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/30/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022]
Abstract
RIT1 is a member of the Ras superfamily of small GTPases involved in regulation of cellular signaling. Mutations to RIT1 are involved in cancer and developmental disorders. Like many Ras subfamily members, RIT1 is localized to the plasma membrane. However, RIT1 lacks the C-terminal prenylation that helps many other subfamily members adhere to cellular membranes. We used molecular dynamics simulations to examine the mechanisms by which the C-terminal peptide (CTP) of RIT1 associates with lipid bilayers. We show that the CTP is unstructured and that its membrane interactions depend on lipid composition. While a 12-residue region of the CTP binds strongly to anionic bilayers containing phosphatidylserine lipids, the CTP termini fray from the membrane allowing for accommodation of the RIT1 globular domain at the membrane-water interface.
Collapse
Affiliation(s)
- Amy D Migliori
- Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, NM, 87545, United States; Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, 87545, United States
| | - Lara A Patel
- Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, NM, 87545, United States; Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, 87545, United States
| | - Chris Neale
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, 87545, United States.
| |
Collapse
|
6
|
McPherson MT, Holub AS, Husbands AY, Petreaca RC. Mutation Spectra of the MRN (MRE11, RAD50, NBS1/NBN) Break Sensor in Cancer Cells. Cancers (Basel) 2020; 12:cancers12123794. [PMID: 33339169 PMCID: PMC7765586 DOI: 10.3390/cancers12123794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/04/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary A DNA double strand break cuts a chromosome in two and is one of the most dangerous forms of DNA damage. Improper repair can lead to various chromosomal re-arrangements that have been detected in almost all cancer cells. A complex of three proteins (MRE11, RAD50, NBS1 or NBN) detects chromosome breaks and orchestrates repair processes. Mutations in these “break sensor” genes have been described in a multitude of cancers. Here, we provide a comprehensive analysis of reported mutations from data deposited on the Catalogue of Somatic Mutations in Cancer (COSMIC) archive. We also undertake an evolutionary analysis of these genes with the aim to understand whether these mutations preferentially accumulate in conserved residues. Interestingly, we find that mutations are overrepresented in evolutionarily conserved residues of RAD50 and NBS1/NBN but not MRE11. Abstract The MRN complex (MRE11, RAD50, NBS1/NBN) is a DNA double strand break sensor in eukaryotes. The complex directly participates in, or coordinates, several activities at the break such as DNA resection, activation of the DNA damage checkpoint, chromatin remodeling and recruitment of the repair machinery. Mutations in components of the MRN complex have been described in cancer cells for several decades. Using the Catalogue of Somatic Mutations in Cancer (COSMIC) database, we characterized all the reported MRN mutations. This analysis revealed several hotspot frameshift mutations in all three genes that introduce premature stop codons and truncate large regions of the C-termini. We also found through evolutionary analyses that COSMIC mutations are enriched in conserved residues of NBS1/NBN and RAD50 but not in MRE11. Given that all three genes are important to carcinogenesis, we propose these differential enrichment patterns may reflect a more severe pleiotropic role for MRE11.
Collapse
|
7
|
Wang R, Chen M, Ye X, Poon K. Role and potential clinical utility of ARID1A in gastrointestinal malignancy. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 787:108360. [PMID: 34083049 DOI: 10.1016/j.mrrev.2020.108360] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/26/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022]
Abstract
ARID1A (AT-rich interactive domain 1A) is a newly discovered tumor suppressor gene, and its encoded product is an important component of the SWI/SNF chromatin remodeling complex. ARID1A plays an important role in cell proliferation, invasion and metastasis, apoptosis, cell cycle regulation, epithelial mesenchymal transition, and the regulation of other of biological behaviors. Recently, ARID1A mutations have been increasingly reported in esophageal adenocarcinoma, gastric cancer, colorectal cancer, hepatocellular carcinoma, cholangiocarcinoma, pancreatic cancer, and other malignant tumors of the digestive system. This article reviews the relationship between ARID1A mutation and the molecular mechanisms of carcinogenesis, including microsatellite instability and the PI3K/ATK signaling pathway, and relates these mechanisms to the prognostic assessment of digestive malignancy. Further, this review describes the potential for molecular pathologic epidemiology (MPE) to provide new insights into environment-tumor-host interactions.
Collapse
Affiliation(s)
- Ruihua Wang
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, Guangdong Province, China.
| | - Mei Chen
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, Guangdong Province, China.
| | - Xiaojun Ye
- Program of Food Science and Technology, Division of Science and Technology, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai, 519085, Guangdong Province, China.
| | - Karen Poon
- Program of Food Science and Technology, Division of Science and Technology, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai, 519085, Guangdong Province, China.
| |
Collapse
|
8
|
Nelakurti DD, Pappula AL, Rajasekaran S, Miles WO, Petreaca RC. Comprehensive Analysis of MEN1 Mutations and Their Role in Cancer. Cancers (Basel) 2020; 12:cancers12092616. [PMID: 32937789 PMCID: PMC7565326 DOI: 10.3390/cancers12092616] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/04/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Cancers are characterized by accumulation of genetic mutations in key cell cycle regulators that alter or disable the function of these genes. Such mutations can be inherited or arise spontaneously during the life of the individual. The MEN1 gene prevents uncontrolled cell division and it is considered a tumor suppressor. Inherited MEN1 mutations are associated with certain parathyroid and pancreatic syndromes while spontaneous mutations have been detected in cancer cells. We investigated whether inherited mutations appear in cancer cells which would suggest that patients with parathyroid and pancreatic syndromes have a predisposition to develop cancer. We find a weak correlation between the spectrum of inherited mutations and those appearing spontaneously. Thus, inherited MEN1 mutations may not be a good predictor of tumorigenesis. Abstract MENIN is a scaffold protein encoded by the MEN1 gene that functions in multiple biological processes, including cell proliferation, migration, gene expression, and DNA damage repair. MEN1 is a tumor suppressor gene, and mutations that disrupts MEN1 function are common to many tumor types. Mutations within MEN1 may also be inherited (germline). Many of these inherited mutations are associated with a number of pathogenic syndromes of the parathyroid and pancreas, and some also predispose patients to hyperplasia. In this study, we cataloged the reported germline mutations from the ClinVar database and compared them with the somatic mutations detected in cancers from the Catalogue of Somatic Mutations in Cancer (COSMIC) database. We then used statistical software to determine the probability of mutations being pathogenic or driver. Our data show that many confirmed germline mutations do not appear in tumor samples. Thus, most mutations that disable MEN1 function in tumors are somatic in nature. Furthermore, of the germline mutations that do appear in tumors, only a fraction has the potential to be pathogenic or driver mutations.
Collapse
Affiliation(s)
- Devi D. Nelakurti
- Biomedical Science Undergraduate Program, The Ohio State University Medical School, Columbus, OH 43210, USA;
| | - Amrit L. Pappula
- Computer Science and Engineering Undergraduate Program, The Ohio State University, Columbus, OH 43210, USA;
| | - Swetha Rajasekaran
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA;
| | - Wayne O. Miles
- Department of Cancer Biology and Genetics, The Ohio State University Medical School, Columbus, OH 43210, USA;
| | - Ruben C. Petreaca
- Department of Molecular Genetics, The Ohio State University, Marion, OH 43302, USA
- Correspondence:
| |
Collapse
|
9
|
Fernandes Q, Gupta I, Vranic S, Al Moustafa AE. Human Papillomaviruses and Epstein-Barr Virus Interactions in Colorectal Cancer: A Brief Review. Pathogens 2020; 9:pathogens9040300. [PMID: 32325943 PMCID: PMC7238043 DOI: 10.3390/pathogens9040300] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023] Open
Abstract
Human papillomaviruses (HPVs) and the Epstein-Barr virus (EBV) are the most common oncoviruses, contributing to approximately 10%-15% of all malignancies. Oncoproteins of high-risk HPVs (E5 and E6/E7), as well as EBV (LMP1, LMP2A and EBNA1), play a principal role in the onset and progression of several human carcinomas, including head and neck, cervical and colorectal. Oncoproteins of high-risk HPVs and EBV can cooperate to initiate and/or enhance epithelial-mesenchymal transition (EMT) events, which represents one of the hallmarks of cancer progression and metastasis. Although the role of these oncoviruses in several cancers is well established, their role in the pathogenesis of colorectal cancer is still nascent. This review presents an overview of the most recent advances related to the presence and role of high-risk HPVs and EBV in colorectal cancer, with an emphasis on their cooperation in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Queenie Fernandes
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (Q.F.); (I.G.)
- Biomedical Research Centre, Qatar University, Doha 2713, Qatar
| | - Ishita Gupta
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (Q.F.); (I.G.)
- Biomedical Research Centre, Qatar University, Doha 2713, Qatar
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (Q.F.); (I.G.)
- Correspondence: (S.V.); (A.-E.A.M.); Tel.:+974-4403-7873 (S.V.); +974-4403-7817 (A.-E.A.M.)
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; (Q.F.); (I.G.)
- Biomedical Research Centre, Qatar University, Doha 2713, Qatar
- Correspondence: (S.V.); (A.-E.A.M.); Tel.:+974-4403-7873 (S.V.); +974-4403-7817 (A.-E.A.M.)
| |
Collapse
|
10
|
Insights on the regulation of the MLL/SET1 family histone methyltransferases. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1863:194561. [PMID: 32304759 DOI: 10.1016/j.bbagrm.2020.194561] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/07/2020] [Accepted: 04/09/2020] [Indexed: 01/09/2023]
Abstract
In eukaryotes, histone H3K4 methylation by the MLL/SET1 family histone methyltransferases is enriched at transcription regulatory elements including gene promoters and enhancers. The level of H3K4 methylation is highly correlated with transcription activation and is one of the most frequently used histone post-translational modifications to predict transcriptional outcome. Recently, it has been shown that rearrangement of the cellular landscape of H3K4 mono-methylation at distal enhancers precedes cell fate transition and is used for identification of novel regulatory elements for development and disease progression. Similarly, broad H3K4 tri-methylation regions have also been used to predict intrinsic tumor suppression properties of regulator regions in a variety of cellular models. Understanding the regulation for how H3K4 methylation is deposited and regulated is of paramount importance. In this review, we will discuss new findings on how the MLL/SET1 family enzymes are regulated on chromatin and their potential functional and regulatory implications. This article is part of a Special Issue entitled: The MLL family of proteins in normal development and disease edited by Thomas A Milne.
Collapse
|
11
|
Park SH, Ayoub A, Lee YT, Xu J, Kim H, Zheng W, Zhang B, Sha L, An S, Zhang Y, Cianfrocco MA, Su M, Dou Y, Cho US. Cryo-EM structure of the human MLL1 core complex bound to the nucleosome. Nat Commun 2019; 10:5540. [PMID: 31804488 PMCID: PMC6895043 DOI: 10.1038/s41467-019-13550-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/14/2019] [Indexed: 12/13/2022] Open
Abstract
Mixed lineage leukemia (MLL) family histone methyltransferases are enzymes that deposit histone H3 Lys4 (K4) mono-/di-/tri-methylation and regulate gene expression in mammals. Despite extensive structural and biochemical studies, the molecular mechanisms whereby the MLL complexes recognize histone H3K4 within nucleosome core particles (NCPs) remain unclear. Here we report the single-particle cryo-electron microscopy (cryo-EM) structure of the NCP-bound human MLL1 core complex. We show that the MLL1 core complex anchors to the NCP via the conserved RbBP5 and ASH2L, which interact extensively with nucleosomal DNA and the surface close to the N-terminal tail of histone H4. Concurrent interactions of RbBP5 and ASH2L with the NCP uniquely align the catalytic MLL1SET domain at the nucleosome dyad, thereby facilitating symmetrical access to both H3K4 substrates within the NCP. Our study sheds light on how the MLL1 complex engages chromatin and how chromatin binding promotes MLL1 tri-methylation activity. MLL family histone methyltransferases deposit histone H3 Lys4 mono-/di-/tri-methylation and regulate gene expression in mammals. Here the authors report the single-particle cryo-EM structure of the NCP-bound human MLL1 core complex, shedding light on how the MLL1 complex engages chromatin and how chromatin binding promotes MLL1 tri-methylation activity.
Collapse
Affiliation(s)
- Sang Ho Park
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Alex Ayoub
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Young-Tae Lee
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Jing Xu
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Hanseong Kim
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Wei Zheng
- Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Biao Zhang
- Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Liang Sha
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Sojin An
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Yang Zhang
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, USA.,Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Michael A Cianfrocco
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Min Su
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Yali Dou
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, USA. .,Department of Pathology, University of Michigan, Ann Arbor, Michigan, 48109, USA.
| | - Uhn-Soo Cho
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, USA.
| |
Collapse
|
12
|
Takaoka S, Hirotsu Y, Ohyama H, Mochizuki H, Amemiya K, Oyama T, Ashizawa H, Yoshimura D, Nakagomi K, Hosoda K, Suzuki Y, Kojima Y, Omata M. Molecular subtype switching in early-stage gastric cancers with multiple occurrences. J Gastroenterol 2019; 54:674-686. [PMID: 30666417 DOI: 10.1007/s00535-019-01547-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/10/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Multiple gastric cancers at the same time (synchronous) or recurrence after 1 year (metachronous) are frequently encountered. Since their genetic profiles were not well elucidated, we molecularly subtyped the genetic events of synchronous and metachronous early-stage gastric cancers. METHODS We studied mismatch repair (MMR) genes in 84 tumors from 31 patients (15 synchronous and 16 metachronous) by immunohistochemistry. We performed microsatellite instability analysis and targeted sequencing of 58 significantly mutated genes (SMGs) in 35 tumors from thirteen patients. Genomic data from TCGA were used for comparisons with advanced-stage cancers. RESULTS Among the 31 patients, at least one deficient-MMR (dMMR) tumor was observed in eight (26%). Of eight patients, seven showed a mixture of proficient-MMR (pMMR) and dMMR tumors. The one case with only dMMR had six recurrent tumors within 2 years. To further subtype, we sequenced 58 SMGs in 35 samples (25 pMMR and 10 dMMR) from thirteen patients. In 35 samples, 163 mutations were identified, but none matched in almost cases, strongly indicating different clonal origins, whether synchronous or metachronous occurrences. Of the 25 pMMR cases, 1 belonged to Epstein-Barr virus (EBV), 24 belonged to chromosomal instability (CIN) subtypes. Of the thirteen cases, repetitive CIN, a mixture of CIN and MSI, a mixture of CIN and EBV, and repetitive MSI were observed in nine (70%), two (15%), one (8%) and one (8%), respectively. CONCLUSIONS Despite multiple tumors occurring in the same patient simultaneously or several years apart, clonal origin was totally different. 'Switching' or 'mixing' of dMMR and pMMR, EBV or CIN occurred, which had clinical relevance with regard to immunotherapy.
Collapse
Affiliation(s)
- Shinya Takaoka
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Yosuke Hirotsu
- Genome Analysis Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan.
| | - Hiroshi Ohyama
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan.,Department of Gastroenterology, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba, Japan
| | - Hitoshi Mochizuki
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan.,Genome Analysis Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Kenji Amemiya
- Genome Analysis Center, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Toshio Oyama
- Department of Pathology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Hiroshi Ashizawa
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Dai Yoshimura
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Keiko Nakagomi
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Kenji Hosoda
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Yoji Suzuki
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Yuichiro Kojima
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan
| | - Masao Omata
- Department of Gastroenterology, Yamanashi Central Hospital, 1-1-1 Fujimi, Kofu, Yamanashi, Japan.,The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
13
|
Aboushousha T, Helal N, Hammam O, Ibrahim M, Khaled S, Mostafa A, Anas A. Overview of MDM2 and B-RAF Expression in Gastric Lesions. Open Access Maced J Med Sci 2018; 6:1795-1802. [PMID: 30455751 PMCID: PMC6236038 DOI: 10.3889/oamjms.2018.338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Globally, gastric cancer (GC) it is the fourth most common cancer and the third cause of cancer-related deaths. Overexpression of MDM2 and B-RAF appeared to be increased in malignancy and associated with poor prognosis in several human tumours, but their role in gastric cancer remains controversial. AIM We had investigated the immunohistochemical expression of MDM2 and B-RAF in 136 gastric lesions with/without H. pylori association. MATERIAL AND METHODS Studied specimens include chronic gastritis (32), intestinal type GC (70), diffuse GC (22) and gastrointestinal stromal tumours (GIST) (12). RESULTS MDM2 expression increased significantly in intestinal GC compared to other groups (p < 0.001), while B-RAF expression increased significantly in GIST compared to other groups (p < 0.001). H. pylori increased expression of MDM2 in intestinal GC cases but did not affect B-RAF expression. MDM2 expression correlated with high grade of tumor differentiation (p < 0.001), deep invasion (p < 0.05), nodal metastases (p < 0.05) and distant metastases (p < 0.1) in intestinal GC, while B-RAF expression did not correlate with TNM stage (p < 0.1). CONCLUSION MDM2 up-regulation was more frequent in intestinal GC, while B-RAF up-regulation was more frequent in GIST compared to other groups; MDM2 expression in intestinal GC was correlated with H. pylori association, high grade of differentiation, deep invasion, nodal and distant metastases, meanwhile, B-RAF expression was correlated with high-grade intestinal GC but did not correlate with H. pylori or TNM stage. The possible role of both MDM2 and B-RAF in predicting progression of gastric tumours and prognosis deserves further investigations.
Collapse
Affiliation(s)
- Tarek Aboushousha
- Department of Pathology, Theodor Bilharz Research Institute, Imbaba, Giza, Egypt
| | - Noha Helal
- Department of Pathology, Theodor Bilharz Research Institute, Imbaba, Giza, Egypt
| | - Olfat Hammam
- Department of Pathology, Theodor Bilharz Research Institute, Imbaba, Giza, Egypt
| | - Manar Ibrahim
- Faculty of Biotechnology, October University of Modern Sciences and Arts, Giza, Egypt
| | - Samar Khaled
- Faculty of Biotechnology, October University of Modern Sciences and Arts, Giza, Egypt
| | - Amr Mostafa
- Department of Surgery, Theodor Bilharz Research Institute, Imbaba, Giza, Egypt
| | - Amgad Anas
- Department of Hepato-Gastroenterology, Theodor Bilharz Research Institute, Giza, Egypt
| |
Collapse
|
14
|
Zhu S, Jin J, Gokhale S, Lu AM, Shan H, Feng J, Xie P. Genetic Alterations of TRAF Proteins in Human Cancers. Front Immunol 2018; 9:2111. [PMID: 30294322 PMCID: PMC6158389 DOI: 10.3389/fimmu.2018.02111] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 08/28/2018] [Indexed: 12/25/2022] Open
Abstract
The tumor necrosis factor receptor (TNF-R)-associated factor (TRAF) family of cytoplasmic adaptor proteins regulate the signal transduction pathways of a variety of receptors, including the TNF-R superfamily, Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I-like receptors (RLRs), and cytokine receptors. TRAF-dependent signaling pathways participate in a diverse array of important cellular processes, including the survival, proliferation, differentiation, and activation of different cell types. Many of these TRAF-dependent signaling pathways have been implicated in cancer pathogenesis. Here we analyze the current evidence of genetic alterations of TRAF molecules available from The Cancer Genome Atlas (TCGA) and the Catalog of Somatic Mutations in Cancer (COSMIC) as well as the published literature, including copy number variations and mutation landscape of TRAFs in various human cancers. Such analyses reveal that both gain- and loss-of-function genetic alterations of different TRAF proteins are commonly present in a number of human cancers. These include pancreatic cancer, meningioma, breast cancer, prostate cancer, lung cancer, liver cancer, head and neck cancer, stomach cancer, colon cancer, bladder cancer, uterine cancer, melanoma, sarcoma, and B cell malignancies, among others. Furthermore, we summarize the key in vivo and in vitro evidence that demonstrates the causal roles of genetic alterations of TRAF proteins in tumorigenesis within different cell types and organs. Taken together, the information presented in this review provides a rationale for the development of therapeutic strategies to manipulate TRAF proteins or TRAF-dependent signaling pathways in different human cancers by precision medicine.
Collapse
Affiliation(s)
- Sining Zhu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Juan Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Angeli M. Lu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Haiyan Shan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jianjun Feng
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education of the People's Republic of China, Fisheries College of Jimei University, Xiamen, China
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Member, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
15
|
Jung SH, Kim SY, An CH, Lee SH, Jung ES, Park HC, Kim MS, Chung YJ, Lee SH. Clonal Structures of Regionally Synchronous Gastric Adenomas and Carcinomas. Clin Cancer Res 2018; 24:4715-4725. [PMID: 29945994 DOI: 10.1158/1078-0432.ccr-18-0345] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/07/2018] [Accepted: 06/22/2018] [Indexed: 01/11/2023]
Abstract
Purpose: Gastric adenoma (GA) is a premalignant lesion that precedes intestinal-type gastric carcinoma (GC). However, genetic progression mechanisms from GA to GC have not been clarified.Experimental Design: We performed whole-exome sequencing-based mutational analyses for 15 synchronous pairs of attached GAs and GCs.Results: There was no significant difference in the number of driver mutations or copy-number alterations between GAs and GCs. Well-known mutations of TP53, APC, RNF43, and RPL22 were recurrently detected in synchronous GA/GC pairs. In addition, we discovered novel KDM6A, PREX2, FAT1, KMT2C, GLI3, and RPL22 mutations and hypermutation in GAs, but did not identify recurrent drivers for GA-to-GC progression. Clonal structure analyses revealed that most GA/GC pairs exhibit parallel evolution with early divergence rather than stepwise evolution during GA-to-GC progression. Of note, three cases were identified as clonally nonrelated GA/GC pairs despite the lack of histologic differences. We found differences in dominant mutational signatures 1, 6, 15, and 17 in GA/GC trunks, GA branches, and GC branches. Compared with our previous work on synchronous colon adenoma/carcinoma genome structures, where most drivers were in the trunk with parallel evolution, synchronous GA/GC genomes showed a different model of parallel evolution, with many drivers in the branches.Conclusions: The preferred sequence of mutational events during GA-to-GC progression might be more context-dependent than colon adenoma progression. Our results show that nonclonal synchronous GA/GC is common and that GA genomes have already acquired distinct genomic alterations, suggesting caution in the diagnosis of synchronous GA and GC, especially in residual or recurrent cases. Clin Cancer Res; 24(19); 4715-25. ©2018 AACR.
Collapse
Affiliation(s)
- Seung-Hyun Jung
- Department of Cancer Evolution Research Center, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea.,Department of Integrated Research Center for Genome Polymorphism, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| | - Shin Young Kim
- Department of Integrated Research Center for Genome Polymorphism, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea.,Department of Surgery, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| | - Chang Hyeok An
- Department of Surgery, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sun Jung
- Department of Hospital Pathology, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| | - Hyeon-Chun Park
- Department of Integrated Research Center for Genome Polymorphism, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| | - Min Sung Kim
- Department of Cancer Evolution Research Center, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea.,Department of Pathology, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| | - Yeun-Jun Chung
- Department of Cancer Evolution Research Center, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea. .,Department of Integrated Research Center for Genome Polymorphism, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea.,Department of Microbiology, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| | - Sug Hyung Lee
- Department of Cancer Evolution Research Center, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea. .,Department of Pathology, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
16
|
Shin S, Park HC, Kim MS, Han MR, Lee SH, Jung SH, Lee SH, Chung YJ. Whole-exome sequencing identified mutational profiles of squamous cell carcinomas of anus. Hum Pathol 2018; 80:1-10. [PMID: 29555573 DOI: 10.1016/j.humpath.2018.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 12/13/2022]
Abstract
Anal squamous cell carcinoma (ASCC), either with human papillomavirus (+) or (-), is a neoplastic disease with frequent recurrence and metastasis. To characterize ASCC genomes, we attempted to disclose novel alterations of ASCC genomes and other genetic features including mutation signatures. We performed whole-exome sequencing and copy number alteration (CNA) profiling for 8 ASCC samples from 6 patients (2 cases with primary and recurrent/metastatic tumors). We found known ASCC mutations (TP53, CDKN2A, and PIK3CA) and CNAs (gains on 3q and 19q and losses on 11q and 13q). In addition, we discovered novel mutations in HRAS and ARID1A and CNAs (gain on 8q and losses 5q, 9p, 10q, and 19p) that had not been reported in ASCCs. We identified 4 signature patterns of the mutations (signatures 1 and 2 with deamination of 5-methyl-cytosin, signature 3 with APOBEC, and signature 4 with mismatch repair) in the ASCCs. Although signatures 1 to 3 have been detected in other SCCs, signature 4 was first identified in ASCCs. In addition, we first found that ASCCs harbored chromothripsis, copy-neutral losses of heterozygosity, and focal amplification of KLF5 super-enhancer. Analyses of primary and recurrent/metastatic pair genomes revealed that driver events in development and progression of ASCC might not be uniform. Our data indicate that ASCCs may have similar mutation and CNA profiles to other SCCs, but that there are unique genomic features of ASCCs as well. Our data may provide useful information for ASCC pathogenesis and for developing clinical strategies for ASCC.
Collapse
Affiliation(s)
- Sun Shin
- Department of Microbiology, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Integrated Research Center for Genome Polymorphism, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Hyeon-Chun Park
- Integrated Research Center for Genome Polymorphism, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Min Sung Kim
- Department of Pathology, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Cancer Evolution Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Mi-Ryung Han
- Integrated Research Center for Genome Polymorphism, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Seung Hyun Jung
- Integrated Research Center for Genome Polymorphism, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Cancer Evolution Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Sug Hyung Lee
- Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Department of Pathology, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Cancer Evolution Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| | - Yeun-Jun Chung
- Department of Microbiology, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Integrated Research Center for Genome Polymorphism, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Cancer Evolution Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea; Precision Medicine Research Center, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
17
|
Jung SH, Kim MS, Jung CK, Park HC, Kim SY, Liu J, Bae JS, Lee SH, Kim TM, Lee SH, Chung YJ. Mutational burdens and evolutionary ages of thyroid follicular adenoma are comparable to those of follicular carcinoma. Oncotarget 2018; 7:69638-69648. [PMID: 27626165 PMCID: PMC5342504 DOI: 10.18632/oncotarget.11922] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 09/02/2016] [Indexed: 12/30/2022] Open
Abstract
Follicular thyroid adenoma (FTA) precedes follicular thyroid carcinoma (FTC) by definition with a favorable prognosis compared to FTC. However, the genetic mechanism of FTA to FTC progression remains unknown. For this, it is required to disclose FTA and FTC genomes in mutational and evolutionary perspectives. We performed whole-exome sequencing and copy number profiling of 14 FTAs and 13 FTCs, which exhibited previously-known gene mutations (NRAS, HRAS, BRAF, TSHR and EIF1AX) and copy number alterations (CNAs) (22q loss and 1q gain) in follicular tumors. In addition, we found eleven potential cancer-related genes with mutations (EZH1, SPOP, NF1, TCF12, IGF2BP3, KMT2C, CNOT1, BRIP1, KDM5C, STAG2 and MAP4K3) that have not been reported in thyroid follicular tumors. Of note, FTA genomes showed comparable levels of mutations to FTC in terms of the number, sequence composition and functional consequences (potential driver mutations) of mutations. Analyses of evolutionary ages using somatic mutations as molecular clocks further identified that FTA genomes were as old as FTC genomes. Whole-transcriptome sequencing did not find any gene fusions with potential significance. Our data indicate that FTA genomes may be as old as FTC genomes, thus suggesting that follicular thyroid tumor genomes during the transition from FTA to FTC may stand stable at genomic levels in contrast to the discernable changes at pathologic and clinical levels. Also, the data suggest a possibility that the mutational profiles obtained from early biopsies may be useful for the molecular diagnosis and therapeutics of follicular tumor patients.
Collapse
Affiliation(s)
- Seung-Hyun Jung
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Department of Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Min Sung Kim
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chan Kwon Jung
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyun-Chun Park
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Department of Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - So Youn Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Department of Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jieying Liu
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Department of Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ja-Seong Bae
- Department of General Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Tae-Min Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sug Hyung Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yeun-Jun Chung
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Department of Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
18
|
Mutational signatures and chromosome alteration profiles of squamous cell carcinomas of the vulva. Exp Mol Med 2018; 50:e442. [PMID: 29422544 PMCID: PMC5903820 DOI: 10.1038/emm.2017.265] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/30/2017] [Accepted: 09/04/2017] [Indexed: 12/30/2022] Open
Abstract
Vulvar squamous cell carcinoma (SCC) consists of two different etiologic categories: human papilloma virus (HPV)-associated (HPV (+)) and HPV-non-associated (HPV (−)). There have been no genome-wide studies on the genetic alterations of vulvar SCCs or on the differences between HPV (+) and HPV (−) vulvar SCCs. In this study, we performed whole-exome sequencing and copy number profiling of 6 HPV (+) and 9 HPV (−) vulvar SCCs and found known mutations (TP53, CDKN2A and HRAS) and copy number alterations (CNAs) (7p and 8q gains and 2q loss) in HPV (−) SCCs. In HPV (+), we found novel mutations in PIK3CA, BRCA2 and FBXW7 that had not been reported in vulvar SCCs. HPV (−) SCCs exhibited more mutational loads (numbers of nonsilent mutations and driver mutations) than HPV (+) SCCs, but the CNA loads and mutation signatures between HPV (+) and HPV (−) SCCs did not differ. Of note, 40% and 40% of the 15 vulvar SCCs harbored PIK3CA and FAT1 alterations, respectively. In addition, we found that the SCCs harbored kataegis (a localized hypermutation) in 2 HPV (+) SCCs and copy-neutral losses of heterozygosity in 4 (one HPV (+) and 3 HPV (−)) SCCs. Our data indicate that HPV (+) and HPV (−) vulvar SCCs may have different mutation and CNA profiles but that there are genomic features common to SCCs. Our data provide useful information for both HPV (+) and HPV (−) vulvar SCCs and may aid in the development of clinical treatment strategies.
Collapse
|
19
|
Lim CH, Cho YK, Kim SW, Choi MG, Rhee JK, Chung YJ, Lee SH, Kim TM. The chronological sequence of somatic mutations in early gastric carcinogenesis inferred from multiregion sequencing of gastric adenomas. Oncotarget 2018; 7:39758-39767. [PMID: 27175599 PMCID: PMC5129968 DOI: 10.18632/oncotarget.9250] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/26/2016] [Indexed: 12/26/2022] Open
Abstract
Mutation profiles and intratumoral heterogeneity are not well understood for benign gastric adenomas, some of which progress into malignant gastric adenocarcinomas. In this study, we performed whole-exome sequencing of three microsatellite stable (MSS) and two microsatellite instability-high (MSI-H) gastric adenomas with three regional tumor biopsies per case. We observed that the mutation abundance of benign gastric adenomas was comparable to those of gastric adenocarcinomas, suggesting that the mutational makeup for gastric carcinogenesis may already be achieved in benign adenomas. The extent of intratumoral heterogeneity was more substantial for MSS genomes in that only 1% - 14% of somatic mutations were common across the regional biopsies or 'public', while 50% - 94% of mutations were public in MSI-H gastric adenomas. We observed biallelic, loss-of-functional events of APC with truncating mutations and/or 5q losses for all cases, mostly as public events. All MSS gastric adenomas also harbored ARID2 truncating mutations, often as multiple, region-specific ones indicative of convergent evolution. Hotspot missense mutations on known cancer genes such as ERBB2 and KRAS were largely observed as region-specific aberrations. These findings suggest that biallelic functional APC inactivation initiates the gastric carcinogenesis and is followed by mutations of histone modifiers and then activation of known cancer-related genes. As the first exome-wide multi-region mutational profiling of gastric adenomas, our study provides clues on the chronological sequence of somatic mutations and their clonal architectures in early gastric carcinogenesis.
Collapse
Affiliation(s)
- Chul-Hyun Lim
- Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Yu Kyung Cho
- Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Sang Woo Kim
- Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Myung-Gyu Choi
- Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Je-Keun Rhee
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Yeun-Jun Chung
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Sug-Hyung Lee
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Tae-Min Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| |
Collapse
|
20
|
Chromatin remodeling gene AT-rich interactive domain-containing protein 1A suppresses gastric cancer cell proliferation by targeting PIK3CA and PDK1. Oncotarget 2018; 7:46127-46141. [PMID: 27323812 PMCID: PMC5216786 DOI: 10.18632/oncotarget.10060] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 06/01/2016] [Indexed: 12/13/2022] Open
Abstract
The tumor suppressor gene AT-rich interactive domain-containing protein 1A (ARID1A) was frequently mutated in cancers. The modulation mechanism of ARID1A for PI3K/AKT signaling in gastric cancer (GC) remains elusive. Here, we found that depletion of endogenous ARID1A enhanced the in vitro proliferation, colony formation, cellular growth, nutrient uptake and in vivo xenograft tumor growth of GC cells. PI3K/AKT activation by ARID1A-silencing was profiled using a phospho-protein antibody array. The phosphorylation of PDK1, AKT, GSK3β and 70S6K, and the protein and mRNA expressions of PI3K and PDK1, were upregulated by ARID1A-silencing. Chromatin immunoprecipitation and luciferase reporter assay revealed that ARID1A-involved SWI/SNF complex inhibited PIK3CA and PDK1 transcription by direct binding to their promoters. Serial deletion mutation analyses revealed that the ARID1A central region containing the HIC1-binding domain, but not the ARID DNA-binding domain and the C-terminal domain, was essential for the inhibition of GC cell growth, PI3K/AKT pathway phosphorylation and its transcriptional modulation activity of PIK3CA and PDK1. The proliferation, cellular growth and glucose consumption of ARID1A-deficient GC cells were efficiently prohibited by allosteric inhibitors mk2206 and LY294002, which targeting AKT and PI3K, respectively. Both inhibitors also downregulated the phosphorylation of PI3K/AKT pathway in ARID1A-deficient GC cells. Such cells were sensitized to the treatment of LY294002, and AT7867, another inhibitor of AKT and p70S6K. The administration of LY294002 alone inhibited the in vivo growth of ARID1A- deficient GC cells in mouse xenograft model. Our study provides a novel insight into the modulatory function and mechanism of ARID1A in PI3K/AKT signaling in GC.
Collapse
|
21
|
Zhu S, Jin J, Gokhale S, Lu AM, Shan H, Feng J, Xie P. Genetic Alterations of TRAF Proteins in Human Cancers. Front Immunol 2018. [PMID: 30294322 DOI: 10.3389/fimmu.2018.02111/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
The tumor necrosis factor receptor (TNF-R)-associated factor (TRAF) family of cytoplasmic adaptor proteins regulate the signal transduction pathways of a variety of receptors, including the TNF-R superfamily, Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I-like receptors (RLRs), and cytokine receptors. TRAF-dependent signaling pathways participate in a diverse array of important cellular processes, including the survival, proliferation, differentiation, and activation of different cell types. Many of these TRAF-dependent signaling pathways have been implicated in cancer pathogenesis. Here we analyze the current evidence of genetic alterations of TRAF molecules available from The Cancer Genome Atlas (TCGA) and the Catalog of Somatic Mutations in Cancer (COSMIC) as well as the published literature, including copy number variations and mutation landscape of TRAFs in various human cancers. Such analyses reveal that both gain- and loss-of-function genetic alterations of different TRAF proteins are commonly present in a number of human cancers. These include pancreatic cancer, meningioma, breast cancer, prostate cancer, lung cancer, liver cancer, head and neck cancer, stomach cancer, colon cancer, bladder cancer, uterine cancer, melanoma, sarcoma, and B cell malignancies, among others. Furthermore, we summarize the key in vivo and in vitro evidence that demonstrates the causal roles of genetic alterations of TRAF proteins in tumorigenesis within different cell types and organs. Taken together, the information presented in this review provides a rationale for the development of therapeutic strategies to manipulate TRAF proteins or TRAF-dependent signaling pathways in different human cancers by precision medicine.
Collapse
Affiliation(s)
- Sining Zhu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Juan Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Angeli M Lu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Haiyan Shan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jianjun Feng
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education of the People's Republic of China, Fisheries College of Jimei University, Xiamen, China
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Member, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
22
|
Gene editing rescue of a novel MPL mutant associated with congenital amegakaryocytic thrombocytopenia. Blood Adv 2017; 1:1815-1826. [PMID: 29296828 DOI: 10.1182/bloodadvances.2016002915] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 08/10/2017] [Indexed: 12/19/2022] Open
Abstract
Thrombopoietin (Tpo) and its receptor (Mpl) are the principal regulators of early and late thrombopoiesis and hematopoietic stem cell maintenance. Mutations in MPL can drastically impair its function and be a contributing factor in multiple hematologic malignancies, including congenital amegakaryocytic thrombocytopenia (CAMT). CAMT is characterized by severe thrombocytopenia at birth, which progresses to bone marrow failure and pancytopenia. Here we report unique familial cases of CAMT that presented with a previously unreported MPL mutation: T814C (W272R) in the background of the activating MPL G117T (K39N or Baltimore) mutation. Confocal microscopy, proliferation and surface biotinylation assays, co-immunoprecipitation, and western blotting analysis were used to elucidate the function and trafficking of Mpl mutants. Results showed that Mpl protein bearing the W272R mutation, alone or together with the K39N mutation, lacks detectable surface expression while being strongly colocalized with the endoplasmic reticulum (ER) marker calreticulin. Both WT and K39N-mutated Mpl were found to be signaling competent, but single or double mutants bearing W272R were unresponsive to Tpo. Function of the deficient Mpl receptor could be rescued by using 2 separate approaches: (1) GRASP55 overexpression, which partially restored Tpo-induced signaling of mutant Mpl by activating an autophagy-dependent secretory pathway and thus forcing ER-trapped immature receptors to traffic to the cell surface; and (2) CRISPR-Cas9 gene editing used to repair MPL T814C mutation in transfected cell lines and primary umbilical cord blood-derived CD34+ cells. We demonstrate proof of principle for rescue of mutant Mpl function by using gene editing of primary hematopoietic stem cells, which indicates direct therapeutic applications for CAMT patients.
Collapse
|
23
|
Abstract
Helicobacter pylori infection is the most important cause of human gastric cancer worldwide. Gastric cancer develops over a long time after H. pylori infection via stepwise accumulation of genetic alterations and positive selection of cells with growth advantages. H. pylori itself and the resultant chronic inflammation lead to the emergence of genetic alterations in gastric epithelial cells via increased susceptibility of these cells to DNA damage. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) in inflammatory and gastric epithelial cells, as well as the expression of cytidine deaminase in gastric epithelial cells, may link H. pylori-related inflammation and DNA damage. Recent comprehensive analyses of gastric cancer genomes provide clues for the possible molecular mechanisms of gastric carcinogenesis. In this chapter, we describe how genetic alterations emerge during gastric carcinogenesis related to H. pylori infection.
Collapse
|
24
|
Kim YS, Jeong H, Choi JW, Oh HE, Lee JH. Unique characteristics of ARID1A mutation and protein level in gastric and colorectal cancer: A meta-analysis. Saudi J Gastroenterol 2017; 23:268-274. [PMID: 28937020 PMCID: PMC5625362 DOI: 10.4103/sjg.sjg_184_17] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND/AIM Recently, AT-rich interactive domain-containing 1A protein (ARID1A) has been identified as a novel tumor suppressor gene in gastric cancer (GC) and colorectal cancer (CRC). However, the clinicopathologic value of ARID1A mutation or protein level in GC and CRC patients is controversial. Hence, we conducted a meta-analysis on the relationship between ARID1A aberrations and clinicopathologic parameters in GC and CRC. MATERIALS AND METHODS Relevant published studies were selected from PubMed and EMBASE. The effect sizes of ARID1A mutation or level on the patient's clinicopathologic parameters were calculated by prevalence rate or odds ratio (OR) or hazard ratio (HR), respectively. The effect sizes were combined using a random-effects model. RESULTS The frequency of ARID1A mutation and loss of ARID1A protein expression in GC patients was 17% and 27%, respectively. The loss of ARID1A protein expression of GC patients was significantly associated with advanced tumor depth (OR = 1.8, P = 0.004), lymph node metastasis (OR = 1.4, P = 0.001), and unfavorable adjusted overall survival (HR = 1.5, P < 0.001). ARID1A mutation of GC was significantly associated with microsatellite instability (MSI) (OR = 24.5, P < 0.001) and EBV infection (OR = 2.6, P = 0.001). The frequency of ARID1A mutation and ARID1A protein expression loss in CRC patients was approximately 12-13%. Interestingly, the loss of ARID1A protein expression in CRC patients was significantly associated with poorly differentiated grade (OR = 4.0, P < 0.001) and advanced tumor depth (OR = 1.8, P = 0.012). CONCLUSION Our meta-analysis revealed that ARID1A alterations may be involved in the carcinogenesis of GC by EBV infection and MSI. The loss of ARID1A protein expression may be a marker of poor prognosis in GC and CRC patients.
Collapse
Affiliation(s)
- Young-Sik Kim
- Department of Pathology, Korea University Ansan Hospital, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do, Republic of Korea
| | - Hoiseon Jeong
- Department of Pathology, Korea University Ansan Hospital, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do, Republic of Korea
| | - Jung-Woo Choi
- Department of Pathology, Korea University Ansan Hospital, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do, Republic of Korea
| | - Hwa Eun Oh
- Department of Pathology, Korea University Ansan Hospital, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do, Republic of Korea
| | - Ju-Han Lee
- Department of Pathology, Korea University Ansan Hospital, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do, Republic of Korea,Address for correspondence: Dr. Ju-Han Lee, Department of Pathology, Korea University Ansan Hospital, 123, Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-do, 15355, Republic of Korea. E-mail:
| |
Collapse
|
25
|
Min BH, Hwang J, Kim NKD, Park G, Kang SY, Ahn S, Ahn S, Ha SY, Lee YK, Kushima R, Van Vrancken M, Kim MJ, Park C, Park HY, Chae J, Jang SS, Kim SJ, Kim YH, Kim JI, Kim KM. Dysregulated Wnt signalling and recurrent mutations of the tumour suppressorRNF43in early gastric carcinogenesis. J Pathol 2016; 240:304-314. [DOI: 10.1002/path.4777] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/06/2016] [Accepted: 08/03/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Byung-Hoon Min
- Department of Medicine, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Jinha Hwang
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
| | - Nayoung KD Kim
- Samsung Genome Institute; Samsung Medical Centre; Seoul Korea
| | - Gibeom Park
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
| | - So Young Kang
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Sangjeong Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Soomin Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Sang Yun Ha
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Yun Kyung Lee
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Ryoji Kushima
- Department of Pathology, Undergraduate School of Medicine; Shiga University of Medical Science; Shiga Japan
| | - Michael Van Vrancken
- Department of Pathology and Laboratory Medicine; Tulane University School of Medicine; New Orleans LA USA
| | - Min Jung Kim
- Cancer Research Institute; Seoul National University College of Medicine; Seoul Korea
| | - Changho Park
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
| | - Ha Young Park
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Jeesoo Chae
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
| | - Se Song Jang
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
| | - Sung Jin Kim
- Samsung Biomedical Research Institute; Samsung Medical Centre; Seoul Korea
| | - Young-Ho Kim
- Department of Medicine, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Jong-Il Kim
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
- Cancer Research Institute; Seoul National University College of Medicine; Seoul Korea
- Genomic Medicine Institute, Medical Research Centre; Seoul National University; Seoul Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| |
Collapse
|
26
|
Whole-exome sequencing identifies recurrent AKT1 mutations in sclerosing hemangioma of lung. Proc Natl Acad Sci U S A 2016; 113:10672-7. [PMID: 27601661 DOI: 10.1073/pnas.1606946113] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pulmonary sclerosing hemangioma (PSH) is a benign tumor with two cell populations (epithelial and stromal cells), for which genomic profiles remain unknown. We conducted exome sequencing of 44 PSHs and identified recurrent somatic mutations of AKT1 (43.2%) and β-catenin (4.5%). We used a second subset of 24 PSHs to confirm the high frequency of AKT1 mutations (overall 31/68, 45.6%; p.E17K, 33.8%) and recurrent β-catenin mutations (overall 3 of 68, 4.4%). Of the PSHs without AKT1 mutations, two exhibited AKT1 copy gain. AKT1 mutations existed in both epithelial and stromal cells. In two separate PSHs from one patient, we observed two different AKT1 mutations, indicating they were not disseminated but independent arising tumors. Because the AKT1 mutations were not found to co-occur with β-catenin mutations (or any other known driver alterations) in any of the PSHs studied, we speculate that this may be the single-most common driver alteration to develop PSHs. Our study revealed genomic differences between PSHs and lung adenocarcinomas, including a high rate of AKT1 mutation in PSHs. These genomic features of PSH identified in the present study provide clues to understanding the biology of PSH and for differential genomic diagnosis of lung tumors.
Collapse
|
27
|
Kloth M, Ruesseler V, Engel C, Koenig K, Peifer M, Mariotti E, Kuenstlinger H, Florin A, Rommerscheidt-Fuss U, Koitzsch U, Wodtke C, Ueckeroth F, Holzapfel S, Aretz S, Propping P, Loeffler M, Merkelbach-Bruse S, Odenthal M, Friedrichs N, Heukamp LC, Zander T, Buettner R. Activating ERBB2/HER2 mutations indicate susceptibility to pan-HER inhibitors in Lynch and Lynch-like colorectal cancer. Gut 2016; 65:1296-305. [PMID: 26001389 DOI: 10.1136/gutjnl-2014-309026] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 04/07/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Microsatellite instability (MSI) is detected in approximately 15% of all colorectal cancers (CRC) and virtually in all cases with Lynch syndrome. The MSI phenotype is caused by dysfunctional mismatch repair (MMR) and leads to accumulation of DNA replication errors. Sporadic MSI CRC often harbours BRAF(V600E); however, no consistent data exist regarding targeted treatment approaches in BRAF(wt) MSI CRC. DESIGN Mutations and quantitative MSI were analysed by deep sequencing in 196 formalin fixed paraffin embedded (FFPE) specimens comprising Lynch and Lynch-like CRCs from the German Hereditary Nonpolyposis Colorectal Cancer registry. Functional relevance of recurrent ERBB2/HER2 mutations was investigated in CRC cell lines using reversible and irreversible HER-targeting inhibitors, EGFR-directed antibody cetuximab, HER2-directed antibody trastuzumab and siRNA-mediated ERBB2/HER2 knockdown. RESULTS Quantification of nucleotide loss in non-coding mononucleotide repeats distinguished microsatellite status with very high accuracy (area under curve=0.9998) and demonstrated progressive losses with deeper invasion of MMR-deficient colorectal neoplasms (p=0.008). Characterisation of BRAF(wt) MSI CRC revealed hot-spot mutations in well-known oncogenic drivers, including KRAS (38.7%), PIK3CA (36.5%), and ERBB2 (15.0%). L755S and V842I substitutions in ERBB2 were highly recurrent. Functional analyses in ERBB2-mutated MSI CRC cell lines revealed a differential response to HER-targeting compounds and superiority of irreversible pan-HER inhibitors. CONCLUSIONS We developed a high-throughput deep sequencing approach for concomitant MSI and mutational analyses in FFPE specimens. We provided novel insights into clinically relevant alterations in MSI CRC and a rationale for targeting ERBB2/HER2 mutations in Lynch and Lynch-like CRC.
Collapse
Affiliation(s)
- Michael Kloth
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Vanessa Ruesseler
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Christoph Engel
- Institute of Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Katharina Koenig
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Martin Peifer
- Department of Translational Genomics, Centre for Molecular Medicine Cologne, Cologne, Germany
| | - Erika Mariotti
- Department of Translational Genomics, Centre for Molecular Medicine Cologne, Cologne, Germany
| | - Helen Kuenstlinger
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Alexandra Florin
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Ursula Rommerscheidt-Fuss
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Ulrike Koitzsch
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Claudia Wodtke
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Frank Ueckeroth
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | | | - Stefan Aretz
- Institute of Human Genetics, University Hospital Bonn, Bonn, Germany
| | - Peter Propping
- Institute of Human Genetics, University Hospital Bonn, Bonn, Germany
| | - Markus Loeffler
- Institute of Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Sabine Merkelbach-Bruse
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Margarete Odenthal
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Nicolaus Friedrichs
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Lukas Carl Heukamp
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Thomas Zander
- Department of Internal Medicine I, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, Centre for Integrated Oncology, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
28
|
Lim B, Kim JH, Kim M, Kim SY. Genomic and epigenomic heterogeneity in molecular subtypes of gastric cancer. World J Gastroenterol 2016; 22:1190-1201. [PMID: 26811657 PMCID: PMC4716030 DOI: 10.3748/wjg.v22.i3.1190] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 09/08/2015] [Accepted: 10/12/2015] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is a complex disease that is affected by multiple genetic and environmental factors. For the precise diagnosis and effective treatment of gastric cancer, the heterogeneity of the disease must be simplified; one way to achieve this is by dividing the disease into subgroups. Toward this effort, recent advances in high-throughput sequencing technology have revealed four molecular subtypes of gastric cancer, which are classified as Epstein-Barr virus-positive, microsatellite instability, genomically stable, and chromosomal instability subtypes. We anticipate that this molecular subtyping will help to extend our knowledge for basic research purposes and will be valuable for clinical use. Here, we review the genomic and epigenomic heterogeneity of the four molecular subtypes of gastric cancer. We also describe a mutational meta-analysis and a reanalysis of DNA methylation that were performed using previously reported gastric cancer datasets.
Collapse
|
29
|
Jung SH, Choi YJ, Kim MS, Baek IP, Lee SH, Lee AW, Hur SY, Kim TM, Lee SH, Chung YJ. Progression of naive intraepithelial neoplasia genome to aggressive squamous cell carcinoma genome of uterine cervix. Oncotarget 2015; 6:4385-93. [PMID: 25738363 PMCID: PMC4414197 DOI: 10.18632/oncotarget.2981] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/20/2014] [Indexed: 01/06/2023] Open
Abstract
Although cervical intraepithelial neoplasia (CIN) is considered a neoplasia, its genomic alterations remain unknown. For this, we performed whole-exome sequencing and copy number profiling of three CINs, a microinvasive carcinoma (MIC) and four cervical squamous cell carcinomas (CSCC). Both total mutation and driver mutation numbers of the CINs were significantly fewer than those of the MIC/CSCCs (P = 0.036 and P = 0.018, respectively). Importantly, PIK3CA was altered in all MIC/CSCCs by either mutation or amplification, but not in CINs. The CINs harbored significantly lower numbers of copy number alterations (CNAs) than the MIC/CSCCs as well (P = 0.036). Pathway analysis predicted that the MIC/CSCCs were enriched with cancer-related signalings such as cell adhesion, mTOR signaling pathway and cell migration that were depleted in the CINs. The mutation-based estimation of evolutionary ages identified that CIN genomes were younger than MIC/CSCC genomes. The data indicate that CIN genomes harbor unfixed mutations in addition to human papilloma virus infection but require additional driver hits such as PIK3CA, TP53, STK11 and MAPK1 mutations for CSCC progression. Taken together, our data may explain the long latency from CIN to CSCC progression and provide useful information for molecular diagnosis of CIN and CSCC.
Collapse
Affiliation(s)
- Seung-Hyun Jung
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Department of Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Youn Jin Choi
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Min Sung Kim
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - In-Pyo Baek
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Department of Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Ah Won Lee
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Soo Young Hur
- Department of Obstetrics/Gynecology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Tae-Min Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Sug Hyung Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Yeun-Jun Chung
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Department of Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| |
Collapse
|
30
|
Genetic Progression of High Grade Prostatic Intraepithelial Neoplasia to Prostate Cancer. Eur Urol 2015; 69:823-30. [PMID: 26542946 DOI: 10.1016/j.eururo.2015.10.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 10/15/2015] [Indexed: 11/22/2022]
Abstract
BACKGROUND Although high grade prostatic intraepithelial neoplasia (HGPIN) is considered a neoplastic lesion that precedes prostate cancer (PCA), the genomic structures of HGPIN remain unknown. OBJECTIVE Identification of the genomic landscape of HGPIN and the genomic differences between HGPIN and PCA that may drive the progression to PCA. DESIGN, SETTINGS, AND PARTICIPANTS We analyzed 20 regions of paired HGPIN and PCA from six patients using whole-exome sequencing and array-comparative genomic hybridization. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Somatic mutation and copy number alteration (CNA) profiles of paired HGPIN and PCA were measured and compared. RESULTS AND LIMITATIONS The number of total mutations and CNAs of HGPINs were significantly fewer than those of PCAs. Mutations in FOXA1 and CNAs (1q and 8q gains) were detected in both HGPIN and PCA ('common'), suggesting their roles in early PCA development. Mutations in SPOP, KDM6A, and KMT2D were 'PCA-specific', suggesting their roles in HGPIN progression to PCA. The 8p loss was either 'common' or 'PCA-specific'. In-silico estimation of evolutionary ages predicted that HGPIN genomes were much younger than PCA genomes. Our data show that PCAs are direct descendants of HGPINs in most cases that require more genomic alterations to progress to PCA. The nature of heterogeneous HGPIN population that might attenuate genomic signals should further be studied. CONCLUSIONS HGPIN genomes harbor relatively fewer mutations and CNAs than PCA but require additional hits for the progression. PATIENT SUMMARY In this study, we suggest a systemic diagram from high grade prostatic intraepithelial neoplasia (HGPIN) to prostate cancer (PCA). Our results provide a clue to explain the long latency from HGPIN to PCA and provide useful information for the genetic diagnosis of HGPIN and PCA.
Collapse
|
31
|
Tan P, Yeoh KG. Genetics and Molecular Pathogenesis of Gastric Adenocarcinoma. Gastroenterology 2015; 149:1153-1162.e3. [PMID: 26073375 DOI: 10.1053/j.gastro.2015.05.059] [Citation(s) in RCA: 357] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/18/2015] [Accepted: 05/20/2015] [Indexed: 02/07/2023]
Abstract
Gastric cancer (GC) is globally the fifth most common cancer and third leading cause of cancer death. A complex disease arising from the interaction of environmental and host-associated factors, key contributors to GC's high mortality include its silent nature, late clinical presentation, and underlying biological and genetic heterogeneity. Achieving a detailed molecular understanding of the various genomic aberrations associated with GC will be critical to improving patient outcomes. The recent years has seen considerable progress in deciphering the genomic landscape of GC, identifying new molecular components such as ARID1A and RHOA, cellular pathways, and tissue populations associated with gastric malignancy and progression. The Cancer Genome Atlas (TCGA) project is a landmark in the molecular characterization of GC. Key challenges for the future will involve the translation of these molecular findings to clinical utility, by enabling novel strategies for early GC detection, and precision therapies for individual GC patients.
Collapse
Affiliation(s)
- Patrick Tan
- Cancer and Stem Cell Biology Program, Duke-National University of Singapore Graduate Medical School, Singapore; Genome Institute of Singapore, Agency for Science, Technology, and Research, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore; Cellular and Molecular Research, National Cancer Centre Singapore, Singapore; Singapore Gastric Cancer Consortium, Singapore.
| | - Khay-Guan Yeoh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Gastroenterology and Hepatology, National University Health System, Singapore; Singapore Gastric Cancer Consortium, Singapore.
| |
Collapse
|
32
|
Lin Y, Wu Z, Guo W, Li J. Gene mutations in gastric cancer: a review of recent next-generation sequencing studies. Tumour Biol 2015; 36:7385-94. [PMID: 26364057 DOI: 10.1007/s13277-015-4002-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/25/2015] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. Although some driver genes have been identified in GC, the molecular compositions of GC have not been fully understood. The development of next-generation sequencing (NGS) provides a high-throughput and systematic method to identify all genetic alterations in the cancer genome, especially in the field of mutation detection. NGS studies in GC have discovered some novel driver mutations. In this review, we focused on novel gene mutations discovered by NGS studies, along with some well-known driver genes in GC. We organized mutated genes from the perspective of related biological pathways. Mutations in genes relating to genome integrity (TP53, BRCA2), chromatin remodeling (ARID1A), cell adhesion (CDH1, FAT4, CTNNA1), cytoskeleton and cell motility (RHOA), Wnt pathway (CTNNB1, APC, RNF43), and RTK pathway (RTKs, RAS family, MAPK pathway, PIK pathway) are discussed. Efforts to establish a molecular classification based on NGS data which is valuable for future targeted therapy for GC are introduced. Comprehensive dissection of the molecular profile of GC cannot only unveil the molecular basis for GC but also identify genes of clinical utility, especially potential and specific therapeutic targets for GC.
Collapse
Affiliation(s)
- Y Lin
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Z Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - W Guo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - J Li
- Tongji University Tianyou Hospital, Shanghai, 200331, China.
| |
Collapse
|
33
|
Abstract
Helicobacter pylori infection plays a crucial role in gastric carcinogenesis. H pylori exerts oncogenic effects on gastric mucosa through complex interaction between bacterial virulence factors and host inflammatory responses. On the other hand, gastric cancer develops via stepwise accumulation of genetic and epigenetic alterations in H pylori-infected gastric mucosa. Recent comprehensive analyses of gastric cancer genomes indicate a multistep process of genetic alterations as well as possible molecular mechanisms of gastric carcinogenesis. Both genetic processes of gastric cancer development and molecular oncogenic pathways related to H pylori infection are important to completely understand the pathogenesis of H pylori-related gastric cancer.
Collapse
|