1
|
Zhao Y, Qin D, Li X, Wang T, Zhang T, Rao X, Min L, Wan Z, Luo C, Xiao M. Identification of NINJ1 as a novel prognostic predictor for retroperitoneal liposarcoma. Discov Oncol 2024; 15:155. [PMID: 38733554 PMCID: PMC11088571 DOI: 10.1007/s12672-024-01016-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Retroperitoneal liposarcoma (RPLS) is known for its propensity for local recurrence and short survival time. We aimed to identify a credible and specific prognostic biomarker for RPLS. METHODS Cases from The Cancer Genome Atlas (TCGA) sarcoma dataset were included as the training group. Co-expression modules were constructed using weighted gene co-expression network analysis (WGCNA) to explore associations between modules and survival. Survival analysis of hub genes was performed using the Kaplan-Meier method. In addition, independent external validation was performed on a cohort of 135 Chinese RPLS patients from the REtroperitoneal SArcoma Registry (RESAR) study (NCT03838718). RESULTS A total of 19 co-expression modules were constructed based on the expression levels of 26,497 RNAs in the TCGA cohort. Among these modules, the green module exhibited a positive correlation with overall survival (OS, p = 0.10) and disease-free survival (DFS, p = 0.06). Gene set enrichment analysis showed that the green module was associated with endocytosis and soft-tissue sarcomas. Survival analysis demonstrated that NINJ1, a hub gene within the green module, was positively associated with OS (p = 0.019) in the TCGA cohort. Moreover, in the validation cohort, patients with higher NINJ1 expression levels displayed a higher probability of survival for both OS (p = 0.023) and DFS (p = 0.012). Multivariable Cox analysis further confirmed the independent prognostic significance of NINJ1. CONCLUSIONS We here provide a foundation for the establishment of a consensus prognostic biomarker for RPLS, which should not only facilitate medical treatment but also guide the development of novel targeted drugs.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Da Qin
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Xiangji Li
- Department of Retroperitoneal Tumor Surgery, International Hospital, Peking University, Beijing, China
| | - Tiange Wang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Tong Zhang
- Department of Pathology, International Hospital, Peking University, Beijing, China
| | - Xiaosong Rao
- Department of Pathology, International Hospital, Peking University, Beijing, China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Zhiyi Wan
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Chenghua Luo
- Department of Retroperitoneal Tumor Surgery, Peking University People's Hospital, Beijing, China.
| | - Mengmeng Xiao
- Department of Retroperitoneal Tumor Surgery, International Hospital, Peking University, Beijing, China.
| |
Collapse
|
2
|
Li N, Huang J, He S, Zheng Q, Ye F, Qin Z, Wang D, Xiao T, Mao M, Zhou Z, Tang T, Zhang L, Wang X, Wang Y, Lyu Y, Liu L, Dai L, Wang J, Guan J. The development of a novel zeolite-based assay for efficient and deep plasma proteomic profiling. J Nanobiotechnology 2024; 22:164. [PMID: 38600601 PMCID: PMC11007927 DOI: 10.1186/s12951-024-02404-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Plasma proteins are considered the most informative source of biomarkers for disease diagnosis and monitoring. Mass spectrometry (MS)-based proteomics has been applied to identify biomarkers in plasma, but the complexity of the plasma proteome and the extremely large dynamic range of protein abundances in plasma make the clinical application of plasma proteomics highly challenging. We designed and synthesized zeolite-based nanoparticles to deplete high-abundance plasma proteins. The resulting novel plasma proteomic assay can measure approximately 3000 plasma proteins in a 45 min chromatographic gradient. Compared to those in neat and depleted plasma, the plasma proteins identified by our assay exhibited distinct biological profiles, as validated in several public datasets. A pilot investigation of the proteomic profile of a hepatocellular carcinoma (HCC) cohort identified 15 promising protein features, highlighting the diagnostic value of the plasma proteome in distinguishing individuals with and without HCC. Furthermore, this assay can be easily integrated with all current downstream protein profiling methods and potentially extended to other biofluids. In conclusion, we established a robust and efficient plasma proteomic assay with unprecedented identification depth, paving the way for the translation of plasma proteomics into clinical applications.
Collapse
Affiliation(s)
- Nan Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jingnan Huang
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Shangwen He
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qiaocong Zheng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Department of Oncology, People's Hospital of YangJiang, Yangjiang, 529500, Guangdong, China
| | - Feng Ye
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhengxing Qin
- State Key Laboratory of Heavy Oil Processing, College of Chemical Engineering, China University of Petroleum (East China), Qingdao, 266580, Shandong, China
| | - Dong Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao, 266580, Shandong, China
| | - Ting Xiao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Mengyuan Mao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhenhua Zhou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Tingxi Tang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Longshan Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiaoqing Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yingqiao Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Ying Lyu
- Department of Traditional Chinese Medicine, Nanfang Hospital,, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Laiyu Liu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Lingyun Dai
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| | - Jigang Wang
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, Shenzhen People's Hospital, (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| | - Jian Guan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
3
|
Denu RA, Dann AM, Keung EZ, Nakazawa MS, Nassif Haddad EF. The Future of Targeted Therapy for Leiomyosarcoma. Cancers (Basel) 2024; 16:938. [PMID: 38473300 PMCID: PMC10930698 DOI: 10.3390/cancers16050938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Leiomyosarcoma (LMS) is an aggressive subtype of soft tissue sarcoma that arises from smooth muscle cells, most commonly in the uterus and retroperitoneum. LMS is a heterogeneous disease with diverse clinical and molecular characteristics that have yet to be fully understood. Molecular profiling has uncovered possible targets amenable to treatment, though this has yet to translate into approved targeted therapies in LMS. This review will explore historic and recent findings from molecular profiling, highlight promising avenues of current investigation, and suggest possible future strategies to move toward the goal of molecularly matched treatment of LMS. We focus on targeting the DNA damage response, the macrophage-rich micro-environment, the PI3K/mTOR pathway, epigenetic regulators, and telomere biology.
Collapse
Affiliation(s)
- Ryan A. Denu
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Amanda M. Dann
- Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Emily Z. Keung
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Michael S. Nakazawa
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elise F. Nassif Haddad
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
4
|
Shaheen A, Bauman G, Cacciotti C, Zelcer S, Ramadan S. Methylation and Molecular Profiling to Aid in Diagnosis and Radiation Treatment for an Intracranial Ewing Sarcoma in a Pediatric Patient: A Case Report. Adv Radiat Oncol 2024; 9:101352. [PMID: 38405311 PMCID: PMC10885575 DOI: 10.1016/j.adro.2023.101352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/09/2023] [Indexed: 02/27/2024] Open
Affiliation(s)
- Amber Shaheen
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University London, Ontario, Canada
| | - Glenn Bauman
- Division of Radiation Oncology, Department of Oncology, London Health Sciences Centre & Western University, London, Ontario, Canada
| | - Chantel Cacciotti
- Division of Hematology/Oncology, Department of Pediatrics, London Health Sciences Centre & Western University, London, Ontario, Canada
| | - Shayna Zelcer
- Division of Hematology/Oncology, Department of Pediatrics, London Health Sciences Centre & Western University, London, Ontario, Canada
| | - Sherif Ramadan
- Division of Radiation Oncology, Department of Oncology, London Health Sciences Centre & Western University, London, Ontario, Canada
| |
Collapse
|
5
|
Lesovaya EA, Fetisov TI, Bokhyan BY, Maksimova VP, Kulikov EP, Belitsky GA, Kirsanov KI, Yakubovskaya MG. Genetic, Epigenetic and Transcriptome Alterations in Liposarcoma for Target Therapy Selection. Cancers (Basel) 2024; 16:271. [PMID: 38254762 PMCID: PMC10813500 DOI: 10.3390/cancers16020271] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/25/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024] Open
Abstract
Liposarcoma (LPS) is one of the most common adult soft-tissue sarcomas (STS), characterized by a high diversity of histopathological features as well as to a lesser extent by a spectrum of molecular abnormalities. Current targeted therapies for STS do not include a wide range of drugs and surgical resection is the mainstay of treatment for localized disease in all subtypes, while many LPS patients initially present with or ultimately progress to advanced disease that is either unresectable, metastatic or both. The understanding of the molecular characteristics of liposarcoma subtypes is becoming an important option for the detection of new potential targets and development novel, biology-driven therapies for this disease. Innovative therapies have been introduced and they are currently part of preclinical and clinical studies. In this review, we provide an analysis of the molecular genetics of liposarcoma followed by a discussion of the specific epigenetic changes in these malignancies. Then, we summarize the peculiarities of the key signaling cascades involved in the pathogenesis of the disease and possible novel therapeutic approaches based on a better understanding of subtype-specific disease biology. Although heterogeneity in liposarcoma genetics and phenotype as well as the associated development of resistance to therapy make difficult the introduction of novel therapeutic targets into the clinic, recently a number of targeted therapy drugs were proposed for LPS treatment. The most promising results were shown for CDK4/6 and MDM2 inhibitors as well as for the multi-kinase inhibitors anlotinib and sunitinib.
Collapse
Affiliation(s)
- Ekaterina A. Lesovaya
- N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (K.I.K.)
- Faculty of Oncology, I.P. Pavlov Ryazan State Medical University, Ministry of Health of Russia, 9 Vysokovol’tnaya St., Ryazan 390026, Russia;
- Laboratory of Single Cell Biology, Peoples’ Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow 117198, Russia
| | - Timur I. Fetisov
- N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (K.I.K.)
| | - Beniamin Yu. Bokhyan
- N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (K.I.K.)
| | - Varvara P. Maksimova
- N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (K.I.K.)
| | - Evgeny P. Kulikov
- Faculty of Oncology, I.P. Pavlov Ryazan State Medical University, Ministry of Health of Russia, 9 Vysokovol’tnaya St., Ryazan 390026, Russia;
| | - Gennady A. Belitsky
- N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (K.I.K.)
| | - Kirill I. Kirsanov
- N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (K.I.K.)
- Laboratory of Single Cell Biology, Peoples’ Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow 117198, Russia
| | - Marianna G. Yakubovskaya
- N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, 24 Kashirskoe Shosse, Moscow 115478, Russia; (E.A.L.); (T.I.F.); (B.Y.B.); (V.P.M.); (K.I.K.)
- Laboratory of Single Cell Biology, Peoples’ Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow 117198, Russia
| |
Collapse
|
6
|
Shen C, Shi X, Wen D, Zhang Y, Du Y, Zhang Y, Ma B, Tang H, Yin M, Huang N, Liao T, Zhang TT, Kong C, Wei W, Ji Q, Wang Y. Comprehensive DNA Methylation Profiling of Medullary Thyroid Carcinoma: Molecular Classification, Potential Therapeutic Target, and Classifier System. Clin Cancer Res 2024; 30:127-138. [PMID: 37931242 DOI: 10.1158/1078-0432.ccr-23-2142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/11/2023] [Accepted: 10/25/2023] [Indexed: 11/08/2023]
Abstract
PURPOSE Medullary thyroid carcinoma (MTC) presents a distinct biological context from other thyroid cancers due to its specific cellular origin. This heterogeneous and rare tumor has a high prevalence of advanced diseases, making it crucial to address the limited therapeutic options and enhance complex clinical management. Given the high clinical accessibility of methylation information, we construct the largest MTC methylation cohort to date. EXPERIMENTAL DESIGN Seventy-eight fresh-frozen MTC samples constituted our methylation cohort. The comprehensive study process incorporated machine learning, statistical analysis, and in vitro experiments. RESULTS Our study pioneered the identification of a three-class clustering system for risk stratification, exhibiting pronounced epigenomic heterogeneity. The elevated overall methylation status in MTC-B, combined with the "mutual exclusivity" of hypomethylated sites displayed by MTC-A and MTC-C, distinctively characterized the MTC-specific methylation pattern. Integrating with the transcriptome, we further depicted the features of these three clusters to scrutinize biological properties. Several MTC-specific aberrant DNA methylation events were emphasized in our study. NNAT expression was found to be notably reduced in poor-prognostic MTC-C, with its promoter region overlapping with an upregulated differentially methylated region. In vitro experiments further affirmed NNAT's therapeutic potential. Moreover, we built an elastic-net logistic regression model with a relatively high AUC encompassing 68 probes, intended for future validation and systematic clinical application. CONCLUSIONS Conducting research on diseases with low incidence poses significant challenges, and we provide a robust resource and comprehensive research framework to assist in ongoing MTC case inclusion and facilitate in-depth dissection of its molecular biological features.
Collapse
Affiliation(s)
- Cenkai Shen
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Xiao Shi
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Duo Wen
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yuqing Zhang
- School of Data Science, Fudan University, Shanghai, P.R. China
| | - Yuxin Du
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yu Zhang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Ben Ma
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Haitao Tang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Min Yin
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Naisi Huang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Tian Liao
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Ting-Ting Zhang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Chang'e Kong
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Wenjun Wei
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Qinghai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| |
Collapse
|
7
|
Toivanen K, Kilpinen S, Ojala K, Merikoski N, Salmikangas S, Sampo M, Böhling T, Sihto H. PDE3A Is a Highly Expressed Therapy Target in Myxoid Liposarcoma. Cancers (Basel) 2023; 15:5308. [PMID: 38001568 PMCID: PMC10669966 DOI: 10.3390/cancers15225308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
Liposarcomas (LPSs) are a heterogeneous group of malignancies that arise from adipose tissue. Although LPSs are among the most common soft-tissue sarcoma subtypes, precision medicine treatments are not currently available. To discover LPS-subtype-specific therapy targets, we investigated RNA sequenced transcriptomes of 131 clinical LPS tissue samples and compared the data with a transcriptome database that contained 20,218 samples from 95 healthy tissues and 106 cancerous tissue types. The identified genes were referred to the NCATS BioPlanet library with Enrichr to analyze upregulated signaling pathways. PDE3A protein expression was investigated with immunohistochemistry in 181 LPS samples, and PDE3A and SLFN12 mRNA expression with RT-qPCR were investigated in 63 LPS samples. Immunoblotting and cell viability assays were used to study LPS cell lines and their sensitivity to PDE3A modulators. We identified 97, 247, and 37 subtype-specific, highly expressed genes in dedifferentiated, myxoid, and pleomorphic LPS subtypes, respectively. Signaling pathway analysis revealed a highly activated hedgehog signaling pathway in dedifferentiated LPS, phospholipase c mediated cascade and insulin signaling in myxoid LPS, and pathways associated with cell proliferation in pleomorphic LPS. We discovered a strong association between high PDE3A expression and myxoid LPS, particularly in high-grade tumors. Moreover, myxoid LPS samples showed elevated expression levels of SLFN12 mRNA. In addition, PDE3A- and SLFN12-coexpressing LPS cell lines SA4 and GOT3 were sensitive to PDE3A modulators. Our results indicate that PDE3A modulators are promising drugs to treat myxoid LPS. Further studies are required to develop these drugs for clinical use.
Collapse
Affiliation(s)
- Kirsi Toivanen
- Department of Pathology, Helsinki University Hospital, University of Helsinki, 00014 Helsinki, Finland; (N.M.); (S.S.); (T.B.); (H.S.)
| | - Sami Kilpinen
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, 00014 Helsinki, Finland;
| | - Kalle Ojala
- HUS Vatsakeskus, Helsinki University Hospital, PL 340, 00290 Helsinki, Finland;
| | - Nanna Merikoski
- Department of Pathology, Helsinki University Hospital, University of Helsinki, 00014 Helsinki, Finland; (N.M.); (S.S.); (T.B.); (H.S.)
| | - Sami Salmikangas
- Department of Pathology, Helsinki University Hospital, University of Helsinki, 00014 Helsinki, Finland; (N.M.); (S.S.); (T.B.); (H.S.)
| | - Mika Sampo
- Department of Pathology, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital, University of Helsinki, 00029 Helsinki, Finland;
| | - Tom Böhling
- Department of Pathology, Helsinki University Hospital, University of Helsinki, 00014 Helsinki, Finland; (N.M.); (S.S.); (T.B.); (H.S.)
| | - Harri Sihto
- Department of Pathology, Helsinki University Hospital, University of Helsinki, 00014 Helsinki, Finland; (N.M.); (S.S.); (T.B.); (H.S.)
| |
Collapse
|
8
|
Mallik S, Seth S, Si A, Bhadra T, Zhao Z. Optimal ranking and directional signature classification using the integral strategy of multi-objective optimization-based association rule mining of multi-omics data. FRONTIERS IN BIOINFORMATICS 2023; 3:1182176. [PMID: 37576714 PMCID: PMC10415913 DOI: 10.3389/fbinf.2023.1182176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/19/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction: Association rule mining (ARM) is a powerful tool for exploring the informative relationships among multiple items (genes) in any dataset. The main problem of ARM is that it generates many rules containing different rule-informative values, which becomes a challenge for the user to choose the effective rules. In addition, few works have been performed on the integration of multiple biological datasets and variable cutoff values in ARM. Methods: To solve all these problems, in this article, we developed a novel framework MOOVARM (multi-objective optimized variable cutoff-based association rule mining) for multi-omics profiles. Results: In this regard, we identified the positive ideal solution (PIS), which maximized the profit and minimized the loss, and negative ideal solution (NIS), which minimized the profit and maximized the loss for all gene sets (item sets), belonging to each extracted rule. Thereafter, we computed the distance (d +) from PIS and distance (d -) from NIS for each gene set or product. These two distances played an important role in determining the optimized associations among various pairs of genes in the multi-omics dataset. We then globally estimated the relative closeness to PIS for ranking the gene sets. When the relative closeness score of the rule is greater than or equal to the pre-defined threshold value, the rule can be considered a final resultant rule. Moreover, MOOVARM evaluated the relative score of the rule based on the status of all genes instead of individual genes. Conclusions: MOOVARM produced the final rank of the extracted (multi-objective optimized) rules of correlated genes which had better disease classification than the state-of-the-art algorithms on gene signature identification.
Collapse
Affiliation(s)
- Saurav Mallik
- Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, United States
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Soumita Seth
- Department of Computer Science and Engineering, Brainware University, Kolkata, India
- Department of Computer Science and Engineering, Aliah University, Kolkata, India
| | - Amalendu Si
- School of Information Technology, Maulana Abul Kalam Azad University of Technology, Haringhata, India
| | - Tapas Bhadra
- Department of Computer Science and Engineering, Aliah University, Kolkata, India
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
9
|
Malvia S, Chintamani C, Sarin R, Dubey US, Saxena S, Bagadi SAR. ABERRANT EXPRESSION OF COL14A1, CELRS3, and CTHRC1 IN BREAST CANCER СELLS. Exp Oncol 2023; 45:28-43. [PMID: 37417284 DOI: 10.15407/exp-oncology.2023.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND Collagens, which are the major components of the extracellular matrix involved in the regulation of tumor microenvironment, could be differentially expressed in breast cancer (BC) with different transcriptome profiling. AIM To analyze the transcript level expression of COL1A1, COL5A1, COL10A1, COL11A1, COL12A1, COL14A1, CTHRC1, and CELRS3 genes and the clinical relevance of their differential expression in BC. MATERIALS AND METHODS The transcript level expression of the genes was analyzed using the quantitative real-time PCR (qPCR) in tumor tissue of 60 BC patients. RESULTS Overexpression of COL1A1, COL5A1, COL10A1, COL11A1, COL12A1, CTHRC, and CELRS3 anddown-regulated expression of COL14A1 were observed. COL14A1 down-regulation was associated with aggressive, basal, and Her-2/neu BC subtypes (p = 0.031). Overexpression of CELSR3 was found to be associated with the older age of the patients (> 55 years, p = 0.049). Further analysis with the TCGA BC data set has shown a concordance in the differential expression of the above genes. Furthermore, overexpression of CTHRC1 was associated with poor overall survival (OS), particularly with poor prognosis (p = 0.00042) for the luminal BC subtype. On the other hand, CELSR3 overexpression was associated with mucinous tumors and poor prognosis in post-menopausal women. In silicotarget prediction identified several BC-associated miRNAs and members of miR-154, -515, and -10 families to perform a likely regulatory role in the above ECM genes. CONCLUSION The present study shows that the expression of COL14A1 and CTHRC1 may serve as potential biological markers for the detection of basal BC and the prognosis of survival for patients with the luminal subtype of BC.
Collapse
Affiliation(s)
- Shreshtha Malvia
- Tumor Biology Division, ICMR-National Institute of Pathology, New Delhi, 110029, India
| | | | - Ramesh Sarin
- Department of Surgery, Indraprastha Apollo Hospital, New Delhi, 110076, India
| | - Uma S Dubey
- Department of Biological Sciences, Birla Institute of Technology and Sciences, Pilani, Rajasthan, 333031
| | - Sunita Saxena
- Consultant, Department of Health Research, New Delhi, 110001 & Ex-Director National Institute of Pathology-ICMR Safdarjang Hospital Campus
| | | |
Collapse
|
10
|
Cai L, DeBerardinis RJ, Xiao G, Minna JD, Xie Y. Dissecting molecular, pathological, and clinical features associated with tumor neural/neuroendocrine heterogeneity. iScience 2023; 26:106983. [PMID: 37378310 PMCID: PMC10291506 DOI: 10.1016/j.isci.2023.106983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/21/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Lineage plasticity, especially transdifferentiation between neural/neuroendocrine (NE) and non-NE lineage, has been observed in multiple cancer types and linked to increased tumor aggressiveness. However, existing NE/non-NE subtype classifications in various cancer types were established through ad hoc approaches in different studies, making it difficult to align findings across cancer types and extend investigations to new datasets. To address this issue, we developed a generalized strategy to generate quantitative NE scores and a web application to facilitate its implementation. We applied this method to nine datasets covering seven cancer types, including two neural cancers, two neuroendocrine cancers, and three non-NE cancers. Our analysis revealed significant NE inter-tumoral heterogeneity and identified strong associations between NE scores and molecular, histological, and clinical features, including prognosis in different cancer types. These results support the translational utility of NE scores. Overall, our work demonstrated a broadly applicable strategy for determining the NE properties of tumors.
Collapse
Affiliation(s)
- Ling Cai
- Quantitative Biomedical Research Center, Peter O’Donnell School of Public Health, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ralph J. DeBerardinis
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Peter O’Donnell School of Public Health, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - John D. Minna
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yang Xie
- Quantitative Biomedical Research Center, Peter O’Donnell School of Public Health, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
11
|
Salgado CM, Alaggio R, Ciolfi A, Zin A, Diomedi Camassei F, Pedace L, Milano GM, Serra A, Di Giannatale A, Mastronuzzi A, Gianatti A, Bisogno G, Ferrari A, Tartaglia M, Reyes-Múgica M, Locatelli F, Miele E. Pediatric BCOR-Altered Tumors From Soft Tissue/Kidney Display Specific DNA Methylation Profiles. Mod Pathol 2023; 36:100039. [PMID: 36853789 DOI: 10.1016/j.modpat.2022.100039] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 09/27/2022] [Accepted: 10/03/2022] [Indexed: 01/11/2023]
Abstract
In the pediatric population, BCL6-correpresor gene (BCOR)-upregulated tumors include primitive myxoid mesenchymal tumors/undifferentiated sarcomas (PMMTI/UND), clear cell sarcomas of the kidney (CCSK), and high-grade neuroepithelial tumors (HG-NET). We investigated DNA methylation (DNAm) and copy number variation (CNV) profiling in these tumors (N = 34) using an Illumina EPIC BeadChip to better define the potential use of these tools to confirm diagnosis and predict outcomes. Twenty-seven tumors from 25 patients (age range, 0-10 years), showed molecular confirmation of genetic abnormalities as follows: BCOR internal tandem duplication in 14 PMMTI/UND, 8 CCSK, and 3 HG-NET and YWHAE fusions in 2 PMMTI/UND. The remaining 7 cases lacking informative molecular data were analyzed by immunophenotyping and were included in the study as a training cohort, clearly separated from the main study group. These were 4 PMMTI, 1 HG-NET, and 1 CCSK in which poor RNA preservation precluded the confirmation of BCOR rearrangements and 1 CCSK in which no rearrangements were found. DNAm data were compared with those of brain tumor and/or sarcoma classifier. Differentially methylated regions (DMRs) were analyzed in the 3 groups. Twenty-two cases of the 24 molecularly confirmed PMMTI/UND and CCSK and 3 of 6 of those with only immunophenotyping were classified within the methylation class "BCOR-altered sarcoma family" with optimal calibrated scores. PMMTI/UND and CCSK showed similar methylation profiles, whereas thousands of DMRs and significantly enriched pathways were evident between soft tissue/kidney tumors and HG-NET. The CNV analysis showed an overall flat profile in 19 of the 31 evaluable tumors (8/10 CCSK; 9/18 PMMTI/UND; 2/4 HG-NET). The most frequent CNVs were 1q gain and 9p and 10q loss. Follow-up time data were available for 20 patients: ≥2 CNV significantly correlated with a worse overall survival rate. In conclusion, soft tissue and kidney BCOR sarcomas matched with BCOR-altered sarcoma methylation class, whereas those from the brain matched with the central nervous system tumor classifier HG-NET BCOR, supporting the notion that DNAm profiling is an informative diagnostic tool. CNV alterations were associated with a more aggressive clinical behavior.
Collapse
Affiliation(s)
- Claudia M Salgado
- Division of Pathology, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rita Alaggio
- Pathology Unit, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.
| | - Andrea Ciolfi
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Angelica Zin
- Clinica di Oncoematologia Pediatrica Azienda Ospedaliera, Università di Padova, Padova, Italy
| | - Francesca Diomedi Camassei
- Pathology Unit, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Lucia Pedace
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Giuseppe Maria Milano
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Annalisa Serra
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Angela Di Giannatale
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Angela Mastronuzzi
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Andrea Gianatti
- Department of Pathology, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Gianni Bisogno
- Department of Pediatric Hematology-Oncology, Università di Padova, Padova, Italy
| | - Andrea Ferrari
- Pediatric Oncology Unit, Medical Oncology and Hematology Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale Tumori, Milano, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Miguel Reyes-Múgica
- Division of Pathology, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Franco Locatelli
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Evelina Miele
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.
| |
Collapse
|
12
|
Deng Y, Lu L, Liang X, Li J, Zhu D, Huang H, Zhang Y, Zhang X, Chen Y, Liu X, Fu Y. DNA methylation-mediated silencing of Neuronatin promotes hepatocellular carcinoma proliferation through the PI3K-Akt signaling pathway. Life Sci 2023; 312:121266. [PMID: 36473542 DOI: 10.1016/j.lfs.2022.121266] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
AIMS To explore the methylation status, function, and underlying mechanism of the imprinted gene Neuronatin (NNAT) in hepatocellular carcinoma (HCC) progression. MAIN METHODS Immunohistochemistry (IHC) was performed to evaluate the expression of NNAT in HCC samples. Bisulfite genomic sequencing PCR (BSP) was applied to examine the methylation status of the NNAT promoter. In addition, colony formation, 5-Ethynyl-20-deoxyuridine (EdU) assays and subcutaneous xenograft nude models were used to explore the roles of NNAT in HCC cell proliferation. Furthermore, RNA-seq and phospho-specific protein microarray assays were conducted to illustrate the underlying mechanism by which NNAT regulates HCC progression. KEY FINDINGS NNAT was obviously downregulated in HCC tissues, and its expression level was closely associated with tumor growth and patient prognosis. The downregulation of NNAT in HCC was induced by hypermethylation of CpG islands in the promoter region, and hypermethylation was correlated with overall survival of HCC. Moreover, the enforced expression of NNAT significantly inhibited HCC cell proliferation in vitro and in vivo. Transcriptome analysis showed that the alteration of NNAT expression was mainly related to dysregulation of the PI3K-Akt signaling pathway. Finally, phospho-specific antibody microarray detection further revealed that overexpressed NNAT can increase the phosphorylation levels of LKB1, Met, and elF4E and decrease the phosphorylation levels of PTEN, which are all involved in the PI3K-Akt signaling pathway. SIGNIFICANCE Our research provides new insights into the epigenetic regulation of imprinted genes in tumorigenesis and implies that the imprinted gene NNAT may act as a prognostic biomarker and tumor suppressor in HCC.
Collapse
Affiliation(s)
- Yalan Deng
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Liqing Lu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xujun Liang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Jingzhi Li
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Obstetrics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Dandan Zhu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Huichao Huang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Department of Infectious Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Ye Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiangqian Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiaojin Liu
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Ying Fu
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
13
|
Ren J, Zhou X, Guo W, Feng K, Huang T, Cai YD. Identification of Methylation Signatures and Rules for Sarcoma Subtypes by Machine Learning Methods. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5297235. [PMID: 36619306 PMCID: PMC9812612 DOI: 10.1155/2022/5297235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/28/2022] [Accepted: 12/08/2022] [Indexed: 12/31/2022]
Abstract
Sarcoma, the second common type of solid tumor in children and adolescents, has a wide variety of subtypes that are often not properly diagnosed at an early stage, leading to late metastases and causing serious loss of life and property to patients and families. It exhibits a high degree of heterogeneity at the cellular, molecular, and epigenetic levels, where DNA methylation has been proposed to play a role in the diagnosis of sarcoma subtypes. Thus, this study is aimed at finding potential biomarkers at the DNA methylation level to distinguish different sarcoma subtypes. A machine learning process was designed to analyse sarcoma samples, each of which was represented by lots of methylation sites. Irrelevant sites were removed using the Boruta method, and remaining sites related to the target variables were kept for further analyses. Afterward, three feature ranking methods (LASSO, LightGBM, and MCFS) were adopted to rank these features, and six classification models were constructed by combining incremental feature selection and two classification algorithms (decision tree and random forest). Among these models, the performance of RF model was higher than that of DT model under all three ranking conditions. The specific expression of genes obtained from the annotation of highly correlated methylation site features, such as PRKAR1B, INPP5A, and GLI3, was proven to be associated with sarcoma by publications. Moreover, the quantitative rules obtained by decision tree algorithm helped us to understand the essential differences between various sarcoma types and classify sarcoma subtypes, providing a new means of clinical identification and determining new therapeutic targets.
Collapse
Affiliation(s)
- Jingxin Ren
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - XianChao Zhou
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200030, China
| | - KaiYan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou 510507, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|
14
|
The Intricate Interplay between the ZNF217 Oncogene and Epigenetic Processes Shapes Tumor Progression. Cancers (Basel) 2022; 14:cancers14246043. [PMID: 36551531 PMCID: PMC9776013 DOI: 10.3390/cancers14246043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
The oncogenic transcription factor ZNF217 orchestrates several molecular signaling networks to reprogram integrated circuits governing hallmark capabilities within cancer cells. High levels of ZNF217 expression provide advantages to a specific subset of cancer cells to reprogram tumor progression, drug resistance and cancer cell plasticity. ZNF217 expression level, thus, provides a powerful biomarker of poor prognosis and a predictive biomarker for anticancer therapies. Cancer epigenetic mechanisms are well known to support the acquisition of hallmark characteristics during oncogenesis. However, the complex interactions between ZNF217 and epigenetic processes have been poorly appreciated. Deregulated DNA methylation status at ZNF217 locus or an intricate cross-talk between ZNF217 and noncoding RNA networks could explain aberrant ZNF217 expression levels in a cancer cell context. On the other hand, the ZNF217 protein controls gene expression signatures and molecular signaling for tumor progression by tuning DNA methylation status at key promoters by interfering with noncoding RNAs or by refining the epitranscriptome. Altogether, this review focuses on the recent advances in the understanding of ZNF217 collaboration with epigenetics processes to orchestrate oncogenesis. We also discuss the exciting burgeoning translational medicine and candidate therapeutic strategies emerging from those recent findings connecting ZNF217 to epigenetic deregulation in cancer.
Collapse
|
15
|
Fuller AM, DeVine A, Murazzi I, Mason NJ, Weber K, Eisinger-Mathason TSK. Comparative oncology reveals DNMT3B as a molecular vulnerability in undifferentiated pleomorphic sarcoma. Cell Oncol (Dordr) 2022; 45:1277-1295. [PMID: 36181640 PMCID: PMC9772002 DOI: 10.1007/s13402-022-00717-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Undifferentiated pleomorphic sarcoma (UPS), an aggressive subtype of soft-tissue sarcoma (STS), is exceedingly rare in humans and lacks effective, well-tolerated therapies. In contrast, STS are relatively common in canine companion animals. Thus, incorporation of veterinary patients into studies of UPS offers an exciting opportunity to develop novel therapeutic strategies for this rare human disease. Genome-wide studies have demonstrated that UPS is characterized by aberrant patterns of DNA methylation. However, the mechanisms and impact of this epigenetic modification on UPS biology and clinical behavior are poorly understood. METHODS DNA methylation in mammalian cells is catalyzed by the canonical DNA methyltransferases DNMT1, DNMT3A and DNMT3B. Therefore, we leveraged cell lines and tissue specimens from human and canine patients, together with an orthotopic murine model, to probe the functional and clinical significance of DNMTs in UPS. RESULTS We found that the DNA methyltransferase DNMT3B is overexpressed in UPS relative to normal mesenchymal tissues and is associated with a poor prognosis. Consistent with these findings, genetic DNMT3B depletion strongly inhibited UPS cell proliferation and tumor progression. However, existing hypomethylating agents, including the clinically approved drug 5-aza-2'-deoxycytidine (DAC) and the DNMT3B-inhibiting tool compound nanaomycin A, were ineffective in UPS due to cellular uptake and toxicity issues. CONCLUSIONS DNMT3B represents a promising molecular susceptibility in UPS, but further development of DNMT3B-targeting strategies for these patients is required.
Collapse
Affiliation(s)
- Ashley M Fuller
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ann DeVine
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ileana Murazzi
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicola J Mason
- Department of Clinical Sciences and Advanced Medicine, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristy Weber
- Penn Sarcoma Program, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - T S Karin Eisinger-Mathason
- Abramson Family Cancer Research Institute, Department of Pathology and Laboratory Medicine, Penn Sarcoma Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
16
|
He X, Zhang W, Fu W, Liu X, Yang P, Wang J, Zhu M, Li S, Zhang W, Zhang X, Dong G, Yan C, Zhao Y, Zeng Z, Jing H. The prognostic value of RASGEF1A RNA expression and DNA methylation in cytogenetically normal acute myeloid leukemia. Cancer Biomark 2022; 36:103-116. [PMID: 36404533 DOI: 10.3233/cbm-210407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a significantly heterogeneous malignancy of the blood. Cytogenetic abnormalities are crucial for the prognosis of AML. However, since more than half of patients with AML are cytogenetically normal AML (CN-AML), predictive prognostic indicators need to be further refined. In recent years, gene abnormalities are considered to be strong prognostic factors of CN-AML, already having clinical significance for treatment. In addition, the relationship of methylation in some genes and AML prognosis predicting has been discovered. RASGEF1A is a guanine nucleotide exchange factors of Ras and widely expressed in brain tissue, bone marrow and 17 other tissues. RASGEF1A has been reported to be associated with a variety of malignant tumors, examples include Hirschsprung disease, renal cell carcinoma, breast cancer, diffuse large B cell lymphoma, intrahepatic cholangiocarcinoma and so on [1, 2]. However, the relationship between the RASGEF1A gene and CN-AML has not been reported. METHODS By integrating the Cancer Genome Atlas (TCGA) database 75 patients with CN-AML and 240 Gene Expression Omnibus (GEO) database CN-AML samples, we examined the association between RASGEF1A's RNA expression level and DNA methylation of and AML patients' prognosis. Then, we investigated the RASGEF1A RNA expression and DNA methylation's prognostic value in 77 patients with AML after allogeneic hematopoietic stem cell transplantation (Allo-HSCT) as well as 101 AML patients after chemotherapy respectively. We investigated the association between sensitivity to Crenolanib and expression level of RASGED1A in patients by integrating 191 CN-AML patients from BeatAML dadataset. We integrated the expression and methylation of RASGEF1A to predict the CN-AML patients' prognosis and investigated the relationship between prognostic of AML patients with different risk classification and expression levels or methylation levels of RASGEF1A. RESULTS We found that RASGEF1A gene high expression group predicted poorer event-free survival (EFS) (P< 0.0001) as well as overall survival (OS) (P< 0.0001) in CN-AML samples, and the identical results were found in AML patients receiving chemotherapy (P< 0.0001) and Allo-HSCT (P< 0.0001). RASGEF1A RNA expression level is an CN-AML patients' independent prognostic factor (EFS: HR = 5.5534, 95% CI: 1.2982-23.756, P= 0.0208; OS: HR = 5.3615, 95% CI: 1.1014-26.099, P= 0.0376). The IC50 (half maximal inhibitory concentration) of Crenolanib of CN-AML samples with RASGEF1A high expression level is lower. In addition, patients with high RASGEF1A methylation level had significant favorable prognosis (EPS: P< 0.0001, OS: P< 0.0001). Furthermore, the integrative analysis of expression and methylation of RASGEF1A could classify CN-AML patients into subgroups with different prognosis (EFS: P= 0.034, OS: P= 0.0024). Expression levels or methylation levels of RASGEF1A help to improve risk classification of 2010 European Leukemia Net. CONCLUSION Higher RASGEF1A RNA expression and lower DNA methylation predicts CN-AML patients' poorer prognosis. The RASGEF1A high expression level from patients with CN-AML have better sensitivity to Crenolanib. The integrative analysis of RASGEF1A RNA expression and DNA methylation can provide a more accurate classification for prognosis. Lower RASGEF1A expression is a favorable prognostic factor for AML patients receiving chemotherapy or Allo-HSCT. 2010 European Leukemia Net's risk classification can be improved by RASGEF1A expression levels or methylation levels.
Collapse
Affiliation(s)
- Xue He
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Pathology, Capital Medical University, Beijing, China.,Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weilong Zhang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China.,Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wei Fu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China.,Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaoni Liu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Ping Yang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Jing Wang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Mingxia Zhu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| | - Shaoxiang Li
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Pathology, Capital Medical University, Beijing, China
| | - Wei Zhang
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Pathology, Capital Medical University, Beijing, China
| | - Xiuru Zhang
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Pathology, Capital Medical University, Beijing, China
| | - Gehong Dong
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Pathology, Capital Medical University, Beijing, China
| | | | - Yali Zhao
- General Practice Medicine, The First People's Hospital of Huzhou, Huzhou, Zhejiang, China
| | - Zhiping Zeng
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Hongmei Jing
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, China
| |
Collapse
|
17
|
In Silico Identification of Key Genes and Immune Infiltration Characteristics in Epicardial Adipose Tissue from Patients with Coronary Artery Disease. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5610317. [PMID: 36345357 PMCID: PMC9637040 DOI: 10.1155/2022/5610317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/04/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022]
Abstract
Background The present study is aimed at identifying the differentially expressed genes (DEGs) and relevant biological processes and pathways associated with epicardial adipose tissue (EAT) from patients with coronary artery disease (CAD). We also explored potential biomarkers using two machine-learning algorithms and calculated the immune cell infiltration in EAT. Materials and Methods Three datasets (GSE120774, GSE64554, and GSE24425) were obtained from the Gene Expression Omnibus (GEO) database. The GSE120774 dataset was used to evaluate DEGs between EAT of CAD patients and the control group. Functional enrichment analyses were conducted to study associated biological functions and mechanisms using the Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO), and Gene Set Enrichment Analysis (GSEA). After this, the least absolute shrinkage and selection operator (LASSO) and support vector machine recursive feature elimination (SVM-RFE) were performed to identify the feature genes related to CAD. The expression level of the feature genes was validated in GSE64554 and GSE24425. Finally, we calculated the immune cell infiltration and evaluated the correlation between the feature genes and immune cells using CIBERSORT. Results We identified a total of 130 upregulated and 107 downregulated genes in GSE120774. Functional enrichment analysis revealed that DEGs are associated with several pathways, including the calcium signaling pathway, complement and coagulation cascades, ferroptosis, fluid shear stress and atherosclerosis, lipid and atherosclerosis, and regulation of lipolysis in adipocytes. TCF21, CDH19, XG, and NNAT were identified as feature genes and validated in the GSE64554 and GSE24425 datasets. Immune cell infiltration analysis showed plasma cells are significantly more numerous in EAT than in the control group (p = 0.001), whereas macrophage M0 (p = 0.024) and resting mast cells (p = 0.036) were significantly less numerous. TCF21, CDH19, XG, and NNAT were correlated with immune cells, including plasma cells, M0 macrophages, and resting mast cells. Conclusion TCF21, CDH19, XG, and NNAT might serve as feature genes for CAD, providing new insights for future research on the pathogenesis of cardiovascular diseases.
Collapse
|
18
|
Dermawan JK, Vanoli F, Herviou L, Sung YS, Zhang L, Singer S, Tap WD, Benayed R, Bale TA, Benhamida JK, Dickson BC, Antonescu CR. Comprehensive genomic profiling of EWSR1/FUS::CREB translocation-associated tumors uncovers prognostically significant recurrent genetic alterations and methylation-transcriptional correlates. Mod Pathol 2022; 35:1055-1065. [PMID: 35347249 PMCID: PMC9329182 DOI: 10.1038/s41379-022-01023-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/22/2022]
Abstract
To elucidate the mechanisms underlying the divergent clinicopathologic spectrum of EWSR1/FUS::CREB translocation-associated tumors, we performed a comprehensive genomic analysis of fusion transcript variants, recurrent genetic alterations (mutations, copy number alterations), gene expression, and methylation profiles across a large cohort of tumor types. The distribution of the EWSR1/FUS fusion partners-ATF1, CREB1, and CREM-and exon involvement was significantly different across different tumor types. Our targeted sequencing showed that secondary genetic events are associated with tumor type rather than fusion type. Of the 39 cases that underwent targeted NGS testing, 18 (46%) had secondary OncoKB mutations or copy number alterations (29 secondary genetic events in total), of which 15 (52%) were recurrent. Secondary recurrent, but mutually exclusive, TERT promoter and CDKN2A mutations were identified only in clear cell sarcoma (CCS) and associated with worse overall survival. CDKN2A/B homozygous deletions were recurrent in angiomatoid fibrous histiocytoma (AFH) and restricted to metastatic cases. mRNA upregulation of MITF, CDH19, PARVB, and PFKP was found in CCS, compared to AFH, and correlated with a hypomethylated profile. In contrast, S100A4 and XAF1 were differentially upregulated and hypomethylated in AFH but not CCS. Unsupervised clustering of methylation profiles revealed that CREB family translocation-associated tumors form neighboring but tight, distinct clusters. A sarcoma methylation classifier was able to accurately match 100% of CCS cases to the correct methylation class; however, it was suboptimal when applied to other histologies. In conclusion, our comprehensive genomic profiling of EWSR1/FUS::CREB translocation-associated tumors uncovered mostly histotype, rather than fusion-type associated correlations in transcript variants, prognostically significant secondary genetic alterations, and gene expression and methylation patterns.
Collapse
Affiliation(s)
| | - Fabio Vanoli
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laurie Herviou
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yun-Shao Sung
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lei Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samuel Singer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William D. Tap
- Department of Medicine, Sarcoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tejus A. Bale
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jamal K. Benhamida
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brendan C. Dickson
- Department of Pathology and Laboratory Medicine, Sinai Health System, Toronto, Ontario, Canada
| | | |
Collapse
|
19
|
Li A, Mueller A, English B, Arena A, Vera D, Kane AE, Sinclair DA. Novel feature selection methods for construction of accurate epigenetic clocks. PLoS Comput Biol 2022; 18:e1009938. [PMID: 35984867 PMCID: PMC9432708 DOI: 10.1371/journal.pcbi.1009938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/31/2022] [Accepted: 07/11/2022] [Indexed: 11/22/2022] Open
Abstract
Epigenetic clocks allow us to accurately predict the age and future health of individuals based on the methylation status of specific CpG sites in the genome and are a powerful tool to measure the effectiveness of longevity interventions. There is a growing need for methods to efficiently construct epigenetic clocks. The most common approach is to create clocks using elastic net regression modelling of all measured CpG sites, without first identifying specific features or CpGs of interest. The addition of feature selection approaches provides the opportunity to optimise the identification of predictive CpG sites. Here, we apply novel feature selection methods and combinatorial approaches including newly adapted neural networks, genetic algorithms, and 'chained' combinations. Human whole blood methylation data of ~470,000 CpGs was used to develop clocks that predict age with R2 correlation scores of greater than 0.73, the most predictive of which uses 35 CpG sites for a R2 correlation score of 0.87. The five most frequent sites across all clocks were modelled to build a clock with a R2 correlation score of 0.83. These two clocks are validated on two external datasets where they maintain excellent predictive accuracy. When compared with three published epigenetic clocks (Hannum, Horvath, Weidner) also applied to these validation datasets, our clocks outperformed all three models. We identified gene regulatory regions associated with selected CpGs as possible targets for future aging studies. Thus, our feature selection algorithms build accurate, generalizable clocks with a low number of CpG sites, providing important tools for the field.
Collapse
Affiliation(s)
- Adam Li
- Blavatnik Institute, Dept. of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, Massachusetts, United States of America
| | - Amber Mueller
- Blavatnik Institute, Dept. of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, Massachusetts, United States of America
| | - Brad English
- Blavatnik Institute, Dept. of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anthony Arena
- Blavatnik Institute, Dept. of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel Vera
- Blavatnik Institute, Dept. of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alice E. Kane
- Blavatnik Institute, Dept. of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, Massachusetts, United States of America
| | - David A. Sinclair
- Blavatnik Institute, Dept. of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
20
|
Li Y, Sena Lopes J, Fuster PC, Rivera RM. Spontaneous and ART-induced large offspring syndrome: similarities and differences in DNA methylome. Epigenetics 2022; 17:1477-1496. [PMID: 35466858 PMCID: PMC9586674 DOI: 10.1080/15592294.2022.2067938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Large/abnormal offspring syndrome (LOS/AOS) is a congenital overgrowth syndrome reported in ruminants produced by assisted reproduction (ART-LOS) which exhibit global disruption of the epigenome and transcriptome. LOS/AOS shares phenotypes and epigenotypes with the human congenital overgrowth condition Beckwith-Wiedemann syndrome. We have reported that LOS occurs spontaneously (SLOS); however, to date, no study has been conducted to determine if SLOS has the same methylome epimutations as ART-LOS. In this study, we performed whole-genome bisulphite sequencing to examine global DNA methylation in bovine SLOS and ART-LOS tissues. We observed unique patterns of global distribution of differentially methylated regions (DMRs) over different genomic contexts, such as promoters, CpG Islands, shores and shelves, as well as at repetitive sequences. In addition, we included data from two previous LOS studies to identify shared vulnerable genomic loci in LOS. Overall, we identified 320 genomic loci in LOS that have alterations in DNA methylation when compared to controls. Specifically, there are 25 highly vulnerable loci that could potentially serve as molecular markers for the diagnosis of LOS, including at the promoters of DMRT2 and TBX18, at the imprinted gene bodies of IGF2R, PRDM8, and BLCAP/NNAT, and at multiple CpG Islands. We also observed tissue-specific DNA methylation patterns between muscle and blood, and conservation of ART-induced DNA methylation changes between muscle and blood. We conclude that as ART-LOS, SLOS is an epigenetic condition. In addition, SLOS and ART-LOS share similarities in methylome epimutations.
Collapse
Affiliation(s)
- Yahan Li
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Jordana Sena Lopes
- Physiology Department. International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Universidad de Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB), Murcia, Spain.,Mediterranean Institute for Agriculture, Environment and Development (MED), University of Évora, Portugal
| | - Pilar Coy Fuster
- Physiology Department. International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Universidad de Murcia, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB), Murcia, Spain
| | | |
Collapse
|
21
|
Bertucci F, Niziers V, de Nonneville A, Finetti P, Mescam L, Mir O, Italiano A, Le Cesne A, Blay JY, Ceccarelli M, Bedognetti D, Birnbaum D, Mamessier E. Immunologic constant of rejection signature is prognostic in soft-tissue sarcoma and refines the CINSARC signature. J Immunother Cancer 2022; 10:jitc-2021-003687. [PMID: 35017155 PMCID: PMC8753443 DOI: 10.1136/jitc-2021-003687] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Soft-tissue sarcomas (STSs) are heterogeneous and aggressive tumors, with high metastatic risk. The immunologic constant of rejection (ICR) 20-gene signature is a signature of cytotoxic immune response. We hypothesized that ICR might improve the prognostic assessment of early-stage STS. METHODS We retrospectively applied ICR to 1455 non-metastatic STS and searched for correlations between ICR classes and clinicopathological and biological variables, including metastasis-free survival (MFS). RESULTS Thirty-four per cent of tumors were classified as ICR1, 27% ICR2, 24% ICR3, and 15% ICR4. These classes were associated with patients' age, pathological type, and tumor depth, and an enrichment from ICR1 to ICR4 of quantitative/qualitative scores of immune response. ICR1 class was associated with a 59% increased risk of metastatic relapse when compared with ICR2-4 class. In multivariate analysis, ICR classification remained associated with MFS, as well as pathological type and Complexity Index in Sarcomas (CINSARC) classification, suggesting independent prognostic value. A prognostic clinicogenomic model, including the three variables, was built in a learning set (n=339) and validated in an independent set (n=339), showing greater prognostic precision than each variable alone or in doublet. Finally, connectivity mapping analysis identified drug classes potentially able to reverse the expression profile of poor-prognosis tumors, such as chemotherapy and targeted therapies. CONCLUSION ICR signature is independently associated with postoperative MFS in early-stage STS, independently from other prognostic features, including CINSARC. We built a robust prognostic clinicogenomic model integrating ICR, CINSARC, and pathological type, and suggested differential vulnerability of each prognostic group to different systemic therapies.
Collapse
Affiliation(s)
- Francois Bertucci
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France .,Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France.,French Sarcoma Group, Lyon, France
| | - Vincent Niziers
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France.,Department of Surgery, Institut Paoli-Calmettes, Marseille, France
| | - Alexandre de Nonneville
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France.,Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Pascal Finetti
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Léna Mescam
- French Sarcoma Group, Lyon, France.,Department of Pathology, Institut Paoli-Calmettes, Marseille, France
| | - Olivier Mir
- French Sarcoma Group, Lyon, France.,Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Antoine Italiano
- French Sarcoma Group, Lyon, France.,Department of Medical Oncology, Institut Bergonie, Bordeaux, France
| | - Axel Le Cesne
- French Sarcoma Group, Lyon, France.,Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Jean-Yves Blay
- French Sarcoma Group, Lyon, France.,Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | - Michele Ceccarelli
- DIETI, University of Naples Federico II Faculty of Engineering, Naples, Italy
| | - Davide Bedognetti
- Cancer Research, Sidra Medicine, Doha, Qatar.,Department of Internal Medicine and Medical Specialties, University of Genova, Genova, Italy
| | - Daniel Birnbaum
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Emilie Mamessier
- Laboratory of Predictive Oncology, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Aix-Marseille Université, INSERM UMR1068, CNRS UMR725, Marseille, France
| |
Collapse
|
22
|
Papanicolau-Sengos A, Aldape K. DNA Methylation Profiling: An Emerging Paradigm for Cancer Diagnosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 17:295-321. [PMID: 34736341 DOI: 10.1146/annurev-pathol-042220-022304] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Histomorphology has been a mainstay of cancer diagnosis in anatomic pathology for many years. DNA methylation profiling is an additional emerging tool that will serve as an adjunct to increase accuracy of pathological diagnosis. Genome-wide interrogation of DNA methylation signatures, in conjunction with machine learning methods, has allowed for the creation of clinical-grade classifiers, most prominently in central nervous system and soft tissue tumors. Tumor DNA methylation profiling has led to the identification of new entities and the consolidation of morphologically disparate cancers into biologically coherent entities, and it will progressively become mainstream in the future. In addition, DNA methylation patterns in circulating tumor DNA hold great promise for minimally invasive cancer detection and classification. Despite practical challenges that accompany any new technology, methylation profiling is here to stay and will become increasingly utilized as a cancer diagnostic tool across a range of tumor types. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 17 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
| | - Kenneth Aldape
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland 20892, USA; ,
| |
Collapse
|
23
|
Hasan NM, Sharma A, Ruzgar NM, Deshpande H, Olino K, Khan S, Ahuja N. Epigenetic signatures differentiate uterine and soft tissue leiomyosarcoma. Oncotarget 2021; 12:1566-1579. [PMID: 34381562 PMCID: PMC8351604 DOI: 10.18632/oncotarget.28032] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/13/2021] [Indexed: 01/12/2023] Open
Abstract
Leiomyosarcomas (LMS) are diverse, rare, and aggressive mesenchymal soft tissue sarcomas. Epigenetic alterations influence multiple aspects of cancer, however epigenetic profiling of LMS has been limited. The goal of this study was to delineate the molecular landscape of LMS for subtype-specific differences (uterine LMS (ULMS) vs soft tissue LMS (STLMS)) based on integrated analysis of DNA methylation and gene expression to identify potential targets for therapeutic intervention and diagnosis. We identified differentially methylated and differentially expressed genes associated with ULMS and STLMS using DNA methylation and RNA-seq data from primary tumors. Two main clusters were identified through unsupervised hierarchical clustering: ULMS-enriched cluster and STLMS-enriched cluster. The integrated analysis demonstrated 34 genes associated with hypermethylation of the promoter CpG islands and downregulation of gene expression in ULMS or STLMS. In summary, these results indicate that differential DNA methylation and gene expression patterns are associated with ULMS and STLMS. Further studies are needed to delineate the contribution of epigenetic regulation to LMS subtype-specific gene expression and determine the roles of the differentially methylated and differentially expressed genes as potential therapeutic targets or biomarkers.
Collapse
Affiliation(s)
- Nesrin M. Hasan
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Anup Sharma
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | | | - Hari Deshpande
- Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Kelly Olino
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Sajid Khan
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Surgery, Section of Hepatopancreatobiliary and Mixed Tumors, Yale University School of Medicine, New Haven, CT, USA
| | - Nita Ahuja
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
24
|
Bellanger A, Le DT, Vendrell J, Wierinckx A, Pongor LS, Solassol J, Lachuer J, Clezardin P, Győrffy B, Cohen PA. Exploring the Significance of the Exon 4-Skipping Isoform of the ZNF217 Oncogene in Breast Cancer. Front Oncol 2021; 11:647269. [PMID: 34277402 PMCID: PMC8283766 DOI: 10.3389/fonc.2021.647269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/31/2021] [Indexed: 12/30/2022] Open
Abstract
Oncogene alternative splicing events can create distinct functional transcripts that offer new candidate prognostic biomarkers for breast cancer. ZNF217 is a well-established oncogene but its exon 4-skipping isoform (ZNF217-ΔE4) has never been investigated in terms of clinical or biological relevance. Using in silico RNA-seq and RT-qPCR analyses, we demonstrated for the first time the existence of ZNF217-ΔE4 transcripts in primary breast tumors, and a positive correlation between ZNF217-ΔE4 mRNA levels and those of the wild-type oncogene (ZNF217-WT). A pilot retrospective analysis revealed that, in the Luminal subclass, the combination of the two ZNF217 variants (the ZNF217-ΔE4-WT gene-expression signature) provided more information than the mRNA expression levels of each isoform alone. Ectopic overexpression of ZNF217-ΔE4 in breast cancer cells promoted an aggressive phenotype and an increase in ZNF217-WT expression levels that was inversely correlated with DNA methylation of the ZNF217 gene. This study provides new insights into the possible role of the ZNF217-ΔE4 splice variant in breast cancer and suggests a close interplay between the ZNF217-WT and ZNF217-ΔE4 isoforms. Our data suggest that a dual signature combining the expression levels of these two isoforms may serve as a novel prognostic biomarker allowing better stratification of breast cancers with good prognosis and aiding clinicians in therapeutic decisions.
Collapse
Affiliation(s)
- Aurélie Bellanger
- Université Lyon 1, Lyon, France.,CRCL-Centre de Recherche en Cancérologie de Lyon-Inserm U1052-CNRS U5286, Lyon, France
| | - Diep T Le
- Université Lyon 1, Lyon, France.,INSERM, UMR1033 LYOS, Lyon, France
| | - Julie Vendrell
- Département de Pathologie et Oncobiologie, Laboratoire de Biologie des Tumeurs Solides, CHU Montpellier, Univ. Montpellier, Montpellier, France
| | - Anne Wierinckx
- Université Lyon 1, Lyon, France.,CRCL-Centre de Recherche en Cancérologie de Lyon-Inserm U1052-CNRS U5286, Lyon, France.,ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7, Lyon, France
| | - Lőrinc S Pongor
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary.,TTK "TermészetTudományi Kutatóközpont" Momentum Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary
| | - Jérôme Solassol
- Département de Pathologie et Oncobiologie, Laboratoire de Biologie des Tumeurs Solides, CHU Montpellier, Univ. Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Univ. Montpellier, Montpellier, France
| | - Joël Lachuer
- Université Lyon 1, Lyon, France.,CRCL-Centre de Recherche en Cancérologie de Lyon-Inserm U1052-CNRS U5286, Lyon, France.,ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7, Lyon, France
| | | | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary.,TTK "TermészetTudományi Kutatóközpont" Momentum Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary
| | - Pascale A Cohen
- Université Lyon 1, Lyon, France.,CRCL-Centre de Recherche en Cancérologie de Lyon-Inserm U1052-CNRS U5286, Lyon, France.,INSERM, UMR1033 LYOS, Lyon, France.,ProfileXpert, SFR-Est, CNRS UMR-S3453, INSERM US7, Lyon, France
| |
Collapse
|
25
|
Pieper W, Ignatov A, Kalinski T, Haybaeck J, Czapiewski P, Nass N. The predictive potential of Neuronatin for neoadjuvant chemotherapy of breast cancer. Cancer Biomark 2021; 32:161-173. [PMID: 34092612 DOI: 10.3233/cbm-203127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Neuronatin (NNAT) determined by immunohistochemistry is a negative prognostic biomarker for breast cancer, independent of the major clinicopathological markers. OBJECTIVE Here, we investigated whether NNAT is also a predictive biomarker for pathological remission after neoadjuvant chemotherapy. METHODS One hundred and four breast cancer patients, treated with systemic neoadjuvant chemotherapy were included in this retrospective study. NNAT was detected in formaldehyde fixed, paraffin embedded primary cancer tissue by immunohistochemistry and an immuno-reactive score (IRS) determined. Pathological remission was scored according to Sinn and by evaluation of cytopathic effects. NNAT-IRS was correlated with clinicopathological parameters as well as relapse free and overall survival and for pathological remission after neoadjuvant therapy. RESULTS NNAT IRS was an independent prognostic marker for relapse free and overall survival and the time from diagnosis to the "tumor-free" state. NNAT IRS was associated with Luminal-A tumors and correlated slightly negative with age and lymph-node metastasis. There was no significant correlation of NNAT-IRS with Sinn's remission score, but with cytopathic effects of chemotherapy. CONCLUSIONS We confirmed the prognostic impact of NNAT-IRS in an independent cohort of neoadjuvantly treated patients. Additionally, a correlation with a score for pathological remission under systemic neoadjuvant chemotherapy for breast cancer was found.
Collapse
Affiliation(s)
- Willi Pieper
- Department of Pathology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Atanas Ignatov
- Department of Obstetrics and Gynecology, Otto von Guericke University, Magdeburg, Germany.,Department of Gynecology and Obstetrics, University Medical Center, Regensburg, Germany
| | - Thomas Kalinski
- Department of Pathology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Johannes Haybaeck
- Department of Pathology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.,Department of Gynecology and Obstetrics, University Medical Center, Regensburg, Germany.,Department of Pathology, Diagnostic and Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz, Austria
| | - Piotr Czapiewski
- Department of Pathology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.,Department of Pathology, Dessau Medical Center, Dessau, Germany.,Department of Pathology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Norbert Nass
- Department of Pathology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.,Department for Internal Medicine I, Dessau Medical Center, Dessau, Germany.,Department of Pathology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
26
|
Lin LL, Liu ZZ, Tian JZ, Zhang X, Zhang Y, Yang M, Zhong HC, Fang W, Wei RX, Hu C. Integrated Analysis of Nine Prognostic RNA-Binding Proteins in Soft Tissue Sarcoma. Front Oncol 2021; 11:633024. [PMID: 34026613 PMCID: PMC8138553 DOI: 10.3389/fonc.2021.633024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/10/2021] [Indexed: 12/24/2022] Open
Abstract
RNA-binding proteins (RBPs) have been shown to be dysregulated in cancer transcription and translation, but few studies have investigated their mechanism of action in soft tissue sarcoma (STS). Here, The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases were used to identify differentially expressed RBPs in STS and normal tissues. Through a series of biological information analyses, 329 differentially expressed RBPs were identified. Functional enrichment analysis showed that differentially expressed RBPs were mainly involved in RNA transport, RNA splicing, mRNA monitoring pathways, ribosome biogenesis and translation regulation. Through Cox regression analyses, 9 RBPs (BYSL, IGF2BP3, DNMT3B, TERT, CD3EAP, SRSF12, TLR7, TRIM21 and MEX3A) were all up-regulated in STS as prognosis-related genes, and a prognostic model was established. The model calculated a risk score based on the expression of 9 hub RBPs. The risk score could be used for risk stratification of patients and had a high prognostic value based on the receiver operating characteristic (ROC) curve. We also established a nomogram containing risk scores and 9 key RBPs to predict the 1-year, 3-year, and 5-year survival rates of patients in STS. Afterwards, methylation analysis showed significant changes in the methylation degree of BYSL, CD3EAP and MEX2A. Furthermore, the expression of 9 hub RBPs was closely related to immune infiltration rather than tumor purity. Based on the above studies, these findings may provide new insights into the pathogenesis of STS and will provide candidate biomarkers for the prognosis of STS.
Collapse
Affiliation(s)
- Lu-Lu Lin
- Department of Pathology and Pathophysiology, School of Basic Medicine, Wuhan University, Wuhan, China
| | - Zi-Zhen Liu
- The Third Clinical School, Hubei University of Medicine, Shiyan, China
| | - Jing-Zhuo Tian
- The Third Clinical School, Hubei University of Medicine, Shiyan, China
| | - Xiao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Zhang
- The Third Clinical School, Hubei University of Medicine, Shiyan, China
| | - Min Yang
- Department of Spine and Orthopedic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hou-Cheng Zhong
- Department of Spine and Orthopedic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Fang
- Hubei University of Medicine, Shiyan, China
| | - Ren-Xiong Wei
- Department of Spine and Orthopedic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chao Hu
- Department of Spine and Orthopedic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
27
|
Ren C, Tang X, Lan H. Comprehensive analysis based on DNA methylation and RNA-seq reveals hypermethylation of the up-regulated WT1 gene with potential mechanisms in PAM50 subtypes of breast cancer. PeerJ 2021; 9:e11377. [PMID: 33987034 PMCID: PMC8103922 DOI: 10.7717/peerj.11377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 04/08/2021] [Indexed: 11/20/2022] Open
Abstract
Background Breast cancer (BC), one of the most widespread cancers worldwide, caused the deaths of more than 600,000 women in 2018, accounting for about 15% of all cancer-associated deaths in women that year. In this study, we aimed to discover potential prognostic biomarkers and explore their molecular mechanisms in different BC subtypes using DNA methylation and RNA-seq. Methods We downloaded the DNA methylation datasets and the RNA expression profiles of primary tissues of the four BC molecular subtypes (luminal A, luminal B, basal-like, and HER2-enriched), as well as the survival information from The Cancer Genome Atlas (TCGA). The highly expressed and hypermethylated genes across all the four subtypes were screened. We examined the methylation sites and the downstream co-expressed genes of the selected genes and validated their prognostic value using a different dataset (GSE20685). For selected transcription factors, the downstream genes were predicted based on the Gene Transcription Regulation Database (GTRD). The tumor microenvironment was also evaluated based on the TCGA dataset. Results We found that Wilms tumor gene 1 (WT1), a transcription factor, was highly expressed and hypermethylated in all the four BC subtypes. All the WT1 methylation sites exhibited hypermethylation. The methylation levels of the TSS200 and 1stExon regions were negatively correlated with WT1 expression in two BC subtypes, while that of the gene body region was positively associated with WT1 expression in three BC subtypes. Patients with low WT1 expression had better overall survival (OS). Five genes including COL11A1, GFAP, FGF5, CD300LG, and IGFL2 were predicted as the downstream genes of WT1. Those five genes were dysregulated in the four BC subtypes. Patients with a favorable 6-gene signature (low expression of WT1 and its five predicted downstream genes) exhibited better OS than that with an unfavorable 6-gene signature. We also found a correlation between WT1 and tamoxifen using STITCH. Higher infiltration rates of CD8 T cells, plasma cells, and monocytes were found in the lower quartile WT1 group and the favorable 6-gene signature group. In conclusion, we demonstrated that WT1 is hypermethylated and up-regulated in the four BC molecular subtypes and a 6-gene signature may predict BC prognosis.
Collapse
Affiliation(s)
- Chongyang Ren
- Department of Breast Cancer, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xiaojiang Tang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, China
| | - Haitao Lan
- Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| |
Collapse
|
28
|
Starzer AM, Berghoff AS, Hamacher R, Tomasich E, Feldmann K, Hatziioannou T, Traint S, Lamm W, Noebauer-Huhmann IM, Furtner J, Müllauer L, Amann G, Bauer S, Schildhaus HU, Preusser M, Heller G, Brodowicz T. Tumor DNA methylation profiles correlate with response to anti-PD-1 immune checkpoint inhibitor monotherapy in sarcoma patients. J Immunother Cancer 2021; 9:jitc-2020-001458. [PMID: 33762319 PMCID: PMC7993298 DOI: 10.1136/jitc-2020-001458] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Some sarcomas respond to immune checkpoint inhibition, but predictive biomarkers are unknown. We analyzed tumor DNA methylation profiles in relation to immunological parameters and response to anti-programmed cell death 1 (anti-PD-1) immune checkpoint inhibitor (ICI) therapy in patients with sarcoma. PATIENTS AND METHODS We retrospectively identified adult patients who had received anti-PD-1 ICI therapy for recurrent sarcoma in two independent centers. We performed (1) blinded radiological response evaluation according to immune response evaluation criteria in solid tumors (iRECIST) ; (2) tumor DNA methylation profiling of >850,000 probes using Infinium MethylationEPIC microarrays; (3) analysis of tumor-infiltrating immune cell subsets (CD3, CD8, CD45RO, FOXP3) and intratumoral expression of immune checkpoint molecules (PD-L1, PD-1, LAG-3) using immunohistochemistry; and (4) evaluation of blood-based systemic inflammation scores (neutrophil-to-lymphocyte ratio, leucocyte-to-lymphocyte ratio, monocyte-to-lymphocyte ratio, platelet-to-lymphocyte ratio). Response to anti-PD-1 ICI therapy was bioinformatically and statistically correlated with DNA methylation profiles and immunological data. RESULTS 35 patients (median age of 50 (23-81) years; 18 females, 17 males; 27 soft tissue sarcomas; 8 osteosarcomas) were included in this study. The objective response rate to anti-PD-1 ICI therapy was 22.9% with complete responses in 3 out of 35 and partial responses in 5 out of 35 patients. Adjustment of DNA methylation data for tumor-infiltrating immune cells resulted in identification of methylation differences between responders and non-responders to anti-PD-1 ICI. 2453 differentially methylated CpG sites (DMPs; 2043 with decreased and 410 with increased methylation) were identified. Clustering of sarcoma samples based on these DMPs revealed two main clusters: methylation cluster 1 (MC1) consisted of 73% responders and methylation cluster 2 (MC2) contained only non-responders to anti-PD-1 ICI. Median progression-free survival from anti-PD-1 therapy start of MC1 and MC2 patients was 16.5 and 1.9 months, respectively (p=0.001). Median overall survival of these patients was 34.4 and 8.0 months, respectively (p=0.029). The most prominent DNA methylation differences were found in pathways implicated in Rap1 signaling, focal adhesion, adherens junction Phosphoinositide 3-kinase (PI3K)-Akt signaling and extracellular matrix (ECM)-receptor interaction. CONCLUSIONS Our data demonstrate that tumor DNA methylation profiles may serve as a predictive marker for response to anti-PD-1 ICI therapy in sarcoma.
Collapse
Affiliation(s)
- Angelika M Starzer
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Rainer Hamacher
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Erwin Tomasich
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Katharina Feldmann
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Teresa Hatziioannou
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Stefan Traint
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Lamm
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Iris M Noebauer-Huhmann
- Department of Biomedical Imaging and Image-guided Therapy, Division of General and Paediatric Radiology, Medical University of Vienna, Vienna, Austria
| | - Julia Furtner
- Department of Biomedical Imaging and Image-guided Therapy, Division of General and Paediatric Radiology, Medical University of Vienna, Vienna, Austria
| | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Gabriele Amann
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | | | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Gerwin Heller
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Thomas Brodowicz
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
29
|
Abstract
Sarcomas are malignant soft tissue and bone tumours affecting adults, adolescents and children. They represent a morphologically heterogeneous class of tumours and some entities lack defining histopathological features. Therefore, the diagnosis of sarcomas is burdened with a high inter-observer variability and misclassification rate. Here, we demonstrate classification of soft tissue and bone tumours using a machine learning classifier algorithm based on array-generated DNA methylation data. This sarcoma classifier is trained using a dataset of 1077 methylation profiles from comprehensively pre-characterized cases comprising 62 tumour methylation classes constituting a broad range of soft tissue and bone sarcoma subtypes across the entire age spectrum. The performance is validated in a cohort of 428 sarcomatous tumours, of which 322 cases were classified by the sarcoma classifier. Our results demonstrate the potential of the DNA methylation-based sarcoma classification for research and future diagnostic applications.
Collapse
|
30
|
Li B, Xu H, He C, Zou W, Tu Y. Lidocaine prevents breast cancer growth by targeting neuronatin to inhibit nerve fibers formation. J Toxicol Sci 2021; 46:329-339. [PMID: 34193770 DOI: 10.2131/jts.46.329] [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] [Indexed: 12/24/2022]
Abstract
Lidocaine has been shown to inhibit the invasion and metastasis of breast cancer, but the mechanism still remains unclear. This study explored the relationship between lidocaine and circulating seeding of breast cancer cells from the perspective of nerve fiber formation. The cell lines MDA-MB-231 and 4T1 were subcutaneously inoculated in mice to simulate the tumor self-seeding by circulating cancer cells. Lidocaine was used to treat these mice and tumor growth was observed. Silver staining was performed to observe the distribution of nerve fibers in tumor-bearing tissues, and immunohistochemical analysis was performed to observe the expression levels of nerve-related proteins. The results showed that lidocaine treatment effectively inhibited tumor growth and nerve fiber formation, and down-regulated the expression levels of protein gene product 9.5, neurofilament, nerve growth factor (NGF), and neuronatin (Nnat). Overexpression NGF and Nnat both could reverse the therapeutic effects of lidocaine. These results suggest that the effect of lidocaine on inhibiting breast cancer invasion and metastasis may be achieved by targeting Nnat, regulating the production of NGFs in cancer cells, and subsequently inhibiting the formation of nerve fibers.
Collapse
Affiliation(s)
- Bingda Li
- Department of Anesthesiology, Jiangxi Cancer Hospital of Nanchang University, China
| | - Hao Xu
- Department of Pediatrics, Wuhan NO.1 Hospital, China
| | - Chongwu He
- Department of Breast Surgery, Jiangxi Cancer Hospital of Nanchang University, China
| | - Wenxiong Zou
- Department of Emergency, Ji'an Central People's Hospital, China
| | - Yun Tu
- Department of Oncology, Jiangxi Cancer Hospital of Nanchang University, China
| |
Collapse
|
31
|
Salta S, Macedo-Silva C, Miranda-Gonçalves V, Lopes N, Gigliano D, Guimarães R, Farinha M, Sousa O, Henrique R, Jerónimo C. A DNA methylation-based test for esophageal cancer detection. Biomark Res 2020; 8:68. [PMID: 33292587 PMCID: PMC7691099 DOI: 10.1186/s40364-020-00248-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022] Open
Abstract
Background Esophageal cancer (ECa) is the 7th most incident cancer and the 6th leading cause of cancer-related death. Most patients are diagnosed with locally advanced or metastatic disease, enduring poor survival. Biomarkers enabling early cancer detection may improve patient management, treatment effectiveness, and survival, are urgently needed. In this context, epigenetic-based biomarkers such as DNA methylation are potential candidates. Methods Herein, we sought to identify and validate DNA methylation-based biomarkers for early detection and prediction of response to therapy in ECa patients. Promoter methylation levels were assessed in a series of treatment-naïve ECa, post-neoadjuvant treatment ECa, and normal esophagus tissues, using quantitative methylation-specific PCR for COL14A1, GPX3, and ZNF569. Results ZNF569 methylation (ZNF569me) levels significantly differed between ECa and normal samples (p < 0.001). Moreover, COL14A1 methylation (COL14A1me) and GPX3 methylation (GPX3me) levels discriminated adenocarcinomas and squamous cell carcinomas, respectively, from normal samples (p = 0.002 and p = 0.009, respectively). COL14A1me & ZNF569me accurately identified adenocarcinomas (82.29%) whereas GPX3me & ZNF569me identified squamous cell carcinomas with 81.73% accuracy. Furthermore, ZNF569me and GPX3me levels significantly differed between normal and pre-treated ECa. Conclusion The biomarker potential of a specific panel of methylated genes for ECa was confirmed. These might prove useful for early detection and might allow for the identification of minimal residual disease after adjuvant therapy.
Collapse
Affiliation(s)
- Sofia Salta
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Catarina Macedo-Silva
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Vera Miranda-Gonçalves
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Nair Lopes
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Davide Gigliano
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, Porto, 4200-072, Portugal
| | - Rita Guimarães
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, Porto, 4200-072, Portugal
| | - Mónica Farinha
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, Porto, 4200-072, Portugal
| | - Olga Sousa
- Department of Radiation Oncology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, Porto, 4200-072, Portugal
| | - Rui Henrique
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal.,Department of Pathology, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino de Almeida, Porto, 4200-072, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar- University of Porto , Rua de Jorge Viterbo Ferreira, 228, Porto, 4050-313, Portugal
| | - Carmen Jerónimo
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal. .,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar- University of Porto , Rua de Jorge Viterbo Ferreira, 228, Porto, 4050-313, Portugal.
| |
Collapse
|
32
|
Sill M, Plass C, Pfister SM, Lipka DB. Molecular tumor classification using DNA methylome analysis. Hum Mol Genet 2020; 29:R205-R213. [PMID: 32657331 DOI: 10.1093/hmg/ddaa147] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/18/2022] Open
Abstract
Tumor classifiers based on molecular patterns promise to define and reliably classify tumor entities. The high tissue- and cell type-specificity of DNA methylation, as well as its high stability, makes DNA methylation an ideal choice for the development of tumor classifiers. Herein, we review existing tumor classifiers using DNA methylome analysis and will provide an overview on their emerging impact on cancer classification, the detection of novel cancer subentities and patient stratification with a focus on brain tumors, sarcomas and hematopoietic malignancies. Furthermore, we provide an outlook on the enormous potential of DNA methylome analysis to complement classical histopathological and genetic diagnostics, including the emerging field of epigenomic analysis in liquid biopsies.
Collapse
Affiliation(s)
- Martin Sill
- Hopp Children's Cancer Center at the National Center for Tumor Diseases (NCT) Heidelberg, 69120 Heidelberg, Germany.,Division of Pediatric Neurooncology (B062), German Cancer Research Center and German Cancer Consortium, 69120 Heidelberg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center at the National Center for Tumor Diseases (NCT) Heidelberg, 69120 Heidelberg, Germany.,Division of Pediatric Neurooncology (B062), German Cancer Research Center and German Cancer Consortium, 69120 Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Daniel B Lipka
- Division of Translational Medical Oncology, Section Translational Cancer Epigenomics, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT) Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
33
|
Saeed H, Sinha S, Mella C, Kuerbitz JS, Cales ML, Steele MA, Stanke J, Damron D, Safadi F, Kuerbitz SJ. Aberrant epigenetic silencing of neuronatin is a frequent event in human osteosarcoma. Oncotarget 2020; 11:1876-1893. [PMID: 32499872 PMCID: PMC7244018 DOI: 10.18632/oncotarget.27583] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 04/03/2020] [Indexed: 11/25/2022] Open
Abstract
The paternally imprinted neuronatin (NNAT) gene has been identified as a target of aberrant epigenetic silencing in diverse cancers, but no association with pediatric bone cancers has been reported to date. In screening childhood cancers, we identified aberrant CpG island hypermethylation in a majority of osteosarcoma (OS) samples and in 5 of 6 human OS cell lines studied but not in normal bone-derived tissue samples. CpG island hypermethylation was associated with transcriptional silencing in human OS cells, and silencing was reversible upon treatment with 5-aza-2'-deoxycytidine. Expression of NNAT was detectable in osteoblasts and chondrocytes of human bone, supporting a potential role in bone homeostasis. Enforced expression of NNAT in human OS cells lacking endogenous expression resulted in significant reduction in colony formation and in vitro migration compared to nonexpressor control cells. We next analyzed the effect of NNAT expression on intracellular calcium homeostasis and found that was associated with an attenuated decay of calcium levels to baseline following ATP-induced release of calcium from endoplasmic reticulum (ER) stores. Furthermore, NNAT expression was associated with increased cytotoxicity in OS cells from thapsigargin, an inhibitor of calcium reuptake into ER and an inducer of the ER stress response. These results suggest a possible tumor suppressor role for NNAT in human osteosarcoma. Additional study is needed ascertain sensitization to ER stress-associated apoptosis as a mechanism of NNAT-dependent cytotoxicity. In that case, epigenetic modification therapy to effect NNAT transcriptional derepression may represent a therapeutic strategy potentially of benefit to a majority of osteosarcoma patients.
Collapse
Affiliation(s)
- Haleema Saeed
- Division of Pediatric Hematology/Oncology, Akron Childrens Hospital, Akron, OH, USA.,Current affiliation: Shaukat Khanum Cancer Hospital and Research Centre, Lahore, Pakistan
| | - Sayantani Sinha
- Department of Biological Sciences, Kent State University, Kent, OH, USA.,Current affiliation: Department of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Christine Mella
- Division of Pediatric Hematology/Oncology, Akron Childrens Hospital, Akron, OH, USA
| | - Jeffrey S Kuerbitz
- Division of Pediatric Hematology/Oncology, Akron Childrens Hospital, Akron, OH, USA.,Current affiliation: Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Monica L Cales
- College of Osteopathic Medicine, University of Pikeville, Pikeville, KY, USA.,Current affiliation: Penn State Health St. Joseph, Reading, PA, USA
| | - Mark A Steele
- Division of Pediatric Hematology/Oncology, Akron Childrens Hospital, Akron, OH, USA
| | - Jennifer Stanke
- Division of Pediatric Hematology/Oncology, Akron Childrens Hospital, Akron, OH, USA.,Current affiliation: Foundation Medicine, Boston, MA, USA
| | - Derek Damron
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Fayez Safadi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, USA.,Musculoskeletal Research Group, Northeast Ohio Medical University, Rootstown, OH, USA.,Rebecca D. Considine Research Institute, Akron Children's Hospital, Akron, OH, USA
| | - Steven J Kuerbitz
- Division of Pediatric Hematology/Oncology, Akron Childrens Hospital, Akron, OH, USA.,Rebecca D. Considine Research Institute, Akron Children's Hospital, Akron, OH, USA.,Department of Pediatrics, Northeast Ohio Medical University, Rootstown, OH, USA
| |
Collapse
|
34
|
Liu S, Yang Z, Li G, Li C, Luo Y, Gong Q, Wu X, Li T, Zhang Z, Xing B, Xu X, Lu X. Multi-omics Analysis of Primary Cell Culture Models Reveals Genetic and Epigenetic Basis of Intratumoral Phenotypic Diversity. GENOMICS PROTEOMICS & BIOINFORMATICS 2020; 17:576-589. [PMID: 32205176 PMCID: PMC7212478 DOI: 10.1016/j.gpb.2018.07.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 05/29/2018] [Accepted: 07/24/2018] [Indexed: 12/27/2022]
Abstract
Uncovering the functionally essential variations related to tumorigenesis and tumor progression from cancer genomics data is still challenging due to the genetic diversity among patients, and extensive inter- and intra-tumoral heterogeneity at different levels of gene expression regulation, including but not limited to the genomic, epigenomic, and transcriptional levels. To minimize the impact of germline genetic heterogeneities, in this study, we establish multiple primary cultures from the primary and recurrent tumors of a single patient with hepatocellular carcinoma (HCC). Multi-omics sequencing was performed for these cultures that encompass the diversity of tumor cells from the same patient. Variations in the genome sequence, epigenetic modification, and gene expression are used to infer the phylogenetic relationships of these cell cultures. We find the discrepancy among the relationships revealed by single nucleotide variations (SNVs) and transcriptional/epigenomic profiles from the cell cultures. We fail to find overlap between sample-specific mutated genes and differentially expressed genes (DEGs), suggesting that most of the heterogeneous SNVs among tumor stages or lineages of the patient are functionally insignificant. Moreover, copy number alterations (CNAs) and DNA methylation variation within gene bodies, rather than promoters, are significantly correlated with gene expression variability among these cell cultures. Pathway analysis of CNA/DNA methylation-related genes indicates that a single cell clone from the recurrent tumor exhibits distinct cellular characteristics and tumorigenicity, and such an observation is further confirmed by cellular experiments both in vitro and in vivo. Our systematic analysis reveals that CNAs and epigenomic changes, rather than SNVs, are more likely to contribute to the phenotypic diversity among subpopulations in the tumor. These findings suggest that new therapeutic strategies targeting gene dosage and epigenetic modification should be considered in personalized cancer medicine. This culture model may be applied to the further identification of plausible determinants of cancer metastasis and relapse.
Collapse
Affiliation(s)
- Sixue Liu
- (1)CAS Key Laboratory of Genomics and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; (2)University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zuyu Yang
- (1)CAS Key Laboratory of Genomics and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; (3)Invasive Pathogens Laboratory, Institute of Environmental Science and Research, Porirua 5022, Wellington, New Zealand
| | - Guanghao Li
- (1)CAS Key Laboratory of Genomics and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; (2)University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunyan Li
- (1)CAS Key Laboratory of Genomics and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; (2)University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanting Luo
- (1)CAS Key Laboratory of Genomics and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; (2)University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiang Gong
- (1)CAS Key Laboratory of Genomics and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xin Wu
- (1)CAS Key Laboratory of Genomics and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Tao Li
- (1)CAS Key Laboratory of Genomics and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; (2)University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiqian Zhang
- (4)Department of Cell Biology, Key Laboratory of Carcinogenesis and Translational Research, Center for Molecular and Translational Medicine, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Baocai Xing
- (5)Department of Hepatobiliary Surgery I, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Xiaolan Xu
- (6)National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Xuemei Lu
- (1)CAS Key Laboratory of Genomics and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; (2)University of Chinese Academy of Sciences, Beijing 100049, China; (7)CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
35
|
Miele E, De Vito R, Ciolfi A, Pedace L, Russo I, De Pasquale MD, Di Giannatale A, Crocoli A, Angelis BD, Tartaglia M, Alaggio R, Milano GM. DNA Methylation Profiling for Diagnosing Undifferentiated Sarcoma with Capicua Transcriptional Receptor ( CIC) Alterations. Int J Mol Sci 2020; 21:ijms21051818. [PMID: 32155762 PMCID: PMC7084764 DOI: 10.3390/ijms21051818] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/15/2020] [Accepted: 03/03/2020] [Indexed: 12/31/2022] Open
Abstract
Undifferentiated soft tissue sarcomas are a group of diagnostically challenging tumors in the pediatric population. Molecular techniques are instrumental for the categorization and differential diagnosis of these tumors. A subgroup of recently identified soft tissue sarcomas with undifferentiated round cell morphology was characterized by Capicua transcriptional receptor (CIC) rearrangements. Recently, an array-based DNA methylation analysis of undifferentiated tumors with small blue round cell histology was shown to provide a highly robust and reproducible approach for precisely classifying this diagnostically challenging group of tumors. We describe the case of an undifferentiated sarcoma of the abdominal wall in a 12-year-old girl. The patient presented with a voluminous mass of the abdominal wall, and multiple micro-nodules in the right lung. The tumor was unclassifiable with current immunohistochemical and molecular approaches. However, DNA methylation profiling allowed us to classify this neoplasia as small blue round cell tumor with CIC alterations. The patient was treated with neoadjuvant chemotherapy followed by complete surgical resection and adjuvant chemotherapy. After 22 months, the patient is disease-free and in good clinical condition. To put our experience in context, we conducted a literature review, analyzing current knowledge and state-of-the-art diagnosis, prognosis, and clinical management of CIC rearranged sarcomas. Our findings further support the use of DNA methylation profiling as an important tool to improve diagnosis of non-Ewing small round cell tumors.
Collapse
Affiliation(s)
- Evelina Miele
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, IRCCS Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (L.P.); (I.R.); (M.D.D.P.); (A.D.G.); (B.D.A.); (G.M.M.)
- Correspondence:
| | - Rita De Vito
- Department of Laboratories, Pathology Unit, IRCCS Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (R.D.V.); (R.A.)
| | - Andrea Ciolfi
- Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital (IRCCS), 00165 Rome, Italy; (A.C.); (M.T.)
| | - Lucia Pedace
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, IRCCS Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (L.P.); (I.R.); (M.D.D.P.); (A.D.G.); (B.D.A.); (G.M.M.)
| | - Ida Russo
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, IRCCS Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (L.P.); (I.R.); (M.D.D.P.); (A.D.G.); (B.D.A.); (G.M.M.)
| | - Maria Debora De Pasquale
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, IRCCS Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (L.P.); (I.R.); (M.D.D.P.); (A.D.G.); (B.D.A.); (G.M.M.)
| | - Angela Di Giannatale
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, IRCCS Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (L.P.); (I.R.); (M.D.D.P.); (A.D.G.); (B.D.A.); (G.M.M.)
| | - Alessandro Crocoli
- Department of Surgery, IRCCS Bambino Gesù Children’s Hospital, 00165 Rome, Italy;
| | - Biagio De Angelis
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, IRCCS Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (L.P.); (I.R.); (M.D.D.P.); (A.D.G.); (B.D.A.); (G.M.M.)
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital (IRCCS), 00165 Rome, Italy; (A.C.); (M.T.)
| | - Rita Alaggio
- Department of Laboratories, Pathology Unit, IRCCS Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (R.D.V.); (R.A.)
| | - Giuseppe Maria Milano
- Department of Pediatric Onco-Hematology and Cell and Gene Therapy, IRCCS Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (L.P.); (I.R.); (M.D.D.P.); (A.D.G.); (B.D.A.); (G.M.M.)
| |
Collapse
|
36
|
Mallik S, Bandyopadhyay S. WeCoMXP: Weighted Connectivity Measure Integrating Co-Methylation, Co-Expression and Protein-Protein Interactions for Gene-Module Detection. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:690-703. [PMID: 30183644 DOI: 10.1109/tcbb.2018.2868348] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The identification of modules (groups of several tightly interconnected genes) in gene interaction network is an essential task for better understanding of the architecture of the whole network. In this article, we develop a novel weighted connectivity measure integrating co-methylation, co-expression, and protein-protein interactions (called WeCoMXP) to detect gene-modules for multi-omics dataset. The proposed measure goes beyond the fundamental degree centrality measure through considering some formulation of higher-order connections. Thereafter, we apply the average linkage clustering method using the corresponding dissimilarity (distance) values of WeCoMXP scores, and utilize a dynamic tree cut method for identifying some gene-modules. We validate the modules through literature search, KEGG pathway, and gene-ontology analyses on the genes representing the modules. Furthermore, the top 10 TFs/miRNAs that are connected with the maximum number of gene-modules and that regulate/target the maximum number of genes from these connected gene-modules, are identified. Moreover, our proposed method provides a better performance than the existing methods in terms of several cluster-validity indices in maximum times.
Collapse
|
37
|
Zhang Y, Kou C, Wang S, Zhang Y. Genome-wide Differential-based Analysis of the Relationship between DNA Methylation and Gene Expression in Cancer. Curr Bioinform 2019. [DOI: 10.2174/1574893614666190424160046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Background::
DNA methylation is an epigenetic modification that plays an important
role in regulating gene expression. There is evidence that the hypermethylation of promoter regions
always causes gene silencing. However, how the methylation patterns of other regions in the
genome, such as gene body and 3’UTR, affect gene expression is unknown.
Objective::
The study aimed to fully explore the relationship between DNA methylation and expression
throughout the genome-wide analysis which is important in understanding the function of
DNA methylation essentially.
Method::
In this paper, we develop a heuristic framework to analyze the relationship between the
methylated change in different regions and that of the corresponding gene expression based on differential
analysis.
Results::
To understande the methylated function of different genomic regions, a gene is divided
into seven functional regions. By applying the method in five cancer datasets from the Synapse database,
it was found that methylated regions with a significant difference between cases and controls
were almost uniformly distributed in the seven regions of the genome. Also, the effect of
DNA methylation in different regions on gene expression was different. For example, there was a
higher percentage of positive relationships in 1stExon, gene body and 3’UTR than in TSS1500 and
TSS200. The functional analysis of genes with a significant positive and negative correlation between
DNA methylation and gene expression demonstrated the epigenetic mechanism of cancerassociated
genes.
Conclusion::
Differential based analysis helps us to recognize the change in DNA methylation and
how this change affects the change in gene expression. It provides a basis for further integrating
gene expression and DNA methylation data to identify disease-associated biomarkers.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- School of information and control engineering, Qingdao University of Technology, Qingdao, Shandong, China
| | - Chuanhua Kou
- School of information and control engineering, Qingdao University of Technology, Qingdao, Shandong, China
| | - Shudong Wang
- College of Computer and Communication Engineering, China University of Petroleum (East China), Qingdao, Shandong, China
| | - Yulin Zhang
- College of Mathematics and Systems Science, Shandong University of Science and Technology, Qingdao, Shandong, China
| |
Collapse
|
38
|
Pemov A, Li H, Presley W, Wallace MR, Miller DT. Genetics of human malignant peripheral nerve sheath tumors. Neurooncol Adv 2019; 2:i50-i61. [PMID: 32642732 PMCID: PMC7317054 DOI: 10.1093/noajnl/vdz049] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are heterogeneous, highly aggressive tumors with no widely effective treatment other than surgery. Genomic architecture of MPNST is similar to other soft tissue sarcomas, with a relatively modest burden of single nucleotide variants and an elevated frequency of copy-number alterations. Recent advances in genomic studies identified previously unrecognized critical involvement of polycomb repressor complex 2 (PRC2) core components SUZ12 and EED in transition to malignancy. Notably, somatic changes in NF1, CDKN2A/B, and PRC2 are found in most MPNST regardless of their etiology (e.g. neurofibromatosis type 1-associated vs. sporadic vs. radiation-induced), indicating that similar molecular mechanisms impact pathogenesis in these neoplasms. The timing and specific order of genetic or epigenetic changes may, however, explain the typically poorer prognosis of NF1-associated MPNSTs. Studies that reveal genes and regulatory pathways uniquely altered in malignancies are essential to development of targeted tumor therapies. Characterization of MPNST molecular profiles may also contribute to tools for earlier detection, and prediction of prognosis or drug response. Here we review the genetic discoveries and their implications in understanding MPNST biology.
Collapse
Affiliation(s)
- Alexander Pemov
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Hua Li
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida
| | - William Presley
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida
| | - Margaret R Wallace
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida.,University of Florida Health Cancer Center, University of Florida, Gainesville, Florida
| | - David T Miller
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
39
|
Jones DTW, Banito A, Grünewald TGP, Haber M, Jäger N, Kool M, Milde T, Molenaar JJ, Nabbi A, Pugh TJ, Schleiermacher G, Smith MA, Westermann F, Pfister SM. Molecular characteristics and therapeutic vulnerabilities across paediatric solid tumours. Nat Rev Cancer 2019; 19:420-438. [PMID: 31300807 DOI: 10.1038/s41568-019-0169-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2019] [Indexed: 02/06/2023]
Abstract
The spectrum of tumours arising in childhood is fundamentally different from that seen in adults, and they are known to be divergent from adult malignancies in terms of cellular origins, epidemiology, genetic complexity, driver mutations and underlying mutational processes. Despite the immense knowledge generated through sequencing efforts and functional characterization of identified (epi-)genetic alterations over the past decade, the clinical implications of this knowledge have so far been limited. Novel preclinical platforms such as the European Innovative Therapies for Children with Cancer-Paediatric Preclinical Proof-of-Concept Platform and the US-based Pediatric Preclinical Testing Consortium are being developed to try to change this by aiming to recapitulate the extensive heterogeneity of paediatric tumours and thereby, hopefully, improve the ability to predict clinical benefit. Numerous studies have also been established worldwide to provide patients with access to real-time molecular profiling and the possibility of more precise mechanism-of-action-based treatments. In addition to tumour-intrinsic findings and mechanisms, ongoing studies are investigating features such as the immune microenvironment of paediatric tumours in comparison with adult cancers - currently of very timely clinical relevance. However, there is an ongoing need for rigorous preclinical biomarker and target validation to feed into the next generation of molecularly stratified clinical trials. This Review aims to provide a comprehensive state-of-the-art overview of the molecular landscape of paediatric solid tumours, including their underlying genomic alterations and interactions with the microenvironment, complemented with our current understanding of potential therapeutic vulnerabilities and how these can be preclinically tested using more accurate predictive methods. Finally, we provide an outlook on the challenges and opportunities associated with translating this overwhelming scientific progress into real clinical benefit.
Collapse
Affiliation(s)
- David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Pediatric Glioma Research Group, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ana Banito
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Pediatric Soft Tissue Sarcoma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas G P Grünewald
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Australia, Randwick, NSW, Australia
- School of Women's & Children's Health, UNSW Australia, Randwick, NSW, Australia
| | - Natalie Jäger
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcel Kool
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Till Milde
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jan J Molenaar
- Princess Maxima Center for Pediatric Cancer, Utrecht, The Netherlands
| | - Arash Nabbi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Gudrun Schleiermacher
- SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer), Institut Curie, Paris, France
- INSERM U830, Laboratoire de Génétique et Biologie des Cancers, Research Center, Institut Curie, Paris, France
| | - Malcolm A Smith
- Cancer Therapy Evaluation Program, National Cancer Institute, Rockville, MD, USA
| | - Frank Westermann
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
40
|
Bertucci F, Finetti P, Monneur A, Perrot D, Chevreau C, Le Cesne A, Blay JY, Mir O, Birnbaum D. PARP1 expression in soft tissue sarcomas is a poor-prognosis factor and a new potential therapeutic target. Mol Oncol 2019; 13:1577-1588. [PMID: 31131495 PMCID: PMC6599836 DOI: 10.1002/1878-0261.12522] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/14/2019] [Accepted: 05/24/2019] [Indexed: 11/10/2022] Open
Abstract
Soft tissue sarcomas (STSs) are aggressive tumors with few efficient systemic therapies. Poly(ADP‐ribose) polymerase‐1 (PARP1) inhibitors represent an emerging therapeutic option in tumors with genomic instability. The genomics of STSs is complex in more than half of cases, suggesting a high level of inherent DNA damage and genomic instability. Thus, STSs could be efficiently targeted with PARP inhibitors. Promising preclinical results have been reported, but few data are available regarding PARP1 expression in clinical samples. We examined PARP1 mRNA expression in 1464 clinical samples of STS, including 1432 primary tumors and 32 relapses, and searched for correlations with clinicopathological features, including metastasis‐free survival (MFS). Expression was heterogeneous across the samples, not different between primary and secondary tumors, and was correlated to PARP1 DNA copy number. In the 1432 primary tumors, the ‘PARP1‐high’ samples were associated with younger patients, more frequent locations at the extremities, superficial trunk and head and neck, more leiomyosarcomas and other STSs and less liposarcomas and myxofibrosarcomas, more grade 3, more high‐risk CINSARC tumors, and more ‘chromosomically instable’ tumors. They were associated with shorter MFS, independently of other significant prognostic features, including the CINSARC signature. We found a strong involvement of genes overexpressed in the ‘PARP1‐high’ samples in cell cycle, DNA replication, and DNA repair. PARP1 expression refines the prediction of MFS in STSs, and similar expression exists in secondary and primary tumors, supporting the development of PARP1 inhibitors.
Collapse
Affiliation(s)
- François Bertucci
- Predictive Oncology Laboratory, Marseille Cancer Research Center (CRCM), Institut Paoli-Calmettes, U1068 INSERM, U7258 CNRS, Aix-Marseille University, Marseille, France.,Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France.,French Sarcoma Group, Lyon, France
| | - Pascal Finetti
- Predictive Oncology Laboratory, Marseille Cancer Research Center (CRCM), Institut Paoli-Calmettes, U1068 INSERM, U7258 CNRS, Aix-Marseille University, Marseille, France
| | - Audrey Monneur
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Delphine Perrot
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Christine Chevreau
- French Sarcoma Group, Lyon, France.,Department of Medical Oncology, IUCT-Oncopole, Institut Claudius-Regaud, Toulouse, France
| | - Axel Le Cesne
- French Sarcoma Group, Lyon, France.,Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Jean-Yves Blay
- French Sarcoma Group, Lyon, France.,Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Olivier Mir
- French Sarcoma Group, Lyon, France.,Department of Ambulatory Care, Gustave Roussy, Villejuif, France
| | - Daniel Birnbaum
- Predictive Oncology Laboratory, Marseille Cancer Research Center (CRCM), Institut Paoli-Calmettes, U1068 INSERM, U7258 CNRS, Aix-Marseille University, Marseille, France
| |
Collapse
|
41
|
Lei W, Xu Y, Su J, Chong CM, Su HX, Luo J, Fang EF, Bao Z, Chen G. Applications of high-throughput ‘omics’ data in the study of frailty. TRANSLATIONAL MEDICINE OF AGING 2019. [DOI: 10.1016/j.tma.2019.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
42
|
Mallik S, Bhadra T, Mukherji A, Mallik S, Bhadra T, Mukherji A, Mallik S, Bhadra T, Mukherji A. DTFP-Growth: Dynamic Threshold-Based FP-Growth Rule Mining Algorithm Through Integrating Gene Expression, Methylation, and Protein-Protein Interaction Profiles. IEEE Trans Nanobioscience 2018; 17:117-125. [PMID: 29870335 DOI: 10.1109/tnb.2018.2803021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Association rule mining is an important technique for identifying interesting relationships between gene pairs in a biological data set. Earlier methods basically work for a single biological data set, and, in maximum cases, a single minimum support cutoff can be applied globally, i.e., across all genesets/itemsets. To overcome this limitation, in this paper, we propose dynamic threshold-based FP-growth rule mining algorithm that integrates gene expression, methylation and protein-protein interaction profiles based on weighted shortest distance to find the novel associations among different pairs of genes in multi-view data sets. For this purpose, we introduce three new thresholds, namely, Distance-based Variable/Dynamic Supports (DVS), Distance-based Variable Confidences (DVC), and Distance-based Variable Lifts (DVL) for each rule by integrating co-expression, co-methylation, and protein-protein interactions existed in the multi-omics data set. We develop the proposed algorithm utilizing these three novel multiple threshold measures. In the proposed algorithm, the values of , , and are computed for each rule separately, and subsequently it is verified whether the support, confidence, and lift of each evolved rule are greater than or equal to the corresponding individual , , and values, respectively, or not. If all these three conditions for a rule are found to be true, the rule is treated as a resultant rule. One of the major advantages of the proposed method compared with other related state-of-the-art methods is that it considers both the quantitative and interactive significance among all pairwise genes belonging to each rule. Moreover, the proposed method generates fewer rules, takes less running time, and provides greater biological significance for the resultant top-ranking rules compared to previous methods.
Collapse
|
43
|
Ma X, Sun P, Zhang ZY. An Integrative Framework for Protein Interaction Network and Methylation Data to Discover Epigenetic Modules. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 16:1855-1866. [PMID: 29994031 DOI: 10.1109/tcbb.2018.2831666] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
DNA methylation is a critical epigenetic modification that plays an important role in cancers. The available algorithms fail to fully characterize epigenetic modules. To address this issue, we first characterize the epigenetic module as a group of well-connected genes in the protein interaction network and are also co-methylated based on gene methylation profiles. Then, the epigenetic module discovery problem is transformed into an optimization problem. Then, a regularized nonnegative matrix factorization algorithm for methylation modules (RNMF-MM) is presented, where the co-methylation constraint is treated as a regularizer. Using the artificial networks with known module structure, we demonstrate that the proposed algorithm outperforms state-of-the-art approaches in terms of accuracy. On the basis of breast cancer methylation data and protein interaction network, the RNMF-MM algorithm discovers methylation modules that are significantly more enriched by the known pathways than those obtained by other algorithms. These modules serve as biomarkers for predicting cancer stages and estimating survival time of patients. The proposed model and algorithm provide an effective way for the integrative analysis of protein interaction network and methylation data.
Collapse
|
44
|
Rahat B, Sharma R, Bagga R, Hamid A, Kaur J. Imbalance between matrix metalloproteinases and their tissue inhibitors in preeclampsia and gestational trophoblastic diseases. Reproduction 2017; 152:11-22. [PMID: 27256632 DOI: 10.1530/rep-16-0060] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/07/2016] [Indexed: 01/16/2023]
Abstract
The invasion cascade exhibited by placental trophoblasts and cancerous cells bears many similarities, and it is attributed to extracellular matrix degradation mediated by matrix metalloproteinases (MMPs). Although proper and controlled invasion by trophoblasts into the maternal uterus is an essential requirement for maintenance of normal pregnancy, any abnormality in this phenomenon results in the development of invasion-related disorders such as gestational trophoblastic diseases (GTDs) and preeclampsia. We studied the epigenetic basis of differential expression of two placental MMPs (MMP2 and MMP9) and tissue inhibitors of metalloproteinases (TIMP2 and TIMP1) during normal gestation and invasion-related disorders, i.e., preeclampsia and GTDs. Our study suggests the association of H3K9/27me3 with differential expression of these MMPs and their inhibitors, which regulate the placental invasion during normal pregnancy, whereas no role of CpG methylation was observed in the differential expression of MMPs/TIMPs. Further, development of GTDs was associated with abnormally higher expression of these MMPs and lower levels of their inhibitors, whereas the reverse trends were observed for MMPs and their TIMPs in case of preeclampsia, in association with abnormal changes in H3K9/27me3. These results suggest the involvement of higher levels of MMPs in an aggressive invasive behavior depicted by GTDs, whereas lower levels of these MMPs in shallow and poor invasive phenotype associated with preeclampsia. Thus, our study shows the significance of a proper balance regulated by histone trimethylation between differential expression of MMPs and their TIMPs for maintaining normal pregnancy and its deregulation as a contributing factor for pathogenesis of invasive disorders during pregnancy.
Collapse
Affiliation(s)
- Beenish Rahat
- Department of BiochemistryPostgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Renuka Sharma
- Department of BiochemistryPostgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rashmi Bagga
- Department of Obstetrics and GynecologyPostgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Abid Hamid
- Cancer Pharmacology DivisionCSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Jyotdeep Kaur
- Department of BiochemistryPostgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
45
|
Wu SP, Cooper BT, Bu F, Bowman CJ, Killian JK, Serrano J, Wang S, Jackson TM, Gorovets D, Shukla N, Meyers PA, Pisapia DJ, Gorlick R, Ladanyi M, Thomas K, Snuderl M, Karajannis MA. DNA Methylation-Based Classifier for Accurate Molecular Diagnosis of Bone Sarcomas. JCO Precis Oncol 2017; 2017. [PMID: 29354796 DOI: 10.1200/po.17.00031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Purpose Pediatric sarcomas provide a unique diagnostic challenge. There is considerable morphologic overlap between entities, increasing the importance of molecular studies in the diagnosis, treatment, and identification of therapeutic targets. We developed and validated a genome-wide DNA methylation based classifier to differentiate between osteosarcoma, Ewing's sarcoma, and synovial sarcoma. Materials and Methods DNA methylation status of 482,421 CpG sites in 10 Ewing's sarcoma, 11 synovial sarcoma, and 15 osteosarcoma samples were determined using the Illumina Infinium HumanMethylation450 array. We developed a random forest classifier trained from the 400 most differentially methylated CpG sites within the training set of 36 sarcoma samples. This classifier was validated on data drawn from The Cancer Genome Atlas (TCGA) synovial sarcoma, TARGET Osteosarcoma, and a recently published series of Ewing's sarcoma. Results Methylation profiling revealed three distinct patterns, each enriched with a single sarcoma subtype. Within the validation cohorts, all samples from TCGA were accurately classified as synovial sarcoma (10/10, sensitivity and specificity 100%), all but one sample from TARGET-OS were classified as osteosarcoma (85/86, sensitivity 98%, specificity 100%) and 14/15 Ewing's sarcoma samples classified correctly (sensitivity 93%, specificity 100%). The single misclassified osteosarcoma sample demonstrated high EWSR1 and ETV1 expression on RNA-seq although no fusion was found on manual curation of the transcript sequence. Two additional clinical samples, that were difficult to classify by morphology and molecular methods, were classified as osteosarcoma when previously suspected to be a synovial sarcoma and Ewing's sarcoma on initial diagnosis, respectively. Conclusion Osteosarcoma, synovial sarcoma, and Ewing's sarcoma have distinct epigenetic profiles. Our validated methylation-based classifier can be used to provide diagnostic assistance when histological and standard techniques are inconclusive.
Collapse
Affiliation(s)
- S Peter Wu
- Department of Radiation Oncology, NYU Langone Medical Center, New York, NY
| | - Benjamin T Cooper
- Department of Radiation Oncology, NYU Langone Medical Center, New York, NY
| | - Fang Bu
- Department of Pathology, NYU Langone Medical Center, New York, NY
| | | | - J Keith Killian
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jonathan Serrano
- Department of Pathology, NYU Langone Medical Center, New York, NY
| | - Shiyang Wang
- Department of Pediatrics, NYU Langone Medical Center, New York, NY
| | - Twana M Jackson
- Department of Pediatrics, NYU Langone Medical Center, New York, NY
| | - Daniel Gorovets
- Department of Radiation Oncology, NYU Langone Medical Center, New York, NY
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Paul A Meyers
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David J Pisapia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Richard Gorlick
- Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kristen Thomas
- Department of Pathology, NYU Langone Medical Center, New York, NY
| | - Matija Snuderl
- Department of Pathology, NYU Langone Medical Center, New York, NY
| | | |
Collapse
|
46
|
Lou S, Balluff B, de Graaff MA, Cleven AHG, Briaire-de Bruijn I, Bovée JVMG, McDonnell LA. High-grade sarcoma diagnosis and prognosis: Biomarker discovery by mass spectrometry imaging. Proteomics 2017; 16:1802-13. [PMID: 27174013 DOI: 10.1002/pmic.201500514] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 05/04/2016] [Accepted: 05/09/2016] [Indexed: 12/24/2022]
Abstract
The combination of high heterogeneity, both intratumoral and intertumoral, with their rarity has made diagnosis, prognosis of high-grade sarcomas difficult. There is an urgent need for more objective molecular biomarkers, to differentiate between the many different subtypes, and to also provide new treatment targets. Mass spectrometry imaging (MSI) has amply demonstrated its ability to identify potential new markers for patient diagnosis, survival, metastasis and response to therapy in cancer research. In this study, we investigated the ability of MALDI-MSI of proteins to distinguish between high-grade osteosarcoma (OS), leiomyosarcoma (LMS), myxofibrosarcoma (MFS) and undifferentiated pleomorphic sarcoma (UPS) (Ntotal = 53). We also investigated if there are individual proteins or protein signatures that are statistically associated with patient survival. Twenty diagnostic protein signals were found characteristic for specific tumors (p ≤ 0.05), amongst them acyl-CoA-binding protein (m/z 11 162), macrophage migration inhibitory factor (m/z 12 350), thioredoxin (m/z 11 608) and galectin-1 (m/z 14 633) were assigned. Another nine protein signals were found to be associated with overall survival (p ≤ 0.05), including proteasome activator complex subunit 1 (m/z 9753), indicative for non-OS patients with poor survival; and two histone H4 variants (m/z 11 314 and 11 355), indicative of poor survival for LMS patients.
Collapse
Affiliation(s)
- Sha Lou
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Benjamin Balluff
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands.,Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Maastricht, The Netherlands
| | - Marieke A de Graaff
- Department of Pathology, Leiden University, Medical Center, Leiden, The Netherlands
| | - Arjen H G Cleven
- Department of Pathology, Leiden University, Medical Center, Leiden, The Netherlands
| | | | - Judith V M G Bovée
- Department of Pathology, Leiden University, Medical Center, Leiden, The Netherlands
| | - Liam A McDonnell
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands.,Department of Pathology, Leiden University, Medical Center, Leiden, The Netherlands.,Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| |
Collapse
|
47
|
Abstract
Aflatoxin B1 (AFB1) is widely distributed in nature, especially in a variety of food commodities. It is confirmed to be the most toxic of all the aflatoxins. The toxicity of AFB1 has been well investigated, and it may result in severe health problems including carcinogenesis, mutagenesis, growth retardation, and immune suppression. Epigenetic modifications including DNA methylation, histone modifications and regulation of non-coding RNA play an important role in AFB1-induced disease and carcinogenesis. To better understand the evidence for AFB1-induced epigenetic alterations and the potential mechanisms of the toxicity of AFB1, we conducted a review of published studies of AFB1-induced epigenetic alterations.
Collapse
Affiliation(s)
- Yaqi Dai
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, 100083, Beijing, China; Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, 100083, Beijing, China
| | - Kunlun Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, 100083, Beijing, China; Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, 100083, Beijing, China; The Supervision, Inspection and Testing Center of Genetically Modified Organisms, Ministry of Agriculture, 100083, Beijing, China
| | - Boyang Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, 100083, Beijing, China; Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, 100083, Beijing, China
| | - Liye Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, 100083, Beijing, China; Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, 100083, Beijing, China
| | - Wentao Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, 100083, Beijing, China; Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, 100083, Beijing, China; The Supervision, Inspection and Testing Center of Genetically Modified Organisms, Ministry of Agriculture, 100083, Beijing, China.
| |
Collapse
|
48
|
Oncopig Soft-Tissue Sarcomas Recapitulate Key Transcriptional Features of Human Sarcomas. Sci Rep 2017; 7:2624. [PMID: 28572589 PMCID: PMC5453942 DOI: 10.1038/s41598-017-02912-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/20/2017] [Indexed: 01/03/2023] Open
Abstract
Human soft-tissue sarcomas (STS) are rare mesenchymal tumors with a 5-year survival rate of 50%, highlighting the need for further STS research. Research has been hampered by limited human sarcoma cell line availability and the large number of STS subtypes, making development of STS cell lines and animal models representative of the diverse human STS subtypes critical. Pigs represent ideal human disease models due to their similar size, anatomy, metabolism, and genetics compared to humans. The Oncopig encodes inducible KRASG12D and TP53R167H transgenes, allowing for STS modeling in a spatial and temporal manner. This study utilized Oncopig STS cell line (fibroblast) and tumor (leiomyosarcoma) RNA-seq data to compare Oncopig and human STS expression profiles. Altered expression of 3,360 and 7,652 genes was identified in Oncopig STS cell lines and leiomyosarcomas, respectively. Transcriptional hallmarks of human STS were observed in Oncopig STS, including altered TP53 signaling, Wnt signaling activation, and evidence of epigenetic reprogramming. Furthermore, master regulators of Oncopig STS expression were identified, including FOSL1, which was previously identified as a potential human STS therapeutic target. These results demonstrate the Oncopig STS model’s ability to mimic human STS transcriptional profiles, providing a valuable resource for sarcoma research and cell line development.
Collapse
|
49
|
Nass N, Walter S, Jechorek D, Weissenborn C, Ignatov A, Haybaeck J, Sel S, Kalinski T. High neuronatin (NNAT) expression is associated with poor outcome in breast cancer. Virchows Arch 2017; 471:23-30. [DOI: 10.1007/s00428-017-2154-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/28/2017] [Accepted: 05/15/2017] [Indexed: 12/14/2022]
|
50
|
Ratovitski EA. Anticancer Natural Compounds as Epigenetic Modulators of Gene Expression. Curr Genomics 2017; 18:175-205. [PMID: 28367075 PMCID: PMC5345332 DOI: 10.2174/1389202917666160803165229] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/24/2015] [Accepted: 11/29/2015] [Indexed: 11/30/2022] Open
Abstract
Accumulating evidence shows that hallmarks of cancer include: "genetic and epigenetic alterations leading to inactivation of cancer suppressors, overexpression of oncogenes, deregulation of intracellular signaling cascades, alterations of cancer cell metabolism, failure to undergo cancer cell death, induction of epithelial to mesenchymal transition, invasiveness, metastasis, deregulation of immune response and changes in cancer microenvironment, which underpin cancer development". Natural compounds as bioactive ingredients isolated from natural sources (plants, fungi, marine life forms) have revolutionized the field of anticancer therapeutics and rapid developments in preclinical studies are encouraging. Natural compounds could affect the epigenetic molecular mechanisms that modulate gene expression, as well as DNA damage and repair mechanisms. The current review will describe the latest achievements in using naturally produced compounds targeting epigenetic regulators and modulators of gene transcription in vitro and in vivo to generate novel anticancer therapeutics.
Collapse
Affiliation(s)
- Edward A. Ratovitski
- Head and Neck Cancer Research Division, Department of Otolaryngology/Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|