1
|
Karkar S, Sharma A, Herrmann C, Blum Y, Richard M. DECOMICS, a shiny application for unsupervised cell type deconvolution and biological interpretation of bulk omic data. BIOINFORMATICS ADVANCES 2024; 4:vbae136. [PMID: 39411450 PMCID: PMC11479579 DOI: 10.1093/bioadv/vbae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/18/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Summary Unsupervised deconvolution algorithms are often used to estimate cell composition from bulk tissue samples. However, applying cell-type deconvolution and interpreting the results remain a challenge, even more without prior training in bioinformatics. Here, we propose a tool for estimating and identifying cell type composition from bulk transcriptomes or methylomes. DECOMICS is a shiny-web application dedicated to unsupervised deconvolution approaches of bulk omic data. It provides (i) a variety of existing algorithms to perform deconvolution on the gene expression or methylation-level matrix, (ii) an enrichment analysis module to aid biological interpretation of the deconvolved components, based on enrichment analysis, and (iii) some visualization tools. Input data can be downloaded in csv format and preprocessed in the web application (normalization, transformation, and feature selection). The results of the deconvolution, enrichment, and visualization processes can be downloaded. Availability and implementation DECOMICS is an R-shiny web application that can be launched (i) directly from a local R session using the R package available here: https://gitlab.in2p3.fr/Magali.Richard/decomics (either by installing it locally or via a virtual machine and a Docker image that we provide); or (ii) in the Biosphere-IFB Clouds Federation for Life Science, a multi-cloud environment scalable for high-performance computing: https://biosphere.france-bioinformatique.fr/catalogue/appliance/193/.
Collapse
Affiliation(s)
- Slim Karkar
- IBGC, UMR 5095, University of Bordeaux, CNRS, Bordeaux Bioinformatic Center, Bordeaux 33077, France
| | - Ashwini Sharma
- Health Data Science Unit, Medical Faculty Heidelberg and BioQuant, Heidelberg 69120, Germany
| | - Carl Herrmann
- Health Data Science Unit, Medical Faculty Heidelberg and BioQuant, Heidelberg 69120, Germany
| | - Yuna Blum
- IGDR (Institut de Genetique et Developpement de Rennes), UMR 6290, ERL U1305, Equipe Labellisée Ligue Nationale contre le Cancer, University of Rennes, CNRS, INSERM, Rennes 35000, France
| | - Magali Richard
- TIMC, UMR 5525, Université Grenoble Alpes, CNRS, Grenoble F-38700, France
| |
Collapse
|
2
|
Bernatz S, Reddin IG, Fenton TR, Vogl TJ, Wild PJ, Köllermann J, Mandel P, Wenzel M, Hoeh B, Mahmoudi S, Koch V, Grünewald LD, Hammerstingl R, Döring C, Harter PN, Weber KJ. Epigenetic profiling of prostate cancer reveals potential prognostic signatures. J Cancer Res Clin Oncol 2024; 150:396. [PMID: 39180680 PMCID: PMC11344710 DOI: 10.1007/s00432-024-05921-0] [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: 07/03/2024] [Accepted: 08/11/2024] [Indexed: 08/26/2024]
Abstract
PURPOSE While epigenetic profiling discovered biomarkers in several tumor entities, its application in prostate cancer is still limited. We explored DNA methylation-based deconvolution of benign and malignant prostate tissue for biomarker discovery and the potential of radiomics as a non-invasive surrogate. METHODS We retrospectively included 30 patients (63 [58-79] years) with prostate cancer (PCa) who had a multiparametric MRI of the prostate before radical prostatectomy between 2014 and 2019. The control group comprised four patients with benign prostate tissue adjacent to the PCa lesions and four patients with benign prostatic hyperplasia. Tissue punches of all lesions were obtained. DNA methylation analysis and reference-free in silico deconvolution were conducted to retrieve Latent Methylation Components (LCMs). LCM-based clustering was analyzed for cellular composition and correlated with clinical disease parameters. Additionally, PCa and adjacent benign lesions were analyzed using radiomics to predict the epigenetic signatures non-invasively. RESULTS LCMs identified two clusters with potential prognostic impact. Cluster one was associated with malignant prostate tissue (p < 0.001) and reduced immune-cell-related signatures (p = 0.004) of CD19 and CD4 cells. Cluster one comprised exclusively malignant prostate tissue enriched for significant prostate cancer and advanced tumor stages (p < 0.03 for both). No radiomics model could non-invasively predict the epigenetic clusters. CONCLUSION Epigenetic clusters were associated with prognostically and clinically relevant metrics in prostate cancer. Further, immune cell-related signatures differed significantly between prognostically favorable and unfavorable clusters. Further research is necessary to explore potential diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Simon Bernatz
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute for Pathology, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
| | - Ian G Reddin
- School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Tim R Fenton
- School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Thomas J Vogl
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
| | - Peter J Wild
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute for Pathology, Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main, Germany
| | - Jens Köllermann
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute for Pathology, Frankfurt am Main, Germany
| | - Philipp Mandel
- Goethe University Frankfurt, University Hospital, Department of Urology, Frankfurt am Main, Germany
| | - Mike Wenzel
- Goethe University Frankfurt, University Hospital, Department of Urology, Frankfurt am Main, Germany
| | - Benedikt Hoeh
- Goethe University Frankfurt, University Hospital, Department of Urology, Frankfurt am Main, Germany
| | - Scherwin Mahmoudi
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
| | - Vitali Koch
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
| | - Leon D Grünewald
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
| | - Renate Hammerstingl
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
| | - Claudia Döring
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute for Pathology, Frankfurt am Main, Germany
| | - Patrick N Harter
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between DKFZ and University/University Hospital, LMU Munich, Munich, Germany
| | - Katharina J Weber
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany.
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany.
| |
Collapse
|
3
|
Chepeleva M, Kaoma T, Zinovyev A, Toth R, Nazarov PV. consICA: an R package for robust reference-free deconvolution of multi-omics data. BIOINFORMATICS ADVANCES 2024; 4:vbae102. [PMID: 39027644 PMCID: PMC11257712 DOI: 10.1093/bioadv/vbae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/25/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Motivation Deciphering molecular signals from omics data helps understanding cellular processes and disease progression. Effective algorithms for extracting these signals are essential, with a strong emphasis on robustness and reproducibility. Results R/Bioconductor package consICA implements consensus independent component analysis (ICA)-a data-driven deconvolution method to decompose heterogeneous omics data and extract features suitable for patient stratification and multimodal data integration. The method separates biologically relevant molecular signals from technical effects and provides information about the cellular composition and biological processes. Build-in annotation, survival analysis, and report generation provide useful tools for the interpretation of extracted signals. The implementation of parallel computing in the package ensures efficient analysis using modern multicore systems. The package offers a reproducible and efficient data-driven solution for the analysis of complex molecular profiles, with significant implications for cancer research. Availability and implementation The package is implemented in R and available under MIT license at Bioconductor (https://bioconductor.org/packages/consICA) or at GitHub (https://github.com/biomod-lih/consICA).
Collapse
Affiliation(s)
- Maryna Chepeleva
- Multiomics Data Science Research Group, Department of Cancer Research, Luxembourg Institute of Health, Strassen L-1445, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette L-4365, Luxembourg
| | - Tony Kaoma
- Bioinformatics and AI Unit, Department of Medical Informatics, Luxembourg Institute of Health, Strassen L-1445, Luxembourg
| | | | - Reka Toth
- Multiomics Data Science Research Group, Department of Cancer Research, Luxembourg Institute of Health, Strassen L-1445, Luxembourg
- Bioinformatics and AI Unit, Department of Medical Informatics, Luxembourg Institute of Health, Strassen L-1445, Luxembourg
| | - Petr V Nazarov
- Multiomics Data Science Research Group, Department of Cancer Research, Luxembourg Institute of Health, Strassen L-1445, Luxembourg
- Bioinformatics and AI Unit, Department of Medical Informatics, Luxembourg Institute of Health, Strassen L-1445, Luxembourg
| |
Collapse
|
4
|
De Ridder K, Che H, Leroy K, Thienpont B. Benchmarking of methods for DNA methylome deconvolution. Nat Commun 2024; 15:4134. [PMID: 38755121 PMCID: PMC11099101 DOI: 10.1038/s41467-024-48466-z] [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: 10/20/2023] [Accepted: 04/30/2024] [Indexed: 05/18/2024] Open
Abstract
Defining the number and abundance of different cell types in tissues is important for understanding disease mechanisms as well as for diagnostic and prognostic purposes. Typically, this is achieved by immunohistological analyses, cell sorting, or single-cell RNA-sequencing. Alternatively, cell-specific DNA methylome information can be leveraged to deconvolve cell fractions from a bulk DNA mixture. However, comprehensive benchmarking of deconvolution methods and modalities was not yet performed. Here we evaluate 16 deconvolution algorithms, developed either specifically for DNA methylome data or more generically. We assess the performance of these algorithms, and the effect of normalization methods, while modeling variables that impact deconvolution performance, including cell abundance, cell type similarity, reference panel size, method for methylome profiling (array or sequencing), and technical variation. We observe differences in algorithm performance depending on each these variables, emphasizing the need for tailoring deconvolution analyses. The complexity of the reference, marker selection method, number of marker loci and, for sequencing-based assays, sequencing depth have a marked influence on performance. By developing handles to select the optimal analysis configuration, we provide a valuable source of information for studies aiming to deconvolve array- or sequencing-based methylation data.
Collapse
Affiliation(s)
- Kobe De Ridder
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
| | - Huiwen Che
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
| | - Kaat Leroy
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
| | - Bernard Thienpont
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium.
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, 3000, Leuven, Belgium.
- KU Leuven Cancer Institute (LKI), KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
5
|
Muench A, Teichmann D, Spille D, Kuzman P, Perez E, May SA, Mueller WC, Kombos T, Nazari-Dehkordi S, Onken J, Vajkoczy P, Ntoulias G, Bettencourt C, von Deimling A, Paulus W, Heppner FL, Koch A, Capper D, Kaul D, Thomas C, Schweizer L. A Novel Type of IDH-wildtype Glioma Characterized by Gliomatosis Cerebri-like Growth Pattern, TERT Promoter Mutation, and Distinct Epigenetic Profile. Am J Surg Pathol 2023; 47:1364-1375. [PMID: 37737691 DOI: 10.1097/pas.0000000000002118] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Diffuse gliomas in adults encompass a heterogenous group of central nervous system neoplasms. In recent years, extensive (epi-)genomic profiling has identified several glioma subgroups characterized by distinct molecular characteristics, most importantly IDH1/2 and histone H3 mutations. A group of 16 diffuse gliomas classified as "adult-type diffuse high-grade glioma, IDH-wildtype, subtype F (HGG-F)" was identified by the DKFZ v12.5 Brain Tumor Classifier . Histopathologic characterization, exome sequencing, and review of clinical data was performed in all cases. Based on unsupervised t -distributed stochastic neighbor embedding and clustering analysis of genome-wide DNA methylation data, HGG-F shows distinct epigenetic profiles separate from established central nervous system tumors. Exome sequencing demonstrated frequent TERT promoter (12/15 cases), PIK3R1 (11/16), and TP53 mutations (5/16). Radiologic characteristics were reminiscent of gliomatosis cerebri in 9/14 cases (64%). Histopathologically, most cases were classified as diffuse gliomas (7/16, 44%) or were suspicious for the infiltration zone of a diffuse glioma (5/16, 31%). None of the cases demonstrated microvascular proliferation or necrosis. Outcome of 14 patients with follow-up data was better compared to IDH-wildtype glioblastomas with a median progression-free survival of 58 months and overall survival of 74 months (both P <0.0001). Our series represents a novel type of adult-type diffuse glioma with distinct molecular and clinical features. Importantly, we provide evidence that TERT promoter mutations in diffuse gliomas without further morphologic or molecular signs of high-grade glioma should be interpreted in the context of the clinicoradiologic presentation as well as epigenetic profile and may not be suitable as a standalone marker for glioblastoma, IDH-wildtype.
Collapse
Affiliation(s)
- Amos Muench
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main
| | | | | | - Peter Kuzman
- Institute of Neuropathology, University Hospital Leipzig, Leipzig
| | | | - Sven-Axel May
- Department of Neurosurgery, Klinikum Chemnitz, Chemnitz
| | - Wolf C Mueller
- Institute of Neuropathology, University Hospital Leipzig, Leipzig
| | | | | | | | | | - Georgios Ntoulias
- Department of Neurosurgery, Schlosspark-Klinik Charlottenburg, Berlin
| | - Conceição Bettencourt
- Queen Square Brain Bank, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Werner Paulus
- Institute of Neuropathology, University Hospital Münster, Münster
| | - Frank L Heppner
- Departments of Neuropathology
- Cluster of Excellence, NeuroCure
- German Center for Neurodegenerative Diseases (DZNE)
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ)
| | - Arend Koch
- Departments of Neuropathology
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ)
| | - David Capper
- Departments of Neuropathology
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ)
| | - David Kaul
- Radiation Oncology and Radiotherapy, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin
| | - Christian Thomas
- Institute of Neuropathology, University Hospital Münster, Münster
| | - Leonille Schweizer
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main
- Frankfurt Cancer Institute (FCI), Frankfurt am Main
- Departments of Neuropathology
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ)
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
6
|
Jain SS, McNamara ME, Varghese RS, Ressom HW. Deconvolution of immune cell composition and biological age of hepatocellular carcinoma using DNA methylation. Methods 2023; 218:125-132. [PMID: 37574160 PMCID: PMC10529003 DOI: 10.1016/j.ymeth.2023.08.004] [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: 04/21/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) has been an approved indication for the administration of immunotherapy since 2017, but biomarkers that predict therapeutic response have remained limited. Understanding and characterizing the tumor immune microenvironment enables better classification of these tumors and may reveal biomarkers that predict immunotherapeutic efficacy. In this paper, we applied a cell-type deconvolution algorithm using DNA methylation array data to investigate the composition of the tumor microenvironment in HCC. Using publicly available and in-house datasets with a total cohort size of 57 patients, each with tumor and matched normal tissue samples, we identified key differences in immune cell composition. We found that NK cell abundance was significantly decreased in HCC tumors compared to adjacent normal tissue. We also applied DNA methylation "clocks" which estimate phenotypic aging and compared these findings to expression-based determinations of cellular senescence. Senescence and epigenetic aging were significantly increased in HCC tumors, and the degree of age acceleration and senescence was strongly associated with decreased NK cell abundance. In summary, we found that NK cell infiltration in the tumor microenvironment is significantly diminished, and that this loss of NK abundance is strongly associated with increased senescence and age-related phenotype. These findings point to key interactions between NK cells and the senescent tumor microenvironment and offer insights into the pathogenesis of HCC as well as potential biomarkers of therapeutic efficacy.
Collapse
Affiliation(s)
- Sidharth S Jain
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Megan E McNamara
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Rency S Varghese
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Habtom W Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
7
|
Bell-Glenn S, Salas LA, Molinaro AM, Butler RA, Christensen BC, Kelsey KT, Wiencke JK, Koestler DC. Calculating detection limits and uncertainty of reference-based deconvolution of whole-blood DNA methylation data. Epigenomics 2023; 15:435-451. [PMID: 37337720 PMCID: PMC10308256 DOI: 10.2217/epi-2023-0006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/16/2023] [Indexed: 06/21/2023] Open
Abstract
DNA methylation (DNAm)-based cell mixture deconvolution (CMD) has become a quintessential part of epigenome-wide association studies where DNAm is profiled in heterogeneous tissue types. Despite being introduced over a decade ago, detection limits, which represent the smallest fraction of a cell type in a mixed biospecimen that can be reliably detected, have yet to be determined in the context of DNAm-based CMD. Moreover, there has been little attention given to approaches for quantifying the uncertainty associated with DNAm-based CMD. Here, analytical frameworks for determining both cell-specific limits of detection and quantification of uncertainty associated with DNAm-based CMD are described. This work may contribute to improved rigor, reproducibility and replicability of epigenome-wide association studies involving CMD.
Collapse
Affiliation(s)
- Shelby Bell-Glenn
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03756, USA
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Rondi A Butler
- Departments of Epidemiology & Pathology & Laboratory Medicine, Brown University, Providence, RI 02912, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03756, USA
- Department of Molecular & Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
- Department of Community & Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Karl T Kelsey
- Departments of Epidemiology & Pathology & Laboratory Medicine, Brown University, Providence, RI 02912, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Devin C Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
8
|
Zhou X, Cheng Z, Dong M, Liu Q, Yang W, Liu M, Tian J, Cheng W. Tumor fractions deciphered from circulating cell-free DNA methylation for cancer early diagnosis. Nat Commun 2022; 13:7694. [PMID: 36509772 PMCID: PMC9744803 DOI: 10.1038/s41467-022-35320-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Tumor-derived circulating cell-free DNA (cfDNA) provides critical clues for cancer early diagnosis, yet it often suffers from low sensitivity. Here, we present a cancer early diagnosis approach using tumor fractions deciphered from circulating cfDNA methylation signatures. We show that the estimated fractions of tumor-derived cfDNA from cancer patients increase significantly as cancer progresses in two independent datasets. Employing the predicted tumor fractions, we establish a Bayesian diagnostic model in which training samples are only derived from late-stage patients and healthy individuals. When validated on early-stage patients and healthy individuals, this model exhibits a sensitivity of 86.1% for cancer early detection and an average accuracy of 76.9% for tumor localization at a specificity of 94.7%. By highlighting the potential of tumor fractions on cancer early diagnosis, our approach can be further applied to cancer screening and tumor progression monitoring.
Collapse
Affiliation(s)
- Xiao Zhou
- grid.12527.330000 0001 0662 3178Department of Automation, Tsinghua University, Beijing, 100084 China
| | - Zhen Cheng
- grid.12527.330000 0001 0662 3178Department of Automation, Tsinghua University, Beijing, 100084 China
| | - Mingyu Dong
- grid.12527.330000 0001 0662 3178Department of Automation, Tsinghua University, Beijing, 100084 China
| | - Qi Liu
- grid.12527.330000 0001 0662 3178Department of Automation, Tsinghua University, Beijing, 100084 China
| | - Weiyang Yang
- grid.12527.330000 0001 0662 3178Department of Automation, Tsinghua University, Beijing, 100084 China
| | - Min Liu
- grid.12527.330000 0001 0662 3178Department of Automation, Tsinghua University, Beijing, 100084 China ,grid.413405.70000 0004 1808 0686Institute for Healthcare Artificial Intelligence Application, Guangdong Second Provincial General Hospital, Guangzhou, 510317 China
| | - Junzhang Tian
- grid.413405.70000 0004 1808 0686Institute for Healthcare Artificial Intelligence Application, Guangdong Second Provincial General Hospital, Guangzhou, 510317 China
| | - Weibin Cheng
- grid.413405.70000 0004 1808 0686Institute for Healthcare Artificial Intelligence Application, Guangdong Second Provincial General Hospital, Guangzhou, 510317 China
| |
Collapse
|
9
|
Zaremba A, Jansen P, Murali R, Mayakonda A, Riedel A, Philip M, Rose C, Schaller J, Müller H, Kutzner H, Möller I, Stadtler N, Kretz J, Sucker A, Bankfalvi A, Livingstone E, Zimmer L, Horn S, Paschen A, Plass C, Schadendorf D, Hadaschik E, Lutsik P, Griewank K. Genetic and methylation profiles distinguish benign, malignant and spitzoid melanocytic tumors. Int J Cancer 2022; 151:1542-1554. [PMID: 35737508 PMCID: PMC9474633 DOI: 10.1002/ijc.34187] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/26/2022] [Accepted: 05/04/2022] [Indexed: 11/07/2022]
Abstract
Accurate classification of melanocytic tumors is important for prognostic evaluation, treatment and follow-up protocols of patients. The majority of melanocytic proliferations can be classified solely based on clinical and pathological criteria, however in select cases a definitive diagnostic assessment remains challenging and additional diagnostic biomarkers would be advantageous. We analyzed melanomas, nevi, Spitz nevi and atypical spitzoid tumors using parallel sequencing (exons of 611 genes and 507 gene translocation analysis) and methylation arrays (850k Illumina EPIC). By combining detailed genetic and epigenetic analysis with reference-based and reference-free DNA methylome deconvolution we compared Spitz nevi to nevi and melanoma and assessed the potential for these methods in classifying challenging spitzoid tumors. Results were correlated with clinical and histologic features. Spitz nevi were found to cluster independently of nevi and melanoma and demonstrated a different mutation profile. Multiple copy number alterations and TERT promoter mutations were identified only in melanomas. Genome-wide methylation in Spitz nevi was comparable to benign nevi while the Leukocytes UnMethylation for Purity (LUMP) algorithm in Spitz nevi was comparable to melanoma. Histologically difficult to classify Spitz tumor cases were assessed which, based on methylation arrays, clustered between Spitz nevi and melanoma and in terms of genetic profile or copy number variations demonstrated worrisome features suggesting a malignant neoplasm. Comprehensive sequencing and methylation analysis verify Spitz nevi as an independent melanocytic entity distinct from both nevi and melanoma. Combined genetic and methylation assays can offer additional insights in diagnosing difficult to classify Spitzoid tumors.
Collapse
Affiliation(s)
- Anne Zaremba
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Philipp Jansen
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Rajmohan Murali
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Anand Mayakonda
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz International Graduate School for Cancer Research, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Anna Riedel
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz International Graduate School for Cancer Research, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Manuel Philip
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | | | | | | | - Heinz Kutzner
- Dermatopathologie Friedrichshafen, Medical faculty of the University Leipzig, Leipzig, Germany
| | - Inga Möller
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Nadine Stadtler
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Julia Kretz
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Agnes Bankfalvi
- Department of Pathology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Susanne Horn
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
- Rudolf-Schönheimer-Institute of Biochemistry, Medical faculty of the University Leipzig, Leipzig, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Eva Hadaschik
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Pavlo Lutsik
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Germany, and German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
10
|
Genetic and Methylation Analysis of CTNNB1 in Benign and Malignant Melanocytic Lesions. Cancers (Basel) 2022; 14:cancers14174066. [PMID: 36077603 PMCID: PMC9454999 DOI: 10.3390/cancers14174066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Recurrent CTNNB1 exon 3 mutations have been recognized in the distinct group of melanocytic tumors showing deep penetrating nevus-like morphology and in 1–2% of advanced melanoma. We performed a detailed genetic analysis of difficult-to-classify nevi and melanomas with CTNNB1 mutations and found that benign tumors (nevi) show characteristic morphological, genetic and epigenetic traits, which distinguish them from other nevi and melanoma. Malignant CTNNB1-mutant tumors (melanoma) demonstrated a different genetic profile, grouping clearly with other non-CTNNB1 melanomas in methylation assays. To further evaluate the role of CTNNB1 mutations in melanoma, we assessed a large cohort of clinically sequenced melanomas, identifying 38 tumors with CTNNB1 exon 3 mutations, including recurrent S45 (n = 13, 34%), G34 (n = 5, 13%), and S27 (n = 5, 13%) mutations. Locations and histological subtype of CTNNB1-mutated melanoma varied; none were reported as showing deep penetrating nevus-like morphology. The most frequent concurrent activating mutations were BRAF V600 (55%) and NRAS Q61 (34%). Abstract Melanocytic neoplasms have been genetically characterized in detail during the last decade. Recurrent CTNNB1 exon 3 mutations have been recognized in the distinct group of melanocytic tumors showing deep penetrating nevus-like morphology. In addition, they have been identified in 1–2% of advanced melanoma. Performing a detailed genetic analysis of difficult-to-classify nevi and melanomas with CTNNB1 mutations, we found that benign tumors (nevi) show characteristic morphological, genetic and epigenetic traits, which distinguish them from other nevi and melanoma. Malignant CTNNB1-mutant tumors (melanomas) demonstrated a different genetic profile, instead grouping clearly with other non-CTNNB1 melanomas in methylation assays. To further evaluate the role of CTNNB1 mutations in melanoma, we assessed a large cohort of clinically sequenced melanomas, identifying 38 tumors with CTNNB1 exon 3 mutations, including recurrent S45 (n = 13, 34%), G34 (n = 5, 13%), and S27 (n = 5, 13%) mutations. Locations and histological subtype of CTNNB1-mutated melanoma varied; none were reported as showing deep penetrating nevus-like morphology. The most frequent concurrent activating mutations were BRAF V600 (n = 21, 55%) and NRAS Q61 (n = 13, 34%). In our cohort, four of seven (58%) and one of nine (11%) patients treated with targeted therapy (BRAF and MEK Inhibitors) or immune-checkpoint therapy, respectively, showed disease control (partial response or stable disease). In summary, CTNNB1 mutations are associated with a unique melanocytic tumor type in benign tumors (nevi), which can be applied in a diagnostic setting. In advanced disease, no clear characteristics distinguishing CTNNB1-mutant from other melanomas were observed; however, studies of larger, optimally prospective, cohorts are warranted.
Collapse
|
11
|
Chen Y, Toth R, Chocarro S, Weichenhan D, Hey J, Lutsik P, Sawall S, Stathopoulos GT, Plass C, Sotillo R. Club cells employ regeneration mechanisms during lung tumorigenesis. Nat Commun 2022; 13:4557. [PMID: 35931677 PMCID: PMC9356049 DOI: 10.1038/s41467-022-32052-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 07/12/2022] [Indexed: 11/09/2022] Open
Abstract
The high plasticity of lung epithelial cells, has for many years, confounded the correct identification of the cell-of-origin of lung adenocarcinoma (LUAD), one of the deadliest malignancies worldwide. Here, we employ lineage-tracing mouse models to investigate the cell of origin of Eml4-Alk LUAD, and show that Club and Alveolar type 2 (AT2) cells give rise to tumours. We focus on Club cell originated tumours and find that Club cells experience an epigenetic switch by which they lose their lineage fidelity and gain an AT2-like phenotype after oncogenic transformation. Single-cell transcriptomic analyses identified two trajectories of Club cell evolution which are similar to the ones used during lung regeneration, suggesting that lung epithelial cells leverage on their plasticity and intrinsic regeneration mechanisms to give rise to a tumour. Together, this study highlights the role of Club cells in LUAD initiation, identifies the mechanism of Club cell lineage infidelity, confirms the presence of these features in human tumours, and unveils key mechanisms conferring LUAD heterogeneity.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Reka Toth
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Bioinformatics Platform, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Sara Chocarro
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Ruprecht Karl University of Heidelberg, Heidelberg, Germany
| | - Dieter Weichenhan
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Joschka Hey
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Ruprecht Karl University of Heidelberg, Heidelberg, Germany
| | - Pavlo Lutsik
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Stefan Sawall
- X-Ray Imaging and CT, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Georgios T Stathopoulos
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), Helmholtz Center Munich-German Research Center for Environmental Health (HMGU), Max-Lebsche-Platz 31, 81377, Munich, Bavaria, Germany.,German Center for Lung Research (DZL), Heidelberg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,German Center for Lung Research (DZL), Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TRLC), Heidelberg, Germany.,German Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany
| | - Rocio Sotillo
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany. .,German Center for Lung Research (DZL), Heidelberg, Germany. .,Translational Lung Research Center Heidelberg (TRLC), Heidelberg, Germany. .,German Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany.
| |
Collapse
|
12
|
Jeong Y, de Andrade e Sousa LB, Thalmeier D, Toth R, Ganslmeier M, Breuer K, Plass C, Lutsik P. Systematic evaluation of cell-type deconvolution pipelines for sequencing-based bulk DNA methylomes. Brief Bioinform 2022; 23:bbac248. [PMID: 35794707 PMCID: PMC9294431 DOI: 10.1093/bib/bbac248] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/18/2022] [Accepted: 05/26/2022] [Indexed: 11/18/2022] Open
Abstract
DNA methylation analysis by sequencing is becoming increasingly popular, yielding methylomes at single-base pair and single-molecule resolution. It has tremendous potential for cell-type heterogeneity analysis using intrinsic read-level information. Although diverse deconvolution methods were developed to infer cell-type composition based on bulk sequencing-based methylomes, systematic evaluation has not been performed yet. Here, we thoroughly benchmark six previously published methods: Bayesian epiallele detection, DXM, PRISM, csmFinder+coMethy, ClubCpG and MethylPurify, together with two array-based methods, MeDeCom and Houseman, as a comparison group. Sequencing-based deconvolution methods consist of two main steps, informative region selection and cell-type composition estimation, thus each was individually assessed. With this elaborate evaluation, we aimed to establish which method achieves the highest performance in different scenarios of synthetic bulk samples. We found that cell-type deconvolution performance is influenced by different factors depending on the number of cell types within the mixture. Finally, we propose a best-practice deconvolution strategy for sequencing data and point out limitations that need to be handled. Array-based methods-both reference-based and reference-free-generally outperformed sequencing-based methods, despite the absence of read-level information. This implies that the current sequencing-based methods still struggle with correctly identifying cell-type-specific signals and eliminating confounding methylation patterns, which needs to be handled in future studies.
Collapse
Affiliation(s)
- Yunhee Jeong
- Division of Cancer Epigenomics, German Cancer Research (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Faculty of Mathematics and Informatics, Heidelberg University, Im Neuenheimer Feld 205, 69120, Heidelberg, Germany
| | | | - Dominik Thalmeier
- Helmholtz AI, Helmholtz Zentrum München, Ingolstädter Landstraβ e 1, 85764, Neuherberg, Germany
| | - Reka Toth
- Division of Cancer Epigenomics, German Cancer Research (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Marlene Ganslmeier
- Division of Cancer Epigenomics, German Cancer Research (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Kersten Breuer
- Division of Cancer Epigenomics, German Cancer Research (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Pavlo Lutsik
- Division of Cancer Epigenomics, German Cancer Research (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| |
Collapse
|
13
|
Woltering N, Albers A, Müther M, Stummer W, Paulus W, Hasselblatt M, Holling M, Thomas C. DNA
methylation profiling of central nervous system hemangioblastomas identifies two distinct subgroups. Brain Pathol 2022; 32:e13083. [PMID: 35637626 PMCID: PMC9616087 DOI: 10.1111/bpa.13083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/10/2022] [Indexed: 12/01/2022] Open
Abstract
Hemangioblastomas (HBs) of the central nervous system are highly vascular neoplasms that occur sporadically or as a manifestation of von Hippel–Lindau (VHL) disease. Despite their benign nature, HBs are clinically heterogeneous and can be associated with significant morbidity due to mass effects of peritumoral cysts or tumor progression. Underlying molecular factors involved in HB tumor biology remain elusive. We investigated genome‐wide DNA methylation profiles and clinical and histopathological features in a series of 47 HBs from 42 patients, including 28 individuals with VHL disease. Thirty tumors occurred in the cerebellum, 8 in the brainstem and 8 HBs were of spinal location, while 1 HB was located in the cerebrum. Histologically, 12 HBs (26%) belonged to the cellular subtype and exclusively occurred in the cerebellum, whereas 35 HBs were reticular (74%). Unsupervised clustering and dimensionality reduction of DNA methylation profiles revealed two distinct subgroups. Methylation cluster 1 comprised 30 HBs of mainly cerebellar location (29/30, 97%), whereas methylation cluster 2 contained 17 HBs predominantly located in non‐cerebellar compartments (16/17, 94%). The sum of chromosomal regions being affected by copy‐number alterations was significantly higher in methylation cluster 1 compared to cluster 2 (mean 262 vs. 109 Mb, p = 0.001). Of note, loss of chromosome 6 occurred in 9/30 tumors (30%) of methylation cluster 1 and was not observed in cluster 2 tumors (p = 0.01). No relevant methylation differences between sporadic and VHL‐related HBs or cystic and non‐cystic HBs could be detected. Deconvolution of the bulk DNA methylation profiles revealed four methylation components that were associated with the two methylation clusters suggesting cluster‐specific cell‐type compositions. In conclusion, methylation profiling of HBs reveals 2 distinct subgroups that mainly associate with anatomical location, cytogenetic profiles and differences in cell type composition, potentially reflecting different cells of origin.
Collapse
Affiliation(s)
- Niklas Woltering
- Institute of Neuropathology University Hospital Münster Münster Germany
| | - Anne Albers
- Institute of Neuropathology University Hospital Münster Münster Germany
| | - Michael Müther
- Department of Neurosurgery University Hospital Münster Münster Germany
| | - Walter Stummer
- Department of Neurosurgery University Hospital Münster Münster Germany
| | - Werner Paulus
- Institute of Neuropathology University Hospital Münster Münster Germany
| | | | - Markus Holling
- Department of Neurosurgery University Hospital Münster Münster Germany
| | - Christian Thomas
- Institute of Neuropathology University Hospital Münster Münster Germany
| |
Collapse
|
14
|
Bell-Glenn S, Thompson JA, Salas LA, Koestler DC. A Novel Framework for the Identification of Reference DNA Methylation Libraries for Reference-Based Deconvolution of Cellular Mixtures. FRONTIERS IN BIOINFORMATICS 2022; 2. [PMID: 35419567 PMCID: PMC9004796 DOI: 10.3389/fbinf.2022.835591] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Reference-based deconvolution methods use reference libraries of cell-specific DNA methylation (DNAm) measurements as a means toward deconvoluting cell proportions in heterogeneous biospecimens (e.g., whole-blood). As the accuracy of such methods depends highly on the CpG loci comprising the reference library, recent research efforts have focused on the selection of libraries to optimize deconvolution accuracy. While existing approaches for library selection work extremely well, the best performing approaches require a training data set consisting of both DNAm profiles over a heterogeneous cell population and gold-standard measurements of cell composition (e.g., flow cytometry) in the same samples. Here, we present a framework for reference library selection without a training dataset (RESET) and benchmark it against the Legacy method (minfi:pickCompProbes), where libraries are constructed based on a pre-specified number of cell-specific differentially methylated loci (DML). RESET uses a modified version of the Dispersion Separability Criteria (DSC) for comparing different libraries and has four main steps: 1) identify a candidate set of cell-specific DMLs, 2) randomly sample DMLs from the candidate set, 3) compute the Modified DSC of the selected DMLs, and 4) update the selection probabilities of DMLs based on their contribution to the Modified DSC. Steps 2–4 are repeated many times and the library with the largest Modified DSC is selected for subsequent reference-based deconvolution. We evaluated RESET using several publicly available datasets consisting of whole-blood DNAm measurements with corresponding measurements of cell composition. We computed the RMSE and R2 between the predicted cell proportions and their measured values. RESET outperformed the Legacy approach in selecting libraries that improve the accuracy of deconvolution estimates. Additionally, reference libraries constructed using RESET resulted in cellular composition estimates that explained more variation in DNAm as compared to the Legacy approach when evaluated in the context of epigenome-wide association studies (EWAS) of several publicly available data sets. This finding has implications for the statistical power of EWAS. RESET combats potential challenges associated with existing approaches for reference library assembly and thus, may serve as a viable strategy for library construction in the absence of a training data set.
Collapse
Affiliation(s)
- Shelby Bell-Glenn
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jeffrey A. Thompson
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, United States
| | - Lucas A. Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
| | - Devin C. Koestler
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, United States
- *Correspondence: Devin C. Koestler,
| |
Collapse
|
15
|
Yates J, Boeva V. Deciphering the etiology and role in oncogenic transformation of the CpG island methylator phenotype: a pan-cancer analysis. Brief Bioinform 2022; 23:6520307. [PMID: 35134107 PMCID: PMC8921629 DOI: 10.1093/bib/bbab610] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/06/2021] [Accepted: 12/30/2021] [Indexed: 12/25/2022] Open
Abstract
Numerous cancer types have shown to present hypermethylation of CpG islands, also known as a CpG island methylator phenotype (CIMP), often associated with survival variation. Despite extensive research on CIMP, the etiology of this variability remains elusive, possibly due to lack of consistency in defining CIMP. In this work, we utilize a pan-cancer approach to further explore CIMP, focusing on 26 cancer types profiled in the Cancer Genome Atlas (TCGA). We defined CIMP systematically and agnostically, discarding any effects associated with age, gender or tumor purity. We then clustered samples based on their most variable DNA methylation values and analyzed resulting patient groups. Our results confirmed the existence of CIMP in 19 cancers, including gliomas and colorectal cancer. We further showed that CIMP was associated with survival differences in eight cancer types and, in five, represented a prognostic biomarker independent of clinical factors. By analyzing genetic and transcriptomic data, we further uncovered potential drivers of CIMP and classified them in four categories: mutations in genes directly involved in DNA demethylation; mutations in histone methyltransferases; mutations in genes not involved in methylation turnover, such as KRAS and BRAF; and microsatellite instability. Among the 19 CIMP-positive cancers, very few shared potential driver events, and those drivers were only IDH1 and SETD2 mutations. Finally, we found that CIMP was strongly correlated with tumor microenvironment characteristics, such as lymphocyte infiltration. Overall, our results indicate that CIMP does not exhibit a pan-cancer manifestation; rather, general dysregulation of CpG DNA methylation is caused by heterogeneous mechanisms.
Collapse
Affiliation(s)
- Josephine Yates
- Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich 8092, Switzerland
| | - Valentina Boeva
- Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich 8092, Switzerland.,Swiss Institute for Bioinformatics (SIB), Zürich, Switzerland.,Cochin Institute, Inserm U1016, CNRS UMR 8104, Paris Descartes University UMR-S1016, Paris 75014, France
| |
Collapse
|
16
|
Scherer M, Gasparoni G, Rahmouni S, Shashkova T, Arnoux M, Louis E, Nostaeva A, Avalos D, Dermitzakis ET, Aulchenko YS, Lengauer T, Lyons PA, Georges M, Walter J. Identification of tissue-specific and common methylation quantitative trait loci in healthy individuals using MAGAR. Epigenetics Chromatin 2021; 14:44. [PMID: 34530905 PMCID: PMC8444396 DOI: 10.1186/s13072-021-00415-6] [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: 04/20/2021] [Accepted: 08/02/2021] [Indexed: 12/18/2022] Open
Abstract
Background Understanding the influence of genetic variants on DNA methylation is fundamental for the interpretation of epigenomic data in the context of disease. There is a need for systematic approaches not only for determining methylation quantitative trait loci (methQTL), but also for discriminating general from cell type-specific effects. Results Here, we present a two-step computational framework MAGAR (https://bioconductor.org/packages/MAGAR), which fully supports the identification of methQTLs from matched genotyping and DNA methylation data, and additionally allows for illuminating cell type-specific methQTL effects. In a pilot analysis, we apply MAGAR on data in four tissues (ileum, rectum, T cells, B cells) from healthy individuals and demonstrate the discrimination of common from cell type-specific methQTLs. We experimentally validate both types of methQTLs in an independent data set comprising additional cell types and tissues. Finally, we validate selected methQTLs located in the PON1, ZNF155, and NRG2 genes by ultra-deep local sequencing. In line with previous reports, we find cell type-specific methQTLs to be preferentially located in enhancer elements. Conclusions Our analysis demonstrates that a systematic analysis of methQTLs provides important new insights on the influences of genetic variants to cell type-specific epigenomic variation. Supplementary Information The online version contains supplementary material available at 10.1186/s13072-021-00415-6.
Collapse
Affiliation(s)
- Michael Scherer
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany.,Computational Biology, Max Planck Institute for Informatics, Saarland Informatics Campus, Saarbrücken, Germany.,Graduate School of Computer Science, Saarland Informatics Campus, Saarbrücken, Germany.,Department of Bioinformatics and Genomics, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Gilles Gasparoni
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany
| | - Souad Rahmouni
- Unit of Animal Genomics, GIGA-Institute & Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Tatiana Shashkova
- Kurchatov Genomics Center of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Research and Training Center on Bioinformatics, A.A. Kharkevich Institute for Information Transmission Problems, Moscow, Russia
| | - Marion Arnoux
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany
| | - Edouard Louis
- Department of Gastroenterology, Liège University Hospital, CHU Liège, Liège, Belgium
| | | | - Diana Avalos
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland.,Swiss Institute of Bioinformatics (SIB), University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics in Geneva, University of Geneva, Geneva, Switzerland
| | - Emmanouil T Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland.,Swiss Institute of Bioinformatics (SIB), University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics in Geneva, University of Geneva, Geneva, Switzerland
| | - Yurii S Aulchenko
- Kurchatov Genomics Center of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Novosibirsk State University, Novosibirsk, Russia.,Moscow Institute of Physics and Technology (State University), Moscow, Russia.,PolyKnomics BV, 's-Hertogenbosch, The Netherlands
| | - Thomas Lengauer
- Computational Biology, Max Planck Institute for Informatics, Saarland Informatics Campus, Saarbrücken, Germany
| | - Paul A Lyons
- Department of Medicine, University of Cambridge School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.,Cambridge Institute for Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, CB2 0AW, UK
| | - Michel Georges
- Unit of Animal Genomics, GIGA-Institute & Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Jörn Walter
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany.
| |
Collapse
|
17
|
Zhang D, Guo S, Schrodi SJ. Mechanisms of DNA Methylation in Virus-Host Interaction in Hepatitis B Infection: Pathogenesis and Oncogenetic Properties. Int J Mol Sci 2021; 22:9858. [PMID: 34576022 PMCID: PMC8466338 DOI: 10.3390/ijms22189858] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/10/2021] [Accepted: 09/10/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatitis B virus (HBV), the well-studied oncovirus that contributes to the majority of hepatocellular carcinomas (HCC) worldwide, can cause a severe inflammatory microenvironment leading to genetic and epigenetic changes in hepatocyte clones. HBV replication contributes to the regulation of DNA methyltransferase gene expression, particularly by X protein (HBx), and subsequent methylation changes may lead to abnormal transcription activation of adjacent genes and genomic instability. Undoubtedly, the altered expression of these genes has been known to cause diverse aspects of infected hepatocytes, including apoptosis, proliferation, reactive oxygen species (ROS) accumulation, and immune responses. Additionally, pollutant-induced DNA methylation changes and aberrant methylation of imprinted genes in hepatocytes also complicate the process of tumorigenesis. Meanwhile, hepatocytes also contribute to epigenetic modification of the viral genome to affect HBV replication or viral protein production. Meanwhile, methylation levels of HBV integrants and surrounding host regions also play crucial roles in their ability to produce viral proteins in affected hepatocytes. Both host and viral changes can provide novel insights into tumorigenesis, individualized responses to therapeutic intervention, disease progress, and early diagnosis. As such, DNA methylation-mediated epigenetic silencing of cancer-related genes and viral replication is a compelling therapeutic goal to reduce morbidity and mortality from liver cancer caused by chronic HBV infection. In this review, we summarize the most recent research on aberrant DNA methylation associated with HBV infection, which is involved in HCC development, and provide an outlook on the future direction of the research.
Collapse
Affiliation(s)
- Dake Zhang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Shicheng Guo
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Steven J. Schrodi
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA;
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
18
|
Filipski K, Scherer M, Zeiner KN, Bucher A, Kleemann J, Jurmeister P, Hartung TI, Meissner M, Plate KH, Fenton TR, Walter J, Tierling S, Schilling B, Zeiner PS, Harter PN. DNA methylation-based prediction of response to immune checkpoint inhibition in metastatic melanoma. J Immunother Cancer 2021; 9:jitc-2020-002226. [PMID: 34281986 PMCID: PMC8291310 DOI: 10.1136/jitc-2020-002226] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2021] [Indexed: 12/18/2022] Open
Abstract
Background Therapies based on targeting immune checkpoints have revolutionized the treatment of metastatic melanoma in recent years. Still, biomarkers predicting long-term therapy responses are lacking. Methods A novel approach of reference-free deconvolution of large-scale DNA methylation data enabled us to develop a machine learning classifier based on CpG sites, specific for latent methylation components (LMC), that allowed for patient allocation to prognostic clusters. DNA methylation data were processed using reference-free analyses (MeDeCom) and reference-based computational tumor deconvolution (MethylCIBERSORT, LUMP). Results We provide evidence that DNA methylation signatures of tumor tissue from cutaneous metastases are predictive for therapy response to immune checkpoint inhibition in patients with stage IV metastatic melanoma. Conclusions These results demonstrate that LMC-based segregation of large-scale DNA methylation data is a promising tool for classifier development and treatment response estimation in cancer patients under targeted immunotherapy.
Collapse
Affiliation(s)
- Katharina Filipski
- Neurological Institute (Edinger Institute), University Hospital, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
| | - Michael Scherer
- Department of Genetics, University of Saarland, Saarbrücken, Germany.,Max Planck Institute for Informatics, Saarland Informatics Campus, Saarbrücken, Germany.,Graduate School of Computer Science, Saarland Informatics Campus, Saabrücken, Germany
| | - Kim N Zeiner
- Department of Dermatology, University Hospital, Frankfurt am Main, Germany
| | - Andreas Bucher
- Department of Radiology, University Hospital, Frankfurt am Main, Germany
| | - Johannes Kleemann
- Department of Dermatology, University Hospital, Frankfurt am Main, Germany
| | - Philipp Jurmeister
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Institute of Pathology, Ludwig Maximilians University Hospital Munich, Munich, Germany
| | - Tabea I Hartung
- Neurological Institute (Edinger Institute), University Hospital, Frankfurt am Main, Germany
| | - Markus Meissner
- Department of Dermatology, University Hospital, Frankfurt am Main, Germany
| | - Karl H Plate
- Neurological Institute (Edinger Institute), University Hospital, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
| | - Tim R Fenton
- School of Biosciences, University of Kent, Kent, UK
| | - Jörn Walter
- Department of Genetics, University of Saarland, Saarbrücken, Germany
| | - Sascha Tierling
- Department of Genetics, University of Saarland, Saarbrücken, Germany
| | - Bastian Schilling
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Pia S Zeiner
- German Cancer Consortium (DKTK) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany.,Dr. Senckenberg Institute of Neurooncology, University Hospital, Frankfurt am Main, Germany
| | - Patrick N Harter
- Neurological Institute (Edinger Institute), University Hospital, Frankfurt am Main, Germany .,German Cancer Consortium (DKTK) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
| |
Collapse
|
19
|
Scherer M, Schmidt F, Lazareva O, Walter J, Baumbach J, Schulz MH, List M. Machine learning for deciphering cell heterogeneity and gene regulation. NATURE COMPUTATIONAL SCIENCE 2021; 1:183-191. [PMID: 38183187 DOI: 10.1038/s43588-021-00038-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022]
Abstract
Epigenetics studies inheritable and reversible modifications of DNA that allow cells to control gene expression throughout their development and in response to environmental conditions. In computational epigenomics, machine learning is applied to study various epigenetic mechanisms genome wide. Its aim is to expand our understanding of cell differentiation, that is their specialization, in health and disease. Thus far, most efforts focus on understanding the functional encoding of the genome and on unraveling cell-type heterogeneity. Here, we provide an overview of state-of-the-art computational methods and their underlying statistical concepts, which range from matrix factorization and regularized linear regression to deep learning methods. We further show how the rise of single-cell technology leads to new computational challenges and creates opportunities to further our understanding of epigenetic regulation.
Collapse
Affiliation(s)
- Michael Scherer
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany
- Computational Biology Group, Max Planck Institute for Informatics, Saarland Informatics Campus, Saarbrücken, Germany
- Graduate School of Computer Science, Saarland Informatics Campus, Saarbrücken, Germany
| | | | - Olga Lazareva
- Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Jörn Walter
- Computational Biology Group, Max Planck Institute for Informatics, Saarland Informatics Campus, Saarbrücken, Germany
| | - Jan Baumbach
- Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Computational BioMedicine Lab, Institute of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
- Chair of Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Marcel H Schulz
- Institute of Cardiovascular Regeneration, University Hospital and Goethe University Frankfurt, Frankfurt, Germany
| | - Markus List
- Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
| |
Collapse
|
20
|
Nazarov PV, Kreis S. Integrative approaches for analysis of mRNA and microRNA high-throughput data. Comput Struct Biotechnol J 2021; 19:1154-1162. [PMID: 33680358 PMCID: PMC7895676 DOI: 10.1016/j.csbj.2021.01.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
Review on tools and databases linking miRNA and its mRNA targetome. Databases show little overlap in miRNA targetome predictions suggesting strong contextual effects. Deconvolution and deep learning approaches are promising new approaches to improve miRNA targetome predictions.
Advanced sequencing technologies such as RNASeq provide the means for production of massive amounts of data, including transcriptome-wide expression levels of coding RNAs (mRNAs) and non-coding RNAs such as miRNAs, lncRNAs, piRNAs and many other RNA species. In silico analysis of datasets, representing only one RNA species is well established and a variety of tools and pipelines are available. However, attaining a more systematic view of how different players come together to regulate the expression of a gene or a group of genes requires a more intricate approach to data analysis. To fully understand complex transcriptional networks, datasets representing different RNA species need to be integrated. In this review, we will focus on miRNAs as key post-transcriptional regulators summarizing current computational approaches for miRNA:target gene prediction as well as new data-driven methods to tackle the problem of comprehensively and accurately dissecting miRNome-targetome interactions.
Collapse
Key Words
- CCA, canonical correlation analysis
- CDS, coding sequence
- CLASH, cross-linking, ligation and sequencing of hybrids
- CLIP, cross-linking immunoprecipitation
- CNN, convolutional neural network
- Data integration
- GO, gene ontology
- ICA, independent component analysis
- Matrix factorization
- NGS, next-generation sequencing
- NMF, non-negative matrix factorization
- PCA, principal component analysis
- RNASeq, high-throughput RNA sequencing
- TDMD, target RNA-directed miRNA degradation
- TF, transcription factors
- Target prediction
- Transcriptomics
- circRNA, circular RNA
- lncRNA, long non-coding RNA
- mRNA, messenger RNA
- miRNA, microRNA
- microRNA
Collapse
Affiliation(s)
- Petr V Nazarov
- Multiomics Data Science Research Group, Department of Oncology & Quantitative Biology Unit, Luxembourg Institute of Health (LIH), Strassen L-1445, Luxembourg
| | - Stephanie Kreis
- Signal Transduction Group, Department of Life Sciences and Medicine, University of Luxembourg, Belvaux L-4367, Luxembourg
| |
Collapse
|