1
|
Theunis K, Vanuytven S, Claes I, Geurts J, Rambow F, Brown D, Van Der Haegen M, Marin-Bejar O, Rogiers A, Van Raemdonck N, Leucci E, Demeulemeester J, Sifrim A, Marine JC, Voet T. Single-cell genome and transcriptome sequencing without upfront whole-genome amplification reveals cell state plasticity of melanoma subclones. Nucleic Acids Res 2025; 53:gkaf173. [PMID: 40138718 PMCID: PMC11941470 DOI: 10.1093/nar/gkaf173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 02/07/2025] [Accepted: 02/21/2025] [Indexed: 03/29/2025] Open
Abstract
Single-cell multi-omics methods enable the study of cell state diversity, which is largely determined by the interplay of the genome, epigenome, and transcriptome. Here, we describe Gtag&T-seq, a genome-and-transcriptome sequencing (G&T-seq) protocol of the same single cells that omits whole-genome amplification (WGA) by using direct genomic tagmentation (Gtag). Gtag drastically decreases the cost and improves coverage uniformity at single-cell and pseudo-bulk levels compared to WGA-based G&T-seq. We also show that transcriptome-based DNA copy number inference has limited resolution and accuracy, underlining the importance of affordable multi-omic approaches. Applying Gtag&T-seq to a melanoma xenograft model before treatment and at minimal residual disease revealed differential cell state plasticity and treatment response between cancer subclones. In summary, Gtag&T-seq is a low-cost and accurate single-cell multi-omics method that explores genetic alterations and their functional consequences in single cells at scale.
Collapse
Affiliation(s)
- Koen Theunis
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Sebastiaan Vanuytven
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Irene Claes
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, 3000 Leuven, Belgium
| | - Jarne Geurts
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, 3000 Leuven, Belgium
| | - Florian Rambow
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Daniel Brown
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, 3052 Parkville, Australia
| | - Michiel Van Der Haegen
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Oskar Marin-Bejar
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Aljosja Rogiers
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Nina Van Raemdonck
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Eleonora Leucci
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- Trace, Leuven Cancer Institute, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Jonas Demeulemeester
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, 3000 Leuven, Belgium
| | - Alejandro Sifrim
- Laboratory of Multi-omic Integrative Bioinformatics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, 3000 Leuven, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Thierry Voet
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
2
|
Jeong D, Lee AC, Shin K, Kim J, Ham MH, Lee C, Lee S, Choi A, Ryu T, Kim O, Jung Y, Kwon S, Lee DS. Hema-seq reveals genomic aberrations in a rare simultaneous occurrence of hematological malignancies. CELL REPORTS METHODS 2023; 3:100617. [PMID: 37852254 PMCID: PMC10626221 DOI: 10.1016/j.crmeth.2023.100617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/24/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023]
Abstract
Co-occurrence of multiple myeloma and acute myelogenous leukemia is rare, with both malignancies often tracing back to multipotent hematopoietic stem cells. Cytogenetic techniques are the established baseline for diagnosis and characterization of complex hematological malignancies. In this study, we develop a workflow called Hema-seq to delineate clonal changes across various hematopoietic lineages through the integration of whole-genome sequencing, copy-number variations, cell morphology, and cytogenetic aberrations. In Hema-seq, cells are selected from Wright-stained slides and fluorescent probe-stained slides for sequencing. This technique therefore enables direct linking of whole-genome sequences to cytogenetic profiles. Through this method, we mapped sequential clonal alterations within the hematopoietic lineage, identifying critical shifts leading to myeloma and acute myeloid leukemia (AML) cell formations. By synthesizing data from each cell lineage, we provided insights into the hematopoietic tree's clonal evolution. Overall, this study highlights Hema-seq's capability in deciphering genomic heterogeneity in complex hematological malignancies, which can enable better diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Dajeong Jeong
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Amos C Lee
- Bio-MAX Institute, Seoul National University, Seoul 08826, Republic of Korea; Meteor Biotech Co., Ltd., Seoul 08826, Republic of Korea
| | - Kyoungseob Shin
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jinhyun Kim
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Myoung Hee Ham
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Changhee Lee
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sumin Lee
- Meteor Biotech Co., Ltd., Seoul 08826, Republic of Korea; Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Ahyoun Choi
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Taehoon Ryu
- ATG Lifetech, Inc., Seoul 08507, Republic of Korea
| | - Okju Kim
- ATG Lifetech, Inc., Seoul 08507, Republic of Korea
| | - Yushin Jung
- ATG Lifetech, Inc., Seoul 08507, Republic of Korea
| | - Sunghoon Kwon
- Bio-MAX Institute, Seoul National University, Seoul 08826, Republic of Korea; Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea; Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea; Institutes of Entrepreneurial BioConvergence, Seoul National University, Seoul 08826, Republic of Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| | - Dong Soon Lee
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| |
Collapse
|
3
|
Valecha M, Posada D. Somatic variant calling from single-cell DNA sequencing data. Comput Struct Biotechnol J 2022; 20:2978-2985. [PMID: 35782734 PMCID: PMC9218383 DOI: 10.1016/j.csbj.2022.06.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 11/03/2022] Open
Abstract
Single-cell sequencing has gained popularity in recent years. Despite its numerous applications, single-cell DNA sequencing data is highly error-prone due to technical biases arising from uneven sequencing coverage, allelic dropout, and amplification error. With these artifacts, the identification of somatic genomic variants becomes a challenging task, and over the years, several methods have been developed explicitly for this type of data. Single-cell variant callers implement distinct strategies, make different use of the data, and typically result in many discordant calls when applied to real data. Here, we review current approaches for single-cell variant calling, emphasizing single nucleotide variants. We highlight their potential benefits and shortcomings to help users choose a suitable tool for their data at hand.
Collapse
Key Words
- ADO, allelic dropout
- Allele dropout
- Amplification error
- CNV, copy number variant
- Indel, short insertion or deletion
- LDO, locus dropout
- SNV, single nucleotide variant
- SV, structural variant
- Single-cell genomics
- Somatic variants
- VAF, variant allele frequency
- Variant calling
- hSNP, heterozygous single-nucleotide polymorphism
- scATAC-seq, single-cell sequencing assay for transposase-accessible chromatin
- scDNA-seq, single-cell DNA sequencing
- scHi-C, single-cell Hi-C sequencing
- scMethyl-seq, single-cell Methylation sequencing
- scRNA-seq, single-cell RNA sequencing
- scWGA, single-cell whole-genome amplification
Collapse
Affiliation(s)
- Monica Valecha
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Spain
| | - David Posada
- CINBIO, Universidade de Vigo, 36310 Vigo, Spain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Spain
- Department of Biochemistry, Genetics, and Immunology, Universidade de Vigo, 36310 Vigo, Spain
| |
Collapse
|
4
|
Feng X, Chen L. SCSilicon: a tool for synthetic single-cell DNA sequencing data generation. BMC Genomics 2022; 23:359. [PMID: 35546390 PMCID: PMC9092674 DOI: 10.1186/s12864-022-08566-w] [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: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Abstract
Background Single-cell DNA sequencing is getting indispensable in the study of cell-specific cancer genomics. The performance of computational tools that tackle single-cell genome aberrations may be nevertheless undervalued or overvalued, owing to the insufficient size of benchmarking data. In silicon simulation is a cost-effective approach to generate as many single-cell genomes as possible in a controlled manner to make reliable and valid benchmarking. Results This study proposes a new tool, SCSilicon, which efficiently generates single-cell in silicon DNA reads with minimum manual intervention. SCSilicon automatically creates a set of genomic aberrations, including SNP, SNV, Indel, and CNV. Besides, SCSilicon yields the ground truth of CNV segmentation breakpoints and subclone cell labels. We have manually inspected a series of synthetic variations. We conducted a sanity check of the start-of-the-art single-cell CNV callers and found SCYN was the most robust one. Conclusions SCSilicon is a user-friendly software package for users to develop and benchmark single-cell CNV callers. Source code of SCSilicon is available at https://github.com/xikanfeng2/SCSilicon. Supplementary Information The online version contains supplementary material available at (10.1186/s12864-022-08566-w).
Collapse
Affiliation(s)
- Xikang Feng
- School of Software, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Lingxi Chen
- Department of Computer Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| |
Collapse
|
5
|
Zhou Y, Jia E, Sheng Y, Qiao Y, Wang Y, Shi H, Liu Z, Pan M, Tu J, Bai Y, Zhao X, Ge Q, Lu Z. Sensitive and Low-Bias Transcriptome Sequencing Using Agarose PCR. ACS APPLIED MATERIALS & INTERFACES 2022; 14:19154-19167. [PMID: 35446027 DOI: 10.1021/acsami.2c02133] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Transcriptome sequencing has emerged as an important research tool for exploring the mysteries of life at the single-cell level. However, its wide application is limited by the bias associated with the amplification reactions which is essential for library building of trace RNA. In this study, low-melting-point agarose was added to the amplification reactions to take advantage of its molecular crowding effect and polymer cross-linked structure to improve the sensitivity of the reactions and reduce bias. To further evaluate the performance of the method, it was applied to transcriptome sequencing of microregion samples from brain tissue sections of mice with Parkinson's disease at the single cell level. The results showed that agarose PCR had better performance than in-tube PCR. Further application of agarose PCR to transcriptome library sequencing could obtain data closer to that of unamplified. With the addition of low melting point agarose, the sensitivity of the amplification reaction was significantly increased, while homogeneity was increased by approximately 2-fold. Not only that, but this work also provides 11% sensitivity improvement for spatial transcriptomic study on Parkinson's disease-associated gene detection. The agarose PCR provides a new tool for efficient and homogeneous amplification of trace samples and can be widely used for spatial transcriptome library sequencing and studies.
Collapse
Affiliation(s)
- Ying Zhou
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Erteng Jia
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yuqi Sheng
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yi Qiao
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ying Wang
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Huajuan Shi
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhiyu Liu
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Min Pan
- School of Medicine, Southeast University, Nanjing 210097, China
| | - Jing Tu
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yunfei Bai
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiangwei Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Qinyu Ge
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zuhong Lu
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
6
|
Multi-Omics Profiling of the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1361:283-326. [DOI: 10.1007/978-3-030-91836-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
7
|
Reconstructing and counting genomic fragments through tagmentation-based haploid phasing. Sci Rep 2021; 11:18907. [PMID: 34556684 PMCID: PMC8460729 DOI: 10.1038/s41598-021-97852-w] [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: 10/29/2020] [Accepted: 08/27/2021] [Indexed: 11/16/2022] Open
Abstract
Single-cell sequencing provides a new level of granularity in studying the heterogeneous nature of cancer cells. For some cancers, this heterogeneity is the result of copy number changes of genes within the cellular genomes. The ability to accurately determine such copy number changes is critical in tracing and understanding tumorigenesis. Current single-cell genome sequencing methodologies infer copy numbers based on statistical approaches followed by rounding decimal numbers to integer values. Such methodologies are sample dependent, have varying calling sensitivities which heavily depend on the sample’s ploidy and are sensitive to noise in sequencing data. In this paper we have demonstrated the concept of integer-counting by using a novel bioinformatic algorithm built on our library construction chemistry in order to detect the discrete nature of the genome.
Collapse
|
8
|
scDPN for High-throughput Single-cell CNV Detection to Uncover Clonal Evolution During HCC Recurrence. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:346-357. [PMID: 34280548 PMCID: PMC8864190 DOI: 10.1016/j.gpb.2021.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 12/10/2020] [Accepted: 03/06/2021] [Indexed: 11/28/2022]
Abstract
Single-cell genomics provides substantial resources for dissecting cellular heterogeneity and cancer evolution. Unfortunately, classical DNA amplification-based methods have low throughput and introduce coverage bias during sample preamplification. We developed a single-cell DNA library preparation method without preamplification in nanolitre scale (scDPN) to address these issues. The method achieved a throughput of up to 1800 cells per run for copy number variation (CNV) detection. Also, our approach demonstrated a lower level of amplification bias and noise than the multiple displacement amplification (MDA) method and showed high sensitivity and accuracy for cell line and tumor tissue evaluation. We used this approach to profile the tumor clones in paired primary and relapsed tumor samples of hepatocellular carcinoma (HCC). We identified three clonal subpopulations with a multitude of aneuploid alterations across the genome. Furthermore, we observed that a minor clone of the primary tumor containing additional alterations in chromosomes 1q, 10q, and 14q developed into the dominant clone in the recurrent tumor, indicating clonal selection during recurrence in HCC. Overall, this approach provides a comprehensive and scalable solution to understand genome heterogeneity and evolution
Collapse
|
9
|
Minussi DC, Nicholson MD, Ye H, Davis A, Wang K, Baker T, Tarabichi M, Sei E, Du H, Rabbani M, Peng C, Hu M, Bai S, Lin YW, Schalck A, Multani A, Ma J, McDonald TO, Casasent A, Barrera A, Chen H, Lim B, Arun B, Meric-Bernstam F, Van Loo P, Michor F, Navin NE. Breast tumours maintain a reservoir of subclonal diversity during expansion. Nature 2021; 592:302-308. [PMID: 33762732 PMCID: PMC8049101 DOI: 10.1038/s41586-021-03357-x] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 02/12/2021] [Indexed: 12/21/2022]
Abstract
Our knowledge of copy number evolution during the expansion of primary breast tumours is limited1,2. Here, to investigate this process, we developed a single-cell, single-molecule DNA-sequencing method and performed copy number analysis of 16,178 single cells from 8 human triple-negative breast cancers and 4 cell lines. The results show that breast tumours and cell lines comprise a large milieu of subclones (7-22) that are organized into a few (3-5) major superclones. Evolutionary analysis suggests that after clonal TP53 mutations, multiple loss-of-heterozygosity events and genome doubling, there was a period of transient genomic instability followed by ongoing copy number evolution during the primary tumour expansion. By subcloning single daughter cells in culture, we show that tumour cells rediversify their genomes and do not retain isogenic properties. These data show that triple-negative breast cancers continue to evolve chromosome aberrations and maintain a reservoir of subclonal diversity during primary tumour growth.
Collapse
Affiliation(s)
- Darlan C Minussi
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth, Houston, TX, USA
| | - Michael D Nicholson
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Hanghui Ye
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth, Houston, TX, USA
| | - Alexander Davis
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth, Houston, TX, USA
| | - Kaile Wang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Toby Baker
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
| | - Maxime Tarabichi
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
| | - Emi Sei
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Haowei Du
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate Program in Diagnostic Genetics, School of Health Professions, MD Anderson Cancer Center, Houston, TX, USA
| | - Mashiat Rabbani
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate Program in Diagnostic Genetics, School of Health Professions, MD Anderson Cancer Center, Houston, TX, USA
| | - Cheng Peng
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate Program in Diagnostic Genetics, School of Health Professions, MD Anderson Cancer Center, Houston, TX, USA
| | - Min Hu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shanshan Bai
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yu-Wei Lin
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth, Houston, TX, USA
| | - Aislyn Schalck
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth, Houston, TX, USA
| | - Asha Multani
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jin Ma
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas O McDonald
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Anna Casasent
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth, Houston, TX, USA
| | - Angelica Barrera
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hui Chen
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Banu Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peter Van Loo
- Cancer Genomics Laboratory, The Francis Crick Institute, London, UK
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA. .,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA. .,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA. .,Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA, USA. .,The Ludwig Center at Harvard, Boston, MA, and the Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Nicholas E Navin
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth, Houston, TX, USA. .,Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
10
|
Zachariadis V, Cheng H, Andrews N, Enge M. A Highly Scalable Method for Joint Whole-Genome Sequencing and Gene-Expression Profiling of Single Cells. Mol Cell 2020; 80:541-553.e5. [PMID: 33068522 DOI: 10.1016/j.molcel.2020.09.025] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/17/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
To address how genetic variation alters gene expression in complex cell mixtures, we developed direct nuclear tagmentation and RNA sequencing (DNTR-seq), which enables whole-genome and mRNA sequencing jointly in single cells. DNTR-seq readily identified minor subclones within leukemia patients. In a large-scale DNA damage screen, DNTR-seq was used to detect regions under purifying selection and identified genes where mRNA abundance was resistant to copy-number alteration, suggesting strong genetic compensation. mRNA sequencing (mRNA-seq) quality equals RNA-only methods, and the low positional bias of genomic libraries allowed detection of sub-megabase aberrations at ultra-low coverage. Each cell library is individually addressable and can be re-sequenced at increased depth, allowing multi-tiered study designs. Additionally, the direct tagmentation protocol enables coverage-independent estimation of ploidy, which can be used to identify cell singlets. Thus, DNTR-seq directly links each cell's state to its corresponding genome at scale, enabling routine analysis of heterogeneous tumors and other complex tissues.
Collapse
Affiliation(s)
- Vasilios Zachariadis
- Department of Oncology-Pathology Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Huaitao Cheng
- Department of Oncology-Pathology Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Nathanael Andrews
- Department of Oncology-Pathology Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Martin Enge
- Department of Oncology-Pathology Karolinska Institutet, 171 64 Stockholm, Sweden.
| |
Collapse
|
11
|
Mallory XF, Edrisi M, Navin N, Nakhleh L. Methods for copy number aberration detection from single-cell DNA-sequencing data. Genome Biol 2020; 21:208. [PMID: 32807205 PMCID: PMC7433197 DOI: 10.1186/s13059-020-02119-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
Copy number aberrations (CNAs), which are pathogenic copy number variations (CNVs), play an important role in the initiation and progression of cancer. Single-cell DNA-sequencing (scDNAseq) technologies produce data that is ideal for inferring CNAs. In this review, we review eight methods that have been developed for detecting CNAs in scDNAseq data, and categorize them according to the steps of a seven-step pipeline that they employ. Furthermore, we review models and methods for evolutionary analyses of CNAs from scDNAseq data and highlight advances and future research directions for computational methods for CNA detection from scDNAseq data.
Collapse
Affiliation(s)
- Xian F. Mallory
- Department of Computer Science, Rice University, Houston, TX USA
- Department of Computer Science, Florida State University, Tallahassee, FL USA
| | | | - Nicholas Navin
- Department of Genetics, the University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Luay Nakhleh
- Department of Computer Science, Rice University, Houston, TX USA
| |
Collapse
|
12
|
Assessing the performance of methods for copy number aberration detection from single-cell DNA sequencing data. PLoS Comput Biol 2020; 16:e1008012. [PMID: 32658894 PMCID: PMC7377518 DOI: 10.1371/journal.pcbi.1008012] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 07/23/2020] [Accepted: 06/03/2020] [Indexed: 12/22/2022] Open
Abstract
Single-cell DNA sequencing technologies are enabling the study of mutations and their evolutionary trajectories in cancer. Somatic copy number aberrations (CNAs) have been implicated in the development and progression of various types of cancer. A wide array of methods for CNA detection has been either developed specifically for or adapted to single-cell DNA sequencing data. Understanding the strengths and limitations that are unique to each of these methods is very important for obtaining accurate copy number profiles from single-cell DNA sequencing data. We benchmarked three widely used methods–Ginkgo, HMMcopy, and CopyNumber–on simulated as well as real datasets. To facilitate this, we developed a novel simulator of single-cell genome evolution in the presence of CNAs. Furthermore, to assess performance on empirical data where the ground truth is unknown, we introduce a phylogeny-based measure for identifying potentially erroneous inferences. While single-cell DNA sequencing is very promising for elucidating and understanding CNAs, our findings show that even the best existing method does not exceed 80% accuracy. New methods that significantly improve upon the accuracy of these three methods are needed. Furthermore, with the large datasets being generated, the methods must be computationally efficient. Copy number aberrations, or CNAs, refer to evolutionary events that act on cancer genomes by deleting segments of the genomes or introducing new copies of existing segments. These events have been implicated in various types of cancer; consequently, their accurate detection could shed light on the initiation and progression of tumor, as well as on the development of potential targeted therapeutics. Single-cell DNA sequencing technologies are now producing the type of data that would allow such detection at the resolution of individual cells. However, to achieve this detection task, methods have to implement several steps of “data wrangling” and dealing with technical artifacts. In this work, we benchmarked three widely used methods for CNA detection from single-cell DNA data, namely Ginkgo, HMMcopy, and CopyNumber. To accomplish this study, we developed a novel simulator and devised a phylogeny-based measure of potentially erroneous CNA calls. We find that none of these methods has high accuracy, and all of them can be computationally very demanding. These findings call for the development of more accurate and more efficient methods for CNA detection from single-cell DNA data.
Collapse
|
13
|
Quirico L, Orso F. The power of microRNAs as diagnostic and prognostic biomarkers in liquid biopsies. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:117-139. [PMID: 35582611 PMCID: PMC9090592 DOI: 10.20517/cdr.2019.103] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/24/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023]
Abstract
In the last decades, progresses in medical oncology have ameliorated the treatment of patients and their outcome. However, further improvements are still necessary, in particular for certain types of tumors such as pancreatic, gastric, and lung cancer as well as acute myeloid leukemia where early detection and monitoring of the disease are crucial for final patient outcome. Liquid biopsy represents a great advance in the field because it is less invasive, less time-consuming, and safer compared to classical biopsies and it can be useful to monitor the evolution of the disease as well as the response of patients to therapy. Liquid biopsy allows the detection of circulating tumor cells, nucleic acids, and exosomes not only in blood but also in different biological fluids: urine, saliva, pleural effusions, cerebrospinal fluid, and stool. Among the potential biomarkers detectable in liquid biopsies, microRNAs (miRNAs) are gaining more and more attention, since they are easily detectable, quite stable in biological fluids, and show high sensitivity. Many data demonstrate that miRNAs alone or in combination with other biomarkers could improve the diagnostic and prognostic power for many different tumors. Despite this, standardization of methods, sample preparation, and analysis remain challenging and a huge effort should be made to address these issues before miRNA biomarkers can enter the clinic. This review summarizes the main findings in the field of circulating miRNAs in both solid and hematological tumors.
Collapse
Affiliation(s)
- Lorena Quirico
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
- Molecular Biotechnology Center (MBC), University of Torino, Torino 10126, Italy
| | - Francesca Orso
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
- Molecular Biotechnology Center (MBC), University of Torino, Torino 10126, Italy
- Center for Complex Systems in Molecular Biology and Medicine, University of Torino, Torino 10126, Italy
| |
Collapse
|
14
|
Lähnemann D, Köster J, Szczurek E, McCarthy DJ, Hicks SC, Robinson MD, Vallejos CA, Campbell KR, Beerenwinkel N, Mahfouz A, Pinello L, Skums P, Stamatakis A, Attolini CSO, Aparicio S, Baaijens J, Balvert M, Barbanson BD, Cappuccio A, Corleone G, Dutilh BE, Florescu M, Guryev V, Holmer R, Jahn K, Lobo TJ, Keizer EM, Khatri I, Kielbasa SM, Korbel JO, Kozlov AM, Kuo TH, Lelieveldt BP, Mandoiu II, Marioni JC, Marschall T, Mölder F, Niknejad A, Rączkowska A, Reinders M, Ridder JD, Saliba AE, Somarakis A, Stegle O, Theis FJ, Yang H, Zelikovsky A, McHardy AC, Raphael BJ, Shah SP, Schönhuth A. Eleven grand challenges in single-cell data science. Genome Biol 2020; 21:31. [PMID: 32033589 PMCID: PMC7007675 DOI: 10.1186/s13059-020-1926-6] [Citation(s) in RCA: 679] [Impact Index Per Article: 135.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/02/2020] [Indexed: 02/08/2023] Open
Abstract
The recent boom in microfluidics and combinatorial indexing strategies, combined with low sequencing costs, has empowered single-cell sequencing technology. Thousands-or even millions-of cells analyzed in a single experiment amount to a data revolution in single-cell biology and pose unique data science problems. Here, we outline eleven challenges that will be central to bringing this emerging field of single-cell data science forward. For each challenge, we highlight motivating research questions, review prior work, and formulate open problems. This compendium is for established researchers, newcomers, and students alike, highlighting interesting and rewarding problems for the coming years.
Collapse
Affiliation(s)
- David Lähnemann
- Algorithms for Reproducible Bioinformatics, Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Paediatric Oncology, Haematology and Immunology, Medical Faculty, Heinrich Heine University, University Hospital, Düsseldorf, Germany
- Computational Biology of Infection Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Johannes Köster
- Algorithms for Reproducible Bioinformatics, Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Ewa Szczurek
- Institute of Informatics, Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warszawa, Poland
| | - Davis J. McCarthy
- Bioinformatics and Cellular Genomics, St Vincent’s Institute of Medical Research, Fitzroy, Australia
- Melbourne Integrative Genomics, School of BioSciences–School of Mathematics & Statistics, Faculty of Science, University of Melbourne, Melbourne, Australia
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins University, Baltimore, MD USA
| | - Mark D. Robinson
- Institute of Molecular Life Sciences and SIB Swiss Institute of Bioinformatics, University of Zürich, Zürich, Switzerland
| | - Catalina A. Vallejos
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
- The Alan Turing Institute, British Library, London, UK
| | - Kieran R. Campbell
- Department of Statistics, University of British Columbia, Vancouver, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, Canada
- Data Science Institute, University of British Columbia, Vancouver, Canada
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Ahmed Mahfouz
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, The Netherlands
- Delft Bioinformatics Lab, Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft, The Netherlands
| | - Luca Pinello
- Molecular Pathology Unit and Center for Cancer Research, Massachusetts General Hospital Research Institute, Charlestown, USA
- Department of Pathology, Harvard Medical School, Boston, USA
- Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Pavel Skums
- Department of Computer Science, Georgia State University, Atlanta, USA
| | - Alexandros Stamatakis
- Computational Molecular Evolution Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
- Institute for Theoretical Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | | | - Samuel Aparicio
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Jasmijn Baaijens
- Life Sciences and Health, Centrum Wiskunde & Informatica, Amsterdam, The Netherlands
| | - Marleen Balvert
- Life Sciences and Health, Centrum Wiskunde & Informatica, Amsterdam, The Netherlands
- Theoretical Biology and Bioinformatics, Science for Life, Utrecht University, Utrecht, The Netherlands
| | - Buys de Barbanson
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Quantitative biology, Hubrecht Institute, Utrecht, The Netherlands
| | - Antonio Cappuccio
- Institute for Advanced Study, University of Amsterdam, Amsterdam, The Netherlands
| | - Giacomo Corleone
- Department of Surgery and Cancer, The Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Bas E. Dutilh
- Theoretical Biology and Bioinformatics, Science for Life, Utrecht University, Utrecht, The Netherlands
- Centre for Molecular and Biomolecular Informatics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maria Florescu
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Quantitative biology, Hubrecht Institute, Utrecht, The Netherlands
| | - Victor Guryev
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rens Holmer
- Bioinformatics Group, Wageningen University, Wageningen, The Netherlands
| | - Katharina Jahn
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Thamar Jessurun Lobo
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Emma M. Keizer
- Biometris, Wageningen University & Research, Wageningen, The Netherlands
| | - Indu Khatri
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Szymon M. Kielbasa
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan O. Korbel
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Alexey M. Kozlov
- Computational Molecular Evolution Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Tzu-Hao Kuo
- Computational Biology of Infection Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Boudewijn P.F. Lelieveldt
- PRB lab, Delft University of Technology, Delft, The Netherlands
- Division of Image Processing, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ion I. Mandoiu
- Computer Science & Engineering Department, University of Connecticut, Storrs, USA
| | - John C. Marioni
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Tobias Marschall
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany
- Max Planck Institute for Informatics, Saarbrücken, Germany
| | - Felix Mölder
- Algorithms for Reproducible Bioinformatics, Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Amir Niknejad
- Computation molecular design, Zuse Institute Berlin, Berlin, Germany
- Mathematics Department, Mount Saint Vincent, New York, USA
| | - Alicja Rączkowska
- Institute of Informatics, Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warszawa, Poland
| | - Marcel Reinders
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, The Netherlands
- Delft Bioinformatics Lab, Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft, The Netherlands
| | - Jeroen de Ridder
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research, Helmholtz-Center for Infection Research, Würzburg, Germany
| | - Antonios Somarakis
- Division of Image Processing, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Oliver Stegle
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center–DKFZ, Heidelberg, Germany
| | - Fabian J. Theis
- Institute of Computational Biology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - Huan Yang
- Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research–LACDR–Leiden University, Leiden, The Netherlands
| | - Alex Zelikovsky
- Department of Computer Science, Georgia State University, Atlanta, USA
- The Laboratory of Bioinformatics, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alice C. McHardy
- Computational Biology of Infection Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Sohrab P. Shah
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Alexander Schönhuth
- Life Sciences and Health, Centrum Wiskunde & Informatica, Amsterdam, The Netherlands
- Theoretical Biology and Bioinformatics, Science for Life, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
15
|
Xi L, Leong P, Mihajlovic A. Preparing Single-cell DNA Library Using Nextera for Detection of CNV. Bio Protoc 2019; 9:e3175. [PMID: 33654981 DOI: 10.21769/bioprotoc.3175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/25/2019] [Accepted: 02/11/2019] [Indexed: 12/26/2022] Open
Abstract
Single-cell DNA sequencing is a powerful tool to evaluate the state of heterogeneity of heterogeneous tissues like cancer in a quantitative manner that bulk sequencing can never achieve. DOP-PCR (Degenerate Oligonucleotide-Primed Polymerase Chain Reaction), MDA (Multiple Displacement Amplification), MALBAC (Multiple Annealing and Looping-Based Amplification Cycles), LIANTI (Linear Amplification via Transposon Insertion) and TnBC (Transposon Barcoded) have been the primary choices to prepare single-cell libraries. TnBC library prep method is a simple and versatile methodology, to detect copy number variations or to obtain the absolute copy numbers of genes per cell.
Collapse
Affiliation(s)
- Larry Xi
- Digenomix Corp, South San Francisco, CA 94080, USA
| | | | | |
Collapse
|
16
|
Molecular heterogeneity and early metastatic clone selection in testicular germ cell cancer development. Br J Cancer 2019; 120:444-452. [PMID: 30739914 PMCID: PMC6461884 DOI: 10.1038/s41416-019-0381-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/27/2018] [Indexed: 02/07/2023] Open
Abstract
Background Testicular germ cell cancer (TGCC), being the most frequent malignancy in young Caucasian males, is initiated from an embryonic germ cell. This study determines intratumour heterogeneity to unravel tumour progression from initiation until metastasis. Methods In total, 42 purified samples of four treatment-resistant nonseminomatous (NS) TGCC were investigated, including the precursor germ cell neoplasia in situ (GCNIS) and metastatic specimens, using whole-genome and targeted sequencing. Their evolution was reconstructed. Results Intratumour molecular heterogeneity did not correspond to the supposed primary tumour histological evolution. Metastases after systemic treatment could be derived from cancer stem cells not identified in the primary cancer. GCNIS mostly lacked the molecular marks of the primary NS and comprised dominant clones that failed to progress. A BRCA-like mutational signature was observed without evidence for direct involvement of BRCA1 and BRCA2 genes. Conclusions Our data strongly support the hypothesis that NS is initiated by whole-genome duplication, followed by chromosome copy number alterations in the cancer stem cell population, and accumulation of low numbers of somatic mutations, even in therapy-resistant cases. These observations of heterogeneity at all stages of tumourigenesis should be considered when treating patients with GCNIS-only disease, or with clinically overt NS.
Collapse
|
17
|
Coller HA. Symposium on single cell analysis and genomic approaches, Experimental Biology 2017 Chicago, Illinois, April 23, 2017. Physiol Genomics 2017; 49:491-495. [PMID: 28802263 PMCID: PMC5625270 DOI: 10.1152/physiolgenomics.00049.2017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 11/22/2022] Open
Abstract
Emerging technologies for the analysis of genome-wide information in single cells have the potential to transform many fields of biology, including our understanding of cell states, the response of cells to external stimuli, mosaicism, and intratumor heterogeneity. At Experimental Biology 2017 in Chicago, Physiological Genomics hosted a symposium in which five leaders in the field of single cell genomics presented their recent research. The speakers discussed emerging methodologies in single cell analysis and critical issues for the analysis of single cell data. Also discussed were applications of single cell genomics to understanding the different types of cells within an organism or tissue and the basis for cell-to-cell variability in response to stimuli.
Collapse
Affiliation(s)
- Hilary A Coller
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles and Department of Biological Chemistry, David Geffen School of Medicine, Los Angeles, California
| |
Collapse
|