Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Med ; 26(7): 1114-1124, 2020 07.
Article in English | MEDLINE | ID: mdl-32483360

ABSTRACT

In many areas of oncology, we lack sensitive tools to track low-burden disease. Although cell-free DNA (cfDNA) shows promise in detecting cancer mutations, we found that the combination of low tumor fraction (TF) and limited number of DNA fragments restricts low-disease-burden monitoring through the prevailing deep targeted sequencing paradigm. We reasoned that breadth may supplant depth of sequencing to overcome the barrier of cfDNA abundance. Whole-genome sequencing (WGS) of cfDNA allowed ultra-sensitive detection, capitalizing on the cumulative signal of thousands of somatic mutations observed in solid malignancies, with TF detection sensitivity as low as 10-5. The WGS approach enabled dynamic tumor burden tracking and postoperative residual disease detection, associated with adverse outcome. Thus, we present an orthogonal framework for cfDNA cancer monitoring via genome-wide mutational integration, enabling ultra-sensitive detection, overcoming the limitation of cfDNA abundance and empowering treatment optimization in low-disease-burden oncology care.


Subject(s)
Biomarkers, Tumor/genetics , Circulating Tumor DNA/blood , DNA, Neoplasm/genetics , Neoplasms/blood , Biomarkers, Tumor/blood , Cell-Free Nucleic Acids/blood , DNA Copy Number Variations/genetics , DNA, Neoplasm/blood , Disease-Free Survival , Female , Genome, Human/genetics , High-Throughput Nucleotide Sequencing , Humans , Kaplan-Meier Estimate , Male , Mutation/genetics , Neoplasms/genetics , Neoplasms/pathology , Tumor Burden/genetics , Whole Genome Sequencing
2.
Nature ; 571(7765): 355-360, 2019 07.
Article in English | MEDLINE | ID: mdl-31270458

ABSTRACT

Defining the transcriptomic identity of malignant cells is challenging in the absence of surface markers that distinguish cancer clones from one another, or from admixed non-neoplastic cells. To address this challenge, here we developed Genotyping of Transcriptomes (GoT), a method to integrate genotyping with high-throughput droplet-based single-cell RNA sequencing. We apply GoT to profile 38,290 CD34+ cells from patients with CALR-mutated myeloproliferative neoplasms to study how somatic mutations corrupt the complex process of human haematopoiesis. High-resolution mapping of malignant versus normal haematopoietic progenitors revealed an increasing fitness advantage with myeloid differentiation of cells with mutated CALR. We identified the unfolded protein response as a predominant outcome of CALR mutations, with a considerable dependency on cell identity, as well as upregulation of the NF-κB pathway specifically in uncommitted stem cells. We further extended the GoT toolkit to genotype multiple targets and loci that are distant from transcript ends. Together, these findings reveal that the transcriptional output of somatic mutations in myeloproliferative neoplasms is dependent on the native cell identity.


Subject(s)
Genotype , Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Neoplasms/genetics , Neoplasms/pathology , Transcriptome/genetics , Animals , Antigens, CD34/metabolism , Calreticulin/genetics , Cell Line , Cell Proliferation , Clone Cells/classification , Clone Cells/metabolism , Clone Cells/pathology , Endoribonucleases/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/classification , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , High-Throughput Nucleotide Sequencing/methods , Humans , Mice , Models, Molecular , Myeloproliferative Disorders/classification , NF-kappa B/metabolism , Neoplasms/classification , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Protein Serine-Threonine Kinases/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Unfolded Protein Response/genetics
3.
Nature ; 569(7757): 576-580, 2019 05.
Article in English | MEDLINE | ID: mdl-31092926

ABSTRACT

Genetic and epigenetic intra-tumoral heterogeneity cooperate to shape the evolutionary course of cancer1. Chronic lymphocytic leukaemia (CLL) is a highly informative model for cancer evolution as it undergoes substantial genetic diversification and evolution after therapy2,3. The CLL epigenome is also an important disease-defining feature4,5, and growing populations of cells in CLL diversify by stochastic changes in DNA methylation known as epimutations6. However, previous studies using bulk sequencing methods to analyse the patterns of DNA methylation were unable to determine whether epimutations affect CLL populations homogeneously. Here, to measure the epimutation rate at single-cell resolution, we applied multiplexed single-cell reduced-representation bisulfite sequencing to B cells from healthy donors and patients with CLL. We observed that the common clonal origin of CLL results in a consistently increased epimutation rate, with low variability in the cell-to-cell epimutation rate. By contrast, variable epimutation rates across healthy B cells reflect diverse evolutionary ages across the trajectory of B cell differentiation, consistent with epimutations serving as a molecular clock. Heritable epimutation information allowed us to reconstruct lineages at high-resolution with single-cell data, and to apply this directly to patient samples. The CLL lineage tree shape revealed earlier branching and longer branch lengths than in normal B cells, reflecting rapid drift after the initial malignant transformation and a greater proliferative history. Integration of single-cell bisulfite sequencing analysis with single-cell transcriptomes and genotyping confirmed that genetic subclones mapped to distinct clades, as inferred solely on the basis of epimutation information. Finally, to examine potential lineage biases during therapy, we profiled serial samples during ibrutinib-associated lymphocytosis, and identified clades of cells that were preferentially expelled from the lymph node after treatment, marked by distinct transcriptional profiles. The single-cell integration of genetic, epigenetic and transcriptional information thus charts the lineage history of CLL and its evolution with therapy.


Subject(s)
Cell Lineage , Epigenesis, Genetic , Evolution, Molecular , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Base Sequence , Biological Clocks , Cell Lineage/genetics , DNA Methylation , Epigenome/genetics , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mutation Rate , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic
4.
Nat Commun ; 10(1): 1289, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894531

ABSTRACT

Cellular proteins continuously undergo non-enzymatic covalent modifications (NECMs) that accumulate under normal physiological conditions and are stimulated by changes in the cellular microenvironment. Glycation, the hallmark of diabetes, is a prevalent NECM associated with an array of pathologies. Histone proteins are particularly susceptible to NECMs due to their long half-lives and nucleophilic disordered tails that undergo extensive regulatory modifications; however, histone NECMs remain poorly understood. Here we perform a detailed analysis of histone glycation in vitro and in vivo and find it has global ramifications on histone enzymatic PTMs, the assembly and stability of nucleosomes, and chromatin architecture. Importantly, we identify a physiologic regulation mechanism, the enzyme DJ-1, which functions as a potent histone deglycase. Finally, we detect intense histone glycation and DJ-1 overexpression in breast cancer tumors. Collectively, our results suggest an additional mechanism for cellular metabolic damage through epigenetic perturbation, with implications in pathogenesis.


Subject(s)
Breast Neoplasms/genetics , Epigenesis, Genetic , Glycation End Products, Advanced/genetics , Histones/metabolism , Nucleosomes/chemistry , Protein Deglycase DJ-1/genetics , Protein Processing, Post-Translational , Acetylation/drug effects , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Glycation End Products, Advanced/metabolism , Glycosylation/drug effects , Heterografts , Histones/genetics , Humans , Mice , Nucleosomes/metabolism , Protein Deglycase DJ-1/metabolism , Pyruvaldehyde/pharmacology , Tumor Microenvironment/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...