Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Genom ; : 100592, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38925122

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) datasets contain true single cells, or singlets, in addition to cells that coalesce during the protocol, or doublets. Identifying singlets with high fidelity in scRNA-seq is necessary to avoid false negative and false positive discoveries. Although several methodologies have been proposed, they are typically tested on highly heterogeneous datasets and lack a priori knowledge of true singlets. Here, we leveraged datasets with synthetically introduced DNA barcodes for a hitherto unexplored application: to extract ground-truth singlets. We demonstrated the feasibility of our framework, "singletCode," to evaluate existing doublet detection methods across a range of contexts. We also leveraged our ground-truth singlets to train a proof-of-concept machine learning classifier, which outperformed other doublet detection algorithms. Our integrative framework can identify ground-truth singlets and enable robust doublet detection in non-barcoded datasets.

2.
Cell Death Dis ; 15(4): 290, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658567

ABSTRACT

High-grade serous ovarian cancer (HGSOC) represents the most common and lethal subtype of ovarian cancer. Despite initial response to platinum-based standard therapy, patients commonly suffer from relapse that likely originates from drug-tolerant persister (DTP) cells. We generated isogenic clones of treatment-naïve and cisplatin-tolerant persister HGSOC cells. In addition, single-cell RNA sequencing of barcoded cells was performed in a xenograft model with HGSOC cell lines after platinum-based therapy. Published single-cell RNA-sequencing data from neo-adjuvant and non-treated HGSOC patients and patient data from TCGA were analyzed. DTP-derived cells exhibited morphological alterations and upregulation of epithelial-mesenchymal transition (EMT) markers. An aggressive subpopulation of DTP-derived cells showed high expression of the stress marker ATF3. Knockdown of ATF3 enhanced the sensitivity of aggressive DTP-derived cells to cisplatin-induced cell death, implying a role for ATF3 stress response in promoting a drug tolerant persister cell state. Furthermore, single cell lineage tracing to detect transcriptional changes in a HGSOC cell line-derived xenograft relapse model showed that cells derived from relapsed solid tumors express increased levels of EMT and multiple endoplasmic reticulum (ER) stress markers, including ATF3. Single cell RNA sequencing of epithelial cells from four HGSOC patients also identified a small cell population resembling DTP cells in all samples. Moreover, analysis of TCGA data from 259 HGSOC patients revealed a significant progression-free survival advantage for patients with low expression of the ATF3-associated partial EMT genes. These findings suggest that increased ATF3 expression together with partial EMT promote the development of aggressive DTP, and thereby relapse in HGSOC patients.


Subject(s)
Activating Transcription Factor 3 , Cisplatin , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Ovarian Neoplasms , Humans , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 3/genetics , Female , Cisplatin/pharmacology , Cisplatin/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Animals , Mice , Xenograft Model Antitumor Assays , Gene Expression Regulation, Neoplastic/drug effects
3.
Cancer Discov ; 12(11): 2666-2683, 2022 11 02.
Article in English | MEDLINE | ID: mdl-35895872

ABSTRACT

Anticancer therapies have been limited by the emergence of mutations and other adaptations. In bacteria, antibiotics activate the SOS response, which mobilizes error-prone factors that allow for continuous replication at the cost of mutagenesis. We investigated whether the treatment of lung cancer with EGFR inhibitors (EGFRi) similarly engages hypermutators. In cycling drug-tolerant persister (DTP) cells and in EGFRi-treated patients presenting residual disease, we observed upregulation of GAS6, whereas ablation of GAS6's receptor, AXL, eradicated resistance. Reciprocally, AXL overexpression enhanced DTP survival and accelerated the emergence of T790M, an EGFR mutation typical to resistant cells. Mechanistically, AXL induces low-fidelity DNA polymerases and activates their organizer, RAD18, by promoting neddylation. Metabolomics uncovered another hypermutator, AXL-driven activation of MYC, and increased purine synthesis that is unbalanced by pyrimidines. Aligning anti-AXL combination treatments with the transition from DTPs to resistant cells cured patient-derived xenografts. Hence, similar to bacteria, tumors tolerate therapy by engaging pharmacologically targetable endogenous mutators. SIGNIFICANCE: EGFR-mutant lung cancers treated with kinase inhibitors often evolve resistance due to secondary mutations. We report that in similarity to the bacterial SOS response stimulated by antibiotics, endogenous mutators are activated in drug-treated cells, and this heralds tolerance. Blocking the process prevented resistance in xenograft models, which offers new treatment strategies. This article is highlighted in the In This Issue feature, p. 2483.


Subject(s)
Drug Resistance, Neoplasm , Lung Neoplasms , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , Humans , Cell Line, Tumor , DNA Replication , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Axl Receptor Tyrosine Kinase
4.
Mol Syst Biol ; 18(7): e11168, 2022 07.
Article in English | MEDLINE | ID: mdl-35866381

ABSTRACT

Similar to persister bacterial cells that survive antibiotic treatments, some cancer cells can evade drug treatments. This Commentary discusses the different classes of persister cells and their implications for developing more efficient cancer treatments.


Subject(s)
Anti-Bacterial Agents , Neoplasms , Microbial Sensitivity Tests , Neoplasms/drug therapy
5.
Nat Rev Cancer ; 22(8): 434-435, 2022 08.
Article in English | MEDLINE | ID: mdl-35538369
6.
Nature ; 596(7873): 576-582, 2021 08.
Article in English | MEDLINE | ID: mdl-34381210

ABSTRACT

Non-genetic mechanisms have recently emerged as important drivers of cancer therapy failure1, where some cancer cells can enter a reversible drug-tolerant persister state in response to treatment2. Although most cancer persisters remain arrested in the presence of the drug, a rare subset can re-enter the cell cycle under constitutive drug treatment. Little is known about the non-genetic mechanisms that enable cancer persisters to maintain proliferative capacity in the presence of drugs. To study this rare, transiently resistant, proliferative persister population, we developed Watermelon, a high-complexity expressed barcode lentiviral library for simultaneous tracing of each cell's clonal origin and proliferative and transcriptional states. Here we show that cycling and non-cycling persisters arise from different cell lineages with distinct transcriptional and metabolic programs. Upregulation of antioxidant gene programs and a metabolic shift to fatty acid oxidation are associated with persister proliferative capacity across multiple cancer types. Impeding oxidative stress or metabolic reprogramming alters the fraction of cycling persisters. In human tumours, programs associated with cycling persisters are induced in minimal residual disease in response to multiple targeted therapies. The Watermelon system enabled the identification of rare persister lineages that are preferentially poised to proliferate under drug pressure, thus exposing new vulnerabilities that can be targeted to delay or even prevent disease recurrence.


Subject(s)
Cell Cycle , Cell Lineage , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Antioxidants/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , DNA Barcoding, Taxonomic , Fatty Acids/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lentivirus/genetics , Neoplasm Recurrence, Local/genetics , Neoplasms/genetics , Neoplasms/metabolism , Oncogene Proteins/antagonists & inhibitors , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism , Transcription, Genetic/drug effects
7.
Cell Rep ; 33(13): 108566, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33378681

ABSTRACT

Aging is closely associated with increased susceptibility to breast cancer, yet there have been limited systematic studies of aging-induced alterations in the mammary gland. Here, we leverage high-throughput single-cell RNA sequencing to generate a detailed transcriptomic atlas of young and aged murine mammary tissues. By analyzing epithelial, stromal, and immune cells, we identify age-dependent alterations in cell proportions and gene expression, providing evidence that suggests alveolar maturation and physiological decline. The analysis also uncovers potential pro-tumorigenic mechanisms coupled to the age-associated loss of tumor suppressor function and change in microenvironment. In addition, we identify a rare, age-dependent luminal population co-expressing hormone-sensing and secretory-alveolar lineage markers, as well as two macrophage populations expressing distinct gene signatures, underscoring the complex heterogeneity of the mammary epithelia and stroma. Collectively, this rich single-cell atlas reveals the effects of aging on mammary physiology and can serve as a useful resource for understanding aging-associated cancer risk.


Subject(s)
Aging/psychology , Epithelial Cells/metabolism , Gene Expression Regulation , Mammary Glands, Animal/metabolism , Stromal Cells/metabolism , Transcriptome , Animals , Biomarkers/metabolism , Cells, Cultured , Cellular Senescence , Dendritic Cells/metabolism , Female , Genes, Tumor Suppressor , High-Throughput Nucleotide Sequencing/methods , Lymphocytes/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Single-Cell Analysis/methods
8.
Cell ; 176(6): 1325-1339.e22, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30827679

ABSTRACT

Lineage tracing provides key insights into the fate of individual cells in complex organisms. Although effective genetic labeling approaches are available in model systems, in humans, most approaches require detection of nuclear somatic mutations, which have high error rates, limited scale, and do not capture cell state information. Here, we show that somatic mutations in mtDNA can be tracked by single-cell RNA or assay for transposase accessible chromatin (ATAC) sequencing. We leverage somatic mtDNA mutations as natural genetic barcodes and demonstrate their utility as highly accurate clonal markers to infer cellular relationships. We track native human cells both in vitro and in vivo and relate clonal dynamics to gene expression and chromatin accessibility. Our approach should allow clonal tracking at a 1,000-fold greater scale than with nuclear genome sequencing, with simultaneous information on cell state, opening the way to chart cellular dynamics in human health and disease.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/genetics , Base Sequence , Cell Lineage , Chromatin , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Genomics/methods , HEK293 Cells , Hematopoietic Stem Cells/physiology , High-Throughput Nucleotide Sequencing/methods , Humans , Mutation , Single-Cell Analysis , Transposases
9.
Nature ; 560(7718): 325-330, 2018 08.
Article in English | MEDLINE | ID: mdl-30089904

ABSTRACT

Human cancer cell lines are the workhorse of cancer research. Although cell lines are known to evolve in culture, the extent of the resultant genetic and transcriptional heterogeneity and its functional consequences remain understudied. Here we use genomic analyses of 106 human cell lines grown in two laboratories to show extensive clonal diversity. Further comprehensive genomic characterization of 27 strains of the common breast cancer cell line MCF7 uncovered rapid genetic diversification. Similar results were obtained with multiple strains of 13 additional cell lines. Notably, genetic changes were associated with differential activation of gene expression programs and marked differences in cell morphology and proliferation. Barcoding experiments showed that cell line evolution occurs as a result of positive clonal selection that is highly sensitive to culture conditions. Analyses of single-cell-derived clones demonstrated that continuous instability quickly translates into heterogeneity of the cell line. When the 27 MCF7 strains were tested against 321 anti-cancer compounds, we uncovered considerably different drug responses: at least 75% of compounds that strongly inhibited some strains were completely inactive in others. This study documents the extent, origins and consequences of genetic variation within cell lines, and provides a framework for researchers to measure such variation in efforts to support maximally reproducible cancer research.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Evolution, Molecular , Genetic Variation/genetics , Genomic Instability/genetics , Transcription, Genetic/genetics , Breast Neoplasms/pathology , Cell Proliferation , Cell Shape , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Genetic Variation/drug effects , Genomic Instability/drug effects , Humans , MCF-7 Cells , Reproducibility of Results
10.
PLoS Genet ; 12(9): e1006280, 2016 09.
Article in English | MEDLINE | ID: mdl-27618184

ABSTRACT

The use of whole-genome phylogenetic analysis has revolutionized our understanding of the evolution and spread of many important bacterial pathogens due to the high resolution view it provides. However, the majority of such analyses do not consider the potential role of accessory genes when inferring evolutionary trajectories. Moreover, the recently discovered importance of the switching of gene regulatory elements suggests that an exhaustive analysis, combining information from core and accessory genes with regulatory elements could provide unparalleled detail of the evolution of a bacterial population. Here we demonstrate this principle by applying it to a worldwide multi-host sample of the important pathogenic E. coli lineage ST131. Our approach reveals the existence of multiple circulating subtypes of the major drug-resistant clade of ST131 and provides the first ever population level evidence of core genome substitutions in gene regulatory regions associated with the acquisition and maintenance of different accessory genome elements.


Subject(s)
Drug Resistance, Microbial/genetics , Escherichia coli Infections/drug therapy , Escherichia coli/genetics , Evolution, Molecular , Escherichia coli/pathogenicity , Escherichia coli Infections/genetics , Genome, Bacterial/drug effects , Humans , Phylogeny , Regulatory Sequences, Nucleic Acid/genetics , Sequence Analysis, DNA
11.
mBio ; 6(1)2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25587010

ABSTRACT

UNLABELLED: Here we present an extensive genomic and genetic analysis of Escherichia coli strains of serotype O78 that represent the major cause of avian colisepticemia, an invasive infection caused by avian pathogenic Escherichia coli (APEC) strains. It is associated with high mortality and morbidity, resulting in significant economic consequences for the poultry industry. To understand the genetic basis of the virulence of avian septicemic E. coli, we sequenced the entire genome of a clinical isolate of serotype O78-O78:H19 ST88 isolate 789 (O78-9)-and compared it with three publicly available APEC O78 sequences and one complete genome of APEC serotype O1 strain. Although there was a large variability in genome content between the APEC strains, several genes were conserved, which are potentially critical for colisepticemia. Some of these genes are present in multiple copies per genome or code for gene products with overlapping function, signifying their importance. A systematic deletion of each of these virulence-related genes identified three systems that are conserved in all septicemic strains examined and are critical for serum survival, a prerequisite for septicemia. These are the plasmid-encoded protein, the defective ETT2 (E. coli type 3 secretion system 2) type 3 secretion system ETT2sepsis, and iron uptake systems. Strain O78-9 is the only APEC O78 strain that also carried the regulon coding for yersiniabactin, the iron binding system of the Yersinia high-pathogenicity island. Interestingly, this system is the only one that cannot be complemented by other iron uptake systems under iron limitation and in serum. IMPORTANCE: Avian colisepticemia is a severe systemic disease of birds causing high morbidity and mortality and resulting in severe economic losses. The bacteria associated with avian colisepticemia are highly antibiotic resistant, making antibiotic treatment ineffective, and there is no effective vaccine due to the multitude of serotypes involved. To understand the disease and work out strategies to combat it, we performed an extensive genomic and genetic analysis of Escherichia coli strains of serotype O78, the major cause of the disease. We identified several potential virulence factors, conserved in all the colisepticemic strains examined, and determined their contribution to growth in serum, an absolute requirement for septicemia. These findings raise the possibility that specific vaccines or drugs can be developed against these critical virulence factors to help combat this economically important disease.


Subject(s)
Escherichia coli Infections/veterinary , Escherichia coli/genetics , Genome, Bacterial , Poultry Diseases/microbiology , Sepsis/veterinary , Animals , Chickens , Escherichia coli/classification , Escherichia coli/isolation & purification , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Genomics , Molecular Sequence Data , Phylogeny , Sepsis/microbiology , Turkeys , Virulence Factors/genetics , Virulence Factors/metabolism
12.
Proc Natl Acad Sci U S A ; 111(45): 16112-7, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25313052

ABSTRACT

Understanding the mechanisms that generate variation is a common pursuit unifying the life sciences. Bacteria represent an especially striking puzzle, because closely related strains possess radically different metabolic and ecological capabilities. Differences in protein repertoire arising from gene transfer are currently considered the primary mechanism underlying phenotypic plasticity in bacteria. Although bacterial coding plasticity has been extensively studied in previous decades, little is known about the role that regulatory plasticity plays in bacterial evolution. Here, we show that bacterial genes can rapidly shift between multiple regulatory modes by acquiring functionally divergent nonhomologous promoter regions. Through analysis of 270,000 regulatory regions across 247 genomes, we demonstrate that regulatory "switching" to nonhomologous alternatives is ubiquitous, occurring across the bacterial domain. Using comparative transcriptomics, we show that at least 16% of the expression divergence between Escherichia coli strains can be explained by this regulatory switching. Further, using an oligonucleotide regulatory library, we establish that switching affects bacterial promoter architecture. We provide evidence that regulatory switching can occur through horizontal regulatory transfer, which allows regulatory regions to move across strains, and even genera, independently from the genes they regulate. Finally, by experimentally characterizing the fitness effect of a regulatory transfer on a pathogenic E. coli strain, we demonstrate that regulatory switching elicits important phenotypic consequences. Taken together, our findings expose previously unappreciated regulatory plasticity in bacteria and provide a gateway for understanding bacterial phenotypic variation and adaptation.


Subject(s)
Adaptation, Physiological/physiology , DNA, Bacterial/genetics , Escherichia coli/genetics , Genome, Bacterial/physiology , Regulatory Sequences, Nucleic Acid/physiology , DNA, Bacterial/metabolism , Escherichia coli/metabolism , Species Specificity
13.
mBio ; 5(4)2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25118243

ABSTRACT

UNLABELLED: Drug-resistant extraintestinal pathogenic Escherichia coli (ExPEC) strains are the major cause of colisepticemia (colibacillosis), a condition that has become an increasing public health problem in recent years. ExPEC strains are characterized by high resistance to serum, which is otherwise highly toxic to most bacteria. To understand how these bacteria survive and grow in serum, we performed system-wide analyses of their response to serum, making a clear distinction between the responses to nutritional immunity and innate immunity. Thus, mild heat inactivation of serum destroys the immune complement and abolishes the bactericidal effect of serum (inactive serum), making it possible to examine nutritional immunity. We used a combination of deep RNA sequencing and proteomics in order to characterize ExPEC genes whose expression is affected by the nutritional stress of serum and by the immune complement. The major change in gene expression induced by serum-active and inactive-involved metabolic genes. In particular, the serum metabolic response is coordinated by three transcriptional regulators, Fur, BasR, and CysB. Fur alone was responsible for more than 80% of the serum-induced transcriptional response. Consistent with its role as a major serum response regulator, deletion of Fur renders the bacteria completely serum sensitive. These results highlight the role of metabolic adaptation in colisepticemia and virulence. IMPORTANCE: Drug-resistant extraintestinal pathogenic Escherichia coli (ExPEC) strains have emerged as major pathogens, especially in community- and hospital-acquired infections. These bacteria cause a large spectrum of syndromes, the most serious of which is septicemia, a condition with a high mortality rate. These bacterial strains are characterized by high resistance to serum, otherwise highly toxic to most bacteria. To understand the basis of this resistance, we carried out system-wide analyses of the response of ExPEC strains to serum by using proteomics and deep RNA sequencing. The major changes in gene expression induced by exposure to serum involved metabolic genes, not necessarily implicated in relation to virulence. One metabolic regulator-Fur-involved in iron metabolism was responsible for more than 80% of the serum-induced response, and its deletion renders the bacteria completely serum sensitive. These results highlight the role of metabolic adaptation in virulence.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/physiology , Escherichia coli Proteins/genetics , Escherichia coli/growth & development , Escherichia coli/pathogenicity , Repressor Proteins/genetics , Repressor Proteins/physiology , Serum/microbiology , Blood Bactericidal Activity , Complement System Proteins/immunology , Escherichia coli/genetics , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genome, Bacterial , Humans , Male , Proteomics , Sepsis/microbiology , Sequence Analysis, RNA
14.
Mol Biol Evol ; 28(12): 3309-18, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21690561

ABSTRACT

Bacterial genes are commonly encoded in clusters, known as operons, which share transcriptional regulatory control and often encode functionally related proteins that take part in certain biological pathways. Operons that are coregulated are known to colocalize in the genome, suggesting that their spatial organization is under selection for efficient expression regulation. However, the internal order of genes within operons is believed to be poorly conserved, and hence expression requirements are claimed to be too weak to oppose gene rearrangements. In light of these opposing views, we set out to investigate whether the internal location of the regulatory genes within operons is under selection. Our analysis shows that transcription factors (TFs) are preferentially encoded as either first or last in their operons, in the two diverged model bacteria Escherichia coli and Bacillus subtilis. In a higher resolution, we find that TFs that repress transcription of the operon in which they are encoded (autorepressors), contribute most of this signal by specific preference of the first operon position. We show that this trend is strikingly conserved throughout highly diverged bacterial phyla. Moreover, these autorepressors regulate operons that carry out highly diverse biological functions. We propose a model according to which autorepressors are selected to be located first in their operons in order to optimize transcription regulation. Specifically, the first operon position helps autorepressors to minimize leaky transcription of the operon structural genes, thus minimizing energy waste. Our analysis provides statistically robust evidence for a paradigm of bacterial autorepressor preferential operonic location. Corroborated with our suggested model, an additional layer of operon expression control that is common throughout the bacterial domain is revealed.


Subject(s)
Bacillus subtilis/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Operon , Regulatory Elements, Transcriptional/genetics , Transcription, Genetic , Bacterial Proteins/genetics , DNA-Binding Proteins/genetics , Escherichia coli Proteins/genetics , Evolution, Molecular , Repressor Proteins/genetics , Selection, Genetic , Signal Transduction , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...