Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell ; 175(7): 1972-1988.e16, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30550791

ABSTRACT

In vitro cancer cultures, including three-dimensional organoids, typically contain exclusively neoplastic epithelium but require artificial reconstitution to recapitulate the tumor microenvironment (TME). The co-culture of primary tumor epithelia with endogenous, syngeneic tumor-infiltrating lymphocytes (TILs) as a cohesive unit has been particularly elusive. Here, an air-liquid interface (ALI) method propagated patient-derived organoids (PDOs) from >100 human biopsies or mouse tumors in syngeneic immunocompetent hosts as tumor epithelia with native embedded immune cells (T, B, NK, macrophages). Robust droplet-based, single-cell simultaneous determination of gene expression and immune repertoire indicated that PDO TILs accurately preserved the original tumor T cell receptor (TCR) spectrum. Crucially, human and murine PDOs successfully modeled immune checkpoint blockade (ICB) with anti-PD-1- and/or anti-PD-L1 expanding and activating tumor antigen-specific TILs and eliciting tumor cytotoxicity. Organoid-based propagation of primary tumor epithelium en bloc with endogenous immune stroma should enable immuno-oncology investigations within the TME and facilitate personalized immunotherapy testing.


Subject(s)
Models, Immunological , Neoplasms, Experimental/immunology , Organoids/immunology , Receptors, Antigen, T-Cell/immunology , Tumor Microenvironment/immunology , Animals , B7-H1 Antigen/immunology , Coculture Techniques , Female , Humans , Immunotherapy , Male , Mice , Mice, Inbred BALB C , Neoplasm Proteins/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Organoids/pathology
2.
Nature ; 618(7964): 383-393, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37258665

ABSTRACT

The earliest events during human tumour initiation, although poorly characterized, may hold clues to malignancy detection and prevention1. Here we model occult preneoplasia by biallelic inactivation of TP53, a common early event in gastric cancer, in human gastric organoids. Causal relationships between this initiating genetic lesion and resulting phenotypes were established using experimental evolution in multiple clonally derived cultures over 2 years. TP53 loss elicited progressive aneuploidy, including copy number alterations and structural variants prevalent in gastric cancers, with evident preferred orders. Longitudinal single-cell sequencing of TP53-deficient gastric organoids similarly indicates progression towards malignant transcriptional programmes. Moreover, high-throughput lineage tracing with expressed cellular barcodes demonstrates reproducible dynamics whereby initially rare subclones with shared transcriptional programmes repeatedly attain clonal dominance. This powerful platform for experimental evolution exposes stringent selection, clonal interference and a marked degree of phenotypic convergence in premalignant epithelial organoids. These data imply predictability in the earliest stages of tumorigenesis and show evolutionary constraints and barriers to malignant transformation, with implications for earlier detection and interception of aggressive, genome-instable tumours.


Subject(s)
Cell Transformation, Neoplastic , Clonal Evolution , Precancerous Conditions , Selection, Genetic , Stomach Neoplasms , Humans , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Clonal Evolution/genetics , Genomic Instability , Mutation , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Organoids/metabolism , Organoids/pathology , Aneuploidy , DNA Copy Number Variations , Single-Cell Analysis , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Disease Progression , Cell Lineage
3.
Nat Mater ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965405

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by its fibrotic and stiff extracellular matrix. However, how the altered cell/extracellular-matrix signalling contributes to the PDAC tumour phenotype has been difficult to dissect. Here we design and engineer matrices that recapitulate the key hallmarks of the PDAC tumour extracellular matrix to address this knowledge gap. We show that patient-derived PDAC organoids from three patients develop resistance to several clinically relevant chemotherapies when cultured within high-stiffness matrices mechanically matched to in vivo tumours. Using genetic barcoding, we find that while matrix-specific clonal selection occurs, cellular heterogeneity is not the main driver of chemoresistance. Instead, matrix-induced chemoresistance occurs within a stiff environment due to the increased expression of drug efflux transporters mediated by CD44 receptor interactions with hyaluronan. Moreover, PDAC chemoresistance is reversible following transfer from high- to low-stiffness matrices, suggesting that targeting the fibrotic extracellular matrix may sensitize chemoresistant tumours. Overall, our findings support the potential of engineered matrices and patient-derived organoids for elucidating extracellular matrix contributions to human disease pathophysiology.

4.
Blood ; 129(7): e13-e25, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28003272

ABSTRACT

Acute myeloid leukemia (AML) is characterized by an impaired differentiation process leading to an accumulation of immature blasts in the blood. One feature of cytogenetically normal AML is alterations to the DNA methylome. We analyzed 57 AML patients with normal karyotype by using Illumina's 450k array and showed that aberrant DNA methylation is significantly altered at enhancer regions and that the methylation levels at specific enhancers predict overall survival of AML patients. The majority of sites that become differentially methylated in AML occur in regulatory elements of the human genome. Hypermethylation associates with enhancer silencing. In addition, chromatin immunoprecipitation sequencing analyses showed that a subset of hypomethylated sites correlate with enhancer activation, indicated by increased H3K27 acetylation. DNA hypomethylation is therefore not sufficient for enhancer activation. Some sites of hypomethylation occur at weak/poised enhancers marked with H3K4 monomethylation in hematopoietic progenitor cells. Other hypomethylated regions occur at sites inactive in progenitors and reflect the de novo acquisition of AML-specific enhancers. Altered enhancer dynamics are reflected in the gene expression of enhancer target genes, including genes involved in oncogenesis and blood cell development. This study demonstrates that histone variants and different histone modifications interact with aberrant DNA methylation and cause perturbed enhancer activity in cytogenetically normal AML that contributes to a leukemic transcriptome.


Subject(s)
DNA Methylation , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Bone Marrow/metabolism , Bone Marrow/pathology , Enhancer Elements, Genetic , Histone Code , Histones/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Promoter Regions, Genetic , Transcriptome
5.
Mol Syst Biol ; 13(5): 930, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28495919

ABSTRACT

Alternative transcription start sites (TSSs) have been extensively studied genome-wide for many cell types and have been shown to be important during development and to regulate transcript abundance between cell types. Likewise, single-cell gene expression has been extensively studied for many cell types. However, how single cells use TSSs has not yet been examined. In particular, it is unknown whether alternative TSSs are independently expressed, or whether they are co-activated or even mutually exclusive in single cells. Here, we use a previously published single-cell RNA-seq dataset, comprising thousands of cells, to study alternative TSS usage. We find that alternative TSS usage is a regulated process, and the correlation between two TSSs expressed in single cells of the same cell type is surprisingly high. Our findings indicate that TSSs generally are regulated by common factors rather than being independently regulated or stochastically expressed.


Subject(s)
Brain/cytology , Transcription Initiation Site , Animals , Base Sequence , Cystatin C/genetics , Gene Expression , Mice , Single-Cell Analysis
6.
BMC Genomics ; 18(1): 126, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28158971

ABSTRACT

BACKGROUND: Alternative mRNA isoform usage is an important source of protein diversity in mammalian cells. This phenomenon has been extensively studied in bulk tissues, however, it remains unclear how this diversity is reflected in single cells. RESULTS: Here we use long-read sequencing technology combined with unique molecular identifiers (UMIs) to reveal patterns of alternative full-length isoform expression in single cells from the mouse brain. We found a surprising amount of isoform diversity, even after applying a conservative definition of what constitutes an isoform. Genes tend to have one or a few isoforms highly expressed and a larger number of isoforms expressed at a low level. However, for many genes, nearly every sequenced mRNA molecule was unique, and many events affected coding regions suggesting previously unknown protein diversity in single cells. Exon junctions in coding regions were less prone to splicing errors than those in non-coding regions, indicating purifying selection on splice donor and acceptor efficiency. CONCLUSIONS: Our findings indicate that mRNA isoform diversity is an important source of biological variability also in single cells.


Subject(s)
Alternative Splicing , Brain/metabolism , RNA, Messenger/genetics , Single-Cell Analysis , Animals , Computational Biology/methods , Genetic Heterogeneity , High-Throughput Nucleotide Sequencing , Mice , RNA Isoforms , Single-Cell Analysis/methods
7.
Genomics ; 105(3): 150-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25543032

ABSTRACT

Cell-free DNA has been used for fetal rhesus factor and sex determination, fetal aneuploidy screening, cancer diagnostics and monitoring, and other applications. However current methods of using cell free DNA require amplification, which leads to allelic dropout and bias especially when starting with small amounts of DNA. Here we describe an amplification-free method for sequencing of cell-free DNA, even from low levels of starting material. We evaluated this method in the context of prenatal diagnosis of fetal aneuploidy and compared it with a PCR-based library preparation method as well as a recently described method using unique molecular identifiers (UMI). All methods performed well, however coverage was increased by the amplification-free method and GC-induced bias was reduced by both the amplification-free method and the UMI method. Future diagnostic applications including whole genome sequencing of cell-free DNA will benefit from amplification-free sequencing.


Subject(s)
Aneuploidy , Genetic Testing/methods , Prenatal Diagnosis/methods , Sequence Analysis, DNA/methods , Female , Humans , Polymerase Chain Reaction , Pregnancy
8.
Nat Methods ; 9(1): 72-4, 2011 Nov 20.
Article in English | MEDLINE | ID: mdl-22101854

ABSTRACT

Counting individual RNA or DNA molecules is difficult because they are hard to copy quantitatively for detection. To overcome this limitation, we applied unique molecular identifiers (UMIs), which make each molecule in a population distinct, to genome-scale human karyotyping and mRNA sequencing in Drosophila melanogaster. Use of this method can improve accuracy of almost any next-generation sequencing method, including chromatin immunoprecipitation-sequencing, genome assembly, diagnostics and manufacturing-process control and monitoring.


Subject(s)
Genomics/methods , Karyotyping/methods , RNA, Messenger/analysis , Sequence Analysis, RNA/methods , Animals , Chromatin Immunoprecipitation/methods , Down Syndrome/genetics , Drosophila melanogaster , Female , Gene Library , Humans , Male
9.
Cell Syst ; 14(9): 764-776.e6, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37734323

ABSTRACT

Organoids are powerful experimental models for studying the ontogeny and progression of various diseases including cancer. Organoids are conventionally cultured in bulk using an extracellular matrix mimic. However, bulk-cultured organoids physically overlap, making it impossible to track the growth of individual organoids over time in high throughput. Moreover, local spatial variations in bulk matrix properties make it difficult to assess whether observed phenotypic heterogeneity between organoids results from intrinsic cell differences or differences in the microenvironment. Here, we developed a microwell-based method that enables high-throughput quantification of image-based parameters for organoids grown from single cells, which can further be retrieved from their microwells for molecular profiling. Coupled with a deep learning image-processing pipeline, we characterized phenotypic traits including growth rates, cellular movement, and apical-basal polarity in two CRISPR-engineered human gastric organoid models, identifying genomic changes associated with increased growth rate and changes in accessibility and expression correlated with apical-basal polarity. A record of this paper's transparent peer review process is included in the supplemental information.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Extracellular Matrix , Humans , Cell Movement , Genomics , Organoids
10.
Cancer Discov ; 11(6): 1562-1581, 2021 06.
Article in English | MEDLINE | ID: mdl-33451982

ABSTRACT

Mutations in ARID1A rank among the most common molecular aberrations in human cancer. However, oncogenic consequences of ARID1A mutation in human cells remain poorly defined due to lack of forward genetic models. Here, CRISPR/Cas9-mediated ARID1A knockout (KO) in primary TP53-/- human gastric organoids induced morphologic dysplasia, tumorigenicity, and mucinous differentiation. Genetic WNT/ß-catenin activation rescued mucinous differentiation, but not hyperproliferation, suggesting alternative pathways of ARID1A KO-mediated transformation. ARID1A mutation induced transcriptional regulatory modules characteristic of microsatellite instability and Epstein-Barr virus-associated subtype human gastric cancer, including FOXM1-associated mitotic genes and BIRC5/survivin. Convergently, high-throughput compound screening indicated selective vulnerability of ARID1A-deficient organoids to inhibition of BIRC5/survivin, functionally implicating this pathway as an essential mediator of ARID1A KO-dependent early-stage gastric tumorigenesis. Overall, we define distinct pathways downstream of oncogenic ARID1A mutation, with nonessential WNT-inhibited mucinous differentiation in parallel with essential transcriptional FOXM1/BIRC5-stimulated proliferation, illustrating the general utility of organoid-based forward genetic cancer analysis in human cells. SIGNIFICANCE: We establish the first human forward genetic modeling of a commonly mutated tumor suppressor gene, ARID1A. Our study integrates diverse modalities including CRISPR/Cas9 genome editing, organoid culture, systems biology, and small-molecule screening to derive novel insights into early transformation mechanisms of ARID1A-deficient gastric cancers.See related commentary by Zafra and Dow, p. 1327.This article is highlighted in the In This Issue feature, p. 1307.


Subject(s)
CRISPR-Cas Systems , Cell Transformation, Neoplastic , DNA-Binding Proteins/genetics , Stomach Neoplasms/genetics , Transcription Factors/genetics , Humans , Models, Biological , Mutation
11.
Nat Cancer ; 1(8): 761-773, 2020 08.
Article in English | MEDLINE | ID: mdl-34142093

ABSTRACT

Organoid technologies enable the creation of in vitro physiologic systems that model tissues of origin more accurately than classical culture approaches. Seminal characteristics, including three-dimensional structure and recapitulation of self-renewal, differentiation, and disease pathology, render organoids eminently suited as hybrids that combine the experimental tractability of traditional 2D cell lines with cellular attributes of in vivo model systems. Here, we describe recent advances in this rapidly evolving field and their applications in cancer biology, clinical translation and precision medicine.


Subject(s)
Neoplasms , Organoids , Biology , Cell Differentiation , Humans , Neoplasms/therapy , Organoids/pathology , Precision Medicine/methods
12.
Stem Cell Res Ther ; 8(1): 250, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29116032

ABSTRACT

Regulation of adipose tissue stem cells (ASCs) and adipogenesis impact the development of excess body fat-related metabolic complications. Animal studies have suggested the presence of distinct subtypes of ASCs with different differentiation properties. In addition, ASCs are becoming the biggest source of mesenchymal stem cells used in therapies, which requires deep characterization. Using unbiased single cell transcriptomics we aimed to characterize ASC populations in human subcutaneous white adipose tissue (scWAT). The transcriptomes of 574 single cells from the WAT total stroma vascular fraction (SVF) of four healthy women were analyzed by clustering and t-distributed stochastic neighbor embedding visualization. The identified cell populations were then mapped to cell types present in WAT using data from gene expression microarray profiling of flow cytometry-sorted SVF. Cells clustered into four distinct populations: three adipose tissue-resident macrophage subtypes and one large, homogeneous population of ASCs. While pseudotemporal ordering analysis indicated that the ASCs were in slightly different differentiation stages, the differences in gene expression were small and could not distinguish distinct ASC subtypes. Altogether, in healthy individuals, ASCs seem to constitute a single homogeneous cell population that cannot be subdivided by single cell transcriptomics, suggesting a common origin for human adipocytes in scWAT.


Subject(s)
Adipocytes/metabolism , Mesenchymal Stem Cells/metabolism , Stem Cells/metabolism , Cell Differentiation , Cells, Cultured , Female , Humans , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL