Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Stem Cell ; 27(2): 284-299.e8, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32693087

ABSTRACT

SMAD pathways govern epithelial proliferation, and transforming growth factor ß (TGF-ß and BMP signaling through SMAD members has distinct effects on mammary development and homeostasis. Here, we show that LEFTY1, a secreted inhibitor of NODAL/SMAD2 signaling, is produced by mammary progenitor cells and, concomitantly, suppresses SMAD2 and SMAD5 signaling to promote long-term proliferation of normal and malignant mammary epithelial cells. In contrast, BMP7, a NODAL antagonist with context-dependent functions, is produced by basal cells and restrains progenitor cell proliferation. In normal mouse epithelium, LEFTY1 expression in a subset of luminal cells and rare basal cells opposes BMP7 to promote ductal branching. LEFTY1 binds BMPR2 to suppress BMP7-induced activation of SMAD5, and this LEFTY1-BMPR2 interaction is specific to tumor-initiating cells in triple-negative breast cancer xenografts that rely on LEFTY1 for growth. These results suggest that LEFTY1 is an endogenous dual-SMAD inhibitor and that suppressing its function may represent a therapeutic vulnerability in breast cancer.


Subject(s)
Signal Transduction , Transforming Growth Factor beta , Animals , Carcinogenesis , Cell Transformation, Neoplastic , Mice
2.
Nat Commun ; 8(1): 1669, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29162812

ABSTRACT

Previous studies have proposed that epithelial to mesenchymal transition (EMT) in breast cancer cells regulates metastasis, stem cell properties and chemo-resistance; most studies were based on in vitro culture of cell lines and mouse transgenic cancer models. However, the identity and function of cells expressing EMT-associated genes in normal murine mammary gland homeostasis and human breast cancer still remains under debate. Using in vivo lineage tracing and triple negative breast cancer (TNBC) patient derived xenografts we demonstrate that the repopulating capacity in normal mammary epithelial cells and tumorigenic capacity in TNBC is independent of expression of EMT-associated genes. In breast cancer, while a subset of cells with epithelial and mesenchymal phenotypes have stem cell activity, in many cells that have lost epithelial characteristics with increased expression of mesenchymal genes, have decreased tumor-initiating capacity and plasticity. These findings have implications for the development of effective therapeutic agents targeting tumor-initiating cells.


Subject(s)
Breast/metabolism , Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Triple Negative Breast Neoplasms/genetics , Animals , Breast/cytology , Breast/physiology , Epithelial Cells/metabolism , Female , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Regeneration/genetics , Transplantation, Heterologous , Triple Negative Breast Neoplasms/pathology
3.
Cell Rep ; 20(13): 3236-3247, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28954238

ABSTRACT

How transcription factors (TFs) reprogram one cell lineage to another remains unclear. Here, we define chromatin accessibility changes induced by the proneural TF Ascl1 throughout conversion of fibroblasts into induced neuronal (iN) cells. Thousands of genomic loci are affected as early as 12 hr after Ascl1 induction. Surprisingly, over 80% of the accessibility changes occur between days 2 and 5 of the 3-week reprogramming process. This chromatin switch coincides with robust activation of endogenous neuronal TFs and nucleosome phasing of neuronal promoters and enhancers. Subsequent morphological and functional maturation of iN cells is accomplished with relatively little chromatin reconfiguration. By integrating chromatin accessibility and transcriptome changes, we built a network model of dynamic TF regulation during iN cell reprogramming and identified Zfp238, Sox8, and Dlx3 as key TFs downstream of Ascl1. These results reveal a singular, coordinated epigenomic switch during direct reprogramming, in contrast to stepwise cell fate transitions in development.


Subject(s)
Chromatin/metabolism , Fibroblasts/metabolism , Neurons/metabolism , Cellular Reprogramming , Humans
4.
Cell Stem Cell ; 20(2): 247-260.e5, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28041896

ABSTRACT

Stem cells in many tissues sustain themselves by entering a quiescent state to avoid genomic insults and to prevent exhaustion caused by excessive proliferation. In the mammary gland, the identity and characteristics of quiescent epithelial stem cells are not clear. Here, we identify a quiescent mammary epithelial cell population expressing high levels of Bcl11b and located at the interface between luminal and basal cells. Bcl11bhigh cells are enriched for cells that can regenerate mammary glands in secondary transplants. Loss of Bcl11b leads to a Cdkn2a-dependent exhaustion of ductal epithelium and loss of epithelial cell regenerative capacity. Gain- and loss-of-function studies show that Bcl11b induces cells to enter the G0 phase of the cell cycle and become quiescent. Taken together, these results suggest that Bcl11b acts as a central intrinsic regulator of mammary epithelial stem cell quiescence and exhaustion and is necessary for long-term maintenance of the mammary gland.


Subject(s)
Cell Cycle , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Repressor Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Antigens, CD/metabolism , Cell Lineage , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/metabolism , Female , Gene Deletion , Homeostasis , Mammary Glands, Animal/growth & development , Mice, Inbred C57BL , Mice, Knockout , Regeneration/physiology
5.
Dev Cell ; 39(4): 491-507, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27840109

ABSTRACT

Embryonic gene expression intricately reflects anatomical context, developmental stage, and cell type. To address whether the precise spatial origins of cardiac cells can be deduced solely from their transcriptional profiles, we established a genome-wide expression database from 118, 949, and 1,166 single murine heart cells at embryonic day 8.5 (e8.5), e9.5, and e10.5, respectively. We segregated these cells by type using unsupervised bioinformatics analysis and identified chamber-specific genes. Using a random forest algorithm, we reconstructed the spatial origin of single e9.5 and e10.5 cardiomyocytes with 92.0% ± 3.2% and 91.2% ± 2.8% accuracy, respectively (99.4% ± 1.0% and 99.1% ± 1.1% if a ±1 zone margin is permitted) and predicted the second heart field distribution of Isl-1-lineage descendants. When applied to Nkx2-5-/- cardiomyocytes from murine e9.5 hearts, we showed their transcriptional alteration and lack of ventricular phenotype. Our database and zone classification algorithm will enable the discovery of novel mechanisms in early cardiac development and disease.


Subject(s)
Gene Expression Profiling , Heart/embryology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Single-Cell Analysis , Algorithms , Animals , Biomarkers/metabolism , Cell Lineage/genetics , Cell Separation , Embryo, Mammalian/cytology , Embryoid Bodies/cytology , Gene Expression Regulation, Developmental , Integrases/metabolism , Mice , Models, Biological , Myocardium/metabolism , Polymerase Chain Reaction , Reproducibility of Results , Sequence Analysis, RNA
6.
Nature ; 534(7607): 391-5, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27281220

ABSTRACT

Direct lineage reprogramming represents a remarkable conversion of cellular and transcriptome states. However, the intermediate stages through which individual cells progress during reprogramming are largely undefined. Here we use single-cell RNA sequencing at multiple time points to dissect direct reprogramming from mouse embryonic fibroblasts to induced neuronal cells. By deconstructing heterogeneity at each time point and ordering cells by transcriptome similarity, we find that the molecular reprogramming path is remarkably continuous. Overexpression of the proneural pioneer factor Ascl1 results in a well-defined initialization, causing cells to exit the cell cycle and re-focus gene expression through distinct neural transcription factors. The initial transcriptional response is relatively homogeneous among fibroblasts, suggesting that the early steps are not limiting for productive reprogramming. Instead, the later emergence of a competing myogenic program and variable transgene dynamics over time appear to be the major efficiency limits of direct reprogramming. Moreover, a transcriptional state, distinct from donor and target cell programs, is transiently induced in cells undergoing productive reprogramming. Our data provide a high-resolution approach for understanding transcriptome states during lineage differentiation.


Subject(s)
Cellular Reprogramming/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Neurons/cytology , Neurons/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/genetics , Cell Lineage/genetics , Cell Transdifferentiation/genetics , Embryo, Mammalian/cytology , Gene Expression Profiling , Gene Silencing , Homeodomain Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , POU Domain Factors/metabolism , Time Factors , Transcription Factors/metabolism , Transcriptome/genetics , Transgenes/genetics
7.
Nat Methods ; 11(1): 41-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24141493

ABSTRACT

Interest in single-cell whole-transcriptome analysis is growing rapidly, especially for profiling rare or heterogeneous populations of cells. We compared commercially available single-cell RNA amplification methods with both microliter and nanoliter volumes, using sequence from bulk total RNA and multiplexed quantitative PCR as benchmarks to systematically evaluate the sensitivity and accuracy of various single-cell RNA-seq approaches. We show that single-cell RNA-seq can be used to perform accurate quantitative transcriptome measurement in individual cells with a relatively small number of sequencing reads and that sequencing large numbers of single cells can recapitulate bulk transcriptome complexity.


Subject(s)
Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Data Interpretation, Statistical , Electronic Data Processing , Gene Expression Profiling , Gene Expression Regulation , HCT116 Cells , Humans , Microfluidics , Nucleic Acid Amplification Techniques , Polymerase Chain Reaction/methods , Reproducibility of Results , Sequence Analysis, DNA , Transcriptome
8.
Gastroenterology ; 142(5): 1195-1205.e6, 2012 May.
Article in English | MEDLINE | ID: mdl-22333952

ABSTRACT

BACKGROUND & AIMS: Paneth cells contribute to the small intestinal niche of Lgr5(+) stem cells. Although the colon also contains Lgr5(+) stem cells, it does not contain Paneth cells. We investigated the existence of colonic Paneth-like cells that have a distinct transcriptional signature and support Lgr5(+) stem cells. METHODS: We used multicolor fluorescence-activated cell sorting to isolate different subregions of colon crypts, based on known markers, from dissociated colonic epithelium of mice. We performed multiplexed single-cell gene expression analysis with quantitative reverse transcriptase polymerase chain reaction followed by hierarchical clustering analysis to characterize distinct cell types. We used immunostaining and fluorescence-activated cell sorting analyses with in vivo administration of a Notch inhibitor and in vitro organoid cultures to characterize different cell types. RESULTS: Multicolor fluorescence-activated cell sorting could isolate distinct regions of colonic crypts. Four major epithelial subtypes or transcriptional states were revealed by gene expression analysis of selected populations of single cells. One of these, the goblet cells, contained a distinct cKit/CD117(+) crypt base subpopulation that expressed Dll1, Dll4, and epidermal growth factor, similar to Paneth cells, which were also marked by cKit. In the colon, cKit(+) goblet cells were interdigitated with Lgr5(+) stem cells. In vivo, this colonic cKit(+) population was regulated by Notch signaling; administration of a γ-secretase inhibitor to mice increased the number of cKit(+) cells. When isolated from mouse colon, cKit(+) cells promoted formation of organoids from Lgr5(+) stem cells, which expressed Kitl/stem cell factor, the ligand for cKit. When organoids were depleted of cKit(+) cells using a toxin-conjugated antibody, organoid formation decreased. CONCLUSIONS: cKit marks small intestinal Paneth cells and a subset of colonic goblet cells that are regulated by Notch signaling and support Lgr5(+) stem cells.


Subject(s)
Colon/cytology , Paneth Cells/chemistry , Paneth Cells/physiology , Proto-Oncogene Proteins c-kit/analysis , Receptors, G-Protein-Coupled/analysis , Stem Cells/physiology , Animals , Antigens, CD/analysis , Cell Adhesion Molecules/analysis , Cells, Cultured , Flow Cytometry , Gene Expression Profiling , Goblet Cells/physiology , Hyaluronan Receptors/analysis , Mice , Mice, Inbred C57BL , Receptors, Notch/physiology , Single-Cell Analysis , Stem Cells/chemistry
9.
Nat Biotechnol ; 29(12): 1120-7, 2011 Nov 13.
Article in English | MEDLINE | ID: mdl-22081019

ABSTRACT

Cancer is often viewed as a caricature of normal developmental processes, but the extent to which its cellular heterogeneity truly recapitulates multilineage differentiation processes of normal tissues remains unknown. Here we implement single-cell PCR gene-expression analysis to dissect the cellular composition of primary human normal colon and colon cancer epithelia. We show that human colon cancer tissues contain distinct cell populations whose transcriptional identities mirror those of the different cellular lineages of normal colon. By creating monoclonal tumor xenografts from injection of a single (n = 1) cell, we demonstrate that the transcriptional diversity of cancer tissues is largely explained by in vivo multilineage differentiation and not only by clonal genetic heterogeneity. Finally, we show that the different gene-expression programs linked to multilineage differentiation are strongly associated with patient survival. We develop two-gene classifier systems (KRT20 versus CA1, MS4A12, CD177, SLC26A3) that predict clinical outcomes with hazard ratios superior to those of pathological grade and comparable to those of microarray-derived multigene expression signatures.


Subject(s)
Adenocarcinoma/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Single-Cell Analysis/methods , Transcription, Genetic , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Disease-Free Survival , Female , Flow Cytometry , HCT116 Cells , Humans , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Neoplasm Staging , Transplantation, Heterologous , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...