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.
Proc Natl Acad Sci U S A ; 116(52): 26599-26605, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31843916

ABSTRACT

Cycling intestinal Lgr5+ stem cells are intermingled with their terminally differentiated Paneth cell daughters at crypt bottoms. Paneth cells provide multiple secreted (e.g., Wnt, EGF) as well as surface-bound (Notch ligand) niche signals. Here we show that ablation of Paneth cells in mice, using a diphtheria toxin receptor gene inserted into the P-lysozyme locus, does not affect the maintenance of Lgr5+ stem cells. Flow cytometry, single-cell sequencing, and histological analysis showed that the ablated Paneth cells are replaced by enteroendocrine and tuft cells. As these cells physically occupy Paneth cell positions between Lgr5 stem cells, they serve as an alternative source of Notch signals, which are essential for Lgr5+ stem cell maintenance. Our combined in vivo results underscore the adaptive flexibility of the intestine in maintaining normal tissue homeostasis.

2.
Dev Cell ; 46(3): 285-301.e9, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30086300

ABSTRACT

Phase separation represents an important form of subcellular compartmentalization. However, relatively little is known about how the formation or disassembly of such compartments is regulated. In zebrafish, the Balbiani body (Bb) and the germ plasm (Gp) are intimately linked phase-separated structures essential for germ cell specification and home to many germ cell-specific mRNAs and proteins. Throughout development, these structures occur as a single large aggregate (Bb), which disperses throughout oogenesis and upon fertilization accumulates again into relatively large assemblies (Gp). Formation of the Bb requires Bucky ball (Buc), a protein with prion-like properties. We found that the multi-tudor domain-containing protein Tdrd6a interacts with Buc, affecting its mobility and aggregation properties. Importantly, lack of this regulatory interaction leads to significant defects in germ cell development. Our work presents insights into how prion-like protein aggregations can be regulated and highlights the biological relevance of such regulatory events.


Subject(s)
Germ Cells/metabolism , Oocytes/metabolism , Oogenesis/physiology , Zebrafish Proteins/metabolism , Animals , Cytoplasm/metabolism , Organelles/metabolism , RNA, Messenger/metabolism , Zebrafish
3.
Proc Natl Acad Sci U S A ; 115(4): E610-E619, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29311336

ABSTRACT

The adult mouse subependymal zone provides a niche for mammalian neural stem cells (NSCs). However, the molecular signature, self-renewal potential, and fate behavior of NSCs remain poorly defined. Here we propose a model in which the fate of active NSCs is coupled to the total number of neighboring NSCs in a shared niche. Using knock-in reporter alleles and single-cell RNA sequencing, we show that the Wnt target Tnfrsf19/Troy identifies both active and quiescent NSCs. Quantitative analysis of genetic lineage tracing of individual NSCs under homeostasis or in response to injury reveals rapid expansion of stem-cell number before some return to quiescence. This behavior is best explained by stochastic fate decisions, where stem-cell number within a shared niche fluctuates over time. Fate mapping proliferating cells using a Ki67iresCreER allele confirms that active NSCs reversibly return to quiescence, achieving long-term self-renewal. Our findings suggest a niche-based mechanism for the regulation of NSC fate and number.


Subject(s)
Lateral Ventricles/cytology , Neural Stem Cells/physiology , Stem Cell Niche , Animals , Cell Lineage , Cell Proliferation , Mice , Neurogenesis , Receptors, Tumor Necrosis Factor/metabolism , Single-Cell Analysis , Transcriptome
4.
Cell Stem Cell ; 20(2): 177-190.e4, 2017 02 02.
Article in English | MEDLINE | ID: mdl-27939219

ABSTRACT

Lgr5+ adult intestinal stem cells are highly proliferative throughout life. Single Lgr5+ stem cells can be cultured into three-dimensional organoids containing all intestinal epithelial cell types at near-normal ratios. Conditions to generate the main cell types (enterocyte, goblet cells, Paneth cells, and M cells) are well established, but signals to induce the spectrum of hormone-producing enteroendocrine cells (EECs) have remained elusive. Here, we induce Lgr5+ stem cell quiescence in vitro by blocking epidermal growth factor receptor (EGFR) or mitogen-associated protein kinase (MAPK) signaling pathways in organoids and show that their quiescent state is readily reverted. Quiescent Lgr5+ stem cells acquire a distinct molecular signature biased toward EEC differentiation. Indeed, combined inhibition of Wnt, Notch, and MAPK pathways efficiently generates a diversity of EEC hormone-expressing subtypes in vitro. Our observations uncouple Wnt-dependent stem cell maintenance from EGF-dependent proliferation and provide an approach for the study of the elusive EECs in a defined environment.


Subject(s)
Cell Cycle , Cell Differentiation , Enteroendocrine Cells/cytology , Hormones/biosynthesis , Intestines/cytology , Organoids/cytology , Receptors, G-Protein-Coupled/metabolism , Stem Cells/cytology , Animals , Cell Lineage , Cell Proliferation , Cells, Cultured , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gene Expression Profiling , MAP Kinase Signaling System , Mice , Receptors, Notch/metabolism , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Wnt Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 113(37): E5399-407, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27573849

ABSTRACT

Leucine-rich repeat-containing G-protein coupled receptor 5-positive (Lgr5(+)) stem cells reside at crypt bottoms of the small and large intestine. Small intestinal Paneth cells supply Wnt3, EGF, and Notch signals to neighboring Lgr5(+) stem cells. Whereas the colon lacks Paneth cells, deep crypt secretory (DCS) cells are intermingled with Lgr5(+) stem cells at crypt bottoms. Here, we report regenerating islet-derived family member 4 (Reg4) as a marker of DCS cells. To investigate a niche function, we eliminated DCS cells by using the diphtheria-toxin receptor gene knocked into the murine Reg4 locus. Ablation of DCS cells results in loss of stem cells from colonic crypts and disrupts gut homeostasis and colon organoid growth. In agreement, sorted Reg4(+) DCS cells promote organoid formation of single Lgr5(+) colon stem cells. DCS cells can be massively produced from Lgr5(+) colon stem cells in vitro by combined Notch inhibition and Wnt activation. We conclude that Reg4(+) DCS cells serve as Paneth cell equivalents in the colon crypt niche.


Subject(s)
Colonic Neoplasms/metabolism , Neoplasm Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Stem Cells/metabolism , Animals , Colon/cytology , Colon/growth & development , Colon/metabolism , Colonic Neoplasms/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Intestine, Small/cytology , Intestine, Small/metabolism , Mice , Neoplasm Proteins/metabolism , Organoids/growth & development , Organoids/metabolism , Pancreatitis-Associated Proteins , Paneth Cells/cytology , Paneth Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Notch/genetics , Stem Cell Niche/genetics , Stem Cells/cytology , Wnt Signaling Pathway/genetics
6.
Cell Stem Cell ; 19(2): 266-277, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27345837

ABSTRACT

Adult mitotic tissues like the intestine, skin, and blood undergo constant turnover throughout the life of an organism. Knowing the identity of the stem cell is crucial to understanding tissue homeostasis and its aberrations upon disease. Here we present a computational method for the derivation of a lineage tree from single-cell transcriptome data. By exploiting the tree topology and the transcriptome composition, we establish StemID, an algorithm for identifying stem cells among all detectable cell types within a population. We demonstrate that StemID recovers two known adult stem cell populations, Lgr5+ cells in the small intestine and hematopoietic stem cells in the bone marrow. We apply StemID to predict candidate multipotent cell populations in the human pancreas, a tissue with largely uncharacterized turnover dynamics. We hope that StemID will accelerate the search for novel stem cells by providing concrete markers for biological follow-up and validation.


Subject(s)
Single-Cell Analysis/methods , Stem Cells/cytology , Transcriptome/genetics , Adult , Algorithms , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Lineage , Entropy , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Intestines/cytology , Mice, Inbred C57BL , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Pancreatic Ducts/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Reproducibility of Results
7.
Cell Stem Cell ; 18(2): 203-13, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26831517

ABSTRACT

Intestinal crypts display robust regeneration upon injury. The relatively rare secretory precursors can replace lost stem cells, but it is unknown if the abundant enterocyte progenitors that express the Alkaline phosphate intestinal (Alpi) gene also have this capacity. We created an Alpi-IRES-CreERT2 (Alpi(CreER)) knockin allele for lineage tracing. Marked clones consist entirely of enterocytes and are all lost from villus tips within days. Genetic fate-mapping of Alpi(+) cells before or during targeted ablation of Lgr5-expressing stem cells generated numerous long-lived crypt-villus "ribbons," indicative of dedifferentiation of enterocyte precursors into Lgr5(+) stems. By single-cell analysis of dedifferentiating enterocytes, we observed the generation of Paneth-like cells and proliferative stem cells. We conclude that the highly proliferative, short-lived enterocyte precursors serve as a large reservoir of potential stem cells during crypt regeneration.


Subject(s)
Cell Lineage , Enterocytes/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Alkaline Phosphatase/metabolism , Animals , Biomarkers/metabolism , Cell Dedifferentiation , Cell Line , Cell Proliferation , Enterocytes/pathology , Integrases/metabolism , Intestinal Neoplasms/pathology , Mice , Mutation/genetics , Organoids , Paneth Cells/metabolism , Paneth Cells/pathology , Regeneration/genetics , Single-Cell Analysis , beta-Galactosidase/metabolism
8.
Nature ; 525(7568): 251-5, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26287467

ABSTRACT

Understanding the development and function of an organ requires the characterization of all of its cell types. Traditional methods for visualizing and isolating subpopulations of cells are based on messenger RNA or protein expression of only a few known marker genes. The unequivocal identification of a specific marker gene, however, poses a major challenge, particularly if this cell type is rare. Identifying rare cell types, such as stem cells, short-lived progenitors, cancer stem cells, or circulating tumour cells, is crucial to acquire a better understanding of normal or diseased tissue biology. To address this challenge we first sequenced the transcriptome of hundreds of randomly selected cells from mouse intestinal organoids, cultured self-organizing epithelial structures that contain all cell lineages of the mammalian intestine. Organoid buds, like intestinal crypts, harbour stem cells that continuously differentiate into a variety of cell types, occurring at widely different abundances. Since available computational methods can only resolve more abundant cell types, we developed RaceID, an algorithm for rare cell type identification in complex populations of single cells. We demonstrate that this algorithm can resolve cell types represented by only a single cell in a population of randomly sampled organoid cells. We use this algorithm to identify Reg4 as a novel marker for enteroendocrine cells, a rare population of hormone-producing intestinal cells. Next, we use Reg4 expression to enrich for these rare cells and investigate the heterogeneity within this population. RaceID confirmed the existence of known enteroendocrine lineages, and moreover discovered novel subtypes, which we subsequently validated in vivo. Having validated RaceID we then applied the algorithm to ex vivo-isolated Lgr5-positive stem cells and their direct progeny. We find that Lgr5-positive cells represent a homogenous abundant population of stem cells mixed with a rare population of Lgr5-positive secretory cells. We envision broad applicability of our method for discovering rare cell types and the corresponding marker genes in healthy and diseased organs.


Subject(s)
Cell Separation/methods , Intestine, Small/cytology , RNA, Messenger/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Algorithms , Animals , Biomarkers/analysis , Cell Differentiation/genetics , Cell Lineage , In Situ Hybridization, Fluorescence , Mice , Neoplasm Proteins/genetics , Organoids/cytology , Pancreatitis-Associated Proteins , Paneth Cells/cytology , Paneth Cells/metabolism , Receptors, G-Protein-Coupled/genetics , Reproducibility of Results , Stem Cells/cytology , Stem Cells/metabolism , Transcriptome/genetics
9.
Nat Methods ; 11(5): 549-551, 2014 May.
Article in English | MEDLINE | ID: mdl-24681693

ABSTRACT

We have developed a quantitative technique for sorting cells on the basis of endogenous RNA abundance, with a molecular resolution of 10-20 transcripts. We demonstrate efficient and unbiased RNA extraction from transcriptionally sorted cells and report a high-fidelity transcriptome measurement of mouse induced pluripotent stem cells (iPSCs) isolated from a heterogeneous reprogramming culture. This method is broadly applicable to profiling transcriptionally distinct cellular states without requiring antibodies or transgenic fluorescent proteins.


Subject(s)
Cell Culture Techniques , Gene Expression Profiling , Induced Pluripotent Stem Cells/cytology , RNA/metabolism , Transcription, Genetic , Alleles , Animals , Cellular Reprogramming , Doxycycline/chemistry , Embryonic Stem Cells/cytology , Fibroblasts/metabolism , Flow Cytometry , Genome-Wide Association Study , Green Fluorescent Proteins/metabolism , In Situ Hybridization, Fluorescence , Mice , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA, Messenger/metabolism , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...