Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










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.
Proc Natl Acad Sci U S A ; 115(52): E12245-E12254, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30530645

ABSTRACT

The significance of cardiac stem cell (CSC) populations for cardiac regeneration remains disputed. Here, we apply the most direct definition of stem cell function (the ability to replace lost tissue through cell division) to interrogate the existence of CSCs. By single-cell mRNA sequencing and genetic lineage tracing using two Ki67 knockin mouse models, we map all proliferating cells and their progeny in homoeostatic and regenerating murine hearts. Cycling cardiomyocytes were only robustly observed in the early postnatal growth phase, while cycling cells in homoeostatic and damaged adult myocardium represented various noncardiomyocyte cell types. Proliferative postdamage fibroblasts expressing follistatin-like protein 1 (FSTL1) closely resemble neonatal cardiac fibroblasts and form the fibrotic scar. Genetic deletion of Fstl1 in cardiac fibroblasts results in postdamage cardiac rupture. We find no evidence for the existence of a quiescent CSC population, for transdifferentiation of other cell types toward cardiomyocytes, or for proliferation of significant numbers of cardiomyocytes in response to cardiac injury.


Subject(s)
Cell Proliferation , Heart Injuries/physiopathology , Animals , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Heart Injuries/genetics , Heart Injuries/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pregnancy , Stem Cells/cytology , Stem Cells/metabolism
3.
Cell ; 175(6): 1591-1606.e19, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30500538

ABSTRACT

The mammalian liver possesses a remarkable regenerative ability. Two modes of damage response have been described: (1) The "oval cell" response emanates from the biliary tree when all hepatocytes are affected by chronic liver disease. (2) A massive, proliferative response of mature hepatocytes occurs upon acute liver damage such as partial hepatectomy (PHx). While the oval cell response has been captured in vitro by growing organoids from cholangiocytes, the hepatocyte proliferative response has not been recapitulated in culture. Here, we describe the establishment of a long-term 3D organoid culture system for mouse and human primary hepatocytes. Organoids can be established from single hepatocytes and grown for multiple months, while retaining key morphological, functional and gene expression features. Transcriptional profiles of the organoids resemble those of proliferating hepatocytes after PHx. Human hepatocyte organoids proliferate extensively after engraftment into mice and thus recapitulate the proliferative damage-response of hepatocytes.


Subject(s)
Cell Proliferation , Hepatocytes/metabolism , Organoids/metabolism , Animals , Cell Culture Techniques , Cells, Cultured , Hepatocytes/cytology , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Organoids/cytology , Stem Cells/cytology , Stem Cells/metabolism , Time Factors
4.
Nat Cell Biol ; 20(8): 909-916, 2018 08.
Article in English | MEDLINE | ID: mdl-30038251

ABSTRACT

Enteroendocrine cells (EECs) control a wide range of physiological processes linked to metabolism1. We show that EEC hormones are differentially expressed between crypts (for example, Glp1) and villi (for example, secretin). As demonstrated by single-cell mRNA sequencing using murine Lgr5+ cell-derived organoids, BMP4 signals alter the hormone expression profiles of individual EECs to resemble those found in the villus. Accordingly, BMP4 induces hormone switching of EECs migrating up the crypt-villus axis in vivo. Our findings imply that EEC lineages in the small intestine exhibit a more flexible hormone repertoire than previously proposed. We also describe a protocol to generate human EECs in organoids and demonstrate a similar regulation of hormone expression by BMP signalling. These findings establish alternative strategies to target EECs with therapeutically relevant hormone production through BMP modulation.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Lineage , Cell Movement/drug effects , Enteroendocrine Cells/metabolism , Gastrointestinal Hormones/metabolism , Intestine, Small/metabolism , Signal Transduction/drug effects , Animals , Humans , Intestine, Small/cytology , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Tissue Culture Techniques
5.
Proc Natl Acad Sci U S A ; 113(42): 11859-11864, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27708166

ABSTRACT

Current mouse models for colorectal cancer often differ significantly from human colon cancer, being largely restricted to the small intestine. Here, we aim to develop a colon-specific inducible mouse model that can faithfully recapitulate human colon cancer initiation and progression. Carbonic anhydrase I (Car1) is a gene expressed uniquely in colonic epithelial cells. We generated a colon-specific inducible Car1CreER knock-in (KI) mouse with broad Cre activity in epithelial cells of the proximal colon and cecum. Deletion of the tumor suppressor gene Apc using the Car1CreER KI caused tumor formation in the cecum but did not yield adenomas in the proximal colon. Mutation of both Apc and Kras yielded microadenomas in both the cecum and the proximal colon, which progressed to macroadenomas with significant morbidity. Aggressive carcinomas with some invasion into lymph nodes developed upon combined induction of oncogenic mutations of Apc, Kras, p53, and Smad4 Importantly, no adenomas were observed in the small intestine. Additionally, we observed tumors from differentiated Car1-expressing cells with Apc/Kras mutations, suggesting that a top-down model of intestinal tumorigenesis can occur with multiple mutations. Our results establish the Car1CreER KI as a valuable mouse model to study colon-specific tumorigenesis and metastasis as well as cancer-cell-of-origin questions.


Subject(s)
Colonic Neoplasms/etiology , Gene Expression Regulation , Integrases/genetics , Mice, Transgenic , Adenoma/etiology , Adenoma/metabolism , Adenoma/pathology , Animals , Biomarkers, Tumor , Carbonic Anhydrase I/genetics , Carbonic Anhydrase I/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Disease Progression , Enzyme Activation , Gene Knock-In Techniques , Gene Targeting , Genes, APC , Genes, ras , Genetic Loci , Humans , Immunohistochemistry , Integrases/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Mutation , Organ Specificity/genetics , Research
6.
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
7.
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
8.
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
9.
Proc Natl Acad Sci U S A ; 112(24): 7548-50, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26023187

ABSTRACT

Rnf43 (RING finger protein 43) and Znrf3 (zinc/RING finger protein 3) (RZ) are two closely related transmembrane E3 ligases, encoded by Wnt target genes, that remove surface Wnt (wingless-int) receptors. The two genes are mutated in various human cancers. Such tumors are predicted to be hypersensitive to, yet still depend on, secreted Wnts. We previously showed that mutation of RZ in the intestine yields rapidly growing adenomas containing LGR5(+) (leucine-rich repeat-containing G-protein coupled receptor 5) stem cells and Wnt3-producing Paneth cells. We now show that removal of Paneth cells by Math1 mutation inhibits RZ(-/-) tumor formation. Similarly, deletion of Wnt3 inhibits tumorigenesis. Treatment of mice carrying RZ(-/-) intestinal neoplasia with a small molecule Wnt secretion inhibitor (porcupine inhibitor C59) strongly inhibited growth, whereas adjacent normal crypts remained intact. These results establish that paracrine Wnt secretion is an essential driver of RZ(-/-) tumor growth and imply that a therapeutic window exists for the use of porcupine inhibitors for RZ-mutant cancers.


Subject(s)
Benzeneacetamides/pharmacology , Intestinal Neoplasms/drug therapy , Membrane Proteins/antagonists & inhibitors , Pyridines/pharmacology , Ubiquitin-Protein Ligases/genetics , Wnt Signaling Pathway/drug effects , Acyltransferases , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Humans , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Paracrine Communication/drug effects , Ubiquitin-Protein Ligases/deficiency , Wnt3 Protein/deficiency , Wnt3 Protein/genetics , Zinc Fingers/genetics
10.
Nat Commun ; 5: 5103, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25267258

ABSTRACT

The transcription factor NF-κB is indispensable for intestinal immune homeostasis, but contributes to chronic inflammation and inflammatory bowel disease (IBD). A20, an inhibitor of both NF-κB and apoptotic signalling, was identified as a susceptibility gene for multiple inflammatory diseases, including IBD. Despite absence of spontaneous intestinal inflammation in intestinal epithelial cell (IEC) specific A20 knockout mice, we found additional myeloid-specific A20 deletion to synergistically drive intestinal pathology through cell-specific mechanisms. A20 ensures intestinal barrier stability by preventing cytokine-induced IEC apoptosis, while A20 prevents excessive cytokine production in myeloid cells. Combining IEC and myeloid A20 deletion induces ileitis and severe colitis, characterized by IEC apoptosis, Paneth and goblet cell loss, epithelial hyperproliferation and intestinal microbiota dysbiosis. Continuous epithelial cell death and regeneration in an inflammatory environment sensitizes cells for neoplastic transformation and the development of colorectal tumours in aged mice.


Subject(s)
Cysteine Endopeptidases/metabolism , Epithelial Cells/enzymology , Intestines/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Apoptosis , Colitis/enzymology , Colitis/genetics , Colitis/pathology , Colitis/physiopathology , Cysteine Endopeptidases/genetics , Epithelial Cells/cytology , Epithelial Cells/pathology , Female , Goblet Cells/cytology , Goblet Cells/enzymology , Goblet Cells/pathology , Homeostasis , Humans , Intestines/pathology , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Paneth Cells/cytology , Paneth Cells/enzymology , Paneth Cells/pathology , Species Specificity , Tumor Necrosis Factor alpha-Induced Protein 3
11.
Cell ; 151(7): 1595-607, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23260145

ABSTRACT

Most studies on TCF7L2 SNP variants in the pathogenesis of type 2 diabetes (T2D) focus on a role of the encoded transcription factor TCF4 in ß cells. Here, a mouse genetics approach shows that removal of TCF4 from ß cells does not affect their function, whereas manipulating TCF4 levels in the liver has major effects on metabolism. In Tcf7l2(-/-) mice, the immediate postnatal surge in liver metabolism does not occur. Consequently, pups die due to hypoglycemia. By combining chromatin immunoprecipitation with gene expression profiling, we identify a TCF4-controlled metabolic gene program that is acutely activated in the postnatal liver. In concordance, adult liver-specific Tcf7l2 knockout mice show reduced hepatic glucose production during fasting and display improved glucose homeostasis when maintained on high-fat diet. Furthermore, liver-specific TCF4 overexpression increases hepatic glucose production. These observations imply that TCF4 directly activates metabolic genes and that inhibition of Wnt signaling may be beneficial in metabolic disease.


Subject(s)
Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Glucose/metabolism , Liver/metabolism , Metabolic Networks and Pathways , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Animals, Newborn , Diet, High-Fat , Fasting/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Transcription Factor 7-Like 2 Protein/genetics , Transcriptional Activation
12.
Nat Cell Biol ; 14(10): 1099-1104, 2012 10.
Article in English | MEDLINE | ID: mdl-23000963

ABSTRACT

Lgr5+ intestinal stem cells generate enterocytes and secretory cells. Secretory lineage commitment requires Notch silencing. The Notch ligand Dll1 is expressed by a subset of immediate stem cell daughters. Lineage tracing in Dll1(GFP-ires-CreERT2) knock-in mice reveals that single Dll1(high) cells generate small, short-lived clones containing all four secretory cell types. Lineage specification thus occurs in immediate stem cell daughters through Notch lateral inhibition. Cultured Dll1(high) cells form long-lived organoids (mini-guts) on brief Wnt3A exposure. When Dll1(high) cells are genetically marked before tissue damage, stem cell tracing events occur. Thus, secretory progenitors exhibit plasticity by regaining stemness on damage.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Intestinal Mucosa/metabolism , Stem Cells/metabolism , Animals , Calcium-Binding Proteins , Cell Lineage , Cells, Cultured , Gene Knock-In Techniques , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/genetics , Mice , Organoids/metabolism , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , Wnt3A Protein/pharmacology
13.
Science ; 337(6095): 730-5, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22855427

ABSTRACT

The concept that tumors are maintained by dedicated stem cells, the so-called cancer stem cell hypothesis, has attracted great interest but remains controversial. Studying mouse models, we provide direct, functional evidence for the presence of stem cell activity within primary intestinal adenomas, a precursor to intestinal cancer. By "lineage retracing" using the multicolor Cre-reporter R26R-Confetti, we demonstrate that the crypt stem cell marker Lgr5 (leucine-rich repeat-containing heterotrimeric guanine nucleotide-binding protein-coupled receptor 5) also marks a subpopulation of adenoma cells that fuel the growth of established intestinal adenomas. These Lgr5(+) cells, which represent about 5 to 10% of the cells in the adenomas, generate additional Lgr5(+) cells as well as all other adenoma cell types. The Lgr5(+) cells are intermingled with Paneth cells near the adenoma base, a pattern reminiscent of the architecture of the normal crypt niche.


Subject(s)
Adenoma/pathology , Intestinal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/physiology , Receptors, G-Protein-Coupled/analysis , Adenoma/genetics , Adenoma/metabolism , Animals , Biomarkers/analysis , Cell Lineage , Cell Transformation, Neoplastic , Gene Expression Profiling , Gene Knock-In Techniques , Genes, Reporter , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Neoplasms/genetics , Mice , Multipotent Stem Cells/pathology , Multipotent Stem Cells/physiology , Paneth Cells/pathology , Stem Cell Niche , Tamoxifen/pharmacology , Tumor Stem Cell Assay
14.
Mol Cell Biol ; 32(10): 1918-27, 2012 May.
Article in English | MEDLINE | ID: mdl-22393260

ABSTRACT

Throughout life, intestinal Lgr5+ stem cells give rise to proliferating transient amplifying cells in crypts, which subsequently differentiate into one of the five main cell types and migrate along the crypt-villus axis. These dynamic processes are coordinated by a relatively small number of evolutionarily conserved signaling pathways, which includes the Wnt signaling pathway. The DNA-binding proteins of the T-cell factor family, Tcf1/Tcf7, Lef, Tcf3/Tcf7l1, and Tcf4/Tcf7l2, constitute the downstream effectors of the Wnt signaling pathway. While Tcf4 is the major member active during embryogenesis, the role of these Wnt effectors in the homeostasis of the adult mouse intestinal epithelium is unresolved. Using Tcf1-/-, Tcf3(flox), and novel Tcf4(flox) mice, we demonstrate an essential role for Tcf4 during homeostasis of the adult mouse intestine.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Intestines/cytology , Stem Cells/metabolism , Wnt Signaling Pathway , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Differentiation , Cell Proliferation , Gene Expression Regulation, Developmental , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Stem Cells/cytology , Transcription Factor 4
15.
PLoS One ; 6(8): e22616, 2011.
Article in English | MEDLINE | ID: mdl-21826198

ABSTRACT

Follistatin-like 1 (Fstl1) is a secreted protein of the BMP inhibitor class. During development, expression of Fstl1 is already found in cleavage stage embryos and becomes gradually restricted to mesenchymal elements of most organs during subsequent development. Knock down experiments in chicken and zebrafish demonstrated a role as a BMP antagonist in early development. To investigate the role of Fstl1 during mouse development, a conditional Fstl1 KO allele as well as a Fstl1-GFP reporter mouse were created. KO mice die at birth from respiratory distress and show multiple defects in lung development. Also, skeletal development is affected. Endochondral bone development, limb patterning as well as patterning of the axial skeleton are perturbed in the absence of Fstl1. Taken together, these observations show that Fstl1 is a crucial regulator in BMP signalling during mouse development.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Follistatin-Related Proteins/metabolism , Lung/embryology , Lung/metabolism , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Organogenesis/physiology , Animals , Female , Follistatin-Related Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organogenesis/genetics
16.
EMBO J ; 30(6): 1104-9, 2011 Mar 16.
Article in English | MEDLINE | ID: mdl-21297579

ABSTRACT

Somatic cells have been proposed to be limited in the number of cell divisions they can undergo. This is thought to be a mechanism by which stem cells retain their integrity preventing disease. However, we have recently discovered intestinal crypt stem cells that persist for the lifetime of a mouse, yet divide every day. We now demonstrate biochemically that primary isolated Lgr5+ve stem cells contain significant telomerase activity. Telomerase activity rapidly decreases in the undifferentiated progeny of these stem cells and is entirely lost in differentiated villus cells. Conversely, asymmetric segregation of chromosomes has been proposed as a mechanism for stem cells to protect their genomes against damage. We determined the average cell cycle length of Lgr5+ve stem cells at 21.5 h and find that Lgr5+ve intestinal stem cells randomly segregate newly synthesized DNA strands, opposing the 'immortal strand' hypothesis.


Subject(s)
Chromosome Segregation , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Stem Cell Niche , Stem Cells/enzymology , Stem Cells/physiology , Telomerase/metabolism , Animals , Cell Cycle , Cell Differentiation , Epithelial Cells/enzymology , Epithelial Cells/physiology , Mice , Time Factors
17.
Nature ; 469(7330): 415-8, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21113151

ABSTRACT

Homeostasis of self-renewing small intestinal crypts results from neutral competition between Lgr5 stem cells, which are small cycling cells located at crypt bottoms. Lgr5 stem cells are interspersed between terminally differentiated Paneth cells that are known to produce bactericidal products such as lysozyme and cryptdins/defensins. Single Lgr5-expressing stem cells can be cultured to form long-lived, self-organizing crypt-villus organoids in the absence of non-epithelial niche cells. Here we find a close physical association of Lgr5 stem cells with Paneth cells in mice, both in vivo and in vitro. CD24(+) Paneth cells express EGF, TGF-α, Wnt3 and the Notch ligand Dll4, all essential signals for stem-cell maintenance in culture. Co-culturing of sorted stem cells with Paneth cells markedly improves organoid formation. This Paneth cell requirement can be substituted by a pulse of exogenous Wnt. Genetic removal of Paneth cells in vivo results in the concomitant loss of Lgr5 stem cells. In colon crypts, CD24(+) cells residing between Lgr5 stem cells may represent the Paneth cell equivalents. We conclude that Lgr5 stem cells compete for essential niche signals provided by a specialized daughter cell, the Paneth cell.


Subject(s)
Intestines/cytology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Paneth Cells/cytology , Receptors, G-Protein-Coupled/metabolism , Stem Cell Niche/cytology , Animals , CD24 Antigen/metabolism , Cell Count , Cell Proliferation , Coculture Techniques , Humans , Mice , Paneth Cells/metabolism , Stem Cell Niche/metabolism , Wnt Proteins/metabolism , Wnt3 Protein
18.
Cell ; 143(1): 134-44, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20887898

ABSTRACT

Intestinal stem cells, characterized by high Lgr5 expression, reside between Paneth cells at the small intestinal crypt base and divide every day. We have carried out fate mapping of individual stem cells by generating a multicolor Cre-reporter. As a population, Lgr5(hi) stem cells persist life-long, yet crypts drift toward clonality within a period of 1-6 months. We have collected short- and long-term clonal tracing data of individual Lgr5(hi) cells. These reveal that most Lgr5(hi) cell divisions occur symmetrically and do not support a model in which two daughter cells resulting from an Lgr5(hi) cell division adopt divergent fates (i.e., one Lgr5(hi) cell and one transit-amplifying [TA] cell per division). The cellular dynamics are consistent with a model in which the resident stem cells double their numbers each day and stochastically adopt stem or TA fates. Quantitative analysis shows that stem cell turnover follows a pattern of neutral drift dynamics.


Subject(s)
Cell Lineage , Intestine, Small/cytology , Stem Cells/cytology , Animals , Clone Cells , Mice , Models, Biological , Receptors, G-Protein-Coupled/metabolism
19.
Science ; 327(5971): 1385-9, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20223988

ABSTRACT

Mammalian epidermis consists of three self-renewing compartments: the hair follicle, the sebaceous gland, and the interfollicular epidermis. We generated knock-in alleles of murine Lgr6, a close relative of the Lgr5 stem cell gene. Lgr6 was expressed in the earliest embryonic hair placodes. In adult hair follicles, Lgr6+ cells resided in a previously uncharacterized region directly above the follicle bulge. They expressed none of the known bulge stem cell markers. Prenatal Lgr6+ cells established the hair follicle, sebaceous gland, and interfollicular epidermis. Postnatally, Lgr6+ cells generated sebaceous gland and interfollicular epidermis, whereas contribution to hair lineages gradually diminished with age. Adult Lgr6+ cells executed long-term wound repair, including the formation of new hair follicles. We conclude that Lgr6 marks the most primitive epidermal stem cell.


Subject(s)
Cell Lineage , Hair Follicle/cytology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Skin/cytology , Stem Cells/cytology , Animals , Epidermal Cells , Gene Expression Profiling , Gene Knock-In Techniques , Hair/cytology , Hair/embryology , Hair/growth & development , Hair Follicle/embryology , Hair Follicle/growth & development , Mice , Mice, Nude , Sebaceous Glands/cytology , Signal Transduction , Stem Cell Transplantation , Stem Cells/metabolism , Wound Healing
20.
Dis Model Mech ; 3(1-2): 104-10, 2010.
Article in English | MEDLINE | ID: mdl-20075383

ABSTRACT

Barrett's esophagus (BE) affects approximately 2% of the Western population and progresses to esophageal adenocarcinoma (EAC) in 0.5% of these patients each year. In BE, the stratified epithelium is replaced by an intestinal-type epithelium owing to chronic gastroduodenal reflux. Since self-renewal of intestinal crypts is driven by Notch signaling, we investigated whether this pathway was active in the proliferative crypts of BE. Immunohistochemistry confirmed the presence of an intact and activated Notch signaling pathway in metaplastic BE epithelium, but not in the normal human esophagus. Similar observations were made in two well-known human Barrett's-derived EAC cell lines, OE33 and SKGT-5. We then sought to investigate the effects of Notch inhibition by systemic treatment with a gamma-secretase inhibitor in a well-validated rodent model for BE. As we have shown previously in normal intestinal epithelium, Notch inhibition converted the proliferative Barrett's epithelial cells into terminally differentiated goblet cells, whereas the squamous epithelium remained intact. These data imply that local application of gamma-secretase inhibitors may present a simple therapeutic strategy for this increasingly common pre-malignant condition.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Barrett Esophagus/enzymology , Barrett Esophagus/pathology , Epithelium/pathology , Goblet Cells/pathology , Mitosis , Animals , Biopsy , Cell Line, Tumor , Cell Proliferation/drug effects , Colon/drug effects , Colon/metabolism , Colon/pathology , Dibenzazepines/pharmacology , Disease Models, Animal , Epithelium/drug effects , Epithelium/metabolism , Gene Expression Regulation/drug effects , Goblet Cells/drug effects , Goblet Cells/metabolism , Humans , Metaplasia , Mitosis/drug effects , Rats , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...