Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters










Publication year range
1.
Cell Stem Cell ; 31(2): 227-243.e12, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38215738

ABSTRACT

The conjunctival epithelium covering the eye contains two main cell types: mucus-producing goblet cells and water-secreting keratinocytes, which present mucins on their apical surface. Here, we describe long-term expanding organoids and air-liquid interface representing mouse and human conjunctiva. A single-cell RNA expression atlas of primary and cultured human conjunctiva reveals that keratinocytes express multiple antimicrobial peptides and identifies conjunctival tuft cells. IL-4/-13 exposure increases goblet and tuft cell differentiation and drastically modifies the conjunctiva secretome. Human NGFR+ basal cells are identified as bipotent conjunctiva stem cells. Conjunctival cultures can be infected by herpes simplex virus 1 (HSV1), human adenovirus 8 (hAdV8), and SARS-CoV-2. HSV1 infection was reversed by acyclovir addition, whereas hAdV8 infection, which lacks an approved drug therapy, was inhibited by cidofovir. We document transcriptional programs induced by HSV1 and hAdV8. Finally, conjunctival organoids can be transplanted. Together, human conjunctiva organoid cultures enable the study of conjunctival (patho)-physiology.


Subject(s)
Conjunctiva , Goblet Cells , Humans , Mice , Animals , Conjunctiva/metabolism , Goblet Cells/metabolism , Epithelium , Interleukin-13 , Homeostasis , Organoids
2.
Br J Cancer ; 130(3): 369-379, 2024 02.
Article in English | MEDLINE | ID: mdl-38102228

ABSTRACT

BACKGROUND: Bladder cancer is one of the most common cancer types worldwide. Generally, research relies on invasive sampling strategies. METHODS: Here, we generate bladder cancer organoids directly from urine (urinoids). In this project, we establish 12 urinoid lines from 22 patients with non-muscle and muscle-invasive bladder tumours, with an efficiency of 55%. RESULTS: The histopathological features of the urinoids accurately resemble those of the original bladder tumours. Genetically, there is a high concordance of single nucleotide polymorphisms (92.56%) and insertions & deletions (91.54%) between urinoids and original tumours from patient 4. Furthermore, these urinoids show sensitivity to bladder cancer drugs, similar to their tissue-derived organoid counterparts. Genetic analysis of longitudinally generated tumoroids and urinoids from one patient receiving systemic immunotherapy, identify alterations that may guide the choice for second-line therapy. Successful treatment adaptation was subsequently demonstrated in the urinoid setting. CONCLUSION: Therefore, urinoids can advance precision medicine in bladder cancer as a non-invasive platform for tumour pathogenesis, longitudinal drug-response monitoring, and therapy adaptation.


Subject(s)
Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder/pathology , Immunotherapy , Precision Medicine , Organoids/pathology
3.
Science ; 382(6669): 451-458, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37883554

ABSTRACT

Enteroendocrine cells (EECs) are hormone-producing cells residing in the epithelium of stomach, small intestine (SI), and colon. EECs regulate aspects of metabolic activity, including insulin levels, satiety, gastrointestinal secretion, and motility. The generation of different EEC lineages is not completely understood. In this work, we report a CRISPR knockout screen of the entire repertoire of transcription factors (TFs) in adult human SI organoids to identify dominant TFs controlling EEC differentiation. We discovered ZNF800 as a master repressor for endocrine lineage commitment, which particularly restricts enterochromaffin cell differentiation by directly controlling an endocrine TF network centered on PAX4. Thus, organoid models allow unbiased functional CRISPR screens for genes that program cell fate.


Subject(s)
CRISPR-Cas Systems , Cell Lineage , Enteroendocrine Cells , Gene Expression Regulation , Repressor Proteins , Zinc Fingers , Humans , Cell Differentiation/genetics , Enteroendocrine Cells/cytology , Enteroendocrine Cells/metabolism , Organoids , Adult , Cell Lineage/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
4.
Nat Commun ; 14(1): 4998, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37591832

ABSTRACT

Optimization of CRISPR/Cas9-mediated genome engineering has resulted in base editors that hold promise for mutation repair and disease modeling. Here, we demonstrate the application of base editors for the generation of complex tumor models in human ASC-derived organoids. First we show efficacy of cytosine and adenine base editors in modeling CTNNB1 hot-spot mutations in hepatocyte organoids. Next, we use C > T base editors to insert nonsense mutations in PTEN in endometrial organoids and demonstrate tumorigenicity even in the heterozygous state. Moreover, drug sensitivity assays on organoids harboring either PTEN or PTEN and PIK3CA mutations reveal the mechanism underlying the initial stages of endometrial tumorigenesis. To further increase the scope of base editing we combine SpCas9 and SaCas9 for simultaneous C > T and A > G editing at individual target sites. Finally, we show that base editor multiplexing allow modeling of colorectal tumorigenesis in a single step by simultaneously transfecting sgRNAs targeting five cancer genes.


Subject(s)
Adult Stem Cells , RNA, Guide, CRISPR-Cas Systems , Adult , Humans , Oncogenes , Carcinogenesis/genetics , Cell Transformation, Neoplastic , Organoids
6.
J Cell Biol ; 219(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32573693

ABSTRACT

In vitro experiments have shown that GRASP65 (GORASP1) and GRASP55 (GORASP2) proteins function in stacking Golgi cisternae. However, in vivo depletion of GORASPs in metazoans has given equivocal results. We have generated a mouse lacking both GORASPs and find that Golgi cisternae remained stacked. However, the stacks are disconnected laterally from each other, and the cisternal cross-sectional diameters are significantly reduced compared with their normal counterparts. These data support earlier findings on the role of GORASPs in linking stacks, and we suggest that unlinking of stacks likely affects dynamic control of COPI budding and vesicle fusion at the rims. The net result is that cisternal cores remain stacked, but cisternal diameter is reduced by rim consumption.


Subject(s)
Golgi Apparatus/metabolism , Golgi Matrix Proteins/metabolism , Animals , COP-Coated Vesicles/metabolism , Female , Intracellular Membranes/metabolism , Membrane Fusion/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL
7.
Nat Commun ; 11(1): 2660, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32461556

ABSTRACT

High-grade serous ovarian cancer (HG-SOC)-often referred to as a "silent killer"-is the most lethal gynecological malignancy. The fallopian tube (murine oviduct) and ovarian surface epithelium (OSE) are considered the main candidate tissues of origin of this cancer. However, the relative contribution of each tissue to HG-SOC is not yet clear. Here, we establish organoid-based tumor progression models of HG-SOC from murine oviductal and OSE tissues. We use CRISPR-Cas9 genome editing to introduce mutations into genes commonly found mutated in HG-SOC, such as Trp53, Brca1, Nf1 and Pten. Our results support the dual origin hypothesis of HG-SOC, as we demonstrate that both epithelia can give rise to ovarian tumors with high-grade pathology. However, the mutated oviductal organoids expand much faster in vitro and more readily form malignant tumors upon transplantation. Furthermore, in vitro drug testing reveals distinct lineage-dependent sensitivities to the common drugs used to treat HG-SOC in patients.


Subject(s)
CRISPR-Cas Systems/genetics , Organoids , Ovarian Neoplasms/etiology , Animals , Antineoplastic Agents/pharmacology , BRCA1 Protein/genetics , CRISPR-Associated Protein 9 , Epithelium/pathology , Fallopian Tubes/pathology , Female , Gene Editing/methods , Mice , Mutation , Neurofibromatosis 1/genetics , Organ Culture Techniques/methods , Organoids/drug effects , Organoids/physiopathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovary/pathology , PTEN Phosphohydrolase/genetics , Tumor Suppressor Protein p53/genetics
8.
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.

9.
Cancer Discov ; 9(7): 852-871, 2019 07.
Article in English | MEDLINE | ID: mdl-31053628

ABSTRACT

Previous studies have described that tumor organoids can capture the diversity of defined human carcinoma types. Here, we describe conditions for long-term culture of human mucosal organoids. Using this protocol, a panel of 31 head and neck squamous cell carcinoma (HNSCC)-derived organoid lines was established. This panel recapitulates genetic and molecular characteristics previously described for HNSCC. Organoids retain their tumorigenic potential upon xenotransplantation. We observe differential responses to a panel of drugs including cisplatin, carboplatin, cetuximab, and radiotherapy in vitro. Additionally, drug screens reveal selective sensitivity to targeted drugs that are not normally used in the treatment of patients with HNSCC. These observations may inspire a personalized approach to the management of HNSCC and expand the repertoire of HNSCC drugs. SIGNIFICANCE: This work describes the culture of organoids derived from HNSCC and corresponding normal epithelium. These tumoroids recapitulate the disease genetically, histologically, and functionally. In vitro drug screening of tumoroids reveals responses to therapies both currently used in the treatment of HNSCC and those not (yet) used in clinical practice.See related commentary by Hill and D'Andrea, p. 828.This article is highlighted in the In This Issue feature, p. 813.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/therapy , Mouth Mucosa/pathology , Organoids/pathology , Precision Medicine/methods , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/therapy , Animals , Carboplatin/administration & dosage , Cetuximab/administration & dosage , Chemoradiotherapy , Cisplatin/administration & dosage , Drug Screening Assays, Antitumor/methods , Head and Neck Neoplasms/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mouth Mucosa/drug effects , Mouth Mucosa/radiation effects , Organoids/drug effects , Organoids/radiation effects , Squamous Cell Carcinoma of Head and Neck/metabolism , Xenograft Model Antitumor Assays
10.
Cell Stem Cell ; 24(6): 927-943.e6, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31130514

ABSTRACT

The deubiquitinating enzyme BAP1 is a tumor suppressor, among others involved in cholangiocarcinoma. BAP1 has many proposed molecular targets, while its Drosophila homolog is known to deubiquitinate histone H2AK119. We introduce BAP1 loss-of-function by CRISPR/Cas9 in normal human cholangiocyte organoids. We find that BAP1 controls the expression of junctional and cytoskeleton components by regulating chromatin accessibility. Consequently, we observe loss of multiple epithelial characteristics while motility increases. Importantly, restoring the catalytic activity of BAP1 in the nucleus rescues these cellular and molecular changes. We engineer human liver organoids to combine four common cholangiocarcinoma mutations (TP53, PTEN, SMAD4, and NF1). In this genetic background, BAP1 loss results in acquisition of malignant features upon xenotransplantation. Thus, control of epithelial identity through the regulation of chromatin accessibility appears to be a key aspect of BAP1's tumor suppressor function. Organoid technology combined with CRISPR/Cas9 provides an experimental platform for mechanistic studies of cancer gene function in a human context.


Subject(s)
Cholangiocarcinoma/genetics , Chromatin/metabolism , Epithelial Cells/physiology , Liver Neoplasms/genetics , Liver/physiology , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Bioengineering , Carcinogenesis , Cells, Cultured , Chromatin/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Cytoskeleton/metabolism , Female , Humans , Loss of Function Mutation/genetics , Mice , Mice, SCID , Organoids , Transplantation, Heterologous , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics
11.
Cell ; 176(5): 1158-1173.e16, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30712869

ABSTRACT

Homeostatic regulation of the intestinal enteroendocrine lineage hierarchy is a poorly understood process. We resolved transcriptional changes during enteroendocrine differentiation in real time at single-cell level using a novel knockin allele of Neurog3, the master regulator gene briefly expressed at the onset of enteroendocrine specification. A bi-fluorescent reporter, Neurog3Chrono, measures time from the onset of enteroendocrine differentiation and enables precise positioning of single-cell transcriptomes along an absolute time axis. This approach yielded a definitive description of the enteroendocrine hierarchy and its sub-lineages, uncovered differential kinetics between sub-lineages, and revealed time-dependent hormonal plasticity in enterochromaffin and L cells. The time-resolved map of transcriptional changes predicted multiple novel molecular regulators. Nine of these were validated by conditional knockout in mice or CRISPR modification in intestinal organoids. Six novel candidate regulators (Sox4, Rfx6, Tox3, Myt1, Runx1t1, and Zcchc12) yielded specific enteroendocrine phenotypes. Our time-resolved single-cell transcriptional map presents a rich resource to unravel enteroendocrine differentiation.


Subject(s)
Cell Lineage/genetics , Enteroendocrine Cells/metabolism , Gene Expression Profiling/methods , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Lineage/physiology , Enteroendocrine Cells/physiology , Fluorescent Dyes , Homeodomain Proteins/genetics , Intestinal Mucosa/cytology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Optical Imaging/methods , Organoids , Phenotype , Single-Cell Analysis/methods , Stem Cells , Transcription Factors/genetics , Transcriptome/genetics
12.
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
13.
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
14.
Science ; 360(6396)2018 06 29.
Article in English | MEDLINE | ID: mdl-29773667

ABSTRACT

The stem cell niche is a specialized environment that dictates stem cell function during development and homeostasis. We show that Dll1, a Notch pathway ligand, is enriched in mammary gland stem cells (MaSCs) and mediates critical interactions with stromal macrophages in the surrounding niche in mouse models. Conditional deletion of Dll1 reduced the number of MaSCs and impaired ductal morphogenesis in the mammary gland. Moreover, MaSC-expressed Dll1 activates Notch signaling in stromal macrophages, increasing their expression of Wnt family ligands such as Wnt3, Wnt10A, and Wnt16, thereby initiating a feedback loop that promotes the function of Dll1-expressing MaSCs. Together, these findings reveal functionally important cross-talk between MaSCs and their macrophageal niche through Dll1-mediated Notch signaling.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Macrophages/physiology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Receptors, Notch/metabolism , Stem Cell Niche/physiology , Stem Cells/physiology , Animals , Calcium-Binding Proteins , Cell Count , Female , Gene Knockout Techniques , Intercellular Signaling Peptides and Proteins/genetics , Ligands , Macrophages/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Morphogenesis , Signal Transduction , Stem Cells/cytology , Stromal Cells/cytology , Stromal Cells/physiology , Wnt Proteins/metabolism
15.
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
16.
Cell Rep ; 21(11): 3271-3284, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29241552

ABSTRACT

Adult neurogenesis in the murine dentate gyrus occurs in a specialized microenvironment that sustains the generation of neurons during life. To fully understand adult neurogenesis, it is essential to determine the neural stem cell (NSC) and progenitor developmental stages, their molecular determinants, and the niche cellular and molecular composition. We report on a single-cell RNA sequencing study of the hippocampal niche, performed by isolating all the non-neuronal cell populations. Our analysis provides a comprehensive description of the dentate gyrus cells, and it allows the identification of exclusive cell-type-specific markers. We define the developmental stages and transcriptional dynamics of NSCs and progenitors, and we find that, while NSCs represent a heterogeneous cellular continuum, progenitors can be grouped into distinct subtypes. We determine the oligodendrocyte lineage and transcriptional dynamics, and we describe the microglia transcriptional profile and activation state. The combined data constitute a valuable resource to understand regulatory mechanisms of adult neurogenesis.


Subject(s)
Dentate Gyrus/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , RNA, Messenger/genetics , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Lineage/genetics , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Female , Hippocampus/cytology , Hippocampus/growth & development , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/metabolism , Molecular Dynamics Simulation , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurons/cytology , Neurons/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , RNA, Messenger/metabolism , Sequence Analysis, RNA , Single-Cell Analysis/methods , Transcription, Genetic
17.
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
18.
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
19.
Cell Stem Cell ; 19(2): 205-216, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27184401

ABSTRACT

The E3 ubiquitin ligase Mule is often overexpressed in human colorectal cancers, but its role in gut tumorigenesis is unknown. Here, we show in vivo that Mule controls murine intestinal stem and progenitor cell proliferation by modulating Wnt signaling via c-Myc. Mule also regulates protein levels of the receptor tyrosine kinase EphB3 by targeting it for proteasomal and lysosomal degradation. In the intestine, EphB/ephrinB interactions position cells along the crypt-villus axis and compartmentalize incipient colorectal tumors. Our study thus unveils an important new avenue by which Mule acts as an intestinal tumor suppressor by regulation of the intestinal stem cell niche.


Subject(s)
Ephrin-B3/metabolism , Intestines/cytology , Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Stem Cell Niche , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway , Adenomatous Polyposis Coli/pathology , Alleles , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Endocytosis , HEK293 Cells , Humans , Mice, Knockout , Models, Biological , Mutation/genetics , Paneth Cells/pathology , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/deficiency
20.
Nat Commun ; 7: 10883, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26955760

ABSTRACT

Constitutive activation of Wnt/ß-catenin inhibits oligodendrocyte myelination. Tcf7l2/Tcf4, a ß-catenin transcriptional partner, is required for oligodendrocyte differentiation. How Tcf7l2 modifies ß-catenin signalling and controls myelination remains elusive. Here we define a stage-specific Tcf7l2-regulated transcriptional circuitry in initiating and sustaining oligodendrocyte differentiation. Multistage genome occupancy analyses reveal that Tcf7l2 serially cooperates with distinct co-regulators to control oligodendrocyte lineage progression. At the differentiation onset, Tcf7l2 interacts with a transcriptional co-repressor Kaiso/Zbtb33 to block ß-catenin signalling. During oligodendrocyte maturation, Tcf7l2 recruits and cooperates with Sox10 to promote myelination. In that context, Tcf7l2 directly activates cholesterol biosynthesis genes and cholesterol supplementation partially rescues oligodendrocyte differentiation defects in Tcf712 mutants. Together, we identify stage-specific co-regulators Kaiso and Sox10 that sequentially interact with Tcf7l2 to coordinate the switch at the transitions of differentiation initiation and maturation during oligodendrocyte development, and point to a previously unrecognized role of Tcf7l2 in control of cholesterol biosynthesis for CNS myelinogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Nervous System/growth & development , Oligodendroglia/metabolism , SOXE Transcription Factors/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Transcription Factors/metabolism , Animals , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System/metabolism , Protein Binding , SOXE Transcription Factors/genetics , Signal Transduction , Species Specificity , Transcription Factor 7-Like 2 Protein/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...