Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Immunity ; 55(4): 701-717.e7, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35364006

ABSTRACT

Bacterial sensing by intestinal tumor cells contributes to tumor growth through cell-intrinsic activation of the calcineurin-NFAT axis, but the role of this pathway in other intestinal cells remains unclear. Here, we found that myeloid-specific deletion of calcineurin in mice activated protective CD8+ T cell responses and inhibited colorectal cancer (CRC) growth. Microbial sensing by myeloid cells promoted calcineurin- and NFAT-dependent interleukin 6 (IL-6) release, expression of the co-inhibitory molecules B7H3 and B7H4 by tumor cells, and inhibition of CD8+ T cell-dependent anti-tumor immunity. Accordingly, targeting members of this pathway activated protective CD8+ T cell responses and inhibited primary and metastatic CRC growth. B7H3 and B7H4 were expressed by the majority of human primary CRCs and metastases, which was associated with low numbers of tumor-infiltrating CD8+ T cells and poor survival. Therefore, a microbiota-, calcineurin-, and B7H3/B7H4-dependent pathway controls anti-tumor immunity, revealing additional targets for immune checkpoint inhibition in microsatellite-stable CRC.


Subject(s)
Colorectal Neoplasms , Microbiota , Animals , B7 Antigens , CD8-Positive T-Lymphocytes , Calcineurin/metabolism , Colorectal Neoplasms/metabolism , Mice , NFATC Transcription Factors/metabolism , V-Set Domain-Containing T-Cell Activation Inhibitor 1
2.
Cell ; 155(2): 357-68, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24120136

ABSTRACT

Proliferation of the self-renewing epithelium of the gastric corpus occurs almost exclusively in the isthmus of the glands, from where cells migrate bidirectionally toward pit and base. The isthmus is therefore generally viewed as the stem cell zone. We find that the stem cell marker Troy is expressed at the gland base by a small subpopulation of fully differentiated chief cells. By lineage tracing with a Troy-eGFP-ires-CreERT2 allele, single marked chief cells are shown to generate entirely labeled gastric units over periods of months. This phenomenon accelerates upon tissue damage. Troy(+) chief cells can be cultured to generate long-lived gastric organoids. Troy marks a specific subset of chief cells that display plasticity in that they are capable of replenishing entire gastric units, essentially serving as quiescent "reserve" stem cells. These observations challenge the notion that stem cell hierarchies represent a "one-way street."


Subject(s)
Chief Cells, Gastric/cytology , Stem Cells/cytology , Stomach/cytology , Animals , Cell Lineage , Chief Cells, Gastric/chemistry , Gastric Mucosa/cytology , Mice , Organoids/cytology , Receptors, Tumor Necrosis Factor/analysis , Wnt Signaling Pathway
3.
Nature ; 594(7863): 442-447, 2021 06.
Article in English | MEDLINE | ID: mdl-34079126

ABSTRACT

Interactions between tumour cells and the surrounding microenvironment contribute to tumour progression, metastasis and recurrence1-3. Although mosaic analyses in Drosophila have advanced our understanding of such interactions4,5, it has been difficult to engineer parallel approaches in vertebrates. Here we present an oncogene-associated, multicolour reporter mouse model-the Red2Onco system-that allows differential tracing of mutant and wild-type cells in the same tissue. By applying this system to the small intestine, we show that oncogene-expressing mutant crypts alter the cellular organization of neighbouring wild-type crypts, thereby driving accelerated clonal drift. Crypts that express oncogenic KRAS or PI3K secrete BMP ligands that suppress local stem cell activity, while changes in PDGFRloCD81+ stromal cells induced by crypts with oncogenic PI3K alter the WNT signalling environment. Together, these results show how oncogene-driven paracrine remodelling creates a niche environment that is detrimental to the maintenance of wild-type tissue, promoting field transformation dominated by oncogenic clones.


Subject(s)
Colorectal Neoplasms/pathology , Intestine, Small/pathology , Neoplastic Stem Cells/pathology , Oncogenes , Stem Cell Niche , Animals , Clone Cells/pathology , Colorectal Neoplasms/genetics , Female , Intestine, Small/metabolism , Male , Mice , Mutation , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Reproducibility of Results , Single-Cell Analysis , Stem Cell Niche/genetics , Tumor Microenvironment , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway
4.
Mol Cancer ; 23(1): 10, 2024 01 10.
Article in English | MEDLINE | ID: mdl-38200602

ABSTRACT

BACKGROUND AND AIMS: This study sought to determine the value of patient-derived organoids (PDOs) from esophago-gastric adenocarcinoma (EGC) for response prediction to neoadjuvant chemotherapy (neoCTx). METHODS: Endoscopic biopsies of patients with locally advanced EGC (n = 120) were taken into culture and PDOs expanded. PDOs' response towards the single substances of the FLOT regimen and the combination treatment were correlated to patients' pathological response using tumor regression grading. A classifier based on FLOT response of PDOs was established in an exploratory cohort (n = 13) and subsequently confirmed in an independent validation cohort (n = 13). RESULTS: EGC PDOs reflected patients' diverse responses to single chemotherapeutics and the combination regimen FLOT. In the exploratory cohort, PDOs response to single 5-FU and FLOT combination treatment correlated with the patients' pathological response (5-FU: Kendall's τ = 0.411, P = 0.001; FLOT: Kendall's τ = 0.694, P = 2.541e-08). For FLOT testing, a high diagnostic precision in receiver operating characteristic (ROC) analysis was reached with an AUCROC of 0.994 (CI 0.980 to 1.000). The discriminative ability of PDO-based FLOT testing allowed the definition of a threshold, which classified in an independent validation cohort FLOT responders from non-responders with high sensitivity (90%), specificity (100%) and accuracy (92%). CONCLUSION: In vitro drug testing of EGC PDOs has a high predictive accuracy in classifying patients' histological response to neoadjuvant FLOT treatment. Taking into account the high rate of successful PDO expansion from biopsies, the definition of a threshold that allows treatment stratification paves the way for an interventional trial exploring PDO-guided treatment of EGC patients.


Subject(s)
Adenocarcinoma , Carbamates , Pyrazines , Pyridines , Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Combined Modality Therapy , Neoadjuvant Therapy , Adenocarcinoma/drug therapy , Organoids , Fluorouracil/pharmacology
5.
Cell ; 136(5): 903-12, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19269367

ABSTRACT

The small intestinal epithelium is the most rapidly self-renewing tissue of mammals. Proliferative cells are confined to crypts, while differentiated cell types predominantly occupy the villi. We recently demonstrated the existence of a long-lived pool of cycling stem cells defined by Lgr5 expression and intermingled with post-mitotic Paneth cells at crypt bottoms. We have now determined a gene signature for these Lgr5 stem cells. One of the genes within this stem cell signature is the Wnt target Achaete scute-like 2 (Ascl2). Transgenic expression of the Ascl2 transcription factor throughout the intestinal epithelium induces crypt hyperplasia and ectopic crypts on villi. Induced deletion of the Ascl2 gene in adult small intestine leads to disappearance of the Lgr5 stem cells within days. The combined results from these gain- and loss-of-function experiments imply that Ascl2 controls intestinal stem cell fate.


Subject(s)
Adult Stem Cells/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Intestine, Small/cytology , Animals , Cell Separation , Gene Deletion , Gene Expression Profiling , Mice , Mice, Transgenic
6.
Gastroenterology ; 162(4): 1067-1087, 2022 04.
Article in English | MEDLINE | ID: mdl-34942172

ABSTRACT

Several genetic and environmental factors increase gastric cancer (GC) risk, with Helicobacter pylori being the main environmental agent. GC is thought to emerge through a sequence of morphological changes that have been elucidated on the molecular level. New technologies have shed light onto pathways that are altered in GC, involving mutational and epigenetic changes and altered signaling pathways. Using various new model systems and innovative approaches, the relevance of such alterations for the emergence and progression of GC has been validated. Here, we highlight the key strategies and the resulting achievements. A major step is the characterization of epithelial stem cell behavior in the healthy stomach. These data, obtained through new reporter mouse lines and lineage tracing, enabled insights into the processes that control cellular proliferation, self-renewal, and differentiation of gastric stem cells. It has become evident that these cells and pathways are often deregulated in carcinogenesis. Second, insights into how H pylori colonizes gastric glands, directly interacts with stem cells, and alters cellular and genomic integrity, as well as the characterization of tissue responses to infection, provide a comprehensive picture of how this bacterium contributes to gastric carcinogenesis. Third, the development of stem cell- and tissue-specific reporter mice have driven our understanding of the signals and mutations that promote different types of GC and now also enable the study of more advanced, metastasized stages. Finally, organoids from human tissue have allowed insights into gastric carcinogenesis by validating mutational and signaling alterations in human primary cells and opening a route to predicting responses to personalized treatment.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Animals , Carcinogenesis/pathology , Gastric Mucosa/pathology , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Humans , Mice , Mutation , Stem Cells/metabolism , Stomach Neoplasms/microbiology
7.
J Pathol ; 257(5): 607-619, 2022 08.
Article in English | MEDLINE | ID: mdl-35373359

ABSTRACT

Drug combination therapies for cancer treatment show high efficacy but often induce severe side effects, resulting in dose or cycle number reduction. We investigated the impact of neoadjuvant chemotherapy (neoCTx) adaptions on treatment outcome in 59 patients with pancreatic ductal adenocarcinoma (PDAC). Resections with tumor-free margins were significantly more frequent when full-dose neoCTx was applied. We determined if patient-derived organoids (PDOs) can be used to personalize poly-chemotherapy regimens by pharmacotyping of treatment-naïve and post-neoCTx PDAC PDOs. Five out of ten CTx-naïve PDO lines exhibited a differential response to either the FOLFIRINOX or the Gem/Pac regimen. NeoCTx PDOs showed a poor response to the neoadjuvant regimen that had been administered to the respective patient in 30% of cases. No significant difference in PDO response was noted when comparing modified treatments in which the least effective single drug was removed from the complete regimen. Drug testing of CTx-naïve PDAC PDOs and neoCTx PDOs may be useful to guide neoadjuvant and adjuvant regimen selection, respectively. Personalizing poly-chemotherapy regimens by omitting substances with low efficacy could potentially result in less severe side effects, thereby increasing the fraction of patients receiving a full course of neoadjuvant treatment. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Adenocarcinoma/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Drug Resistance , Humans , Neoadjuvant Therapy , Organoids/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms
8.
Langenbecks Arch Surg ; 407(4): 1-11, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35501604

ABSTRACT

PURPOSE: Robotic-assisted minimally invasive esophagectomy (RAMIE) has become one standard approach for the operative treatment of esophageal tumors at specialized centers. Here, we report the results of a prospective multicenter registry for standardized RAMIE. METHODS: The German da Vinci Xi registry trial included all consecutive patients who underwent RAMIE at five tertiary university centers between Oct 17, 2017, and Jun 5, 2020. RAMIE was performed according to a standard technique using an intrathoracic circular stapled esophagogastrostomy. RESULTS: A total of 220 patients were included. The median age was 64 years. Total minimally invasive RAMIE was accomplished in 85.9%; hybrid resection with robotic-assisted thoracic approach was accomplished in an additional 11.4%. A circular stapler size of ≥28 mm was used in 84%, and the median blood loss and operative time were 200 (IQR: 80-400) ml and 425 (IQR: 335-527) min, respectively. The rate of anastomotic leakage was 13.2% (n=29), whereas the two centers with >70 cases each had rates of 7.0% and 12.0%. Pneumonia occurred in 19.5% of patients, and the 90-day mortality was 3.6%. Cumulative sum analysis of the operative time indicated the end of the learning curve after 22 cases. CONCLUSIONS: High-quality multicenter registry data confirm that RAMIE is a safe procedure and can be reproduced with acceptable leak rates in a multicenter setting. The learning curve is comparably low for experienced robotic surgeons.


Subject(s)
Esophageal Neoplasms , Robotic Surgical Procedures , Esophageal Neoplasms/pathology , Esophageal Neoplasms/surgery , Esophagectomy/methods , Humans , Middle Aged , Minimally Invasive Surgical Procedures/methods , Prospective Studies , Registries , Robotic Surgical Procedures/methods
9.
Gut ; 70(9): 1632-1641, 2021 09.
Article in English | MEDLINE | ID: mdl-33199443

ABSTRACT

OBJECTIVE: Epidermal growth factor receptor (EGFR) inhibition may be effective in biomarker-selected populations of advanced gastro-oesophageal adenocarcinoma (aGEA) patients. Here, we tested the association between outcome and EGFR copy number (CN) in pretreatment tissue and plasma cell-free DNA (cfDNA) of patients enrolled in a randomised first-line phase III clinical trial of chemotherapy or chemotherapy plus the anti-EGFR monoclonal antibody panitumumab in aGEA (NCT00824785). DESIGN: EGFR CN by either fluorescence in situ hybridisation (n=114) or digital-droplet PCR in tissues (n=250) and plasma cfDNAs (n=354) was available for 474 (86%) patients in the intention-to-treat (ITT) population. Tissue and plasma low-pass whole-genome sequencing was used to screen for coamplifications in receptor tyrosine kinases. Interaction between chemotherapy and EGFR inhibitors was modelled in patient-derived organoids (PDOs) from aGEA patients. RESULTS: EGFR amplification in cfDNA correlated with poor survival in the ITT population and similar trends were observed when the analysis was conducted in tissue and plasma by treatment arm. EGFR inhibition in combination with chemotherapy did not correlate with improved survival, even in patients with significant EGFR CN gains. Addition of anti-EGFR inhibitors to the chemotherapy agent epirubicin in PDOs, resulted in a paradoxical increase in viability and accelerated progression through the cell cycle, associated with p21 and cyclin B1 downregulation and cyclin E1 upregulation, selectively in organoids from EGFR-amplified aGEA. CONCLUSION: EGFR CN can be accurately measured in tissue and liquid biopsies and may be used for the selection of aGEA patients. EGFR inhibitors may antagonise the antitumour effect of anthracyclines with important implications for the design of future combinatorial trials.


Subject(s)
Adenocarcinoma/drug therapy , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Epirubicin/therapeutic use , ErbB Receptors/antagonists & inhibitors , Esophageal Neoplasms/drug therapy , Panitumumab/therapeutic use , Stomach Neoplasms/drug therapy , Adenocarcinoma/chemistry , Aged , Antibiotics, Antineoplastic/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , Biomarkers, Tumor/analysis , Epirubicin/administration & dosage , ErbB Receptors/analysis , Esophageal Neoplasms/chemistry , Humans , Male , Middle Aged , Panitumumab/administration & dosage , Stomach Neoplasms/chemistry
10.
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
11.
Gastroenterology ; 157(6): 1599-1614.e2, 2019 12.
Article in English | MEDLINE | ID: mdl-31585123

ABSTRACT

BACKGROUND & AIMS: Patterns of genetic alterations characterize different molecular subtypes of human gastric cancer. We aimed to establish mouse models of these subtypes. METHODS: We searched databases to identify genes with unique expression in the stomach epithelium, resulting in the identification of Anxa10. We generated mice with tamoxifen-inducible Cre recombinase (CreERT2) in the Anxa10 gene locus. We created 3 mouse models with alterations in pathways that characterize the chromosomal instability (CIN) and the genomically stable (GS) subtypes of human gastric cancer: Anxa10-CreERT2;KrasG12D/+;Tp53R172H/+;Smad4fl/f (CIN mice), Anxa10-CreERT2;Cdh1fl/fl;KrasG12D/+;Smad4fl/fl (GS-TGBF mice), and Anxa10-CreERT2;Cdh1fl/fl;KrasG12D/+;Apcfl/fl (GS-Wnt mice). We analyzed tumors that developed in these mice by histology for cell types and metastatic potential. We derived organoids from the tumors and tested their response to chemotherapeutic agents and the epithelial growth factor receptor signaling pathway inhibitor trametinib. RESULTS: The gastric tumors from the CIN mice had an invasive phenotype and formed liver and lung metastases. The tumor cells had a glandular morphology, similar to human intestinal-type gastric cancer. The gastric tumors from the GS-TGFB mice were poorly differentiated with diffuse morphology and signet ring cells, resembling human diffuse-type gastric cancer. Cells from these tumors were invasive, and mice developed peritoneal carcinomatosis and lung metastases. GS-Wnt mice developed adenomatous tooth-like gastric cancer. Organoids derived from tumors of GS-TGBF and GS-Wnt mice were more resistant to docetaxel, whereas organoids from the CIN tumors were more resistant to trametinib. CONCLUSIONS: Using a stomach-specific CreERT2 system, we created mice that develop tumors with morphologic similarities to subtypes of human gastric cancer. These tumors have different patterns of local growth, metastasis, and response to therapeutic agents. They can be used to study different subtypes of human gastric cancer.


Subject(s)
Disease Models, Animal , Gastric Mucosa/pathology , Genetic Loci/genetics , Stomach Neoplasms/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , Annexins/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Transformation, Neoplastic/genetics , Drug Resistance, Neoplasm/genetics , Female , Humans , Integrases/genetics , Male , Mice , Mice, Transgenic , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Smad4 Protein/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/genetics
12.
Gut ; 68(2): 207-217, 2019 02.
Article in English | MEDLINE | ID: mdl-29703791

ABSTRACT

OBJECTIVE: Gastric cancer is the second leading cause of cancer-related deaths and the fifth most common malignancy worldwide. In this study, human and mouse gastric cancer organoids were generated to model the disease and perform drug testing to delineate treatment strategies. DESIGN: Human gastric cancer organoid cultures were established, samples classified according to their molecular profile and their response to conventional chemotherapeutics tested. Targeted treatment was performed according to specific druggable mutations. Mouse gastric cancer organoid cultures were generated carrying molecular subtype-specific alterations. RESULTS: Twenty human gastric cancer organoid cultures were established and four selected for a comprehensive in-depth analysis. Organoids demonstrated divergent growth characteristics and morphologies. Immunohistochemistry showed similar characteristics to the corresponding primary tissue. A divergent response to 5-fluoruracil, oxaliplatin, irinotecan, epirubicin and docetaxel treatment was observed. Whole genome sequencing revealed a mutational spectrum that corresponded to the previously identified microsatellite instable, genomic stable and chromosomal instable subtypes of gastric cancer. The mutational landscape allowed targeted therapy with trastuzumab for ERBB2 alterations and palbociclib for CDKN2A loss. Mouse cancer organoids carrying Kras and Tp53 or Apc and Cdh1 mutations were characterised and serve as model system to study the signalling of induced pathways. CONCLUSION: We generated human and mouse gastric cancer organoids modelling typical characteristics and altered pathways of human gastric cancer. Successful interference with activated pathways demonstrates their potential usefulness as living biomarkers for therapy response testing.


Subject(s)
Disease Models, Animal , Organoids/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Animals , Cdh1 Proteins/genetics , Genes, APC , Humans , Immunohistochemistry , Mice , Mutation , Organ Culture Techniques , Piperazines/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Pyridines/pharmacology , Stomach Neoplasms/drug therapy , Trastuzumab/pharmacology , Tumor Suppressor Protein p53/genetics
13.
Nature ; 488(7413): 665-9, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22895187

ABSTRACT

LGR5+ stem cells reside at crypt bottoms, intermingled with Paneth cells that provide Wnt, Notch and epidermal growth factor signals. Here we find that the related RNF43 and ZNRF3 transmembrane E3 ubiquitin ligases are uniquely expressed in LGR5+ stem cells. Simultaneous deletion of the two genes encoding these proteins in the intestinal epithelium of mice induces rapidly growing adenomas containing high numbers of Paneth and LGR5+ stem cells. In vitro, growth of organoids derived from these adenomas is arrested when Wnt secretion is inhibited, indicating a dependence of the adenoma stem cells on Wnt produced by adenoma Paneth cells. In the HEK293T human cancer cell line, expression of RNF43 blocks Wnt responses and targets surface-expressed frizzled receptors to lysosomes. In the RNF43-mutant colorectal cancer cell line HCT116, reconstitution of RNF43 expression removes its response to exogenous Wnt. We conclude that RNF43 and ZNRF3 reduce Wnt signals by selectively ubiquitinating frizzled receptors, thereby targeting these Wnt receptors for degradation.


Subject(s)
DNA-Binding Proteins/metabolism , Endocytosis , Oncogene Proteins/metabolism , Receptors, Wnt/metabolism , Stem Cells/enzymology , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway , Adenoma/metabolism , Adenoma/pathology , Animals , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Frizzled Receptors/metabolism , HEK293 Cells , Humans , Lysosomes/metabolism , Mice , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Organoids/cytology , Organoids/metabolism , Organoids/pathology , Paneth Cells/metabolism , Paneth Cells/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Wnt/antagonists & inhibitors , Stem Cells/cytology , Stem Cells/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
14.
Dev Biol ; 420(2): 239-250, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27521455

ABSTRACT

The gastrointestinal tract is characterized by a self-renewing epithelium fueled by adult stem cells residing at the bottom of the intestinal crypt and gastric glands. Their activity and proliferation is strongly dependent on complex signaling pathways involving other crypt/gland cells as well as surrounding stromal cells. In recent years organoids are becoming increasingly popular as a new and powerful tool to study developmental or other biological processes. Organoids retain morphological and molecular patterns of the tissue they are derived from, are self-organizing, relatively simple to handle and accessible to genetic engineering. This review focuses on the developmental processes and signaling molecules involved in epithelial homeostasis and how a profound knowledge of these mechanisms allowed the establishment of a three dimensional organoid culture derived from adult gastrointestinal stem cells.


Subject(s)
Gastrointestinal Tract/growth & development , Organoids/growth & development , Adult Stem Cells/cytology , Animals , Cell Differentiation , Gastrointestinal Tract/cytology , Gastrointestinal Tract/physiology , Humans , Mice , Models, Biological , Organ Culture Techniques/methods , Organogenesis , Organoids/cytology , Organoids/physiology , Regeneration , Signal Transduction
15.
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
16.
Nature ; 476(7360): 293-7, 2011 Jul 04.
Article in English | MEDLINE | ID: mdl-21727895

ABSTRACT

The adult stem cell marker Lgr5 and its relative Lgr4 are often co-expressed in Wnt-driven proliferative compartments. We find that conditional deletion of both genes in the mouse gut impairs Wnt target gene expression and results in the rapid demise of intestinal crypts, thus phenocopying Wnt pathway inhibition. Mass spectrometry demonstrates that Lgr4 and Lgr5 associate with the Frizzled/Lrp Wnt receptor complex. Each of the four R-spondins, secreted Wnt pathway agonists, can bind to Lgr4, -5 and -6. In HEK293 cells, RSPO1 enhances canonical WNT signals initiated by WNT3A. Removal of LGR4 does not affect WNT3A signalling, but abrogates the RSPO1-mediated signal enhancement, a phenomenon rescued by re-expression of LGR4, -5 or -6. Genetic deletion of Lgr4/5 in mouse intestinal crypt cultures phenocopies withdrawal of Rspo1 and can be rescued by Wnt pathway activation. Lgr5 homologues are facultative Wnt receptor components that mediate Wnt signal enhancement by soluble R-spondin proteins. These results will guide future studies towards the application of R-spondins for regenerative purposes of tissues expressing Lgr5 homologues.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Thrombospondins/metabolism , Wnt Proteins/metabolism , Adult Stem Cells/metabolism , Animals , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Frizzled Receptors/metabolism , Gene Deletion , HEK293 Cells , Humans , Mice , Protein Binding , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Regeneration , Signal Transduction/genetics , Wnt Proteins/genetics , Wnt3 Protein , Wnt3A Protein
17.
EMBO J ; 31(14): 3079-91, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22692129

ABSTRACT

Two types of stem cells are currently defined in small intestinal crypts: cycling crypt base columnar (CBC) cells and quiescent '+4' cells. Here, we combine transcriptomics with proteomics to define a definitive molecular signature for Lgr5(+) CBC cells. Transcriptional profiling of FACS-sorted Lgr5(+) stem cells and their daughters using two microarray platforms revealed an mRNA stem cell signature of 384 unique genes. Quantitative mass spectrometry on the same cell populations identified 278 proteins enriched in intestinal stem cells. The mRNA and protein data sets showed a high level of correlation and a combined signature of 510 stem cell-enriched genes was defined. Spatial expression patterns were further characterized by mRNA in-situ hybridization, revealing that approximately half of the genes were expressed in a gradient with highest levels at the crypt bottom, while the other half was expressed uniquely in Lgr5(+)stem cells. Lineage tracing using a newly established knock-in mouse for one of the signature genes, Smoc2, confirmed its stem cell specificity. Using this resource, we find-and confirm by independent approaches-that the proposed quiescent/'+4' stem cell markers Bmi1, Tert, Hopx and Lrig1 are robustly expressed in CBC cells.


Subject(s)
Antigens, Differentiation/metabolism , Gene Expression Regulation , Intestinal Mucosa/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Animals , Antigens, Differentiation/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Gene Expression Profiling , Intestines/cytology , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/genetics , Stem Cells/cytology
18.
Nature ; 459(7244): 262-5, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19329995

ABSTRACT

The intestinal epithelium is the most rapidly self-renewing tissue in adult mammals. We have recently demonstrated the presence of about six cycling Lgr5(+) stem cells at the bottoms of small-intestinal crypts. Here we describe the establishment of long-term culture conditions under which single crypts undergo multiple crypt fission events, while simultanously generating villus-like epithelial domains in which all differentiated cell types are present. Single sorted Lgr5(+) stem cells can also initiate these cryptvillus organoids. Tracing experiments indicate that the Lgr5(+) stem-cell hierarchy is maintained in organoids. We conclude that intestinal cryptvillus units are self-organizing structures, which can be built from a single stem cell in the absence of a non-epithelial cellular niche.


Subject(s)
Cell Culture Techniques/methods , Intestines/anatomy & histology , Intestines/cytology , Organoids/cytology , Receptors, G-Protein-Coupled/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Lineage , Cell Separation , Gene Expression Regulation, Developmental , Intestinal Mucosa/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Organoids/growth & development , Organoids/metabolism , Paneth Cells/metabolism , Receptors, Notch/metabolism , Regeneration , Stem Cell Niche
19.
Nat Methods ; 9(1): 81-3, 2011 Dec 04.
Article in English | MEDLINE | ID: mdl-22138822

ABSTRACT

The study of gene function in endodermal epithelia such as of stomach, small intestine and colon relies heavily on transgenic approaches. Establishing such animal models is laborious, expensive and time-consuming. We present here a method based on Cre recombinase-inducible retrovirus vectors that allows the conditional manipulation of gene expression in primary mouse organoid culture systems.


Subject(s)
Intestinal Mucosa/cytology , Organoids/cytology , Receptors, G-Protein-Coupled/physiology , Animals , Gene Expression , Gene Knockdown Techniques/methods , Green Fluorescent Proteins/genetics , Humans , Integrases/metabolism , Mice , Organoids/metabolism , Receptors, Notch/physiology , Retroviridae/genetics , Stem Cells/virology , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Transduction, Genetic/methods
20.
Stem Cells ; 31(11): 2287-95, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23836510

ABSTRACT

The intestine has developed over the last few years into a prime model system for adult stem cell research. Intestinal cells have an average lifetime of 5 days, moving within this time from the bottom of intestinal crypts to the top of villi. This rapid self-renewal capacity combined with an easy to follow (mostly) unidirectional movement of cells offers an ideal site to conduct adult stem cell research. The delineation of the active pathways in the intestinal epithelium together with the development of molecular techniques to prove stemness laid the grounds for the identification of the intestinal stem cell. In vitro systems and transgenic mouse models broaden our knowledge on the role of the stem cell niche and those cells that reestablish homeostasis after perturbation of the system. These insights expedited also research on the role of normal adult stem cells in cancer initiation and the factors influencing the maintenance of cancer stem cells.


Subject(s)
Adult Stem Cells/cytology , Intestinal Mucosa/cytology , Intestinal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Adult Stem Cells/pathology , Animals , Cell Growth Processes/physiology , Humans , Intestinal Mucosa/pathology , Stem Cell Niche
SELECTION OF CITATIONS
SEARCH DETAIL