Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 15 de 15
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Methods Mol Biol ; 2811: 137-154, 2024.
Article de Anglais | MEDLINE | ID: mdl-39037655

RÉSUMÉ

The integration of CRISPR/Cas9 genome editing techniques with organoid technology has revolutionized the field of tumor modeling, enabling the creation of diverse tumor models with distinct mutational profiles. This protocol details the application of CRISPR knock-ins to engineer tumor organoids with reporter cassettes, which are regulated by endogenous promoters of specific genes of interest. This approach facilitates the precise fluorescent labeling, isolation, and subsequent manipulation of targeted tumor cell subpopulations. The utilization of these knock-in reporter cassettes not only allows the visualization and purification of specific tumor cell subsets but also enables conditional cell ablation and lineage tracing studies. In this chapter, we provide a comprehensive guide for the design, construction, delivery, and validation of CRISPR/Cas9 tools tailored for knock-in reporter cassette integration into specific marker genes of interest. By following this protocol, researchers can harness the potential of engineered tumor organoids to decipher intricate tumor heterogeneity, track metastatic trajectories, and unveil novel therapeutic vulnerabilities linked to specific tumor cell subpopulations.


Sujet(s)
Systèmes CRISPR-Cas , Édition de gène , Techniques de knock-in de gènes , Organoïdes , Organoïdes/métabolisme , Organoïdes/anatomopathologie , Humains , Techniques de knock-in de gènes/méthodes , Édition de gène/méthodes , Animaux , Tumeurs/génétique , Tumeurs/anatomopathologie , Gènes rapporteurs
2.
Nat Commun ; 15(1): 3363, 2024 Apr 18.
Article de Anglais | MEDLINE | ID: mdl-38637494

RÉSUMÉ

Colorectal cancer (CRC) tumors are composed of heterogeneous and plastic cell populations, including a pool of cancer stem cells that express LGR5. Whether these distinct cell populations display different mechanical properties, and how these properties might contribute to metastasis is poorly understood. Using CRC patient derived organoids (PDOs), we find that compared to LGR5- cells, LGR5+ cancer stem cells are stiffer, adhere better to the extracellular matrix (ECM), move slower both as single cells and clusters, display higher nuclear YAP, show a higher survival rate in response to mechanical confinement, and form larger transendothelial gaps. These differences are largely explained by the downregulation of the membrane to cortex attachment proteins Ezrin/Radixin/Moesin (ERMs) in the LGR5+ cells. By analyzing single cell RNA-sequencing (scRNA-seq) expression patterns from a patient cohort, we show that this downregulation is a robust signature of colorectal tumors. Our results show that LGR5- cells display a mechanically dynamic phenotype suitable for dissemination from the primary tumor whereas LGR5+ cells display a mechanically stable and resilient phenotype suitable for extravasation and metastatic growth.


Sujet(s)
Tumeurs colorectales , Récepteurs couplés aux protéines G , Humains , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Tumeurs colorectales/anatomopathologie , Cellules souches tumorales/métabolisme , Phénotype
3.
Nature ; 611(7936): 603-613, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-36352230

RÉSUMÉ

Around 30-40% of patients with colorectal cancer (CRC) undergoing curative resection of the primary tumour will develop metastases in the subsequent years1. Therapies to prevent disease relapse remain an unmet medical need. Here we uncover the identity and features of the residual tumour cells responsible for CRC relapse. An analysis of single-cell transcriptomes of samples from patients with CRC revealed that the majority of genes associated with a poor prognosis are expressed by a unique tumour cell population that we named high-relapse cells (HRCs). We established a human-like mouse model of microsatellite-stable CRC that undergoes metastatic relapse after surgical resection of the primary tumour. Residual HRCs occult in mouse livers after primary CRC surgery gave rise to multiple cell types over time, including LGR5+ stem-like tumour cells2-4, and caused overt metastatic disease. Using Emp1 (encoding epithelial membrane protein 1) as a marker gene for HRCs, we tracked and selectively eliminated this cell population. Genetic ablation of EMP1high cells prevented metastatic recurrence and mice remained disease-free after surgery. We also found that HRC-rich micrometastases were infiltrated with T cells, yet became progressively immune-excluded during outgrowth. Treatment with neoadjuvant immunotherapy eliminated residual metastatic cells and prevented mice from relapsing after surgery. Together, our findings reveal the cell-state dynamics of residual disease in CRC and anticipate that therapies targeting HRCs may help to avoid metastatic relapse.


Sujet(s)
Tumeurs colorectales , Métastase tumorale , Protéines tumorales , Récidive tumorale locale , Maladie résiduelle , Récepteurs de surface cellulaire , Animaux , Humains , Souris , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/thérapie , Évolution de la maladie , Protéines tumorales/déficit , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Récidive tumorale locale/génétique , Récidive tumorale locale/anatomopathologie , Récidive tumorale locale/prévention et contrôle , Récidive tumorale locale/thérapie , Maladie résiduelle/génétique , Maladie résiduelle/anatomopathologie , Récepteurs de surface cellulaire/déficit , Récepteurs de surface cellulaire/génétique , Récepteurs de surface cellulaire/métabolisme , Métastase tumorale/génétique , Métastase tumorale/anatomopathologie , Métastase tumorale/prévention et contrôle , Métastase tumorale/thérapie , Modèles animaux de maladie humaine , Lymphocytes T/cytologie , Lymphocytes T/immunologie , Lymphocytes TIL/cytologie , Lymphocytes TIL/immunologie , Traitement néoadjuvant , Immunothérapie
4.
J Exp Clin Cancer Res ; 41(1): 315, 2022 Oct 26.
Article de Anglais | MEDLINE | ID: mdl-36289544

RÉSUMÉ

BACKGROUND: Tumor-initiating cells (TIC), also known as cancer stem cells, are considered a specific subpopulation of cells necessary for cancer initiation and metastasis; however, the mechanisms by which they acquire metastatic traits are not well understood. METHODS: LAMC2 transcriptional levels were evaluated using publicly available transcriptome data sets, and LAMC2 immunohistochemistry was performed using a tissue microarray composed of PDAC and normal pancreas tissues. Silencing and tracing of LAMC2 was performed using lentiviral shRNA constructs and CRISPR/Cas9-mediated homologous recombination, respectively. The contribution of LAMC2 to PDAC tumorigenicity was explored in vitro by tumor cell invasion, migration, sphere-forming and organoids assays, and in vivo by tumor growth and metastatic assays. mRNA sequencing was performed to identify key cellular pathways upregulated in LAMC2 expressing cells. Metastatic spreading induced by LAMC2- expressing cells was blocked by pharmacological inhibition of transforming growth factor beta (TGF-ß) signaling. RESULTS: We report a LAMC2-expressing cell population, which is endowed with enhanced self-renewal capacity, and is sufficient for tumor initiation and differentiation, and drives metastasis. mRNA profiling of these cells indicates a prominent squamous signature, and differentially activated pathways critical for tumor growth and metastasis, including deregulation of the TGF-ß signaling pathway. Treatment with Vactosertib, a new small molecule inhibitor of the TGF-ß type I receptor (activin receptor-like kinase-5, ALK5), completely abrogated lung metastasis, primarily originating from LAMC2-expressing cells. CONCLUSIONS: We have identified a highly metastatic subpopulation of TICs marked by LAMC2. Strategies aimed at targeting the LAMC2 population may be effective in reducing tumor aggressiveness in PDAC patients. Our results prompt further study of this TIC population in pancreatic cancer and exploration as a potential therapeutic target and/or biomarker.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Humains , Carcinome du canal pancréatique/anatomopathologie , Récepteur de type I du facteur de croissance transformant bêta , Petit ARN interférent , Tumeurs du pancréas/anatomopathologie , Cellules souches tumorales/métabolisme , Facteur de croissance transformant bêta , ARN messager , Récepteur activine , Mouvement cellulaire/génétique , Lignée cellulaire tumorale , Laminine/génétique , Laminine/métabolisme , Tumeurs du pancréas
5.
Nat Cancer ; 3(9): 1052-1070, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35773527

RÉSUMÉ

Colorectal cancer (CRC) patient-derived organoids predict responses to chemotherapy. Here we used them to investigate relapse after treatment. Patient-derived organoids expand from highly proliferative LGR5+ tumor cells; however, we discovered that lack of optimal growth conditions specifies a latent LGR5+ cell state. This cell population expressed the gene MEX3A, is chemoresistant and regenerated the organoid culture after treatment. In CRC mouse models, Mex3a+ cells contributed marginally to metastatic outgrowth; however, after chemotherapy, Mex3a+ cells produced large cell clones that regenerated the disease. Lineage-tracing analysis showed that persister Mex3a+ cells downregulate the WNT/stem cell gene program immediately after chemotherapy and adopt a transient state reminiscent to that of YAP+ fetal intestinal progenitors. In contrast, Mex3a-deficient cells differentiated toward a goblet cell-like phenotype and were unable to resist chemotherapy. Our findings reveal that adaptation of cancer stem cells to suboptimal niche environments protects them from chemotherapy and identify a candidate cell of origin of relapse after treatment in CRC.


Sujet(s)
Tumeurs colorectales , Organoïdes , Animaux , Différenciation cellulaire , Tumeurs colorectales/traitement médicamenteux , Souris , Cellules souches tumorales , Récidive
6.
Nat Cancer ; 3(4): 418-436, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35469014

RÉSUMÉ

Patient-derived organoids (PDOs) recapitulate tumor architecture, contain cancer stem cells and have predictive value supporting personalized medicine. Here we describe a large-scale functional screen of dual-targeting bispecific antibodies (bAbs) on a heterogeneous colorectal cancer PDO biobank and paired healthy colonic mucosa samples. More than 500 therapeutic bAbs generated against Wingless-related integration site (WNT) and receptor tyrosine kinase (RTK) targets were functionally evaluated by high-content imaging to capture the complexity of PDO responses. Our drug discovery strategy resulted in the generation of MCLA-158, a bAb that specifically triggers epidermal growth factor receptor degradation in leucine-rich repeat-containing G-protein-coupled receptor 5-positive (LGR5+) cancer stem cells but shows minimal toxicity toward healthy LGR5+ colon stem cells. MCLA-158 exhibits therapeutic properties such as growth inhibition of KRAS-mutant colorectal cancers, blockade of metastasis initiation and suppression of tumor outgrowth in preclinical models for several epithelial cancer types.


Sujet(s)
Anticorps bispécifiques , Tumeurs épithéliales épidermoïdes et glandulaires , Anticorps bispécifiques/pharmacologie , Récepteurs ErbB/métabolisme , Humains , Imidazoles , Tumeurs épithéliales épidermoïdes et glandulaires/métabolisme , Cellules souches tumorales/métabolisme , Organoïdes , Pyrazines , Récepteurs couplés aux protéines G/métabolisme
7.
Cell Stem Cell ; 26(6): 845-861.e12, 2020 06 04.
Article de Anglais | MEDLINE | ID: mdl-32396863

RÉSUMÉ

Colorectal cancers (CRCs) are composed of an amalgam of cells with distinct genotypes and phenotypes. Here, we reveal a previously unappreciated heterogeneity in the biosynthetic capacities of CRC cells. We discover that the majority of ribosomal DNA transcription and protein synthesis in CRCs occurs in a limited subset of tumor cells that localize in defined niches. The rest of the tumor cells undergo an irreversible loss of their biosynthetic capacities as a consequence of differentiation. Cancer cells within the biosynthetic domains are characterized by elevated levels of the RNA polymerase I subunit A (POLR1A). Genetic ablation of POLR1A-high cell population imposes an irreversible growth arrest on CRCs. We show that elevated biosynthesis defines stemness in both LGR5+ and LGR5- tumor cells. Therefore, a common architecture in CRCs is a simple cell hierarchy based on the differential capacity to transcribe ribosomal DNA and synthesize proteins.


Sujet(s)
Tumeurs colorectales , Cellules souches tumorales , Lignée cellulaire tumorale , Tumeurs colorectales/génétique , ADN ribosomique , Humains , Récepteurs couplés aux protéines G
8.
EMBO Mol Med ; 9(7): 869-879, 2017 07.
Article de Anglais | MEDLINE | ID: mdl-28468934

RÉSUMÉ

The analysis of stem cell hierarchies in human cancers has been hampered by the impossibility of identifying or tracking tumor cell populations in an intact environment. To overcome this limitation, we devised a strategy based on editing the genomes of patient-derived tumor organoids using CRISPR/Cas9 technology to integrate reporter cassettes at desired marker genes. As proof of concept, we engineered human colorectal cancer (CRC) organoids that carry EGFP and lineage-tracing cassettes knocked in the LGR5 locus. Analysis of LGR5-EGFP+ cells isolated from organoid-derived xenografts demonstrated that these cells express a gene program similar to that of normal intestinal stem cells and that they propagate the disease to recipient mice very efficiently. Lineage-tracing experiments showed that LGR5+ CRC cells self-renew and generate progeny over long time periods that undergo differentiation toward mucosecreting- and absorptive-like phenotypes. These genetic experiments confirm that human CRCs adopt a hierarchical organization reminiscent of that of the normal colonic epithelium. The strategy described herein may have broad applications to study cell heterogeneity in human tumors.


Sujet(s)
Techniques de culture cellulaire/méthodes , Tumeurs colorectales/physiopathologie , Cellules souches tumorales/physiologie , Organoïdes , Animaux , Différenciation cellulaire , Prolifération cellulaire , Femelle , Édition de gène/méthodes , Techniques de knock-in de gènes , Gènes rapporteurs , Protéines à fluorescence verte/analyse , Protéines à fluorescence verte/génétique , Hétérogreffes , Humains , Souris SCID , Récepteurs couplés aux protéines G/génétique , Coloration et marquage/méthodes
9.
Cancer Res ; 77(7): 1730-1740, 2017 04 01.
Article de Anglais | MEDLINE | ID: mdl-28108514

RÉSUMÉ

The contribution of somatic mutations to metastasis of colorectal cancers is currently unknown. To find mutations involved in the colorectal cancer metastatic process, we performed deep mutational analysis of 676 genes in 107 stages II to IV primary colorectal cancer, of which half had metastasized. The mutation prevalence in the ephrin (EPH) family of tyrosine kinase receptors was 10-fold higher in primary tumors of metastatic colorectal than in nonmetastatic cases and preferentially occurred in stage III and IV tumors. Mutational analyses in situ confirmed expression of mutant EPH receptors. To enable functional studies of EPHB1 mutations, we demonstrated that DLD-1 colorectal cancer cells expressing EPHB1 form aggregates upon coculture with ephrin B1 expressing cells. When mutations in the fibronectin type III and kinase domains of EPHB1 were compared with wild-type EPHB1 in DLD-1 colorectal cancer cells, they decreased ephrin B1-induced compartmentalization. These observations provide a mechanistic link between EPHB receptor mutations and metastasis in colorectal cancer. Cancer Res; 77(7); 1730-40. ©2017 AACR.


Sujet(s)
Tumeurs colorectales/anatomopathologie , Mutation , Métastase tumorale , Récepteur EphB1/génétique , Lignée cellulaire tumorale , Tumeurs colorectales/génétique , Domaine fibronectine de type III/génétique , Humains , Stadification tumorale , Protein-tyrosine kinases/génétique
10.
Nat Genet ; 47(4): 320-9, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25706628

RÉSUMÉ

Recent molecular classifications of colorectal cancer (CRC) based on global gene expression profiles have defined subtypes displaying resistance to therapy and poor prognosis. Upon evaluation of these classification systems, we discovered that their predictive power arises from genes expressed by stromal cells rather than epithelial tumor cells. Bioinformatic and immunohistochemical analyses identify stromal markers that associate robustly with disease relapse across the various classifications. Functional studies indicate that cancer-associated fibroblasts (CAFs) increase the frequency of tumor-initiating cells, an effect that is dramatically enhanced by transforming growth factor (TGF)-ß signaling. Likewise, we find that all poor-prognosis CRC subtypes share a gene program induced by TGF-ß in tumor stromal cells. Using patient-derived tumor organoids and xenografts, we show that the use of TGF-ß signaling inhibitors to block the cross-talk between cancer cells and the microenvironment halts disease progression.


Sujet(s)
Tumeurs colorectales/diagnostic , Tumeurs colorectales/génétique , Fibroblastes/métabolisme , Cellules souches tumorales/métabolisme , Animaux , Analyse de regroupements , Tumeurs colorectales/classification , Tumeurs colorectales/anatomopathologie , Fibroblastes/anatomopathologie , Régulation de l'expression des gènes tumoraux , Cellules HT29 , Humains , Souris , Souris nude , Analyse sur microréseau , Invasion tumorale , Métastase tumorale , Cellules souches tumorales/anatomopathologie , Pronostic , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie , Transcriptome
11.
Nat Cell Biol ; 16(7): 695-707, 2014 Jul.
Article de Anglais | MEDLINE | ID: mdl-24952462

RÉSUMÉ

Aberrant activation of WNT signalling and loss of BMP signals represent the two main alterations leading to the initiation of colorectal cancer (CRC). Here we screen for genes required for maintaining the tumour stem cell phenotype and identify the zinc-finger transcription factor GATA6 as a key regulator of the WNT and BMP pathways in CRC. GATA6 directly drives the expression of LGR5 in adenoma stem cells whereas it restricts BMP signalling to differentiated tumour cells. Genetic deletion of Gata6 from mouse colon adenomas increases the levels of BMP factors, which signal to block self-renewal of tumour stem cells. In human tumours, GATA6 competes with ß-catenin/TCF4 for binding to a distal regulatory region of the BMP4 locus that has been linked to increased susceptibility to development of CRC. Hence, GATA6 creates an environment permissive for CRC initiation by lowering the threshold of BMP signalling required for tumour stem cell expansion.


Sujet(s)
Adénomes , Récepteurs de la protéine morphogénique osseuse/génétique , Tumeurs colorectales/physiopathologie , Facteur de transcription GATA-6/métabolisme , Régulation de l'expression des gènes tumoraux , Cellules souches/cytologie , Cellules souches/métabolisme , Adénomes/anatomopathologie , Animaux , Antinéoplasiques/pharmacologie , Récepteurs de la protéine morphogénique osseuse/métabolisme , Prolifération cellulaire , Femelle , Technique d'immunofluorescence , Facteur de transcription GATA-6/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Mâle , Souris , Pyrazoles/pharmacologie , Pyrimidines/pharmacologie , Cellules souches/effets des médicaments et des substances chimiques , Protéines de type Wingless/métabolisme
12.
Cell Stem Cell ; 13(3): 341-50, 2013 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-23871606

RÉSUMÉ

Since the initial discovery that OCT4, SOX2, KLF4, and c-MYC overexpression sufficed for the induction of pluripotency in somatic cells, methodologies replacing the original factors have enhanced our understanding of the reprogramming process. However, unlike in mouse, OCT4 has not been replaced successfully during reprogramming of human cells. Here we report on a strategy to accomplish this replacement. Through a combination of transcriptome and bioinformatic analysis we have identified factors previously characterized as being lineage specifiers that are able to replace OCT4 and SOX2 in the reprogramming of human fibroblasts. Our results show that it is possible to replace OCT4 and SOX2 simultaneously with alternative lineage specifiers in the reprogramming of human cells. At a broader level, they also support a model in which counteracting lineage specification networks underlies the induction of pluripotency.


Sujet(s)
Dédifférenciation cellulaire , Fibroblastes/physiologie , Facteur de transcription GATA-3/métabolisme , Facteur de transcription Oct-3/métabolisme , Cellules souches pluripotentes/physiologie , Dédifférenciation cellulaire/génétique , Lignage cellulaire , Cellules cultivées , Biologie informatique/méthodes , Facteur de transcription GATA-3/génétique , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes/génétique , Régénération tissulaire guidée , Humains , Facteur-4 de type Kruppel , Facteur de transcription Oct-3/génétique , Petit ARN interférent/génétique , Facteurs de transcription SOX-B1/génétique , Facteurs de transcription SOX-B1/métabolisme , Transgènes/génétique
13.
J Biol Chem ; 287(29): 24131-8, 2012 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-22613719

RÉSUMÉ

The tubular epithelium of the kidney is susceptible to injury from a number of different causes, including inflammatory and immune disorders, oxidative stress, and nephrotoxins, among others. Primary renal epithelial cells remain one of the few tools for studying the biochemical and physiological characteristics of the renal tubular system. Nevertheless, differentiated primary cells are not suitable for recapitulation of disease properties that might arise during embryonic kidney formation and further maturation. Thus, cellular systems resembling kidney characteristics are in urgent need to model disease as well as to establish reliable drug-testing platforms. Induced pluripotent stem cells (iPSCs) bear the capacity to differentiate into every cell lineage comprising the adult organism. Thus, iPSCs bring the possibility for recapitulating embryonic development by directed differentiation into specific lineages. iPSC differentiation ultimately allows for both disease modeling in vitro and the production of cellular products with potential for regenerative medicine. Here, we describe the rapid, reproducible, and highly efficient generation of iPSCs derived from endogenous kidney tubular renal epithelial cells with only two transcriptional factors, OCT4 and SOX2. Kidney-derived iPSCs may provide a reliable cellular platform for the development of kidney differentiation protocols allowing drug discovery studies and the study of kidney pathology.


Sujet(s)
Différenciation cellulaire/physiologie , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Tubules contournés proximaux/cytologie , Tubules contournés proximaux/métabolisme , Facteur de transcription Oct-3/métabolisme , Facteurs de transcription SOX-B1/métabolisme , Différenciation cellulaire/génétique , Cellules cultivées , Reprogrammation cellulaire/génétique , Reprogrammation cellulaire/physiologie , Technique d'immunofluorescence , Humains , Mâle , Adulte d'âge moyen , Facteur de transcription Oct-3/génétique , Facteurs de transcription SOX-B1/génétique
14.
Nat Cell Biol ; 13(9): 1100-7, 2011 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-21804545

RÉSUMÉ

The formation and maintenance of complex organs requires segregation of distinct cell populations into defined territories (that is, cell sorting) and the establishment of boundaries between them. Here we have investigated the mechanism by which Eph/ephrin signalling controls the compartmentalization of cells in epithelial tissues. We show that EphB/ephrin-B signalling in epithelial cells regulates the formation of E-cadherin-based adhesions. EphB receptors interact with E-cadherin and with the metalloproteinase ADAM10 at sites of adhesion and their activation induces shedding of E-cadherin by ADAM10 at interfaces with ephrin-B1-expressing cells. This process results in asymmetric localization of E-cadherin and, as a consequence, in differences in cell affinity between EphB-positive and ephrin-B-positive cells. Furthermore, genetic inhibition of ADAM10 activity in the intestine of mice results in a lack of compartmentalization of Paneth cells within the crypt stem cell niche, a defect that phenocopies that of EphB3-null mice. These results provide important insights into the regulation of cell migration in the intestinal epithelium and may help in the understanding of the nature of the cell sorting process in other epithelial tissues where Eph-ephrin interactions play a central role.


Sujet(s)
Protéines ADAM/métabolisme , Amyloid precursor protein secretases/métabolisme , Cadhérines/métabolisme , Cellules épithéliales/métabolisme , Protéines membranaires/métabolisme , Famille des récepteurs Eph/métabolisme , Transduction du signal , Protéines ADAM/génétique , Protéine ADAM10 , Amyloid precursor protein secretases/génétique , Animaux , Technique de Western , Cadhérines/génétique , Adhérence cellulaire , Lignée cellulaire , Lignée cellulaire tumorale , Cellules HEK293 , Humains , Muqueuse intestinale/métabolisme , Intestins/cytologie , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Protéines membranaires/génétique , Souris , Souris knockout , Souris transgéniques , Microscopie confocale , Microscopie de fluorescence , Cellules de Paneth/métabolisme , Liaison aux protéines , Interférence par ARN , Récepteur EphB1/génétique , Récepteur EphB1/métabolisme , Récepteur EphB3/génétique , Récepteur EphB3/métabolisme , Famille des récepteurs Eph/génétique , Niche de cellules souches
15.
Nat Genet ; 39(11): 1376-83, 2007 Nov.
Article de Anglais | MEDLINE | ID: mdl-17906625

RÉSUMÉ

The genes encoding tyrosine kinase receptors EphB2 and EphB3 are beta-catenin and Tcf4 target genes in colorectal cancer (CRC) and in normal intestinal cells. In the intestinal epithelium, EphB signaling controls the positioning of cell types along the crypt-villus axis. In CRC, EphB activity suppresses tumor progression beyond the earliest stages. Here we show that EphB receptors compartmentalize the expansion of CRC cells through a mechanism dependent on E-cadherin-mediated adhesion. We demonstrate that EphB-mediated compartmentalization restricts the spreading of EphB-expressing tumor cells into ephrin-B1-positive territories in vitro and in vivo. Our results indicate that CRC cells must silence EphB expression to avoid repulsive interactions imposed by normal ephrin-B1-expressing intestinal cells at the onset of tumorigenesis.


Sujet(s)
Tumeurs colorectales/métabolisme , Tumeurs colorectales/prévention et contrôle , Éphrine B1/métabolisme , Régulation de l'expression des gènes tumoraux , Adénomes/métabolisme , Adénomes/anatomopathologie , Adénomes/prévention et contrôle , Animaux , Lignée cellulaire tumorale , Tumeurs colorectales/anatomopathologie , Évolution de la maladie , Éphrine B1/antagonistes et inhibiteurs , Éphrine B1/génétique , Femelle , Gènes APC/physiologie , Humains , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Tumeurs de l'intestin/métabolisme , Tumeurs de l'intestin/anatomopathologie , Tumeurs de l'intestin/prévention et contrôle , Mâle , Souris , Souris de lignée C57BL , Souris knockout , ARN messager/antagonistes et inhibiteurs , ARN messager/génétique , ARN messager/métabolisme , Famille des récepteurs Eph/antagonistes et inhibiteurs , Famille des récepteurs Eph/génétique , Famille des récepteurs Eph/métabolisme , Transduction du signal , Fractions subcellulaires , Facteurs de transcription TCF/métabolisme , Protéine-2 de type facteur-7 de transcription , bêta-Caténine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE